US20190031784A1 - T cell activating bispecific antigen binding molecules - Google Patents

T cell activating bispecific antigen binding molecules Download PDF

Info

Publication number
US20190031784A1
US20190031784A1 US16/016,209 US201816016209A US2019031784A1 US 20190031784 A1 US20190031784 A1 US 20190031784A1 US 201816016209 A US201816016209 A US 201816016209A US 2019031784 A1 US2019031784 A1 US 2019031784A1
Authority
US
United States
Prior art keywords
seq
antigen binding
folr1
antigen
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/016,209
Inventor
Marina Bacac
Peter Bruenker
Anne Freimoser-Grundschober
Ralf Hosse
Christian Klein
Ekkehard Moessner
Pablo Umana
Tina Weinzierl
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.)
Hoffmann La Roche Inc
Original Assignee
Hoffmann La Roche 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 Hoffmann La Roche Inc filed Critical Hoffmann La Roche Inc
Priority to US16/016,209 priority Critical patent/US20190031784A1/en
Publication of US20190031784A1 publication Critical patent/US20190031784A1/en
Priority to US17/580,248 priority patent/US20220220224A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • 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
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/66Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a swap of domains, e.g. CH3-CH2, VH-CL or VL-CH1
    • 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
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention generally relates to bispecific antigen binding molecules for activating T cells, in particular to bispecific antibodies targeting CD3 and Folate Receptor 1 (FolR1).
  • the present invention relates to polynucleotides encoding such bispecific antigen binding molecules, and vectors and host cells comprising such polynucleotides.
  • the invention further relates to methods for producing the bispecific antigen binding molecules of the invention, and to methods of using these bispecific antigen binding molecules in the treatment of disease.
  • the selective destruction of an individual cell or a specific cell type is often desirable in a variety of clinical settings. For example, it is a primary goal of cancer therapy to specifically destroy tumor cells, while leaving healthy cells and tissues intact and undamaged.
  • CTLs constitute the most potent effector cells of the immune system, however they cannot be activated by the effector mechanism mediated by the Fc domain of conventional therapeutic antibodies.
  • bispecific antibodies designed to bind with one “arm” to a surface antigen on target cells, and with the second “arm” to an activating, invariant component of the T cell receptor (TCR) complex, have become of interest in recent years.
  • TCR T cell receptor
  • the simultaneous binding of such an antibody to both of its targets will force a temporary interaction between target cell and T cell, causing activation of any cytotoxic T cell and subsequent lysis of the target cell.
  • the immune response is re-directed to the target cells and is independent of peptide antigen presentation by the target cell or the specificity of the T cell as would be relevant for normal MHC-restricted activation of CTLs.
  • CTLs are only activated when a target cell is presenting the bispecific antibody to them, i.e. the immunological synapse is mimicked.
  • bispecific antibodies that do not require lymphocyte preconditioning or co-stimulation in order to elicit efficient lysis of target cells.
  • FOLR1 is expressed on epithelial tumor cells of various origins, e.g., ovarian cancer, lung cancer, breast cancer, renal cancer, colorectal cancer, endometrial cancer.
  • ovarian cancer ovarian cancer
  • lung cancer breast cancer
  • renal cancer colorectal cancer
  • endometrial cancer endometrial cancer.
  • therapeutic antibodies such as farletuzumab, antibody drug conjugates, or adoptive T cell therapy for imaging of tumors
  • the present invention provides bispecific antigen binding molecules designed for targeted T cell activation, particularly, bispecific antigen binding molecules suitable as effective and safe therapeutics that can be readily produced and dosed.
  • the invention provides a T cell activating bispecific antigen binding molecule comprising
  • the T cell activating bispecific antigen binding molecule comprises a first antigen binding moiety that comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • the T cell activating bispecific antigen binding molecule additionally comprises (iii) a third antigen binding moiety capable of specific binding to FolR1.
  • the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences.
  • the third antigen binding moiety is identical to the second antigen binding moiety.
  • the T cell activating bispecific antigen binding molecule of the above embodiments at least one of the second and third antigen binding moiety is a Fab molecule.
  • the T cell activating bispecific antigen binding molecule of the above embodiments additionally comprises an Fc domain composed of a first and a second subunit capable of stable association.
  • the first antigen binding moiety and the second antigen binding moiety are each connected at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.
  • a third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, optionally via a peptide linker.
  • the antigen binding moiety capable of specific binding to Folate Receptor 1 comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • the antigen binding moiety capable of specific binding to Folate Receptor 1 comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
  • the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • the antigen binding moiety capable of specific binding to Folate Receptor 1 comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 50 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
  • CDR heavy chain complementarity determining region
  • the antigen binding moiety capable of specific binding to FolR1 comprises (a) a complementarity determining region heavy chain 1 (CDR-H1) amino acid sequences of SEQ ID NO: 8; (b) a CDR-H2 amino acid sequence of SEQ ID NO: 9; (c) a CDR-H3 amino acid sequence of SEQ ID NO: 50; (d) a complementarity determining region light chain 1 (CDR-L1) amino acid sequence of SEQ ID NO: 52; (e) a CDR-L2 amino acid sequence of SEQ ID NO: 53, and (f) a CDR-L3 amino acid sequence of SEQ ID NO: 54.
  • the antigen binding moiety capable of specific binding to FolR1 comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • the antigen binding moiety capable of specific binding to Folate Receptor 1 comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 275 and SEQ ID NO: 315 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • CDR heavy chain complementarity determining region
  • the antigen binding moiety capable of specific binding to FolR1 comprises (a) a complementarity determining region heavy chain 1 (CDR-H1) amino acid sequences of SEQ ID NO: 16; (b) a CDR-H2 amino acid sequence of SEQ ID NO: 275; (c) a CDR-H3 amino acid sequence of SEQ ID NO: 315; (d) a complementarity determining region light chain 1 (CDR-L1) amino acid sequence of SEQ ID NO: 32; (e) a CDR-L2 amino acid sequence of SEQ ID NO: 33, and (f) a CDR-L3 amino acid sequence of SEQ ID NO: 34.
  • the antigen binding moiety capable of specific binding to FolR1 comprises a variable heavy chain domain (VH) comprising an amino acid sequence of SEQ ID NO: 274 and a variable light chain domain (VL) comprising an amino acid sequence of SEQ ID NO: 31.
  • VH variable heavy chain domain
  • VL variable light chain domain
  • the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1 and a cynomolgus monkey FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1, a cynomolgus monkey FolR1 and a murine FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1, a cynomolgus monkey FolR1 and not a murine FolR1.
  • the first antigen binding moiety is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments comprises not more than one antigen binding moiety capable of specific binding to CD3.
  • the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule.
  • the Fc domain is an IgG class immunoglobulin, specifically an IgG 1 or IgG 4 , Fc domain.
  • the Fc domain is a human Fc domain.
  • the Fc domain comprises a modification promoting the association of the first and the second subunit of the Fc domain.
  • an amino acid residue in the CH3 domain of the first subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.
  • the Fc domain comprises at least one amino acid substitution that reduces binding to an Fc receptor and/or effector function, as compared to a native IgG 1 Fc domain.
  • each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G (Kabat numbering).
  • the Fc receptor is an Fe ⁇ receptor.
  • the effector function is antibody-dependent cell-mediated cytotoxicity (ADCC).
  • the T cell activating bispecific antigen binding molecule induces proliferation of a human CD3 positive T cell in vitro.
  • the T cell activating bispecific antigen binding molecule induces human peripheral blood mononuclear cell mediated killing of a FolR1-expressing human tumor cell in vitro.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of a FolR1-expressing human tumor cell in vitro.
  • the T cell is a CD8 positive T cell.
  • the FolR1-expressing human tumor cell is a Hela, Skov-3, HT-29, or HRCEpiC cell.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing human tumor cell in vitro with an EC50 of between about 36 pM and about 39573 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 36 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 178.4 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 134.5 pM or greater after 48 hours.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments induces upregulation of cell surface expression of at least one of CD25 and CD69 on the T cell as measured by flow cytometry.
  • the T cell is a CD4 positive T cell or a CD8 positive T cell.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments binds human FolR1 with an apparent K D of about 5.36 pM to about 4 nM.
  • the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent K D of about 4 nM.
  • the T cell activating bispecific antigen binding molecule binds murine FolR1 with an apparent K D of about 1.5 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of at least about 1000 nM.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments is specific for FolR1 and does not bind to FolR2 or FolR3. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments has an affinity (monovalent binding) of 1 ⁇ M or greater. In one embodiment, the affinity is around 1.4 ⁇ M for human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments has an avidity (bivalent binding) of about 1-100 nM or lower. In one embodiment, the avidity is about 10 nM or less. In one embodiment, the avidity is 10 nM.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to FolR1 expressed on a human tumor cell. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to a conformational epitope on human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3).
  • the antigen binding moiety binds to a FolR1 polypeptide comprising the amino acids 25 to 234 of human FolR1 (SEQ ID NO:227).
  • the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NOs: 227, 230 and 231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 and 229.
  • the invention provides for a bispecific antibody comprising a) a first antigen-binding site that competes for binding to human FolR1 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 49 and a variable light chain domain of SEQ ID NO: 51; and b) a second antigen-binding site that competes for binding to human CD3 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31, wherein binding competition is measured using a surface plasmon resonance assay.
  • the invention provides for a bispecific antibody comprising a) a first antigen-binding site that competes for binding to human FolR1 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31; and b) a second antigen-binding site that competes for binding to human CD3 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31, wherein binding competition is measured using a surface plasmon resonance assay.
  • the invention provides for a T cell activating bispecific antigen binding molecule comprising a first antigen binding moiety capable of specific binding to CD3, and a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1), wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human FolR1 as a first reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 49 and a variable light chain domain of SEQ ID NO: 51; and wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human CD3 as a second reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31.
  • VH variable heavy chain domain
  • the invention provides for a T cell activating bispecific antigen binding molecule comprising a first antigen binding moiety capable of specific binding to CD3, and a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1), wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human FolR1 as a first reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain (VL) of SEQ ID NO: 31; and wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human CD3 as a second reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain (VL) of SEQ ID NO: 31.
  • VH variable heavy chain domain
  • VL variable light chain domain
  • the invention relates to an antibody or an antigen-binding fragment thereof that competes for binding to human FolR1 with an antibody that comprises a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31, wherein binding competition is measured using a surface plasmon resonance assay.
  • VH variable heavy chain domain
  • the invention provides for a T cell activating bispecific antigen binding molecule, wherein the antigen binding molecule comprises a first, second, third, fourth and fifth polypeptide chain that form a first, a second and a third antigen binding moiety, wherein the first antigen binding moiety is capable of binding CD3 and the second and the third antigen binding moiety each are capable of binding Folate Receptor 1 (FolR1), wherein a) the first and the second polypeptide chain comprise, in amino (N)-terminal to carboxyl (C)-terminal direction, VLD1 and CLD1; b) the third polypeptide chain comprises, in N-terminal to C-terminal direction, VLD2 and CH1D2; c) the fourth polypeptide chain comprises, in N-terminal to C-terminal direction, VHD1, CH1D1, CH2D1 and CH3D1; d) the fifth polypeptide chain comprises VHD1, CH1D1, VHD2, CLD2, CH2D2 and CH3D1;
  • the third polypeptide chain and VHD2 and CLD2 of the fifth polypeptide chain form the first antigen binding moiety capable of binding CD3;
  • the first polypeptide chain and VHD1 and CH1D1 of the fourth polypeptide chain form the second binding moiety capable of binding to FolR1;
  • the second polypeptide chain and VHD1 and CH1D1 of the fifth polypeptide chain form the third binding moiety capable of binding to FolR1.
  • CH2D1, CH3D1, CH2D2 and CH3D2 form an Fc domain of an IgG class immunoglobulin.
  • the Fc domain is a human Fc domain.
  • the Fc domain comprises a modification promoting the association of the first and the second subunit of the Fc domain.
  • CH3D2 comprises an amino acid residue having a larger side chain volume, which is positionable in a cavity within CH3D1.
  • the Fc domain comprises at least one amino acid substitution that reduces binding to an Fc receptor and/or effector function, as compared to a native IgG 1 Fc domain.
  • each subunit of the Fc domain comprises three amino acid substitutions that reduce at least one of binding to an activating Fc receptor and effector function wherein said amino acid substitutions are L234A, L235A and P329G according to Kabat numbering.
  • the Fc receptor is an Fc ⁇ receptor.
  • the T cell activating bispecific antigen binding molecule induces proliferation of a human CD3 positive T cell in vitro. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces human peripheral blood mononuclear cell mediated killing of a FolR1-expressing human tumor cell in vitro. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of a FolR1-expressing human tumor cell in vitro. In one such embodiment, the FolR1-expressing human tumor cell is a Hela, Skov-3, HT-29, or HRCEpiC cell.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of between about 36 pM and about 39573 pM after 24 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 36 pM after 24 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 178.4 pM after 24 hours.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 134.5 pM or greater after 48 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces upregulation of cell surface expression of at least one of CD25 and CD69 on the T cell as measured by flow cytometry. In one such embodiments, the T cell is a CD4 positive T cell or a CD8 positive T cell. In one of the above embodiments, wherein the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent K D of about 5.36 pM to about 4 nM.
  • the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent K D of about 4 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds murine FolR1 with an apparent K D of about 1.5 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of at least about 1000 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds to FolR1 expressed on a human tumor cell.
  • the T cell activating bispecific antigen binding molecule binds to a conformational epitope on human FolR1. In one of the above embodiments, the T cell activating bispecific antigen binding molecule does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3). In one of the above embodiments, the antigen binding moiety binds to a FolR1 polypeptide comprising the amino acids 25 to 234 of human FolR1 (SEQ ID NO:227).
  • the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NOs:227, 230 and 231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 and 229.
  • the T cell activating bispecific antigen binding molecule is a humanized or a chimeric molecule.
  • VHD2 and CH1D1 are linked through a peptide linker.
  • the first and second polypeptide chain comprise the amino acid sequence of SEQ ID NO:230.
  • the third polypeptide chain comprises the amino acid sequence of SEQ ID NO:86.
  • the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO:309.
  • the fifth polypeptide chain comprises the amino acid sequence of SEQ ID NO:308.
  • the first and second polypeptide chain comprise the amino acid sequence of SEQ ID NO:230; the third polypeptide chain comprises the amino acid sequence of SEQ ID NO:86; the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO:309; and the fifth polypeptide chain comprise the amino acid sequence of SEQ ID NO:308.
  • the invention provides for a T cell activating bispecific antigen binding molecule comprising the amino acid sequence of SEQ ID NO:308.
  • the T cell activating bispecific antigen binding molecule of the above embodiment further comprises the amino acid sequence of SEQ ID NO:230 and of SEQ ID NO:86.
  • the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:308. In one aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:309.
  • the invention provides for a T cell activating bispecific antigen binding molecule comprising the amino acid sequence of SEQ ID NO:276.
  • the T cell activating bispecific antigen binding molecule of the above embodiment further comprises the amino acid sequence of SEQ ID NO:277 and of SEQ ID NO:35.
  • the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:277. In one aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:276.
  • the invention provides for an isolated polynucleotide encoding the T cell activating bispecific antigen binding molecule of any one of the embodiments disclosed herein. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:169. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:246.
  • the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:247. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:97. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:198.
  • the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:287. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:288. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:289.
  • the invention provides for an isolated polypeptide encoded by the polynucleotide of the above embodiment.
  • the invention provides for a vector, particularly an expression vector, comprising the polynucleotide encoding the T cell activating bispecific antigen binding molecule of any one of the embodiments disclosed herein.
  • the invention provides for a host cell comprising a polynucleotide or a vector of any of the embodiments disclosed herein.
  • the invention provides for a method of producing the T cell activating bispecific antigen binding molecule capable of specific binding to CD3 and a target cell antigen, comprising the steps of a) culturing the host cell of the above embodiments under conditions suitable for the expression of the T cell activating bispecific antigen binding molecule and b) recovering the T cell activating bispecific antigen binding molecule.
  • the invention provides for T cell activating bispecific antigen binding molecule produced by the method of the above embodiment.
  • the invention provides for a pharmaceutical composition comprising the T cell activating bispecific antigen binding molecule of any one of the above embodiments and a pharmaceutically acceptable carrier. In one aspect, the invention provides for the T cell activating bispecific antigen binding molecule of any one of the above embodiments or the pharmaceutical composition of any of the above embodiments for use as a medicament.
  • the invention provides for the T cell activating bispecific antigen binding molecule of any one of the above embodiments or the pharmaceutical composition of any one of the above embodiments for use in the treatment of a disease in an individual in need thereof.
  • the disease is cancer.
  • the invention provides for a use of the T cell activating bispecific antigen binding molecule of any one of the above embodiments for the manufacture of a medicament for the treatment of a disease in an individual in need thereof.
  • the invention provides for a method of treating a disease in an individual, comprising administering to said individual a therapeutically effective amount of a composition comprising the T cell activating bispecific antigen binding molecule of any one of the above embodiments in a pharmaceutically acceptable form.
  • said disease is a cancer.
  • the invention provides for a method for inducing lysis of a target cell, comprising contacting a target cell with the T cell activating bispecific antigen binding molecule of any one of the above embodiments in the presence of a T cell.
  • the invention provides for a the invention as described hereinbefore.
  • FIGS. 1A-I illustrate exemplary configurations of the T cell activating bispecific antigen binding molecules (TCBs) of the invention.
  • All constructs except the kappa-lambda format in ( FIG. 1I ) have P329G LALA mutations and comprise knob-into-hole Fc fragments with knob-into-hole modifications.
  • FIG. 1A Illustration of the “FolR1 TCB 2+1 inverted (common light chain)”.
  • the FolR1 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification.
  • FIG. 1B Illustration of the “FolR1 TCB 1+1 head-to-tail (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain.
  • FIG. 1C Illustration of the “FolR1 TCB 1+1 classical (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain.
  • FIG. 1D Illustration of the “FolR1 TCB 2+1 classical (common light chain)”. The CD3 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. These constructs are not crossed and have three times the same VLCL light chain.
  • FIG. 1C Illustration of the “FolR1 TCB 1+1 classical (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain.
  • FIG. 1D Illustration of the “FolR1 TCB 2+1 classical (common light chain
  • FIG. 1E Illustration of the “FolR1 TCB 2+1 crossfab classical”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain.
  • the CD3 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification.
  • FIG. 1F Illustration of the “FolR1 TCB 2+1 crossfab inverted”.
  • These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain.
  • the FolR1 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification.
  • FIG. 1G Illustration of the “FolR1 TCB 1+1 crossfab head-to-tail”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain.
  • FIG. 1H Illustration of the “FolR1 TCB 1+1 crossfab classical”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain.
  • FIG. 1I illustrates the CD3/FolR1 kappa-lambda antibody format. These constructs comprise a crossed common light chain VLCH1 and one crossed VHCL chain specific for CD3 and one crossed VHCL chain specific for FolR1.
  • FIGS. 2A-C depict graphs summarizing Binding of FoLR1 IgG binders to HeLa cells. Binding of newly generated FolR1 binders to FolR1 expressed on HeLa cells were determined by flow cytometry. Bound antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 3A-B depict graphs summarizing specificity of FolR1 binders for FolR1. Binding of FolR1 IgGs to HEK cells transiently transfected with either FolR1 or FolR2 was analyzed by flow cytometry to identify clones which bind specifically to FolR1 and not to FolR2. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 4A-B depict graphs summarizing cross-reactivity of FolR1 binders to cyFoLR1.
  • Cross-reactivity of the FolR1 antibodies to cyno FolR1 was addressed on HEK cells transiently transfected with cyFolR1 by flow cytometry. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIG. 5 depicts a graph illustrating internalization of FolR1 TCBs after binding. Internalization of the four FolR1 TCBs after binding to FolR1 was tested on HeLa cells. Remaining FolR1 TCBs on the surface were detected with a fluorescently labeled anti-human secondary antibody after indicated time points of incubation at 37° C. Percentage of internalization was calculated.
  • FIGS. 6A-E depict graphs summarizing binding of FolR1 IgGs to cells with different FolR1 expression levels. Binding of 9D11, 16D5 and Mov19 IgG to tumor cells with different FolR1 expression levels was analyzed by flow cytometry. DP47 IgG was included as isotype control and MKN-45 were included as FolR1 negative cell line. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 7A-L depict graphs summarizing T cell mediated killing of HT-29 and SKOV3 cells.
  • FolR1 TCBs were used to test T cell mediated killing of HT-29 and SKOV3 tumor cells and upregulation of activation marker on T cells upon killing.
  • FIGS. 7A-D T cell mediated killing of HT-29 and SKOV3 cells in the presence of 9D11 FolR1 TCB and 16D5 FolR1 TCB was measured by LDH release after 24 h and 48 h. DP47 TCB was included as negative control. After 48 h incubation upregulation of the activation marker CD25 and CD69 on CD8 T cells and CD4 T cells upon killing of SKOV3 ( FIG. 7E-H ) or HT-29 ( FIG. 7I-L ) tumor cells was assessed by flow cytometry.
  • FIG. 8 depicts a graph showing absence of anti-FolR1 binding to erythrocytes.
  • Erythrocytes were gated as CD235a positive population and binding of 9D11 IgG, 16D5 IgG, Mov19 IgG and DP47 IgG to this population was determined by flow cytometry.
  • the antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 9A-D depict graphs summarizing activation marker upregulation in whole blood.
  • FIG. 10 Binding of 9D11 TCB a-glyco variants to HeLa cells. Binding of 9D11 FolR1 TCB a-glyco variants to Hela cells was compared to binding of the original 9D11 TCB on HeLa cells. The antibodies were detected with a fluorescently labeled anti-human secondary antibody and binding was determined by flow cytometry.
  • FIGS. 11A-F depict graphs summarizing T cell mediated killing with 9D11 FolR1 TCB a-glyco variants of tumor cells.
  • 9D11 FolR1 TCB a-glyco variants were used to test T cell mediated killing of ( FIG. 11A-D ) SKOV3, MKN-45 (as FolR1 negative control) and ( FIG. 11E-F ) HT-29 tumor cells in comparison to killing with the original 9D11 FolR1 TCB.
  • FIGS. 12A-X depict graphs summarizing T cell mediated killing of primary epithelial cells.
  • Primary epithelial cells with very low levels of FolR1 were used to test T cell mediated killing with 16D5 FolR1 TCB and 9D11 FolR1 TCB, DP47 TCB was included as a negative control and HT29 cells were included as positive control.
  • FIGS. 12A-H LDH release of human retinal pigment (HRP), human renal cortical (HRC), human bronchial (HB) and of HT29 cells was determined after 24 h and 48 h.
  • CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells upon killing of FIGS. 12I-L ) HRP, ( FIGS. 12M-P ) HRC, ( FIGS. 12Q-T ) HB and ( FIG. 12 U-X) HT29 was determined after 48 h by flow cytometry.
  • FIGS. 13A-C show a comparison of different TCB formats with 16D5.
  • Four different TCB formats containing the FolR1 binder 16D5 were compared in FIG. 13A binding to HeLa cells, in FIG. 14 B T cell mediated killing of SKOV3 cells after 24 h and 48 h and in FIG. 14C CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells 48 h after killing.
  • FIGS. 14A-C depict a comparison of different TCB formats with 9D11.
  • Three different TCB formats containing the FolR1 binder 9D11 were compared in A) binding to HeLa cells, in B) T cell mediated killing of SKOV3 cells after 24 h and 48 h and in C) CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells 48 h after killing.
  • FIG. 15 depicts a PK-profile of FOLR1 TCB in NOG mice for three different doses.
  • FIG. 16 illustrates an experimental protocol for efficacy study with FOLR1 TCB.
  • FIGS. 17A-B depict tumor growth curves.
  • FIG. 17A Mean values and SEM of tumor volumes in the different treatment groups.
  • FIG. 17B Tumor growth of single mice in all treatment groups. TGI (tumor growth inhibition) give the percentage of the Mean tumor volume compared to vehicle group.
  • FIG. 18 shows tumor weights at study termination.
  • FIGS. 19A-B show FACS analysis of tumor infiltrating T-cells at study day 32.
  • FIG. 19A Tumor single cells suspensions were stained with anti-human CD3/CD4/CD8 and analyzed by flow cytometry.
  • FIG. 19B Mean values and SEM of T-cell counts per mg tumor tissue in different treatment groups.
  • FIGS. 20A-B show FACS analysis for T-cell activation/degranulation and cytokine secretion at study day 32.
  • FIGS. 21A-B show percent tumor lysis.
  • SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 24 h ( FIG. 21A ) and 48 h ( FIG. 21B ) killing of tumor cells was determined by measuring LDH release.
  • FIGS. 22A-D show CD25 and CD69 upregulation on CD4 T cells.
  • SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 48 h CD25 and CD69 upregulation on CD4 T cells ( FIG. 22A-B ) and CD8 T cells ( FIG. 22C-D ) was measured by flow cytometry.
  • DP47 TCB was included as non-binding control.
  • FIG. 27 depicts a table summarizing quantification of FolR1 binding sites on various normal and cancer cells lines.
  • FIGS. 28A-B show binding of 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA to human CD3 expressed on Jurkat cells.
  • DP47 TCB was included as non-binding control.
  • DP47 TCB was included as non-binding control.
  • FIGS. 31A-C show mean fluorescence intensity and tumor cell lysis.
  • FIGS. 32A-E shows binding of 36F2 TCB, 16D5 TCB and 16D5 HC/LC variants to human FolR1 expressed on Hela cells.
  • FIG. 33 shows binding of 36F2 TCB, 16D5 TCB and the two 16D5 affinity reduced variants 16D5 W96Y/D52E TCB and 16D5 G49S/S93A TCB to human FolR1 on Hela cells.
  • FIGS. 34A-E show binding of 36F2 TCB, 16D5 TCB and 16D5 HC/LC variants to human FolR1 expressed on HT-29 cells.
  • FIGS. 35A-D show binding of intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), 16D5 TCB and 36F2 TCB to HEK293T cells expressing either human or mouse FolR1 or FolR2.
  • FIG. 36A-F show T-cell killing of Hela (high FolR1 expression), SKov-3 (medium FolR1 expression) and HT-29 (low FolR1 expression) human tumor cells induced by intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), 16D5 TCB and 36F2 TCB after 24 h (A-C) and 48 h (D-F) of incubation.
  • HT-29 cells low FolR1 expression
  • DP47 TCB served as non-binding control.
  • FIGS. 39A-B show single dose PK of FOLR1 TCB constructs in female NOG mice.
  • FIGS. 40A-G show in vivo efficacy of FOLR1 TCB constructs (16D5, 16D5 G49S/S93A and 16D5 W96Y/D52E) after human PBMC transfer in Hela-bearing NOG mice.
  • FIG. 41 shows that Farletuzumab (dark green, second from the top) and Mov19 (grey, top) are able to bind on huFolR1 that is captured on 16D5, demonstrating that the 16D5 series binders recognize an epitope distinct from that recognized by either Farletuzumab or Mov19.
  • antigen binding molecule refers in its broadest sense to a molecule that specifically binds an antigenic determinant.
  • antigen binding molecules are immunoglobulins and derivatives, e.g. fragments, thereof.
  • bispecific means that the antigen binding molecule is able to specifically bind to at least two distinct antigenic determinants.
  • a bispecific antigen binding molecule comprises at least two antigen binding sites, each of which is specific for a different antigenic determinant.
  • the bispecific antigen binding molecule is capable of simultaneously binding two antigenic determinants, particularly two antigenic determinants expressed on two distinct cells.
  • valent denotes the presence of a specified number of antigen binding sites in an antigen binding molecule.
  • monovalent binding to an antigen denotes the presence of one (and not more than one) antigen binding site specific for the antigen in the antigen binding molecule.
  • an “antigen binding site” refers to the site, i.e. one or more amino acid residues, of an antigen binding molecule which provides interaction with the antigen.
  • the antigen binding site of an antibody comprises amino acid residues from the complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • a native immunoglobulin molecule typically has two antigen binding sites, a Fab molecule typically has a single antigen binding site.
  • an antigen binding moiety refers to a polypeptide molecule that specifically binds to an antigenic determinant.
  • an antigen binding moiety is able to direct the entity to which it is attached (e.g. a second antigen binding moiety) to a target site, for example to a specific type of tumor cell or tumor stroma bearing the antigenic determinant.
  • an antigen binding moiety is able to activate signaling through its target antigen, for example a T cell receptor complex antigen.
  • Antigen binding moieties include antibodies and fragments thereof as further defined herein. Particular antigen binding moieties include an antigen binding domain of an antibody, comprising an antibody heavy chain variable region and an antibody light chain variable region.
  • the antigen binding moieties may comprise antibody constant regions as further defined herein and known in the art.
  • Useful heavy chain constant regions include any of the five isotypes: ⁇ , ⁇ , ⁇ , ⁇ , or ⁇ .
  • Useful light chain constant regions include any of the two isotypes: ⁇ and ⁇ .
  • antigenic determinant is synonymous with “antigen” and “epitope,” and refers to a site (e.g. a contiguous stretch of amino acids or a conformational configuration made up of different regions of non-contiguous amino acids) on a polypeptide macromolecule to which an antigen binding moiety binds, forming an antigen binding moiety-antigen complex.
  • Useful antigenic determinants can be found, for example, on the surfaces of tumor cells, on the surfaces of virus-infected cells, on the surfaces of other diseased cells, on the surface of immune cells, free in blood serum, and/or in the extracellular matrix (ECM).
  • ECM extracellular matrix
  • the proteins referred to as antigens herein can be any native form the proteins from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats), unless otherwise indicated.
  • the antigen is a human protein.
  • the term encompasses the “full-length”, unprocessed protein as well as any form of the protein that results from processing in the cell.
  • the term also encompasses naturally occurring variants of the protein, e.g. splice variants or allelic variants.
  • Exemplary human proteins useful as antigens include, but are not limited to: FolR1 (Folate receptor alpha (FRA); Folate binding protein (FBP); human FolR1 UniProt no.: P15328; murine FolR1 UniProt no.: P35846; cynomolgus FolR1 UniProt no.: G7PR14) and CD3, particularly the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP 000724.1, SEQ ID NO:150 for the human sequence; or UniProt no. Q95LI5 (version 49), NCBI GenBank no.
  • the T cell activating bispecific antigen binding molecule of the invention binds to an epitope of CD3 or a target cell antigen that is conserved among the CD3 or target antigen from different species. In certain embodiments the T cell activating bispecific antigen binding molecule of the invention binds to CD3 and FolR1, but does not bind to FolR2 (Folate receptor beta; FRB; human FolR2 UniProt no.: P14207) or FolR3 (Folate receptor gamma; human FolR3 UniProt no.: P41439).
  • FolR2 FolR2
  • FolR3 FolR3 (Folate receptor gamma; human FolR3 UniProt no.: P41439).
  • ELISA enzyme-linked immunosorbent assay
  • SPR surface plasmon resonance
  • an antigen binding moiety that binds to the antigen, or an antigen binding molecule comprising that antigen binding moiety has a dissociation constant (K D ) of ⁇ 1 ⁇ M, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 ⁇ 8 M or less, e.g. from 10 ⁇ 8 M to 10 ⁇ 13 M, e.g., from 10 ⁇ 9 M to 10 ⁇ 13 M).
  • K D dissociation constant
  • Binding affinity refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., an antigen binding moiety and an antigen, or a receptor and its ligand).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (K D ), which is the ratio of dissociation and association rate constants (k off and k on , respectively).
  • affinities may comprise different rate constants, as long as the ratio of the rate constants remains the same.
  • Affinity can be measured by well-established methods known in the art, including those described herein.
  • a particular method for measuring affinity is Surface Plasmon Resonance (SPR).
  • Reduced binding for example reduced binding to an Fc receptor, refers to a decrease in affinity for the respective interaction, as measured for example by SPR.
  • the term includes also reduction of the affinity to zero (or below the detection limit of the analytic method), i.e. complete abolishment of the interaction.
  • increased binding refers to an increase in binding affinity for the respective interaction.
  • T cell activation refers to one or more cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers.
  • the T cell activating bispecific antigen binding molecules of the invention are capable of inducing T cell activation. Suitable assays to measure T cell activation are known in the art described herein.
  • target cell antigen refers to an antigenic determinant presented on the surface of a target cell, for example a cell in a tumor such as a cancer cell or a cell of the tumor stroma.
  • target cell antigen refers to Folate Receptor 1.
  • first and second with respect to antigen binding moieties etc., are used for convenience of distinguishing when there is more than one of each type of moiety. Use of these terms is not intended to confer a specific order or orientation of the T cell activating bispecific antigen binding molecule unless explicitly so stated.
  • a “Fab molecule” refers to a protein consisting of the VH and CH1 domain of the heavy chain (the “Fab heavy chain”) and the VL and CL domain of the light chain (the “Fab light chain”) of an immunoglobulin.
  • Fab molecules having identical VLCL light chains refers to binders that share one light chain but still have separate specificities, e.g., can bind CD3 or FolR1.
  • the T-cell activating bispecific molecules comprise at least two Fab molecules having identical VLCL light chains. The corresponding heavy chains are remodeled and confer specific binding to the T cell activating bispecific antigen CD3 and the target cell antigen FolR1, respectively.
  • fused is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.
  • single-chain refers to a molecule comprising amino acid monomers linearly linked by peptide bonds.
  • one of the antigen binding moieties is a single-chain Fab molecule, i.e. a Fab molecule wherein the Fab light chain and the Fab heavy chain are connected by a peptide linker to form a single peptide chain.
  • the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain in the single-chain Fab molecule.
  • crossover Fab molecule also termed “Crossfab” is meant a Fab molecule wherein either the variable regions or the constant regions of the Fab heavy and light chain are exchanged, i.e. the crossover Fab molecule comprises a peptide chain composed of the light chain variable region and the heavy chain constant region, and a peptide chain composed of the heavy chain variable region and the light chain constant region.
  • the peptide chain comprising the heavy chain constant region is referred to herein as the “heavy chain” of the crossover Fab molecule.
  • CrossFab an antibody that comprises one or more CrossFabs.
  • a “conventional” Fab molecule is meant a Fab molecule in its natural format, i.e. comprising a heavy chain composed of the heavy chain variable and constant regions (VH-CH1), and a light chain composed of the light chain variable and constant regions (VL-CL).
  • immunoglobulin molecule refers to a protein having the structure of a naturally occurring antibody.
  • immunoglobulins of the IgG class are heterotetrameric glycoproteins of about 150,000 daltons, composed of two light chains and two heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3), also called a heavy chain constant region.
  • VH variable region
  • CH1, CH2, and CH3 constant domains
  • each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain, also called a light chain constant region.
  • VL variable region
  • the heavy chain of an immunoglobulin may be assigned to one of five types, called ⁇ (IgA), ⁇ (IgD), ⁇ (IgE), ⁇ (IgG), or ⁇ (IgM), some of which may be further divided into subtypes, e.g. ⁇ 1 (IgG 1 ), ⁇ 2 (IgG 2 ), ⁇ 3 (IgG 3 ), ⁇ 4 (IgG 4 ), ⁇ 1 (IgA 1 ) and ⁇ 2 (IgA 2 ).
  • the light chain of an immunoglobulin may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • An immunoglobulin essentially consists of two Fab molecules and an Fc domain, linked via the immunoglobulin hinge region.
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab′, Fab′-SH, F(ab′) 2 , diabodies, linear antibodies, single-chain antibody molecules (e.g. scFv), and single-domain antibodies.
  • scFv single-chain antibody molecules
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific.
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, Mass.; see e.g. U.S. Pat. No. 6,248,516 B1).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • an antigen binding domain refers to the part of an antibody that comprises the area which specifically binds to and is complementary to part or all of an antigen.
  • An antigen binding domain may be provided by, for example, one or more antibody variable domains (also called antibody variable regions).
  • an antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). See, e.g., Kindt et al., Kuby Immunology, 6 th ed., W.H. Freeman and Co., page 91 (2007).
  • a single VH or VL domain may be sufficient to confer antigen-binding specificity.
  • hypervariable region refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops (“hypervariable loops”).
  • native four-chain antibodies comprise six HVRs; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3).
  • HVRs generally comprise amino acid residues from the hypervariable loops and/or from the complementarity determining regions (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops.
  • Hypervariable regions are also referred to as “complementarity determining regions” (CDRs), and these terms are used herein interchangeably in reference to portions of the variable region that form the antigen binding regions.
  • CDRs complementarity determining regions
  • This particular region has been described by Kabat et al., U.S. Dept. of Health and Human Services, Sequences of Proteins of Immunological Interest (1983) and by Chothia et al., J Mol Biol 196:901-917 (1987), where the definitions include overlapping or subsets of amino acid residues when compared against each other. Nevertheless, application of either definition to refer to a CDR of an antibody or variants thereof is intended to be within the scope of the term as defined and used herein.
  • Kabat et al. also defined a numbering system for variable region sequences that is applicable to any antibody.
  • One of ordinary skill in the art can unambiguously assign this system of “Kabat numbering” to any variable region sequence, without reliance on any experimental data beyond the sequence itself.
  • “Kabat numbering” refers to the numbering system set forth by Kabat et al., U.S. Dept. of Health and Human Services, “Sequence of Proteins of Immunological Interest” (1983). Unless otherwise specified, references to the numbering of specific amino acid residue positions in an antibody variable region are according to the Kabat numbering system.
  • polypeptide sequences of the sequence listing are not numbered according to the Kabat numbering system. However, it is well within the ordinary skill of one in the art to convert the numbering of the sequences of the Sequence Listing to Kabat numbering.
  • “Framework” or “FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.
  • the “class” of an antibody or immunoglobulin refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • Fc domain or “Fc region” herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to extend from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • a “subunit” of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association.
  • a subunit of an IgG Fc domain comprises an IgG CH2 and an IgG CH3 constant domain.
  • a “modification promoting the association of the first and the second subunit of the Fc domain” is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer.
  • a modification promoting association as used herein particularly includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the modifications are complementary to each other so as to promote association of the two Fc domain subunits.
  • a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their association sterically or electrostatically favorable, respectively.
  • (hetero)dimerization occurs between a polypeptide comprising the first Fc domain subunit and a polypeptide comprising the second Fc domain subunit, which might be non-identical in the sense that further components fused to each of the subunits (e.g. antigen binding moieties) are not the same.
  • the modification promoting association comprises an amino acid mutation in the Fc domain, specifically an amino acid substitution.
  • the modification promoting association comprises a separate amino acid mutation, specifically an amino acid substitution, in each of the two subunits of the Fc domain.
  • effector functions refers to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype.
  • antibody effector functions include: C1q binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor), and B cell activation.
  • engine engineered, engineering
  • engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches.
  • amino acid mutation as used herein is meant to encompass amino acid substitutions, deletions, insertions, and modifications. Any combination of substitution, deletion, insertion, and modification can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., reduced binding to an Fc receptor, or increased association with another peptide.
  • Amino acid sequence deletions and insertions include amino- and/or carboxy-terminal deletions and insertions of amino acids.
  • Particular amino acid mutations are amino acid substitutions.
  • non-conservative amino acid substitutions i.e. replacing one amino acid with another amino acid having different structural and/or chemical properties, are particularly preferred.
  • Amino acid substitutions include replacement by non-naturally occurring amino acids or by naturally occurring amino acid derivatives of the twenty standard amino acids (e.g. 4-hydroxyproline, 3-methylhistidine, ornithine, homoserine, 5-hydroxylysine).
  • Amino acid mutations can be generated using genetic or chemical methods well known in the art. Genetic methods may include site-directed mutagenesis, PCR, gene synthesis and the like. It is contemplated that methods of altering the side chain group of an amino acid by methods other than genetic engineering, such as chemical modification, may also be useful. Various designations may be used herein to indicate the same amino acid mutation. For example, a substitution from proline at position 329 of the Fc domain to glycine can be indicated as 329G, G329, G 329 , P329G, or Pro329Gly.
  • polypeptide refers to a molecule composed of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds).
  • polypeptide refers to any chain of two or more amino acids, and does not refer to a specific length of the product.
  • peptides, dipeptides, tripeptides, oligopeptides, “protein,” “amino acid chain,” or any other term used to refer to a chain of two or more amino acids are included within the definition of “polypeptide,” and the term “polypeptide” may be used instead of, or interchangeably with any of these terms.
  • polypeptide is also intended to refer to the products of post-expression modifications of the polypeptide, including without limitation glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
  • a polypeptide may be derived from a natural biological source or produced by recombinant technology, but is not necessarily translated from a designated nucleic acid sequence. It may be generated in any manner, including by chemical synthesis.
  • a polypeptide of the invention may be of a size of about 3 or more, 5 or more, 10 or more, 20 or more, 25 or more, 50 or more, 75 or more, 100 or more, 200 or more, 500 or more, 1,000 or more, or 2,000 or more amino acids.
  • Polypeptides may have a defined three-dimensional structure, although they do not necessarily have such structure. Polypeptides with a defined three-dimensional structure are referred to as folded, and polypeptides which do not possess a defined three-dimensional structure, but rather can adopt a large number of different conformations, and are referred to as unfolded.
  • an “isolated” polypeptide or a variant, or derivative thereof is intended a polypeptide that is not in its natural milieu. No particular level of purification is required.
  • an isolated polypeptide can be removed from its native or natural environment.
  • Recombinantly produced polypeptides and proteins expressed in host cells are considered isolated for the purpose of the invention, as are native or recombinant polypeptides which have been separated, fractionated, or partially or substantially purified by any suitable technique.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, Calif., or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • polynucleotide refers to an isolated nucleic acid molecule or construct, e.g. messenger RNA (mRNA), virally-derived RNA, or plasmid DNA (pDNA).
  • mRNA messenger RNA
  • pDNA virally-derived RNA
  • a polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond (e.g. an amide bond, such as found in peptide nucleic acids (PNA).
  • PNA peptide nucleic acids
  • nucleic acid molecule refers to any one or more nucleic acid segments, e.g. DNA or RNA fragments, present in a polynucleotide.
  • isolated nucleic acid molecule or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment.
  • a recombinant polynucleotide encoding a polypeptide contained in a vector is considered isolated for the purposes of the present invention.
  • Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution.
  • An isolated polynucleotide includes a polynucleotide molecule contained in cells that ordinarily contain the polynucleotide molecule, but the polynucleotide molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts of the present invention, as well as positive and negative strand forms, and double-stranded forms. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically.
  • a polynucleotide or a nucleic acid may be or may include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator.
  • a nucleic acid or polynucleotide having a nucleotide sequence at least, for example, 95% “identical” to a reference nucleotide sequence of the present invention it is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence.
  • a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • These alterations of the reference sequence may occur at the 5′ or 3′ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • any particular polynucleotide sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be determined conventionally using known computer programs, such as the ones discussed above for polypeptides (e.g. ALIGN-2).
  • expression cassette refers to a polynucleotide generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a target cell.
  • the recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus, or nucleic acid fragment.
  • the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter.
  • the expression cassette of the invention comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
  • vector or “expression vector” is synonymous with “expression construct” and refers to a DNA molecule that is used to introduce and direct the expression of a specific gene to which it is operably associated in a target cell.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • the expression vector of the present invention comprises an expression cassette. Expression vectors allow transcription of large amounts of stable mRNA. Once the expression vector is inside the target cell, the ribonucleic acid molecule or protein that is encoded by the gene is produced by the cellular transcription and/or translation machinery.
  • the expression vector of the invention comprises an expression cassette that comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a host cell is any type of cellular system that can be used to generate the bispecific antigen binding molecules of the present invention.
  • Host cells include cultured cells, e.g.
  • mammalian cultured cells such as CHO cells, BHK cells, NS0 cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
  • an “activating Fc receptor” is an Fc receptor that following engagement by an Fc domain of an antibody elicits signaling events that stimulate the receptor-bearing cell to perform effector functions.
  • Human activating Fc receptors include Fc ⁇ RIIIa (CD16a), Fc ⁇ RI (CD64), Fc ⁇ RIIa (CD32), and Fc ⁇ RI (CD89).
  • Antibody-dependent cell-mediated cytotoxicity is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells.
  • the target cells are cells to which antibodies or derivatives thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region.
  • reduced ADCC is defined as either a reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or an increase in the concentration of antibody in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC.
  • the reduction in ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered.
  • the reduction in ADCC mediated by an antibody comprising in its Fc domain an amino acid substitution that reduces ADCC is relative to the ADCC mediated by the same antibody without this amino acid substitution in the Fc domain.
  • Suitable assays to measure ADCC are well known in the art (see e.g. PCT publication no. WO 2006/082515 or PCT publication no. WO 2012/130831).
  • an “effective amount” of an agent refers to the amount that is necessary to result in a physiological change in the cell or tissue to which it is administered.
  • a “therapeutically effective amount” of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • a therapeutically effective amount of an agent for example eliminates, decreases, delays, minimizes or prevents adverse effects of a disease.
  • mammals include, but are not limited to, domesticated animals (e.g. cows, sheep, cats, dogs, and horses), primates (e.g. humans and non-human primates such as monkeys), rabbits, and rodents (e.g. mice and rats). Particularly, the individual or subject is a human.
  • domesticated animals e.g. cows, sheep, cats, dogs, and horses
  • primates e.g. humans and non-human primates such as monkeys
  • rabbits e.g. mice and rats
  • rodents e.g. mice and rats
  • composition refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical composition, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • treatment refers to clinical intervention in an attempt to alter the natural course of a disease in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • T cell activating bispecific antigen binding molecules of the invention are used to delay development of a disease or to slow the progression of a disease.
  • antagonist is used in the broadest sense, and includes any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity of a native polypeptide disclosed herein.
  • agonist is used in the broadest sense and includes any molecule that induces a biological activity of a native polypeptide disclosed herein.
  • Suitable agonist or antagonist molecules specifically include agonist or antagonist antibodies or antibody fragments, including engineered antibody fragments, fragments or amino acid sequence variants of native polypeptides, peptides, antisense oligonucleotides, small organic molecules, etc.
  • Methods for identifying agonists or antagonists of a polypeptide may comprise contacting a polypeptide with a candidate agonist or antagonist molecule and measuring a detectable change in one or more biological activities normally associated with the polypeptide.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • the T cell activating bispecific antigen binding molecule of the invention is bispecific, i.e. it comprises at least two antigen binding moieties capable of specific binding to two distinct antigenic determinants, i.e. to CD3 and to FolR1.
  • the antigen binding moieties are Fab molecules (i.e. antigen binding domains composed of a heavy and a light chain, each comprising a variable and a constant region).
  • said Fab molecules are human.
  • said Fab molecules are humanized.
  • said Fab molecules comprise human heavy and light chain constant regions.
  • the T cell activating bispecific antigen binding molecule of the invention is capable of simultaneous binding to the target cell antigen FolR1 and CD3.
  • the T cell activating bispecific antigen binding molecule is capable of crosslinking a T cell and a FolR1 expressing target cell by simultaneous binding to the target cell antigen FolR1 and CD3.
  • simultaneous binding results in lysis of the FolR1 expressing target cell, particularly a FolR1 expressing tumor cell.
  • such simultaneous binding results in activation of the T cell.
  • such simultaneous binding results in a cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers.
  • a T lymphocyte particularly a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers.
  • binding of the T cell activating bispecific antigen binding molecule to CD3 without simultaneous binding to the target cell antigen FolR1 does not result in T cell activation.
  • the T cell activating bispecific antigen binding molecule is capable of re-directing cytotoxic activity of a T cell to a FolR1 expressing target cell.
  • said re-direction is independent of MHC-mediated peptide antigen presentation by the target cell and and/or specificity of the T cell.
  • a T cell is a cytotoxic T cell.
  • the T cell is a CD4 + or a CD8 + T cell, particularly a CD8 + T cell.
  • the T cell activating bispecific antigen binding molecule of the invention comprises at least one antigen binding moiety capable of binding to CD3 (also referred to herein as an “CD3 antigen binding moiety” or “first antigen binding moiety”).
  • the T cell activating bispecific antigen binding molecule comprises not more than one antigen binding moiety capable of specific binding to CD3.
  • the T cell activating bispecific antigen binding molecule provides monovalent binding to CD3.
  • CD3 is human CD3 or cynomolgus CD3, most particularly human CD3.
  • the CD3 antigen binding moiety is cross-reactive for (i.e. specifically binds to) human and cynomolgus CD3.
  • the first antigen binding moiety is capable of specific binding to the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP_000724.1, SEQ ID NO:150 for the human sequence; UniProt no. Q95LI5 (version 49), NCBI GenBank no. BAB71849.1, for the cynomolgus [ Macaca fascicularis ] sequence).
  • the CD3 antigen binding moiety comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • CDR heavy chain complementarity determining region
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • the CD3 antigen binding moiety comprises a variable heavy chain comprising an amino acid sequence of: SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of: SEQ ID NO: 31.
  • the CD3 antigen binding moiety comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31.
  • the T cell activating bispecific antigen binding molecule of the invention comprises at least one antigen binding moiety capable of binding to the target cell antigen FolR1 (also referred to herein as an “FolR1 binding moiety” or “second” or “third” antigen binding moiety).
  • the antigen binding moiety capable of binding to the target cell antigen FolR1 does not bind to FolR2 or FolR3.
  • the FolR1 antigen binding moiety is cross-reactive for (i.e. specifically binds to) human and cynomolgus FolR1.
  • the T cell activating bispecific antigen binding molecule comprises two antigen binding moieties capable of binding to the target cell antigen FolR1.
  • each of these antigen binding moieties specifically binds to the same antigenic determinant. In an even more particular embodiment, all of these antigen binding moieties are identical. In one embodiment the T cell activating bispecific antigen binding molecule comprises not more than two antigen binding moieties capable of binding to FolR1.
  • the FolR1 binding moiety is generally a Fab molecule that specifically binds to FolR1 and is able to direct the T cell activating bispecific antigen binding molecule to which it is connected to a target site, for example to a specific type of tumor cell that expresses FolR1.
  • the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • the first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences.
  • the third antigen binding moiety is identical to the second antigen binding moiety.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments additionally comprises an Fc domain composed of a first and a second subunit capable of stable association.
  • first antigen binding moiety and the second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.
  • the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, optionally via a peptide linker.
  • not more than one antigen binding moiety capable of specific binding to CD3 is present in the T cell activating bispecific antigen binding molecule (i.e. the T cell activating bispecific antigen binding molecule provides monovalent binding to CD3).
  • the inventors of the present invention generated a bispecific antibody wherein the binding moieties share a common light chain that retains the specificity and efficacy of the parent monospecific antibody for CD3 and can bind a second antigen (e.g., FolR1) using the same light chain.
  • a bispecific molecule with a common light chain that retains the binding properties of the parent antibody is not straight-forward as the common CDRs of the hybrid light chain have to effectuate the binding specificity for both targets.
  • the present invention provides a T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, one of which is a Fab molecule capable of specific binding to CD3 and the other one of which is a Fab molecule capable of specific binding to FolR1, wherein the first and the second Fab molecule have identical VLCL light chains.
  • said identical light chain (VLCL) comprises the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.
  • said identical light chain (VLCL) comprises SEQ ID NO. 35.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 16, the heavy chain CDR2 of SEQ ID NO: 17, the heavy chain CDR3 of SEQ ID NO:18, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 275 and SEQ ID NO: 315 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises the heavy chain complementarity determining region (CDR) amino acid sequences of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39, and the light chain CDR amino acid sequences of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34;
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises the heavy chain complementarity determining region (CDR) amino acid sequences of SEQ ID NO: 16, SEQ ID NO: 275 and SEQ ID NO: 315, and the light chain CDR amino acid sequences of SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31;
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 274 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • the antigen binding moiety that is specific for FolR1 comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:15 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31 or variants thereof that retain functionality.
  • the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:15, and a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31.
  • a third antigen binding moiety (which is a Fab molecule) capable of specific binding to FolR1.
  • the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences.
  • the third antigen binding moiety is identical to the second antigen binding moiety.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • CDR heavy chain complementarity determining region
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 16, the heavy chain CDR2 of SEQ ID NO: 17, the heavy chain CDR3 of SEQ ID NO:18, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • the invention relates to bispecific molecules wherein at least two binding moieties have identical light chains and corresponding remodeled heavy chains that confer the specific binding to the T cell activating antigen CD3 and the target cell antigen FolR1, respectively.
  • the use of this so-called ‘common light chain’ principle i.e. combining two binders that share one light chain but still have separate specificities, prevents light chain mispairing.
  • there are less side products during production facilitating the homogenous preparation of T cell activating bispecific antigen binding molecules.
  • T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations are depicted in FIGS. 1A-I and are further described below.
  • said T cell activating bispecific antigen binding molecule further comprises an Fc domain composed of a first and a second subunit capable of stable association.
  • Fc domain composed of a first and a second subunit capable of stable association.
  • the inventors of the present invention generated a second bispecific antibody format wherein one of the binding moieties is a crossover Fab fragment.
  • a monovalent bispecific antibody is provided, wherein one of the Fab fragments of an IgG molecule is replaced by a crossover Fab fragment.
  • Crossover Fab fragments are Fab fragments wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
  • Bispecific antibody formats comprising crossover Fab fragments have been described, for example, in WO2009080252, WO2009080253, WO2009080251, WO2009080254, WO2010/136172, WO2010/145792 and WO2013/026831.
  • the first antigen binding moiety is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged.
  • Such modification prevent mispairing of heavy and light chains from different Fab molecules, thereby improving the yield and purity of the T cell activating bispecific antigen binding molecule of the invention in recombinant production.
  • the variable regions of the Fab light chain and the Fab heavy chain are exchanged.
  • the constant regions of the Fab light chain and the Fab heavy chain are exchanged.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 56, the heavy chain CDR3 of SEQ ID NO:57, the light chain CDR1 of SEQ ID NO: 59, the light chain CDR2 of SEQ ID NO: 60, and the light chain CDR3 of SEQ ID NO:65.
  • the second antigen binding moiety is a conventional Fab molecule.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • the second antigen binding moiety is a conventional Fab molecule.
  • the antigen binding moiety that is specific for FolR1 comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:55 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 64 or variants thereof that retain functionality.
  • the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:55, and a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 64.
  • the third antigen binding moiety is a conventional Fab molecule. In one embodiment, the third antigen binding moiety is a crossover Fab molecule.
  • the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences.
  • the third antigen binding moiety is identical to the second antigen binding moiety.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
  • a third antigen binding moiety capable of specific binding to Folate Receptor 1 comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
  • CDR Folate Receptor 1
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 56, the heavy chain CDR3 of SEQ ID NO:57, the light chain CDR1 of SEQ ID NO: 59, the light chain CDR2 of SEQ ID NO: 60, and the light chain CDR3 of SEQ ID NO:65.
  • the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 50 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • the second antigen binding moiety is a conventional Fab molecule. In one embodiment, the second antigen binding moiety is a crossover Fab molecule.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • the second antigen binding moiety is a conventional Fab molecule. In one embodiment, the second antigen binding moiety is a crossover Fab molecule.
  • the antigen binding moiety that is specific for FolR1 comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:49 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 51 or variants thereof that retain functionality.
  • the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:49, and a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 51.
  • the third antigen binding moiety is a conventional Fab molecule. In one embodiment, the second antigen binding moiety is a crossover Fab molecule.
  • the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences.
  • the third antigen binding moiety is identical to the second antigen binding moiety.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
  • a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 49 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
  • a third antigen binding moiety capable of specific binding to Folate Receptor 1 comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 50 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
  • CDR heavy chain complementarity determining region
  • the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate
  • Receptor 1 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • the invention relates to bispecific molecules wherein two binding moieties confer specific binding to FolR1 and one binding moiety confers specificity to the T cell activating antigen CD3.
  • One of the heavy chains is modified to ensure proper pairing of the heavy and light chains, thus eliminating the need for a common light chain approach.
  • the presence of two FolR1 binding sites enables appropriate engagement with the target antigen FolR1 and the activation of T cells.
  • T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations are depicted in FIGS. 1A-I and are further described below.
  • said T cell activating bispecific antigen binding molecule further comprises an Fc domain composed of a first and a second subunit capable of stable association.
  • Fc domain composed of a first and a second subunit capable of stable association.
  • the T cell activating bispecific antigen binding molecules comprise at least two Fab fragments having identical light chains (VLCL) and having different heavy chains (VHCL) which confer the specificities to two different antigens, i.e. one Fab fragment is capable of specific binding to a T cell activating antigen CD3 and the other Fab fragment is capable of specific binding to the target cell antigen FolR1.
  • VLCL identical light chains
  • VHCL variable heavy chains
  • the T cell activating bispecific antigen binding molecule comprises at least two antigen binding moieties (Fab molecules), one of which is a crossover Fab molecule and one of which is a conventional Fab molecule.
  • Fab molecules antigen binding moieties
  • the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule
  • the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations are depicted in FIGS. 1A-I .
  • the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • the first and second antigen binding moiety both are Fab fragments and have identical light chains (VLCL).
  • the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety.
  • the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • the first and second antigen binding moiety both are Fab fragments and have identical light chains (VLCL).
  • the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • the Fab light chain of the first antigen binding moiety and the Fab light chain of the second antigen binding moiety may additionally be fused to each other.
  • the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.
  • the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety.
  • the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • the first and second antigen binding moiety both are Fab fragments and have identical light chains (VLCL).
  • the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • the Fab light chain of the first antigen binding moiety and the Fab light chain of the second antigen binding moiety may additionally be fused to each other.
  • the antigen binding moieties may be fused to the Fc domain or to each other directly or through a peptide linker, comprising one or more amino acids, typically about 2-20 amino acids.
  • Peptide linkers are known in the art and are described herein. Suitable, non-immunogenic peptide linkers include, for example, (G 4 S) n (SEQ ID NO: 300), (SG 4 ) n (SEQ ID NO: 301), (G 4 S) n (SEQ ID NO: 300) or G 4 (SG 4 ) n (SEQ ID NO: 302) peptide linkers.
  • “n” is generally a number between 1 and 10, typically between 2 and 4.
  • a particularly suitable peptide linker for fusing the Fab light chains of the first and the second antigen binding moiety to each other is (G 4 S) 2 (SEQ ID NO: 303).
  • An exemplary peptide linker suitable for connecting the Fab heavy chains of the first and the second antigen binding moiety is EPKSC(D)-(G 4 S) 2 (SEQ ID NOS 304 and 305).
  • linkers may comprise (a portion of) an immunoglobulin hinge region. Particularly where an antigen binding moiety is fused to the N-terminus of an Fc domain subunit, it may be fused via an immunoglobulin hinge region or a portion thereof, with or without an additional peptide linker.
  • T cell activating bispecific antigen binding molecule comprising two binding moieties specific for the target cell antigen FolR have superior characteristics compared to T cell activating bispecific antigen binding molecule comprising only one binding moiety specific for the target cell antigen FolR.
  • the T cell activating bispecific antigen binding molecule of the invention further comprises a third antigen binding moiety which is a Fab molecule capable of specific binding to FolR.
  • the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences.
  • the third antigen binding moiety is identical to the second antigen binding moiety (i.e. they comprise the same amino acid sequences).
  • the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus to the N-terminus of the Fab heavy chain of the first antigen binding moiety.
  • the T cell activating bispecific antigen binding molecule essentially consists of a first, a second and a third antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety.
  • the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL).
  • the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second and third antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • the Fab light chain of the first antigen binding moiety and the Fab light chain of the third antigen binding moiety may additionally be fused to each other.
  • the T cell activating bispecific antigen binding molecule of the invention comprises five polypeptide chains that form a first, a second and a third antigen binding moiety wherein the first antigen binding moiety is capable of binding CD3 and the second and the third antigen binding moiety each are capable of binding Folate Receptor 1 (FolR1).
  • the first and the second polypeptide chain comprise, in amino (N)-terminal to carboxyl (C)-terminal direction, a first light chain variable domain (VLD1) and a first light chain constant domain (CLD1).
  • the third polypeptide chain comprises, in N-terminal to C-terminal direction, second light chain variable domain (VLD2) and a second heavy chain constant domain 1 (CH1D2).
  • the fourth polypeptide chain comprises, in N-terminal to C-terminal direction, a first heavy chain variable domain (VHD1), a first heavy chain constant domain 1 (CH1D1), a first heavy chain constant domain 2 (CH2D1) and a first heavy chain constant domain 3 (CH3D1).
  • the fifth polypeptide chain comprises VHD1, CH1D1, a second heavy chain variable domain (VHD2), a second light chain constant domain (CLD2), a second heavy chain constant domain 2 (CH2D2) and a second heavy chain constant domain 3 (CH3D2).
  • the third polypeptide chain and VHD2 and CLD2 of the fifth polypeptide chain form the first antigen binding moiety capable of binding CD3.
  • the second polypeptide chain and VHD1 and CH1D1 of the fifth polypeptide chain form the third binding moiety capable of binding to FolR1.
  • the first polypeptide chain and VHD1 and CH1D1 of the fourth polypeptide chain form the second binding moiety capable of binding to FolR1.
  • the second and the third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety.
  • the T cell activating bispecific antigen binding molecule essentially consists of a first, a second and a third antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second and third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the third antigen binding moiety.
  • the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL).
  • the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second and third antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • the Fab light chain of the first antigen binding moiety and the Fab light chain of the second antigen binding moiety may additionally be fused to each other.
  • the antigen binding moieties may be fused to the Fc domain directly or through a peptide linker.
  • the antigen binding moieties are each fused to the Fc domain through an immunoglobulin hinge region.
  • the immunoglobulin hinge region is a human IgG 1 hinge region.
  • the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule.
  • the immunoglobulin molecule is an IgG class immunoglobulin.
  • the immunoglobulin is an IgG 1 subclass immunoglobulin.
  • the immunoglobulin is an IgG 4 subclass immunoglobulin.
  • the immunoglobulin is a human immunoglobulin.
  • the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
  • said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, wherein the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL), wherein the first antigen binding moiety capable of specific binding to CD3 comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO:
  • said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, wherein the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL), wherein the first antigen binding moiety capable of specific binding to CD3 comprises a variable heavy chain comprising a sequence of SEQ ID NO: 36, a variable light chain comprising a sequence of SEQ ID NO: 31; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise a variable heavy chain comprising a sequence of SEQ ID NO: 15, a variable light chain comprising a sequence of SEQ ID NO: 31.
  • said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety and the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SDR) selected from
  • said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety and the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged, wherein the first antigen binding moiety capable of specific binding to CD3 comprises a variable heavy chain comprising a sequence of SEQ ID NO: 36, a variable light chain comprising a sequence of SEQ ID NO: 31; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise a variable heavy chain comprising a sequence of SEQ ID NO: 55, a variable light chain comprising a sequence of SEQ ID NO: 65.
  • the T cell activating bispecific antigen binding molecule is monovalent for each antigen.
  • the T cell activating bispecific antigen binding molecule can bind to human CD3 and human folate receptor alpha (FolR1) and was made without employing a hetero-dimerization approach, such as, e.g., knob-into-hole technology.
  • the molecule can be produced by employing a common light chain library and CrossMab technology.
  • the variable region of the CD3 binder is fused to the CH1 domain of a standard human IgG 1 antibody to form the VLVH crossed molecule (fused to Fc) which is common for both specificities.
  • VHCL To generate the crossed counterparts (VHCL), a CD3 specific variable heavy chain domain is fused to a constant human ⁇ light chain whereas a variable heavy chain domain specific for human FolR1 (e.g., isolated from a common light chain library) is fused to a constant human ⁇ light chain.
  • the resulting desired molecule with correctly paired chains comprises both kappa and lambda light chains or fragments thereof. Consequently, this desired bispecific molecule species can be purified from mispaired or homodimeric species with sequential purification steps selecting for kappa and lambda light chain, in either sequence.
  • purification of the desired bispecific antibody employs subsequent purification steps with KappaSelect and LambdaFabSelect columns (GE Healthcare) to remove undesired homodimeric antibodies.
  • the Fc domain of the T cell activating bispecific antigen binding molecule consists of a pair of polypeptide chains comprising heavy chain domains of an immunoglobulin molecule.
  • the Fc domain of an immunoglobulin G (IgG) molecule is a dimer, each subunit of which comprises the CH2 and CH3 IgG heavy chain constant domains.
  • the two subunits of the Fc domain are capable of stable association with each other.
  • the T cell activating bispecific antigen binding molecule of the invention comprises not more than one Fc domain.
  • the Fc domain of the T cell activating bispecific antigen binding molecule is an IgG Fc domain.
  • the Fc domain is an IgG 1 Fc domain.
  • the Fc domain is an IgG 4 Fc domain.
  • the Fc domain is an IgG 4 Fc domain comprising an amino acid substitution at position S228 (Kabat numbering), particularly the amino acid substitution S228P. This amino acid substitution reduces in vivo Fab arm exchange of IgG 4 antibodies (see Stubenrauch et al., Drug Metabolism and Disposition 38, 84-91 (2010)).
  • the Fc domain is human. An exemplary sequence of a human IgG 1 Fc region is given in SEQ ID NO:245.
  • T cell activating bispecific antigen binding molecules comprise different antigen binding moieties, fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised in two non-identical polypeptide chains. Recombinant co-expression of these polypeptides and subsequent dimerization leads to several possible combinations of the two polypeptides. To improve the yield and purity of T cell activating bispecific antigen binding molecules in recombinant production, it will thus be advantageous to introduce in the Fc domain of the T cell activating bispecific antigen binding molecule a modification promoting the association of the desired polypeptides.
  • the Fc domain of the T cell activating bispecific antigen binding molecule comprises a modification promoting the association of the first and the second subunit of the Fc domain.
  • the site of most extensive protein-protein interaction between the two subunits of a human IgG Fc domain is in the CH3 domain of the Fc domain.
  • said modification is in the CH3 domain of the Fc domain.
  • said modification is a so-called “knob-into-hole” modification, comprising a “knob” modification in one of the two subunits of the Fc domain and a “hole” modification in the other one of the two subunits of the Fc domain.
  • the knob-into-hole technology is described e.g. in U.S. Pat. No. 5,731,168; U.S. Pat. No. 7,695,936; Ridgway et al., Prot Eng 9, 617-621 (1996) and Carter, J Immunol Meth 248, 7-15 (2001).
  • the method involves introducing a protuberance (“knob”) at the interface of a first polypeptide and a corresponding cavity (“hole”) in the interface of a second polypeptide, such that the protuberance can be positioned in the cavity so as to promote heterodimer formation and hinder homodimer formation.
  • Protuberances are constructed by replacing small amino acid side chains from the interface of the first polypeptide with larger side chains (e.g.
  • Compensatory cavities of identical or similar size to the protuberances are created in the interface of the second polypeptide by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine).
  • an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.
  • the protuberance and cavity can be made by altering the nucleic acid encoding the polypeptides, e.g. by site-specific mutagenesis, or by peptide synthesis.
  • the threonine residue at position 366 in the CH3 domain of the first subunit of the Fc domain the threonine residue at position 366 is replaced with a tryptophan residue (T366W), and in the CH3 domain of the second subunit of the Fc domain the tyrosine residue at position 407 is replaced with a valine residue (Y407V).
  • the threonine residue at position 366 in the second subunit of the Fc domain additionally the threonine residue at position 366 is replaced with a serine residue (T366S) and the leucine residue at position 368 is replaced with an alanine residue (L368A).
  • the serine residue at position 354 is replaced with a cysteine residue (S354C)
  • the tyrosine residue at position 349 is replaced by a cysteine residue (Y349C).
  • the antigen binding moiety capable of binding to CD3 is fused (optionally via the antigen binding moiety capable of binding to FolR1 on a target cell antigen) to the first subunit of the Fc domain (comprising the “knob” modification).
  • fusion of the antigen binding moiety capable of binding to CD3 to the knob-containing subunit of the Fc domain will (further) minimize the generation of antigen binding molecules comprising two antigen binding moieties capable of binding to CD3 (steric clash of two knob-containing polypeptides).
  • a modification promoting association of the first and the second subunit of the Fc domain comprises a modification mediating electrostatic steering effects, e.g. as described in PCT publication WO 2009/089004.
  • this method involves replacement of one or more amino acid residues at the interface of the two Fc domain subunits by charged amino acid residues so that homodimer formation becomes electrostatically unfavorable but heterodimerization electrostatically favorable.
  • the Fc domain confers to the T cell activating bispecific antigen binding molecule favorable pharmacokinetic properties, including a long serum half-life which contributes to good accumulation in the target tissue and a favorable tissue-blood distribution ratio. At the same time it may, however, lead to undesirable targeting of the T cell activating bispecific antigen binding molecule to cells expressing Fc receptors rather than to the preferred antigen-bearing cells. Moreover, the co-activation of Fc receptor signaling pathways may lead to cytokine release which, in combination with the T cell activating properties and the long half-life of the antigen binding molecule, results in excessive activation of cytokine receptors and severe side effects upon systemic administration. Activation of (Fc receptor-bearing) immune cells other than T cells may even reduce efficacy of the T cell activating bispecific antigen binding molecule due to the potential destruction of T cells e.g. by NK cells.
  • the Fc domain of the T cell activating bispecific antigen binding molecules according to the invention exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG 1 Fc domain.
  • the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits less than 50%, preferably less than 20%, more preferably less than 10% and most preferably less than 5% of the binding affinity to an Fc receptor, as compared to a native IgG 1 Fc domain (or a T cell activating bispecific antigen binding molecule comprising a native IgG 1 Fc domain), and/or less than 50%, preferably less than 20%, more preferably less than 10% and most preferably less than 5% of the effector function, as compared to a native IgG 1 Fc domain domain (or a T cell activating bispecific antigen binding molecule comprising a native IgG 1 Fc domain).
  • the Fc domain domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) does not substantially bind to an Fc receptor and/or induce effector function.
  • the Fc receptor is an Fc ⁇ receptor.
  • the Fc receptor is a human Fc receptor.
  • the Fc receptor is an activating Fc receptor.
  • the Fc receptor is an activating human Fc ⁇ receptor, more specifically human Fc ⁇ RIIIa, Fc ⁇ RI or Fc ⁇ RIIa, most specifically human Fc ⁇ RIIIa.
  • the effector function is one or more selected from the group of CDC, ADCC, ADCP, and cytokine secretion. In a particular embodiment the effector function is ADCC.
  • the Fc domain domain exhibits substantially similar binding affinity to neonatal Fc receptor (FcRn), as compared to a native IgG 1 Fc domain domain.
  • FcRn neonatal Fc receptor
  • Substantially similar binding to FcRn is achieved when the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits greater than about 70%, particularly greater than about 80%, more particularly greater than about 90% of the binding affinity of a native IgG 1 Fc domain (or the T cell activating bispecific antigen binding molecule comprising a native IgG 1 Fc domain) to FcRn.
  • the Fc domain is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain.
  • the Fc domain of the T cell activating bispecific antigen binding molecule comprises one or more amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function. Typically, the same one or more amino acid mutation is present in each of the two subunits of the Fc domain.
  • the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor.
  • the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor by at least 2-fold, at least 5-fold, or at least 10-fold.
  • the combination of these amino acid mutations may reduce the binding affinity of the Fc domain to an Fc receptor by at least 10-fold, at least 20-fold, or even at least 50-fold.
  • the T cell activating bispecific antigen binding molecule comprising an engineered Fc domain exhibits less than 20%, particularly less than 10%, more particularly less than 5% of the binding affinity to an Fc receptor as compared to a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain.
  • the Fc receptor is an Fe ⁇ receptor.
  • the Fc receptor is a human Fc receptor.
  • the Fc receptor is an activating Fc receptor.
  • the Fc receptor is an activating human Fc ⁇ receptor, more specifically human Fc ⁇ RIIIa, Fc ⁇ RI or Fc ⁇ RIIa, most specifically human Fc ⁇ RIIIa.
  • binding to each of these receptors is reduced.
  • binding affinity to a complement component, specifically binding affinity to C1q is also reduced.
  • binding affinity to neonatal Fc receptor (FcRn) is not reduced. Substantially similar binding to FcRn, i.e.
  • the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits greater than about 70% of the binding affinity of a non-engineered form of the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said non-engineered form of the Fc domain) to FcRn.
  • the Fc domain, or T cell activating bispecific antigen binding molecules of the invention comprising said Fc domain may exhibit greater than about 80% and even greater than about 90% of such affinity.
  • the Fc domain of the T cell activating bispecific antigen binding molecule is engineered to have reduced effector function, as compared to a non-engineered Fc domain.
  • the reduced effector function can include, but is not limited to, one or more of the following: reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen-presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced crosslinking of target-bound antibodies, reduced dendritic cell maturation, or reduced T cell priming.
  • CDC complement dependent cytotoxicity
  • ADCC reduced antibody-dependent cell-mediated cytotoxicity
  • ADCP reduced antibody-dependent cellular phagocytosis
  • reduced immune complex-mediated antigen uptake by antigen-presenting cells reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing
  • the reduced effector function is one or more selected from the group of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In a particular embodiment the reduced effector function is reduced ADCC. In one embodiment the reduced ADCC is less than 20% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain).
  • the amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function is an amino acid substitution.
  • the Fc domain comprises an amino acid substitution at a position selected from the group of E233, L234, L235, N297, P331 and P329.
  • the Fc domain comprises an amino acid substitution at a position selected from the group of L234, L235 and P329.
  • the Fc domain comprises the amino acid substitutions L234A and L235A.
  • the Fc domain is an IgG 1 Fc domain, particularly a human IgG 1 Fc domain.
  • the Fc domain comprises an amino acid substitution at position P329.
  • the amino acid substitution is P329A or P329G, particularly P329G.
  • the Fc domain comprises an amino acid substitution at position P329 and a further amino acid substitution at a position selected from E233, L234, L235, N297 and P331.
  • the further amino acid substitution is E233P, L234A, L235A, L235E, N297A, N297D or P331S.
  • the Fc domain comprises amino acid substitutions at positions P329, L234 and L235.
  • the Fc domain comprises the amino acid mutations L234A, L235A and P329G (“P329G LALA”).
  • the Fc domain is an IgG 1 Fc domain, particularly a human IgG 1 Fc domain.
  • the “P329G LALA” combination of amino acid substitutions almost completely abolishes Fc ⁇ receptor binding of a human IgG 1 Fc domain, as described in PCT publication no. WO 2012/130831, incorporated herein by reference in its entirety.
  • WO 2012/130831 also describes methods of preparing such mutant Fc domains and methods for determining its properties such as Fc receptor binding or effector functions.
  • the Fc domain of the T cell activating bispecific antigen binding molecules of the invention is an IgG 4 Fc domain, particularly a human IgG 4 Fc domain.
  • the IgG 4 Fc domain comprises amino acid substitutions at position 5228, specifically the amino acid substitution S228P.
  • the IgG 4 Fc domain comprises an amino acid substitution at position L235, specifically the amino acid substitution L235E.
  • the IgG 4 Fc domain comprises an amino acid substitution at position P329, specifically the amino acid substitution P329G.
  • the IgG 4 Fc domain comprises amino acid substitutions at positions S228, L235 and P329, specifically amino acid substitutions S228P, L235E and P329G.
  • IgG 4 Fc domain mutants and their Fe ⁇ receptor binding properties are described in PCT publication no. WO 2012/130831, incorporated herein by reference in its entirety.
  • the Fc domain exhibiting reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG 1 Fc domain is a human IgG 1 Fc domain comprising the amino acid substitutions L234A, L235A and optionally P329G, or a human IgG 4 Fc domain comprising the amino acid substitutions S228P, L235E and optionally P329G.
  • N-glycosylation of the Fc domain has been eliminated.
  • the Fc domain comprises an amino acid mutation at position N297, particularly an amino acid substitution replacing asparagine by alanine (N297A) or aspartic acid (N297D).
  • Fc domains with reduced Fc receptor binding and/or effector function also include those with substitution of one or more of Fc domain residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Pat. No. 6,737,056).
  • Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called “DANA” Fc mutant with substitution of residues 265 and 297 to alanine (U.S. Pat. No. 7,332,581).
  • Mutant Fc domains can be prepared by amino acid deletion, substitution, insertion or modification using genetic or chemical methods well known in the art. Genetic methods may include site-specific mutagenesis of the encoding DNA sequence, PCR, gene synthesis, and the like. The correct nucleotide changes can be verified for example by sequencing.
  • Binding to Fc receptors can be easily determined e.g. by ELISA, or by Surface Plasmon Resonance (SPR) using standard instrumentation such as a BIAcore instrument (GE Healthcare), and Fc receptors such as may be obtained by recombinant expression. A suitable such binding assay is described herein. Alternatively, binding affinity of Fc domains or cell activating bispecific antigen binding molecules comprising an Fc domain for Fc receptors may be evaluated using cell lines known to express particular Fc receptors, such as human NK cells expressing Fc ⁇ IIIa receptor.
  • Effector function of an Fc domain, or a T cell activating bispecific antigen binding molecule comprising an Fc domain can be measured by methods known in the art.
  • a suitable assay for measuring ADCC is described herein.
  • Other examples of in vitro assays to assess ADCC activity of a molecule of interest are described in U.S. Pat. No. 5,500,362; Hellstrom et al. Proc Natl Acad Sci USA 83, 7059-7063 (1986) and Hellstrom et al., Proc Natl Acad Sci USA 82, 1499-1502 (1985); U.S. Pat. No. 5,821,337; Bruggemann et al., J Exp Med 166, 1351-1361 (1987).
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, Calif.); and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, Wis.)).
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g. in a animal model such as that disclosed in Clynes et al., Proc Natl Acad Sci USA 95, 652-656 (1998).
  • binding of the Fc domain to a complement component, specifically to C1q is reduced.
  • said reduced effector function includes reduced CDC.
  • C1q binding assays may be carried out to determine whether the T cell activating bispecific antigen binding molecule is able to bind C1q and hence has CDC activity. See e.g., C1q and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J Immunol Methods 202, 163 (1996); Cragg et al., Blood 101, 1045-1052 (2003); and Cragg and Glennie, Blood 103, 2738-2743 (2004)).
  • One of skill in the art can appreciate the advantageous efficiency of a molecule that selectively distinguishes between cancerous and non-cancerous, healthy cells.
  • One way to accomplish this goal is by appropriate target selection. Markers expressed exclusively on tumor cells can be employed to selectively target effector molecules or cells to tumor cells while sparing normal cells that do not express such marker. However, in some instances, so called tumor cell markers are also expressed in normal tissue, albeit at lower levels. This expression in normal tissue raises the possibility of toxicity. Thus, there was a need in the art for molecules that can more selectively target tumor cells.
  • the invention described herein provides for T cell activating bispecific antigen binding molecules that selectively target FolR1-positive tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all.
  • the T cell activating bispecific antigen binding molecule comprises at least two, preferably two, FolR1 binding moieties of relatively low affinity that confer an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this embodiment of the invention selectively binds to, and/or induces killing of, tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all. In one embodiment, the T cell activating bispecific antigen binding molecule is in the 2+1 inverted format.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of FolR1-positive tumor cells and not non-tumor cells and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO:
  • the T cell activating bispecific antigen binding molecule does not induce killing of a normal cells having less than about 1000 copies of FolR1 its surface.
  • the invention does not require chemical cross linking or a hybrid approach to be produced. Accordingly, in one embodiment, the invention provides for T cell activating bispecific antigen binding molecule capable of production in CHO cells. In one embodiment, the T cell activating bispecific antigen binding molecule comprises humanized and human polypeptides. In one embodiment, the T cell activating bispecific antigen binding molecule does not cause FcgR crosslinking.
  • the T cell activating bispecific antigen binding molecule is capable of production in CHO cells and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38
  • some embodiments contemplated herein include T cell activating bispecific antigen binding molecules having two binding moieties that confer specific binding to FolR1 and one binding moiety that confers specificity to the T cell activating antigen CD3, wherein each individual FolR1 binding moiety engages the antigen with low affinity. Because the molecule comprises two antigen binding moieties that confer binding to FolR1, the overall avidity of the molecule, nevertheless, provides effective binding to FolR1-expressing target cells and activation of T cells to induce T cell effector function.
  • the T cell activating bispecific antigen binding molecules have a monovalent binding affinity to huFolR1 in the micromolar range and an avidity to huFolR1 in the nanomolar range.
  • the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent K D of about 10 nM to about 40 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent K D of about 10 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent K D of about 10 nM and about 30 nM, respectively. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of at least about 1000 nM.
  • the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of about 1400 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of about 1400 nM and to cynomolgus FolR1 with a monovalent binding K D of about 5600 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent K D of about 10 nM and with a monovalent binding K D of about 1400 nM.
  • the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent K D of about 5.36 pM to about 4 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent K D of about 4 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds murine FolR1 with an apparent K D of about 1.5 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of at least about 1000 nM.
  • the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent K D of about 4 nM, binds murine FolR1 with an apparent K D of about 1.5 nM, and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ
  • the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding K D of at least about 1000 nM and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO:
  • the T cell activating bispecific antigen binding molecules contemplated herein can induce T cell effector function, e.g., cell surface marker expression, cytokine production, T cell mediated killing.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing target cell, such as a human tumor cell, in vitro.
  • the T cell is a CD8 positive T cell.
  • FolR1-expressing human tumor cells include but are not limited to Hela, Skov-3, HT-29, and HRCEpiC cells. Other FolR1 positive human cancer cells that can be used for in vitro testing are readily available to the skilled artisan.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing human tumor cell in vitro with an EC50 of between about 36 pM and about 39573 pM after 24 hours.
  • T cell activating bispecific antigen binding molecules that induce T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 36 pM after 24 hours.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 178.4 pM after 24 hours.
  • the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 134.5 pM or greater after 48 hours.
  • the EC50 can be measure by methods known in the art, for example by methods disclosed herein by the examples.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments induces upregulation of cell surface expression of at least one of CD25 and CD69 on the T cell as measured by flow cytometry.
  • the T cell is a CD4 positive T cell or a CD8 positive T cell.
  • the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to FolR1 expressed on a human tumor cell. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to a conformational epitope on human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3).
  • the antigen binding moiety binds to a FolR1 polypeptide comprising the amino acids 25 to 234 of human FolR1 (SEQ ID NO:227).
  • the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:227, to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:230 and to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 or 229.
  • the T cell activating bispecific antigen binding molecule comprises a FolR1 antigen binding moiety that binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NOs:227, 230 and 231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 or 229, and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50
  • the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:227 and to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228, 229 or 230.
  • the T cell activating bispecific antigen binding molecule comprises a FolR1 antigen binding moiety that binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:227 and to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228, 229 or 230, and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 16, the heavy chain CDR2 of
  • the T cell activating bispecific antigen binding molecules contemplated herein can have agonist, antagonist or neutral effect.
  • agonist effect include induction or enhancement of signaling through the FolR1 upon engagement by the FolR1 binding moiety with the FolR1 receptor on the target cell.
  • antagonist activity include abrogation or reduction of signaling through the FolR1 upon engagement by the FolR1 binding moiety with the FolR1 receptor on the target cell. This can, for example, occur by blocking or reducing the interaction between folate with FolR1. Sequence variants of the embodiments disclosed herein having lower affinity while retaining the above described biological properties are specifically contemplated.
  • the invention also pertains to immunoconjugates comprising a T cell activating bispecific antigen binding molecule conjugated to a cytotoxic agent such as a chemotherapeutic agent, a growth inhibitory agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • a cytotoxic agent such as a chemotherapeutic agent, a growth inhibitory agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • the invention further provides isolated polynucleotides encoding a T cell activating bispecific antigen binding molecule as described herein or a fragment thereof.
  • Polynucleotides of the invention include those that are at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequences set forth in SEQ ID NOs:151-226 including functional fragments or variants thereof.
  • the polynucleotides encoding T cell activating bispecific antigen binding molecules of the invention may be expressed as a single polynucleotide that encodes the entire T cell activating bispecific antigen binding molecule or as multiple (e.g., two or more) polynucleotides that are co-expressed. Polypeptides encoded by polynucleotides that are co-expressed may associate through, e.g., disulfide bonds or other means to form a functional T cell activating bispecific antigen binding molecule.
  • the light chain portion of an antigen binding moiety may be encoded by a separate polynucleotide from the portion of the T cell activating bispecific antigen binding molecule comprising the heavy chain portion of the antigen binding moiety, an Fc domain subunit and optionally (part of) another antigen binding moiety.
  • the heavy chain polypeptides When co-expressed, the heavy chain polypeptides will associate with the light chain polypeptides to form the antigen binding moiety.
  • the portion of the T cell activating bispecific antigen binding molecule comprising one of the two Fc domain subunits and optionally (part of) one or more antigen binding moieties could be encoded by a separate polynucleotide from the portion of the T cell activating bispecific antigen binding molecule comprising the the other of the two Fc domain subunits and optionally (part of) an antigen binding moiety. When co-expressed, the Fc domain subunits will associate to form the Fc domain.
  • the isolated polynucleotide encodes the entire T cell activating bispecific antigen binding molecule according to the invention as described herein. In other embodiments, the isolated polynucleotide encodes a polypeptides comprised in the T cell activating bispecific antigen binding molecule according to the invention as described herein.
  • the present invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable region sequence as shown in SEQ ID NOs 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182 and 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223.
  • the present invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule or fragment thereof, wherein the polynucleotide comprises a sequence that encodes a polypeptide sequence as shown in SEQ ID NOs:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 1, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94,
  • the invention is further directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to a nucleotide sequence shown in SEQ ID NOs 97, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 20
  • the invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a nucleic acid sequence shown in SEQ ID NOs 97, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 12, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222,
  • the invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs 1, 2, 3, 4, 5, 6, 7, 11, 13, 15, 19, 21, 12, 25, 27, 29, 31, 36, 41, 45, 49, 51, 55, 58, 62, 64, 66, 68, 70, 72, 74, 76, 78, 82, 113, 114, 115, 116, 117, 118, 119, 12, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135.
  • the invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule or fragment thereof, wherein the polynucleotide comprises a sequence that encodes a polypeptide comprising one or more sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs: 8, 9, 50, 37, 38, and 39.
  • the invention encompasses an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes the variable region sequence of SEQ ID NOs 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182 and 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223 with conservative amino acid substitutions.
  • the invention also encompasses an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or fragment thereof, wherein the polynucleotide comprises a sequence that encodes the polypeptide sequence of SEQ ID NOs 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 1, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96
  • RNA for example, in the form of messenger RNA (mRNA).
  • mRNA messenger RNA
  • RNA of the present invention may be single stranded or double stranded.
  • T cell activating bispecific antigen binding molecules of the invention may be obtained, for example, by solid-state peptide synthesis (e.g. Merrifield solid phase synthesis) or recombinant production.
  • solid-state peptide synthesis e.g. Merrifield solid phase synthesis
  • Such polynucleotide may be readily isolated and sequenced using conventional procedures.
  • a vector, preferably an expression vector, comprising one or more of the polynucleotides of the invention is provided.
  • Methods which are well known to those skilled in the art can be used to construct expression vectors containing the coding sequence of a T cell activating bispecific antigen binding molecule (fragment) along with appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al., M OLECULAR C LONING : A L ABORATORY M ANUAL , Cold Spring Harbor Laboratory, N.Y.
  • the expression vector can be part of a plasmid, virus, or may be a nucleic acid fragment.
  • the expression vector includes an expression cassette into which the polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment) (i.e. the coding region) is cloned in operable association with a promoter and/or other transcription or translation control elements.
  • a “coding region” is a portion of nucleic acid which consists of codons translated into amino acids.
  • a “stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, if present, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, 5′ and 3′ untranslated regions, and the like, are not part of a coding region.
  • Two or more coding regions can be present in a single polynucleotide construct, e.g. on a single vector, or in separate polynucleotide constructs, e.g. on separate (different) vectors.
  • any vector may contain a single coding region, or may comprise two or more coding regions, e.g.
  • a vector of the present invention may encode one or more polypeptides, which are post- or co-translationally separated into the final proteins via proteolytic cleavage.
  • a vector, polynucleotide, or nucleic acid of the invention may encode heterologous coding regions, either fused or unfused to a polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment) of the invention, or variant or derivative thereof.
  • Heterologous coding regions include without limitation specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain. An operable association is when a coding region for a gene product, e.g.
  • a polypeptide is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s).
  • Two DNA fragments (such as a polypeptide coding region and a promoter associated therewith) are “operably associated” if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not interfere with the ability of the expression regulatory sequences to direct the expression of the gene product or interfere with the ability of the DNA template to be transcribed.
  • a promoter region would be operably associated with a nucleic acid encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid.
  • the promoter may be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells.
  • Other transcription control elements besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription.
  • Suitable promoters and other transcription control regions are disclosed herein.
  • a variety of transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions, which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (e.g. the immediate early promoter, in conjunction with intron-A), simian virus 40 (e.g.
  • transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit a-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as inducible promoters (e.g. promoters inducible tetracyclins). Similarly, a variety of translation control elements are known to those of ordinary skill in the art.
  • the expression cassette may also include other features such as an origin of replication, and/or chromosome integration elements such as retroviral long terminal repeats (LTRs), or adeno-associated viral (AAV) inverted terminal repeats (ITRs).
  • LTRs retroviral long terminal repeats
  • AAV adeno-associated viral
  • Polynucleotide and nucleic acid coding regions of the present invention may be associated with additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide of the present invention.
  • additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide of the present invention.
  • DNA encoding a signal sequence may be placed upstream of the nucleic acid encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof.
  • proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated.
  • polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the translated polypeptide to produce a secreted or “mature” form of the polypeptide.
  • the native signal peptide e.g.
  • an immunoglobulin heavy chain or light chain signal peptide is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it.
  • a heterologous mammalian signal peptide, or a functional derivative thereof may be used.
  • the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator (TPA) or mouse ⁇ -glucuronidase.
  • DNA encoding a short protein sequence that could be used to facilitate later purification (e.g. a histidine tag) or assist in labeling the T cell activating bispecific antigen binding molecule may be included within or at the ends of the T cell activating bispecific antigen binding molecule (fragment) encoding polynucleotide.
  • a host cell comprising one or more polynucleotides of the invention.
  • a host cell comprising one or more vectors of the invention.
  • the polynucleotides and vectors may incorporate any of the features, singly or in combination, described herein in relation to polynucleotides and vectors, respectively.
  • a host cell comprises (e.g. has been transformed or transfected with) a vector comprising a polynucleotide that encodes (part of) a T cell activating bispecific antigen binding molecule of the invention.
  • the term “host cell” refers to any kind of cellular system which can be engineered to generate the T cell activating bispecific antigen binding molecules of the invention or fragments thereof.
  • Host cells suitable for replicating and for supporting expression of T cell activating bispecific antigen binding molecules are well known in the art. Such cells may be transfected or transduced as appropriate with the particular expression vector and large quantities of vector containing cells can be grown for seeding large scale fermenters to obtain sufficient quantities of the T cell activating bispecific antigen binding molecule for clinical applications.
  • Suitable host cells include prokaryotic microorganisms, such as E. coli , or various eukaryotic cells, such as Chinese hamster ovary cells (CHO), insect cells, or the like.
  • polypeptides may be produced in bacteria in particular when glycosylation is not needed. After expression, the polypeptide may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been “humanized”, resulting in the production of a polypeptide with a partially or fully human glycosylation pattern. See Gerngross, Nat Biotech 22, 1409-1414 (2004), and Li et al., Nat Biotech 24, 210-215 (2006).
  • Suitable host cells for the expression of (glycosylated) polypeptides are also derived from multicellular organisms (invertebrates and vertebrates).
  • invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures can also be utilized as hosts. See e.g. U.S. Pat. Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293T cells as described, e.g., in Graham et al., J Gen Virol 36, 59 (1977)), baby hamster kidney cells (BHK), mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol Reprod 23, 243-251 (1980)), monkey kidney cells (CV1), African green monkey kidney cells (VERO-76), human cervical carcinoma cells (HELA), canine kidney cells (MDCK), buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver cells (Hep G2), mouse mammary tumor cells (MMT 060562), TRI cells (as described, e.g., in Mather et al., Annals N.Y.
  • MRC 5 cells MRC 5 cells
  • FS4 cells Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including dhfr ⁇ CHO cells (Urlaub et al., Proc Natl Acad Sci USA 77, 4216 (1980)); and myeloma cell lines such as YO, NS0, P3X63 and Sp2/0.
  • CHO Chinese hamster ovary
  • dhfr ⁇ CHO cells Urlaub et al., Proc Natl Acad Sci USA 77, 4216 (1980)
  • myeloma cell lines such as YO, NS0, P3X63 and Sp2/0.
  • Yazaki and Wu Methods in Molecular Biology, Vol. 248 (B. K. C. Lo, ed., Humana Press, Totowa, N.J.), pp. 255-268 (2003).
  • Host cells include cultured cells, e.g., mammalian cultured cells, yeast cells, insect cells, bacterial cells and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
  • the host cell is a eukaryotic cell, preferably a mammalian cell, such as a Chinese Hamster Ovary (CHO) cell, a human embryonic kidney (HEK) cell or a lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • CHO Chinese Hamster Ovary
  • HEK human embryonic kidney
  • a lymphoid cell e.g., Y0, NS0, Sp20 cell.
  • Cells expressing a polypeptide comprising either the heavy or the light chain of an antigen binding domain such as an antibody may be engineered so as to also express the other of the antibody chains such that the expressed product is an antibody that has both a heavy and a light chain.
  • a method of producing a T cell activating bispecific antigen binding molecule according to the invention comprises culturing a host cell comprising a polynucleotide encoding the T cell activating bispecific antigen binding molecule, as provided herein, under conditions suitable for expression of the T cell activating bispecific antigen binding molecule, and recovering the T cell activating bispecific antigen binding molecule from the host cell (or host cell culture medium).
  • T cell activating bispecific antigen binding molecule The components of the T cell activating bispecific antigen binding molecule are genetically fused to each other.
  • T cell activating bispecific antigen binding molecule can be designed such that its components are fused directly to each other or indirectly through a linker sequence.
  • the composition and length of the linker may be determined in accordance with methods well known in the art and may be tested for efficacy. Examples of linker sequences between different components of T cell activating bispecific antigen binding molecules are found in the sequences provided herein. Additional sequences may also be included to incorporate a cleavage site to separate the individual components of the fusion if desired, for example an endopeptidase recognition sequence.
  • the one or more antigen binding moieties of the T cell activating bispecific antigen binding molecules comprise at least an antibody variable region capable of binding an antigenic determinant.
  • Variable regions can form part of and be derived from naturally or non-naturally occurring antibodies and fragments thereof.
  • Methods to produce polyclonal antibodies and monoclonal antibodies are well known in the art (see e.g. Harlow and Lane, “Antibodies, a laboratory manual”, Cold Spring Harbor Laboratory, 1988).
  • Non-naturally occurring antibodies can be constructed using solid phase-peptide synthesis, can be produced recombinantly (e.g. as described in U.S. Pat. No. 4,186,567) or can be obtained, for example, by screening combinatorial libraries comprising variable heavy chains and variable light chains (see e.g. U.S. Pat. No. 5,969,108, McCafferty).
  • any animal species of antibody, antibody fragment, antigen binding domain or variable region can be used in the T cell activating bispecific antigen binding molecules of the invention.
  • Non-limiting antibodies, antibody fragments, antigen binding domains or variable regions useful in the present invention can be of murine, primate, or human origin. If the T cell activating bispecific antigen binding molecule is intended for human use, a chimeric form of antibody may be used wherein the constant regions of the antibody are from a human.
  • a humanized or fully human form of the antibody can also be prepared in accordance with methods well known in the art (see e.g. U.S. Pat. No. 5,565,332 to Winter).
  • Humanization may be achieved by various methods including, but not limited to (a) grafting the non-human (e.g., donor antibody) CDRs onto human (e.g. recipient antibody) framework and constant regions with or without retention of critical framework residues (e.g. those that are important for retaining good antigen binding affinity or antibody functions), (b) grafting only the non-human specificity-determining regions (SDRs or a-CDRs; the residues critical for the antibody-antigen interaction) onto human framework and constant regions, or (c) transplanting the entire non-human variable domains, but “cloaking” them with a human-like section by replacement of surface residues.
  • a grafting the non-human (e.g., donor antibody) CDRs onto human (e.g. recipient antibody) framework and constant regions with or without retention of critical framework residues (e.g. those that are important for retaining good antigen binding affinity or antibody functions)
  • SDRs or a-CDRs the residues critical for the antibody-antigen interaction
  • Human antibodies and human variable regions can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr Opin Pharmacol 5, 368-74 (2001) and Lonberg, Curr Opin Immunol 20, 450-459 (2008). Human variable regions can form part of and be derived from human monoclonal antibodies made by the hybridoma method (see e.g. Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)). Human antibodies and human variable regions may also be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge (see e.g.
  • Human antibodies and human variable regions may also be generated by isolating Fv clone variable region sequences selected from human-derived phage display libraries (see e.g., Hoogenboom et al. in Methods in Molecular Biology 178, 1-37 (O'Brien et al., ed., Human Press, Totowa, N.J., 2001); and McCafferty et al., Nature 348, 552-554; Clackson et al., Nature 352, 624-628 (1991)). Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • scFv single-chain Fv
  • the antigen binding moieties useful in the present invention are engineered to have enhanced binding affinity according to, for example, the methods disclosed in U.S. Pat. Appl. Publ. No. 2004/0132066, the entire contents of which are hereby incorporated by reference.
  • the ability of the T cell activating bispecific antigen binding molecule of the invention to bind to a specific antigenic determinant can be measured either through an enzyme-linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g.
  • Competition assays may be used to identify an antibody, antibody fragment, antigen binding domain or variable domain that competes with a reference antibody for binding to a particular antigen, e.g. an antibody that competes with the V9 antibody for binding to CD3.
  • a competing antibody binds to the same epitope (e.g. a linear or a conformational epitope) that is bound by the reference antibody.
  • immobilized antigen e.g. CD3
  • a first labeled antibody that binds to the antigen (e.g. V9 antibody, described in U.S. Pat. No. 6,054,297)
  • a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to the antigen.
  • the second antibody may be present in a hybridoma supernatant.
  • immobilized antigen is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to the antigen, excess unbound antibody is removed, and the amount of label associated with immobilized antigen is measured. If the amount of label associated with immobilized antigen is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to the antigen. See Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.).
  • T cell activating bispecific antigen binding molecules prepared as described herein may be purified by art-known techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography, and the like.
  • the actual conditions used to purify a particular protein will depend, in part, on factors such as net charge, hydrophobicity, hydrophilicity etc., and will be apparent to those having skill in the art.
  • affinity chromatography purification an antibody, ligand, receptor or antigen can be used to which the T cell activating bispecific antigen binding molecule binds.
  • a matrix with protein A or protein G may be used for affinity chromatography purification of T cell activating bispecific antigen binding molecules of the invention.
  • Sequential Protein A or G affinity chromatography and size exclusion chromatography can be used to isolate a T cell activating bispecific antigen binding molecule essentially as described in the Examples.
  • the purity of the T cell activating bispecific antigen binding molecule can be determined by any of a variety of well known analytical methods including gel electrophoresis, high pressure liquid chromatography, and the like.
  • the heavy chain fusion proteins expressed as described in the Examples were shown to be intact and properly assembled as demonstrated by reducing SDS-PAGE (see e.g. FIG. 2 ). Three bands were resolved at approximately Mr 25,000, Mr 50,000 and Mr 75,000, corresponding to the predicted molecular weights of the T cell activating bispecific antigen binding molecule light chain, heavy chain and heavy chain/light chain fusion protein.
  • T cell activating bispecific antigen binding molecules provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
  • the affinity of the T cell activating bispecific antigen binding molecule for an Fc receptor or a target antigen can be determined in accordance with the methods set forth in the Examples by surface plasmon resonance (SPR), using standard instrumentation such as a BIAcore instrument (GE Healthcare), and receptors or target proteins such as may be obtained by recombinant expression.
  • SPR surface plasmon resonance
  • GE Healthcare BIAcore instrument
  • receptors or target proteins such as may be obtained by recombinant expression.
  • binding of T cell activating bispecific antigen binding molecules for different receptors or target antigens may be evaluated using cell lines expressing the particular receptor or target antigen, for example by flow cytometry (FACS).
  • FACS flow cytometry
  • K D is measured by surface plasmon resonance using a BIACORE® T100 machine (GE Healthcare) at 25° C.
  • CMS carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N′-(3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Penta-His antibody (“Penta-His” disclosed as SEQ ID NO: 306) is diluted with 10 mM sodium acetate, pH 5.0, to 40 ⁇ g/ml before injection at a flow rate of 5 ⁇ l/min to achieve approximately 6500 response units (RU) of coupled protein. Following the injection of the ligand, 1 M ethanolamine is injected to block unreacted groups. Subsequently the Fc-receptor is captured for 60 s at 4 or 10 nM.
  • HBS-EP GE Healthcare, 10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% Surfactant P20, pH 7.4
  • HBS-EP GE Healthcare, 10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% Surfactant P20, pH 7.4
  • bispecific constructs are captured by an anti human Fab specific antibody (GE Healthcare) that is immobilized on an activated CM5-sensor chip surface as described for the anti Penta-His antibody (“Penta-His” disclosed as SEQ ID NO: 306).
  • the final amount of coupled protein is approximately 12000 R U.
  • the bispecific constructs are captured for 90 s at 300 nM.
  • the target antigens are passed through the flow cells for 180 s at a concentration range from 250 to 1000 nM with a flowrate of 30 ⁇ l/min.
  • the dissociation is monitored for 180 s. Bulk refractive index differences are corrected for by subtracting the response obtained on reference flow cell.
  • the steady state response was used to derive the dissociation constant K D by non-linear curve fitting of the Langmuir binding isotherm.
  • Association rates (k on ) and dissociation rates (k off ) are calculated using a simple one-to-one Langmuir binding model (BIACORE® T100 Evaluation Software version 1.1.1) by simultaneously fitting the association and dissociation sensorgrams.
  • the equilibrium dissociation constant (K D ) is calculated as the ratio k off /k on . See, e.g., Chen et al., J Mol Biol 293, 865-881 (1999).
  • Biological activity of the T cell activating bispecific antigen binding molecules of the invention can be measured by various assays as described in the Examples. Biological activities may for example include the induction of proliferation of T cells, the induction of signaling in T cells, the induction of expression of activation markers in T cells, the induction of cytokine secretion by T cells, the induction of lysis of target cells such as tumor cells, and the induction of tumor regression and/or the improvement of survival.
  • compositions Compositions, Formulations, and Routes of Administration
  • the invention provides pharmaceutical compositions comprising any of the T cell activating bispecific antigen binding molecules provided herein, e.g., for use in any of the below therapeutic methods.
  • a pharmaceutical composition comprises any of the T cell activating bispecific antigen binding molecules provided herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprises any of the T cell activating bispecific antigen binding molecules provided herein and at least one additional therapeutic agent, e.g., as described below.
  • a method of producing a T cell activating bispecific antigen binding molecule of the invention in a form suitable for administration in vivo comprising (a) obtaining a T cell activating bispecific antigen binding molecule according to the invention, and (b) formulating the T cell activating bispecific antigen binding molecule with at least one pharmaceutically acceptable carrier, whereby a preparation of T cell activating bispecific antigen binding molecule is formulated for administration in vivo.
  • compositions of the present invention comprise a therapeutically effective amount of one or more T cell activating bispecific antigen binding molecule dissolved or dispersed in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to molecular entities and compositions that are generally non-toxic to recipients at the dosages and concentrations employed, i.e. do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate.
  • the preparation of a pharmaceutical composition that contains at least one T cell activating bispecific antigen binding molecule and optionally an additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 18th Ed.
  • compositions are lyophilized formulations or aqueous solutions.
  • pharmaceutically acceptable carrier includes any and all solvents, buffers, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g.
  • antibacterial agents antifungal agents
  • isotonic agents absorption delaying agents, salts, preservatives, antioxidants, proteins, drugs, drug stabilizers, polymers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329, incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
  • T cell activating bispecific antigen binding molecules of the present invention can be administered intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrasplenically, intrarenally, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, intratumorally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, topically, locally, by inhalation (e.g.
  • aerosol inhalation injection, infusion, continuous infusion, localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g. liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference).
  • Parenteral administration in particular intravenous injection, is most commonly used for administering polypeptide molecules such as the T cell activating bispecific antigen binding molecules of the invention.
  • compositions include those designed for administration by injection, e.g. subcutaneous, intradermal, intralesional, intravenous, intraarterial intramuscular, intrathecal or intraperitoneal injection.
  • the T cell activating bispecific antigen binding molecules of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • the solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the T cell activating bispecific antigen binding molecules may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • Sterile injectable solutions are prepared by incorporating the T cell activating bispecific antigen binding molecules of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated below, as required. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and/or the other ingredients. In the case of sterile powders for the preparation of sterile injectable solutions, suspensions or emulsion, the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered liquid medium thereof.
  • the liquid medium should be suitably buffered if necessary and the liquid diluent first rendered isotonic prior to injection with sufficient saline or glucose.
  • the composition must be stable under the conditions of manufacture and storage, and preserved against the contaminating action of microorganisms, such as bacteria and fungi. It will be appreciated that endotoxin contamination should be kept minimally at a safe level, for example, less that 0.5 ng/mg protein.
  • Suitable pharmaceutically acceptable carriers include, but are not limited to: 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
  • Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl cleats or triglycerides, or liposomes.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) 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.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • prolonged absorption of an injectable composition can be brought about by the use in the compositions of agents delaying absorption, such as, for example, aluminum monostearate, gelatin or combinations thereof.
  • the T cell activating bispecific antigen binding molecules may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the T cell activating bispecific antigen binding molecules may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions comprising the T cell activating bispecific antigen binding molecules of the invention may be manufactured by means of conventional mixing, dissolving, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the proteins into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the T cell activating bispecific antigen binding molecules may be formulated into a composition in a free acid or base, neutral or salt form.
  • Pharmaceutically acceptable salts are salts that substantially retain the biological activity of the free acid or base. These include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
  • T cell activating bispecific antigen binding molecules may be used in therapeutic methods.
  • T cell activating bispecific antigen binding molecules of the invention can be used as immunotherapeutic agents, for example in the treatment of cancers.
  • T cell activating bispecific antigen binding molecules of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • T cell activating bispecific antigen binding molecules of the invention for use as a medicament are provided.
  • T cell activating bispecific antigen binding molecules of the invention for use in treating a disease are provided.
  • T cell activating bispecific antigen binding molecules of the invention for use in a method of treatment are provided.
  • the invention provides a T cell activating bispecific antigen binding molecule as described herein for use in the treatment of a disease in an individual in need thereof.
  • the invention provides a T cell activating bispecific antigen binding molecule for use in a method of treating an individual having a disease comprising administering to the individual a therapeutically effective amount of the T cell activating bispecific antigen binding molecule.
  • the disease to be treated is a proliferative disorder.
  • the disease is cancer.
  • the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer.
  • the invention provides a T cell activating bispecific antigen binding molecule as described herein for use in inducing lysis of a target cell, particularly a tumor cell.
  • the invention provides a T cell activating bispecific antigen binding molecule for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the T cell activating bispecific antigen binding molecule to induce lysis of a target cell.
  • An “individual” according to any of the above embodiments is a mammal, preferably a human.
  • the invention provides for the use of a T cell activating bispecific antigen binding molecule of the invention in the manufacture or preparation of a medicament.
  • the medicament is for the treatment of a disease in an individual in need thereof.
  • the medicament is for use in a method of treating a disease comprising administering to an individual having the disease a therapeutically effective amount of the medicament.
  • the disease to be treated is a proliferative disorder.
  • the disease is cancer.
  • the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer.
  • the medicament is for inducing lysis of a target cell, particularly a tumor cell.
  • the medicament is for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the medicament to induce lysis of a target cell.
  • An “individual” according to any of the above embodiments may be a mammal, preferably a human.
  • the invention provides a method for treating a disease.
  • the method comprises administering to an individual having such disease a therapeutically effective amount of a T cell activating bispecific antigen binding molecule of the invention.
  • a composition is administered to said individual, comprising the T cell activating bispecific antigen binding molecule of the invention in a pharmaceutically acceptable form.
  • the disease to be treated is a proliferative disorder.
  • the disease is cancer.
  • the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer.
  • An “individual” according to any of the above embodiments may be a mammal, preferably a human.
  • the invention provides a method for inducing lysis of a target cell, particularly a tumor cell.
  • the method comprises contacting a target cell with a T cell activating bispecific antigen binding molecule of the invention in the presence of a T cell, particularly a cytotoxic T cell.
  • a method for inducing lysis of a target cell, particularly a tumor cell, in an individual is provided.
  • the method comprises administering to the individual an effective amount of a T cell activating bispecific antigen binding molecule to induce lysis of a target cell.
  • an “individual” is a human.
  • the disease to be treated is a proliferative disorder, particularly cancer.
  • cancers include bladder cancer, brain cancer, head and neck cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, uterine cancer, cervical cancer, endometrial cancer, esophageal cancer, colon cancer, colorectal cancer, rectal cancer, gastric cancer, prostate cancer, blood cancer, skin cancer, squamous cell carcinoma, bone cancer, and kidney cancer.
  • the cancer is chosen from the group consisting of renal cell cancer, skin cancer, lung cancer, colorectal cancer, breast cancer, brain cancer, head and neck cancer.
  • T cell activating bispecific antigen binding molecule may not provide a cure but may only provide partial benefit.
  • a physiological change having some benefit is also considered therapeutically beneficial.
  • an amount of T cell activating bispecific antigen binding molecule that provides a physiological change is considered an “effective amount” or a “therapeutically effective amount”.
  • the subject, patient, or individual in need of treatment is typically a mammal, more specifically a human.
  • an effective amount of a T cell activating bispecific antigen binding molecule of the invention is administered to a cell. In other embodiments, a therapeutically effective amount of a T cell activating bispecific antigen binding molecule of the invention is administered to an individual for the treatment of disease.
  • the appropriate dosage of a T cell activating bispecific antigen binding molecule of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the route of administration, the body weight of the patient, the type of T cell activating bispecific antigen binding molecule, the severity and course of the disease, whether the T cell activating bispecific antigen binding molecule is administered for preventive or therapeutic purposes, previous or concurrent therapeutic interventions, the patient's clinical history and response to the T cell activating bispecific antigen binding molecule, and the discretion of the attending physician.
  • the practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • the T cell activating bispecific antigen binding molecule is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.1 mg/kg-10 mg/kg) of T cell activating bispecific antigen binding molecule can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10 milligram/kg body weight, about 50 milligram/kg body weight, about 100 milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram/kg body weight to about 500 milligram/kg body weight, etc. can be administered, based on the numbers described above.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the T cell activating bispecific antigen binding molecule).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • the T cell activating bispecific antigen binding molecules of the invention will generally be used in an amount effective to achieve the intended purpose.
  • the T cell activating bispecific antigen binding molecules of the invention, or pharmaceutical compositions thereof are administered or applied in a therapeutically effective amount. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • a therapeutically effective dose can be estimated initially from in vitro assays, such as cell culture assays.
  • a dose can then be formulated in animal models to achieve a circulating concentration range that includes the IC 50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the T cell activating bispecific antigen binding molecules which are sufficient to maintain therapeutic effect.
  • Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day.
  • Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
  • the effective local concentration of the T cell activating bispecific antigen binding molecules may not be related to plasma concentration.
  • One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
  • a therapeutically effective dose of the T cell activating bispecific antigen binding molecules described herein will generally provide therapeutic benefit without causing substantial toxicity.
  • Toxicity and therapeutic efficacy of a T cell activating bispecific antigen binding molecule can be determined by standard pharmaceutical procedures in cell culture or experimental animals. Cell culture assays and animal studies can be used to determine the LD 50 (the dose lethal to 50% of a population) and the ED 50 (the dose therapeutically effective in 50% of a population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD 50 /ED 50 . T cell activating bispecific antigen binding molecules that exhibit large therapeutic indices are preferred.
  • the T cell activating bispecific antigen binding molecule according to the present invention exhibits a high therapeutic index.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans.
  • the dosage lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al., 1975, in: The Pharmacological Basis of Therapeutics, Ch. 1, p.
  • the attending physician for patients treated with T cell activating bispecific antigen binding molecules of the invention would know how and when to terminate, interrupt, or adjust administration due to toxicity, organ dysfunction, and the like. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.
  • the T cell activating bispecific antigen binding molecules of the invention may be administered in combination with one or more other agents in therapy.
  • a T cell activating bispecific antigen binding molecule of the invention may be co-administered with at least one additional therapeutic agent.
  • therapeutic agent encompasses any agent administered to treat a symptom or disease in an individual in need of such treatment.
  • additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • an additional therapeutic agent is an immunomodulatory agent, a cytostatic agent, an inhibitor of cell adhesion, a cytotoxic agent, an activator of cell apoptosis, or an agent that increases the sensitivity of cells to apoptotic inducers.
  • the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.
  • Such other agents are suitably present in combination in amounts that are effective for the purpose intended.
  • the effective amount of such other agents depends on the amount of T cell activating bispecific antigen binding molecule used, the type of disorder or treatment, and other factors discussed above.
  • the T cell activating bispecific antigen binding molecules are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate compositions), and separate administration, in which case, administration of the T cell activating bispecific antigen binding molecule of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • T cell activating bispecific antigen binding molecules of the invention can also be used in combination with radiation therapy.
  • the invention provides for a bispecific antibody comprising a) a first antigen-binding site that comprises a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31; and b) a second antigen-binding site that comprises a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31 for use in combination with an antibody to PD-L1 or FAP-4-1BBL.
  • the bispecific antibody further comprises a third antigen-binding site that comprises a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and 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).
  • At least one active agent in the composition is a T cell activating bispecific antigen binding molecule of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a T cell activating bispecific antigen binding molecule of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • Ringer's solution such as phosphate
  • DNA sequences were determined by standard double strand sequencing at Synergene (Schlieren).
  • Desired gene segments where required were either generated by PCR using appropriate templates or were synthesized by Geneart AG (Regensburg, Germany) from synthetic oligonucleotides and PCR products by automated gene synthesis. In cases where no exact gene sequence was available, oligonucleotide primers were designed based on sequences from closest homologues and the genes were isolated by RT-PCR from RNA originating from the appropriate tissue. The gene segments flanked by singular restriction endonuclease cleavage sites were cloned into standard cloning/sequencing vectors. The plasmid DNA was purified from transformed bacteria and concentration determined by UV spectroscopy. The DNA sequence of the subcloned gene fragments was confirmed by DNA sequencing. Gene segments were designed with suitable restriction sites to allow sub-cloning into the respective expression vectors. All constructs were designed with a 5′-end DNA sequence coding for a leader peptide which targets proteins for secretion in eukaryotic cells.
  • PBMCs Peripheral blood mononuclear cells
  • enriched lymphocyte preparations obtained from local blood banks or from fresh blood from healthy human donors. Briefly, blood was diluted with sterile PBS and carefully layered over a Histopaque gradient (Sigma, H8889). After centrifugation for 30 minutes at 450 ⁇ g at room temperature (brake switched off), part of the plasma above the PBMC containing interphase was discarded. The PBMCs were transferred into new 50 ml Falcon tubes and tubes were filled up with PBS to a total volume of 50 ml. The mixture was centrifuged at room temperature for 10 minutes at 400 ⁇ g (brake switched on).
  • PBMC pellet washed twice with sterile PBS (centrifugation steps at 4° C. for 10 minutes at 350 ⁇ g).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium, containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO 2 in the incubator until assay start.
  • T cell enrichment from PBMCs was performed using the Pan T Cell Isolation Kit II (Miltenyi Biotec #130-091-156), according to the manufacturer's instructions.
  • the cell pellets were diluted in 40 ⁇ l cold buffer per 10 million cells (PBS with 0.5% BSA, 2 mM EDTA, sterile filtered) and incubated with 10 ⁇ l Biotin-Antibody Cocktail per 10 million cells for 10 min at 4° C. 30 ⁇ l cold buffer and 20 ⁇ l Anti-Biotin magnetic beads per 10 million cells were added, and the mixture incubated for another 15 min at 4° C. Cells were washed by adding 10-20 ⁇ the current volume and a subsequent centrifugation step at 300 ⁇ g for 10 min. Up to 100 million cells were resuspended in 500 ⁇ l buffer.
  • Magnetic separation of unlabeled human pan T cells was performed using LS columns (Miltenyi Biotec #130-042-401) according to the manufacturer's instructions. The resulting T cell population was counted automatically (ViCell) and stored in AIM-V medium at 37° C., 5% CO 2 in the incubator until assay start (not longer than 24 h).
  • PBMCs Peripheral blood mononuclar cells
  • enriched lymphocyte preparations obtained from local blood banks or from fresh blood from healthy human donors.
  • T-cell enrichment from PBMCs was performed using the Naive CD8 + T cell isolation Kit from Miltenyi Biotec (#130-093-244), according to the manufacturer's instructions, but skipping the last isolation step of CD8 + T cells (also see description for the isolation of primary human pan T cells).
  • Spleens were isolated from C57BL/6 mice, transferred into a GentleMACS C-tube (Miltenyi Biotech #130-093-237) containing MACS buffer (PBS+0.5% BSA+2 mM EDTA) and dissociated with the GentleMACS Dissociator to obtain single-cell suspensions according to the manufacturer's instructions.
  • the cell suspension was passed through a pre-separation filter to remove remaining undissociated tissue particles. After centrifugation at 400 ⁇ g for 4 min at 4° C., ACK Lysis Buffer was added to lyse red blood cells (incubation for 5 min at room temperature). The remaining cells were washed with MACS buffer twice, counted and used for the isolation of murine pan T cells.
  • the negative (magnetic) selection was performed using the Pan T Cell Isolation Kit from Miltenyi Biotec (#130-090-861), following the manufacturer's instructions.
  • the resulting T cell population was automatically counted (ViCell) and immediately used for further assays.
  • PBMCs Peripheral blood mononuclar cells
  • Heparinized blood was diluted 1:3 with sterile PBS
  • Lymphoprep medium (Axon Lab #1114545) was diluted to 90% with sterile PBS.
  • Two volumes of the diluted blood were layered over one volume of the diluted density gradient and the PBMC fraction was separated by centrifugation for 30 min at 520 ⁇ g, without brake, at room temperature.
  • the PBMC band was transferred into a fresh 50 ml Falcon tube and washed with sterile PBS by centrifugation for 10 min at 400 ⁇ g at 4° C.
  • One low-speed centrifugation was performed to remove the platelets (15 min at 150 ⁇ g, 4° C.), and the resulting PBMC population was automatically counted (ViCell) and immediately used for further assays.
  • the following antigens and screening tools were generated as monovalent Fc fusion proteins (the extracellular domain of the antigen linked to the hinge region of Fc-knob which is co-expressed with an Fc-hole molecule).
  • the antigen genes were synthesized (Geneart, Regensburg, Germany) based on sequences obtained from GenBank or SwissProt and inserted into expression vectors to generate fusion proteins with Fc-knob with a C-terminal Avi-tag for in vivo or in vitro biotinylation. In vivo biotinylation was achieved by co-expression of the bacterial birA gene encoding a bacterial biotin ligase during production. Expression of all genes was under control of a chimeric MPSV promoter on a plasmid containing an oriP element for stable maintenance of the plasmids in EBNA containing cell lines.
  • HEK293 EBNA cells were seeded 24 hours before transfection.
  • transfection cells were centrifuged for 5 minutes at 210 g, and supernatant was replaced by pre-warmed CD CHO medium.
  • Expression vectors were resuspended in 20 mL of CD CHO medium containing 200 ⁇ g of vector DNA. After addition of 540 ⁇ L of polyethylenimine (PEI), the solution was mixed for 15 seconds and incubated for 10 minutes at room temperature. Afterwards, cells were mixed with the DNA/PEI solution, transferred to a 500 mL shake flask and incubated for 3 hours at 37° C. in an incubator with a 5% CO 2 atmosphere.
  • PEI polyethylenimine
  • F17 medium After the incubation, 160 mL of F17 medium was added and cells were cultured for 24 hours. One day after transfection, 1 mM valproic acid and 7% Feed 1 (Lonza) were added to the culture. The production medium was also supplemented with 100 ⁇ M biotin. After 7 days of culturing, the cell supernatant was collected by spinning down cells for 15 min at 210 g. The solution was sterile filtered (0.22 ⁇ m filter), supplemented with sodium azide to a final concentration of 0.01% (w/v), and kept at 4° C.
  • Secreted proteins were purified from cell culture supernatants by affinity chromatography using Protein A, followed by size exclusion chromatography.
  • the column was then washed with 10 column volumes of 20 mM sodium citrate, 100 mM sodium chloride, 100 mM glycine, pH 3.0. pH of collected fractions was adjusted by adding 1/10 (v/v) of 0.5 M sodium phosphate, pH 8.0.
  • the protein was concentrated and filtered prior to loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM histidine, 140 mM sodium chloride, pH 6.0.
  • the protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the FolR1-Fc-fusion was analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer).
  • the aggregate content of samples was analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C.
  • Purified antigen-Fc-fusion proteins were analyzed by surface plasmon resonance assays using commercially available antibodies to confirm correct and natural conformation of the antigens (data not shown).
  • the T cell activating bispecific molecules described herein comprise at least one CD3 binding moiety.
  • This moiety can be generated by immunizing laboratory animals, screening phage library or using known anti-CD3 antibodies.
  • the common light chain with CD3c specificity was generated by humanizing the light chain of a murine parental anti-CD3c antibody (CH2527).
  • CH2527 murine parental anti-CD3c antibody
  • acceptor framework sequences are selected by aligning the sequence of the donor to a collection of potential acceptor sequences and choosing one that has either reasonable homology to the donor, or shows similar amino acids at some positions critical for structure and activity.
  • the search for the antibody acceptor framework was performed by aligning the mouse VL-domain sequence of the parental antibody to a collection of human germline sequences and choosing the human sequence that showed high sequence identity.
  • a good match in terms of framework sequence homology was found in a rather infrequent human light chain belonging to the V-domain family 7 of the lambda type, more precisely, hVL_7_46 (IMGT nomenclature, GenBank Acc No. Z73674).
  • This infrequent human light chain was subsequently chosen as acceptor framework for humanization of the light chain of CH2527.
  • the three complementarity determining regions (CDRs) of the mouse light chain variable domain were grafted onto this acceptor framework. Since the framework 4 region is not part of the variable region of the germline V-gene, the alignment for this region (J-element) was done individually. Hence the IGLJ3-02 sequence was chosen for humanization of this light chain.
  • Humanized VL variants were assessed as chimera in a 2+1 classical format ( FIG. 1D ), i.e. humanized light chain V-domains were paired with murine heavy chain V-domains. SPR assessment was carried out on a ProteOn XPR36 instrument (Bio-Rad). More precisely, the variants were captured directly from the culture supernatant on an anti-Fab derivatized GLM sensorchip (Goat Anti-Human IgG, F(ab′)2 Fragment Specific, Jackson ImmunoResearch) in vertical orientation.
  • the light chain V-domain variant that was chosen for the humanized lead molecule is VL7_46-13.
  • the degree of humanness i.e. the sequence homology of the humanized V-domain to the human germline V-domain sequence was determined.
  • the overall sequence identity with the closest human germline homolog is 65% before humanization and 80% afterwards. Omitting the CDR regions, the sequence identity is 92% to the closest human germline homolog.
  • VL7_46-13 is the only humanized VL variant out of a panel of 13 variants that showed comparable binding to the parental murine antibody and also retained its cross-reactivity to cynomolgus CD3c.
  • the humanized VL-domain VL7_46-13 based on an infrequent human germline belonging to the V-domain family 7 of the lambda type and retaining affinity and specificity for CD3c, is also suitable to be used as a common light chain in phage-displayed antibody libraries of the Fab-format and enables successful selection for novel specificities which greatly facilitates the generation and production of bispecific molecules binding to CD3 ⁇ and e.g. a tumor target and sharing the same ‘common’ light chain.
  • IGHV1-46*01 (X92343) (SEQ ID NO: 104), IGHV1-69*06 (L22583), (SEQ ID NO: 105) IGHV3-15*01 (X92216), (SEQ ID NO: 106) IGHV3-23*01 (M99660), (SEQ ID NO: 107) IGHV4-59*01 (AB019438), (SEQ ID NO: 108) IGHV5-51*01 (M99686), (SEQ ID NO: 109)
  • All heavy chains use the IGHJ2 as J-element, except the IGHV1-69*06 which uses IGHJ6 sequence.
  • the design of the randomization included the CDR-H1, CDR-H2, and CDR-H3.
  • CDR-H1 and CDR-H2 a “soft” randomization strategy was chosen, and the randomization oligonucleotides were such that the codon for the amino acid of the germ-line sequence was present at 50%. All other amino acids, except cysteine, were summing up for the remaining 50%.
  • oligonucleotides were designed that allow for the usage of randomized inserts between the V-element and the J-element of 4 to 9 amino acids in length. Those oligonucleotides contained in their randomized part e.g. The three amino acids G/Y/S are present to 15% each, those amino acids A/D/T/R/P/L/V/N/W/F/I/E are present to 4,6% each.
  • the light chain is derived from the human sequence hVK1-39, and is used in an unmodified and non-randomized fashion. This will ensure that the same light chain can be used for other projects without additional modifications.
  • 10 ⁇ 12 phagemid particles of each library are bound to 100 nM biotinylated soluble antigen for 0.5 h in a total volume of 1 ml.
  • Biotinylated antigen is captured and specifically bound phage particles are isolated by addition of ⁇ 5 ⁇ 10 ⁇ 7 streptavidin-coated magnetic beads for 10 min.
  • Beads are washed using 5-10 ⁇ 1 ml PBS/Tween20 and 5-10 ⁇ 1 ml PBS.
  • Elution of phage particles is done by addition of 1 ml 100 mM TEA (triethylamine) for 10 min and neutralization by addition of 500 ul 1 M Tris/HCl pH 7.4 and 5. Re-infection of exponentially growing E.
  • coli TG1 bacteria infection with helper phage VCSM13 and subsequent PEG/NaCl precipitation of phagemid particles is applied in subsequent selection rounds. Selections are carried out over 3-5 rounds using either constant or decreasing (from 10 ⁇ 7 M to 2 ⁇ 10 ⁇ 9M) antigen concentrations.
  • capture of antigen/phage complexes is performed using neutravidin plates instead of streptavidin beads. All binding reactions are supplemented either with 100 nM bovine serum albumin, or with non-fat milk powder in order to compete for unwanted clones arising from mere sticky binding of the antibodies to the plastic support.
  • Specific binders are defined as signals ca. 5 ⁇ higher than background and are identified by ELISA. Specific binders are identified by ELISA as follows: 100 ⁇ l of 10 nM biotinylated antigen per well are coated on neutravidin plates. Fab-containing bacterial supernatants are added and binding Fabs are detected via their Flag-tags by using an anti-Flag/HRP secondary antibody.
  • ELISA-positive clones are bacterially expressed as soluble Fab fragments in 96-well format and supernatants are subjected to a kinetic screening experiment by SPR-analysis using ProteOn XPR36 (BioRad).
  • Clones expressing Fabs with the highest affinity constants are identified and the corresponding phagemids are sequenced.
  • the Fab sequences are amplified via PCR from the phagemid and cloned via appropriate restriction sites into human IgG1 expression vectors for mammalian production.
  • an antibody library for the display on M13 phage is described.
  • a multi framework library for the heavy chain with one constant (or “common”) light chain was designed for the generation of Fc-containing, but FcgR binding inactive T cell bispecific antibodies of IgG1 P329G LALA or IgG4 SPLE PG isotype in which one or two Fab recognize a tumor surface antigen expressed on a tumor cell whereas the remaining Fab arm of the antibody recognizes CD3e on a T cell.
  • an antibody library for the display on M13 phage is described.
  • This library is designed solely for the generation of Fc-containing, but FcgR binding inactive T cell bispecific antibodies of IgG1 P329G LALA or IgG4 SPLE PG isotype. Diversity was introduced via randomization oligonucleotides only in the CDR3 of the different heavy chains. Methods for generation of antibody libraries are well known in the art and are described in (Hoogenboom et al., Nucleic Acids Res. 1991, 19, 4133-413; or in: Lee et., al J. Mol. Biol. (2004) 340, 1073-1093).
  • IGHV1-46*01 (X92343), (SEQ ID NO: 104) IGHV1-69*06 (L22583), (SEQ ID NO: 105) IGHV3-15*01 (X92216), (SEQ ID NO: 106) IGHV3-23*01 (M99660), (SEQ ID NO: 107) IGHV4-59*01 (AB019438), (SEQ ID NO: 108) IGHV5-51*01 (M99686), (SEQ ID NO: 109)
  • the antibodies 16A3, 15A1, 18D3, 19E5, 19A4, 15H7, 15B6, 16D5, 15E12, 21D1, 16F12, 21A5, 21G8, 19H3, 20G6, and 20H7 comprising the common light chain VL7_46-13 with CD3 ⁇ specificity were obtained by phage display selections against different species (human, cynomolgus and murine) of FolR1.
  • Clones 16A3, 15A1, 18D3, 19E5, 19A4, 15H7, 15B6, 21D1, 16F12, 19H3, 20G6, and 20H7 were selected from a sub-library in which the common light chain was paired with a heavy chain repertoire based on the human germline VH1_46.
  • CDR3 of VH1_46 has been randomized based on 6 different CDR3 lengths.
  • Clones 16D5, 15E12, 21A5, and 21G8 were selected from a sub-library in which the common light chain was paired with a heavy chain repertoire based on the human germline VH3_15.
  • CDR3 of VH3_15 has been randomized based on 6 different CDR3 lengths.
  • Clones exhibiting signals on human FolR1 and being negative on human IgG were short-listed for further analyses and were also tested in a similar fashion against the remaining two species of FolR1. They were bacterially expressed in a 0.5 liter culture volume, affinity purified and further characterized by SPR-analysis using BioRad's ProteOn XPR36 biosensor.
  • K D Affinities (K D ) of selected clones were measured by surface plasmon resonance (SPR) using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated human, cynomolgus, and murine FolR1 as well as human FolR2 and FolR3 (negative controls) immobilized on NLC chips by neutravidin capture.
  • Immobilization of antigens (ligand) Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 ⁇ g/ml, then injected at 30 ⁇ l/minute in vertical orientation.
  • KD Equilibrium dissociation constants
  • the antibodies 11F8, 36F2, 9D11, 5D9, 6B6, and 14E4 were obtained by phage display selections based on generic multi-framework sub-libraries against different species (human, cynomolgus and murine) of FolR1.
  • different VL-domains with randomized CDR3 (3 different lengths) are paired with different VH-domains with randomized CDR3 (6 different lengths).
  • the selected clones are of the following VL/VH pairings: 11F8 (Vk_1_5/VH_1_69), 36F2 (Vk_3_20/VH_1_46), 9D11 (Vk2D_28/VH1_46), 5D9 (Vk3_20/VH1_46), 6B6 (Vk3_20/VH1_46), and 14E4 (Vk3_20/VH3_23).
  • Clones exhibiting signals on human FolR1 and being negative on human IgG were short-listed for further analyses and were also tested in a similar fashion against the remaining two species of FolR1. They were bacterially expressed in a 0.5 liter culture volume, affinity purified and further characterized by SPR-analysis using BioRad's ProteOn XPR36 biosensor.
  • K D Affinities (K D ) of selected clones were measured by surface plasmon resonance (SPR) using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated human, cynomolgus, and murine FolR1 as well as human FolR2 and FolR3 (negative controls) immobilized on NLC chips by neutravidin capture.
  • Immobilization of antigens (ligand) Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 ⁇ g/ml, then injected at 30 ⁇ l/minute in vertical orientation.
  • K D Equilibrium dissociation constants (K D ) for anti-FolR1 antibodies (Fab-format) selected by phage display from generic multi-framework sub-libraries.
  • K D (nM) Clone huFolR1 cyFolR1 muFolR1 huFolR2 huFolR3 11F8 632 794 1200 no binding no binding 36F2 1810 1640 737 no binding no binding 9D11 8.64 5.29 no binding no binding no binding 5D9 8.6 5.9 no binding no binding 6B6 14.5 9.4 no binding no binding no binding 14E4 no binding no binding 6.09 no binding no binding no binding
  • the antibodies isolated from a common light chain- or Fab-library were converted into the corresponding human IgG1 format.
  • the variable heavy and variable light chains of unique FolR1 binders from phage display were amplified by standard PCR reactions using the Fab clones as the template.
  • the PCR products were purified and inserted (either by restriction endonuclease and ligase based cloning, or by ‘recombineering’ using the InFusion kit from Invitrogen) into suitable expression vectors in which they are fused to the appropriate human constant heavy or human constant light chain.
  • the expression cassettes in these vectors consist of a chimeric MPSV promoter and a synthetic polyadenylation site.
  • the plasmids contain the oriP region from the Epstein Barr virus for the stable maintenance of the plasmids in HEK293 cells harboring the EBV nuclear antigen (EBNA).
  • EBNA EBV nuclear antigen
  • HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells are centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 ⁇ g DNA.
  • PEI solution After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added.
  • Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N 2 and stored at ⁇ 80° C.
  • binders were converted into a T-cell bispecific format.
  • the FolR1:CD3 binding moieties are arranged in a 2:1 order with the FolR1 Fabs being located at the N-terminus.
  • the CD3 binding part was generated as a CrossFab (CHICK crossing) while for the clones from the common light chain library no crossing was necessary.
  • IgG TCB Yield Monomer Yield Monomer # Clone Library [mg/L] [%] [mg/L ⁇ [%] 1 11F8 Fab 8.03 96.26 — — 2 14E4 Fab 8.90 98.12 — — 3 15B6 CLC 7.72 100.00 — — 4 15E12 CLC 6.19 100.00 — — 5 15H7 CLC 8.94 100.00 — — 6 16A3 CLC 0.60 n.d.
  • T-cell bispecific formats were prepared for one common light chain binder (16D5) and three formats for one binder from the Fab library (9D11) to compare their killing properties in vitro.
  • the standard format is the 2+1 inverted format as already described (FolR1:CD3 binding moieties arranged in a 2:1 order with the FolR1 Fabs located at the N-terminus).
  • the FolR1:CD3 binding moieties are arranged in a 2:1 order with the CD3 Fab being located at the N-terminus.
  • Two monovalent formats were also prepared.
  • the 1+1 head-to-tail has the FolR1:CD3 binding moieties arranged in a 1:1 order on the same arm of the molecule with the FolR1 Fab located at the N-terminus.
  • the FolR1:CD3 binding moieties are present once, each on one arm of the molecule.
  • the IgGs were injected for 60 seconds at a concentration of 500 nM. IgGs binding to huFolR2 and huFolR3 were rejected for lack of specificity. Most of the binders are only crossreactive between human and cyno FolR1, additional crossreactivity to murine FolR1 went most of the time hand in hand with loss of specificity.
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany).
  • the immobilization level was about 300-400 RU.
  • the anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 2.1 to 500 nM with a flow of 30 ⁇ L/minutes through the flow cells over 180 seconds. The dissociation was monitored for 600 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated IL2 receptor Fc fusion.
  • folate (Sigma F7876) was added in the HBS-EP running buffer at a concentration of 2.3 ⁇ M.
  • the binding curves resulting from the bivalent binding of the IgGs or T cell bispecifics were approximated to a 1:1 Langmuir binding and fitted with that model (which is not correct, but gives an idea of the avidity).
  • the apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).
  • the dissociation phase was monitored for 240 s and triggered by switching from the sample solution to HBS-EP.
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1 or pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • the CD3 binding part is identical for all constructs and the affinity is similar for the measured T cell bispecifics (K D range between 60 and 90 nM).
  • the anti-FolR1 T cell bispecifics were injected for 60 s at 500 nM with a flow of 30 ⁇ L/minutes through the flow cells, followed by an injection of hu CD ⁇ -Fc for 60 s at 500 nM. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated IL2 receptor Fc fusion.
  • the four T cell bispecifics tested (16D5 TCB, 21A5 TCB, 51C7 TCB and 45D2 TCB) were able to bind simultaneously to Folate Receptor 1 and human CD3 as expected.
  • the anti-FolR1 IgGs or T cell bispecifics were directly immobilized on a CMS chip at pH 5.0 using the standard amine coupling kit (GE Healthcare), with a final response around 700 RU.
  • 500 nM huFolR1-Fc was then captured for 60 s, followed by 500 nM of the different binders for 30 s.
  • the surface was regenerated with two injections of 10 mM glycine pH 2 for 30 s each. It is assessed if the different binders can bind to huFolR1 captured on immobilized binders (Table 12).
  • FolR1 binders in the IgG formats were screened by surface plasmon resonance (SPR) and by in vitro assay on cells to select the best candidates.
  • the anti-FolR1 IgGs were analyzed by SPR to characterize their crossreactivity (to human, murine and cynomolgus FolR1) and specificity (to human FolR1, human FolR2, human FolR3). Unspecific binding to human FolR2 and 3 was considered an exclusion factor. Binding and specificity to human FolR1 was confirmed on cells. Some binders did not bind on cells expressing FolR1 even though they recognized the recombinant human FolR1 in SPR. Aggregation temperature was determined but was not an exclusion factor because the selected binders were all stable. Selected binders were tested in a polyreactivity ELISA to check for unspecific binding, which led to the exclusion of four binders.
  • New FolR1 binders were generated via Phage Display using either a Fab library or a common light chain library using the CD3 light chain. The identified binders were converted into a human IgG1 format and binding to FolR1 high expressing HeLa cells was addressed. As reference molecule the human FolR1 binder Mov19 was included. Most of the binders tested in this assay showed intermediate to good binding to FolR1 with some clones binding equally well as Mov19 (see FIG. 2 ). The clones 16A3, 18D3, 15H7, 15B6, 21D1, 14E4 and 16F12 were excluded because binding to FolR1 on cells could not be confirmed by flow cytometry.
  • HEK cells were transiently transfected with either human FolR1 or human FolR2 to address specificity.
  • the clones 36F2 and 9D11 derived from the Fab library and the clones 16D5 and 21A5 derived from the CLC library bind specifically to human FolR1 and not to human FolR2 (see FIGS. 3A-B ). All the other tested clones showed at least some binding to human FolR2 (see FIGS. 3A-B ). Therefore these clones were excluded from further characterization.
  • Mov19 FolR1 TCB was included in all studies. These FolR1 TCBs were then used to compare induction of internalization after binding to FolR1 on HeLa cells. All three tested clones are internalized upon binding to FolR1 comparable to internalization upon binding of Mov19 FoLR1 TCB ( FIG. 5 ). 21A5 FolR1 TCB was discontinued due to signs of polyreactivity.
  • the FolR1 TCBs were used to determine T cell mediated killing of tumor cells expressing FoLR1.
  • a panel of potential target cell lines was used to determine FoLR1 binding sites by Qifikit analysis.
  • the used panel of tumor cells contains FolR1 high, intermediate and low expressing tumor cells and a FolR1 negative cell line.
  • Binding of the three different FoLR1 TCBs (containing 9D11, 16D5 and Mov19 binders) to this panel of tumor cell lines was determined showing that the FoLR1 TCBs bind specifically to FolR1 expressing tumor cells and not to a FoLR1 negative tumor cell line.
  • the amount of bound construct is proportional to the FolR1 expression level and there is still good binding of the constructs to the FolR1 low cell line HT-29 detectable.
  • the intermediate expressing cell line SKOV3 and the low expressing cell line HT-29 were further on used to test T cell mediated killing and T cell activation using 16D5 TCB and 9D11 TCB; DP47 TCB was included as negative control. Both cell lines were killed in the presence of already very low levels of 16D5 TCB and 9D11 TCB and there was no difference in activity between both TCBs even though 9D11 TCB binds stronger to FolR1 than 16D5 TCB. Overall killing of SKOV3 cells was higher compared to HT-29 which reflects the higher expression levels of FolR1 on SKOV3 cells ( FIGS. 7A-D ).
  • the four T cell bispecifics were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below.
  • HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium.
  • 400 million HEK293 EBNA cells were seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly).
  • For transfection cells were centrifuged for 5 min by 210 ⁇ g, supernatant was replaced by pre-warmed 20 ml CD CHO medium.
  • Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200m DNA.
  • PEI solution After addition of 540 ⁇ l PEI solution was vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added.
  • Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 2 M Tris pH 8.0). Samples were concentrated to 1 ml using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a SuperdexTM 200 10/300 GL (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography.
  • Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N 2 and stored at ⁇ 80° C.
  • N100S N95S
  • N100Q N95Q
  • T102A T97A
  • T102N T97N
  • FolR1 is expressed at low levels on primary epithelial cells. Here we wanted to test if these levels are sufficient to induce T cell mediated killing in the presence of the FolR1 TCBs. To test this we used primary human bronchial epithelial cells, primary human choroid plexus epithelial cell, primary human renal cortical epithelial cells and primary human retinal pigment epithelial cells. As positive control either FolR1 positive SKOV3 cells or HT-29 cells were included. First we verified FolR1 expression on the used primary cells and determined the amount of FolR1 binding sites on these cells.
  • Bronchial epithelial cells, renal cortical epithelial cells and retinal pigment epithelial cells express very low but significant levels of FolR1 compared to the levels expressed on tumor cells.
  • the choroid plexus epithelial cells do not express significant levels of FolR1.
  • the primary epithelial cells that demonstrated FolR1 expression on the surface were used to address the question if these cells can be killed by T cells in the presence of FoLR1 TCBs. No significant levels of killing could be measured but induction of T cell activation in the presence of retinal pigment epithelial cells, bronchial epithelial cells and renal cortical cells resulting in upregulation of CD25 and CD69 was detected. The strongest activation is seen with retinal pigment epithelial cells resulting in upregulation of CD25 and CD69 both on CD4 + T cells and CD8 + T cells.
  • T cells In the presence of bronchial epithelial cells lower activation of T cells is induced with upregulation of CD69 on CD4 + T cells and CD8 + T cells but very low upregulation of CD25 only on CD4 + T cells but not on CD8 + T cells.
  • the lowest activation of T cells is obtained in the presence of renal epithelial cells with no upregulation of CD25 on CD4 T ⁇ cells and CD8 + T cells and CD69 been only upregulated on CD8 + T cells ( FIGS. 12A-X ).
  • TCB 2+1 inverted format is the most active format with the selected FolR1 binder
  • different formats containing either 16D5 or 9D11 were produced and compared in target cell binding, T cell mediated killing and T cell activation.
  • the 16D5 binder was tested in the TCB 2+1 inverted ( FIG. 1A ), TCB 2+1 classical ( FIG. 1D ), TCB 1+1 classical ( FIG. 1C ) and TCB 1+1 head-to-tail ( FIG. 1B ) format
  • the 9D11 binder was tested in the TCB 2+1 inverted ( FIG. 1A ), TCB 1+1 classical ( FIG. 1C ) and TCB 1+1 head-to-tail ( FIG. 1B ) format.
  • HeLa cells were obtained from ATCC and cultured in DMEM with 10% FCS and 2 mM Glutamine
  • SKOV3 HBV-77
  • OVCAR5 were obtained from NCI and cultured in RPMI with 10% FCS and 2 mM Glutamine
  • HT-29 ACC-299
  • MKN-45 ACC-409 were obtained from DSMZ and cultured in RPMI with 10% FCS and 2 mM Glutamine.
  • HRCEpiC Human Renal Cortical Epithelial Cells (HRCEpiC), Catalog Number 4110, were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell).
  • HRCEpiC Human Choroid Plexus Epithelial Cells (HCPEpiC), Catalog Number 1310 were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell).
  • Target cells as indicated were harvested with Cell Dissociation Buffer, washed with PBS and resuspended in FACS buffer. The antibody staining was performed in a 96 well round bottom plate. Therefore 200′000 cells were seeded per well. The plate was centrifuged for 4 min at 400 g and the supernatant was removed. The test antibodies were diluted in FACS buffer and 20 ⁇ l of the antibody solution were added to the cells for 30 min at 4° C.
  • the cells were washed twice with FACS buffer before addition of the diluted secondary antibody (FITC conjugated AffiniPure F(ab′)2 fragment goat anti-human IgG, Fcg Fragment, Jackson ImmunoResearch #109-096-098 or PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific, Jackson ImmunoResearch #109-116-170. After 30 min incubation on 4° C. unbound secondary antibody was washed away. Before measurement the cells were resuspended in 200 ⁇ l FACS buffer and analyzed by flow cytometry using BD Canto II or BD Fortessa.
  • the cells were harvested and the viability was determined.
  • the cells were re-suspended in fresh cold medium at 2 Mio cells per ml and the cell suspension was transferred in a 15 ml falcon tube for each antibody.
  • the antibodies that should be tested for internalization were added with a final concentration of 20 ⁇ g per ml to the cells.
  • the tubes were incubated for 45 min in the cold room on a shaker. After incubation the cells were washed three times with cold PBS to remove unbound antibodies. 0.2 Mio cells per well were transfer to the FACS plate as time point zero.
  • the labeled cells were re-suspended in warm medium and incubated at 37° C.
  • Mio cells per well were transferred in cold PBS, washed in plated on the FACS plate. To detect the constructs that remain on the surface the cells were stained with PE-labeled anti-human Fc secondary antibody. Therefore 20 ⁇ l of the diluted antibody were added per well and the plate was incubated for 30 min at 4° C. Then the cells were washed twice to remove unbound antibodies and then fixed with 1% PFA to prevent any further internalization. The fluorescence was measured using BD FACS CantoII.
  • QIFIKIT® contains a series of beads, 10 ⁇ m in diameter and coated with different, but well-defined quantities of mouse Mab molecules (high-affinity anti-human CD5, Clone CRIS-1, isotype IgG 2 a).
  • the beads mimic cells with different antigen densities which have been labeled with a primary mouse Mab, isotype IgG. Briefly, cells were labeled with primary mouse monoclonal antibody directed against the antigen of interest. In a separate test well, cells were labeled with irrelevant mouse monoclonal antibody (isotype control). Then, cells, Set-Up Beads and Calibration Beads were labeled with a fluorescein-conjugated anti-mouse secondary antibody included in the kit.
  • the primary antibody used for labeling of the cells has to be used at saturating concentration.
  • the primary antibody may be of any mouse IgG isotype. Under these conditions, the number of bound primary antibody molecules corresponds to the number of antigenic sites present on the cell surface.
  • the secondary antibody is also used at saturating concentration. Consequently, the fluorescence is correlated with the number of bound primary antibody molecules on the cells and on the beads.
  • Target cells were harvested with Trypsin/EDTA, counted and viability was checked. The cells were resuspended in their respective medium with a final concentration of 300′000 cells per ml. Then 100 ⁇ l of the target cell suspension was transferred into each well of a 96-flat bottom plate. The plate was incubated overnight at 37° C. in the incubator to allow adherence of the cells to the plate. On the next day PBMCs were isolated from whole blood from healthy donors. The blood was diluted 2:1 with PBS and overlayed on 15 ml Histopaque-1077 (#10771, Sigma-Aldrich) in Leucosep tubes and centrifuged for 30 min at 450 g without break.
  • the band containing the cells was collected with a 10 ml pipette and transferred into 50 ml tubes.
  • the tubes were filled up with PBS until 50 ml and centrifuged (400 g, 10 min, room temperature). The supernatant was removed and the pellet resuspended in PBS. After centrifugation (300 g, 10 min, room temperature), supernatants were discarded, 2 tubes were pooled and the washing step was repeated (this time centrifugation 350 ⁇ g, 10 min, room temperature). Afterwards the cells were resuspended and the pellets pooled in 50 ml PBS for cell counting.
  • Activation marker upregulation on T cells after tumor cell killing was measured by flow cytometry. Briefly PBMCs were harvested, transferred into a 96 well round bottom plate and stained with CD4 PE-Cy7 (#3557852, BD Bioscience), CD8 FITC (#555634, BD Bioscience), CD25 APC (#555434, BD Bioscience), CD69 PE (#310906, BioLegend) antibodies diluted in FACS buffer. After 30 min incubation at 4° C. the cells were washed twice with FACS buffer. Before measuring the fluorescence using BD Canto II the cells were resuspended in 200 ⁇ l FACS buffer.
  • 280 ⁇ l of fresh blood were added into a 96 well conical deep well plate. Then 20 ⁇ l of the diluted TCBs were added to the blood and mixed well by shaking the plate. After 24 h incubation at 37° C. in an incubator the blood was mixed and 35 ⁇ l were transferred to a 96 well round bottom plate. Then 20 ⁇ l of the antibody staining mix were added consisting of CD4 PE-Cy7 (#3557852, BD Bioscience), CD8 FITC (#555634, BD Bioscience), CD25 APC (#555434, BD Bioscience), CD69 PE (#310906, BioLegend) and CD45 V500 (#560777, BD Horizon) and incubated for 15 min in the dark at room temperature. Before measuring 200 ⁇ l of the freshly prepared BD FACS lysing solution (#349202, BD FCAS) was added to the blood. After 15 min incubation at room temperature the cells were measured with BD Fortessa.
  • mice Female NOD/Shi-scid/IL-2R ⁇ null (NOG) mice, age 6-7 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (P 2011/128). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.
  • mice were injected i.v. with 10/1/0.1 ⁇ g/mouse of the FOLR1 TCB whereas 3 mice were bled per group and time point. All mice were injected with a total volume of 200 ⁇ l of the appropriate solution. To obtain the proper amount of the FOLR1 TCB per 200 ⁇ l, the stock solutions were diluted with PBS when necessary. Serum samples were collected 5 min, 1 h, 3 h, 8 h, 24 h, 48 h, 72 h, 96 h and 168 h after therapy injection.
  • FIG. 15 shows that the 16D5 FOLR1 TCB shows typical and dose proportional IgG-like PK properties in NOG mice with slow clearance.
  • the FOLR1 TCB was tested in the human ovarian carcinoma cell line Skov3, injected s.c. into PBMC engrafted NOG mice.
  • the Skov3 ovarian carcinoma cells were obtained from ATCC (HTB-77).
  • the tumor cell line was cultured in RPMI containing 10% FCS (Gibco) at 37° C. in a water-saturated atmosphere at 5% CO 2 .
  • Passage 35 was used for transplantation, at a viability >95%.
  • 5 ⁇ 10 6 cells per animal were injected s.c. into the right flank of the animals in a total of 100 ⁇ l of RPMI cell culture medium (Gibco).
  • mice Female NOD/Shi-scid/IL-2R ⁇ null (NOG) mice, age 6-7 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (P 2011/128). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.
  • mice were injected s.c. on study day 0 with 5 ⁇ 10 6 of the Skov3.
  • human PBMC of a healthy donor were isolated via the Ficoll method and 10 ⁇ 10 6 cells were injected i.p. into the tumor-bearing mice.
  • the mice in the vehicle group were injected with PBS.
  • the stock solutions were diluted with PBS when necessary. Tumor growth was measured once weekly using a caliper ( FIG. 17 ) and tumor volume was calculated as followed:
  • mice per treatment group were sacrificed at study day 32, tumors were removed and single cell suspensions were prepared through an enzymatic digestion with Collagenase V, Dispase II and DNAse for subsequent FACS-analysis ( FIGS. 19 and 20 ).
  • Single cells where either used directly for staining of extracellular antigens and activation markers or were re-stimulated using 5 ng/ml PMA and 500 ng/ml lonomycin in the presence of a protein transport inhibitor Monensin for 5h in normal culture medium. After re-stimulation, cells were stained for surface antigens, followed by a fixation and permeabilization step.
  • TNF- ⁇ , IFN- ⁇ and IL-2 producing as well as perforin + /granzym-B + CD4 + and CD8 + T-cells were detected in FOLR1 TCB treated tumors.
  • Tumor infiltrating T-cells treated with FOLR1 TCB also showed higher degranulation rates compared to control groups.
  • a bispecific antibody (monovalent for each antigen) that simultaneously can bind to human CD3 and human folate receptor alpha (FolR1) without using any hetero-dimerization approach (e.g. knob-into-hole technology)
  • a combination of a common light chain library with the so-called CrossMab technology was applied:
  • the variable region of the humanized CD3 binder (CH2527_VL7_46/13) was fused to the CH1 domain of a standard human IgG1 antibody to form the VLVH crossed molecule (fused to Fc) which is common for both specificities.
  • VHCL a CD3 specific variable heavy chain domain (CH2527_VH_23/12) was fused to a constant human ⁇ light chain whereas a variable heavy chain domain specific for human FolR1 (clone 16D5, isolated from common light chain library) was fused to a constant human ⁇ light chain.
  • This enables the purification of the desired bispecific antibody by applying subsequent purification steps with KappaSelect and LambdaFabSelect columns (GE Healthcare) to remove undesired homodimeric antibodies.
  • HEK293 EBNA cells were transfected with plasmids encoding the respective genes using a standard polyethlenimine (PEI) based method.
  • PEI polyethlenimine
  • the used plasmid ratio of the three expression vectors was 1:1:1.
  • Transfected cells were cultivated for 7 days before supernatants were harvested for purification.
  • the bispecific FolR1/CD3-kappa-lambda antibodies were produced and purified as follows.
  • HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells were centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200m DNA.
  • PEI solution After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added.
  • the kappa-lambda bispecific antibody was purified in three steps, using an affinity step specific for kappa light chains, followed by an affinity step specific for lambda light chains and finally by a size exclusion chromatography step for removal of aggregates.
  • the protein was eluted using a pH gradient to buffer B (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0) over 25 cv.
  • buffer B 50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0
  • Fractions containing the protein of interest were pooled and the pH of the solution was adjusted to pH 8.0 (using 2 M Tris pH 8.0).
  • the protein was eluted using a pH gradient to buffer B (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0) over 25 cv.
  • buffer B 50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0
  • Fractions containing the protein of interest were pooled and the pH of the solution was adjusted to pH 8.0 (using 2 M Tris pH 8.0).
  • This solution was concentrated using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a SuperdexTM 200 10/300 GL (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20.
  • the pooled fractions after size exclusion were again concentrated using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius).
  • the protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Only small amounts of protein could be purified with a final yield of 0.17 mg/L.
  • T cell mediated killing of SKOV3 cells was determined after 24 h and 48 h by LDH release. After 48 h the T cells were harvested and CD69 and CD25 upregulation on CD4 T cells and CD8 T cells was measured by flow cytometry.
  • the kappa lambda FolR1 construct induces killing of SKOV3 cells in a concentration dependent manner which is accompanied by CD69 and CD25 upregulation both on CD4 T cells and on CD8 T cells.
  • SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 24 h and 48 h killing of tumor cells was determined by measuring LDH release ( FIG. 21 ). SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 48 h CD25 and CD69 upregulation on CD4 T cells and CD8 T cells was measured by flow cytometry ( FIG. 22 ).
  • Binding of anti-FolR1 16D5 in different monovalent or bivalent T-cell bispecific formats and of anti-FolR1 36F2 as IgG or as T-cell bispecific to recombinant human, cynomolgus and murine folate receptor 1 (all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, GE Healthcare).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, GE Healthcare).
  • the immobilization level was about 300-400 RU.
  • the anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 3.7 to 900 nM with a flow of 30 ⁇ L/minutes through the flow cells over 180 seconds. The dissociation was monitored for 240 or 600 seconds.
  • the chip surface was regenerated after every cycle using a double injection of 30 sec 10 mM Glycine-HCl pH 2.
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • the affinity (monovalent binding) to human and cyno FolR1-Fc of 36F2 TCB is similar and around 1000 nM for both, whereas the affinity to murine FolR1-Fc is slightly better and around 300 nM.
  • the 36F2 can be used in murine and primate models, there is no need for a surrogate.
  • the avidity (apparent K D ) of 36F2 TCB to human FolR1 is around 30 times lower than the affinity of the 16D5 TCB to human FolR1.
  • 36F2 TCB is in the low nanomolar range
  • 16D5 TCB is in the low picomolar range (1000 fold difference).
  • FolR1 is expressed on tumor cells overexpressed, at intermittent and high levels, on the surface of cancer cells in a spectrum of epithelial malignancies, including ovarian, breast, renal, colorectal, lung and other solid cancers and is also expressed on the apical surface of a limited subset of polarized epithelial cells in normal tissue.
  • These non-tumorous, normal cells express FolR1 only at low levels, and include, e.g., bronchiolal epithelial cells on alveolar surface, renal cortical luminal border of tubular cells, retinal pigment epithelium (basolateral membrane) and choroid plexus.
  • 16D5 TCB binds to normal tissues cells expressing low amounts of FolR1 which results in their T cell mediated killing. This might, at least in part, account for limited tolerance observed at 10 ⁇ g/kg in cynomolgus monkeys.
  • the inventors wanted to determine if lowering the affinity of the T cell bispecific molecule could increase the differentiation between high and low target density tissues and, thereby, lower toxicity by making use of bivalent binding and avidity.
  • Low affinity binders are ordinarily not selected as suitable candidates for further analysis because low affinity is often associated with low potency and efficacy. Nevertheless, the low affinity FolR1 binder 36F2 was developed in several formats and characterized for its biological properties.
  • the avidity effect (difference between monovalent and bivalent binding) is around 250 fold (1000 nM versus 4 nM).
  • affinity defined the interaction and with 1000 nM led to a low potency of the TCB.
  • molecule's avidity comes into play and with 4 nM led to a high activity of the TCB (see Example 32).
  • the inventors generated monovalent formats of 16D5 and low affinity variant of 16D5 (affinity about 10-40 nM) in a bivalent format.
  • the 16D5 binder used in a monovalent format (1+1) has an affinity of about 50 nM.
  • the differentiation between high and low target density tissues can be better achieved by taking advantage of the avidity effect.
  • PBMCs peripheral blood mononuclear cells
  • Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450 ⁇ g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400 ⁇ g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350 ⁇ g, 10 minutes).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h).
  • the antibody was added at the indicated concentrations (range of 0.005 pM-5 nM in triplicates).
  • PBMCs were added to target cells at final E:T ratio of 10:1.
  • DP47 TCB antibody was included as negative control. Human PBMCs were used as effectors and the killing was detected at 24 h of incubation with the bispecific antibody.
  • PBMCs Peripheral blood mononuclear cells
  • Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450 ⁇ g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS.
  • the mixture was centrifuged (400 ⁇ g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350 ⁇ g, 10 minutes).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h).
  • the antibody was added at the indicated concentrations (range of 0.01 pM-100 nM in triplicates).
  • PBMCs were added to target cells at final E:T ratio of 10:1.
  • this data shows that using the 36F2 FolR1 binder in the bivalent 2+1 TCB format widens the therapeutic window compared to the 16D5 FOLR1 TCB ( FIG. 24A-C ).
  • the 36F2 FOLR1 TCB differentiates between high and low expressing cells which is of special importance to reduce toxicity as the cells of some normal, non-tumorous tissues express very low levels of FolR1 (approximately less than 1000 copies per cell). Consistent with this observation, the results discussed in Example 35 below show that 36F2 TCB does not induce T-cell killing of primary cells ( FIGS. 26A-D ) whereas for 16D5 TCB some killing can be observed on HRCEpiC and HRPEpiC cells after 48 h of incubation ( FIGS. 26B and C).
  • 36F2 TCB This important characteristic of 36F2 TCB allows for dosing for the treatment of FolR1-positive tumors so that it mediates potent killing of tumor tissues with high or medium FOLR1 expression, but not of normal tissues with low (partially polarized) expression. Notably, this characteristic appears to be mediated by the avidity of 36F2 TCB in the bivalent 2+1 inverted format, as it was not observed when using the 1+1 monovalent formats carrying the same low affinity 36F2 binder.
  • 36F2 TCB in the bivalent 2+1 format comprises FolR1 binding moieties of relatively low affinity but it possesses an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this TCB selectively binds to tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all.
  • the 36F2 TCB in the bivalent 2+1 inverted format also has the advantage that it does not require chemical cross linking or other hybrid approach. This makes it suitable for manufacture of a medicament to treat patients, for example patients having FolR1-positive cancerous tumors.
  • the 36F2 TCB in the bivalent 2+1 inverted format can be produced using standard CHO processes with low aggregates.
  • the 36F2 TCB in the bivalent 2+1 comprises human and humanized sequences making it superior to molecules that employ rat and murine polypeptides that are highly immunogenic when administered to humans.
  • the 36F2 TCB in the bivalent 2+1 format was engineered to abolish FcgR binding and, as such, does not cause FcgR crosslinking and infusion reactions, further enhancing its safety when administered to patients.
  • a further advantage of the 36F2 TCB in the bivalent 2+1 format and other embodiments disclosed herein is that their clinical development does not require the use of surrogate molecules as they bind to human, cynomous and murine FolR1. As such, the molecules disclosed herein recognize a different epitope than antibodies to FolR1 previously described that do not recognize FolR1 from all three species.
  • Table 29 shows a comparison of EC50 values of 16D5 TCB and 36F2 TCB on the different cell lines tested. Out of the obtained EC50 values the delta (EC50 of 16D5 TCB minus EC50 of 36F2 TCB) and the x-fold difference (EC50 of 16D5 TCB divided by the EC50 of 36F2 TCB) was calculated.
  • Tables 30 and 31 show the comparisons of the EC50 values of 16D5 TCB vs 16D5 TCB HT (Table 30) and 16D5 TCB vs 16D5 TCB 1+1 (Table 31) as well as the corresponding deltas (EC50 of 16D5 TCB minus EC50 of 16D5 TCB HT/1+1) and the x-fold differences (EC50 of 16D5 TCB divided by the EC50 of 16D5 TCB HT/1+1).
  • Activation of CD8 + and CD4 + T cells after T-cell killing of FolR1-expressing Hela, SKov-3 and HT-29 tumor cells mediated by 36F2 TCB and 16D5 TCB was assessed by FACS analysis using antibodies recognizing the T cell activation markers CD25 (late activation marker) and CD69 (early activation marker).
  • DP47 TCB was included as non-binding control.
  • the antibody and the killing assay conditions were essentially as described above (Example 32) using the same antibody concentration range (0.01 pM-100 nM in triplicates), E:T ratio 10:1 and an incubation time of 48h.
  • PBMCs were transferred to a round-bottom 96-well plate, centrifuged at 400 ⁇ g for 4 min and washed twice with PBS containing 0.1% BSA.
  • T-cell killing mediated by 36F2 TCB and 16D5 TCB was assessed on primary cells (Human Renal Cortical Epithelial Cells (HRCEpiC) (ScienCell Research Laboratories; Cat No 4110) and Human Retinal Pigment Epithelial Cells (HRPEpiC) (ScienCell Research Laboratories; Cat No 6540)).
  • HRCEpiC Human Renal Cortical Epithelial Cells
  • HRPEpiC Human Retinal Pigment Epithelial Cells
  • HT-29 cells low FolR1
  • DP47 TCB served as non-binding control.
  • Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies.
  • PBMCs Peripheral blood mononuclear cells
  • Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450 ⁇ g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS.
  • the mixture was centrifuged (400 ⁇ g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350 ⁇ g, 10 minutes).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h).
  • the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates).
  • PBMCs were added to target cells at final E:T ratio of 10:1.
  • the “untargeted TCB” was used as a control in the above experiments.
  • the bispecific antibody engages CD3e but does not bind to any other antigen and therefore cannot crosslink T cells to any target cells (and subsequently cannot induce any killing). It was therefore used as negative control in the assays to monitor any unspecific T cell activation.
  • This untargeted TCB was prepared as described in WO2014/131712. In brief, the variable region of heavy and light chain DNA sequences have been subcloned in frame with either the constant heavy chain or the constant light chain pre-inserted into the respective recipient mammalian expression vector. The antibody expression was driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS. In addition each vector contains an EBV OriP sequence.
  • the molecule was produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using polyethylenimine. The cells were transfected with the corresponding expression vectors in a 1:2:1:1 ratio (“vector heavy chain Fc(hole)”:“vector light chain”:“vector light chain Crossfab”:“vector heavy chain Fc(knob)-FabCrossfab”).
  • HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection. For transfection cells were centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200 g DNA. After addition of 540 ⁇ l PEI solution was vortexed for 15 s and subsequently incubated for 10 min at room temperature.
  • cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210 ⁇ g, the solution was sterile filtered (0.22 ⁇ m filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C.
  • the secreted protein was purified from cell culture supernatants by affinity chromatography using ProteinA.
  • Target protein was eluted during a gradient over 20 column volume from 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5 to 20 mM sodium citrate, 0.5 M sodium chloride, pH 2.5.
  • Protein solution was neutralized by adding 1/10 of 0.5 M sodium phosphate, pH 8.
  • Target protein was concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM Histidine, 140 mM sodium chloride solution of pH 6.0.
  • the protein concentration of purified protein samples was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence.
  • the aggregate content of antibody samples was analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-Arginine Monohydrocloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C.
  • the binding of 16D5 TCB and the corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and the demidation variants 9D11 TCB N100A and 9D11 TCB S100aA to human CD3 was assessed on a CD3-expressing immortalized T lymphocyte line (Jurkat). Briefly, cells were harvested, counted, checked for viability and resuspended at 2 ⁇ 10 6 cells/ml in FACS buffer (100 ⁇ l PBS 0.1% BSA). 100 ⁇ l of cell suspension (containing 0.2 ⁇ 10 6 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C.
  • results show reduced binding of the deamidation variants N100A and S100aA to CD3 compared to the parental antibodies 16D5 TCB ( FIG. 28A ) and 9D11 TCB ( FIG. 28B ).
  • T-cell killing mediated by 16D5 TCB and the corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and the demidation variants 9D11 TCB N100A and 9D11 TCB S100aA was assessed on SKov-3 (medium FolR1) and HT-29 (low FolR1) cells.
  • Human PBMCs were used as effectors and the killing was detected at 24 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at a density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight.
  • PBMCs Peripheral blood mononuclear cells
  • enriched lymphocyte preparations obtained from healthy human donors.
  • Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450 ⁇ g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400 ⁇ g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350 ⁇ g, 10 minutes).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h).
  • the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1.
  • Binding of two 16D5 TCBs with CD3 binder variants (N100A or S100aA) to human recombinant CD3 (CD3epsilon-CD3delta heterodimer as Fc fusion) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • the two CD3 deamidation variants have a slightly reduced affinity compared to the wild-type CD3 binder (CH2527), but the difference is not grave.
  • the two deamidation variants 16D5 TCBs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below.
  • HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium.
  • 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly).
  • For transfection cells are centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium.
  • Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200m DNA.
  • PEI solution After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added.
  • the two deamidation variants 16D5 TCBs were purified in two steps using standard procedures, such as protein A affinity purification ( ⁇ kta Explorer) and size exclusion chromatography.
  • the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine) over 20 cv.
  • buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine)
  • Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 0.5 M Na 2 HPO4 pH 8.0).
  • Samples were concentrated to 1 ml using ultra-concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore) and subsequently applied to a HiLoadTM 16/60 SuperdexTM 200 preparative grade (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl.
  • the aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 ⁇ l of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled. The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence.
  • OD optical density
  • Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at ⁇ 80° C.
  • Both TCBs were produced in good quality, similar to the construct with the wild-type CD3 binder.
  • HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium.
  • HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly).
  • transfection cells are centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium.
  • Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 ⁇ g DNA.
  • PEI solution After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added.
  • the two IgGs were purified in two steps using standard procedures, such as protein A affinity purification ( ⁇ kta Explorer) and size exclusion chromatography.
  • the 5 ml containing the protein of interest are stored in a loop on the Akta Explorer and subsequently applied to a HiLoad 16/60 SuperdexTM 200 (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl (no TWEEn was used).
  • Fractions containing the IgGs were pooled and concentrated using ultra concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore). The aggregate content of the final pool was analyzed by analytical size exclusion chromatography.
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany).
  • the immobilization level was about 160.
  • the anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 3.7 to 900 nM with a flow of 30 ⁇ L/minutes through the flow cells over 180 seconds. The dissociation was monitored for 600 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine IL2 receptor Fc fusion.
  • the binding curves resulting from the bivalent binding of the IgG or T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding.
  • the apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • the two 16D5 hot spot variants have similar avidity (bivalent binding) than the wild-type 16D5 binder.
  • the avidity is slightly decreased for the D52dQ variant and this difference is even more visible in affinity (monovalent binding).
  • the twelve IgGs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below.
  • HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium.
  • 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly).
  • For transfection cells are centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium.
  • Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 ⁇ g DNA.
  • PEI solution After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added.
  • the 5 ml containing the protein of interest are stored in a loop on the ⁇ kta Explorer and subsequently applied to a HiLoad 16/60 SuperdexTM 200 (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20.
  • Fractions containing the IgGs were pooled and concentrated using ultra concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore). The aggregate content of the final pool was analyzed by analytical size exclusion chromatography.
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany).
  • the immobilization level was about 300.
  • the anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 11.1 to 900 nM with a flow of 30 ⁇ L/minutes through the flow cells over 180 seconds. The dissociation was monitored for 240 or 600 seconds.
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • 16D5 T-Cell Bispecific with Mutations to Reduce the Affinity to Human and Cynomolgus FolR1 16D5 TCB G49S/S93A, G49S/K53A, W96Y, W96Y/D52E
  • HEK293 EBNA cells Four additional variants of 16D5 TCBs having reduced affinity to FolR1 were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below.
  • HEK293 EBNA cells are cultivated in suspension serum free in Excell culture medium containing 6 mM L-Glutamine and 250 mg/l G418 culture medium.
  • 600 ml tubespin flask maximum. working volume 400 mL
  • 600 million HEK293 EBNA cells are seeded 24 hours before transfection.
  • transfection cells are centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium.
  • Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 400 ⁇ g DNA. After addition of 1080 ⁇ l PEI solution (2.7 ⁇ g/ml) is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 600 ml tubespin flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 360 ml Excell+6 mM L-Glutamine+5 g/L Pepsoy+1.0 mM VPA medium is added and cells are cultivated for 24 hours. One day after transfection 7% Feed 7 is added.
  • the reduced affinity variants 16D5 TCBs were purified in two steps using standard procedures, such as protein A affinity purification ( ⁇ kta Explorer) and size exclusion chromatography.
  • Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at ⁇ 80° C.
  • the intermediate affinity TCBs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below.
  • HEK293 EBNA cells are cultivated in suspension serum free in Excell culture medium containing 6 mM L-Glutamine and 250 mg/l G418 culture medium.
  • 600 ml tubespin flask max. working volume 400 mL
  • 600 million HEK293 EBNA cells are seeded 24 hours before transfection.
  • For transfection cells are centrifuged for 5 min by 210 ⁇ g, supernatant is replaced by pre-warmed 20 ml CD CHO medium.
  • Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 400 ⁇ g DNA. After addition of 1080 ⁇ l PEI solution (2.7 ⁇ g/ml) is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 600 ml tubespin flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 360 ml Excell+6 mM L-Glutamine+5 g/L Pepsoy+1.0 mM VPA medium is added and cells are cultivated for 24 hours. One day after transfection 7% Feed 7 is added.
  • the intermediate affinity TCBs were purified in two steps using standard procedures, such as protein A affinity purification ( ⁇ kta Explorer) and size exclusion chromatography.
  • Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N 2 and stored at ⁇ 80° C.
  • FIG. 34A-E The binding of 36F2 TCB, 16D5 TCB and various HC/LC variants ( FIG. 34A-E ) of 16D5 to human FolR1 was assessed on HT-29 cells. Briefly, cells were harvested, counted, checked for viability and resuspended at 2 ⁇ 10 6 cells/ml in FACS buffer (100 ⁇ l PBS 0.1% BSA). 100 ⁇ l of cell suspension (containing 0.2 ⁇ 10 6 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with different concentrations of the bispecific antibodies (229 pM-500 nM). After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C.
  • FACS buffer 100 ⁇ l PBS 0.1% BSA. 100 ⁇ l of cell suspension (containing 0.2 ⁇ 10 6 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with 100 nM of the bispecific antibodies. After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a Fluorescein (FITC) AffiniPure F(ab′) 2 Fragment Goat Anti-Human IgG, Fc ⁇ Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-096-098).
  • FITC Fluorescein
  • Binding of anti-FolR1 16D5 reduced affinity variants and additional intermediate affinity binders in the bivalent T-cell bispecific format to recombinant human, cynomolgus and murine folate receptor 1 (all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, GE Healthcare). The molecules used for affinity and avidity determination are described in Table 45.
  • FIG. reference 16D5 reduced affinity variants 2 + 1 T-cell bispecific
  • FIG. 1A 16D5 TCB inverted format 16D5 G49S/S93A TCB (common light chain) 16D5 G49S/K53A TCB 16D5 W96Y TCB 16D5 W96Y/D52E TCB
  • FIG. 1F 36F2 TCB inverted format, 6E10 TCB crossfab 14B1 TCB 9C7 TCB
  • the anti-FolR1 TCBs were analyzed by single injections (Table 46) to characterize their crossreactivity (to human, murine and cyno FolR1) and specificity (to human FolR1, human FolR2, human FolR3).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) or human Folate Receptor 2 and 3 (FolR2-Fc, FolR3-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU.
  • the TCBs were injected for 60 seconds at a concentration of 500 nM.
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, GE Healthcare).
  • the immobilization level was about 200-300 RU.
  • the anti-FolR1 T cell bispecifics were passed at a concentration range from 11.1 to 900 nM (for the 16D5 reduced affinity variants) or 0.2 to 500 nM (for the additional intermediate affinity binders and 36F2) with a flow of 30 ⁇ L/minutes through the flow cells over 180 seconds.
  • the dissociation was monitored for 240 or 600 seconds.
  • the chip surface was regenerated after every cycle using a double injection of 30 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine IL2R Fc fusion (unrelated Fc fused receptor).
  • the binding curves resulting from the bivalent binding of the T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding.
  • the apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).
  • the chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1.
  • the affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • the steady state analysis model was applied using the Bia Evaluation software (GE Healthcare). The steady state analysis gives the KD of the binding reaction at equilibrium.
  • the mutations introduced into the 16D5 binders reduce its affinity to human and cynomolgus FolR1 as determined by surface plasmon resonance.
  • the ranking with decreasing affinity is 16D5 WT (57 nM)>W96Y (6.5 fold lower)>G49S/S93A (14.5 fold lower)>W96Y/D52E (24.5 fold lower)>G49S/K53A (30 fold lower).
  • the same ranking is visible in the avidity values, however the fold differences are smaller 16D5 WT (3 nM)>W96Y, G49S/S93A, W96Y/D52E (3 fold lower)>G49S/K53A (13 fold lower).
  • the intermediate affinity binders have following ranking in affinity 16D5 (57 nM)>14B1 (8.5 fold lower)>9C7 (15 fold lower)>6E10 (21 fold lower)>36F2 (24.5 fold lower). These differences however disappear in the avidity measurement 14B1, 9C7, 6E10 (1 nM)>16D5 (3 nM)>36F2 (7 nM).
  • 16D5 W96Y/D52E TCB addresses the problems observed with previous candidates.
  • 16D5 W96Y/D52E TCB is based on the common light chain 16D5 binder and has two point mutations on the heavy chain with respect to the parental 16D5 binder. The W96Y mutation reduces the affinity of the binder to FolR1 compared to the parental binder and the D52E mutation removes a deamidation site and also contributes to the reduction in affinity.
  • 16D5 W96Y/D52E TCB binds to human and cynomolgus FolR1, but not to murine FolR1. It is specific for FolR1 and does not bind to recombinant human FolR2 or human FolR3.
  • the affinity (monovalent binding) of 16D5 W96Y/D52E is around 1.4 ⁇ M for human FolR1 (24.5 fold lower than the parental 16D5 binder) and the avidity (bivalent binding) is around 10 nM (3 fold lower than the parental 16D5 binder).
  • T-cell killing mediated by intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), was assessed on Hela (high FolR1), SKov-3 (medium FolR1) and HT-29 (low FolR1) cells. 16D5 TCB and 36F2 TCB were included as benchmarks. Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at a density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight.
  • PBMCs Peripheral blood mononuclear cells
  • enriched lymphocyte preparations obtained from healthy human donors.
  • Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450 ⁇ g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400 ⁇ g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350 ⁇ g, 10 minutes).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h).
  • the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1.
  • T-cell killing mediated by affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5-G49S/K53A TCB, 16D5 W96Y TCB, 16D5 W96Y/D52E TCB) was assessed on Hela (high FolR1), SKov-3 (medium FolR1) and HT-29 (low FolR1) cells. 16D5 TCB and 36F2 TCB were included as benchmarks. The assay was performed as described above (Example 52).
  • the 16D5 W96Y/D52E TCB differentiates between high and low expressing cells which is of special importance to reduce toxicity as the cells of some normal, non-tumorous tissues express very low levels of FolR1 (approximately less than 1000 copies per cell). Consistent with this observation, the results discussed in Example 54 below show that 16D5 W96Y/D52E TCB induces much lower levels of T-cell-mediated killing of primary cells ( FIG. 38A-F ) compared to the parental 16D5 TCB. As such, 16D5 W96Y/D52E TCB mediates potent killing of tumor tissues with high or medium FOLR1 expression, but not of normal tissues with low expression.
  • 16D5 W96Y/D52E TCB in the bivalent 2+1 format comprises FolR1 binding moieties of relatively low affinity but it possesses an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this TCB selectively binds to tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all. As an additional advantage over the 36F2 FOLR1 TCB described above, the 16D5 W96Y/D52E TCB binds specifically to FolR1 and not to FolR2 or FolR3, further enhancing its safety for in vivo treatment.
  • the 16D5 W96Y/D52E TCB in the bivalent 2+1 inverted format also has the advantage that it does not require chemical cross linking or other hybrid approach. This makes it suitable for manufacture of a medicament to treat patients, for example patients having FolR1-positive cancerous tumors.
  • the 16D5 W96Y/D52E TCB in the bivalent 2+1 inverted format can be produced using standard CHO processes with low aggregates.
  • the 16D5 W96Y/D52E TCB in the bivalent 2+1 comprises human and humanized sequences making it superior to molecules that employ rat and murine polypeptides that are highly immunogenic when administered to humans.
  • the 16D5 W96Y/D52E TCB in the bivalent 2+1 format was engineered to abolish FcgR binding and, as such, does not cause FcgR crosslinking and infusion reactions, further enhancing its safety when administered to patients.
  • a further advantage of the 16D5 W96Y/D52E TCB in the bivalent 2+1 format and other embodiments disclosed herein is that their clinical development does not require the use of surrogate molecules as they bind to human and cynomous FolR1. As such, the molecules disclosed herein recognize a different epitope than antibodies to FolR1 previously described that do not recognize FolR1 from both species (see also FIG. 41 ).
  • HRCEpiC Human Renal Cortical Epithelial Cells
  • HRPEpiC Human Retinal Pigment Epithelial Cells
  • HT-29 cells low FolR1 were included as control cell line.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

The present invention generally relates to novel bispecific antigen binding molecules for T cell activation and re-direction to specific target cells. In addition, the present invention relates to polynucleotides encoding such bispecific antigen binding molecules, and vectors and host cells comprising such polynucleotides. The invention further relates to methods for producing the bispecific antigen binding molecules of the invention, and to methods of using these bispecific antigen binding molecules in the treatment of disease.

Description

    SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Nov. 9, 2015, is named 32186_SL.txt and is 445,570 bytes in size.
  • FIELD OF THE INVENTION
  • The present invention generally relates to bispecific antigen binding molecules for activating T cells, in particular to bispecific antibodies targeting CD3 and Folate Receptor 1 (FolR1). In addition, the present invention relates to polynucleotides encoding such bispecific antigen binding molecules, and vectors and host cells comprising such polynucleotides. The invention further relates to methods for producing the bispecific antigen binding molecules of the invention, and to methods of using these bispecific antigen binding molecules in the treatment of disease.
  • BACKGROUND
  • The selective destruction of an individual cell or a specific cell type is often desirable in a variety of clinical settings. For example, it is a primary goal of cancer therapy to specifically destroy tumor cells, while leaving healthy cells and tissues intact and undamaged.
  • An attractive way of achieving this is by inducing an immune response against the tumor, to make immune effector cells such as natural killer (NK) cells or cytotoxic T lymphocytes (CTLs) attack and destroy tumor cells. CTLs constitute the most potent effector cells of the immune system, however they cannot be activated by the effector mechanism mediated by the Fc domain of conventional therapeutic antibodies.
  • In this regard, bispecific antibodies designed to bind with one “arm” to a surface antigen on target cells, and with the second “arm” to an activating, invariant component of the T cell receptor (TCR) complex, have become of interest in recent years. The simultaneous binding of such an antibody to both of its targets will force a temporary interaction between target cell and T cell, causing activation of any cytotoxic T cell and subsequent lysis of the target cell. Hence, the immune response is re-directed to the target cells and is independent of peptide antigen presentation by the target cell or the specificity of the T cell as would be relevant for normal MHC-restricted activation of CTLs. In this context it is crucial that CTLs are only activated when a target cell is presenting the bispecific antibody to them, i.e. the immunological synapse is mimicked. Particularly desirable are bispecific antibodies that do not require lymphocyte preconditioning or co-stimulation in order to elicit efficient lysis of target cells.
  • FOLR1 is expressed on epithelial tumor cells of various origins, e.g., ovarian cancer, lung cancer, breast cancer, renal cancer, colorectal cancer, endometrial cancer. Several approaches to target FOLR1 with therapeutic antibodies, such as farletuzumab, antibody drug conjugates, or adoptive T cell therapy for imaging of tumors have been described (Kandalaft et al., J Transl Med. 2012 Aug. 3; 10:157. doi: 10.1186/1479-5876-10-157; van Dam et al., Nat Med. 2011 Sep. 18; 17(10):1315-9. doi: 10.1038/nm.2472; Clifton et al., Hum Vaccin. 2011 February; 7(2):183-90. Epub 2011 Feb. 1; Kelemen et al., Int J Cancer. 2006 Jul. 15; 119(2):243-50; Vaitilingam et al., J Nucl Med. 2012 July; 53(7); Teng et al., 2012 August; 9(8):901-8. doi: 10.1517/17425247.2012.694863. Epub 2012 Jun. 5. Some attempts have been made to target folate receptor-positive tumors with constructs that target the folate receptor and CD3 (Kranz et al., Proc Natl Acad Sci USA. Sep. 26, 1995; 92(20): 9057-9061; Roy et al., Adv Drug Deliv Rev. 2004 Apr. 29; 56(8):1219-31; Huiting Cui et al Biol Chem. Aug. 17, 2012; 287(34): 28206-28214; Lamers et al., Int. J. Cancer. 60(4):450 (1995); Thompson et al., MAbs. 2009 July-August; 1(4):348-56. Epub 2009 Jul. 19; Mezzanzanca et al., Int. J. Cancer, 41, 609-615 (1988). However, the approaches taken so far have many disadvantages. The molecules used thus far could not be readily and reliably produced as they require chemical cross linking. Similarly, hybrid molecules cannot be produced at large scale as human proteins and require the use of rat, murine or other proteins that are highly immunogenic when administered to humans and, thus, of limited therapeutic value. Further, many of the existing molecules retained FcgR binding.
  • Thus, there remains a need for novel, improved bispecific antibodies for targeted T cell mediated immunotherapy. The present invention provides bispecific antigen binding molecules designed for targeted T cell activation, particularly, bispecific antigen binding molecules suitable as effective and safe therapeutics that can be readily produced and dosed.
  • SUMMARY OF THE INVENTION
  • In one aspect, the invention provides a T cell activating bispecific antigen binding molecule comprising
      • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34; and
      • (ii) a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1).
  • In one embodiment, the T cell activating bispecific antigen binding molecule comprises a first antigen binding moiety that comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31. In one embodiment, the T cell activating bispecific antigen binding molecule additionally comprises (iii) a third antigen binding moiety capable of specific binding to FolR1. In one embodiment, the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences. In one embodiment, the third antigen binding moiety is identical to the second antigen binding moiety.
  • In one embodiment of the T cell activating bispecific antigen binding molecule of the above embodiments, at least one of the second and third antigen binding moiety is a Fab molecule. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments, additionally comprises an Fc domain composed of a first and a second subunit capable of stable association. In some embodiments, the first antigen binding moiety and the second antigen binding moiety are each connected at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain. In some embodiments, a third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, optionally via a peptide linker.
  • In one embodiment of the T cell activating bispecific antigen binding molecule of the above embodiments, the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34. In one embodiment, the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31. In one embodiment, the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65. In one embodiment, the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • In another embodiment, the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 50 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54. In one embodiment, the antigen binding moiety capable of specific binding to FolR1 comprises (a) a complementarity determining region heavy chain 1 (CDR-H1) amino acid sequences of SEQ ID NO: 8; (b) a CDR-H2 amino acid sequence of SEQ ID NO: 9; (c) a CDR-H3 amino acid sequence of SEQ ID NO: 50; (d) a complementarity determining region light chain 1 (CDR-L1) amino acid sequence of SEQ ID NO: 52; (e) a CDR-L2 amino acid sequence of SEQ ID NO: 53, and (f) a CDR-L3 amino acid sequence of SEQ ID NO: 54. In one embodiment, the antigen binding moiety capable of specific binding to FolR1 comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • In another embodiment, the antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprises at least one heavy chain complementarity determining region (CDR) amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 275 and SEQ ID NO: 315 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34. In one embodiment, the antigen binding moiety capable of specific binding to FolR1 comprises (a) a complementarity determining region heavy chain 1 (CDR-H1) amino acid sequences of SEQ ID NO: 16; (b) a CDR-H2 amino acid sequence of SEQ ID NO: 275; (c) a CDR-H3 amino acid sequence of SEQ ID NO: 315; (d) a complementarity determining region light chain 1 (CDR-L1) amino acid sequence of SEQ ID NO: 32; (e) a CDR-L2 amino acid sequence of SEQ ID NO: 33, and (f) a CDR-L3 amino acid sequence of SEQ ID NO: 34. In one embodiment, the antigen binding moiety capable of specific binding to FolR1 comprises a variable heavy chain domain (VH) comprising an amino acid sequence of SEQ ID NO: 274 and a variable light chain domain (VL) comprising an amino acid sequence of SEQ ID NO: 31.
  • In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1 and a cynomolgus monkey FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1, a cynomolgus monkey FolR1 and a murine FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiments binds to a human FolR1, a cynomolgus monkey FolR1 and not a murine FolR1.
  • In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the first antigen binding moiety is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments comprises not more than one antigen binding moiety capable of specific binding to CD3. In one embodiment of the T cell activating bispecific antigen binding molecule, the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule. In one embodiment, the Fc domain is an IgG class immunoglobulin, specifically an IgG1 or IgG4, Fc domain. In one embodiment, the Fc domain is a human Fc domain.
  • In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the Fc domain comprises a modification promoting the association of the first and the second subunit of the Fc domain. In one embodiment, in the CH3 domain of the first subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable. In one embodiment, the Fc domain comprises at least one amino acid substitution that reduces binding to an Fc receptor and/or effector function, as compared to a native IgG1 Fc domain. In one embodiment, each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G (Kabat numbering). In one embodiment, the Fc receptor is an Feγ receptor. In one embodiment, the effector function is antibody-dependent cell-mediated cytotoxicity (ADCC).
  • In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the T cell activating bispecific antigen binding molecule induces proliferation of a human CD3 positive T cell in vitro. In one embodiment, the T cell activating bispecific antigen binding molecule induces human peripheral blood mononuclear cell mediated killing of a FolR1-expressing human tumor cell in vitro. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of a FolR1-expressing human tumor cell in vitro. In one embodiment, the T cell is a CD8 positive T cell. In one embodiment, the FolR1-expressing human tumor cell is a Hela, Skov-3, HT-29, or HRCEpiC cell. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing human tumor cell in vitro with an EC50 of between about 36 pM and about 39573 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 36 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 178.4 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 134.5 pM or greater after 48 hours.
  • In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments induces upregulation of cell surface expression of at least one of CD25 and CD69 on the T cell as measured by flow cytometry. In one embodiment, the T cell is a CD4 positive T cell or a CD8 positive T cell. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds human FolR1 with an apparent KD of about 5.36 pM to about 4 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent KD of about 4 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds murine FolR1 with an apparent KD of about 1.5 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of at least about 1000 nM.
  • In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments is specific for FolR1 and does not bind to FolR2 or FolR3. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments has an affinity (monovalent binding) of 1 μM or greater. In one embodiment, the affinity is around 1.4 μM for human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments has an avidity (bivalent binding) of about 1-100 nM or lower. In one embodiment, the avidity is about 10 nM or less. In one embodiment, the avidity is 10 nM.
  • In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to FolR1 expressed on a human tumor cell. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to a conformational epitope on human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3). In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the antigen binding moiety binds to a FolR1 polypeptide comprising the amino acids 25 to 234 of human FolR1 (SEQ ID NO:227). In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NOs: 227, 230 and 231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 and 229.
  • In another aspect, the invention provides for a bispecific antibody comprising a) a first antigen-binding site that competes for binding to human FolR1 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 49 and a variable light chain domain of SEQ ID NO: 51; and b) a second antigen-binding site that competes for binding to human CD3 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31, wherein binding competition is measured using a surface plasmon resonance assay.
  • In another aspect, the invention provides for a bispecific antibody comprising a) a first antigen-binding site that competes for binding to human FolR1 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31; and b) a second antigen-binding site that competes for binding to human CD3 with a reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31, wherein binding competition is measured using a surface plasmon resonance assay.
  • In another aspect, the invention provides for a T cell activating bispecific antigen binding molecule comprising a first antigen binding moiety capable of specific binding to CD3, and a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1), wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human FolR1 as a first reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 49 and a variable light chain domain of SEQ ID NO: 51; and wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human CD3 as a second reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31.
  • In another aspect, the invention provides for a T cell activating bispecific antigen binding molecule comprising a first antigen binding moiety capable of specific binding to CD3, and a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1), wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human FolR1 as a first reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain (VL) of SEQ ID NO: 31; and wherein the T cell activating bispecific antigen binding molecule binds to the same epitope on human CD3 as a second reference antibody comprising a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain (VL) of SEQ ID NO: 31.
  • In another aspect, the invention relates to an antibody or an antigen-binding fragment thereof that competes for binding to human FolR1 with an antibody that comprises a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31, wherein binding competition is measured using a surface plasmon resonance assay.
  • In one aspect, the invention provides for a T cell activating bispecific antigen binding molecule, wherein the antigen binding molecule comprises a first, second, third, fourth and fifth polypeptide chain that form a first, a second and a third antigen binding moiety, wherein the first antigen binding moiety is capable of binding CD3 and the second and the third antigen binding moiety each are capable of binding Folate Receptor 1 (FolR1), wherein a) the first and the second polypeptide chain comprise, in amino (N)-terminal to carboxyl (C)-terminal direction, VLD1 and CLD1; b) the third polypeptide chain comprises, in N-terminal to C-terminal direction, VLD2 and CH1D2; c) the fourth polypeptide chain comprises, in N-terminal to C-terminal direction, VHD1, CH1D1, CH2D1 and CH3D1; d) the fifth polypeptide chain comprises VHD1, CH1D1, VHD2, CLD2, CH2D2 and CH3D2; wherein VLD1 is a first light chain variable domain, VLD2 is a second light chain variable domain, CLD1 is a first light chain constant domain, CLD2 is a second light chain constant domain, VHD1 is a first heavy chain variable domain, VHD2 is a second heavy chain variable domain, CH1D1 is a first heavy chain constant domain 1, CH1D2 is a second heavy chain constant domain 1, CH2D1 is a first heavy chain constant domain 2, CH2D2 is a second heavy chain constant domain 2, CH3D1 is a first heavy chain constant domain 3, and CH3D2 is a second heavy chain constant domain 3.
  • In one embodiment of the T cell activating bispecific antigen binding molecule, (i) the third polypeptide chain and VHD2 and CLD2 of the fifth polypeptide chain form the first antigen binding moiety capable of binding CD3; (ii) the first polypeptide chain and VHD1 and CH1D1 of the fourth polypeptide chain form the second binding moiety capable of binding to FolR1; and (iii) the second polypeptide chain and VHD1 and CH1D1 of the fifth polypeptide chain form the third binding moiety capable of binding to FolR1. In one embodiment, CH2D1, CH3D1, CH2D2 and CH3D2 form an Fc domain of an IgG class immunoglobulin. In one embodiment, the Fc domain is a human Fc domain. In one embodiment, the Fc domain comprises a modification promoting the association of the first and the second subunit of the Fc domain. In one embodiment, CH3D2 comprises an amino acid residue having a larger side chain volume, which is positionable in a cavity within CH3D1. In one embodiment, the Fc domain comprises at least one amino acid substitution that reduces binding to an Fc receptor and/or effector function, as compared to a native IgG1 Fc domain. In one embodiment, each subunit of the Fc domain comprises three amino acid substitutions that reduce at least one of binding to an activating Fc receptor and effector function wherein said amino acid substitutions are L234A, L235A and P329G according to Kabat numbering. In one embodiment, the Fc receptor is an Fcγ receptor. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces proliferation of a human CD3 positive T cell in vitro. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces human peripheral blood mononuclear cell mediated killing of a FolR1-expressing human tumor cell in vitro. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of a FolR1-expressing human tumor cell in vitro. In one such embodiment, the FolR1-expressing human tumor cell is a Hela, Skov-3, HT-29, or HRCEpiC cell. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of between about 36 pM and about 39573 pM after 24 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 36 pM after 24 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 178.4 pM after 24 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 134.5 pM or greater after 48 hours. In one of the above embodiments, the T cell activating bispecific antigen binding molecule induces upregulation of cell surface expression of at least one of CD25 and CD69 on the T cell as measured by flow cytometry. In one such embodiments, the T cell is a CD4 positive T cell or a CD8 positive T cell. In one of the above embodiments, wherein the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent KD of about 5.36 pM to about 4 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent KD of about 4 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds murine FolR1 with an apparent KD of about 1.5 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of at least about 1000 nM. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds to FolR1 expressed on a human tumor cell. In one of the above embodiments, the T cell activating bispecific antigen binding molecule binds to a conformational epitope on human FolR1. In one of the above embodiments, the T cell activating bispecific antigen binding molecule does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3). In one of the above embodiments, the antigen binding moiety binds to a FolR1 polypeptide comprising the amino acids 25 to 234 of human FolR1 (SEQ ID NO:227). In one of the above embodiments, the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NOs:227, 230 and 231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 and 229. In one of the above embodiments, the T cell activating bispecific antigen binding molecule is a humanized or a chimeric molecule. In one of the above embodiments, VHD2 and CH1D1 are linked through a peptide linker.
  • In one of the above embodiments of the T cell activating bispecific antigen binding molecule, the first and second polypeptide chain comprise the amino acid sequence of SEQ ID NO:230. In one of the above embodiments of the T cell activating bispecific antigen binding molecule, the third polypeptide chain comprises the amino acid sequence of SEQ ID NO:86. In one of the above embodiments of the T cell activating bispecific antigen binding molecule, the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO:309. In one of the above embodiments of the T cell activating bispecific antigen binding molecule, the fifth polypeptide chain comprises the amino acid sequence of SEQ ID NO:308. In one of the above embodiments of the T cell activating bispecific antigen binding molecule, the first and second polypeptide chain comprise the amino acid sequence of SEQ ID NO:230; the third polypeptide chain comprises the amino acid sequence of SEQ ID NO:86; the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO:309; and the fifth polypeptide chain comprise the amino acid sequence of SEQ ID NO:308.
  • In one aspect, the invention provides for a T cell activating bispecific antigen binding molecule comprising the amino acid sequence of SEQ ID NO:308. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiment further comprises the amino acid sequence of SEQ ID NO:230 and of SEQ ID NO:86.
  • In one aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:308. In one aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:309.
  • In one aspect, the invention provides for a T cell activating bispecific antigen binding molecule comprising the amino acid sequence of SEQ ID NO:276. In one embodiment, the T cell activating bispecific antigen binding molecule of the above embodiment further comprises the amino acid sequence of SEQ ID NO:277 and of SEQ ID NO:35.
  • In one aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:277. In one aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:276.
  • In one aspect, the invention provides for an isolated polynucleotide encoding the T cell activating bispecific antigen binding molecule of any one of the embodiments disclosed herein. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:169. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:246. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:247. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:97. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:198.
  • In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:287. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:288. In one embodiment, the invention provides for an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule comprising the nucleotide sequence of SEQ ID NO:289.
  • In one aspect, the invention provides for an isolated polypeptide encoded by the polynucleotide of the above embodiment. In another aspect, the invention provides for a vector, particularly an expression vector, comprising the polynucleotide encoding the T cell activating bispecific antigen binding molecule of any one of the embodiments disclosed herein. In another aspect, the invention provides for a host cell comprising a polynucleotide or a vector of any of the embodiments disclosed herein.
  • In one aspect, the invention provides for a method of producing the T cell activating bispecific antigen binding molecule capable of specific binding to CD3 and a target cell antigen, comprising the steps of a) culturing the host cell of the above embodiments under conditions suitable for the expression of the T cell activating bispecific antigen binding molecule and b) recovering the T cell activating bispecific antigen binding molecule.
  • In one aspect, the invention provides for T cell activating bispecific antigen binding molecule produced by the method of the above embodiment.
  • In one aspect, the invention provides for a pharmaceutical composition comprising the T cell activating bispecific antigen binding molecule of any one of the above embodiments and a pharmaceutically acceptable carrier. In one aspect, the invention provides for the T cell activating bispecific antigen binding molecule of any one of the above embodiments or the pharmaceutical composition of any of the above embodiments for use as a medicament.
  • In one aspect, the invention provides for the T cell activating bispecific antigen binding molecule of any one of the above embodiments or the pharmaceutical composition of any one of the above embodiments for use in the treatment of a disease in an individual in need thereof. In some embodiments, the disease is cancer. In one aspect, the invention provides for a use of the T cell activating bispecific antigen binding molecule of any one of the above embodiments for the manufacture of a medicament for the treatment of a disease in an individual in need thereof.
  • In one aspect, the invention provides for a method of treating a disease in an individual, comprising administering to said individual a therapeutically effective amount of a composition comprising the T cell activating bispecific antigen binding molecule of any one of the above embodiments in a pharmaceutically acceptable form. In some embodiments, said disease is a cancer.
  • In one aspect, the invention provides for a method for inducing lysis of a target cell, comprising contacting a target cell with the T cell activating bispecific antigen binding molecule of any one of the above embodiments in the presence of a T cell.
  • In one aspect, the invention provides for a the invention as described hereinbefore.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-I illustrate exemplary configurations of the T cell activating bispecific antigen binding molecules (TCBs) of the invention. All constructs except the kappa-lambda format in (FIG. 1I) have P329G LALA mutations and comprise knob-into-hole Fc fragments with knob-into-hole modifications. (FIG. 1A) Illustration of the “FolR1 TCB 2+1 inverted (common light chain)”. The FolR1 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. These constructs are not crossed and have three times the same VLCL light chain. (FIG. 1B) Illustration of the “FolR1 TCB 1+1 head-to-tail (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain. (FIG. 1C) Illustration of the “FolR1 TCB 1+1 classical (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain. (FIG. 1D) Illustration of the “FolR1 TCB 2+1 classical (common light chain)”. The CD3 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. These constructs are not crossed and have three times the same VLCL light chain. (FIG. 1E) Illustration of the “FolR1 TCB 2+1 crossfab classical”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. The CD3 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. (FIG. 1F) Illustration of the “FolR1 TCB 2+1 crossfab inverted”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. The FolR1 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. (FIG. 1G) Illustration of the “FolR1 TCB 1+1 crossfab head-to-tail”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. (FIG. 1H) Illustration of the “FolR1 TCB 1+1 crossfab classical”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. FIG. 1I illustrates the CD3/FolR1 kappa-lambda antibody format. These constructs comprise a crossed common light chain VLCH1 and one crossed VHCL chain specific for CD3 and one crossed VHCL chain specific for FolR1.
  • FIGS. 2A-C depict graphs summarizing Binding of FoLR1 IgG binders to HeLa cells. Binding of newly generated FolR1 binders to FolR1 expressed on HeLa cells were determined by flow cytometry. Bound antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 3A-B depict graphs summarizing specificity of FolR1 binders for FolR1. Binding of FolR1 IgGs to HEK cells transiently transfected with either FolR1 or FolR2 was analyzed by flow cytometry to identify clones which bind specifically to FolR1 and not to FolR2. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 4A-B depict graphs summarizing cross-reactivity of FolR1 binders to cyFoLR1. Cross-reactivity of the FolR1 antibodies to cyno FolR1 was addressed on HEK cells transiently transfected with cyFolR1 by flow cytometry. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIG. 5 depicts a graph illustrating internalization of FolR1 TCBs after binding. Internalization of the four FolR1 TCBs after binding to FolR1 was tested on HeLa cells. Remaining FolR1 TCBs on the surface were detected with a fluorescently labeled anti-human secondary antibody after indicated time points of incubation at 37° C. Percentage of internalization was calculated.
  • FIGS. 6A-E depict graphs summarizing binding of FolR1 IgGs to cells with different FolR1 expression levels. Binding of 9D11, 16D5 and Mov19 IgG to tumor cells with different FolR1 expression levels was analyzed by flow cytometry. DP47 IgG was included as isotype control and MKN-45 were included as FolR1 negative cell line. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 7A-L depict graphs summarizing T cell mediated killing of HT-29 and SKOV3 cells. FolR1 TCBs were used to test T cell mediated killing of HT-29 and SKOV3 tumor cells and upregulation of activation marker on T cells upon killing. (FIGS. 7A-D) T cell mediated killing of HT-29 and SKOV3 cells in the presence of 9D11 FolR1 TCB and 16D5 FolR1 TCB was measured by LDH release after 24 h and 48 h. DP47 TCB was included as negative control. After 48 h incubation upregulation of the activation marker CD25 and CD69 on CD8 T cells and CD4 T cells upon killing of SKOV3 (FIG. 7E-H) or HT-29 (FIG. 7I-L) tumor cells was assessed by flow cytometry.
  • FIG. 8 depicts a graph showing absence of anti-FolR1 binding to erythrocytes. Erythrocytes were gated as CD235a positive population and binding of 9D11 IgG, 16D5 IgG, Mov19 IgG and DP47 IgG to this population was determined by flow cytometry. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.
  • FIGS. 9A-D depict graphs summarizing activation marker upregulation in whole blood. CD25 and CD69 activation marker upregulation of CD4 T cells and CD8 T cells 24 h after addition of 9D11 FolR1 TCB, 16D5 FolR1 TCB, Mov19 FolR1 TCB and DP47 TCB was analyzed by flow cytometry.
  • FIG. 10 Binding of 9D11 TCB a-glyco variants to HeLa cells. Binding of 9D11 FolR1 TCB a-glyco variants to Hela cells was compared to binding of the original 9D11 TCB on HeLa cells. The antibodies were detected with a fluorescently labeled anti-human secondary antibody and binding was determined by flow cytometry.
  • FIGS. 11A-F depict graphs summarizing T cell mediated killing with 9D11 FolR1 TCB a-glyco variants of tumor cells. 9D11 FolR1 TCB a-glyco variants were used to test T cell mediated killing of (FIG. 11A-D) SKOV3, MKN-45 (as FolR1 negative control) and (FIG. 11E-F) HT-29 tumor cells in comparison to killing with the original 9D11 FolR1 TCB. As read-out LDH release after 24 h and 48 h was used.
  • FIGS. 12A-X depict graphs summarizing T cell mediated killing of primary epithelial cells. Primary epithelial cells with very low levels of FolR1 were used to test T cell mediated killing with 16D5 FolR1 TCB and 9D11 FolR1 TCB, DP47 TCB was included as a negative control and HT29 cells were included as positive control. (FIGS. 12A-H) LDH release of human retinal pigment (HRP), human renal cortical (HRC), human bronchial (HB) and of HT29 cells was determined after 24 h and 48 h. CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells upon killing of (FIGS. 12I-L) HRP, (FIGS. 12M-P) HRC, (FIGS. 12Q-T) HB and (FIG. 12 U-X) HT29 was determined after 48 h by flow cytometry.
  • FIGS. 13A-C show a comparison of different TCB formats with 16D5. Four different TCB formats containing the FolR1 binder 16D5 were compared in FIG. 13A binding to HeLa cells, in FIG. 14 B T cell mediated killing of SKOV3 cells after 24 h and 48 h and in FIG. 14C CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells 48 h after killing.
  • FIGS. 14A-C depict a comparison of different TCB formats with 9D11. Three different TCB formats containing the FolR1 binder 9D11 were compared in A) binding to HeLa cells, in B) T cell mediated killing of SKOV3 cells after 24 h and 48 h and in C) CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells 48 h after killing.
  • FIG. 15 depicts a PK-profile of FOLR1 TCB in NOG mice for three different doses.
  • FIG. 16 illustrates an experimental protocol for efficacy study with FOLR1 TCB.
  • FIGS. 17A-B depict tumor growth curves. (FIG. 17A) Mean values and SEM of tumor volumes in the different treatment groups. (FIG. 17B) Tumor growth of single mice in all treatment groups. TGI (tumor growth inhibition) give the percentage of the Mean tumor volume compared to vehicle group.
  • FIG. 18 shows tumor weights at study termination.
  • FIGS. 19A-B show FACS analysis of tumor infiltrating T-cells at study day 32. (FIG. 19A) Tumor single cells suspensions were stained with anti-human CD3/CD4/CD8 and analyzed by flow cytometry. (FIG. 19B) Mean values and SEM of T-cell counts per mg tumor tissue in different treatment groups.
  • FIGS. 20A-B show FACS analysis for T-cell activation/degranulation and cytokine secretion at study day 32. CD4+ (FIG. 20A) and CD8+ (FIG. 20B) tumor infiltrating T-cells were stained for cytokines, activation and degranulation markers. Displayed are the mean values and SEM of T-cell counts per mg tumor tissue in different treatment groups.
  • FIGS. 21A-B show percent tumor lysis. SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 24 h (FIG. 21A) and 48 h (FIG. 21B) killing of tumor cells was determined by measuring LDH release.
  • FIGS. 22A-D show CD25 and CD69 upregulation on CD4 T cells. SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 48 h CD25 and CD69 upregulation on CD4 T cells (FIG. 22A-B) and CD8 T cells (FIG. 22C-D) was measured by flow cytometry.
  • FIGS. 23A-B show percent tumor lysis. T-cell killing of SKov-3 cells (medium FolR1) induced by 36F2 TCB, Mov19 TCB and 21A5 TCB after 24h (FIG. 23A) and 48 h (FIG. 23B) of incubation (E:T=10:1, effectors human PBMCs).
  • FIGS. 24A-C show T-cell killing induced by 36F2 TCB, 16D5 TCB, 16D5 TCB classical, 16D5 TCB 1+1 and 16D5 TCB HT of Hela (high FolR1) (FIG. 24A), Skov-3 (medium FolR1) (FIG. 24B) and HT-29 (low FolR1) (FIG. 24C) human tumor cells (E:T=10:1, effectors human PBMCs, incubation time 24 h). DP47 TCB was included as non-binding control.
  • FIGS. 25A-C show upregulation of CD25 and CD69 on human CD8+(FIG. 25A, B) and CD4+(FIG. 25C), T cells after T cell-mediated killing of Hela cells (high FolR1) (FIG. 25A), SKov-3 cells (medium FolR1) (FIG. 25B) and HT-29 cells (low FolR1) (FIG. 25C) (E:T=10:1, 48 h incubation) induced by 36F2 TCB, 16D5 TCB and DP47 TCB (non-binding control).
  • FIGS. 26A-F show T-cell killing induced by 36F2 TCB, 16D5 TCB and DP47 TCB of human Renal Cortical Epithelial Cells (FIG. 26A, B), human Retinal Pigment Epithelial Cells (FIG. 26C, D) and HT-29 cells (FIG. 26E, F) cells after 24h (FIG. 26A, C, E) and 48 h (FIG. 26B, D, F) of incubation (E:T=10:1, effectors human PBMCs).
  • FIG. 27 depicts a table summarizing quantification of FolR1 binding sites on various normal and cancer cells lines.
  • FIGS. 28A-B show binding of 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA to human CD3 expressed on Jurkat cells.
  • FIGS. 29A-B show T-cell killing of SKov-3 (medium FolR1) human tumor cells induced by 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA (FIG. 29A) and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA (FIG. 29B) (E:T=10:1, effectors human PBMCs, incubation time 24 h). DP47 TCB was included as non-binding control.
  • FIG. 30A-B show T-cell killing of HT-29 (low FolR1) human tumor cells induced by 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA (FIG. 30A) and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA (FIG. 30B) (E:T=10:1, effectors human PBMCs, incubation time 24 h). DP47 TCB was included as non-binding control.
  • FIGS. 31A-C show mean fluorescence intensity and tumor cell lysis.
  • FIGS. 32A-E shows binding of 36F2 TCB, 16D5 TCB and 16D5 HC/LC variants to human FolR1 expressed on Hela cells.
  • FIG. 33 shows binding of 36F2 TCB, 16D5 TCB and the two 16D5 affinity reduced variants 16D5 W96Y/D52E TCB and 16D5 G49S/S93A TCB to human FolR1 on Hela cells.
  • FIGS. 34A-E show binding of 36F2 TCB, 16D5 TCB and 16D5 HC/LC variants to human FolR1 expressed on HT-29 cells.
  • FIGS. 35A-D show binding of intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), 16D5 TCB and 36F2 TCB to HEK293T cells expressing either human or mouse FolR1 or FolR2.
  • FIG. 36A-F show T-cell killing of Hela (high FolR1 expression), SKov-3 (medium FolR1 expression) and HT-29 (low FolR1 expression) human tumor cells induced by intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), 16D5 TCB and 36F2 TCB after 24 h (A-C) and 48 h (D-F) of incubation. Human PBMCs were used as effector cells (E:T=10:1).
  • FIG. 37A-F shows T-cell killing of Hela (high FolR1 expression), SKov-3 (medium FolR1 expression) and HT-29 (low FolR1 expression) human tumor cells induced by affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5-G49S/K53A TCB, 16D5 W96Y TCB, 16D5 W96Y/D52E TCB), 16D5 TCB and 36F2 TCB after 24 h (FIG. 38A-C) and 48 h (FIG. 38D-F) of incubation. Human PBMCs were used as effector cells (E:T=10:1).
  • FIG. 38A-F show T-cell killing of primary human cells from retinal pigment epithelium and renal cortical epithelium induced by affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5 W96Y/D52E TCB), 16D5 TCB, 36F2 TCB and the intermediate FolR1 binder 14B1 TCB was assessed after 24 h (FIG. 39A-C) and 48 h (FIG. 39D-F) of incubation (E:T=10:1, effectors human PBMCs). HT-29 cells (low FolR1 expression) were included as control cell line and DP47 TCB served as non-binding control.
  • FIGS. 39A-B show single dose PK of FOLR1 TCB constructs in female NOG mice.
  • FIGS. 40A-G show in vivo efficacy of FOLR1 TCB constructs (16D5, 16D5 G49S/S93A and 16D5 W96Y/D52E) after human PBMC transfer in Hela-bearing NOG mice.
  • FIG. 41 shows that Farletuzumab (dark green, second from the top) and Mov19 (grey, top) are able to bind on huFolR1 that is captured on 16D5, demonstrating that the 16D5 series binders recognize an epitope distinct from that recognized by either Farletuzumab or Mov19.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • Terms are used herein as generally used in the art, unless otherwise defined in the following.
  • As used herein, the term “antigen binding molecule” refers in its broadest sense to a molecule that specifically binds an antigenic determinant. Examples of antigen binding molecules are immunoglobulins and derivatives, e.g. fragments, thereof.
  • The term “bispecific” means that the antigen binding molecule is able to specifically bind to at least two distinct antigenic determinants. Typically, a bispecific antigen binding molecule comprises at least two antigen binding sites, each of which is specific for a different antigenic determinant. In certain embodiments the bispecific antigen binding molecule is capable of simultaneously binding two antigenic determinants, particularly two antigenic determinants expressed on two distinct cells.
  • The term “valent” as used herein denotes the presence of a specified number of antigen binding sites in an antigen binding molecule. As such, the term “monovalent binding to an antigen” denotes the presence of one (and not more than one) antigen binding site specific for the antigen in the antigen binding molecule.
  • An “antigen binding site” refers to the site, i.e. one or more amino acid residues, of an antigen binding molecule which provides interaction with the antigen. For example, the antigen binding site of an antibody comprises amino acid residues from the complementarity determining regions (CDRs). A native immunoglobulin molecule typically has two antigen binding sites, a Fab molecule typically has a single antigen binding site.
  • As used herein, the term “antigen binding moiety” refers to a polypeptide molecule that specifically binds to an antigenic determinant. In one embodiment, an antigen binding moiety is able to direct the entity to which it is attached (e.g. a second antigen binding moiety) to a target site, for example to a specific type of tumor cell or tumor stroma bearing the antigenic determinant. In another embodiment an antigen binding moiety is able to activate signaling through its target antigen, for example a T cell receptor complex antigen. Antigen binding moieties include antibodies and fragments thereof as further defined herein. Particular antigen binding moieties include an antigen binding domain of an antibody, comprising an antibody heavy chain variable region and an antibody light chain variable region. In certain embodiments, the antigen binding moieties may comprise antibody constant regions as further defined herein and known in the art. Useful heavy chain constant regions include any of the five isotypes: α, δ, ε, γ, or μ. Useful light chain constant regions include any of the two isotypes: κ and λ.
  • As used herein, the term “antigenic determinant” is synonymous with “antigen” and “epitope,” and refers to a site (e.g. a contiguous stretch of amino acids or a conformational configuration made up of different regions of non-contiguous amino acids) on a polypeptide macromolecule to which an antigen binding moiety binds, forming an antigen binding moiety-antigen complex. Useful antigenic determinants can be found, for example, on the surfaces of tumor cells, on the surfaces of virus-infected cells, on the surfaces of other diseased cells, on the surface of immune cells, free in blood serum, and/or in the extracellular matrix (ECM). The proteins referred to as antigens herein, e.g., FolR1 and CD3, can be any native form the proteins from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats), unless otherwise indicated. In a particular embodiment the antigen is a human protein. Where reference is made to a specific protein herein, the term encompasses the “full-length”, unprocessed protein as well as any form of the protein that results from processing in the cell. The term also encompasses naturally occurring variants of the protein, e.g. splice variants or allelic variants. Exemplary human proteins useful as antigens include, but are not limited to: FolR1 (Folate receptor alpha (FRA); Folate binding protein (FBP); human FolR1 UniProt no.: P15328; murine FolR1 UniProt no.: P35846; cynomolgus FolR1 UniProt no.: G7PR14) and CD3, particularly the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP 000724.1, SEQ ID NO:150 for the human sequence; or UniProt no. Q95LI5 (version 49), NCBI GenBank no. BAB71849.1, for the cynomolgus [Macaca fascicularis] sequence). The T cell activating bispecific antigen binding molecule of the invention binds to an epitope of CD3 or a target cell antigen that is conserved among the CD3 or target antigen from different species. In certain embodiments the T cell activating bispecific antigen binding molecule of the invention binds to CD3 and FolR1, but does not bind to FolR2 (Folate receptor beta; FRB; human FolR2 UniProt no.: P14207) or FolR3 (Folate receptor gamma; human FolR3 UniProt no.: P41439).
  • By “specific binding” is meant that the binding is selective for the antigen and can be discriminated from unwanted or non-specific interactions. The ability of an antigen binding moiety to bind to a specific antigenic determinant can be measured either through an enzyme-linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance (SPR) technique (analyzed on a BIAcore instrument) (Liljeblad et al., Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)). In one embodiment, the extent of binding of an antigen binding moiety to an unrelated protein is less than about 10% of the binding of the antigen binding moiety to the antigen as measured, e.g., by SPR. In certain embodiments, an antigen binding moiety that binds to the antigen, or an antigen binding molecule comprising that antigen binding moiety, has a dissociation constant (KD) of ≤1 μM, ≤100 nM, ≤10 nM, ≤1 nM, ≤0.1 nM, ≤0.01 nM, or ≤0.001 nM (e.g. 10−8M or less, e.g. from 10−8M to 10−13M, e.g., from 10−9M to 10−13 M).
  • “Affinity” refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., a receptor) and its binding partner (e.g., a ligand). Unless indicated otherwise, as used herein, “binding affinity” refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., an antigen binding moiety and an antigen, or a receptor and its ligand). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD), which is the ratio of dissociation and association rate constants (koff and kon, respectively). Thus, equivalent affinities may comprise different rate constants, as long as the ratio of the rate constants remains the same. Affinity can be measured by well-established methods known in the art, including those described herein. A particular method for measuring affinity is Surface Plasmon Resonance (SPR).
  • “Reduced binding”, for example reduced binding to an Fc receptor, refers to a decrease in affinity for the respective interaction, as measured for example by SPR. For clarity the term includes also reduction of the affinity to zero (or below the detection limit of the analytic method), i.e. complete abolishment of the interaction. Conversely, “increased binding” refers to an increase in binding affinity for the respective interaction.
  • “T cell activation” as used herein refers to one or more cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. The T cell activating bispecific antigen binding molecules of the invention are capable of inducing T cell activation. Suitable assays to measure T cell activation are known in the art described herein.
  • A “target cell antigen” as used herein refers to an antigenic determinant presented on the surface of a target cell, for example a cell in a tumor such as a cancer cell or a cell of the tumor stroma. In particular “target cell antigen” refers to Folate Receptor 1.
  • As used herein, the terms “first” and “second” with respect to antigen binding moieties etc., are used for convenience of distinguishing when there is more than one of each type of moiety. Use of these terms is not intended to confer a specific order or orientation of the T cell activating bispecific antigen binding molecule unless explicitly so stated.
  • A “Fab molecule” refers to a protein consisting of the VH and CH1 domain of the heavy chain (the “Fab heavy chain”) and the VL and CL domain of the light chain (the “Fab light chain”) of an immunoglobulin.
  • The term “Fab molecules having identical VLCL light chains” as used therein refers to binders that share one light chain but still have separate specificities, e.g., can bind CD3 or FolR1. In some embodiments the T-cell activating bispecific molecules comprise at least two Fab molecules having identical VLCL light chains. The corresponding heavy chains are remodeled and confer specific binding to the T cell activating bispecific antigen CD3 and the target cell antigen FolR1, respectively.
  • By “fused” is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.
  • As used herein, the term “single-chain” refers to a molecule comprising amino acid monomers linearly linked by peptide bonds. In certain embodiments, one of the antigen binding moieties is a single-chain Fab molecule, i.e. a Fab molecule wherein the Fab light chain and the Fab heavy chain are connected by a peptide linker to form a single peptide chain. In a particular such embodiment, the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain in the single-chain Fab molecule.
  • By a “crossover” Fab molecule (also termed “Crossfab”) is meant a Fab molecule wherein either the variable regions or the constant regions of the Fab heavy and light chain are exchanged, i.e. the crossover Fab molecule comprises a peptide chain composed of the light chain variable region and the heavy chain constant region, and a peptide chain composed of the heavy chain variable region and the light chain constant region. For clarity, in a crossover Fab molecule wherein the variable regions of the Fab light chain and the Fab heavy chain are exchanged, the peptide chain comprising the heavy chain constant region is referred to herein as the “heavy chain” of the crossover Fab molecule. Conversely, in a crossover Fab molecule wherein the constant regions of the Fab light chain and the Fab heavy chain are exchanged, the peptide chain comprising the heavy chain variable region is referred to herein as the “heavy chain” of the crossover Fab molecule. An antibody that comprises one or more CrossFabs is referred to herein as “CrossMab.”
  • In contrast thereto, by a “conventional” Fab molecule is meant a Fab molecule in its natural format, i.e. comprising a heavy chain composed of the heavy chain variable and constant regions (VH-CH1), and a light chain composed of the light chain variable and constant regions (VL-CL).
  • The term “immunoglobulin molecule” refers to a protein having the structure of a naturally occurring antibody. For example, immunoglobulins of the IgG class are heterotetrameric glycoproteins of about 150,000 daltons, composed of two light chains and two heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3), also called a heavy chain constant region. Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain, also called a light chain constant region. The heavy chain of an immunoglobulin may be assigned to one of five types, called α (IgA), δ (IgD), ε (IgE), γ (IgG), or μ (IgM), some of which may be further divided into subtypes, e.g. γ1 (IgG1), γ2 (IgG2), γ3 (IgG3), γ4 (IgG4), α1 (IgA1) and α2 (IgA2). The light chain of an immunoglobulin may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain. An immunoglobulin essentially consists of two Fab molecules and an Fc domain, linked via the immunoglobulin hinge region.
  • The term “antibody” herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • An “antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab′, Fab′-SH, F(ab′)2, diabodies, linear antibodies, single-chain antibody molecules (e.g. scFv), and single-domain antibodies. For a review of certain antibody fragments, see Hudson et al., Nat Med 9, 129-134 (2003). For a review of scFv fragments, see e.g. Plückthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994); see also WO 93/16185; and U.S. Pat. Nos. 5,571,894 and 5,587,458. For discussion of Fab and F(ab′)2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Pat. No. 5,869,046. Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al., Nat Med 9, 129-134 (2003); and Hollinger et al., Proc Natl Acad Sci USA 90, 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat Med 9, 129-134 (2003). Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, Mass.; see e.g. U.S. Pat. No. 6,248,516 B1). Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • The term “antigen binding domain” refers to the part of an antibody that comprises the area which specifically binds to and is complementary to part or all of an antigen. An antigen binding domain may be provided by, for example, one or more antibody variable domains (also called antibody variable regions). Particularly, an antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).
  • The term “variable region” or “variable domain” refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). See, e.g., Kindt et al., Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007). A single VH or VL domain may be sufficient to confer antigen-binding specificity.
  • The term “hypervariable region” or “HVR”, as used herein, refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops (“hypervariable loops”). Generally, native four-chain antibodies comprise six HVRs; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the complementarity determining regions (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. Hypervariable regions (HVRs) are also referred to as “complementarity determining regions” (CDRs), and these terms are used herein interchangeably in reference to portions of the variable region that form the antigen binding regions. This particular region has been described by Kabat et al., U.S. Dept. of Health and Human Services, Sequences of Proteins of Immunological Interest (1983) and by Chothia et al., J Mol Biol 196:901-917 (1987), where the definitions include overlapping or subsets of amino acid residues when compared against each other. Nevertheless, application of either definition to refer to a CDR of an antibody or variants thereof is intended to be within the scope of the term as defined and used herein. The appropriate amino acid residues which encompass the CDRs as defined by each of the above cited references are set forth below in Table A as a comparison. The exact residue numbers which encompass a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can routinely determine which residues comprise a particular CDR given the variable region amino acid sequence of the antibody.
  • TABLE A
    CDR Definitions1
    CDR Kabat Chothia AbM2
    VH CDR1 31-35 26-32 26-35
    VH CDR2 50-65 52-58 50-58
    VH CDR3  95-102  95-102  95-102
    VL CDR1 24-34 26-32 24-34
    VL CDR2 50-56 50-52 50-56
    VL CDR3 89-97 91-96 89-97
    1Numbering of all CDR definitions in Table A is according to the numbering conventions set forth by Kabat et al. (see below).
    2“AbM” with a lowercase “b” as used in Table A refers to the CDRs as defined by Oxford Molecular's “AbM” antibody modeling software.
  • Kabat et al. also defined a numbering system for variable region sequences that is applicable to any antibody. One of ordinary skill in the art can unambiguously assign this system of “Kabat numbering” to any variable region sequence, without reliance on any experimental data beyond the sequence itself. As used herein, “Kabat numbering” refers to the numbering system set forth by Kabat et al., U.S. Dept. of Health and Human Services, “Sequence of Proteins of Immunological Interest” (1983). Unless otherwise specified, references to the numbering of specific amino acid residue positions in an antibody variable region are according to the Kabat numbering system.
  • The polypeptide sequences of the sequence listing are not numbered according to the Kabat numbering system. However, it is well within the ordinary skill of one in the art to convert the numbering of the sequences of the Sequence Listing to Kabat numbering.
  • “Framework” or “FR” refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.
  • The “class” of an antibody or immunoglobulin refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: 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 domains that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively.
  • The term “Fc domain” or “Fc region” herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. Although the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to extend from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991. A “subunit” of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association. For example, a subunit of an IgG Fc domain comprises an IgG CH2 and an IgG CH3 constant domain.
  • A “modification promoting the association of the first and the second subunit of the Fc domain” is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer. A modification promoting association as used herein particularly includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the modifications are complementary to each other so as to promote association of the two Fc domain subunits. For example, a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their association sterically or electrostatically favorable, respectively. Thus, (hetero)dimerization occurs between a polypeptide comprising the first Fc domain subunit and a polypeptide comprising the second Fc domain subunit, which might be non-identical in the sense that further components fused to each of the subunits (e.g. antigen binding moieties) are not the same. In some embodiments the modification promoting association comprises an amino acid mutation in the Fc domain, specifically an amino acid substitution. In a particular embodiment, the modification promoting association comprises a separate amino acid mutation, specifically an amino acid substitution, in each of the two subunits of the Fc domain.
  • The term “effector functions” refers to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor), and B cell activation.
  • As used herein, the terms “engineer, engineered, engineering”, are considered to include any manipulation of the peptide backbone or the post-translational modifications of a naturally occurring or recombinant polypeptide or fragment thereof. Engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches.
  • The term “amino acid mutation” as used herein is meant to encompass amino acid substitutions, deletions, insertions, and modifications. Any combination of substitution, deletion, insertion, and modification can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., reduced binding to an Fc receptor, or increased association with another peptide. Amino acid sequence deletions and insertions include amino- and/or carboxy-terminal deletions and insertions of amino acids. Particular amino acid mutations are amino acid substitutions. For the purpose of altering e.g. the binding characteristics of an Fc region, non-conservative amino acid substitutions, i.e. replacing one amino acid with another amino acid having different structural and/or chemical properties, are particularly preferred. Amino acid substitutions include replacement by non-naturally occurring amino acids or by naturally occurring amino acid derivatives of the twenty standard amino acids (e.g. 4-hydroxyproline, 3-methylhistidine, ornithine, homoserine, 5-hydroxylysine). Amino acid mutations can be generated using genetic or chemical methods well known in the art. Genetic methods may include site-directed mutagenesis, PCR, gene synthesis and the like. It is contemplated that methods of altering the side chain group of an amino acid by methods other than genetic engineering, such as chemical modification, may also be useful. Various designations may be used herein to indicate the same amino acid mutation. For example, a substitution from proline at position 329 of the Fc domain to glycine can be indicated as 329G, G329, G329, P329G, or Pro329Gly.
  • As used herein, term “polypeptide” refers to a molecule composed of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds). The term “polypeptide” refers to any chain of two or more amino acids, and does not refer to a specific length of the product. Thus, peptides, dipeptides, tripeptides, oligopeptides, “protein,” “amino acid chain,” or any other term used to refer to a chain of two or more amino acids, are included within the definition of “polypeptide,” and the term “polypeptide” may be used instead of, or interchangeably with any of these terms. The term “polypeptide” is also intended to refer to the products of post-expression modifications of the polypeptide, including without limitation glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids. A polypeptide may be derived from a natural biological source or produced by recombinant technology, but is not necessarily translated from a designated nucleic acid sequence. It may be generated in any manner, including by chemical synthesis. A polypeptide of the invention may be of a size of about 3 or more, 5 or more, 10 or more, 20 or more, 25 or more, 50 or more, 75 or more, 100 or more, 200 or more, 500 or more, 1,000 or more, or 2,000 or more amino acids. Polypeptides may have a defined three-dimensional structure, although they do not necessarily have such structure. Polypeptides with a defined three-dimensional structure are referred to as folded, and polypeptides which do not possess a defined three-dimensional structure, but rather can adopt a large number of different conformations, and are referred to as unfolded.
  • By an “isolated” polypeptide or a variant, or derivative thereof is intended a polypeptide that is not in its natural milieu. No particular level of purification is required. For example, an isolated polypeptide can be removed from its native or natural environment. Recombinantly produced polypeptides and proteins expressed in host cells are considered isolated for the purpose of the invention, as are native or recombinant polypeptides which have been separated, fractionated, or partially or substantially purified by any suitable technique.
  • “Percent (%) amino acid sequence identity” with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, Calif., or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary. In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:

  • 100 times the fraction X/Y
  • where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program.
  • The term “polynucleotide” refers to an isolated nucleic acid molecule or construct, e.g. messenger RNA (mRNA), virally-derived RNA, or plasmid DNA (pDNA). A polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond (e.g. an amide bond, such as found in peptide nucleic acids (PNA). The term “nucleic acid molecule” refers to any one or more nucleic acid segments, e.g. DNA or RNA fragments, present in a polynucleotide.
  • By “isolated” nucleic acid molecule or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment. For example, a recombinant polynucleotide encoding a polypeptide contained in a vector is considered isolated for the purposes of the present invention. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution. An isolated polynucleotide includes a polynucleotide molecule contained in cells that ordinarily contain the polynucleotide molecule, but the polynucleotide molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the present invention, as well as positive and negative strand forms, and double-stranded forms. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically. In addition, a polynucleotide or a nucleic acid may be or may include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator. By a nucleic acid or polynucleotide having a nucleotide sequence at least, for example, 95% “identical” to a reference nucleotide sequence of the present invention, it is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the 5′ or 3′ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence. As a practical matter, whether any particular polynucleotide sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be determined conventionally using known computer programs, such as the ones discussed above for polypeptides (e.g. ALIGN-2).
  • The term “expression cassette” refers to a polynucleotide generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a target cell. The recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus, or nucleic acid fragment. Typically, the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter. In certain embodiments, the expression cassette of the invention comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
  • The term “vector” or “expression vector” is synonymous with “expression construct” and refers to a DNA molecule that is used to introduce and direct the expression of a specific gene to which it is operably associated in a target cell. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. The expression vector of the present invention comprises an expression cassette. Expression vectors allow transcription of large amounts of stable mRNA. Once the expression vector is inside the target cell, the ribonucleic acid molecule or protein that is encoded by the gene is produced by the cellular transcription and/or translation machinery. In one embodiment, the expression vector of the invention comprises an expression cassette that comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
  • The terms “host cell”, “host cell line,” and “host cell culture” are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein. A host cell is any type of cellular system that can be used to generate the bispecific antigen binding molecules of the present invention. Host cells include cultured cells, e.g. mammalian cultured cells, such as CHO cells, BHK cells, NS0 cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
  • An “activating Fc receptor” is an Fc receptor that following engagement by an Fc domain of an antibody elicits signaling events that stimulate the receptor-bearing cell to perform effector functions. Human activating Fc receptors include FcγRIIIa (CD16a), FcγRI (CD64), FcγRIIa (CD32), and FcαRI (CD89).
  • Antibody-dependent cell-mediated cytotoxicity (ADCC) is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells. The target cells are cells to which antibodies or derivatives thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region. As used herein, the term “reduced ADCC” is defined as either a reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or an increase in the concentration of antibody in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC. The reduction in ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered. For example the reduction in ADCC mediated by an antibody comprising in its Fc domain an amino acid substitution that reduces ADCC, is relative to the ADCC mediated by the same antibody without this amino acid substitution in the Fc domain. Suitable assays to measure ADCC are well known in the art (see e.g. PCT publication no. WO 2006/082515 or PCT publication no. WO 2012/130831).
  • An “effective amount” of an agent refers to the amount that is necessary to result in a physiological change in the cell or tissue to which it is administered.
  • A “therapeutically effective amount” of an agent, e.g. a pharmaceutical composition, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. A therapeutically effective amount of an agent for example eliminates, decreases, delays, minimizes or prevents adverse effects of a disease.
  • An “individual” or “subject” is a mammal. Mammals include, but are not limited to, domesticated animals (e.g. cows, sheep, cats, dogs, and horses), primates (e.g. humans and non-human primates such as monkeys), rabbits, and rodents (e.g. mice and rats). Particularly, the individual or subject is a human.
  • The term “pharmaceutical composition” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical composition, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • As used herein, “treatment” (and grammatical variations thereof such as “treat” or “treating”) refers to clinical intervention in an attempt to alter the natural course of a disease in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, T cell activating bispecific antigen binding molecules of the invention are used to delay development of a disease or to slow the progression of a disease.
  • The term “antagonist” is used in the broadest sense, and includes any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity of a native polypeptide disclosed herein. In a similar manner, the term “agonist” is used in the broadest sense and includes any molecule that induces a biological activity of a native polypeptide disclosed herein. Suitable agonist or antagonist molecules specifically include agonist or antagonist antibodies or antibody fragments, including engineered antibody fragments, fragments or amino acid sequence variants of native polypeptides, peptides, antisense oligonucleotides, small organic molecules, etc. Methods for identifying agonists or antagonists of a polypeptide may comprise contacting a polypeptide with a candidate agonist or antagonist molecule and measuring a detectable change in one or more biological activities normally associated with the polypeptide.
  • The term “package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • All references, publication, patents and patent applications disclosed herein are hereby incorporated by reference in their entirety.
  • DETAILED DESCRIPTION OF THE EMBODIMENTS
  • The T cell activating bispecific antigen binding molecule of the invention is bispecific, i.e. it comprises at least two antigen binding moieties capable of specific binding to two distinct antigenic determinants, i.e. to CD3 and to FolR1. According to the invention, the antigen binding moieties are Fab molecules (i.e. antigen binding domains composed of a heavy and a light chain, each comprising a variable and a constant region). In one embodiment said Fab molecules are human. In another embodiment said Fab molecules are humanized. In yet another embodiment said Fab molecules comprise human heavy and light chain constant regions.
  • The T cell activating bispecific antigen binding molecule of the invention is capable of simultaneous binding to the target cell antigen FolR1 and CD3. In one embodiment, the T cell activating bispecific antigen binding molecule is capable of crosslinking a T cell and a FolR1 expressing target cell by simultaneous binding to the target cell antigen FolR1 and CD3. In an even more particular embodiment, such simultaneous binding results in lysis of the FolR1 expressing target cell, particularly a FolR1 expressing tumor cell. In one embodiment, such simultaneous binding results in activation of the T cell. In other embodiments, such simultaneous binding results in a cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. In one embodiment, binding of the T cell activating bispecific antigen binding molecule to CD3 without simultaneous binding to the target cell antigen FolR1 does not result in T cell activation.
  • In one embodiment, the T cell activating bispecific antigen binding molecule is capable of re-directing cytotoxic activity of a T cell to a FolR1 expressing target cell. In a particular embodiment, said re-direction is independent of MHC-mediated peptide antigen presentation by the target cell and and/or specificity of the T cell.
  • Particularly, a T cell according to some of the embodiments of the invention is a cytotoxic T cell. In some embodiments the T cell is a CD4+ or a CD8+ T cell, particularly a CD8+ T cell. The T cell activating bispecific antigen binding molecule of the invention comprises at least one antigen binding moiety capable of binding to CD3 (also referred to herein as an “CD3 antigen binding moiety” or “first antigen binding moiety”). In a particular embodiment, the T cell activating bispecific antigen binding molecule comprises not more than one antigen binding moiety capable of specific binding to CD3. In one embodiment the T cell activating bispecific antigen binding molecule provides monovalent binding to CD3. In a particular embodiment CD3 is human CD3 or cynomolgus CD3, most particularly human CD3. In a particular embodiment the CD3 antigen binding moiety is cross-reactive for (i.e. specifically binds to) human and cynomolgus CD3. In some embodiments, the first antigen binding moiety is capable of specific binding to the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP_000724.1, SEQ ID NO:150 for the human sequence; UniProt no. Q95LI5 (version 49), NCBI GenBank no. BAB71849.1, for the cynomolgus [Macaca fascicularis] sequence).
  • In some embodiments, the CD3 antigen binding moiety comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • In one embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • In one embodiment the CD3 antigen binding moiety comprises a variable heavy chain comprising an amino acid sequence of: SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of: SEQ ID NO: 31.
  • In one embodiment the CD3 antigen binding moiety comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31.
  • The T cell activating bispecific antigen binding molecule of the invention comprises at least one antigen binding moiety capable of binding to the target cell antigen FolR1 (also referred to herein as an “FolR1 binding moiety” or “second” or “third” antigen binding moiety). In one embodiment, the antigen binding moiety capable of binding to the target cell antigen FolR1 does not bind to FolR2 or FolR3. In a particular embodiment the FolR1 antigen binding moiety is cross-reactive for (i.e. specifically binds to) human and cynomolgus FolR1. In certain embodiments, the T cell activating bispecific antigen binding molecule comprises two antigen binding moieties capable of binding to the target cell antigen FolR1. In a particular such embodiment, each of these antigen binding moieties specifically binds to the same antigenic determinant. In an even more particular embodiment, all of these antigen binding moieties are identical. In one embodiment the T cell activating bispecific antigen binding molecule comprises not more than two antigen binding moieties capable of binding to FolR1.
  • The FolR1 binding moiety is generally a Fab molecule that specifically binds to FolR1 and is able to direct the T cell activating bispecific antigen binding molecule to which it is connected to a target site, for example to a specific type of tumor cell that expresses FolR1.
  • In one aspect the present invention provides a T cell activating bispecific antigen binding molecule comprising
      • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34; and
      • (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1).
  • In one embodiment the first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • In one embodiment the T cell activating bispecific antigen binding molecule additionally comprises
      • (iii) a third antigen binding moiety which is a Fab molecule capable of specific binding to FolR1.
  • In one such embodiment the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences. In one such embodiment the third antigen binding moiety is identical to the second antigen binding moiety.
  • In one embodiment the T cell activating bispecific antigen binding molecule of any of the above embodiments additionally comprises an Fc domain composed of a first and a second subunit capable of stable association.
  • In one embodiment the first antigen binding moiety and the second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.
  • In one embodiment the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, optionally via a peptide linker.
  • In a further particular embodiment, not more than one antigen binding moiety capable of specific binding to CD3 is present in the T cell activating bispecific antigen binding molecule (i.e. the T cell activating bispecific antigen binding molecule provides monovalent binding to CD3).
  • T Cell Activating Bispecific Antigen Binding Molecule with a Common Light Chain
  • The inventors of the present invention generated a bispecific antibody wherein the binding moieties share a common light chain that retains the specificity and efficacy of the parent monospecific antibody for CD3 and can bind a second antigen (e.g., FolR1) using the same light chain. The generation of a bispecific molecule with a common light chain that retains the binding properties of the parent antibody is not straight-forward as the common CDRs of the hybrid light chain have to effectuate the binding specificity for both targets. In one aspect the present invention provides a T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, one of which is a Fab molecule capable of specific binding to CD3 and the other one of which is a Fab molecule capable of specific binding to FolR1, wherein the first and the second Fab molecule have identical VLCL light chains. In one embodiment said identical light chain (VLCL) comprises the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34. In one embodiment said identical light chain (VLCL) comprises SEQ ID NO. 35.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • In one such embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 16, the heavy chain CDR2 of SEQ ID NO: 17, the heavy chain CDR3 of SEQ ID NO:18, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 275 and SEQ ID NO: 315 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises the heavy chain complementarity determining region (CDR) amino acid sequences of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39, and the light chain CDR amino acid sequences of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34;
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises the heavy chain complementarity determining region (CDR) amino acid sequences of SEQ ID NO: 16, SEQ ID NO: 275 and SEQ ID NO: 315, and the light chain CDR amino acid sequences of SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31;
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 274 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • In a further embodiment, the antigen binding moiety that is specific for FolR1 comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:15 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31 or variants thereof that retain functionality. In one embodiment the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:15, and a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31.
  • In one embodiment the T cell activating bispecific antigen binding molecule additionally comprises
  • (iii) a third antigen binding moiety (which is a Fab molecule) capable of specific binding to FolR1.
  • In one such embodiment the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences. In one such embodiment the third antigen binding moiety is identical to the second antigen binding moiety.
  • Hence in one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3, and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
    (iii) a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) and which comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34.
  • In one such embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 16, the heavy chain CDR2 of SEQ ID NO: 17, the heavy chain CDR3 of SEQ ID NO:18, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • In one embodiment the present invention provides a T cell activating bispecific antigen binding molecule comprising
  • (i) a first antigen binding moiety which is a Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (iii) a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
  • Thus, in one embodiment, the invention relates to bispecific molecules wherein at least two binding moieties have identical light chains and corresponding remodeled heavy chains that confer the specific binding to the T cell activating antigen CD3 and the target cell antigen FolR1, respectively. The use of this so-called ‘common light chain’ principle, i.e. combining two binders that share one light chain but still have separate specificities, prevents light chain mispairing. Thus, there are less side products during production, facilitating the homogenous preparation of T cell activating bispecific antigen binding molecules.
  • The components of the T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations are depicted in FIGS. 1A-I and are further described below.
  • In some embodiments, said T cell activating bispecific antigen binding molecule further comprises an Fc domain composed of a first and a second subunit capable of stable association. Below exemplary embodiments of T cell activating bispecific antigen binding molecule comprising an Fc domain are described.
  • T Cell Activating Bispecific Antigen Binding Molecule with a Crossover Fab Fragment
  • The inventors of the present invention generated a second bispecific antibody format wherein one of the binding moieties is a crossover Fab fragment. In one aspect of the invention a monovalent bispecific antibody is provided, wherein one of the Fab fragments of an IgG molecule is replaced by a crossover Fab fragment. Crossover Fab fragments are Fab fragments wherein either the variable regions or the constant regions of the heavy and light chain are exchanged. Bispecific antibody formats comprising crossover Fab fragments have been described, for example, in WO2009080252, WO2009080253, WO2009080251, WO2009080254, WO2010/136172, WO2010/145792 and WO2013/026831. In a particular embodiment, the first antigen binding moiety is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged. Such modification prevent mispairing of heavy and light chains from different Fab molecules, thereby improving the yield and purity of the T cell activating bispecific antigen binding molecule of the invention in recombinant production. In a particular crossover Fab molecule useful for the T cell activating bispecific antigen binding molecule of the invention, the variable regions of the Fab light chain and the Fab heavy chain are exchanged. In another crossover Fab molecule useful for the T cell activating bispecific antigen binding molecule of the invention, the constant regions of the Fab light chain and the Fab heavy chain are exchanged.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
  • In one such embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 56, the heavy chain CDR3 of SEQ ID NO:57, the light chain CDR1 of SEQ ID NO: 59, the light chain CDR2 of SEQ ID NO: 60, and the light chain CDR3 of SEQ ID NO:65.
  • In one embodiment, the second antigen binding moiety is a conventional Fab molecule.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • In one embodiment, the second antigen binding moiety is a conventional Fab molecule.
  • In a further embodiment, the antigen binding moiety that is specific for FolR1 comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:55 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 64 or variants thereof that retain functionality. In one embodiment the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:55, and a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 64.
  • In one embodiment the T cell activating bispecific antigen binding molecule additionally comprises
  • (iii) a third antigen binding moiety capable of specific binding to FolR1.
  • In one embodiment, the third antigen binding moiety is a conventional Fab molecule. In one embodiment, the third antigen binding moiety is a crossover Fab molecule.
  • In one such embodiment the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences. In one such embodiment the third antigen binding moiety is identical to the second antigen binding moiety.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
    (iii) a third antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 65.
  • In one such embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 56, the heavy chain CDR3 of SEQ ID NO:57, the light chain CDR1 of SEQ ID NO: 59, the light chain CDR2 of SEQ ID NO: 60, and the light chain CDR3 of SEQ ID NO:65.
  • In one embodiment, the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
    (iii) a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 64.
  • In one embodiment, the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 50 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
  • In one such embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • In one embodiment, the second antigen binding moiety is a conventional Fab molecule. In one embodiment, the second antigen binding moiety is a crossover Fab molecule.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • In one embodiment, the second antigen binding moiety is a conventional Fab molecule. In one embodiment, the second antigen binding moiety is a crossover Fab molecule.
  • In a further embodiment, the antigen binding moiety that is specific for FolR1 comprises a heavy chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:49 and a light chain variable region sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 51 or variants thereof that retain functionality.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31, a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:49, and a polypeptide sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 51.
  • In one embodiment the T cell activating bispecific antigen binding molecule additionally comprises
  • (iii) a third antigen binding moiety capable of specific binding to FolR1.
  • In one embodiment, the third antigen binding moiety is a conventional Fab molecule. In one embodiment, the second antigen binding moiety is a crossover Fab molecule.
  • In one such embodiment the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences. In one such embodiment the third antigen binding moiety is identical to the second antigen binding moiety.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34;
    (ii) a second antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 49 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
    (iii) a third antigen binding moiety capable of specific binding to Folate Receptor 1 (FolR1) comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9 and SEQ ID NO: 50 and at least one light chain CDR selected from the group of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54.
  • In one such embodiment the CD3 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and the FolR1 antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • In one embodiment, the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • In one embodiment the T cell activating bispecific antigen binding molecule comprises
  • (i) a first antigen binding moiety which is a crossover Fab molecule capable of specific binding to CD3 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 36 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 31.
    (ii) a second antigen binding moiety which is a Fab molecule capable of specific binding to Folate
  • Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • (iii) a third antigen binding moiety which is a Fab molecule capable of specific binding to Folate Receptor 1 (FolR1) comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising an amino acid sequence of SEQ ID NO: 51.
  • In one embodiment, the second antigen binding moiety and the third antigen binding moiety are both a conventional Fab molecule.
  • Thus, in one embodiment, the invention relates to bispecific molecules wherein two binding moieties confer specific binding to FolR1 and one binding moiety confers specificity to the T cell activating antigen CD3. One of the heavy chains is modified to ensure proper pairing of the heavy and light chains, thus eliminating the need for a common light chain approach. The presence of two FolR1 binding sites enables appropriate engagement with the target antigen FolR1 and the activation of T cells.
  • The components of the T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations are depicted in FIGS. 1A-I and are further described below.
  • In some embodiments, said T cell activating bispecific antigen binding molecule further comprises an Fc domain composed of a first and a second subunit capable of stable association. Below exemplary embodiments of T cell activating bispecific antigen binding molecule comprising an Fc domain are described.
  • T Cell Activating Bispecific Antigen Binding Molecule Formats
  • As depicted above and in FIGS. 1A-I, in one embodiment the T cell activating bispecific antigen binding molecules comprise at least two Fab fragments having identical light chains (VLCL) and having different heavy chains (VHCL) which confer the specificities to two different antigens, i.e. one Fab fragment is capable of specific binding to a T cell activating antigen CD3 and the other Fab fragment is capable of specific binding to the target cell antigen FolR1.
  • In another embodiment the T cell activating bispecific antigen binding molecule comprises at least two antigen binding moieties (Fab molecules), one of which is a crossover Fab molecule and one of which is a conventional Fab molecule. In one such embodiment the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • These components of the T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations are depicted in FIGS. 1A-I.
  • In some embodiments, the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain. In one such embodiment the first and second antigen binding moiety both are Fab fragments and have identical light chains (VLCL). In another such embodiment the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule.
  • In one embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain. In one such embodiment the first and second antigen binding moiety both are Fab fragments and have identical light chains (VLCL). In another such embodiment the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule. Optionally, the Fab light chain of the first antigen binding moiety and the Fab light chain of the second antigen binding moiety may additionally be fused to each other.
  • In other embodiments, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain. In a particular such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain. In one such embodiment the first and second antigen binding moiety both are Fab fragments and have identical light chains (VLCL). In another such embodiment the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule. Optionally, the Fab light chain of the first antigen binding moiety and the Fab light chain of the second antigen binding moiety may additionally be fused to each other.
  • The antigen binding moieties may be fused to the Fc domain or to each other directly or through a peptide linker, comprising one or more amino acids, typically about 2-20 amino acids. Peptide linkers are known in the art and are described herein. Suitable, non-immunogenic peptide linkers include, for example, (G4S)n (SEQ ID NO: 300), (SG4)n (SEQ ID NO: 301), (G4S)n (SEQ ID NO: 300) or G4(SG4)n (SEQ ID NO: 302) peptide linkers. “n” is generally a number between 1 and 10, typically between 2 and 4. A particularly suitable peptide linker for fusing the Fab light chains of the first and the second antigen binding moiety to each other is (G4S)2 (SEQ ID NO: 303). An exemplary peptide linker suitable for connecting the Fab heavy chains of the first and the second antigen binding moiety is EPKSC(D)-(G4S)2 (SEQ ID NOS 304 and 305). Additionally, linkers may comprise (a portion of) an immunoglobulin hinge region. Particularly where an antigen binding moiety is fused to the N-terminus of an Fc domain subunit, it may be fused via an immunoglobulin hinge region or a portion thereof, with or without an additional peptide linker.
  • It has been found by the inventors of the present invention that T cell activating bispecific antigen binding molecule comprising two binding moieties specific for the target cell antigen FolR have superior characteristics compared to T cell activating bispecific antigen binding molecule comprising only one binding moiety specific for the target cell antigen FolR.
  • Accordingly, in certain embodiments, the T cell activating bispecific antigen binding molecule of the invention further comprises a third antigen binding moiety which is a Fab molecule capable of specific binding to FolR. In one such embodiment the second and third antigen binding moiety capable of specific binding to FolR1 comprise identical heavy chain complementarity determining region (CDR) and light chain CDR sequences. In one such embodiment the third antigen binding moiety is identical to the second antigen binding moiety (i.e. they comprise the same amino acid sequences).
  • In one embodiment, the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first, a second and a third antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first and second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In one such embodiment the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL). In another such embodiment the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second and third antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule. Optionally, the Fab light chain of the first antigen binding moiety and the Fab light chain of the third antigen binding moiety may additionally be fused to each other.
  • Accordingly, in certain embodiments, the T cell activating bispecific antigen binding molecule of the invention comprises five polypeptide chains that form a first, a second and a third antigen binding moiety wherein the first antigen binding moiety is capable of binding CD3 and the second and the third antigen binding moiety each are capable of binding Folate Receptor 1 (FolR1). The first and the second polypeptide chain comprise, in amino (N)-terminal to carboxyl (C)-terminal direction, a first light chain variable domain (VLD1) and a first light chain constant domain (CLD1). The third polypeptide chain comprises, in N-terminal to C-terminal direction, second light chain variable domain (VLD2) and a second heavy chain constant domain 1 (CH1D2). The fourth polypeptide chain comprises, in N-terminal to C-terminal direction, a first heavy chain variable domain (VHD1), a first heavy chain constant domain 1 (CH1D1), a first heavy chain constant domain 2 (CH2D1) and a first heavy chain constant domain 3 (CH3D1). The fifth polypeptide chain comprises VHD1, CH1D1, a second heavy chain variable domain (VHD2), a second light chain constant domain (CLD2), a second heavy chain constant domain 2 (CH2D2) and a second heavy chain constant domain 3 (CH3D2). The third polypeptide chain and VHD2 and CLD2 of the fifth polypeptide chain form the first antigen binding moiety capable of binding CD3. The second polypeptide chain and VHD1 and CH1D1 of the fifth polypeptide chain form the third binding moiety capable of binding to FolR1. The first polypeptide chain and VHD1 and CH1D1 of the fourth polypeptide chain form the second binding moiety capable of binding to FolR1.
  • In another embodiment, the second and the third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first, a second and a third antigen binding moiety, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second and third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the third antigen binding moiety. In one such embodiment the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL). In another such embodiment the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule and the second and third antigen binding moiety capable of specific binding to FolR is a conventional Fab molecule. Optionally, the Fab light chain of the first antigen binding moiety and the Fab light chain of the second antigen binding moiety may additionally be fused to each other.
  • The antigen binding moieties may be fused to the Fc domain directly or through a peptide linker. In a particular embodiment the antigen binding moieties are each fused to the Fc domain through an immunoglobulin hinge region. In a specific embodiment, the immunoglobulin hinge region is a human IgG1 hinge region.
  • In one embodiment the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
  • In a particular embodiment said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, wherein the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL), wherein the first antigen binding moiety capable of specific binding to CD3 comprises at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.
  • In a particular embodiment said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, wherein the first, second and third antigen binding moiety are conventional Fab fragments and have identical light chains (VLCL), wherein the first antigen binding moiety capable of specific binding to CD3 comprises a variable heavy chain comprising a sequence of SEQ ID NO: 36, a variable light chain comprising a sequence of SEQ ID NO: 31; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise a variable heavy chain comprising a sequence of SEQ ID NO: 15, a variable light chain comprising a sequence of SEQ ID NO: 31.
  • In a particular embodiment said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety and the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged, comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39 and at least one light chain CDR selected from the group of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise at least one heavy chain complementarity determining region (CDR) selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56 and SEQ ID NO: 57 and at least one light chain CDR selected from the group of SEQ ID NO: 59, SEQ ID NO: 60 and SEQ ID NO: 65.
  • In a particular embodiment said T cell activating bispecific antigen binding molecule the first and the second antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, and the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety and the first antigen binding moiety capable of specific binding to CD3 is a crossover Fab molecule wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged, wherein the first antigen binding moiety capable of specific binding to CD3 comprises a variable heavy chain comprising a sequence of SEQ ID NO: 36, a variable light chain comprising a sequence of SEQ ID NO: 31; and the second and the third antigen binding moiety capable of specific binding to FolR1 comprise a variable heavy chain comprising a sequence of SEQ ID NO: 55, a variable light chain comprising a sequence of SEQ ID NO: 65.
  • In one embodiment the T cell activating bispecific antigen binding molecule is monovalent for each antigen. In a particular embodiment the T cell activating bispecific antigen binding molecule can bind to human CD3 and human folate receptor alpha (FolR1) and was made without employing a hetero-dimerization approach, such as, e.g., knob-into-hole technology. For example, the molecule can be produced by employing a common light chain library and CrossMab technology. In a particular embodiment, The variable region of the CD3 binder is fused to the CH1 domain of a standard human IgG1 antibody to form the VLVH crossed molecule (fused to Fc) which is common for both specificities. To generate the crossed counterparts (VHCL), a CD3 specific variable heavy chain domain is fused to a constant human λ light chain whereas a variable heavy chain domain specific for human FolR1 (e.g., isolated from a common light chain library) is fused to a constant human κ light chain. The resulting desired molecule with correctly paired chains comprises both kappa and lambda light chains or fragments thereof. Consequently, this desired bispecific molecule species can be purified from mispaired or homodimeric species with sequential purification steps selecting for kappa and lambda light chain, in either sequence. In one particular embodiment, purification of the desired bispecific antibody employs subsequent purification steps with KappaSelect and LambdaFabSelect columns (GE Healthcare) to remove undesired homodimeric antibodies.
  • Fc Domain
  • The Fc domain of the T cell activating bispecific antigen binding molecule consists of a pair of polypeptide chains comprising heavy chain domains of an immunoglobulin molecule. For example, the Fc domain of an immunoglobulin G (IgG) molecule is a dimer, each subunit of which comprises the CH2 and CH3 IgG heavy chain constant domains. The two subunits of the Fc domain are capable of stable association with each other. In one embodiment the T cell activating bispecific antigen binding molecule of the invention comprises not more than one Fc domain.
  • In one embodiment according the invention the Fc domain of the T cell activating bispecific antigen binding molecule is an IgG Fc domain. In a particular embodiment the Fc domain is an IgG1 Fc domain. In another embodiment the Fc domain is an IgG4 Fc domain. In a more specific embodiment, the Fc domain is an IgG4 Fc domain comprising an amino acid substitution at position S228 (Kabat numbering), particularly the amino acid substitution S228P. This amino acid substitution reduces in vivo Fab arm exchange of IgG4 antibodies (see Stubenrauch et al., Drug Metabolism and Disposition 38, 84-91 (2010)). In a further particular embodiment the Fc domain is human. An exemplary sequence of a human IgG1 Fc region is given in SEQ ID NO:245.
  • Fc Domain Modifications Promoting Heterodimerization
  • T cell activating bispecific antigen binding molecules according to the invention comprise different antigen binding moieties, fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised in two non-identical polypeptide chains. Recombinant co-expression of these polypeptides and subsequent dimerization leads to several possible combinations of the two polypeptides. To improve the yield and purity of T cell activating bispecific antigen binding molecules in recombinant production, it will thus be advantageous to introduce in the Fc domain of the T cell activating bispecific antigen binding molecule a modification promoting the association of the desired polypeptides.
  • Accordingly, in particular embodiments the Fc domain of the T cell activating bispecific antigen binding molecule according to the invention comprises a modification promoting the association of the first and the second subunit of the Fc domain. The site of most extensive protein-protein interaction between the two subunits of a human IgG Fc domain is in the CH3 domain of the Fc domain. Thus, in one embodiment said modification is in the CH3 domain of the Fc domain.
  • In a specific embodiment said modification is a so-called “knob-into-hole” modification, comprising a “knob” modification in one of the two subunits of the Fc domain and a “hole” modification in the other one of the two subunits of the Fc domain.
  • The knob-into-hole technology is described e.g. in U.S. Pat. No. 5,731,168; U.S. Pat. No. 7,695,936; Ridgway et al., Prot Eng 9, 617-621 (1996) and Carter, J Immunol Meth 248, 7-15 (2001). Generally, the method involves introducing a protuberance (“knob”) at the interface of a first polypeptide and a corresponding cavity (“hole”) in the interface of a second polypeptide, such that the protuberance can be positioned in the cavity so as to promote heterodimer formation and hinder homodimer formation. Protuberances are constructed by replacing small amino acid side chains from the interface of the first polypeptide with larger side chains (e.g. tyrosine or tryptophan). Compensatory cavities of identical or similar size to the protuberances are created in the interface of the second polypeptide by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). Accordingly, in a particular embodiment, in the CH3 domain of the first subunit of the Fc domain of the T cell activating bispecific antigen binding molecule an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.
  • The protuberance and cavity can be made by altering the nucleic acid encoding the polypeptides, e.g. by site-specific mutagenesis, or by peptide synthesis.
  • In a specific embodiment, in the CH3 domain of the first subunit of the Fc domain the threonine residue at position 366 is replaced with a tryptophan residue (T366W), and in the CH3 domain of the second subunit of the Fc domain the tyrosine residue at position 407 is replaced with a valine residue (Y407V). In one embodiment, in the second subunit of the Fc domain additionally the threonine residue at position 366 is replaced with a serine residue (T366S) and the leucine residue at position 368 is replaced with an alanine residue (L368A).
  • In yet a further embodiment, in the first subunit of the Fc domain additionally the serine residue at position 354 is replaced with a cysteine residue (S354C), and in the second subunit of the Fc domain additionally the tyrosine residue at position 349 is replaced by a cysteine residue (Y349C). Introduction of these two cysteine residues results in formation of a disulfide bridge between the two subunits of the Fc domain, thus further stabilizing the dimer (Carter, J Immunol Methods 248, 7-15 (2001)).
  • In a particular embodiment the antigen binding moiety capable of binding to CD3 is fused (optionally via the antigen binding moiety capable of binding to FolR1 on a target cell antigen) to the first subunit of the Fc domain (comprising the “knob” modification). Without wishing to be bound by theory, fusion of the antigen binding moiety capable of binding to CD3 to the knob-containing subunit of the Fc domain will (further) minimize the generation of antigen binding molecules comprising two antigen binding moieties capable of binding to CD3 (steric clash of two knob-containing polypeptides).
  • In an alternative embodiment a modification promoting association of the first and the second subunit of the Fc domain comprises a modification mediating electrostatic steering effects, e.g. as described in PCT publication WO 2009/089004. Generally, this method involves replacement of one or more amino acid residues at the interface of the two Fc domain subunits by charged amino acid residues so that homodimer formation becomes electrostatically unfavorable but heterodimerization electrostatically favorable.
  • Fc Domain Modifications Abolishing Fc Receptor Binding and/or Effector Function
  • The Fc domain confers to the T cell activating bispecific antigen binding molecule favorable pharmacokinetic properties, including a long serum half-life which contributes to good accumulation in the target tissue and a favorable tissue-blood distribution ratio. At the same time it may, however, lead to undesirable targeting of the T cell activating bispecific antigen binding molecule to cells expressing Fc receptors rather than to the preferred antigen-bearing cells. Moreover, the co-activation of Fc receptor signaling pathways may lead to cytokine release which, in combination with the T cell activating properties and the long half-life of the antigen binding molecule, results in excessive activation of cytokine receptors and severe side effects upon systemic administration. Activation of (Fc receptor-bearing) immune cells other than T cells may even reduce efficacy of the T cell activating bispecific antigen binding molecule due to the potential destruction of T cells e.g. by NK cells.
  • Accordingly, in particular embodiments the Fc domain of the T cell activating bispecific antigen binding molecules according to the invention exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain. In one such embodiment the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits less than 50%, preferably less than 20%, more preferably less than 10% and most preferably less than 5% of the binding affinity to an Fc receptor, as compared to a native IgG1 Fc domain (or a T cell activating bispecific antigen binding molecule comprising a native IgG1 Fc domain), and/or less than 50%, preferably less than 20%, more preferably less than 10% and most preferably less than 5% of the effector function, as compared to a native IgG1 Fc domain domain (or a T cell activating bispecific antigen binding molecule comprising a native IgG1 Fc domain). In one embodiment, the Fc domain domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) does not substantially bind to an Fc receptor and/or induce effector function. In a particular embodiment the Fc receptor is an Fcγ receptor. In one embodiment the Fc receptor is a human Fc receptor. In one embodiment the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fcγ receptor, more specifically human FcγRIIIa, FcγRI or FcγRIIa, most specifically human FcγRIIIa. In one embodiment the effector function is one or more selected from the group of CDC, ADCC, ADCP, and cytokine secretion. In a particular embodiment the effector function is ADCC. In one embodiment the Fc domain domain exhibits substantially similar binding affinity to neonatal Fc receptor (FcRn), as compared to a native IgG1 Fc domain domain. Substantially similar binding to FcRn is achieved when the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits greater than about 70%, particularly greater than about 80%, more particularly greater than about 90% of the binding affinity of a native IgG1 Fc domain (or the T cell activating bispecific antigen binding molecule comprising a native IgG1 Fc domain) to FcRn.
  • In certain embodiments the Fc domain is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain. In particular embodiments, the Fc domain of the T cell activating bispecific antigen binding molecule comprises one or more amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function. Typically, the same one or more amino acid mutation is present in each of the two subunits of the Fc domain. In one embodiment the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor. In one embodiment the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor by at least 2-fold, at least 5-fold, or at least 10-fold. In embodiments where there is more than one amino acid mutation that reduces the binding affinity of the Fc domain to the Fc receptor, the combination of these amino acid mutations may reduce the binding affinity of the Fc domain to an Fc receptor by at least 10-fold, at least 20-fold, or even at least 50-fold. In one embodiment the T cell activating bispecific antigen binding molecule comprising an engineered Fc domain exhibits less than 20%, particularly less than 10%, more particularly less than 5% of the binding affinity to an Fc receptor as compared to a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain. In a particular embodiment the Fc receptor is an Feγ receptor. In some embodiments the Fc receptor is a human Fc receptor. In some embodiments the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fcγ receptor, more specifically human FcγRIIIa, FcγRI or FcγRIIa, most specifically human FcγRIIIa. Preferably, binding to each of these receptors is reduced. In some embodiments binding affinity to a complement component, specifically binding affinity to C1q, is also reduced. In one embodiment binding affinity to neonatal Fc receptor (FcRn) is not reduced. Substantially similar binding to FcRn, i.e. preservation of the binding affinity of the Fc domain to said receptor, is achieved when the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits greater than about 70% of the binding affinity of a non-engineered form of the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said non-engineered form of the Fc domain) to FcRn. The Fc domain, or T cell activating bispecific antigen binding molecules of the invention comprising said Fc domain, may exhibit greater than about 80% and even greater than about 90% of such affinity. In certain embodiments the Fc domain of the T cell activating bispecific antigen binding molecule is engineered to have reduced effector function, as compared to a non-engineered Fc domain. The reduced effector function can include, but is not limited to, one or more of the following: reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen-presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced crosslinking of target-bound antibodies, reduced dendritic cell maturation, or reduced T cell priming. In one embodiment the reduced effector function is one or more selected from the group of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In a particular embodiment the reduced effector function is reduced ADCC. In one embodiment the reduced ADCC is less than 20% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain).
  • In one embodiment the amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function is an amino acid substitution. In one embodiment the Fc domain comprises an amino acid substitution at a position selected from the group of E233, L234, L235, N297, P331 and P329. In a more specific embodiment the Fc domain comprises an amino acid substitution at a position selected from the group of L234, L235 and P329. In some embodiments the Fc domain comprises the amino acid substitutions L234A and L235A. In one such embodiment, the Fc domain is an IgG1 Fc domain, particularly a human IgG1 Fc domain. In one embodiment the Fc domain comprises an amino acid substitution at position P329. In a more specific embodiment the amino acid substitution is P329A or P329G, particularly P329G. In one embodiment the Fc domain comprises an amino acid substitution at position P329 and a further amino acid substitution at a position selected from E233, L234, L235, N297 and P331. In a more specific embodiment the further amino acid substitution is E233P, L234A, L235A, L235E, N297A, N297D or P331S. In particular embodiments the Fc domain comprises amino acid substitutions at positions P329, L234 and L235. In more particular embodiments the Fc domain comprises the amino acid mutations L234A, L235A and P329G (“P329G LALA”). In one such embodiment, the Fc domain is an IgG1 Fc domain, particularly a human IgG1 Fc domain. The “P329G LALA” combination of amino acid substitutions almost completely abolishes Fcγ receptor binding of a human IgG1 Fc domain, as described in PCT publication no. WO 2012/130831, incorporated herein by reference in its entirety. WO 2012/130831 also describes methods of preparing such mutant Fc domains and methods for determining its properties such as Fc receptor binding or effector functions.
  • IgG4 antibodies exhibit reduced binding affinity to Fc receptors and reduced effector functions as compared to IgG1 antibodies. Hence, in some embodiments the Fc domain of the T cell activating bispecific antigen binding molecules of the invention is an IgG4 Fc domain, particularly a human IgG4 Fc domain. In one embodiment the IgG4 Fc domain comprises amino acid substitutions at position 5228, specifically the amino acid substitution S228P. To further reduce its binding affinity to an Fc receptor and/or its effector function, in one embodiment the IgG4 Fc domain comprises an amino acid substitution at position L235, specifically the amino acid substitution L235E. In another embodiment, the IgG4 Fc domain comprises an amino acid substitution at position P329, specifically the amino acid substitution P329G. In a particular embodiment, the IgG4 Fc domain comprises amino acid substitutions at positions S228, L235 and P329, specifically amino acid substitutions S228P, L235E and P329G. Such IgG4 Fc domain mutants and their Feγ receptor binding properties are described in PCT publication no. WO 2012/130831, incorporated herein by reference in its entirety.
  • In a particular embodiment the Fc domain exhibiting reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain, is a human IgG1 Fc domain comprising the amino acid substitutions L234A, L235A and optionally P329G, or a human IgG4 Fc domain comprising the amino acid substitutions S228P, L235E and optionally P329G.
  • In certain embodiments N-glycosylation of the Fc domain has been eliminated. In one such embodiment the Fc domain comprises an amino acid mutation at position N297, particularly an amino acid substitution replacing asparagine by alanine (N297A) or aspartic acid (N297D).
  • In addition to the Fc domains described hereinabove and in PCT publication no. WO 2012/130831, Fc domains with reduced Fc receptor binding and/or effector function also include those with substitution of one or more of Fc domain residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Pat. No. 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called “DANA” Fc mutant with substitution of residues 265 and 297 to alanine (U.S. Pat. No. 7,332,581).
  • Mutant Fc domains can be prepared by amino acid deletion, substitution, insertion or modification using genetic or chemical methods well known in the art. Genetic methods may include site-specific mutagenesis of the encoding DNA sequence, PCR, gene synthesis, and the like. The correct nucleotide changes can be verified for example by sequencing.
  • Binding to Fc receptors can be easily determined e.g. by ELISA, or by Surface Plasmon Resonance (SPR) using standard instrumentation such as a BIAcore instrument (GE Healthcare), and Fc receptors such as may be obtained by recombinant expression. A suitable such binding assay is described herein. Alternatively, binding affinity of Fc domains or cell activating bispecific antigen binding molecules comprising an Fc domain for Fc receptors may be evaluated using cell lines known to express particular Fc receptors, such as human NK cells expressing FcγIIIa receptor.
  • Effector function of an Fc domain, or a T cell activating bispecific antigen binding molecule comprising an Fc domain, can be measured by methods known in the art. A suitable assay for measuring ADCC is described herein. Other examples of in vitro assays to assess ADCC activity of a molecule of interest are described in U.S. Pat. No. 5,500,362; Hellstrom et al. Proc Natl Acad Sci USA 83, 7059-7063 (1986) and Hellstrom et al., Proc Natl Acad Sci USA 82, 1499-1502 (1985); U.S. Pat. No. 5,821,337; Bruggemann et al., J Exp Med 166, 1351-1361 (1987). Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, Calif.); and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, Wis.)). Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g. in a animal model such as that disclosed in Clynes et al., Proc Natl Acad Sci USA 95, 652-656 (1998).
  • In some embodiments, binding of the Fc domain to a complement component, specifically to C1q, is reduced. Accordingly, in some embodiments wherein the Fc domain is engineered to have reduced effector function, said reduced effector function includes reduced CDC. C1q binding assays may be carried out to determine whether the T cell activating bispecific antigen binding molecule is able to bind C1q and hence has CDC activity. See e.g., C1q and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J Immunol Methods 202, 163 (1996); Cragg et al., Blood 101, 1045-1052 (2003); and Cragg and Glennie, Blood 103, 2738-2743 (2004)).
  • Biological Properties and Functional Characteristics of T Cell Activating Bispecific Antigen Binding Molecules
  • One of skill in the art can appreciate the advantageous efficiency of a molecule that selectively distinguishes between cancerous and non-cancerous, healthy cells. One way to accomplish this goal is by appropriate target selection. Markers expressed exclusively on tumor cells can be employed to selectively target effector molecules or cells to tumor cells while sparing normal cells that do not express such marker. However, in some instances, so called tumor cell markers are also expressed in normal tissue, albeit at lower levels. This expression in normal tissue raises the possibility of toxicity. Thus, there was a need in the art for molecules that can more selectively target tumor cells. The invention described herein provides for T cell activating bispecific antigen binding molecules that selectively target FolR1-positive tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all. In one embodiment, the T cell activating bispecific antigen binding molecule comprises at least two, preferably two, FolR1 binding moieties of relatively low affinity that confer an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this embodiment of the invention selectively binds to, and/or induces killing of, tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all. In one embodiment, the T cell activating bispecific antigen binding molecule is in the 2+1 inverted format. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of FolR1-positive tumor cells and not non-tumor cells and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • In one specific embodiment, the T cell activating bispecific antigen binding molecule does not induce killing of a normal cells having less than about 1000 copies of FolR1 its surface.
  • In addition to the above advantageous characteristics, one embodiment of the invention does not require chemical cross linking or a hybrid approach to be produced. Accordingly, in one embodiment, the invention provides for T cell activating bispecific antigen binding molecule capable of production in CHO cells. In one embodiment, the T cell activating bispecific antigen binding molecule comprises humanized and human polypeptides. In one embodiment, the T cell activating bispecific antigen binding molecule does not cause FcgR crosslinking. In one such embodiment, the T cell activating bispecific antigen binding molecule is capable of production in CHO cells and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • As noted above, some embodiments contemplated herein include T cell activating bispecific antigen binding molecules having two binding moieties that confer specific binding to FolR1 and one binding moiety that confers specificity to the T cell activating antigen CD3, wherein each individual FolR1 binding moiety engages the antigen with low affinity. Because the molecule comprises two antigen binding moieties that confer binding to FolR1, the overall avidity of the molecule, nevertheless, provides effective binding to FolR1-expressing target cells and activation of T cells to induce T cell effector function. Considering that while FolR1 is expressed at various level on tumor cells, it is also expressed at very low levels (e.g., less than about 1000 copies on the cell surface) in certain normal cells, one of skill in the art can readily recognize the advantageous efficiency of such a molecule for use as a therapeutic agent. Such molecule selectively targets tumor cells over normal cells. Such molecule, thus, can be administered to an individual in need thereof with significantly less concern about toxicity resulting from FolR1 positive normal cells compared to molecules that bind to FolR1 with high affinity to induce effector function. In a preferred embodiment, the T cell activating bispecific antigen binding molecules have a monovalent binding affinity to huFolR1 in the micromolar range and an avidity to huFolR1 in the nanomolar range.
  • In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent KD of about 10 nM to about 40 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent KD of about 10 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent KD of about 10 nM and about 30 nM, respectively. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of at least about 1000 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of about 1400 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of about 1400 nM and to cynomolgus FolR1 with a monovalent binding KD of about 5600 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent KD of about 10 nM and with a monovalent binding KD of about 1400 nM.
  • In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with an apparent KD of about 5.36 pM to about 4 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent KD of about 4 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds murine FolR1 with an apparent KD of about 1.5 nM. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of at least about 1000 nM. In a specific embodiment, the T cell activating bispecific antigen binding molecule binds human and cynomolgus FolR1 with an apparent KD of about 4 nM, binds murine FolR1 with an apparent KD of about 1.5 nM, and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54. In one embodiment, the T cell activating bispecific antigen binding molecule binds human FolR1 with a monovalent binding KD of at least about 1000 nM and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • As described above, the T cell activating bispecific antigen binding molecules contemplated herein can induce T cell effector function, e.g., cell surface marker expression, cytokine production, T cell mediated killing. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing target cell, such as a human tumor cell, in vitro. In one embodiment, the T cell is a CD8 positive T cell. Examples of FolR1-expressing human tumor cells include but are not limited to Hela, Skov-3, HT-29, and HRCEpiC cells. Other FolR1 positive human cancer cells that can be used for in vitro testing are readily available to the skilled artisan. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing human tumor cell in vitro with an EC50 of between about 36 pM and about 39573 pM after 24 hours. Specifically contemplated are T cell activating bispecific antigen binding molecules that induce T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 36 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 178.4 pM after 24 hours. In one embodiment, the T cell activating bispecific antigen binding molecule induces T cell mediated killing of the FolR1-expressing tumor cell in vitro with an EC50 of about 134.5 pM or greater after 48 hours. The EC50 can be measure by methods known in the art, for example by methods disclosed herein by the examples.
  • In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments induces upregulation of cell surface expression of at least one of CD25 and CD69 on the T cell as measured by flow cytometry. In one embodiment, the T cell is a CD4 positive T cell or a CD8 positive T cell.
  • In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to FolR1 expressed on a human tumor cell. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments binds to a conformational epitope on human FolR1. In one embodiment, the T cell activating bispecific antigen binding molecule of any of the above embodiments does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3). In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the antigen binding moiety binds to a FolR1 polypeptide comprising the amino acids 25 to 234 of human FolR1 (SEQ ID NO:227). In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:227, to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:230 and to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 or 229. In one specific embodiment, the T cell activating bispecific antigen binding molecule comprises a FolR1 antigen binding moiety that binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NOs:227, 230 and 231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228 or 229, and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 8, the heavy chain CDR2 of SEQ ID NO: 9, the heavy chain CDR3 of SEQ ID NO:50, the light chain CDR1 of SEQ ID NO: 52, the light chain CDR2 of SEQ ID NO: 53, and the light chain CDR3 of SEQ ID NO:54.
  • In one embodiment of the T cell activating bispecific antigen binding molecule of any of the above embodiments, the FolR1 antigen binding moiety binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:227 and to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228, 229 or 230. In one specific embodiment, the T cell activating bispecific antigen binding molecule comprises a FolR1 antigen binding moiety that binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:227 and to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO:231, and wherein the FolR1 antigen binding moiety does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NOs:228, 229 or 230, and comprises a CD3 antigen binding moiety that comprises the heavy chain CDR1 of SEQ ID NO: 37, the heavy chain CDR2 of SEQ ID NO: 38, the heavy chain CDR3 of SEQ ID NO:39, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34 and two FolR1 antigen binding moieties that each comprise the heavy chain CDR1 of SEQ ID NO: 16, the heavy chain CDR2 of SEQ ID NO: 275, the heavy chain CDR3 of SEQ ID NO:315, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO:34.
  • With respect to the FolR1, the T cell activating bispecific antigen binding molecules contemplated herein can have agonist, antagonist or neutral effect. Examples of agonist effect include induction or enhancement of signaling through the FolR1 upon engagement by the FolR1 binding moiety with the FolR1 receptor on the target cell. Examples of antagonist activity include abrogation or reduction of signaling through the FolR1 upon engagement by the FolR1 binding moiety with the FolR1 receptor on the target cell. This can, for example, occur by blocking or reducing the interaction between folate with FolR1. Sequence variants of the embodiments disclosed herein having lower affinity while retaining the above described biological properties are specifically contemplated.
  • Immunoconjugates
  • The invention also pertains to immunoconjugates comprising a T cell activating bispecific antigen binding molecule conjugated to a cytotoxic agent such as a chemotherapeutic agent, a growth inhibitory agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • Polynucleotides
  • The invention further provides isolated polynucleotides encoding a T cell activating bispecific antigen binding molecule as described herein or a fragment thereof.
  • Polynucleotides of the invention include those that are at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequences set forth in SEQ ID NOs:151-226 including functional fragments or variants thereof.
  • The polynucleotides encoding T cell activating bispecific antigen binding molecules of the invention may be expressed as a single polynucleotide that encodes the entire T cell activating bispecific antigen binding molecule or as multiple (e.g., two or more) polynucleotides that are co-expressed. Polypeptides encoded by polynucleotides that are co-expressed may associate through, e.g., disulfide bonds or other means to form a functional T cell activating bispecific antigen binding molecule. For example, the light chain portion of an antigen binding moiety may be encoded by a separate polynucleotide from the portion of the T cell activating bispecific antigen binding molecule comprising the heavy chain portion of the antigen binding moiety, an Fc domain subunit and optionally (part of) another antigen binding moiety. When co-expressed, the heavy chain polypeptides will associate with the light chain polypeptides to form the antigen binding moiety. In another example, the portion of the T cell activating bispecific antigen binding molecule comprising one of the two Fc domain subunits and optionally (part of) one or more antigen binding moieties could be encoded by a separate polynucleotide from the portion of the T cell activating bispecific antigen binding molecule comprising the the other of the two Fc domain subunits and optionally (part of) an antigen binding moiety. When co-expressed, the Fc domain subunits will associate to form the Fc domain.
  • In some embodiments, the isolated polynucleotide encodes the entire T cell activating bispecific antigen binding molecule according to the invention as described herein. In other embodiments, the isolated polynucleotide encodes a polypeptides comprised in the T cell activating bispecific antigen binding molecule according to the invention as described herein.
  • In another embodiment, the present invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable region sequence as shown in SEQ ID NOs 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182 and 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223. In another embodiment, the present invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule or fragment thereof, wherein the polynucleotide comprises a sequence that encodes a polypeptide sequence as shown in SEQ ID NOs:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 1, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111,112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244. In another embodiment, the invention is further directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to a nucleotide sequence shown in SEQ ID NOs 97, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 12, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 246, 247. In another embodiment, the invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a nucleic acid sequence shown in SEQ ID NOs 97, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 12, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 246, 247. In another embodiment, the invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs 1, 2, 3, 4, 5, 6, 7, 11, 13, 15, 19, 21, 12, 25, 27, 29, 31, 36, 41, 45, 49, 51, 55, 58, 62, 64, 66, 68, 70, 72, 74, 76, 78, 82, 113, 114, 115, 116, 117, 118, 119, 12, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135. In another embodiment, the invention is directed to an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule or fragment thereof, wherein the polynucleotide comprises a sequence that encodes a polypeptide comprising one or more sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs: 8, 9, 50, 37, 38, and 39. The invention encompasses an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes the variable region sequence of SEQ ID NOs 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182 and 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223 with conservative amino acid substitutions. The invention also encompasses an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or fragment thereof, wherein the polynucleotide comprises a sequence that encodes the polypeptide sequence of SEQ ID NOs 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 1, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111,112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138 and 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244 with conservative amino acid substitutions.
  • In certain embodiments the polynucleotide or nucleic acid is DNA. In other embodiments, a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA (mRNA). RNA of the present invention may be single stranded or double stranded.
  • Recombinant Methods
  • T cell activating bispecific antigen binding molecules of the invention may be obtained, for example, by solid-state peptide synthesis (e.g. Merrifield solid phase synthesis) or recombinant production. For recombinant production one or more polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment), e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such polynucleotide may be readily isolated and sequenced using conventional procedures. In one embodiment a vector, preferably an expression vector, comprising one or more of the polynucleotides of the invention is provided. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the coding sequence of a T cell activating bispecific antigen binding molecule (fragment) along with appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al., MOLECULAR CLONING: A LABORATORY MANUAL, Cold Spring Harbor Laboratory, N.Y. (1989); and Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Greene Publishing Associates and Wiley Interscience, N.Y (1989). The expression vector can be part of a plasmid, virus, or may be a nucleic acid fragment. The expression vector includes an expression cassette into which the polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment) (i.e. the coding region) is cloned in operable association with a promoter and/or other transcription or translation control elements. As used herein, a “coding region” is a portion of nucleic acid which consists of codons translated into amino acids. Although a “stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, if present, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, 5′ and 3′ untranslated regions, and the like, are not part of a coding region. Two or more coding regions can be present in a single polynucleotide construct, e.g. on a single vector, or in separate polynucleotide constructs, e.g. on separate (different) vectors. Furthermore, any vector may contain a single coding region, or may comprise two or more coding regions, e.g. a vector of the present invention may encode one or more polypeptides, which are post- or co-translationally separated into the final proteins via proteolytic cleavage. In addition, a vector, polynucleotide, or nucleic acid of the invention may encode heterologous coding regions, either fused or unfused to a polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment) of the invention, or variant or derivative thereof. Heterologous coding regions include without limitation specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain. An operable association is when a coding region for a gene product, e.g. a polypeptide, is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s). Two DNA fragments (such as a polypeptide coding region and a promoter associated therewith) are “operably associated” if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not interfere with the ability of the expression regulatory sequences to direct the expression of the gene product or interfere with the ability of the DNA template to be transcribed. Thus, a promoter region would be operably associated with a nucleic acid encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid. The promoter may be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells. Other transcription control elements, besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription. Suitable promoters and other transcription control regions are disclosed herein. A variety of transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions, which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (e.g. the immediate early promoter, in conjunction with intron-A), simian virus 40 (e.g. the early promoter), and retroviruses (such as, e.g. Rous sarcoma virus). Other transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit a-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as inducible promoters (e.g. promoters inducible tetracyclins). Similarly, a variety of translation control elements are known to those of ordinary skill in the art. These include, but are not limited to ribosome binding sites, translation initiation and termination codons, and elements derived from viral systems (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence). The expression cassette may also include other features such as an origin of replication, and/or chromosome integration elements such as retroviral long terminal repeats (LTRs), or adeno-associated viral (AAV) inverted terminal repeats (ITRs).
  • Polynucleotide and nucleic acid coding regions of the present invention may be associated with additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide of the present invention. For example, if secretion of the T cell activating bispecific antigen binding molecule is desired, DNA encoding a signal sequence may be placed upstream of the nucleic acid encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof. According to the signal hypothesis, proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated. Those of ordinary skill in the art are aware that polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the translated polypeptide to produce a secreted or “mature” form of the polypeptide. In certain embodiments, the native signal peptide, e.g. an immunoglobulin heavy chain or light chain signal peptide is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it. Alternatively, a heterologous mammalian signal peptide, or a functional derivative thereof, may be used. For example, the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator (TPA) or mouse β-glucuronidase.
  • DNA encoding a short protein sequence that could be used to facilitate later purification (e.g. a histidine tag) or assist in labeling the T cell activating bispecific antigen binding molecule may be included within or at the ends of the T cell activating bispecific antigen binding molecule (fragment) encoding polynucleotide.
  • In a further embodiment, a host cell comprising one or more polynucleotides of the invention is provided. In certain embodiments a host cell comprising one or more vectors of the invention is provided. The polynucleotides and vectors may incorporate any of the features, singly or in combination, described herein in relation to polynucleotides and vectors, respectively. In one such embodiment a host cell comprises (e.g. has been transformed or transfected with) a vector comprising a polynucleotide that encodes (part of) a T cell activating bispecific antigen binding molecule of the invention. As used herein, the term “host cell” refers to any kind of cellular system which can be engineered to generate the T cell activating bispecific antigen binding molecules of the invention or fragments thereof. Host cells suitable for replicating and for supporting expression of T cell activating bispecific antigen binding molecules are well known in the art. Such cells may be transfected or transduced as appropriate with the particular expression vector and large quantities of vector containing cells can be grown for seeding large scale fermenters to obtain sufficient quantities of the T cell activating bispecific antigen binding molecule for clinical applications. Suitable host cells include prokaryotic microorganisms, such as E. coli, or various eukaryotic cells, such as Chinese hamster ovary cells (CHO), insect cells, or the like. For example, polypeptides may be produced in bacteria in particular when glycosylation is not needed. After expression, the polypeptide may be isolated from the bacterial cell paste in a soluble fraction and can be further purified. In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been “humanized”, resulting in the production of a polypeptide with a partially or fully human glycosylation pattern. See Gerngross, Nat Biotech 22, 1409-1414 (2004), and Li et al., Nat Biotech 24, 210-215 (2006). Suitable host cells for the expression of (glycosylated) polypeptides are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures can also be utilized as hosts. See e.g. U.S. Pat. Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293T cells as described, e.g., in Graham et al., J Gen Virol 36, 59 (1977)), baby hamster kidney cells (BHK), mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol Reprod 23, 243-251 (1980)), monkey kidney cells (CV1), African green monkey kidney cells (VERO-76), human cervical carcinoma cells (HELA), canine kidney cells (MDCK), buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver cells (Hep G2), mouse mammary tumor cells (MMT 060562), TRI cells (as described, e.g., in Mather et al., Annals N.Y. Acad Sci 383, 44-68 (1982)), MRC 5 cells, and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including dhfrCHO cells (Urlaub et al., Proc Natl Acad Sci USA 77, 4216 (1980)); and myeloma cell lines such as YO, NS0, P3X63 and Sp2/0. For a review of certain mammalian host cell lines suitable for protein production, see, e.g., Yazaki and Wu, Methods in Molecular Biology, Vol. 248 (B. K. C. Lo, ed., Humana Press, Totowa, N.J.), pp. 255-268 (2003). Host cells include cultured cells, e.g., mammalian cultured cells, yeast cells, insect cells, bacterial cells and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue. In one embodiment, the host cell is a eukaryotic cell, preferably a mammalian cell, such as a Chinese Hamster Ovary (CHO) cell, a human embryonic kidney (HEK) cell or a lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • Standard technologies are known in the art to express foreign genes in these systems. Cells expressing a polypeptide comprising either the heavy or the light chain of an antigen binding domain such as an antibody, may be engineered so as to also express the other of the antibody chains such that the expressed product is an antibody that has both a heavy and a light chain.
  • In one embodiment, a method of producing a T cell activating bispecific antigen binding molecule according to the invention is provided, wherein the method comprises culturing a host cell comprising a polynucleotide encoding the T cell activating bispecific antigen binding molecule, as provided herein, under conditions suitable for expression of the T cell activating bispecific antigen binding molecule, and recovering the T cell activating bispecific antigen binding molecule from the host cell (or host cell culture medium).
  • The components of the T cell activating bispecific antigen binding molecule are genetically fused to each other. T cell activating bispecific antigen binding molecule can be designed such that its components are fused directly to each other or indirectly through a linker sequence. The composition and length of the linker may be determined in accordance with methods well known in the art and may be tested for efficacy. Examples of linker sequences between different components of T cell activating bispecific antigen binding molecules are found in the sequences provided herein. Additional sequences may also be included to incorporate a cleavage site to separate the individual components of the fusion if desired, for example an endopeptidase recognition sequence.
  • In certain embodiments the one or more antigen binding moieties of the T cell activating bispecific antigen binding molecules comprise at least an antibody variable region capable of binding an antigenic determinant. Variable regions can form part of and be derived from naturally or non-naturally occurring antibodies and fragments thereof. Methods to produce polyclonal antibodies and monoclonal antibodies are well known in the art (see e.g. Harlow and Lane, “Antibodies, a laboratory manual”, Cold Spring Harbor Laboratory, 1988). Non-naturally occurring antibodies can be constructed using solid phase-peptide synthesis, can be produced recombinantly (e.g. as described in U.S. Pat. No. 4,186,567) or can be obtained, for example, by screening combinatorial libraries comprising variable heavy chains and variable light chains (see e.g. U.S. Pat. No. 5,969,108, McCafferty).
  • Any animal species of antibody, antibody fragment, antigen binding domain or variable region can be used in the T cell activating bispecific antigen binding molecules of the invention. Non-limiting antibodies, antibody fragments, antigen binding domains or variable regions useful in the present invention can be of murine, primate, or human origin. If the T cell activating bispecific antigen binding molecule is intended for human use, a chimeric form of antibody may be used wherein the constant regions of the antibody are from a human. A humanized or fully human form of the antibody can also be prepared in accordance with methods well known in the art (see e.g. U.S. Pat. No. 5,565,332 to Winter). Humanization may be achieved by various methods including, but not limited to (a) grafting the non-human (e.g., donor antibody) CDRs onto human (e.g. recipient antibody) framework and constant regions with or without retention of critical framework residues (e.g. those that are important for retaining good antigen binding affinity or antibody functions), (b) grafting only the non-human specificity-determining regions (SDRs or a-CDRs; the residues critical for the antibody-antigen interaction) onto human framework and constant regions, or (c) transplanting the entire non-human variable domains, but “cloaking” them with a human-like section by replacement of surface residues. Humanized antibodies and methods of making them are reviewed, e.g., in Almagro and Fransson, Front Biosci 13, 1619-1633 (2008), and are further described, e.g., in Riechmann et al., Nature 332, 323-329 (1988); Queen et al., Proc Natl Acad Sci USA 86, 10029-10033 (1989); U.S. Pat. Nos. 5,821,337, 7,527,791, 6,982,321, and 7,087,409; Jones et al., Nature 321, 522-525 (1986); Morrison et al., Proc Natl Acad Sci 81, 6851-6855 (1984); Morrison and Oi, Adv Immunol 44, 65-92 (1988); Verhoeyen et al., Science 239, 1534-1536 (1988); Padlan, Molec Immun 31(3), 169-217 (1994); Kashmiri et al., Methods 36, 25-34 (2005) (describing SDR (a-CDR) grafting); Padlan, Mol Immunol 28, 489-498 (1991) (describing “resurfacing”); Dall'Acqua et al., Methods 36, 43-60 (2005) (describing “FR shuffling”); and Osbourn et al., Methods 36, 61-68 (2005) and Klimka et al., Br J Cancer 83, 252-260 (2000) (describing the “guided selection” approach to FR shuffling). Human antibodies and human variable regions can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr Opin Pharmacol 5, 368-74 (2001) and Lonberg, Curr Opin Immunol 20, 450-459 (2008). Human variable regions can form part of and be derived from human monoclonal antibodies made by the hybridoma method (see e.g. Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)). Human antibodies and human variable regions may also be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge (see e.g. Lonberg, Nat Biotech 23, 1117-1125 (2005). Human antibodies and human variable regions may also be generated by isolating Fv clone variable region sequences selected from human-derived phage display libraries (see e.g., Hoogenboom et al. in Methods in Molecular Biology 178, 1-37 (O'Brien et al., ed., Human Press, Totowa, N.J., 2001); and McCafferty et al., Nature 348, 552-554; Clackson et al., Nature 352, 624-628 (1991)). Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • In certain embodiments, the antigen binding moieties useful in the present invention are engineered to have enhanced binding affinity according to, for example, the methods disclosed in U.S. Pat. Appl. Publ. No. 2004/0132066, the entire contents of which are hereby incorporated by reference. The ability of the T cell activating bispecific antigen binding molecule of the invention to bind to a specific antigenic determinant can be measured either through an enzyme-linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance technique (analyzed on a BIACORE T100 system) (Liljeblad, et al., Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)). Competition assays may be used to identify an antibody, antibody fragment, antigen binding domain or variable domain that competes with a reference antibody for binding to a particular antigen, e.g. an antibody that competes with the V9 antibody for binding to CD3. In certain embodiments, such a competing antibody binds to the same epitope (e.g. a linear or a conformational epitope) that is bound by the reference antibody. Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) “Epitope Mapping Protocols,” in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, N.J.). In an exemplary competition assay, immobilized antigen (e.g. CD3) is incubated in a solution comprising a first labeled antibody that binds to the antigen (e.g. V9 antibody, described in U.S. Pat. No. 6,054,297) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to the antigen. The second antibody may be present in a hybridoma supernatant. As a control, immobilized antigen is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to the antigen, excess unbound antibody is removed, and the amount of label associated with immobilized antigen is measured. If the amount of label associated with immobilized antigen is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to the antigen. See Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.).
  • T cell activating bispecific antigen binding molecules prepared as described herein may be purified by art-known techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography, and the like. The actual conditions used to purify a particular protein will depend, in part, on factors such as net charge, hydrophobicity, hydrophilicity etc., and will be apparent to those having skill in the art. For affinity chromatography purification an antibody, ligand, receptor or antigen can be used to which the T cell activating bispecific antigen binding molecule binds. For example, for affinity chromatography purification of T cell activating bispecific antigen binding molecules of the invention, a matrix with protein A or protein G may be used. Sequential Protein A or G affinity chromatography and size exclusion chromatography can be used to isolate a T cell activating bispecific antigen binding molecule essentially as described in the Examples. The purity of the T cell activating bispecific antigen binding molecule can be determined by any of a variety of well known analytical methods including gel electrophoresis, high pressure liquid chromatography, and the like. For example, the heavy chain fusion proteins expressed as described in the Examples were shown to be intact and properly assembled as demonstrated by reducing SDS-PAGE (see e.g. FIG. 2). Three bands were resolved at approximately Mr 25,000, Mr 50,000 and Mr 75,000, corresponding to the predicted molecular weights of the T cell activating bispecific antigen binding molecule light chain, heavy chain and heavy chain/light chain fusion protein.
  • Assays
  • T cell activating bispecific antigen binding molecules provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
  • Affinity Assays
  • The affinity of the T cell activating bispecific antigen binding molecule for an Fc receptor or a target antigen can be determined in accordance with the methods set forth in the Examples by surface plasmon resonance (SPR), using standard instrumentation such as a BIAcore instrument (GE Healthcare), and receptors or target proteins such as may be obtained by recombinant expression. Alternatively, binding of T cell activating bispecific antigen binding molecules for different receptors or target antigens may be evaluated using cell lines expressing the particular receptor or target antigen, for example by flow cytometry (FACS). A specific illustrative and exemplary embodiment for measuring binding affinity is described in the following and in the Examples below. According to one embodiment, KD is measured by surface plasmon resonance using a BIACORE® T100 machine (GE Healthcare) at 25° C.
  • To analyze the interaction between the Fc-portion and Fc receptors, His-tagged recombinant Fc-receptor is captured by an anti-Penta His antibody (“Penta His” disclosed as SEQ ID NO: 306) (Qiagen) immobilized on CMS chips and the bispecific constructs are used as analytes. Briefly, carboxymethylated dextran biosensor chips (CMS, GE Healthcare) are activated with N-ethyl-N′-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Anti Penta-His antibody (“Penta-His” disclosed as SEQ ID NO: 306) is diluted with 10 mM sodium acetate, pH 5.0, to 40 μg/ml before injection at a flow rate of 5 μl/min to achieve approximately 6500 response units (RU) of coupled protein. Following the injection of the ligand, 1 M ethanolamine is injected to block unreacted groups. Subsequently the Fc-receptor is captured for 60 s at 4 or 10 nM. For kinetic measurements, four-fold serial dilutions of the bispecific construct (range between 500 nM and 4000 nM) are injected in HBS-EP (GE Healthcare, 10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% Surfactant P20, pH 7.4) at 25° C. at a flow rate of 30 μl/min for 120 s.
  • To determine the affinity to the target antigen, bispecific constructs are captured by an anti human Fab specific antibody (GE Healthcare) that is immobilized on an activated CM5-sensor chip surface as described for the anti Penta-His antibody (“Penta-His” disclosed as SEQ ID NO: 306). The final amount of coupled protein is is approximately 12000 R U. The bispecific constructs are captured for 90 s at 300 nM. The target antigens are passed through the flow cells for 180 s at a concentration range from 250 to 1000 nM with a flowrate of 30 μl/min. The dissociation is monitored for 180 s. Bulk refractive index differences are corrected for by subtracting the response obtained on reference flow cell. The steady state response was used to derive the dissociation constant KD by non-linear curve fitting of the Langmuir binding isotherm. Association rates (kon) and dissociation rates (koff) are calculated using a simple one-to-one Langmuir binding model (BIACORE® T100 Evaluation Software version 1.1.1) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) is calculated as the ratio koff/kon. See, e.g., Chen et al., J Mol Biol 293, 865-881 (1999).
  • Activity Assays
  • Biological activity of the T cell activating bispecific antigen binding molecules of the invention can be measured by various assays as described in the Examples. Biological activities may for example include the induction of proliferation of T cells, the induction of signaling in T cells, the induction of expression of activation markers in T cells, the induction of cytokine secretion by T cells, the induction of lysis of target cells such as tumor cells, and the induction of tumor regression and/or the improvement of survival.
  • Compositions, Formulations, and Routes of Administration
  • In a further aspect, the invention provides pharmaceutical compositions comprising any of the T cell activating bispecific antigen binding molecules provided herein, e.g., for use in any of the below therapeutic methods. In one embodiment, a pharmaceutical composition comprises any of the T cell activating bispecific antigen binding molecules provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical composition comprises any of the T cell activating bispecific antigen binding molecules provided herein and at least one additional therapeutic agent, e.g., as described below.
  • Further provided is a method of producing a T cell activating bispecific antigen binding molecule of the invention in a form suitable for administration in vivo, the method comprising (a) obtaining a T cell activating bispecific antigen binding molecule according to the invention, and (b) formulating the T cell activating bispecific antigen binding molecule with at least one pharmaceutically acceptable carrier, whereby a preparation of T cell activating bispecific antigen binding molecule is formulated for administration in vivo.
  • Pharmaceutical compositions of the present invention comprise a therapeutically effective amount of one or more T cell activating bispecific antigen binding molecule dissolved or dispersed in a pharmaceutically acceptable carrier. The phrases “pharmaceutical or pharmacologically acceptable” refers to molecular entities and compositions that are generally non-toxic to recipients at the dosages and concentrations employed, i.e. do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate. The preparation of a pharmaceutical composition that contains at least one T cell activating bispecific antigen binding molecule and optionally an additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference. Moreover, for animal (e.g., human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards or corresponding authorities in other countries. Preferred compositions are lyophilized formulations or aqueous solutions. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, buffers, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g. antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, antioxidants, proteins, drugs, drug stabilizers, polymers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329, incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
  • The composition may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection. T cell activating bispecific antigen binding molecules of the present invention (and any additional therapeutic agent) can be administered intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrasplenically, intrarenally, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, intratumorally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, topically, locally, by inhalation (e.g. aerosol inhalation), injection, infusion, continuous infusion, localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g. liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference). Parenteral administration, in particular intravenous injection, is most commonly used for administering polypeptide molecules such as the T cell activating bispecific antigen binding molecules of the invention.
  • Parenteral compositions include those designed for administration by injection, e.g. subcutaneous, intradermal, intralesional, intravenous, intraarterial intramuscular, intrathecal or intraperitoneal injection. For injection, the T cell activating bispecific antigen binding molecules of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer. The solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the T cell activating bispecific antigen binding molecules may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. Sterile injectable solutions are prepared by incorporating the T cell activating bispecific antigen binding molecules of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated below, as required. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and/or the other ingredients. In the case of sterile powders for the preparation of sterile injectable solutions, suspensions or emulsion, the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered liquid medium thereof. The liquid medium should be suitably buffered if necessary and the liquid diluent first rendered isotonic prior to injection with sufficient saline or glucose. The composition must be stable under the conditions of manufacture and storage, and preserved against the contaminating action of microorganisms, such as bacteria and fungi. It will be appreciated that endotoxin contamination should be kept minimally at a safe level, for example, less that 0.5 ng/mg protein. Suitable pharmaceutically acceptable carriers include, but are not limited to: 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 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 polyethylene glycol (PEG). Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl cleats or triglycerides, or liposomes.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) 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's Pharmaceutical Sciences (18th Ed. Mack Printing Company, 1990). Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide, which matrices are in the form of shaped articles, e.g. films, or microcapsules. In particular embodiments, prolonged absorption of an injectable composition can be brought about by the use in the compositions of agents delaying absorption, such as, for example, aluminum monostearate, gelatin or combinations thereof.
  • In addition to the compositions described previously, the T cell activating bispecific antigen binding molecules may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the T cell activating bispecific antigen binding molecules may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • Pharmaceutical compositions comprising the T cell activating bispecific antigen binding molecules of the invention may be manufactured by means of conventional mixing, dissolving, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the proteins into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • The T cell activating bispecific antigen binding molecules may be formulated into a composition in a free acid or base, neutral or salt form. Pharmaceutically acceptable salts are salts that substantially retain the biological activity of the free acid or base. These include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
  • Therapeutic Methods and Compositions
  • Any of the T cell activating bispecific antigen binding molecules provided herein may be used in therapeutic methods. T cell activating bispecific antigen binding molecules of the invention can be used as immunotherapeutic agents, for example in the treatment of cancers.
  • For use in therapeutic methods, T cell activating bispecific antigen binding molecules of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • In one aspect, T cell activating bispecific antigen binding molecules of the invention for use as a medicament are provided. In further aspects, T cell activating bispecific antigen binding molecules of the invention for use in treating a disease are provided. In certain embodiments, T cell activating bispecific antigen binding molecules of the invention for use in a method of treatment are provided. In one embodiment, the invention provides a T cell activating bispecific antigen binding molecule as described herein for use in the treatment of a disease in an individual in need thereof. In certain embodiments, the invention provides a T cell activating bispecific antigen binding molecule for use in a method of treating an individual having a disease comprising administering to the individual a therapeutically effective amount of the T cell activating bispecific antigen binding molecule. In certain embodiments the disease to be treated is a proliferative disorder. In a particular embodiment the disease is cancer. In certain embodiments the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer. In further embodiments, the invention provides a T cell activating bispecific antigen binding molecule as described herein for use in inducing lysis of a target cell, particularly a tumor cell. In certain embodiments, the invention provides a T cell activating bispecific antigen binding molecule for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the T cell activating bispecific antigen binding molecule to induce lysis of a target cell. An “individual” according to any of the above embodiments is a mammal, preferably a human.
  • In a further aspect, the invention provides for the use of a T cell activating bispecific antigen binding molecule of the invention in the manufacture or preparation of a medicament. In one embodiment the medicament is for the treatment of a disease in an individual in need thereof. In a further embodiment, the medicament is for use in a method of treating a disease comprising administering to an individual having the disease a therapeutically effective amount of the medicament. In certain embodiments the disease to be treated is a proliferative disorder. In a particular embodiment the disease is cancer. In one embodiment, the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer. In a further embodiment, the medicament is for inducing lysis of a target cell, particularly a tumor cell. In still a further embodiment, the medicament is for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the medicament to induce lysis of a target cell. An “individual” according to any of the above embodiments may be a mammal, preferably a human.
  • In a further aspect, the invention provides a method for treating a disease. In one embodiment, the method comprises administering to an individual having such disease a therapeutically effective amount of a T cell activating bispecific antigen binding molecule of the invention. In one embodiment a composition is administered to said individual, comprising the T cell activating bispecific antigen binding molecule of the invention in a pharmaceutically acceptable form. In certain embodiments the disease to be treated is a proliferative disorder. In a particular embodiment the disease is cancer. In certain embodiments the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer. An “individual” according to any of the above embodiments may be a mammal, preferably a human.
  • In a further aspect, the invention provides a method for inducing lysis of a target cell, particularly a tumor cell. In one embodiment the method comprises contacting a target cell with a T cell activating bispecific antigen binding molecule of the invention in the presence of a T cell, particularly a cytotoxic T cell. In a further aspect, a method for inducing lysis of a target cell, particularly a tumor cell, in an individual is provided. In one such embodiment, the method comprises administering to the individual an effective amount of a T cell activating bispecific antigen binding molecule to induce lysis of a target cell. In one embodiment, an “individual” is a human.
  • In certain embodiments the disease to be treated is a proliferative disorder, particularly cancer. Non-limiting examples of cancers include bladder cancer, brain cancer, head and neck cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, uterine cancer, cervical cancer, endometrial cancer, esophageal cancer, colon cancer, colorectal cancer, rectal cancer, gastric cancer, prostate cancer, blood cancer, skin cancer, squamous cell carcinoma, bone cancer, and kidney cancer. Other cell proliferation disorders that can be treated using a T cell activating bispecific antigen binding molecule of the present invention include, but are not limited to neoplasms located in the: abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic region, and urogenital system. Also included are pre-cancerous conditions or lesions and cancer metastases. In certain embodiments the cancer is chosen from the group consisting of renal cell cancer, skin cancer, lung cancer, colorectal cancer, breast cancer, brain cancer, head and neck cancer. A skilled artisan readily recognizes that in many cases the T cell activating bispecific antigen binding molecule may not provide a cure but may only provide partial benefit. In some embodiments, a physiological change having some benefit is also considered therapeutically beneficial. Thus, in some embodiments, an amount of T cell activating bispecific antigen binding molecule that provides a physiological change is considered an “effective amount” or a “therapeutically effective amount”. The subject, patient, or individual in need of treatment is typically a mammal, more specifically a human.
  • In some embodiments, an effective amount of a T cell activating bispecific antigen binding molecule of the invention is administered to a cell. In other embodiments, a therapeutically effective amount of a T cell activating bispecific antigen binding molecule of the invention is administered to an individual for the treatment of disease.
  • For the prevention or treatment of disease, the appropriate dosage of a T cell activating bispecific antigen binding molecule of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the route of administration, the body weight of the patient, the type of T cell activating bispecific antigen binding molecule, the severity and course of the disease, whether the T cell activating bispecific antigen binding molecule is administered for preventive or therapeutic purposes, previous or concurrent therapeutic interventions, the patient's clinical history and response to the T cell activating bispecific antigen binding molecule, and the discretion of the attending physician. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject. Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • The T cell activating bispecific antigen binding molecule is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1 mg/kg-10 mg/kg) of T cell activating bispecific antigen binding molecule can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the T cell activating bispecific antigen binding molecule would be in the range from about 0.005 mg/kg to about 10 mg/kg. In other non-limiting examples, a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10 milligram/kg body weight, about 50 milligram/kg body weight, about 100 milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram/kg body weight to about 500 milligram/kg body weight, etc., can be administered, based on the numbers described above. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the T cell activating bispecific antigen binding molecule). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • The T cell activating bispecific antigen binding molecules of the invention will generally be used in an amount effective to achieve the intended purpose. For use to treat or prevent a disease condition, the T cell activating bispecific antigen binding molecules of the invention, or pharmaceutical compositions thereof, are administered or applied in a therapeutically effective amount. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • For systemic administration, a therapeutically effective dose can be estimated initially from in vitro assays, such as cell culture assays. A dose can then be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the T cell activating bispecific antigen binding molecules which are sufficient to maintain therapeutic effect. Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day. Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
  • In cases of local administration or selective uptake, the effective local concentration of the T cell activating bispecific antigen binding molecules may not be related to plasma concentration. One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
  • A therapeutically effective dose of the T cell activating bispecific antigen binding molecules described herein will generally provide therapeutic benefit without causing substantial toxicity. Toxicity and therapeutic efficacy of a T cell activating bispecific antigen binding molecule can be determined by standard pharmaceutical procedures in cell culture or experimental animals. Cell culture assays and animal studies can be used to determine the LD50 (the dose lethal to 50% of a population) and the ED50 (the dose therapeutically effective in 50% of a population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50. T cell activating bispecific antigen binding molecules that exhibit large therapeutic indices are preferred. In one embodiment, the T cell activating bispecific antigen binding molecule according to the present invention exhibits a high therapeutic index. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans. The dosage lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al., 1975, in: The Pharmacological Basis of Therapeutics, Ch. 1, p. 1, incorporated herein by reference in its entirety). The attending physician for patients treated with T cell activating bispecific antigen binding molecules of the invention would know how and when to terminate, interrupt, or adjust administration due to toxicity, organ dysfunction, and the like. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.
  • Other Agents and Treatments
  • The T cell activating bispecific antigen binding molecules of the invention may be administered in combination with one or more other agents in therapy. For instance, a T cell activating bispecific antigen binding molecule of the invention may be co-administered with at least one additional therapeutic agent. The term “therapeutic agent” encompasses any agent administered to treat a symptom or disease in an individual in need of such treatment. Such additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. In certain embodiments, an additional therapeutic agent is an immunomodulatory agent, a cytostatic agent, an inhibitor of cell adhesion, a cytotoxic agent, an activator of cell apoptosis, or an agent that increases the sensitivity of cells to apoptotic inducers. In a particular embodiment, the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.
  • Such other agents are suitably present in combination in amounts that are effective for the purpose intended. The effective amount of such other agents depends on the amount of T cell activating bispecific antigen binding molecule used, the type of disorder or treatment, and other factors discussed above. The T cell activating bispecific antigen binding molecules are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate compositions), and separate administration, in which case, administration of the T cell activating bispecific antigen binding molecule of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. T cell activating bispecific antigen binding molecules of the invention can also be used in combination with radiation therapy.
  • In another aspect, the invention provides for a bispecific antibody comprising a) a first antigen-binding site that comprises a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31; and b) a second antigen-binding site that comprises a variable heavy chain domain (VH) of SEQ ID NO: 36 and a variable light chain domain of SEQ ID NO: 31 for use in combination with an antibody to PD-L1 or FAP-4-1BBL. In one embodiment, the bispecific antibody further comprises a third antigen-binding site that comprises a variable heavy chain domain (VH) of SEQ ID NO: 274 and a variable light chain domain of SEQ ID NO: 31.
  • Articles of Manufacture
  • In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and 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). At least one active agent in the composition is a T cell activating bispecific antigen binding molecule of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a T cell activating bispecific antigen binding molecule of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • EXAMPLES
  • The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.
  • General Methods
  • Recombinant DNA Techniques
  • Standard methods were used to manipulate DNA as described in Sambrook et al., Molecular cloning: A laboratory manual; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. The molecular biological reagents were used according to the manufacturers' instructions. General information regarding the nucleotide sequences of human immunoglobulins light and heavy chains is given in: Kabat, E. A. et al., (1991) Sequences of Proteins of Immunological Interest, 5th ed., NIH Publication No. 91-3242.
  • DNA Sequencing
  • DNA sequences were determined by standard double strand sequencing at Synergene (Schlieren).
  • Gene Synthesis
  • Desired gene segments where required were either generated by PCR using appropriate templates or were synthesized by Geneart AG (Regensburg, Germany) from synthetic oligonucleotides and PCR products by automated gene synthesis. In cases where no exact gene sequence was available, oligonucleotide primers were designed based on sequences from closest homologues and the genes were isolated by RT-PCR from RNA originating from the appropriate tissue. The gene segments flanked by singular restriction endonuclease cleavage sites were cloned into standard cloning/sequencing vectors. The plasmid DNA was purified from transformed bacteria and concentration determined by UV spectroscopy. The DNA sequence of the subcloned gene fragments was confirmed by DNA sequencing. Gene segments were designed with suitable restriction sites to allow sub-cloning into the respective expression vectors. All constructs were designed with a 5′-end DNA sequence coding for a leader peptide which targets proteins for secretion in eukaryotic cells.
  • Isolation of Primary Human Pan T Cells from PBMCs
  • Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation from enriched lymphocyte preparations (buffy coats) obtained from local blood banks or from fresh blood from healthy human donors. Briefly, blood was diluted with sterile PBS and carefully layered over a Histopaque gradient (Sigma, H8889). After centrifugation for 30 minutes at 450×g at room temperature (brake switched off), part of the plasma above the PBMC containing interphase was discarded. The PBMCs were transferred into new 50 ml Falcon tubes and tubes were filled up with PBS to a total volume of 50 ml. The mixture was centrifuged at room temperature for 10 minutes at 400×g (brake switched on). The supernatant was discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps at 4° C. for 10 minutes at 350×g). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium, containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in the incubator until assay start. T cell enrichment from PBMCs was performed using the Pan T Cell Isolation Kit II (Miltenyi Biotec #130-091-156), according to the manufacturer's instructions. Briefly, the cell pellets were diluted in 40 μl cold buffer per 10 million cells (PBS with 0.5% BSA, 2 mM EDTA, sterile filtered) and incubated with 10 μl Biotin-Antibody Cocktail per 10 million cells for 10 min at 4° C. 30 μl cold buffer and 20 μl Anti-Biotin magnetic beads per 10 million cells were added, and the mixture incubated for another 15 min at 4° C. Cells were washed by adding 10-20× the current volume and a subsequent centrifugation step at 300×g for 10 min. Up to 100 million cells were resuspended in 500 μl buffer. Magnetic separation of unlabeled human pan T cells was performed using LS columns (Miltenyi Biotec #130-042-401) according to the manufacturer's instructions. The resulting T cell population was counted automatically (ViCell) and stored in AIM-V medium at 37° C., 5% CO2 in the incubator until assay start (not longer than 24 h).
  • Isolation of Primary Human Naive T Cells from PBMCs
  • Peripheral blood mononuclar cells (PBMCs) were prepared by Histopaque density centrifugation from enriched lymphocyte preparations (buffy coats) obtained from local blood banks or from fresh blood from healthy human donors. T-cell enrichment from PBMCs was performed using the Naive CD8+ T cell isolation Kit from Miltenyi Biotec (#130-093-244), according to the manufacturer's instructions, but skipping the last isolation step of CD8+ T cells (also see description for the isolation of primary human pan T cells).
  • Isolation of Murine Pan T Cells from Splenocytes
  • Spleens were isolated from C57BL/6 mice, transferred into a GentleMACS C-tube (Miltenyi Biotech #130-093-237) containing MACS buffer (PBS+0.5% BSA+2 mM EDTA) and dissociated with the GentleMACS Dissociator to obtain single-cell suspensions according to the manufacturer's instructions. The cell suspension was passed through a pre-separation filter to remove remaining undissociated tissue particles. After centrifugation at 400×g for 4 min at 4° C., ACK Lysis Buffer was added to lyse red blood cells (incubation for 5 min at room temperature). The remaining cells were washed with MACS buffer twice, counted and used for the isolation of murine pan T cells. The negative (magnetic) selection was performed using the Pan T Cell Isolation Kit from Miltenyi Biotec (#130-090-861), following the manufacturer's instructions. The resulting T cell population was automatically counted (ViCell) and immediately used for further assays.
  • Isolation of Primary Cynomolgus PBMCs from Heparinized Blood
  • Peripheral blood mononuclar cells (PBMCs) were prepared by density centrifugation from fresh blood from healthy cynomolgus donors, as follows: Heparinized blood was diluted 1:3 with sterile PBS, and Lymphoprep medium (Axon Lab #1114545) was diluted to 90% with sterile PBS. Two volumes of the diluted blood were layered over one volume of the diluted density gradient and the PBMC fraction was separated by centrifugation for 30 min at 520×g, without brake, at room temperature. The PBMC band was transferred into a fresh 50 ml Falcon tube and washed with sterile PBS by centrifugation for 10 min at 400×g at 4° C. One low-speed centrifugation was performed to remove the platelets (15 min at 150×g, 4° C.), and the resulting PBMC population was automatically counted (ViCell) and immediately used for further assays.
  • Example 1 Purification of Biotinylated Folate Receptor-Fc Fusions
  • To generate new antibodies against human FolR1 the following antigens and screening tools were generated as monovalent Fc fusion proteins (the extracellular domain of the antigen linked to the hinge region of Fc-knob which is co-expressed with an Fc-hole molecule). The antigen genes were synthesized (Geneart, Regensburg, Germany) based on sequences obtained from GenBank or SwissProt and inserted into expression vectors to generate fusion proteins with Fc-knob with a C-terminal Avi-tag for in vivo or in vitro biotinylation. In vivo biotinylation was achieved by co-expression of the bacterial birA gene encoding a bacterial biotin ligase during production. Expression of all genes was under control of a chimeric MPSV promoter on a plasmid containing an oriP element for stable maintenance of the plasmids in EBNA containing cell lines.
  • For preparation of the biotinylated monomeric antigen/Fc fusion molecules, exponentially growing suspension HEK293 EBNA cells were co-transfected with three vectors encoding the two components of fusion protein (knob and hole chains) as well as BirA, an enzyme necessary for the biotinylation reaction. The corresponding vectors were used at a 9.5:9.5:1 ratio (“antigen ECD-Fc knob-avi tag”:“Fc hole”:“BirA”).
  • For protein production in 500 ml shake flasks, 400 million HEK293 EBNA cells were seeded 24 hours before transfection. For transfection cells were centrifuged for 5 minutes at 210 g, and supernatant was replaced by pre-warmed CD CHO medium. Expression vectors were resuspended in 20 mL of CD CHO medium containing 200 μg of vector DNA. After addition of 540 μL of polyethylenimine (PEI), the solution was mixed for 15 seconds and incubated for 10 minutes at room temperature. Afterwards, cells were mixed with the DNA/PEI solution, transferred to a 500 mL shake flask and incubated for 3 hours at 37° C. in an incubator with a 5% CO2 atmosphere. After the incubation, 160 mL of F17 medium was added and cells were cultured for 24 hours. One day after transfection, 1 mM valproic acid and 7% Feed 1 (Lonza) were added to the culture. The production medium was also supplemented with 100 μM biotin. After 7 days of culturing, the cell supernatant was collected by spinning down cells for 15 min at 210 g. The solution was sterile filtered (0.22 μm filter), supplemented with sodium azide to a final concentration of 0.01% (w/v), and kept at 4° C.
  • Secreted proteins were purified from cell culture supernatants by affinity chromatography using Protein A, followed by size exclusion chromatography. For affinity chromatography, the supernatant was loaded on a HiTrap ProteinA HP column (CV=5 mL, GE Healthcare) equilibrated with 40 mL 20 mM sodium phosphate, 20 mM sodium citrate pH 7.5. Unbound protein was removed by washing with at least 10 column volumes of 20 mM sodium phosphate, 20 mM sodium citrate pH 7.5. The bound protein was eluted using a linear pH-gradient created over 20 column volumes of 20 mM sodium citrate, 100 mM sodium chloride, 100 mM glycine, pH 3.0. The column was then washed with 10 column volumes of 20 mM sodium citrate, 100 mM sodium chloride, 100 mM glycine, pH 3.0. pH of collected fractions was adjusted by adding 1/10 (v/v) of 0.5 M sodium phosphate, pH 8.0. The protein was concentrated and filtered prior to loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM histidine, 140 mM sodium chloride, pH 6.0.
  • The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the FolR1-Fc-fusion was analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). The aggregate content of samples was analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Purified antigen-Fc-fusion proteins were analyzed by surface plasmon resonance assays using commercially available antibodies to confirm correct and natural conformation of the antigens (data not shown).
  • TABLE 1
    Antigens produced for isolation, selection and counter
    selection of human FolR1 antibodies
    ECD Accession Seq ID
    Antigen (aa) number Sequence No
    human 25-234 P15328 RIAWARTELLNVCMNAKHHKEKPGPEDKLHEQCRPWR 227
    FolR1 KNACCSTNTSQEAHKDVSYLYRFNWNHCGEMAPACKR
    HFIQDTCLYECSPNLGPWIQQVDQSWRKERVLNVPLC
    KEDCEQWWEDCRTSYTCKSNWHKGWNWTSGFNKCAVG
    AACQPFHFYFPTPTVLCNEIWTHSYKVSNYSRGSGRC
    IQMWFDPAQGNPNEEVARFYAAAM
    human 17-230 P14207 TMCSAQDRTDLLNVCMDAKHHKTKPGPEDKLHDQCSP 228
    FolR2 WKKNACCTASTSQELHKDTSRLYNFNWDHCGKMEPAC
    KRHFIQDTCLYECSPNLGPWIQQVNQSWRKERFLDVP
    LCKEDCQRWWEDCHTSHTCKSNWHRGWDWTSGVNKCP
    AGALCRTFESYFPTPAALCEGLWSHSYKVSNYSRGSG
    RCIQMWFDSAQGNPNEEVARFYAAAMHVN
    human 24-243 P41439 SARARTDLLNVCMNAKHHKTQPSPEDELYGQCSPWKK 229
    FolR3 NACCTASTSQELHKDTSRLYNFNWDHCGKMEPTCKRH
    FIQDSCLYECSPNLGPWIRQVNQSWRKERILNVPLCK
    EDCERWWEDCRTSYTCKSNWHKGWNWTSGINECPAGA
    LCSTFESYFPTPAALCEGLWSHSFKVSNYSRGSGRCI
    QMWFDSAQGNPNEEVAKFYAAAMNAGAPSRGIIDS
    murine 25-232 P35846 TRARTELLNVCMDAKHHKEKPGPEDNLHDQCSPWKTN 230
    FolR1 SCCSTNTSQEAHKDISYLYRFNWNHCGTMTSECKRHF
    IQDTCLYECSPNLGPWIQQVDQSWRKERILDVPLCKE
    DCQQWWEDCQSSFTCKSNWHKGWNWSSGHNECPVGAS
    CHPFTFYFPTSAALCEEIWSHSYKLSNYSRGSGRCIQ
    MWFDPAQGNPNEEVARFYAEAMS
    cynomolgus 25-234 G7PR14 EAQTRTARARTELLNVCMNAKHHKEKPGPEDKLHEQC 231
    FolR1 RPWKKNACCSTNTSQEAHKDVSYLYRFNWNHCGEMAP
    ACKRHFIQDTCLYECSPNLGPWIQQVDQSWRKERVLN
    VPLCKEDCERWWEDCRTSYCKSNWHKGWNWTSGFNKC
    PVGAACQPFHFYFPTPTVLCNEIWTYSYKVSNYSRGS
    GRCIQMWFDPAQGNPNEEVARFYAAAMS
  • TABLE 2
    Summary of the yield and final monomer
    content of the FolR-Fc-fusions.
    Monomer
    [%]
    Antigen (SEC) Yield
    huFolR1
    100 30 mg/L
    cyFolR1
    100 32 mg/L
    muFolR1
    100 31 mg/L
    huFolR2
    100 16 mg/L
    huFolR3 95 38 mg/L
  • Example 2 Generation of Common Light Chain with CD3ε Specificity
  • The T cell activating bispecific molecules described herein comprise at least one CD3 binding moiety. This moiety can be generated by immunizing laboratory animals, screening phage library or using known anti-CD3 antibodies. The common light chain with CD3c specificity was generated by humanizing the light chain of a murine parental anti-CD3c antibody (CH2527). For humanization of an antibody of non-human origin, the CDR residues from the non-human antibody (donor) have to be transplanted onto the framework of a human (acceptor) antibody. Generally, acceptor framework sequences are selected by aligning the sequence of the donor to a collection of potential acceptor sequences and choosing one that has either reasonable homology to the donor, or shows similar amino acids at some positions critical for structure and activity. In the present case, the search for the antibody acceptor framework was performed by aligning the mouse VL-domain sequence of the parental antibody to a collection of human germline sequences and choosing the human sequence that showed high sequence identity. Surprisingly, a good match in terms of framework sequence homology was found in a rather infrequent human light chain belonging to the V-domain family 7 of the lambda type, more precisely, hVL_7_46 (IMGT nomenclature, GenBank Acc No. Z73674). This infrequent human light chain was subsequently chosen as acceptor framework for humanization of the light chain of CH2527. The three complementarity determining regions (CDRs) of the mouse light chain variable domain were grafted onto this acceptor framework. Since the framework 4 region is not part of the variable region of the germline V-gene, the alignment for this region (J-element) was done individually. Hence the IGLJ3-02 sequence was chosen for humanization of this light chain.
  • Thirteen humanized variants were generated (CH2527-VL7_46-1 to VL7_46-10, VL7_46-12 to VL7_46-14). These differ in framework residues (and combinations thereof) that were back-mutated to the murine V-domain sequence or in CDR-residues (Kabat definition) that could be kept identical to the human germline sequence. The following framework residues outside the CDRs were back-mutated to the murine residues in the final humanized VL-domain variant VL7_46-13 (murine residues listed): V36, E38, F44, G46, G49, and G57, respectively. The human J-element IGLJ3-02 was 100% identical to the J-element of the murine parental antibody.
  • Example 3 SPR Assessment of Humanized Variants with CD3ε Specificity
  • Humanized VL variants were assessed as chimera in a 2+1 classical format (FIG. 1D), i.e. humanized light chain V-domains were paired with murine heavy chain V-domains. SPR assessment was carried out on a ProteOn XPR36 instrument (Bio-Rad). More precisely, the variants were captured directly from the culture supernatant on an anti-Fab derivatized GLM sensorchip (Goat Anti-Human IgG, F(ab′)2 Fragment Specific, Jackson ImmunoResearch) in vertical orientation. The following analytes were subsequently injected horizontally as single concentrations to assess binding to human and cynomolgus CD3ε: 3 μM hu CD3ε(−1-26)-Fc(knob)-avi (ID807) and 2.5 μM cy CD3ε-(−1-26)-Fc(knob)-Avi-Fc(hole) (ID873), respectively. Binding responses were qualitatively compared to binding of the murine control construct and graded+(comparable binding observed), +/−(reduced binding observed) and −(no binding observed). The capture antibody was regenerated after each cycle of ligand capture and analyte binding and the murine construct was re-injected at the end of the study to confirm the activity of the capture surface. The results are summarized in Table 3.
  • TABLE 3
    Qualitative binding assessment based on SPR for the humanized
    light chain variants combined with the murine heavy chain of
    CH2527. Only the humanized light chain variant that was finally
    chosen, CH2527-VL7_46-13, highlighted in bold letters, exhibited
    comparable binding to human and cynomolgus CD3ε.
    humanized VL variant binding to CD3ε
    murine_CH2527-VL +
    CH2527-VL7_46-1
    CH2527-VL7_46-2
    CH2527-VL7_46-3
    CH2527-VL7_46-4
    CH2527-VL7_46-5
    CH2527-VL7_46-6
    CH2527-VL7_46-7
    CH2527-VL7_46-8
    CH2527-VL7_46-9
    CH2527-VL7_46-10
    CH2527-VL7_46-12 +/−
    CH2527-VL7_46-13 +
    CH2527-VL7_46-14
  • Example 4 Properties of Humanized Common Light Chain with CD3ε Specificity
  • The light chain V-domain variant that was chosen for the humanized lead molecule is VL7_46-13. The degree of humanness, i.e. the sequence homology of the humanized V-domain to the human germline V-domain sequence was determined. For VL7_46-13, the overall sequence identity with the closest human germline homolog is 65% before humanization and 80% afterwards. Omitting the CDR regions, the sequence identity is 92% to the closest human germline homolog. As can be seen from Table 3, VL7_46-13 is the only humanized VL variant out of a panel of 13 variants that showed comparable binding to the parental murine antibody and also retained its cross-reactivity to cynomolgus CD3c. This result indicates that it was not trivial to humanize the murine VL-domain without losing binding affinity to CD3c which required several back-mutations to murine framework residues (in particular G46) while retaining G24 in CDR1. In addition, this result shows that the VL-domain plays a crucial role in target recognition. Importantly, the humanized VL-domain VL7_46-13 based on an infrequent human germline belonging to the V-domain family 7 of the lambda type and retaining affinity and specificity for CD3c, is also suitable to be used as a common light chain in phage-displayed antibody libraries of the Fab-format and enables successful selection for novel specificities which greatly facilitates the generation and production of bispecific molecules binding to CD3ε and e.g. a tumor target and sharing the same ‘common’ light chain.
  • Example 5 Generation of a Phage Displayed Antibody Library Using a Human Germ-Line Common Light Chain Derived from HVK1-39
  • Several approaches to generate bispecific antibodies that resemble full length human IgG utilize modifications in the Fc region that induce heterodimerization of two distinct heavy chains. Such examples include knobs-into-holes (Merchant et al., Nat Biotechnol. 1998 July; 16(7):677-81) SEED (Davis et al., Protein Eng Des Sel. 2010 April; 23(4):195-202) and electrostatic steering technologies (Gunasekaran et al., J Biol Chem. 2010 Jun. 18; 285(25):19637-46). Although these approaches enable effective heterodimerization of two distinct heavy chains, appropriate pairing of cognate light and heavy chains remains a problem. Usage of a common light chain (LC) can solve this issue (Merchant, et al. Nat Biotech 16, 677-681 (1998)).
  • Here, we describe the generation of an antibody library for the display on a M13 phage. Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain. This library is designed for generating multispecific antibodies without the need to use sophisticated technologies to avoid light chain mispairing.
  • By using a common light chain the production of these molecules can be facilitated as no mispairing occurs any longer and the isolation of a highly pure bispecific antibody is facilitated. As compared to other formats the use of Fab fragments as building blocks as opposed to e.g. the use of scFv fragments results in higher thermal stability and the lack of scFv aggregation and intermolecular scFv formation.
  • Library Generation
  • In the following the generation of an antibody library for the display on M13 phage is described. Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain.
  • We used these heavy chains in the library (GenBank Accession Numbers in brackets):
  • IGHV1-46*01 (X92343) (SEQ ID NO: 104),
    IGHV1-69*06 (L22583), (SEQ ID NO: 105)
    IGHV3-15*01 (X92216), (SEQ ID NO: 106)
    IGHV3-23*01 (M99660), (SEQ ID NO: 107)
    IGHV4-59*01 (AB019438), (SEQ ID NO: 108)
    IGHV5-51*01 (M99686), (SEQ ID NO: 109)
  • All heavy chains use the IGHJ2 as J-element, except the IGHV1-69*06 which uses IGHJ6 sequence. The design of the randomization included the CDR-H1, CDR-H2, and CDR-H3. For CDR-H1 and CDR-H2 a “soft” randomization strategy was chosen, and the randomization oligonucleotides were such that the codon for the amino acid of the germ-line sequence was present at 50%. All other amino acids, except cysteine, were summing up for the remaining 50%. In CDR-H3, where no germ-line amino acid is present due to the presence of the genetic D-element, oligonucleotides were designed that allow for the usage of randomized inserts between the V-element and the J-element of 4 to 9 amino acids in length. Those oligonucleotides contained in their randomized part e.g. The three amino acids G/Y/S are present to 15% each, those amino acids A/D/T/R/P/L/V/N/W/F/I/E are present to 4,6% each.
  • Exemplary methods for generation of antibody libraries are described in Hoogenboom et al., Nucleic Acids Res. 1991, 19, 4133-413; Lee et., al J. Mol. Biol. (2004) 340, 1073-1093.
  • The light chain is derived from the human sequence hVK1-39, and is used in an unmodified and non-randomized fashion. This will ensure that the same light chain can be used for other projects without additional modifications.
  • Exemplary Library Selection:
  • Selections with all affinity maturation libraries are carried out in solution according to the following procedure using a monomeric and biotinylated extracellular domain of a target antigen X.
  • 1. 10̂12 phagemid particles of each library are bound to 100 nM biotinylated soluble antigen for 0.5 h in a total volume of 1 ml. 2. Biotinylated antigen is captured and specifically bound phage particles are isolated by addition of ˜5×10̂7 streptavidin-coated magnetic beads for 10 min. 3. Beads are washed using 5-10×1 ml PBS/Tween20 and 5-10×1 ml PBS. 4. Elution of phage particles is done by addition of 1 ml 100 mM TEA (triethylamine) for 10 min and neutralization by addition of 500 ul 1 M Tris/HCl pH 7.4 and 5. Re-infection of exponentially growing E. coli TG1 bacteria, infection with helper phage VCSM13 and subsequent PEG/NaCl precipitation of phagemid particles is applied in subsequent selection rounds. Selections are carried out over 3-5 rounds using either constant or decreasing (from 10̂−7 M to 2×10̂−9M) antigen concentrations. In round 2, capture of antigen/phage complexes is performed using neutravidin plates instead of streptavidin beads. All binding reactions are supplemented either with 100 nM bovine serum albumin, or with non-fat milk powder in order to compete for unwanted clones arising from mere sticky binding of the antibodies to the plastic support.
  • Selections are being carried out over three or four rounds using decreasing antigen concentrations of the antigen starting from 100 nM and going down to 5 nM in the final selection round. Specific binders are defined as signals ca. 5× higher than background and are identified by ELISA. Specific binders are identified by ELISA as follows: 100 μl of 10 nM biotinylated antigen per well are coated on neutravidin plates. Fab-containing bacterial supernatants are added and binding Fabs are detected via their Flag-tags by using an anti-Flag/HRP secondary antibody. ELISA-positive clones are bacterially expressed as soluble Fab fragments in 96-well format and supernatants are subjected to a kinetic screening experiment by SPR-analysis using ProteOn XPR36 (BioRad). Clones expressing Fabs with the highest affinity constants are identified and the corresponding phagemids are sequenced. For further characterization, the Fab sequences are amplified via PCR from the phagemid and cloned via appropriate restriction sites into human IgG1 expression vectors for mammalian production.
  • Generation of a Phage Displayed Antibody Library Using a Humanized CD3ε Specific Common Light Chain
  • Here, the generation of an antibody library for the display on M13 phage is described. Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain. This library was designed for the generation of Fc-containing, but FcgR binding inactive T cell bispecific antibodies of IgG1 P329G LALA or IgG4 SPLE PG isotype in which one or two Fab recognize a tumor surface antigen expressed on a tumor cell whereas the remaining Fab arm of the antibody recognizes CD3e on a T cell.
  • Library Generation
  • In the following the generation of an antibody library for the display on M13 phage is described. Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain. This library is designed solely for the generation of Fc-containing, but FcgR binding inactive T cell bispecific antibodies of IgG1 P329G LALA or IgG4 SPLE PG isotype. Diversity was introduced via randomization oligonucleotides only in the CDR3 of the different heavy chains. Methods for generation of antibody libraries are well known in the art and are described in (Hoogenboom et al., Nucleic Acids Res. 1991, 19, 4133-413; or in: Lee et., al J. Mol. Biol. (2004) 340, 1073-1093).
  • We used these heavy chains in the library:
  • IGHV1-46*01 (X92343), (SEQ ID NO: 104)
    IGHV1-69*06 (L22583), (SEQ ID NO: 105)
    IGHV3-15*01 (X92216), (SEQ ID NO: 106)
    IGHV3-23*01 (M99660), (SEQ ID NO: 107)
    IGHV4-59*01 (AB019438), (SEQ ID NO: 108)
    IGHV5-51*01 (M99686), (SEQ ID NO: 109)
  • We used the light chain derived from the humanized human and Cynomolgus CD3 ε specific antibody CH2527 in the library: (VL7_46-13; SEQ ID NO:112). This light chain was not randomized and used without any further modifications in order to ensure compatibility with different bispecific binders.
  • All heavy chains use the IGHJ2 as J-element, except the IGHV1-69*06 which uses IGHJ6 sequence. The design of the randomization focused on the CDR-H3 only, and PCR oligonucleotides were designed that allow for the usage of randomized inserts between the V-element and the J-element of 4 to 9 amino acids in length.
  • Example 6
  • Selection of Antibody Fragments from Common Light Chain Libraries (Comprising Light Chain with CD3ε Specificity) to FolR1
  • The antibodies 16A3, 15A1, 18D3, 19E5, 19A4, 15H7, 15B6, 16D5, 15E12, 21D1, 16F12, 21A5, 21G8, 19H3, 20G6, and 20H7 comprising the common light chain VL7_46-13 with CD3ε specificity were obtained by phage display selections against different species (human, cynomolgus and murine) of FolR1. Clones 16A3, 15A1, 18D3, 19E5, 19A4, 15H7, 15B6, 21D1, 16F12, 19H3, 20G6, and 20H7 were selected from a sub-library in which the common light chain was paired with a heavy chain repertoire based on the human germline VH1_46. In this sub-library, CDR3 of VH1_46 has been randomized based on 6 different CDR3 lengths. Clones 16D5, 15E12, 21A5, and 21G8 were selected from a sub-library in which the common light chain was paired with a heavy chain repertoire based on the human germline VH3_15. In this sub-library, CDR3 of VH3_15 has been randomized based on 6 different CDR3 lengths. In order to obtain species cross-reactive (or murine FolR1-reactive) antibodies, the different species of FolR1 were alternated (or kept constant) in different ways over 3 rounds of biopanning: 16A3 and 15A1 (human-cynomolgus-human FolR1); 18D3 (cynomolgus-human-murine FolR1); 19E5 and 19A4 (3 rounds against murine FolR1); 15H7, 15B6, 16D5, 15E12, 21D1, 16F12, 21A5, 21G8 (human-cynomolgus-human FolR1); 19H3, 20G6, and 20H7 (3 rounds against murine FolR1).
  • Human, murine and cynomolgus FolR1 as antigens for the phage display selections as well as ELISA- and SPR-based screenings were transiently expressed as N-terminal monomeric Fc-fusion in HEK EBNA cells and in vivo site-specifically biotinylated via co-expression of BirA biotin ligase at the avi-tag recognition sequence located at the C-terminus of the Fc portion carrying the receptor chain (Fc knob chain). In order to assess the specificity to FolR1, two related receptors, human FolR2 and FolR3 were generated in the same way.
  • Selection rounds (biopanning) were performed in solution according to the following pattern:
  • 1. Pre-clearing of ˜1012 phagemid particles on maxisorp plates coated with 10 ug/ml of an unrelated human IgG to deplete the libraries of antibodies recognizing the Fc-portion of the antigen.
    2. Incubating the non-Fc-binding phagemid particles with 100 nM biotinylated human, cynomolgus, or murine FolR1 for 0.5h in the presence of 100 nM unrelated non-biotinylated Fc knob-into-hole construct for further depletion of Fc-binders in a total volume of 1 ml.
    3. Capturing the biotinylated FolR1 and attached specifically binding phage by transfer to 4 wells of a neutravidin pre-coated microtiter plate for 10 min (in rounds 1 & 3).
    4. Washing the respective wells using 5×PBS/Tween20 and 5×PBS.
    5. Eluting the phage particles by addition of 250 ul 100 mM TEA (triethylamine) per well for 10 min and neutralization by addition of 500 ul 1 M Tris/HCl pH 7.4 to the pooled eluates from 4 wells.
    6. Post-clearing of neutralized eluates by incubation on neutravidin pre-coated microtiter plate with 100 nM biotin-captured FolR2 or FolR3 for final removal of Fc- and unspecific binders.
    7. Re-infection of log-phase E. coli TG1 cells with the supernatant of eluted phage particles, infection with helperphage VCSM13, incubation on a shaker at 30° C. over night and subsequent PEG/NaCl precipitation of phagemid particles to be used in the next selection round.
  • Selections were carried out over 3 rounds using constant antigen concentrations of 100 nM. In round 2, in order to avoid enrichment of binders to neutravidin, capture of antigen:phage complexes was performed by addition of 5.4×107 streptavidin-coated magnetic beads. Specific binders were identified by ELISA as follows: 100 ul of 25 nM biotinylated human, cynomolgus, or murine FolR1 and 10 ug/ml of human IgG were coated on neutravidin plates and maxisorp plates, respectively. Fab-containing bacterial supernatants were added and binding Fabs were detected via their Flag-tags using an anti-Flag/HRP secondary antibody. Clones exhibiting signals on human FolR1 and being negative on human IgG were short-listed for further analyses and were also tested in a similar fashion against the remaining two species of FolR1. They were bacterially expressed in a 0.5 liter culture volume, affinity purified and further characterized by SPR-analysis using BioRad's ProteOn XPR36 biosensor.
  • Affinities (KD) of selected clones were measured by surface plasmon resonance (SPR) using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated human, cynomolgus, and murine FolR1 as well as human FolR2 and FolR3 (negative controls) immobilized on NLC chips by neutravidin capture. Immobilization of antigens (ligand): Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 μg/ml, then injected at 30 μl/minute in vertical orientation. Injection of analytes: For ‘one-shot kinetics’ measurements, injection direction was changed to horizontal orientation, two-fold dilution series of purified Fab (varying concentration ranges) were injected simultaneously along separate channels 1-5, with association times of 200 s, and dissociation times of 600 s. Buffer (PBST) was injected along the sixth channel to provide an “in-line” blank for referencing. Association rate constants (kon) and dissociation rate constants (koff) were calculated using a simple one-to-one Langmuir binding model in ProteOn Manager v3.1 software by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) was calculated as the ratio koff/kon. Table 4 lists the equilibrium dissociation constants (KD) of the selected clones specific for FolR1.
  • TABLE 4
    Equilibrium dissociation constants (KD) for anti-FolR1 antibodies
    (Fab-format) selected by phage display from common light chain
    sub-libraries comprising VL7_46-13, a humanized light
    chain specific for CD3ε. KD in nM.
    huFolR1 cyFolR1 muFolR1 huFolR2
    Clone [nM] [nM] [nM] [nM] huFolR3 [nM]
    16A3 21.7 18 very weak no binding no binding
    15A1 30.9 17.3 very weak no binding no binding
    18D3 93.6 40.2 very weak no binding no binding
    19E5 522 276 19.4 no binding no binding
    19A4 2050 4250 43.1 no binding no binding
    15H7 13.4 72.5 no binding no binding no binding
    15B6 19.1 13.9 no binding no binding no binding
    16D5 39.5 114 no binding no binding no binding
    15E12 55.7 137 no binding no binding no binding
    21D1 62.6 32.1 no binding no binding no binding
    16F12 68 90.9 no binding no binding no binding
    21A5 68.8 131 no binding no binding no binding
    21G8 130 261 no binding no binding no binding
    19H3 no binding no binding 89.7 no binding no binding
    20G6 no binding no binding 78.5 no binding no binding
  • Example 7 Selection of Antibody Fragments from Generic Multi-Framework Libraries to FolR1
  • The antibodies 11F8, 36F2, 9D11, 5D9, 6B6, and 14E4 were obtained by phage display selections based on generic multi-framework sub-libraries against different species (human, cynomolgus and murine) of FolR1. In these multi-framework sub-libraries, different VL-domains with randomized CDR3 (3 different lengths) are paired with different VH-domains with randomized CDR3 (6 different lengths). The selected clones are of the following VL/VH pairings: 11F8 (Vk_1_5/VH_1_69), 36F2 (Vk_3_20/VH_1_46), 9D11 (Vk2D_28/VH1_46), 5D9 (Vk3_20/VH1_46), 6B6 (Vk3_20/VH1_46), and 14E4 (Vk3_20/VH3_23). In order to obtain species cross-reactive (or murine FolR1-reactive) antibodies, the different species of FolR1 were alternated (or kept constant) in different ways over 3 or 4 rounds of biopanning: 11F8 (cynomolgus-murine-human FolR1); 36F2 (human-murine-cynomolgus-murine FolR1); 9D11 (cynomolgus-human-cynomolgus FolR1); 5D9 (human-cynomolgus-human FolR1); 6B6 (human-cynomolgus-human FolR1) and 14E4 (3 rounds against murine FolR1).
  • Human, murine and cynomolgus FolR1 as antigens for the phage display selections as well as ELISA- and SPR-based screenings were transiently expressed as N-terminal monomeric Fc-fusion in HEK EBNA cells and in vivo site-specifically biotinylated via co-expression of BirA biotin ligase at the avi-tag recognition sequence located at the C-terminus of the Fc portion carrying the receptor chain (Fc knob chain). In order to assess the specificity to FolR1, two related receptors, human FolR2 and FolR3 were generated in the same way.
  • Selection rounds (biopanning) were performed in solution according to the following pattern:
  • 1. Pre-clearing of ˜1012 phagemid particles on maxisorp plates coated with 10 ug/ml of an unrelated human IgG to deplete the libraries of antibodies recognizing the Fc-portion of the antigen.
    2. Incubating the non-Fc-binding phagemid particles with 100 nM biotinylated human, cynomolgus, or murine FolR1 for 0.5h in the presence of 100 nM unrelated non-biotinylated Fc knob-into-hole construct for further depletion of Fc-binders in a total volume of 1 ml.
    3. Capturing the biotinylated FolR1 and attached specifically binding phage by transfer to 4 wells of a neutravidin pre-coated microtiter plate for 10 min (in rounds 1 & 3).
    4. Washing the respective wells using 5×PBS/Tween20 and 5×PBS.
    5. Eluting the phage particles by addition of 250 ul 100 mM TEA (triethylamine) per well for 10 min and neutralization by addition of 500 ul 1 M Tris/HCl pH 7.4 to the pooled eluates from 4 wells.
    6. Post-clearing of neutralized eluates by incubation on neutravidin pre-coated microtiter plate with 100 nM biotin-captured FolR2 or FolR3 for final removal of Fc- and unspecific binders.
    7. Re-infection of log-phase E. coli TG1 cells with the supernatant of eluted phage particles, infection with helperphage VCSM13, incubation on a shaker at 30° C. over night and subsequent PEG/NaCl precipitation of phagemid particles to be used in the next selection round.
  • Selections were carried out over 3 rounds using constant antigen concentrations of 100 nM. In round 2 and 4, in order to avoid enrichment of binders to neutravidin, capture of antigen:phage complexes was performed by addition of 5.4×107 streptavidin-coated magnetic beads. Specific binders were identified by ELISA as follows: 100 ul of 25 nM biotinylated human, cynomolgus, or murine FolR1 and 10 ug/ml of human IgG were coated on neutravidin plates and maxisorp plates, respectively. Fab-containing bacterial supernatants were added and binding Fabs were detected via their Flag-tags using an anti-Flag/HRP secondary antibody. Clones exhibiting signals on human FolR1 and being negative on human IgG were short-listed for further analyses and were also tested in a similar fashion against the remaining two species of FolR1. They were bacterially expressed in a 0.5 liter culture volume, affinity purified and further characterized by SPR-analysis using BioRad's ProteOn XPR36 biosensor.
  • Affinities (KD) of selected clones were measured by surface plasmon resonance (SPR) using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated human, cynomolgus, and murine FolR1 as well as human FolR2 and FolR3 (negative controls) immobilized on NLC chips by neutravidin capture. Immobilization of antigens (ligand): Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 μg/ml, then injected at 30 μl/minute in vertical orientation. Injection of analytes: For ‘one-shot kinetics’ measurements, injection direction was changed to horizontal orientation, two-fold dilution series of purified Fab (varying concentration ranges) were injected simultaneously along separate channels 1-5, with association times of 150 or 200 s, and dissociation times of 200 or 600 s, respectively. Buffer (PBST) was injected along the sixth channel to provide an “in-line” blank for referencing. Association rate constants (kon) and dissociation rate constants (koff) were calculated using a simple one-to-one Langmuir binding model in ProteOn Manager v3.1 software by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) was calculated as the ratio koff/kon. Table 5 lists the equilibrium dissociation constants (KD) of the selected clones specific for FolR1.
  • TABLE 5
    Equilibrium dissociation constants (KD) for anti-FolR1 antibodies
    (Fab-format) selected by phage display from generic multi-framework
    sub-libraries. KD in nM.
    KD (nM)
    Clone huFolR1 cyFolR1 muFolR1 huFolR2 huFolR3
    11F8 632 794 1200 no binding no binding
    36F2 1810 1640 737 no binding no binding
    9D11 8.64 5.29 no binding no binding no binding
    5D9 8.6 5.9 no binding no binding no binding
    6B6 14.5 9.4 no binding no binding no binding
    14E4 no binding no binding 6.09 no binding no binding
  • Example 8 Production and Purification of Novel FolR1 Binders in IgG and T-Cell Bispecific Formats
  • To identify FolR1 binders which are able to induce T-cell dependent killing of selected target cells the antibodies isolated from a common light chain- or Fab-library were converted into the corresponding human IgG1 format. In brief, the variable heavy and variable light chains of unique FolR1 binders from phage display were amplified by standard PCR reactions using the Fab clones as the template. The PCR products were purified and inserted (either by restriction endonuclease and ligase based cloning, or by ‘recombineering’ using the InFusion kit from Invitrogen) into suitable expression vectors in which they are fused to the appropriate human constant heavy or human constant light chain. The expression cassettes in these vectors consist of a chimeric MPSV promoter and a synthetic polyadenylation site. In addition, the plasmids contain the oriP region from the Epstein Barr virus for the stable maintenance of the plasmids in HEK293 cells harboring the EBV nuclear antigen (EBNA). After PEI mediated transfection the antibodies were transiently produced in HEK293 EBNA cells and purified by standard ProteinA affinity chromatography followed by size exclusion chromatography as described:
  • Transient Transfection and Production
  • All (bispecific) antibodies (if not obtained from a commercial source) used herein were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 μg DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C. After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 μm sterile filters and stored at 4° C. until purification.
  • Antibody Purification
  • All molecules were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to HiTrap PA FF (GE Healthcare, column volume (cv)=5 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate, 20 mM sodium citrate, pH 7.5). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 3, 100 mM NaCl, 100 mM glycine) over 12 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 0.5 M Na2HPO4 pH 8.0). Samples were concentrated to 2 ml using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a HiLoad™ 16/60 Superdex™ 200 preparative grade (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled and concentrated to final concentration of 1-1.5 mg/ml using ultra concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius). The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at −80° C.
  • Based on in vitro characterization results selected binders were converted into a T-cell bispecific format. In these molecules the FolR1:CD3 binding moieties are arranged in a 2:1 order with the FolR1 Fabs being located at the N-terminus. For clones isolated from the standard Fab library the CD3 binding part was generated as a CrossFab (CHICK crossing) while for the clones from the common light chain library no crossing was necessary. These bispecific molecules were produced and purified analogously to the IgGs.
  • TABLE 6
    Yield and monomer content of novel FolR1
    binders in IgG and TCB format, respectively.
    IgG TCB
    Yield Monomer Yield Monomer
    # Clone Library [mg/L] [%] [mg/L} [%]
    1 11F8 Fab 8.03 96.26
    2 14E4 Fab 8.90 98.12
    3 15B6 CLC 7.72 100.00
    4 15E12 CLC 6.19 100.00
    5 15H7 CLC 8.94 100.00
    6 16A3 CLC 0.60 n.d.
    7 16D5 CLC 36.50 96.96 4.36 97.19
    8 16F12 CLC 5.73 97.17
    9 18D3 CLC 0.90 n.d.
    10 19A4 CLC 38.32 100.00 37.50 100.00
    11 19E5 CLC 46.09 100.00
    12 19H3 CLC 7.64 100.00
    13 20G6 CLC 24.00 100.00
    14 20H7 CLC 45.39 100.00
    15 21A5 CLC 1.38 98.56 47.31 95.08
    16 21D1 CLC 5.47 100.00
    17 21G8 CLC 6.14 97.28 9.27 100.00
    18 36F2 Fab 11.22 100.00 18.00 100.00
    19 5D9 Fab 20.50 100.00 0.93 97.32
    20 6B6 Fab 3.83 100.00 4.17 91.53
    21 9D11 Fab 14.61 100.00 2.63 100.00
    CLC: Common light chain
  • Example 9 2+1 and 1+1 T-Cell Bispecific Formats
  • Four different T-cell bispecific formats were prepared for one common light chain binder (16D5) and three formats for one binder from the Fab library (9D11) to compare their killing properties in vitro.
  • The standard format is the 2+1 inverted format as already described (FolR1:CD3 binding moieties arranged in a 2:1 order with the FolR1 Fabs located at the N-terminus). In the 2+1 classical format the FolR1:CD3 binding moieties are arranged in a 2:1 order with the CD3 Fab being located at the N-terminus. Two monovalent formats were also prepared. The 1+1 head-to-tail has the FolR1:CD3 binding moieties arranged in a 1:1 order on the same arm of the molecule with the FolR1 Fab located at the N-terminus. In the 1+1 classical format the FolR1:CD3 binding moieties are present once, each on one arm of the molecule. For the 9D11 clone isolated from the standard Fab library the CD3 binding part was generated as a CrossFab (CHICK crossing) while for the 16D5 from the common light chain library no crossing was necessary. These bispecific molecules were produced and purified analogously to the standard inverted T-cell bispecific format.
  • TABLE 7
    Summary of the yield and final monomer content of the different
    T-cell bispecific formats.
    Monomer
    [%]
    Construct (SEC) Yield
    16D5 FolR1 TCB 2 + 1 (inverted) 96% 5.4 mg/L
    16D5 FolR1 TCB 2 + 1 (classical) 90% 4.6 mg/L
    16D5 FolR1 TCB 1 + 1 (head-to- 100% 5.4 mg/L
    tail)
    16D5 FolR1 TCB 1 + 1 (classical) 100% 0.7 mg/L
    9D11 FolR1 TCB 2 + 1 (inverted) 100% 2.6 mg/L
    9D11 FolR1 TCB 1 + 1 (head-to- 100% 6.1 mg/L
    tail)
    9D11 FolR1 TCB 1 + 1 (classical) 96% 1.3 mg/L
    Mov19 FolR1 TCB 2 + 1 (inverted) 98%   3 mg/L
    Mov19 FolR1 TCB 1 + 1 (head-to- 100% 5.2 mg/L
    tail)
  • Example 10 Biochemical Characterization of FolR1 Binders by Surface Plasmon Resonance
  • Binding of FolR1 binders as IgG or in the T-cell bispecific format to different recombinant folate receptors (human FolR1, 2 and 3, murine FolR1 and cynomolgus FolR1; all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).
  • Single Injections
  • First the anti-FolR1 IgGs were analyzed by single injections (Table 1) to characterize their crossreactivity (to human, murine and cyno FolR1) and specificity (to human FolR1, human FolR2, human FolR3). Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) or human Folate Receptor 2 and 3 (FolR2-Fc, FolR3-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The IgGs were injected for 60 seconds at a concentration of 500 nM. IgGs binding to huFolR2 and huFolR3 were rejected for lack of specificity. Most of the binders are only crossreactive between human and cyno FolR1, additional crossreactivity to murine FolR1 went most of the time hand in hand with loss of specificity.
  • TABLE 8
    Crossreactivity and specificity of 25 new folate receptor 1 binders (as IgGs) as well as of
    two control IgGs (Mov19 and Farletuzumab). + means binding, − means no binding,
    +/− means weak binding.
    Binding Binding Binding Binding Binding
    Clone name to huFolR1 to cyFolR1 to muFolR1 to huFolR2 to huFolR3
    Mov19 + +
    Farletuzumab + +
    16A3 + + +/−
    18D3 + +
    19E5 + + + + +
    19A4 + + +
    15H7 + + +
    15B6 + +
    16D5 + +
    15E12 + + +/− + +
    21D1 + + +/−
    16F12 + +
    21A5 + + +/−
    21G8 + + + +
    19H3 +
    20G6 +
    20H7 +
    9D11 + +
    5D9 + + + +
    6B6 + + + +
    11F8 + + + + +
    36F2 + + +
    14E4 +
  • Avidity to Folate Receptor 1
  • The avidity of the interaction between the anti-FolR1 IgGs or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 9).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 2.1 to 500 nM with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 600 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated IL2 receptor Fc fusion. For the analysis of the interaction of 19H3 IgG and murine folate receptor 1, folate (Sigma F7876) was added in the HBS-EP running buffer at a concentration of 2.3 μM. The binding curves resulting from the bivalent binding of the IgGs or T cell bispecifics were approximated to a 1:1 Langmuir binding and fitted with that model (which is not correct, but gives an idea of the avidity). The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).
  • TABLE 9
    Bivalent binding (avidity with apparent KD) of selected FolR1 binders
    as IgGs or as T-cell bispecifics (TCB) on human and cyno FolR1.
    Apparent
    Analyte Ligand ka (1/Ms) kd (1/s) KD (M)
    16D5 TCB huFolR1 8.31E+04 3.53E−04 4.24E−09
    cyFolR1 1.07E+05 3.70E−04 3.45E−09
    9D11 TCB huFolR1 1.83E+05 9.83E−05 5.36E−10
    cyFolR1 2.90E+05 6.80E−05 2.35E−10
    21A5 TCB huFolR1 2.43E+05 2.64E−04 1.09E−09
    cyFolR1 2.96E+05 2.76E−04 9.32E−10
    36F2 IgG huFolR1 2.62E+06 1.51E−02 5.74E−9 
    cyFolR1 3.02E+06 1.60E−02 5.31E−9 
    muFolR1  3.7E+05 6.03E−04 1.63E−9 
    Mov19 IgG huFolR1 8.61E+05 1.21E−04  1.4E−10
    cyFolR1 1.29E+06 1.39E−04 1.08E−10
    Farletuzumab huFolR1 1.23E+06   9E−04  7.3E−10
    cyFolR1 1.33E+06 8.68E−04  6.5E−10
    19H3 IgG muFolR1  7.1E+05  1.1E−03 1.55E−09
  • 1. Affinity to Folate Receptor 1
  • The affinity of the interaction between the anti-FolR1 IgGs or the T cell bispecifics and the recombinant folate receptors was determined as described below (Table 10).
  • For affinity measurement, direct coupling of around 6000-7000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 IgGs or T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 20 or 40 sec, the reference flow cell was left without capture. Dilution series (6.17 to 500 nM or 12.35 to 3000 nM) of human or cyno Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 120 or 240 sec to record the association phase. The dissociation phase was monitored for 240 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1 or pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • TABLE 10
    Monovalent binding (affinity) of selected FolR1 binders as IgGs or
    as T-cell bispecifics (TCB) on human and cyno FolR1.
    Ligand Analyte ka (1/Ms) kd(1/s) KD (M)
    16D5 TCB huFolR1 1.53E+04 6.88E−04 4.49E−08
    cyFolR1 1.32E+04 1.59E−03 1.21E−07
    9D11 TCB huFolR1 3.69E+04 3.00E−04 8.13E−09
    cyFolR1 3.54E+04 2.06E−04 5.82E−09
    21A5 TCB huFolR1 1.79E+04  1.1E−03 6.16E−08
    cyFolR1 1.48E+04 2.06E−03  1.4E−07
    Mov19 IgG huFolR1 2.89E+05 1.59E−04  5.5E−10
    cyFolR1 2.97E+05 1.93E−04  6.5E−10
    Farletuzumab huFolR1 4.17E+05 2.30E−02 5.53E−08
    cyFolR1 5.53E+05 3.73E−02 6.73E−08
  • 2. Affinity to CD3
  • The affinity of the interaction between the anti-FolR1 T cell bispecifics and the recombinant human CD3εδ-Fc was determined as described below (Table 11).
  • For affinity measurement, direct coupling of around 9000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 40 sec, the reference flow cell was left without capture. Dilution series (6.17 to 500 nM) of human CD3εδ-Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 240 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • TABLE 11
    Monovalent binding (affinity) of selected FolR1 T-cell bispecifics (TCB)
    on human CD3-Fc.
    Ligand Analyte ka (1/Ms) kd (1/s) KD (M)
    16D5 TCB huCD3 4.25E+04 3.46E−03 8.14E−08
    21A5 TCB huCD3 3.72E+04 3.29E−03  8.8E−08
  • The CD3 binding part is identical for all constructs and the affinity is similar for the measured T cell bispecifics (KD range between 60 and 90 nM).
  • Example 11 Simultaneous Binding T Cell Bispecifics on Folate Receptor 1 and CD3
  • Simultaneous binding of the anti-FolR1 T cell bispecifics on recombinant Folate Receptor 1 and recombinant human CD3εδ-Fc was determined by surface plasmon resonance as described below. Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The anti-FolR1 T cell bispecifics were injected for 60 s at 500 nM with a flow of 30 μL/minutes through the flow cells, followed by an injection of hu CDεδ-Fc for 60 s at 500 nM. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated IL2 receptor Fc fusion. The four T cell bispecifics tested (16D5 TCB, 21A5 TCB, 51C7 TCB and 45D2 TCB) were able to bind simultaneously to Folate Receptor 1 and human CD3 as expected.
  • Example 12 Epitope Binning
  • For epitope binning, the anti-FolR1 IgGs or T cell bispecifics were directly immobilized on a CMS chip at pH 5.0 using the standard amine coupling kit (GE Healthcare), with a final response around 700 RU. 500 nM huFolR1-Fc was then captured for 60 s, followed by 500 nM of the different binders for 30 s. The surface was regenerated with two injections of 10 mM glycine pH 2 for 30 s each. It is assessed if the different binders can bind to huFolR1 captured on immobilized binders (Table 12).
  • TABLE 12
    Epitope characterization of selected FolR1 binders as
    IgGs or as T-cell bispecifics (TCB) on human FolR1.
    Analytes in solution
    On 16D5 21A5 9D11 36F2 Mov19
    huFolR1 TCB TCB TCB IgG IgG Farletuzumab
    Immobilized 16D5 + + +
    TCB
    21A5 + + +
    TCB
    9D11 No additional binding on FolR1 possible once captured on
    TCB 9D11
    36F2 IgG Measure not possible, huFolR1 dissociates too rapidly
    Mov19 + + +/−
    IgG
    + means binding,
    − means no binding,
    +/− means weak binding
  • Based on these results and additional data with simultaneous binding on immobilized huFolR1, the binders were separated in three groups. It is not clear if 9D11 has a separate epitope because it displaces all the other binders. 16D5 and 21A5 seem to be in the same group and Mov19, Farletuzumab (Coney et al., Cancer Res. 1991 Nov. 15; 51(22):6125-32; Kalli et al., Curr Opin Investig Drugs. 2007 December; 8(12):1067-73) and 36F2 in another (Table 13). However, 36F2 binds to a different epitope than Mov 19 and Farletuzumab as it binds to human, cynomous and murine FolR1.
  • TABLE 13
    Epitope grouping of selected FolR1 binders as IgGs or as T-cell
    bispecifics (TCB) on human FolR1
    Epitope
    1 Epitope 2 Epitope 3
    16D5 9D11 Mov19
    21A5 Farletuzumab
    36F2
  • Example 13 Selection of Binders
  • FolR1 binders in the IgG formats were screened by surface plasmon resonance (SPR) and by in vitro assay on cells to select the best candidates.
  • The anti-FolR1 IgGs were analyzed by SPR to characterize their crossreactivity (to human, murine and cynomolgus FolR1) and specificity (to human FolR1, human FolR2, human FolR3). Unspecific binding to human FolR2 and 3 was considered an exclusion factor. Binding and specificity to human FolR1 was confirmed on cells. Some binders did not bind on cells expressing FolR1 even though they recognized the recombinant human FolR1 in SPR. Aggregation temperature was determined but was not an exclusion factor because the selected binders were all stable. Selected binders were tested in a polyreactivity ELISA to check for unspecific binding, which led to the exclusion of four binders. This process resulted in an initial selection of three binders: 36F2 (Fab library), 9D11 (Fab library) and 16D5 (common light chain). 36F2 dissociated rapidly from huFolR1 and was, therefore, initially not favored.
  • Example 14 Specific Binding of Newly Generated FolR1 Binders to Human FolR1 Positive Tumor Cells
  • New FolR1 binders were generated via Phage Display using either a Fab library or a common light chain library using the CD3 light chain. The identified binders were converted into a human IgG1 format and binding to FolR1 high expressing HeLa cells was addressed. As reference molecule the human FolR1 binder Mov19 was included. Most of the binders tested in this assay showed intermediate to good binding to FolR1 with some clones binding equally well as Mov19 (see FIG. 2). The clones 16A3, 18D3, 15H7, 15B6, 21D1, 14E4 and 16F12 were excluded because binding to FolR1 on cells could not be confirmed by flow cytometry. In a next step the selected clones were tested for specificity to human FolR1 by excluding binding to the closely related human FolR2. HEK cells were transiently transfected with either human FolR1 or human FolR2 to address specificity. The clones 36F2 and 9D11 derived from the Fab library and the clones 16D5 and 21A5 derived from the CLC library bind specifically to human FolR1 and not to human FolR2 (see FIGS. 3A-B). All the other tested clones showed at least some binding to human FolR2 (see FIGS. 3A-B). Therefore these clones were excluded from further characterization. In parallel cross-reactivity of the FolR1 clones to cyno FolR1 was addressed by performing binding studies to HEK cells transiently transfected with cyno FolR1. All tested clones were able to bind cyno FolR1 and the four selected human FoLR1 specific clones 36F2, 9D11, 16D5 and 21A5 bind comparably well human and cyno FoLR1 (FIG. 4). Subsequently three human FolR1 specific cyno cross-reactive binders were converted into TCB format and tested for induction of T cell killing and T cell activation. These clones were 9D11 from the Fab library and 16D5 and 21A5 from the CLC library. As reference molecule Mov19 FolR1 TCB was included in all studies. These FolR1 TCBs were then used to compare induction of internalization after binding to FolR1 on HeLa cells. All three tested clones are internalized upon binding to FolR1 comparable to internalization upon binding of Mov19 FoLR1 TCB (FIG. 5). 21A5 FolR1 TCB was discontinued due to signs of polyreactivity.
  • Example 15 T Cell-Mediated Killing of FolR1-Expressing Tumor Target Cells Induced by FolR1 TCB Antibodies
  • The FolR1 TCBs were used to determine T cell mediated killing of tumor cells expressing FoLR1. A panel of potential target cell lines was used to determine FoLR1 binding sites by Qifikit analysis. The used panel of tumor cells contains FolR1 high, intermediate and low expressing tumor cells and a FolR1 negative cell line.
  • TABLE 14
    FolR1 binding sites on tumor cells
    Cell line Origin FolR1 binding sites
    Hela Cervix adenocarcinoma 2′240′716
    Skov3 Ovarian adenocarcinoma 91′510
    OVCAR5 Ovarian adenocarcinoma 22′077
    HT29 Colorectal adenocarcinoma 10′135
    MKN45 Gastric adenocarcinoma 54
  • Binding of the three different FoLR1 TCBs (containing 9D11, 16D5 and Mov19 binders) to this panel of tumor cell lines was determined showing that the FoLR1 TCBs bind specifically to FolR1 expressing tumor cells and not to a FoLR1 negative tumor cell line. The amount of bound construct is proportional to the FolR1 expression level and there is still good binding of the constructs to the FolR1 low cell line HT-29 detectable. In addition there is no binding of the negative control DP47 TCB to any of the used cell lines (FIGS. 6A-E).
  • The intermediate expressing cell line SKOV3 and the low expressing cell line HT-29 were further on used to test T cell mediated killing and T cell activation using 16D5 TCB and 9D11 TCB; DP47 TCB was included as negative control. Both cell lines were killed in the presence of already very low levels of 16D5 TCB and 9D11 TCB and there was no difference in activity between both TCBs even though 9D11 TCB binds stronger to FolR1 than 16D5 TCB. Overall killing of SKOV3 cells was higher compared to HT-29 which reflects the higher expression levels of FolR1 on SKOV3 cells (FIGS. 7A-D). In line with this, a strong upregulation of the activation marker CD25 and CD69 on CD4+ T cells and CD8+ T cells was detected. Activation of T cells was very similar in the presence of SKOV3 cells and HT-29 cells. The negative control DP47 TCB does not induce any killing at the used concentrations and there was no significant upregulation of CD25 and CD69 on T cells.
  • TABLE 15
    EC50 values of tumor cell killing and T cell
    activation with SKOV3 cells
    CD4+ CD4+ CD8+ CD8+
    Killing Killing CD69+ CD25+ CD69+ CD25+
    Construct
    24 h (pM) 48 h (pM) (%) (%) (%) (%)
    9D11 1.1 0.03 0.51 0.46 0.019 0.03
    FolR1
    TCB
    16D5 0.7 0.04 0.34 0.33 0.025 0.031
    FolR1
    TCB
  • TABLE 16
    EC50 values of tumor cell killing and T cell
    activation with HT-29 cells
    CD4+ CD4+ CD8+ CD8+
    Killing Killing CD69+ CD25+ CD69+ CD25+
    Construct
    24 h (pM) 48 h (pM) (%) (%) (%) (%)
    9D11 2.3 0.1 1.22 1.11 0.071 0.084
    FolR1
    TCB
    16D5 2.8 0.1 0.69 0.62 0.021 0.028
    FolR1
    TCB
  • Example 16 Binding to Erythrocytes and T Cell Activation in Whole Blood
  • To prove that there is no spontaneous activation in the absence of FoLR1 expressing tumor cells we tested if there is binding of the FolR1 clones to erythrocytes which might potentially express FolR1. We could not observe any specific binding of 9D11 IgG, 16D5 IgG and Mov19 IgG to erythrocytes, as negative control DP47 IgG was included (FIG. 8).
  • To exclude any further unspecific binding to blood cells or unspecific activation via FoLR1 TCB, 9D11 TCB, 16D5 TCB and Mov19 TCB were added into whole blood and upregulation of CD25 and CD69 on CD4+ T cells and CD8+ T cells was analyzed by flow cytometry. DP47 TCB was included as negative control. No activation of T cells with any of the tested constructs could be observed by analyzing upregulation of CD25 and CD69 on CD4+ T cells and CD8+ T cells (FIG. 9).
  • Example 17 Removal of the N-Glycosylation Site in 9D11 Light Chain
  • During analysis of the different FolR1 binders to identify potential sequence hot spots, at the end of CDR L3 of the clone 9D11 a putative N-glycosylation site was identified. Usually the consensus motif for N-glycosylation is defined as N-X-S/T-X (where X is not P). The sequence of CDR L3 (MQASIMNRT) (SEQ ID NO: 61) perfectly matches this consensus motif having the sequence N-R-T. Since glycosylation might not be completely reproducible among different production batches this could have an impact on FolR1 binding, if the glycosylation in CDR L3 contributes to antigen binding. To evaluate if this N-glycosylation site is important for FolR1 binding, or could be replaced without impairing binding, different variants of the 9D11 light chain were generated in which the N-glycosylation site was exchanged by site specific mutagenesis.
  • 1. Transient Transfection and Production
  • The four T cell bispecifics were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells were centrifuged for 5 min by 210×g, supernatant was replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200m DNA. After addition of 540 μl PEI solution was vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C. After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 μm sterile filters and stored at 4° C. until purification.
  • 2. Antibody purification
  • All molecules were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to HiTrap PA HP (GE Healthcare, column volume (cv)=5 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M NaCl, 0.01% Tween-20, pH 7.5). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 2.5, 0.5 M NaCl, 0.01% Tween-20) over 20 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 2 M Tris pH 8.0). Samples were concentrated to 1 ml using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a Superdex™ 200 10/300 GL (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled and concentrated to final concentration of 1-1.5 mg/ml using ultra concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius). The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at −80° C.
  • 3. Aggregation Temperature
  • Stability of the four constructs was tested on an Optim1000 (Avacta, PALL Corporation) by a gradient heating from 25° to 80° at 0.1° C./min. The temperature at onset of aggregation is recorded.
  • TABLE 34
    Yield, monomer content and aggregation temperature of four
    N-glycosylation site knock-out mutant of the 9D11 binder in the 2 + 1
    inverted T-cell bispecific format. All four mutants behaved
    similarly to the wild-type 9D11 binder
    Yield Monomer Aggregation
    Clone Mutation [mg/L} [%] temperature
    9D11 T102N 1.34 97 56°
    9D11 T102A 1.29 100 56°
    9D11 N100Q 2.5 100 56°
    9D11 N100S 2.05 100 56°
    9D11 2.6 100 57°
  • The following variants were generated: N100S (N95S); N100Q (N95Q), T102A (T97A) and T102N (T97N) (Kabat numbering indicated in parenthesis) and converted into the T-cell bispecific format. After transient production in HEK293 EBNA cells and purification the different variants were analyzed for target binding and cell killing activity in comparison to the original 9D11 clone.
  • TABLE 17
    Primers used for removal of N-glycosylation site in CDR L3 of 9D11
    (sequences see below)
    # Amino acid exchange Mutagenesis primer
    1 N95S GAB-7735
    2 N95Q GAB-7734
    3 T97A GAB7736
    4 T97N GAB-7737
  • Example 18 Binding and T Cell Mediated Killing with 9D11 a-Glyco Variants
  • Due to a glycosylation site in the CDRs four different 9D11 variants were produced with a mutation removing the glycosylation site (Example 17). These four variants were tested in comparison to the original 9D11 for binding to FolR1 on HeLa cells (FIG. 10) and induction of tumor cell killing on SKOV3 and HT-29 (FIG. 11A-B, E-F). None of the variants showed differences in binding or induction of tumor cell killing. In parallel unspecific killing of the FolR1 negative cell lines MKN-45 was addressed (FIGS. 11C-D). Also, no differences between the variants and the original binder could be observed. None of the constructs induced unspecific killing on FoLR1 negative tumor cells.
  • Example 19 FolR1 Expression on Primary Epithelial Cells
  • FolR1 is expressed at low levels on primary epithelial cells. Here we wanted to test if these levels are sufficient to induce T cell mediated killing in the presence of the FolR1 TCBs. To test this we used primary human bronchial epithelial cells, primary human choroid plexus epithelial cell, primary human renal cortical epithelial cells and primary human retinal pigment epithelial cells. As positive control either FolR1 positive SKOV3 cells or HT-29 cells were included. First we verified FolR1 expression on the used primary cells and determined the amount of FolR1 binding sites on these cells. Bronchial epithelial cells, renal cortical epithelial cells and retinal pigment epithelial cells express very low but significant levels of FolR1 compared to the levels expressed on tumor cells. The choroid plexus epithelial cells do not express significant levels of FolR1.
  • TABLE 18
    FolR1 binding sites on primary epithelial cells
    Cell line Binding sites
    Bronchial epithelium 492
    Choroid plexus epithelium 104
    Renal cortical epithelium 312
    Retinal pigment epithelium 822
    Skov3 69′890
  • The primary epithelial cells that demonstrated FolR1 expression on the surface were used to address the question if these cells can be killed by T cells in the presence of FoLR1 TCBs. No significant levels of killing could be measured but induction of T cell activation in the presence of retinal pigment epithelial cells, bronchial epithelial cells and renal cortical cells resulting in upregulation of CD25 and CD69 was detected. The strongest activation is seen with retinal pigment epithelial cells resulting in upregulation of CD25 and CD69 both on CD4+ T cells and CD8+ T cells. In the presence of bronchial epithelial cells lower activation of T cells is induced with upregulation of CD69 on CD4+ T cells and CD8+ T cells but very low upregulation of CD25 only on CD4+ T cells but not on CD8+ T cells. The lowest activation of T cells is obtained in the presence of renal epithelial cells with no upregulation of CD25 on CD4 T±cells and CD8+ T cells and CD69 been only upregulated on CD8+ T cells (FIGS. 12A-X).
  • Example 20 Comparison of Different TCB Formats Containing Either 16D5 or 9D11 Binder
  • To determine if the TCB 2+1 inverted format is the most active format with the selected FolR1 binder, different formats containing either 16D5 or 9D11 were produced and compared in target cell binding, T cell mediated killing and T cell activation. The 16D5 binder was tested in the TCB 2+1 inverted (FIG. 1A), TCB 2+1 classical (FIG. 1D), TCB 1+1 classical (FIG. 1C) and TCB 1+1 head-to-tail (FIG. 1B) format; the 9D11 binder was tested in the TCB 2+1 inverted (FIG. 1A), TCB 1+1 classical (FIG. 1C) and TCB 1+1 head-to-tail (FIG. 1B) format.
  • All constructs were tested for binding to FolR1 on HeLa cells. The molecules bivalent for binding to FolR1 bind stronger compared to the monovalent constructs due to avidity. The difference between the bivalent vs. monovalent constructs is more pronounced for 16D5. The reason might be that due to the lower affinity of 16D5 the avidity effect for this binder is stronger. Between the two 1+1 TCBs there is no significant difference in binding but there is a difference between the two 2+1 constructs. The inverted 2+1 construct binds stronger to FolR1 than the classical 2+1 construct. This indicates that in the classical 2+1 construct the binding to FoLR1 is influenced by the presence of the CD3 Fab whereas in the inverted construct binding is less influenced.
  • By testing T cell mediated killing with these constructs we could show that stronger binding of the 2+1 inverted TCB in converted into stronger tumor cell killing and T cell activation compared to the 2+1 classical TCB. The 16D5 FoLR1 TCB 2+1 classical is only a little bit more active than the respective 1+1 head-to-tail construct. The 1+1 head-to-tail construct is significantly more active than the 1+1 classical construct. This does not reflect the situation seen in binding and might be due to better crosslinking with the head-to-tail construct. Overall tumor cell killing and T cell activation is comparable with all tested constructs, the differences in potency seen with the differences are only in terms of EC50 values. In general it can be concluded that the FolR1 TCB 2+1 inverted independent of the used binder is the preferred format to induce T cell mediated tumor cell killing and T cell activation (see FIG. 13A-C and FIG. 14A-C).
  • TABLE 19
    EC50 values of target cell binding and T cell mediated killing
    with different TCB formats
    Binding
    Construct EC50 (nM) Killing 24 h (pM) Killing 48 h (pM)
    16D5 FolR1 TCB 11.03 1.43 0.18
    2 + 1 inverted
    16D5 FolR1 TCB 17.07 5.60 2.18
    2 + 1 classical
    16D5 FolR1 TCB 107.3 n.d. n.d.
    1 + 1 classical
    16D5 FoLR1 TCB 102.6 26.24 6.06
    1 + 1 head-to-tail
    9D11 FoLR1 TCB 17.52 0.74 0.14
    2 + 1 inverted
    9D11 FoLR1 TCB 38.57 20.92 n.d.
    1 + 1 classical
    9D11 FoLR1 TCB 44.20 4.73 n.d.
    1 + 1 head-to-tail
  • TABLE 20
    EC50 values of T cell activation in the presence of SKOV3 cells
    with different TCB formats
    CD4+CD25+ CD4+CD69+ CD8+CD25+ CD8+CD69+
    Construct (%) (%) (%) (%)
    16D5 FolR1 1.96 0.33 2.10 n.d.
    TCB
    2 + 1 inverted
    16D5 FolR1 13.83 3.67 12.88 4.47
    TCB
    2 + 1 classical
    16D5 FolR1 38.54 n.d. n.d. n.d.
    TCB
    1 + 1 classical
    16D5 FoLR1 17.14 7.47 25.15 n.d.
    TCB 1 + 1
    head-to-tail
    9D11 FoLR1 1.41 0.27 1.24 0.35
    TCB 2 + 1
    inverted
    9D11 FoLR1 34.01 n.d. 34.39 7.40
    TCB 1 + 1
    classical
    9D11 FoLR1 3.73 2.47 4.98 2.89
    TCB 1 + 1
    head-to-tail
  • Example 21 Tumor Cell Lines and Primary Cells
  • HeLa cells (CCL-2) were obtained from ATCC and cultured in DMEM with 10% FCS and 2 mM Glutamine, SKOV3 (HTB-77) were obtained from ATCC and cultured in RPMI with 10% FCS and 2 mM Glutamine, OVCAR5 were obtained from NCI and cultured in RPMI with 10% FCS and 2 mM Glutamine, HT-29 (ACC-299) were obtained from DSMZ and cultured in McCoy's 5A medium with 10% FCS and 2 mM Glutamine, MKN-45 (ACC-409) were obtained from DSMZ and cultured in RPMI with 10% FCS and 2 mM Glutamine.
  • All tested primary epithelial cells were obtained from ScienCell Research Laboratories. Human Bronchial Epithelium Cells (HBEpiC, Catalog Number 3210 were cultured in Bronchial Epithelial Cell Medium (BEpiCM, Cat. No. 3211, ScienCell). Human Colonic Epithelial Cells (HCoEpiC), Catalog Number 2950 were cultured in Colonic Epithelial Cell Medium (CoEpiCM, Cat. No. 2951, ScienCell). Human Retinal Pigment Epithelial Cells (HRPEpiC), Catalog Number 6540 were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell). Human Renal Cortical Epithelial Cells (HRCEpiC), Catalog Number 4110, were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell). Human Choroid Plexus Epithelial Cells (HCPEpiC), Catalog Number 1310 were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell).
  • Example 22 Target Binding by Flow Cytometry
  • Target cells as indicated were harvested with Cell Dissociation Buffer, washed with PBS and resuspended in FACS buffer. The antibody staining was performed in a 96 well round bottom plate. Therefore 200′000 cells were seeded per well. The plate was centrifuged for 4 min at 400 g and the supernatant was removed. The test antibodies were diluted in FACS buffer and 20 μl of the antibody solution were added to the cells for 30 min at 4° C. To remove unbound antibody the cells were washed twice with FACS buffer before addition of the diluted secondary antibody (FITC conjugated AffiniPure F(ab′)2 fragment goat anti-human IgG, Fcg Fragment, Jackson ImmunoResearch #109-096-098 or PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific, Jackson ImmunoResearch #109-116-170. After 30 min incubation on 4° C. unbound secondary antibody was washed away. Before measurement the cells were resuspended in 200 μl FACS buffer and analyzed by flow cytometry using BD Canto II or BD Fortessa.
  • Example 23 Internalization
  • The cells were harvested and the viability was determined. The cells were re-suspended in fresh cold medium at 2 Mio cells per ml and the cell suspension was transferred in a 15 ml falcon tube for each antibody. The antibodies that should be tested for internalization were added with a final concentration of 20 μg per ml to the cells. The tubes were incubated for 45 min in the cold room on a shaker. After incubation the cells were washed three times with cold PBS to remove unbound antibodies. 0.2 Mio cells per well were transfer to the FACS plate as time point zero. The labeled cells were re-suspended in warm medium and incubated at 37° C. At the indicated time-points 0.2 Mio cells per well were transferred in cold PBS, washed in plated on the FACS plate. To detect the constructs that remain on the surface the cells were stained with PE-labeled anti-human Fc secondary antibody. Therefore 20 μl of the diluted antibody were added per well and the plate was incubated for 30 min at 4° C. Then the cells were washed twice to remove unbound antibodies and then fixed with 1% PFA to prevent any further internalization. The fluorescence was measured using BD FACS CantoII.
  • Example 24 QIFIKIT® Analysis
  • QIFIKIT® contains a series of beads, 10 μm in diameter and coated with different, but well-defined quantities of mouse Mab molecules (high-affinity anti-human CD5, Clone CRIS-1, isotype IgG2a). The beads mimic cells with different antigen densities which have been labeled with a primary mouse Mab, isotype IgG. Briefly, cells were labeled with primary mouse monoclonal antibody directed against the antigen of interest. In a separate test well, cells were labeled with irrelevant mouse monoclonal antibody (isotype control). Then, cells, Set-Up Beads and Calibration Beads were labeled with a fluorescein-conjugated anti-mouse secondary antibody included in the kit. The primary antibody used for labeling of the cells has to be used at saturating concentration. The primary antibody may be of any mouse IgG isotype. Under these conditions, the number of bound primary antibody molecules corresponds to the number of antigenic sites present on the cell surface. The secondary antibody is also used at saturating concentration. Consequently, the fluorescence is correlated with the number of bound primary antibody molecules on the cells and on the beads.
  • Example 25 T Cell Mediated Tumor Cell Killing and T Cell Activation
  • Target cells were harvested with Trypsin/EDTA, counted and viability was checked. The cells were resuspended in their respective medium with a final concentration of 300′000 cells per ml. Then 100 μl of the target cell suspension was transferred into each well of a 96-flat bottom plate. The plate was incubated overnight at 37° C. in the incubator to allow adherence of the cells to the plate. On the next day PBMCs were isolated from whole blood from healthy donors. The blood was diluted 2:1 with PBS and overlayed on 15 ml Histopaque-1077 (#10771, Sigma-Aldrich) in Leucosep tubes and centrifuged for 30 min at 450 g without break. After centrifugation the band containing the cells was collected with a 10 ml pipette and transferred into 50 ml tubes. The tubes were filled up with PBS until 50 ml and centrifuged (400 g, 10 min, room temperature). The supernatant was removed and the pellet resuspended in PBS. After centrifugation (300 g, 10 min, room temperature), supernatants were discarded, 2 tubes were pooled and the washing step was repeated (this time centrifugation 350×g, 10 min, room temperature). Afterwards the cells were resuspended and the pellets pooled in 50 ml PBS for cell counting. After counting cells were centrifuged (350 g, 10 min, room temperature) and resuspended at 6 Mio cells per ml in RPMI with 2% FCS and 2 nM Glutamine. Medium was removed from plated target cells and the test antibodies diluted in RPMI with 2% FCS and 2 nM Glutamine were added as well as. 300′000 cells of the effector cell solution were transferred to each well resulting in a E:T ratio of 10:1. To determine the maximal release target cells were lysed with Triton X-100. LDH release was determined after 24 h and 48 h using Cytotoxicity Detection Kit (#1644793, Roche Applied Science). Activation marker upregulation on T cells after tumor cell killing was measured by flow cytometry. Briefly PBMCs were harvested, transferred into a 96 well round bottom plate and stained with CD4 PE-Cy7 (#3557852, BD Bioscience), CD8 FITC (#555634, BD Bioscience), CD25 APC (#555434, BD Bioscience), CD69 PE (#310906, BioLegend) antibodies diluted in FACS buffer. After 30 min incubation at 4° C. the cells were washed twice with FACS buffer. Before measuring the fluorescence using BD Canto II the cells were resuspended in 200 μl FACS buffer.
  • Example 26 T Cell Activation in Whole Blood
  • 280 μl of fresh blood were added into a 96 well conical deep well plate. Then 20 μl of the diluted TCBs were added to the blood and mixed well by shaking the plate. After 24 h incubation at 37° C. in an incubator the blood was mixed and 35 μl were transferred to a 96 well round bottom plate. Then 20 μl of the antibody staining mix were added consisting of CD4 PE-Cy7 (#3557852, BD Bioscience), CD8 FITC (#555634, BD Bioscience), CD25 APC (#555434, BD Bioscience), CD69 PE (#310906, BioLegend) and CD45 V500 (#560777, BD Horizon) and incubated for 15 min in the dark at room temperature. Before measuring 200 μl of the freshly prepared BD FACS lysing solution (#349202, BD FCAS) was added to the blood. After 15 min incubation at room temperature the cells were measured with BD Fortessa.
  • Example 27 SDPK (Single Dose Pharmacokinetics) Study of Humanized FOLR1 TCB (Clone 16D5) in Immunodeficient NOD/Shi-Scid/IL-2Rγnull (NOG) Mice
  • Female NOD/Shi-scid/IL-2Rγnull (NOG) mice, age 6-7 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (P 2011/128). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.
  • Mice were injected i.v. with 10/1/0.1 μg/mouse of the FOLR1 TCB whereas 3 mice were bled per group and time point. All mice were injected with a total volume of 200 μl of the appropriate solution. To obtain the proper amount of the FOLR1 TCB per 200 μl, the stock solutions were diluted with PBS when necessary. Serum samples were collected 5 min, 1 h, 3 h, 8 h, 24 h, 48 h, 72 h, 96 h and 168 h after therapy injection.
  • FIG. 15 shows that the 16D5 FOLR1 TCB shows typical and dose proportional IgG-like PK properties in NOG mice with slow clearance.
  • TABLE 21
    Experimental conditions.
    Formulation Concentration
    Compound Dose buffer (mg/mL)
    FOLR1 TCB 10 μg 20 mM Histidine, 5.43
    (16D5) (corresponding 140 mM NaCl, (=stock solution)
    to ca. 0.5 mg/kg) pH 6.0
    FOLR1 TCB 1 μg 20 mM Histidine, 5.43
    (16D5) (corresponding 140 mM NaCl, (=stock solution)
    to ca. 0.05 mg/kg) pH 6.0
    FOLR1 TCB 0.1 μg 20 mM Histidine, 5.43
    (16D5) (corresponding 140 mM NaCl, (=stock solution)
    to ca. 0.005 mg/kg) pH 6.0
  • Example 28 In Vivo Efficacy of FOLR1 TCB (Clone 16D5) after Human PBMC Transfer in Skov3-Bearing NOG Mice
  • The FOLR1 TCB was tested in the human ovarian carcinoma cell line Skov3, injected s.c. into PBMC engrafted NOG mice.
  • The Skov3 ovarian carcinoma cells were obtained from ATCC (HTB-77). The tumor cell line was cultured in RPMI containing 10% FCS (Gibco) at 37° C. in a water-saturated atmosphere at 5% CO2. Passage 35 was used for transplantation, at a viability >95%. 5×106 cells per animal were injected s.c. into the right flank of the animals in a total of 100 μl of RPMI cell culture medium (Gibco).
  • Female NOD/Shi-scid/IL-2Rγnull (NOG) mice, age 6-7 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (P 2011/128). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.
  • According to the protocol (FIG. 16), mice were injected s.c. on study day 0 with 5×106 of the Skov3. At study day 21, human PBMC of a healthy donor were isolated via the Ficoll method and 10×106 cells were injected i.p. into the tumor-bearing mice. Two days after, mice were randomized and equally distributed in five treatment groups (n=12) followed by i.v. injection with either 10/1/0.1 μg/mouse of the FOLR1 TCB or 10 μg/mouse of the DP47 control TCB once weekly for three weeks. All mice were injected i.v. with 200 μl of the appropriate solution. The mice in the vehicle group were injected with PBS. To obtain the proper amount of TCB per 200 μl, the stock solutions were diluted with PBS when necessary. Tumor growth was measured once weekly using a caliper (FIG. 17) and tumor volume was calculated as followed:

  • T v:(W 2/2)×L(W:Width,L:Length)
  • The once weekly injection of the FOLR1 TCB resulted in a dose-dependent anti-tumoral effect. Whereas a dose of 10 μg/mouse and 1 μg/mouse induced tumor shrinkage and 0.1 μg/mouse a tumor stasis (FIG. 17, Table 22). Maximal tumor shrinkage was achieved at a dose of 10 μg/mouse as compared to a non-targeted control DP47 TCB.
  • TABLE 22
    In vivo efficacy.
    Tumor growth
    Compound Dose inhibition
    DP47 TCB
    10 μg 7%
    control TCB (corresponding
    to ca. 0.5 mg/kg)
    FOLR1 TCB 10 μg 90%
    (16D5) (corresponding
    to ca. 0.5 mg/kg)
    FOLR1 TCB 1 μg 74%
    (16D5) (corresponding
    to ca. 0.05 mg/kg)
    FOLR1 TCB 0.1 μg 56%
    (16D5) (corresponding
    to ca. 0.005 mg/kg)
  • For PD read-outs, three mice per treatment group were sacrificed at study day 32, tumors were removed and single cell suspensions were prepared through an enzymatic digestion with Collagenase V, Dispase II and DNAse for subsequent FACS-analysis (FIGS. 19 and 20). Single cells where either used directly for staining of extracellular antigens and activation markers or were re-stimulated using 5 ng/ml PMA and 500 ng/ml lonomycin in the presence of a protein transport inhibitor Monensin for 5h in normal culture medium. After re-stimulation, cells were stained for surface antigens, followed by a fixation and permeabilization step. Fix samples were then stained intracellulary for TNF-α, IFN-γ, IL-10 and IL-2 and analyzed by flow cytometry. Same procedure was used for the degranulation of cells, but an anti-CD107a antibody was added during the restimulation period and fixed samples were staining for intracellular perforin and granzyme-B contents. The FACS analysis revealed statistically higher number of infiltrating CD4+ and CD8+ T-cells in the tumor tissue upon treatment with FOLR1 TCB compared to vehicle and untargeted control TCB. Furthermore, higher numbers of TNF-α, IFN-γ and IL-2 producing as well as perforin+/granzym-B+ CD4+ and CD8+ T-cells were detected in FOLR1 TCB treated tumors. Tumor infiltrating T-cells treated with FOLR1 TCB also showed higher degranulation rates compared to control groups.
  • At study termination day 38, all animals were sacrificed; tumors were removed and weight (FIG. 18). The weight of the tumors treated with 10 and 1 μg/mouse of the FOLR1 TCB showed a statistically significant difference compared to the control groups.
  • TABLE 23
    Experimental conditions.
    Concentration
    Compound Dose Formulation buffer (mg/mL)
    PBS
    FOLR1 TCB
    10 μg 20 mM Histidine, 3.88
    (16D5) 140 mM NaCl, (= stock solution)
    pH6.0
    FOLR1 TCB  1 μg 20 mM Histidine, 3.88
    (16D5) 140 mM NaCl, (= stock solution)
    pH6.0
    FOLR1 TCB 0.1 μg  20 mM Histidine, 3.88
    (16D5) 140 mM NaCl, (= stock solution)
    pH6.0
    DP47 TCB 10 μg 20 mM Histidine, 4.35
    140 mM NaCl, (= stock solution)
    pH6.0
  • Example 29 Generation of a Bispecific FolR1/CD3-Kappa-Lambda Antibody
  • To generate a bispecific antibody (monovalent for each antigen) that simultaneously can bind to human CD3 and human folate receptor alpha (FolR1) without using any hetero-dimerization approach (e.g. knob-into-hole technology), a combination of a common light chain library with the so-called CrossMab technology was applied: The variable region of the humanized CD3 binder (CH2527_VL7_46/13) was fused to the CH1 domain of a standard human IgG1 antibody to form the VLVH crossed molecule (fused to Fc) which is common for both specificities. To generate the crossed counterparts (VHCL), a CD3 specific variable heavy chain domain (CH2527_VH_23/12) was fused to a constant human λ light chain whereas a variable heavy chain domain specific for human FolR1 (clone 16D5, isolated from common light chain library) was fused to a constant human κ light chain. This enables the purification of the desired bispecific antibody by applying subsequent purification steps with KappaSelect and LambdaFabSelect columns (GE Healthcare) to remove undesired homodimeric antibodies.
  • All antibody expression vectors were generated using standard recombinant DNA technology as described in Sambrook, J. et al., Molecular cloning: A laboratory manual; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. Molecular biological reagents were used according the manufacturer's recommendations. Genes or gene fragments were either amplified by polymerase chain reaction (PCR) or generated from synthetic oligonucleotides at Geneart AG (Regensburg, Germany) by automated gene synthesis. PCR-amplified or subcloned DNA fragments were confirmed by DNA sequencing (Synergene GmbH, Switzerland). Plasmid DNA was transformed into and amplified in suitable E. coli host strains for preparation of transfection-grade plasmid DNA using standard Maxiprep kits (Qiagen). For production of the bispecific molecules HEK293 EBNA cells were transfected with plasmids encoding the respective genes using a standard polyethlenimine (PEI) based method. The used plasmid ratio of the three expression vectors was 1:1:1. Transfected cells were cultivated for 7 days before supernatants were harvested for purification. The bispecific FolR1/CD3-kappa-lambda antibodies were produced and purified as follows.
  • 1. Transient Transfection and Production
  • The kappa-lambda bispecific antibody was transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells were centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200m DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C.
  • 2. Purification
  • The kappa-lambda bispecific antibody was purified in three steps, using an affinity step specific for kappa light chains, followed by an affinity step specific for lambda light chains and finally by a size exclusion chromatography step for removal of aggregates. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to Capture Select kappa affinity matrix, or HiTrap KappaSelect, GE Healthcare, column volume (cv)=1 ml, equilibrated with 5 column volumes (cv) buffer A (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 8.0). After washing with 15 cv of buffer A, the protein was eluted using a pH gradient to buffer B (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0) over 25 cv. Fractions containing the protein of interest were pooled and the pH of the solution was adjusted to pH 8.0 (using 2 M Tris pH 8.0). The neutralized pooled fractions were applied to Capture Select lambda affinity matrix (now: HiTrap LambdaFabSelect, GE Healthcare, column volume (cv)=1 ml) equilibrated with 5 column volumes (cv) buffer A (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 8.0). After washing with 15 cv of buffer A, the protein was eluted using a pH gradient to buffer B (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0) over 25 cv. Fractions containing the protein of interest were pooled and the pH of the solution was adjusted to pH 8.0 (using 2 M Tris pH 8.0). This solution was concentrated using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a Superdex™ 200 10/300 GL (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The pooled fractions after size exclusion were again concentrated using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius).
  • The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Only small amounts of protein could be purified with a final yield of 0.17 mg/L.
  • Example 30 T Cell Mediated Killing with Bispecific FolR1/CD3-Kappa-Lambda Antibody
  • Activity of kappa lambda FolR1 TCB was tested on SKOV3 cells in the presence of freshly isolated PBMCs. As negative control DP47 TCB was included. T cell mediated killing of SKOV3 cells was determined after 24 h and 48 h by LDH release. After 48 h the T cells were harvested and CD69 and CD25 upregulation on CD4 T cells and CD8 T cells was measured by flow cytometry.
  • The kappa lambda FolR1 construct induces killing of SKOV3 cells in a concentration dependent manner which is accompanied by CD69 and CD25 upregulation both on CD4 T cells and on CD8 T cells.
  • SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 24 h and 48 h killing of tumor cells was determined by measuring LDH release (FIG. 21). SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 48 h CD25 and CD69 upregulation on CD4 T cells and CD8 T cells was measured by flow cytometry (FIG. 22).
  • Example 31 Biochemical Characterization of 16D5 and 36F2 FolR1 Binders by Surface Plasmon Resonance
  • Binding of anti-FolR1 16D5 in different monovalent or bivalent T-cell bispecific formats and of anti-FolR1 36F2 as IgG or as T-cell bispecific to recombinant human, cynomolgus and murine folate receptor 1 (all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, GE Healthcare).
  • 1. Molecules Tested
  • The molecules used for affinity and avidity determination are described in Table 24.
  • TABLE 24
    Name and description of the 6 constructs used in SPR analysis
    Name Description
    16D5 TCB
    2 + 1 T-cell bispecific, inverted
    format (common light chain)
    16D5 TCB classical 2 + 1 T-cell bispecific, classical
    format (common light chain)
    16D5 TCB 1 + 1 1 + 1 T-cell bispecific
    (common light chain)
    16D5 TCB 1 + 1 HT 1 + 1 T-cell bispecific head-to-
    tail (common light chain)
    36F2 IgG Human IgG1 with P329G
    LALA
    36F2 TCB
    2 + 1 T-cell bispecific, inverted
    format, crossfab
  • 2. Avidity to Folate Receptor 1
  • The avidity of the interaction between the anti-FolR1 IgG or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 25).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, GE Healthcare). The immobilization level was about 300-400 RU. The anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 3.7 to 900 nM with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 240 or 600 seconds. The chip surface was regenerated after every cycle using a double injection of 30 sec 10 mM Glycine-HCl pH 2. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine CD134 Fc fusion. The binding curves resulting from the bivalent binding of the IgG or T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding. The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare). For the 1+1 T cell bispecifics format the interaction is a real 1:1 and the KD represents affinity since there is only one FolR1 binder in this construct.
  • TABLE 25
    Bivalent binding (avidity with apparent KD) of anti-FolR1 16D5 and 36F2
    as IgG or as T-cell bispecifics (TCB) on human, cyno and murine FolR1.
    Apparent
    Analyte Ligand ka (1/Ms) kd (1/s) KD
    36F2 IgG huFolR1 2.07E+06  1.3E−02 6 nM
    cyFolR1 2.78E+06 1.75E−02 6 nM
    muFolR1 4.28E+05 8.23E−04 2 nM
    36F2 TCB huFolR1 2.45E+06 9.120E−03  4 nM
    cyFolR1 4.31E+06 1.45E−02 3 nM
    muFolR1 6.97E+05 9.51E−04 1 nM
    16D5 TCB huFolR1 1.57E+05 3.92E−04 3 nM
    cyFolR1 2.01E+05 3.81E−04 2 nM
    16D5 TCB classical huFolR1 2.04E+05 1.84E−04 0.9 nM  
    cyFolR1 2.50E+05 3.05E−04 1 nM
    16D5 TCB
    1 + 1 HT huFolR1 5.00E+04 2.25E−03 45 nM 
    cyFolR1 5.75E+04 4.10E−03 70 nM 
    16D5 TCB
    1 + 1 huFolR1 3.65E+04 2.04E−03 56 nM 
    cyFolR1 4.09E+04 3.60E−03 90 nM 
  • 3. Affinity to Folate Receptor 1
  • The affinity of the interaction between the anti-FolR1 IgG or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 26).
  • For affinity measurement, direct coupling of around 12000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 IgG or T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 40 sec, the reference flow cell was left without capture. Dilution series (12.3 to 3000 nM) of human, cyno or murine Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 300 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • TABLE 26
    Monovalent binding (affinity) of anti-FolR1 16D5 and 36F2 as IgG or
    as T-cell bispecifics (TCB) on human, cyno and murine FolR1.
    Analyte Ligand ka (1/Ms) kd (1/s) KD
    36F2 IgG huFolR1 9.10E+04 6.65E−02 730 nM
    cyFolR1 1.02E+05 5.78E−02 570 nM
    muFolR1 8.32E+04 1.78E−02 210 nM
    36F2 TCB huFolR1 5.94E+04 6.13E−02 1000 nM 
    cyFolR1 6.29E+04 5.42E−02 860 nM
    muFolR1 5.68E+04 1.75E−02 300 nM
    16D5 TCB huFolR1 2.23E+04 7.33E−04  33 nM
    cyFolR1 1.57E+04 1.60E−03 100 nM
    16D5 TCB classical huFolR1 1.03E+04 7.59E−04  74 nM
    cyFolR1 9.18E+03 1.61E−03 175 nM
    16D5 TCB 1 + 1 HT huFolR1 2.05E+04 7.08E−04  35 nM
    cyFolR1 1.67E+04 1.53E−03  92 nM
    16D5 TCB 1 + 1 huFolR1 1.43E+04 9.91E−04  69 nM
    cyFolR1 1.20E+04 1.80E−03 150 nM
  • The affinity (monovalent binding) to human and cyno FolR1-Fc of 36F2 TCB is similar and around 1000 nM for both, whereas the affinity to murine FolR1-Fc is slightly better and around 300 nM. The 36F2 can be used in murine and primate models, there is no need for a surrogate.
  • The avidity (apparent KD) of 36F2 TCB to human FolR1 is around 30 times lower than the affinity of the 16D5 TCB to human FolR1. In the bivalent format, 36F2 TCB is in the low nanomolar range, whereas 16D5 TCB is in the low picomolar range (1000 fold difference).
  • FolR1 is expressed on tumor cells overexpressed, at intermittent and high levels, on the surface of cancer cells in a spectrum of epithelial malignancies, including ovarian, breast, renal, colorectal, lung and other solid cancers and is also expressed on the apical surface of a limited subset of polarized epithelial cells in normal tissue. These non-tumorous, normal cells express FolR1 only at low levels, and include, e.g., bronchiolal epithelial cells on alveolar surface, renal cortical luminal border of tubular cells, retinal pigment epithelium (basolateral membrane) and choroid plexus. 16D5 TCB binds to normal tissues cells expressing low amounts of FolR1 which results in their T cell mediated killing. This might, at least in part, account for limited tolerance observed at 10 μg/kg in cynomolgus monkeys. The inventors wanted to determine if lowering the affinity of the T cell bispecific molecule could increase the differentiation between high and low target density tissues and, thereby, lower toxicity by making use of bivalent binding and avidity. Low affinity binders are ordinarily not selected as suitable candidates for further analysis because low affinity is often associated with low potency and efficacy. Nevertheless, the low affinity FolR1 binder 36F2 was developed in several formats and characterized for its biological properties. For the 36F2 used in the bivalent T cell bispecific format the avidity effect (difference between monovalent and bivalent binding) is around 250 fold (1000 nM versus 4 nM). At low target density the affinity defined the interaction and with 1000 nM led to a low potency of the TCB. However, at high target density the molecule's avidity comes into play and with 4 nM led to a high activity of the TCB (see Example 32).
  • In an alternatively approach, the inventors generated monovalent formats of 16D5 and low affinity variant of 16D5 (affinity about 10-40 nM) in a bivalent format. The 16D5 binder used in a monovalent format (1+1) has an affinity of about 50 nM. The differentiation between high and low target density tissues can be better achieved by taking advantage of the avidity effect.
  • Example 32 T-Cell Killing of SKov-3 Cells Induced by 36F2 TCB, Mov19 TCB and 21A5 TCB
  • T-cell killing mediated by 36F2 TCB, Mov19 TCB and 21A5 TCB was assessed on SKov-3 cells (medium FolR1). Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.005 pM-5 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h and 48 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.
  • The results show that the killing induced by 36F2 is strongly reduced in comparison to Mov19 TCB and 21A5 TCB (FIGS. 23A-B). The EC50 values related to killing assays, calculated using GraphPadPrism6 are summarized in Table 27.
  • TABLE 27
    EC50 values (pM) for T-cell mediated killing of FolR1-expressing
    SKov-3 cells induced by 36F2 TCB, Mov19 TCB and 21A5 TCB.
    EC50 [pM]
    Antibody 24 h 48 h
    36F2 TCB 1406.07* 134.5
    Mov19 TCB 0.75 0.05
    21A5 TCB 2.83 0.10
    *curve did not reach saturation, value is hypothetical
  • Example 33 T-Cell Killing Induced by 36F2 TCB and 16D5 TCB in Different Monovalent and Bivalent T-Cell Bispecific Formats
  • T-cell killing mediated by 36F2 TCB, 16D5 TCB, 16D5 TCB classical, 16D5 TCB 1+1 and 16D5 TCB HT antibodies of Hela (high FolR1, about 2 million copies, Table 14, FIG. 27), Skov-3 (medium FolR1, about 70000-90000 copies, Table 14, FIG. 27) and HT-29 (low FolR1, about 10000, Table 14, FIG. 27) human tumor cells was assessed. DP47 TCB antibody was included as negative control. Human PBMCs were used as effectors and the killing was detected at 24 h of incubation with the bispecific antibody. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-100 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.
  • The results show that target-specific killing of all three FolR1+ target cell lines induced by 36F2 TCB is much weaker compared to the killing induced by 16D5 TCB (FIGS. 24A-C, Table 29). Target-specific killing induced by the monovalent 16D5 TCBs (16D5 HT and 16D5 1+1) is worse compared to the bivalent 16D5 TCBs (16D5 TCB and 16D5 TCB classical). The EC50 values related to killing assays, calculated using GraphPadPrism6, are summarized in Table 28. Importantly, this data shows that using the 36F2 FolR1 binder in the bivalent 2+1 TCB format widens the therapeutic window compared to the 16D5 FOLR1 TCB (FIG. 24A-C). Whereas the potency reduction between 16D5 and 36F2 FOLR1 TCB is approximately 45-fold for Hela cells (high FOLR1 expression, see Table 28: 16D5 TCB=0.8 versus 36F2 TCB 36.0) and approximately 297-fold for Skov3 cells (medium FOLR1 expression, see Table 28: 16D5 TCB=0.6 versus 36F2 TCB 178.4), this reduction is almost 7000-fold for HT29 with low FOLR1 expression (see Table 28: 16D5 TCB=5.7 versus 36F2 TCB 39573). Thus, the 36F2 FOLR1 TCB differentiates between high and low expressing cells which is of special importance to reduce toxicity as the cells of some normal, non-tumorous tissues express very low levels of FolR1 (approximately less than 1000 copies per cell). Consistent with this observation, the results discussed in Example 35 below show that 36F2 TCB does not induce T-cell killing of primary cells (FIGS. 26A-D) whereas for 16D5 TCB some killing can be observed on HRCEpiC and HRPEpiC cells after 48 h of incubation (FIGS. 26B and C). This important characteristic of 36F2 TCB allows for dosing for the treatment of FolR1-positive tumors so that it mediates potent killing of tumor tissues with high or medium FOLR1 expression, but not of normal tissues with low (partially polarized) expression. Notably, this characteristic appears to be mediated by the avidity of 36F2 TCB in the bivalent 2+1 inverted format, as it was not observed when using the 1+1 monovalent formats carrying the same low affinity 36F2 binder.
  • Stated another way, 36F2 TCB in the bivalent 2+1 format comprises FolR1 binding moieties of relatively low affinity but it possesses an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this TCB selectively binds to tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all.
  • In addition to the above advantageous characteristics, the 36F2 TCB in the bivalent 2+1 inverted format also has the advantage that it does not require chemical cross linking or other hybrid approach. This makes it suitable for manufacture of a medicament to treat patients, for example patients having FolR1-positive cancerous tumors. The 36F2 TCB in the bivalent 2+1 inverted format can be produced using standard CHO processes with low aggregates. Further, the 36F2 TCB in the bivalent 2+1 comprises human and humanized sequences making it superior to molecules that employ rat and murine polypeptides that are highly immunogenic when administered to humans. Furthermore, the 36F2 TCB in the bivalent 2+1 format was engineered to abolish FcgR binding and, as such, does not cause FcgR crosslinking and infusion reactions, further enhancing its safety when administered to patients.
  • As demonstrated by the results described above, its head-to-tail geometry make the 36F2 TCB in the bivalent 2+1 inverted format a highly potent molecule that induces absolute target cell killing. Its bivalency enhance avidity and potency, but also allow for differentiation between high and low expressing cells. Its preference for high or medium target expressing cells due to its avidity affect reduce toxicity resulting from T cell mediated killing of normal cells that express FolR1 at low levels.
  • A further advantage of the 36F2 TCB in the bivalent 2+1 format and other embodiments disclosed herein is that their clinical development does not require the use of surrogate molecules as they bind to human, cynomous and murine FolR1. As such, the molecules disclosed herein recognize a different epitope than antibodies to FolR1 previously described that do not recognize FolR1 from all three species.
  • TABLE 28
    EC50 values (pM) for T-cell mediated killing of FolR1-expressing tumor
    cells induced by 36F2 TCB and 16D5 TCB in different monovalent and
    bivalent T-cell bispecific formats after 24 h of incubation.
    Skov-3
    Hela (FolR1 HT-29
    Antibody (FolR1 high) medium) (FolR1 low)
    16D5 TCB 0.8 0.6 5.7
    16D5 TCB 4.6 2.0 13.0
    classical
    16D5 TCB 11.6 12.3 15.1
    HT
    16D5 TCB 23.8 48.9 883.8*
    1 + 1
    36F2 TCB 36.0 178.4 39573.0*
    *curve did not reach saturation, only hypothetical value
  • Table 29 shows a comparison of EC50 values of 16D5 TCB and 36F2 TCB on the different cell lines tested. Out of the obtained EC50 values the delta (EC50 of 16D5 TCB minus EC50 of 36F2 TCB) and the x-fold difference (EC50 of 16D5 TCB divided by the EC50 of 36F2 TCB) was calculated.
  • TABLE 29
    Comparison of EC50 values of 16D5 TCB and 36F2 TCB.
    Hela Skov-3 HT-29
    Antibody (FolR1 high) (FolR1 medium) (FolR1 low)
    16D5 TCB 0.82 0.63 5.73
    36F2 TCB 35.99 178.40 39573.00*
    Δ 35.17 177.77 39567.27
    x-fold 43.83 284.61 6906.58
    *curve did not reach saturation, only hypothetical value
  • The calculated EC50 values clearly show that the difference between 36F2 TCB and 16D5 TCB gets larger the lower the FolR1 expression on the target cells is.
  • The same calculations as done for the comparison of the EC50 values of 16D5 TCB and 36F2 TCB were done for 16D5 TCB and the two monovalent 16D5 TCBs (16D5 TCB HT and 16D5 1+1). Tables 30 and 31 show the comparisons of the EC50 values of 16D5 TCB vs 16D5 TCB HT (Table 30) and 16D5 TCB vs 16D5 TCB 1+1 (Table 31) as well as the corresponding deltas (EC50 of 16D5 TCB minus EC50 of 16D5 TCB HT/1+1) and the x-fold differences (EC50 of 16D5 TCB divided by the EC50 of 16D5 TCB HT/1+1).
  • TABLE 30
    Comparison of EC50 values of 16D5 TCB (2 + 1 inverted) and
    16D5 TCB HT.
    Hela Skov-3 HT-29
    Antibody (FolR1 high) (FolR1 medium) (FolR1 low)
    16D5 TCB 0.82 0.63 5.73
    16D5 TCB HT 11.61 12.27 15.11
    Δ 10.79 11.65 9.38
    x-fold 14.14 19.58 2.64
  • TABLE 31
    Comparison of EC50 values of 16D5 TCB and 16D5 TCB 1 + 1.
    Hela Skov-3 HT-29
    Antibody (FolR1 high) (FolR1 medium) (FolR1 low)
    16D5 TCB 0.82 0.63 5.73
    16D5 TCB 1 + 1 23.84 48.86 883.78*
    Δ 23.02 48.24 878.05
    x-fold 29.03 77.95 154.24
    *curve did not reach saturation, only hypothetical value
  • The comparison of the EC50 values of 16D5 TCB and 36F2 TCB (Table 29) shows that the difference in the EC50 values gets larger the lower the FolR1 expression on the target cells is. This effect cannot be seen in the comparison of 16D5 TCB and the monovalent 16D5 TCBs (Table 29 and Table 30). For 16D5 TCB 1+1 (Table 31) there is also a slight increase in the difference between the EC50 of 16D5 TCB and 16D5 TCB 1+1 with decreasing FolR1 expression but by far not as pronounced as can be seen in the comparison of 16D5 TCB vs 36F2 TCB.
  • Example 34 CD25 and CD69 Upregulation on CD8+ and CD4+ Effector Cells after T Cell-Killing of FolR1-Expressing Tumor Cells Induced by 36F2 TCB and 16D5 TCB Antibody
  • Activation of CD8+ and CD4+ T cells after T-cell killing of FolR1-expressing Hela, SKov-3 and HT-29 tumor cells mediated by 36F2 TCB and 16D5 TCB was assessed by FACS analysis using antibodies recognizing the T cell activation markers CD25 (late activation marker) and CD69 (early activation marker). DP47 TCB was included as non-binding control. The antibody and the killing assay conditions were essentially as described above (Example 32) using the same antibody concentration range (0.01 pM-100 nM in triplicates), E:T ratio 10:1 and an incubation time of 48h. After the incubation, PBMCs were transferred to a round-bottom 96-well plate, centrifuged at 400×g for 4 min and washed twice with PBS containing 0.1% BSA. Surface staining for CD8 (PE anti-human CD8, BD #555635), CD4 (Brilliant Violet 421™ anti-human CD4, Biolegend #300532), CD69 (FITC anti-human CD69, BD #555530) and CD25 (APC anti-human CD25 BD #555434) was performed according to the manufacturer's instructions. Cells were washed twice with 150 μl/well PBS containing 0.1% BSA. After centrifugation, the samples were resuspended in 200 μl/well PBS 0.1% for the FACS measurement. Samples were analyzed at BD FACS Canto II. 36F2 TCB induced a target-specific up-regulation of activation markers (CD25, CD69) on CD8+ and CD4+ T cells after killing of Hela (FIG. 25A) and SKov-3 (FIG. 25B) cells. In comparison to 16D5 TCB the up-regulation of CD25 and CD69 on CD8+ and CD4+ T cells induced by 36F2 is much weaker.
  • On HT-29 (low FolR1) an up-regulation of activation markers can only be seen at the highest concentration of 36F2 TCB. In contrast, with 16D5 TCB up-regulation of CD25 and CD69 can be seen already at much lower antibody concentrations (FIG. 25C).
  • As seen as well in the tumor lysis experiment, the analysis of activation markers (CD25 and CD69) on T cells (CD4+ and CD8+) after killing clearly shows that the difference between 16D5 TCB and 36F2 TCB becomes larger the lower the FolR1 expression level on the target cells is.
  • Example 35 T-Cell Killing of Primary Cells Induced by 36F2 TCB and 16D5 TCB
  • T-cell killing mediated by 36F2 TCB and 16D5 TCB was assessed on primary cells (Human Renal Cortical Epithelial Cells (HRCEpiC) (ScienCell Research Laboratories; Cat No 4110) and Human Retinal Pigment Epithelial Cells (HRPEpiC) (ScienCell Research Laboratories; Cat No 6540)). HT-29 cells (low FolR1) were included as control cell line. DP47 TCB served as non-binding control. Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h and 48 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.
  • The results show that 36F2 TCB does not induce T-cell killing of primary cells (FIG. 26A-D) whereas for 16D5 TCB some killing can be observed on HRCEpiC and HRPEpiC cells after 48 h of incubation (FIGS. 26B and D). As described above, a strong difference in T-cell killing between of HT-29 cells was observed between 16D5 TCB and 36F2 TCB (FIG. 26E, F).
  • Example 36 Preparation of DP47 GS TCB (2+1 Crossfab-IgG P329G LALA Inverted=“Untargeted TCB”)
  • The “untargeted TCB” was used as a control in the above experiments. The bispecific antibody engages CD3e but does not bind to any other antigen and therefore cannot crosslink T cells to any target cells (and subsequently cannot induce any killing). It was therefore used as negative control in the assays to monitor any unspecific T cell activation. This untargeted TCB was prepared as described in WO2014/131712. In brief, the variable region of heavy and light chain DNA sequences have been subcloned in frame with either the constant heavy chain or the constant light chain pre-inserted into the respective recipient mammalian expression vector. The antibody expression was driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS. In addition each vector contains an EBV OriP sequence.
  • The molecule was produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using polyethylenimine. The cells were transfected with the corresponding expression vectors in a 1:2:1:1 ratio (“vector heavy chain Fc(hole)”:“vector light chain”:“vector light chain Crossfab”:“vector heavy chain Fc(knob)-FabCrossfab”).
  • For transfection HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection. For transfection cells were centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200 g DNA. After addition of 540 μl PEI solution was vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210×g, the solution was sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C.
  • The secreted protein was purified from cell culture supernatants by affinity chromatography using ProteinA. Supernatant was loaded on a HiTrap ProteinA HP column (CV=5 mL, GE Healthcare) equilibrated with 40 ml 20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5. Unbound protein was removed by washing with at least 10 column volume 20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5. Target protein was eluted during a gradient over 20 column volume from 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5 to 20 mM sodium citrate, 0.5 M sodium chloride, pH 2.5. Protein solution was neutralized by adding 1/10 of 0.5 M sodium phosphate, pH 8. Target protein was concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM Histidine, 140 mM sodium chloride solution of pH 6.0.
  • The protein concentration of purified protein samples was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence.
  • Purity and molecular weight of molecules were analyzed by CE-SDS analyses in the presence and absence of a reducing agent. The Caliper LabChip GXII system (Caliper lifescience) was used according to the manufacturer's instruction. 2 ug sample is used for analyses.
  • The aggregate content of antibody samples was analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-Arginine Monohydrocloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C.
  • TABLE 32
    Summary production and purification of DP47 GS TCB.
    Aggregate
    after 1st Mono-
    Titer Yield purification HMW LMW mer
    Construct [mg/l] [mg/l] step [%] [%] [%] [%]
    DP47 GS 103.7 8.04 8 2.3 6.9 91.8
    TCB
  • TABLE 33
    CE-SDS analyses of DP47 GS TCB.
    Peak kDa Corresponding Chain
    DP47 GS TCB non reduced 1 165.22 Molecule with 2 missing
    (A) light chains
    2 181.35 Molecule with 1 missing
    light chain
    3 190.58 Correct molecule without
    N-linked glycosylation
    4 198.98 Correct molecule
    DP47 GS TCB reduced (B) 1 27.86 Light chain DP47 GS
    2 35.74 Light chain huCH2527
    3 63.57 Fc (hole)
    4 93.02 Fc (knob)
  • Example 37 Binding of 16D5 TCB and 9D11 TCB and their Corresponding CD3 Deamidation Variants N100A and S100aA to CD3-Expressing Jurkat Cells
  • The binding of 16D5 TCB and the corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and the demidation variants 9D11 TCB N100A and 9D11 TCB S100aA to human CD3 was assessed on a CD3-expressing immortalized T lymphocyte line (Jurkat). Briefly, cells were harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with different concentrations of the bispecific antibodies (686 pM-500 nM). After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-116-170). After washing the samples twice with cold PBS 0.1% BSA they were immediately analyzed by FACS using a FACS CantoII (Software FACS Diva). Binding curves were obtained using GraphPadPrism6 (FIG. 28A-B).
  • The results show reduced binding of the deamidation variants N100A and S100aA to CD3 compared to the parental antibodies 16D5 TCB (FIG. 28A) and 9D11 TCB (FIG. 28B).
  • Example 38 T-Cell Killing of SKov-3 and HT-29 Cells Induced by 16D5 TCB and 9D11 TCB and their CD3 Deamidation Variants N100A and S100aA
  • T-cell killing mediated by 16D5 TCB and the corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and the demidation variants 9D11 TCB N100A and 9D11 TCB S100aA was assessed on SKov-3 (medium FolR1) and HT-29 (low FolR1) cells. Human PBMCs were used as effectors and the killing was detected at 24 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at a density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.
  • The results show that on SKov-3 cells the killing induced by the CD3 deamidation variants 16D5 TCB N100A and 16D5 S100aA is comparable to the one induced by 16D5 TCB (FIG. 29A). The same is true for 9D11 TCB and its variants 9D11 TCB N100A and 9D11 TCB S100aA (FIG. 29B). On FolR1 low expressing HT-29 cells the S100aA variant shows an impaired killing efficiency which is the case for 16D5 TCB (FIG. 30A) as well as for 9D11 TCB (FIG. 30B). The EC50 values related to killing assays, calculated using GraphPadPrism6 are given in Table 35.
  • TABLE 35
    EC50 values (pM) for T-cell mediated killing of FolR1-expressing SKov-3
    and HT-29 cells induced by 16D5 TCB and 9D11 TCB and their
    deamidation variants N100A and A100aA.
    EC50 [pM]
    Antibody SKov-3 HT-29
    16D5 TCB 1.283 56.67
    16D5 TCB 1.886 91.95
    N100A
    16D5 TCB 1.939 165.6
    S100aA
    9D11 TCB 1.283 2.827
    9D11 TCB 1.886 37.72
    N100A
    9D11 TCB 1.939 n.d.*
    S100aA
    *not determined
  • Example 39 Biochemical Characterization by Surface Plasmon Resonance as TCBs of Two CD3 Binder Variants (N100A and S100aA) to Remove a Deamidation Site
  • Binding of two 16D5 TCBs with CD3 binder variants (N100A or S100aA) to human recombinant CD3 (CD3epsilon-CD3delta heterodimer as Fc fusion) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).
  • Affinity to CD3ed-Fc
  • The affinity of the interaction between the anti-FolR1 T cell bispecifics and the recombinant CD3 epsilon-delta heterodimer was determined as described below (Table 36).
  • For affinity measurement, direct coupling of around 6000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CMS chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 T cell bispecifics were captured at 200 nM with a flow rate of 20 μl/min for 60 sec, the reference flow cell was left without capture. Dilution series (4.1 to 3000 nM) of human and cyno Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 240 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • TABLE 36
    Monovalent binding (affinity) of two 16D5 CD3 deamidation variants
    as TCBs on human CD3ed-Fc.
    Ligand Analyte ka (1/Ms) kd (1/s) KD
    16D5 TCB N100A huCD3 1.23E+04 4.67E−03 380 nM
    16D5 TCB S100aA huCD3 1.21E+04 5.49E−03 460 nM
    16D5 TCB huCD3 2.03E+04 4.41E−03 220 nM
  • The two CD3 deamidation variants have a slightly reduced affinity compared to the wild-type CD3 binder (CH2527), but the difference is not grave.
  • Example 40 Production and Purification of Two Variants of the 16D5 T-Cell Bispecific with Mutations to Remove the Deamidation Site in the CD3 Binder: 16D5 TCB N100A, 16D5 TCB S100aA Transient Transfection and Production
  • The two deamidation variants 16D5 TCBs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200m DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C. After production the supernatant was harvested, filtered through 0.22 μm sterile filters and stored at 4° C. until purification.
  • Purification
  • The two deamidation variants 16D5 TCBs were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to MabSelect SuRe (GE Healthcare, column volume (cv)=2 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate pH 7.5, 20 mM sodium citrate). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine) over 20 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 0.5 M Na2HPO4 pH 8.0). Samples were concentrated to 1 ml using ultra-concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore) and subsequently applied to a HiLoad™ 16/60 Superdex™ 200 preparative grade (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled. The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at −80° C.
  • TABLE 36
    Yield, monomer content and purity by CE-SDS of the two 16D5
    deamidation variants in the T cell bispecific format.
    Mutation in the Yield Purity by CE-
    Name CD3 binder [mg/L] Monomer [%] SDS [%]
    16D5 TCB N100A 9 100 89
    16D5 TCB S100aA 23 100 83
    16D5 TCB Wild-type 9 100 93
  • Both TCBs were produced in good quality, similar to the construct with the wild-type CD3 binder.
  • Example 41 Production and Purification of Two 16D5 Binder Variants (D52dE and D52dQ) as IgGs to Remove a Hotspot in the CDR
  • Transient Transfection and Production
  • The two IgGs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 μg DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C. After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 μm sterile filters and stored at 4° C. until purification.
  • Antibody Purification
  • The two IgGs were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to POROS MabCapture A (Applied Biosystems, column volume (cv)=1 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate, 20 mM sodium citrate, pH 7.5). After washing with 10 cv of buffer A, the protein was eluted using a pH step to buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine) over 5 cv. The 5 ml containing the protein of interest are stored in a loop on the Akta Explorer and subsequently applied to a HiLoad 16/60 Superdex™ 200 (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl (no TWEEn was used). Fractions containing the IgGs were pooled and concentrated using ultra concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore). The aggregate content of the final pool was analyzed by analytical size exclusion chromatography. Therefore, 30 μl were applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were stored at 4° C.
  • TABLE 37
    Yield, monomer content and purity by CE-SDS of two 16D5 IgG
    hotspot variants.
    Mutation Monomer Purity by CE-
    Clone HC/LC Yield [mg/L} [%] SDS [%]
    16D5 D52dE 24 100 96
    16D5 D52dQ 20 100 96
  • Both IgGs produced well and in good quality.
  • Example 42 Biochemical Characterization by Surface Plasmon Resonance of Two 16D5 Binder Variants (D52dE and D52dQ) as IgGs to Remove a Hotspot in the CDR
  • Binding of two 16D5 binder variants (D52dE and D52dQ) as IgGs to human and cyno recombinant folate receptor 1 (both as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).
  • 1. Avidity to Folate Receptor 1
  • The avidity of the interaction between the anti-FolR1 IgGs or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 38).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 160. The anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 3.7 to 900 nM with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 600 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine IL2 receptor Fc fusion. The binding curves resulting from the bivalent binding of the IgG or T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding. The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).
  • TABLE 38
    Bivalent binding (avidity with apparent KD) of two 16D5
    hot spot variants as IgGs on human, murine and cyno FolR1
    (no binding on muFolR1 as expected).
    Analyte Ligand ka (1/Ms) kd (1/s) Apparent KD
    16D5 D52dE IgG huFolR1 1.62E+05 5.45E−04 3.4 nM
    cyFolR1 2.98E+06 7.47E−03 2.5 nM
    16D5 D52dQ IgG huFolR1 8.40E+04 7.75E−04 9.2 nM
    cyFolR1 4.12E+05 2.04E−03   5 nM
    16D5 TCB huFolR1 2.25E+05 5.00E−04 2.2 nM
    cyFolR1 2.71E+05 6.63E−04 2.5 nM
  • 2. Affinity to Folate Receptor 1
  • The affinity of the interaction between the anti-FolR1 IgGs or the T cell bispecifics and the recombinant folate receptors was determined as described below (Table 39).
  • For affinity measurement, direct coupling of around 10000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 IgGs or T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 40 sec, the reference flow cell was left without capture. Dilution series (12.35 to 3000 nM) of human and cyno Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 300 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • TABLE 39
    Monovalent binding (affinity) of two 16D5 hot spot
    variants as IgGs on human and cyno FolR1.
    Ligand Analyte ka (1/Ms) kd (1/s) KD
    16D5 D52dE IgG huFolR1 2.40E+04 2.27E−03  95 nM
    cyFolR1 2.25E+04 1.20E−02 530 nM
    16D5 D52dQ IgG huFolR1 6.97E+03 1.62E−03 230 nM
    cyFolR1 8.20E+03 3.32E−03 410 nM
    16D5 TCB huFolR1 2.05E+04 7.05E−04  35 nM
    cyFolR1 1.72E+04 1.62E−03  90 nM
  • The two 16D5 hot spot variants have similar avidity (bivalent binding) than the wild-type 16D5 binder. The avidity is slightly decreased for the D52dQ variant and this difference is even more visible in affinity (monovalent binding).
  • Example 43
  • Production and Purification as IgGs of Twelve Variants of the 16D5 Binder with Mutations in the Heavy and Light Chain to Reduce Affinity to FolR1
  • Transient Transfection and Production
  • The twelve IgGs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 μg DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C. After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 μm sterile filters and stored at 4° C. until purification.
  • Antibody Purification
  • All molecules were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to POROS MabCapture A (Applied Biosystems, column volume (cv)=1 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate, 20 mM sodium citrate, pH 7.5). After washing with 10 cv of buffer A, the protein was eluted using a pH step to buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine) over 5 cv. The 5 ml containing the protein of interest are stored in a loop on the Äkta Explorer and subsequently applied to a HiLoad 16/60 Superdex™ 200 (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. Fractions containing the IgGs were pooled and concentrated using ultra concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore). The aggregate content of the final pool was analyzed by analytical size exclusion chromatography. Therefore, 30 μl were applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were stored at 4° C.
  • TABLE 40
    Yield, monomer content and purity by
    CE-SDS of twelve 16D5 IgG variants
    Mutation Monomer Purity by CE-
    Clone HC/LC Yield [mg/L} [%] SDS [%]
    16D5 W98Y/wt 32 100 100
    16D5 W98Y/K53A 24 100 100
    16D5 S35H/wt 21 100 100
    16D5 S35H/K53A 18 100 100
    16D5 S35H/S93A 18 100 100
    16D5 W96Y/wt 40 100 100
    16D5 W96Y/K53A 21 100 100
    16D5 W96Y/S93A 25 98 100
    16D5 R50S/K53A 10 98 100
    16D5 R50S/S93A 7 100 100
    16D5 G49S/K53A 42 100 100
    16D5 G49S/S93A 45 100 100
  • All twelve IgGs produced well and in good quality.
  • Example 44 Biochemical Characterization of 16D5 Heavy and Light Chain Combination Variants as IgG by Surface Plasmon Resonance
  • Binding of FolR1 16D5 heavy and light chain combination variants binders as IgG to different recombinant folate receptors (human, murine and cynomolgus FolR1; all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).
  • Avidity to Folate Receptor 1
  • The avidity of the interaction between the anti-FolR1 IgGs or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 41).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300. The anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 11.1 to 900 nM with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 240 or 600 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine IL2 receptor Fc fusion. The binding curves resulting from the bivalent binding of the IgG or T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding. The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare). For low affinity kinetics with association and dissociation phases too fast to be fitted by the 1:1 Langmuir binding model, the steady state analysis model was applied using the Bia Evaluation software (GE Healthcare). The steady state analysis gives the KD of the binding reaction at equilibrium.
  • TABLE 41
    Bivalent binding (avidity with apparent KD) of twelve 16D5 variants
    binders as IgGs on human, murine and cyno FolR1.
    Analyte
    HC variant/LC Apparent
    variant Ligand ka (1/Ms) kd (1/s) KD
    W98Y/K53A huFolR1 Weak binding
    cyFolR1 Weak binding
    S35H/K53A huFolR1 2.10E+04 2.91E−02 1400 nM 
    cyFolR1 3.47E+04 4.04E−02 1100 nM 
    W96Y/K53A huFolR1 580 nM 
    (steady state)
    cyFolR1 660 nM 
    (steady state)
    W98Y/wt huFolR1 1.36E+05 3.28E−02 240 nM 
    cyFolR1 1.71E+05 3.61E−02 200 nM 
    S35H/S93A huFolR1 2.43E+05 2.20E−02 90 nM
    cyFolR1 6.12E+05 6.77E−02 110 nM 
    G49S/K53A huFolR1 1.90E+05 1.15E−02 60 nM
    cyFolR1 3.93E+05 3.28E−02 80 nM
    R50S/K53A huFolR1 3.28E+05 1.97E−02 60 nM
    cyFolR1 5.50E+05 4.55E−02 80 nM
    S35H/wt huFolR1 1.32E+05 5.68E−03 40 nM
    cyFolR1 2.23E+05 1.24E−02 55 nM
    R50S/S93A huFolR1 1.25E+05 3.23E−03 30 nM
    cyFolR1 4.39E+05 7.80E−03 20 nM
    W96Y/S93A huFolR1 6.55E+05 1.89E−02 30 nM
    cyFolR1 6.25E+05 1.74E−02 30 nM
    G49S/S93A huFolR1 1.52E+05 3.06E−03 20 nM
    cyFolR1 3.58E+05 6.22E−03 20 nM
    W96Y/wt huFolR1 1.29E+05 2.13E−03 20 nM
    cyFolR1 1.73E+05 2.11E−03 10 nM
    36F2 TCB huFolR1 2.44E+06 1.37E−02  6 nM
    cyFolR1 4.12E+06 2.15E−02  5 nM
    muFolR1 4.86E+05 1.20E−03 2.5 nM 
    16D5 TCB huFolR1 1.41E+05 4.25E−04  3 nM
    cyFolR1 1.78E+05 6.39E−04 3.5 nM 
  • Affinity to Folate Receptor 1
  • The affinity of the interaction between the anti-FolR1 IgGs or the T cell bispecifics and the recombinant folate receptors was determined as described below (Table 42).
  • For affinity measurement, direct coupling of around 10000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 IgGs or T cell bispecifics were captured at 200 nM with a flow rate of 10 ul/min for 40 sec, the reference flow cell was left without capture. Dilution series (12.35 to 3000 nM) of human Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 300 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).
  • TABLE 42
    Monovalent binding (affinity) of twelve 16D5 variants FolR1 binders
    as IgGs on human, cyno and murine FolR1.
    Ligand
    HC variant/LC
    variant Analyte ka (1/Ms) kd (1/s) KD
    W98Y/K53A huFolR1 No binding
    S35H/K53A huFolR1 Weak binding
    W96Y/K53A huFolR1 Weak binding
    W98Y/wt huFolR1 5400 nM
    (steady state)
    G49S/K53A huFolR1 9.19E+03 1.74E−02 1900 nM
    R50S/K53A huFolR1 1.35E+04 2.45E−02 1800 nM
    36F2 TCB huFolR1 5.00E+04 8.57E−02 1700 nM
    S35H/S93A huFolR1 8.43E+03 1.12E−02 1300 nM
    S35H/wt huFolR1 8.96E+03 1.13E−02 1200 nM
    R50S/S93A huFolR1 1.57E+04 1.23E−02  780 nM
    G49S/S93A huFolR1 1.05E+04 7.99E−03  760 nM
    W96Y/wt huFolR1 9.95E+03 5.44E−03  550 nM
    W96Y/S93A huFolR1 4.05E+04 1.72E−02  420 nM
    16D5 TCB huFolR1 1.18E+04 7.22E−04  60 nM
  • Twelve “affinity reduced” variants of the 16D5 FolR1 binder were analyzed by surface plasmon resonance in comparison to the 16D5 wild-type binder and the 36F2 binder. The goal was to find a 16D5 variant with an affinity and an avidity comparable to 36F2. When measuring monovalent binding (affinity) there were variants with a higher and variants with a lower affinity than 36F2. However in the bivalent binding (avidity) all the variants have a higher apparent KD value than 36F2. This is mainly due to the fast association rate (ka) of 36F2 that results in a small apparent KD for 36F2. The big avidity effect when 36F2 binds bivalently seems to be unique to this binder. As noted above, 36F2 was the only human, murine and cyno crossreactive binder that could be identified.
  • Example 45 Binding of 16D5 HC/LC Variants to Human FolR1 Expressed on Hela Cells
  • The binding of 36F2 TCB, 16D5 TCB and various HC/LC variants of 16D5 to human FolR1 was assessed on Hela cells. Briefly, cells were harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with different concentrations of the bispecific antibodies (229 pM-500 nM). After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-116-170). After washing the samples twice with cold PBS 0.1% BSA they were fixed with 1% PFA overnight. Afterwards samples were centrifuged, resuspended in PBS 0.1% BSA and analyzed by FACS using a FACS CantoII (Software FACS Diva). Binding curves were obtained using GraphPadPrism6 (FIG. 32A-E). The 36F2 TCB bound FolR2, was not well tolerated in mice, and did not demonstrate the desired efficacy.
  • Example 46 Production and Purification of Four Variants of the 16D5 T-Cell Bispecific with Mutations to Reduce the Affinity to Human and Cynomolgus FolR1: 16D5 TCB G49S/S93A, G49S/K53A, W96Y, W96Y/D52E
  • Transient Transfection and Production
  • Four additional variants of 16D5 TCBs having reduced affinity to FolR1 were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. For transfection HEK293 EBNA cells are cultivated in suspension serum free in Excell culture medium containing 6 mM L-Glutamine and 250 mg/l G418 culture medium. For the production in 600 ml tubespin flask (max. working volume 400 mL) 600 million HEK293 EBNA cells are seeded 24 hours before transfection. For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 400 μg DNA. After addition of 1080 μl PEI solution (2.7 μg/ml) is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 600 ml tubespin flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 360 ml Excell+6 mM L-Glutamine+5 g/L Pepsoy+1.0 mM VPA medium is added and cells are cultivated for 24 hours. One day after transfection 7% Feed 7 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 20-30 min at 3600×g (Sigma 8K centrifuge), the solution is sterile filtered (0.22 mm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C.
  • Purification
  • The reduced affinity variants 16D5 TCBs were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to HiTrap Protein A (GE Healthcare, column volume (cv)=5 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate pH 7.5, 20 mM sodium citrate). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine) over 20 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 0.5 M Na2HPO4 pH 8.0). Samples were concentrated to 1 ml using ultra-concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore) and subsequently applied to a HiLoad™ 16/60 Superdex™ 200 preparative grade (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween 20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled. The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at −80° C.
  • TABLE 43
    Yield, monomer content and purity by CE-SDS of the reduced
    affinity 16D5 variants in the T cell bispecific format.
    Mutations to Yield Purity by CE-
    Name reduce affinity [mg/L] Monomer [%] SDS [%]
    16D5 TCB G49S S93A 10.3 100 88
    16D5 TCB G49S K53A 22.3 98.5 96
    16D5 TCB W96Y 15.2 98.7 92.5
    16D5 TCB W96Y D52E 9.9 99.3 92.9
    16D5 TCB Wild-type 5.4 96 91.6
  • All variants with reduced affinity could be produced in good quality.
  • Example 47 Binding of 36F2 TCB, 16D5 TCB and the Two 16D5 Affinity Reduced Variants 16D5 W96Y/D52E TCB and 16D5 G49S/S93A TCB to Human FolR1 Expressed on Hela Cells
  • The binding of 36F2 TCB, 16D5 TCB and the two 16D5 affinity reduced variants 16D5 W96Y/D52E TCB and 16D5 G49S/S93A TCB to human FolR1 was assessed on Hela cells. Briefly, cells were harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with different concentrations of the bispecific antibodies (30 pM-500 nM). After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a FITC-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-096-098). After washing the samples twice with cold PBS 0.1% BSA samples were centrifuged, resuspended in PBS 0.1% BSA and analyzed by FACS using a FACS CantoII (Software FACS Diva). Binding curves were obtained using GraphPadPrism6 (FIG. 33).
  • Example 48 Production and Purification of Three T-Cell Bispecifics with Intermediate Affinity to Human and Cynomolgus FolR1: 14B1, 6E10, 2C7
  • Transient Transfection and Production
  • The intermediate affinity TCBs were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. For transfection HEK293 EBNA cells are cultivated in suspension serum free in Excell culture medium containing 6 mM L-Glutamine and 250 mg/l G418 culture medium. For the production in 600 ml tubespin flask (max. working volume 400 mL) 600 million HEK293 EBNA cells are seeded 24 hours before transfection. For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 400 μg DNA. After addition of 1080 μl PEI solution (2.7 μg/ml) is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 600 ml tubespin flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 360 ml Excell+6 mM L-Glutamine+5 g/L Pepsoy+1.0 mM VPA medium is added and cells are cultivated for 24 hours. One day after transfection 7% Feed 7 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 20-30 min at 3600×g (Sigma 8K centrifuge), the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C.
  • Purification
  • The intermediate affinity TCBs were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to HiTrap Protein A (GE Healthcare, column volume (cv)=5 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate pH 7.5, 20 mM sodium citrate). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 3.0, 100 mM NaCl, 100 mM glycine) over 20 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 0.5 M Na2HPO4 pH 8.0). Samples were concentrated to 1 ml using ultra-concentrators (Amicon Ultra-15, 30.000 MWCO, Millipore) and subsequently applied to a HiLoad™ 16/60 Superdex™ 200 preparative grade (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween 20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Toso h) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled. The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis (CE-SDS) in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at −80° C.
  • TABLE 44
    Yield, monomer content and purity by CE-SDS
    of the intermediate affinity TCBs.
    Purity by CE-
    Name Yield [mg/L] Monomer [%] SDS [%]
    6E10 TCB 2.3 93 95
    14B1 TCB 1.8 94 70
    9C7 TCB 3.4 98 99
  • All intermediate affinity T cell bispecifics could be produced. The yields are not high. The quality is good for 9C7 and acceptable for 14B1 and 6E10.
  • Example 49 Binding of 16D5 HC/LC Variants to Human FolR1 Expressed on HT-29 Cells
  • The binding of 36F2 TCB, 16D5 TCB and various HC/LC variants (FIG. 34A-E) of 16D5 to human FolR1 was assessed on HT-29 cells. Briefly, cells were harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with different concentrations of the bispecific antibodies (229 pM-500 nM). After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-116-170). After washing the samples twice with cold PBS 0.1% BSA they were fixed with 1% PFA overnight. Afterwards samples were centrifuged, resuspended in PBS 0.1% BSA and analyzed by FACS using a FACS CantoII (Software FACS Diva). Binding curves were obtained using GraphPadPrism6 (FIG. 34A-E).
  • Example 50 Binding of Intermediate FolR1 Binders to Human and Mouse FolR1 and FolR2
  • Cross-reactivity of the intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), as well as 16D5 TCB and 36F2 TCB to human and mouse FolR1 and FolR2 was assessed in a FACS binding assay on transfected HEK293T cells.
  • Briefly, cells were harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with 100 nM of the bispecific antibodies. After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a Fluorescein (FITC) AffiniPure F(ab′)2 Fragment Goat Anti-Human IgG, Fcγ Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-096-098). After washing the samples twice with cold PBS 0.1% BSA they were fixed with 1% PFA overnight. Afterwards samples were centrifuged, resuspended in PBS 0.1% BSA and analyzed by FACS using a FACS CantoII (Software FACS Diva). Graphs were obtained using GraphPadPrism6 (FIG. 35A-D).
  • The results show that 36F2 TCB and 14B1 TCB are cross-reactive to mouse FolR1 and human and mouse FolR2. For 6E10 TCB a weak binding to human FolR2 can be observed. 16D5 TCB and 9C7 TCB are specific for human FolR1 and show no cross-reactivity to mouse FolR1 or human and mouse FolR2.
  • Example 51 Biochemical Characterization by Surface Plasmon Resonance of 16D5 Reduced Affinity Variants and Additional Intermediate Affinity Binders in the T-Cell Bispecific Format
  • Binding of anti-FolR1 16D5 reduced affinity variants and additional intermediate affinity binders in the bivalent T-cell bispecific format to recombinant human, cynomolgus and murine folate receptor 1 (all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, GE Healthcare). The molecules used for affinity and avidity determination are described in Table 45.
  • TABLE 45
    Name, description and figure reference
    of the nine constructs used in SPR analysis.
    Name Description FIG. reference
    16D5 reduced affinity variants 2 + 1 T-cell bispecific, FIG. 1A
    16D5 TCB inverted format
    16D5 G49S/S93A TCB (common light chain)
    16D5 G49S/K53A TCB
    16D5 W96Y TCB
    16D5 W96Y/D52E TCB
    Intermediate affinity binders 2 + 1 T-cell bispecific, FIG. 1F
    36F2 TCB inverted format,
    6E10 TCB crossfab
    14B1 TCB
    9C7 TCB
  • Single Injections
  • First the anti-FolR1 TCBs were analyzed by single injections (Table 46) to characterize their crossreactivity (to human, murine and cyno FolR1) and specificity (to human FolR1, human FolR2, human FolR3). Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) or human Folate Receptor 2 and 3 (FolR2-Fc, FolR3-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The TCBs were injected for 60 seconds at a concentration of 500 nM.
  • TABLE 46
    Crossreactivity and specificity of 7 folate receptor 1 T cell
    bispecifics. + means binding, − means no binding,
    +/− means weak binding.
    Binding to Binding to Binding to Binding to Binding to
    Clone name huFolR1 cyFolR1 muFolR1 huFolR2 huFolR3
    16D5 TCB + +
    16D5 + +
    G49S/S93A
    TCB
    16D5 + +
    W96Y/D52E
    TCB
    36F2 TCB + + + +/−
    6E10 TCB + +
    14B1 TCB + + + +/−
    9C7 TCB + +
  • Avidity to Folate Receptor 1
  • The avidity of the interaction between the anti-FolR1 T cell bispecifics and the recombinant folate receptors was determined as described below (Table 47).
  • Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, GE Healthcare). The immobilization level was about 200-300 RU. The anti-FolR1 T cell bispecifics were passed at a concentration range from 11.1 to 900 nM (for the 16D5 reduced affinity variants) or 0.2 to 500 nM (for the additional intermediate affinity binders and 36F2) with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 240 or 600 seconds. The chip surface was regenerated after every cycle using a double injection of 30 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine IL2R Fc fusion (unrelated Fc fused receptor). The binding curves resulting from the bivalent binding of the T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding. The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).
  • TABLE 47
    Bivalent binding (avidity with apparent KD) of anti-FolR1 T-cell
    bispecifics (TCB) on human, cyno and murine FolR1.
    Apparent
    Analyte Ligand ka (1/Ms) kd (1/s) KD
    16D5 TCB huFolR1 1.68E+05 4.33E−04 3 nM
    cyFolR1 2.08E+05 6.95E−04 3 nM
    16D5 G49S/S93A huFolR1 1.49E+05 2.09E−03 10 nM 
    TCB cyFolR1 4.54E+05 7.84E−03 20 nM 
    16D5 G49S/K53A huFolR1 1.32E+05 5.86E−03 40 nM 
    TCB cyFolR1 3.73E+05 2.56E−02 70 nM 
    16D5 W96Y TCB huFolR1 1.15E+05 1.44E−03 10 nM 
    cyFolR1 1.37E+05 1.68E−03 10 nM 
    16D5 W96Y/D52E huFolR1 1.24E+05 1.40E−03 10 nM 
    TCB cyFolR1 5.17E+05 1.41E−02 30 nM 
    36F2 TCB huFolR1 1.12E+06 7.90E−03 7 nM
    cyFolR1 1.97E+06 1.10E−02 6 nM
    muFolR1 5.54E+05 1.47E−03 3 nM
    6E10 TCB huFolR1 7.93E+06 8.74E−03 1 nM
    cyFolR1 5.56E+06 5.72E−03 1 nM
    14B1 TCB huFolR1 1.12E+06 1.40E−03 1 nM
    cyFolR1 1.02E+06 1.66E−03 2 nM
    muFolR1 8.03E+06 8.20E−04 0.1 nM  
    9C7 TCB huFolR1 1.18E+06 1.42E−03 1 nM
    cyFolR1 4.98E+06 4.82E−03 1 nM
  • 3. Affinity to Folate Receptor 1
  • The affinity of the interaction between the anti-FolR1 T cell bispecifics and the recombinant folate receptors was determined as described below (Table 48).
  • For affinity measurement, direct coupling of around 12000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 40 sec, the reference flow cell was left without capture. Dilution series (12.3 to 3000 nM) of human, cyno or murine Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 300 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare). For low affinity kinetics with association and dissociation phases too fast to be fitted by the 1:1 Langmuir binding model, the steady state analysis model was applied using the Bia Evaluation software (GE Healthcare). The steady state analysis gives the KD of the binding reaction at equilibrium.
  • TABLE 48
    Monovalent binding (affinity) of anti-FolR1 T-cell bispecifics
    (TCB) on human, cyno and murine FolR1.
    Analyte Ligand ka (1/Ms) kd (1/s) KD
    16D5 TCB huFolR1 1.22E+04 7.02E−04  57 nM
    cyFolR1 1.29E+04 1.71E−03  130 nM
    16D5 G49S/ huFolR1 1.01E+04 8.37E−03  830 nM
    S93A TCB cyFolR1 2.05E+04 8.60E−03  420 nM
    16D5 G49S/ huFolR1 9.17E+03 1.59E−02 1700 nM
    K53A TCB cyFolR1 1900 nM
    (steady state analysis)
    16D5 W96Y huFolR1 1.11E+04 4.05E−03  370 nM
    TCB cyFolR1 1.17E+04 5.16E−03  440 nM
    16D5 huFolR1 1400 nM
    W96Y/ (steady state analysis)
    D52E TCB cyFolR1 5600 nM
    (steady state analysis)
    36F2 TCB huFolR1 1400 nM
    (steady state analysis)
    cyFolR1 1500 nM
    (steady state analysis)
    muFolR1 3.50E+04 1.73E−02  490 nM
    6E10 TCB huFolR1 1200 nM
    (steady state analysis)
    cyFolR1 1500 nM
    (steady state analysis)
    14B1 TCB huFolR1 6.16E+04 3.03E−02  490 nM
    cyFolR1 1200 nM
    (steady state analysis)
    muFolR1 7.03E+04 2.28E−03  30 nM
    9C7 TCB huFolR1  840 nM
    (steady state analysis)
    cyFolR1 1400 nM
    (steady state analysis)
  • The mutations introduced into the 16D5 binders reduce its affinity to human and cynomolgus FolR1 as determined by surface plasmon resonance. The ranking with decreasing affinity is 16D5 WT (57 nM)>W96Y (6.5 fold lower)>G49S/S93A (14.5 fold lower)>W96Y/D52E (24.5 fold lower)>G49S/K53A (30 fold lower). The same ranking is visible in the avidity values, however the fold differences are smaller 16D5 WT (3 nM)>W96Y, G49S/S93A, W96Y/D52E (3 fold lower)>G49S/K53A (13 fold lower).
  • The intermediate affinity binders have following ranking in affinity 16D5 (57 nM)>14B1 (8.5 fold lower)>9C7 (15 fold lower)>6E10 (21 fold lower)>36F2 (24.5 fold lower). These differences however disappear in the avidity measurement 14B1, 9C7, 6E10 (1 nM)>16D5 (3 nM)>36F2 (7 nM).
  • 16D5 W96Y/D52E TCB addresses the problems observed with previous candidates. 16D5 W96Y/D52E TCB is based on the common light chain 16D5 binder and has two point mutations on the heavy chain with respect to the parental 16D5 binder. The W96Y mutation reduces the affinity of the binder to FolR1 compared to the parental binder and the D52E mutation removes a deamidation site and also contributes to the reduction in affinity. 16D5 W96Y/D52E TCB binds to human and cynomolgus FolR1, but not to murine FolR1. It is specific for FolR1 and does not bind to recombinant human FolR2 or human FolR3. The affinity (monovalent binding) of 16D5 W96Y/D52E is around 1.4 μM for human FolR1 (24.5 fold lower than the parental 16D5 binder) and the avidity (bivalent binding) is around 10 nM (3 fold lower than the parental 16D5 binder).
  • Example 52 T-Cell Killing of Hela, SKov-3 and HT-29 Cells Induced by Intermediate FolR1 TCBs
  • T-cell killing mediated by intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB), was assessed on Hela (high FolR1), SKov-3 (medium FolR1) and HT-29 (low FolR1) cells. 16D5 TCB and 36F2 TCB were included as benchmarks. Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at a density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h and 48 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.
  • The results show that tumor lysis induced by the intermediate FolR1 binders (6E10 TCB, 14B1 TCB and 9C7 TCB) ranges between the one obtained for the high affinity 16D5 TCB and the low affinity 36F2 TCB (FIG. 36A-F). Among the intermediate FolR1 binders, 14B1 TCB shows the strongest killing as can be seen after 48h of incubation (FIG. 36D-F). The EC50 values related to killing assays after 24 h and 48 h of incubation, calculated using GraphPadPrism6, are given in Table 49 and Table 50.
  • TABLE 49
    EC50 values (pM) for T-cell mediated killing of Hela, SKov-3
    and HT-29 cells induced by intermediate FolR1 TCBs after
    24 h of incubation.
    EC50 [pM]
    Antibody Hela SKov-3 HT-29
    6E10 TCB 6.5 n.d. *n.d.
    14B1 TCB 8.5 30.1 *n.d.
    9C7 TCB 2.8 741.4 *n.d.
    16D5 TCB 2.2 1.5 *n.d.
    36F2 TCB 31.1 *n.d. *n.d.
    *not determined
  • TABLE 50
    EC50 values (pM) for T-cell mediated killing of Hela,
    SKov-3 and HT-29 cells induced by intermediate FolR1
    TCBs after 48 h of incubation.
    EC50 [pM]
    Antibody Hela SKov-3 HT-29
    6E10 TCB 2.1 2164.0 *n.d.
    14B1 TCB 5.5 4.7 397.7
    9C7 TCB 4.3 519.6 *n.d.
    16D5 TCB 2.3 *n.d.  4.9
    36F2 TCB 10.5 *n.d. n.d.
    *not determined
  • Example 53 T-Cell Killing of Hela, SKov-3 and HT-29 Cells Induced by Affinity Reduced 16D5 Variants
  • T-cell killing mediated by affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5-G49S/K53A TCB, 16D5 W96Y TCB, 16D5 W96Y/D52E TCB) was assessed on Hela (high FolR1), SKov-3 (medium FolR1) and HT-29 (low FolR1) cells. 16D5 TCB and 36F2 TCB were included as benchmarks. The assay was performed as described above (Example 52).
  • The results show that tumor lysis induced by affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5-G49S/K53A TCB, 16D5 W96Y TCB, 16D5 W96Y/D52E TCB), ranges between the one obtained for the high affinity 16D5 TCB and the low affinity 36F2 TCB. The EC50 values related to killing assays after 24 h and 48 h of incubation, calculated using GraphPadPrism6, are given in Table 51 and Table 52 (FIG. 37A-F).
  • TABLE 51
    EC50 values (pM) for T-cell mediated killing of FolR1-
    expressing Hela, SKov-3 and HT-29 cells induced by 16D5
    TCB and its affinity reduced variants after 24 h of incubation.
    EC50 [pM]
    Antibody Hela SKov-3 HT-29
    16D5 TCB 2.2 1.5 *n.d.
    16D5- 2.3 430.4 *n.d.
    G49S/S93A
    TCB
    16D5- 4.4 1701.9 *n.d.
    G49S/K53A
    TCB
    16D5 W96Y 3.0 164.5 *n.d.
    TCB
    16D5 1.3 235.4 *n.d.
    W96Y/D52E
    TCB
    36F2 TCB 31.1 *n.d. *n.d.
    *not determined
  • TABLE 52
    EC50 values (pM) for T-cell mediated killing of FolR1-
    expressing Hela, SKov-3 and HT-29 cells induced by 16D5
    TCB and its affinity reduced variants after 48 h of incubation.
    EC50 [pM]
    Antibody Hela SKov-3 HT-29
    16D5 TCB 2.3 0.1 4.9
    16D5- 0.9 95.9 99.3
    G49S/S93A
    TCB
    16D5- 0.5 950.4 1790.7
    G49S/K53A
    TCB
    16D5 W96Y 1.8 24.7 99.3
    TCB
    16D5 0.9 93.0 399.4
    W96Y/D52E
    TCB
    36F2 TCB 10.5 968.5 *n.d.
    *not determined
  • Thus, as with 36F2 FOLR1 TCB described above, the 16D5 W96Y/D52E TCB differentiates between high and low expressing cells which is of special importance to reduce toxicity as the cells of some normal, non-tumorous tissues express very low levels of FolR1 (approximately less than 1000 copies per cell). Consistent with this observation, the results discussed in Example 54 below show that 16D5 W96Y/D52E TCB induces much lower levels of T-cell-mediated killing of primary cells (FIG. 38A-F) compared to the parental 16D5 TCB. As such, 16D5 W96Y/D52E TCB mediates potent killing of tumor tissues with high or medium FOLR1 expression, but not of normal tissues with low expression. 16D5 W96Y/D52E TCB in the bivalent 2+1 format comprises FolR1 binding moieties of relatively low affinity but it possesses an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this TCB selectively binds to tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all. As an additional advantage over the 36F2 FOLR1 TCB described above, the 16D5 W96Y/D52E TCB binds specifically to FolR1 and not to FolR2 or FolR3, further enhancing its safety for in vivo treatment.
  • In addition to the above advantageous characteristics, the 16D5 W96Y/D52E TCB in the bivalent 2+1 inverted format also has the advantage that it does not require chemical cross linking or other hybrid approach. This makes it suitable for manufacture of a medicament to treat patients, for example patients having FolR1-positive cancerous tumors. The 16D5 W96Y/D52E TCB in the bivalent 2+1 inverted format can be produced using standard CHO processes with low aggregates. Further, the 16D5 W96Y/D52E TCB in the bivalent 2+1 comprises human and humanized sequences making it superior to molecules that employ rat and murine polypeptides that are highly immunogenic when administered to humans. Furthermore, the 16D5 W96Y/D52E TCB in the bivalent 2+1 format was engineered to abolish FcgR binding and, as such, does not cause FcgR crosslinking and infusion reactions, further enhancing its safety when administered to patients.
  • As demonstrated by the results described above, its head-to-tail geometry make the 16D5 W96Y/D52E TCB in the bivalent 2+1 inverted format a highly potent molecule that induces absolute target cell killing. Its bivalency enhance avidity and potency, but also allow for differentiation between high and low expressing cells. Its preference for high or medium target expressing cells due to its avidity affect reduce toxicity resulting from T cell mediated killing of normal cells that express FolR1 at low levels.
  • A further advantage of the 16D5 W96Y/D52E TCB in the bivalent 2+1 format and other embodiments disclosed herein is that their clinical development does not require the use of surrogate molecules as they bind to human and cynomous FolR1. As such, the molecules disclosed herein recognize a different epitope than antibodies to FolR1 previously described that do not recognize FolR1 from both species (see also FIG. 41).
  • Example 54
  • T-Cell Killing of Primary Cells Induced by Affinity Reduced 16D5 Variants and Intermediate FolR1 TCBs
  • T-cell killing mediated by affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5 W96Y/D52E TCB) and the intermediate FolR1 binder 14B1 TCB was assessed on primary cells (Human Renal Cortical Epithelial Cells (HRCEpiC) (ScienCell Research Laboratories; Cat No 4110) and Human Retinal Pigment Epithelial Cells (HRPEpiC) (ScienCell Research Laboratories; Cat No 6540)). HT-29 cells (low FolR1) were included as control cell line. 16D5 TCB and 36F2 TCB were included as benchmarks and DP47 TCB served as non-binding control.
  • The assay was performed as described in Example 52, with a concentration range of the antibodies of 0.1 pM-100 nM (in triplicates).
  • When human primary cells are used as targets, the overall lysis is much lower due to a lower expression rate of FolR1 on these cells (FIG. 38A-F). For the high affinity FolR1 binder 16D5 TCB a T-cell mediated lysis can be observed on both primary cell types used. As observed previously when tumor cell lines were used as targets, lysis induced by the intermediate FolR1 binder 14B1 TCB and the affinity reduced 16D5 variants (16D5-G49S/S93A TCB, 16D5 W96Y/D52E TCB), ranges between the one obtained for the high affinity 16D5 TCB and the low affinity 36F2 TCB. The significantly reduced lysis of cells that express FolR1 at low levels is consistent with low off target activity and the affinity reduced 16D5 variants 16D5-G49S/S93A TCB and 16D5 W96Y/D52E TCB are, thus, expected to be well tolerated in vivo.
  • Example 55 Single Dose PK of FOLR1 TCB Constructs in Female NOG Mice
  • Female NOD/Shi-scid/IL-2Rγnull (NOG) mice at an average ager of 8-10 weeks at start of experiment (purchased from Taconic, SOPF facility) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). Experimental study protocol was reviewed and approved by local government (ZH193/2014). After arrival animals were maintained for one week to get accustomed to new environment and for observation. Continuous health monitoring was carried out on regular basis.
  • A single dose pharmacokinetic study (SDPK) was performed to evaluate exposure of FOLR1 TCB constructs (36F2, 16D5, 16D5 G49S/S93A and 16D5 W96Y/D52E). An i.v. bolus administration of 0.5 mg/kg was administered to NOG mice and blood samples were taken at selected time points for pharmacokinetic evaluation. Mouse serum samples were analyzed by ELISA. Biotinylated a-huCD3-CDR (mAb<ID-mAb<CD3>>M-4.25.93-IgG-Bi), test samples, Digoxygenin labelled a-huFc antibody (mAb<H-FC pan>M-R10Z8E9-IgG-Dig) and anti-Digoxygenin detection antibody (POD) were added stepwise to a 96-well streptavidin-coated microtiter plate and incubated after every step for 1 h at room temperature. The plate is washed three times after each step to remove unbound substances. Finally, the peroxidase-bound complex is visualized by adding ABTS substrate solution to form a colored reaction product. The reaction product intensity, which is photometrically determined at 405 nm (with reference wavelength at 490 nm), is proportional to the analyte concentration in the serum sample. The calibration range of the standard curve for the constructs is was 0.078 to 5 ng/ml, where 1.5 ng/ml is the lower limit of quantification (LLOQ).
  • The SDPK study revealed an IgG-like PK-profile for the 16D5, 16D5 W96Y/D52E and 16D5 G49S/S93A constructs (FIG. 39A-B). Because of that, a once per weeks scheduling was chosen for the efficacy study (FIG. 40B). The half-life for 36F2 is lower as compared to the other clones. 36F2 is the only out of the four molecules tested that is cross-reactive to mouse FOLR1, which might explain the lower half-life for this molecule and indicates a TMDD (Target Mediated Drug Disposition).
  • Example 56 In Vivo Efficacy of FOLR1 TCB Constructs (16D5, 16D5 G49S/S93A and 16D5 W96Y/D52E) after Human PBMC Transfer in Hela-Bearing NOG Mice
  • The FOLR1 TCB constructs were tested in the FOLR1-expressing human cervical cancer cell line Hela, injected s.c. into PBMC engrafted NOG mice.
  • Hela cells were originally obtained from ATCC (CCL2) and after expansion deposited in the Roche-Glycart internal cell bank. The tumor cell line was routinely cultured in RPMI containing 10% FCS (Gibco) at 37° C. in a water-saturated atmosphere at 5% CO2. Passage 13 was used for transplantation, at a viability >95%. 1×106 cells per animal were injected s.c. into the right flank of the animals in a total of 100 μl of RPMI cell culture medium (Gibco).
  • 60 female NOG mice, age 8-10 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (ZH193/2014). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.
  • According to the study protocol (FIG. 40B), mice were injected s.c. on study day 0 with 1×106 Hela cells. At study day 30, when tumor reached a size of app. 150 mm3, human PBMC of a healthy donor were isolated via the Ficoll method and 10×106 cells were injected i.v. into the tumor-bearing mice. Two days after (day 32), mice were randomized and equally distributed in six treatment groups (n=10) followed by i.v. injection with either 16D5 (0.5 mg/kg), 16D5 G49S/S93A (2.5 or 0.5 mg/kg) and 16D5 W96Y/D52E (2.5 or 0.5 mg/kg). All treatments group were injected once weekly for three weeks in total. Mice were injected i.v. with 200 μl of the appropriate solution. The mice in the vehicle group were injected with PBS. To obtain the proper amount of TCB per 200 μl, the stock solutions were diluted with PBS when necessary. Tumor growth was measured once weekly using a caliper (FIG. 40C-E) and tumor volume was calculated as followed:

  • Tv:(W2/2)×L(W:Width,L:Length)
  • The once weekly injection of the FOLR1 TCB constructs resulted in significant tumor regression (FIG. 40C-E). The efficacy of 16D5 (0.5 mg/kg) and 16D5 W96Y/D52E16D5 (0.5 mg/kg) was comparable, whereas 16D5 G49S/S93A (0.5 mg/kg) showed slight less potency. The higher doses of 2.5 mg/kg of 16D5 W96Y/D52E16D5 and 16D5 G49S/S93A didn't show increased efficacy compared to 0.5 mg/kg doses. For PD read-outs, mice were sacrificed at study day 52, tumors were removed, weighted and single cell suspensions were prepared through an enzymatic digestion with Collagenase V, Dispase II and DNAse for subsequent FACS-analysis. Explanted tumors of all treatment groups showed significant lower tumor weight at study termination as compared to vehicle control tumors (FIG. 40F). Single cell suspensions from tumors where stained for huCD45 and huCD3 and DAPI for dead cell exclusion and were analyzed at the BD Fortessa. The FACS analysis revealed statistically higher numbers of infiltrated CD3-positive human T-cells in the tumor tissue upon treatment with 16D5 as well as 16D5 W96Y/D52E16D5 compared to vehicle control tumors (FIG. 40C).
  • Example 57 Toxicity Study in Cynomolgus Monkey
  • A pharmacokinetic (PK), pharmacodynamic (PD) and tolerability study is performed to investigate the tolerability, PK and PD effects of a single intravenous dose of affinity reduced 16D5 variant TCBs (e.g., 16D5-G49S/S93A TCB, 16D5 W96Y/D52E TCB) in cynomolgus monkeys. In this study, naïve cynomolgus monkeys, (1 male and 1 female monkey/group), receive a single intravenous dose of affinity reduced 16D5 variant TCBs, including 16D5 W96Y/D52E TCB, following a dose escalating protocol. Exemplary dose levels include 0.003, 0.03, and 0.09 mg/kg. Standard toxicity parameters (clinical signs, body weights, hematology & clinical chemistry) and the kinetics of T cell numbers and activation status in blood and the kinetics of cytokine release are assessed. Blood samples are also taken for PK for a period of 28 days for the measurement of affinity reduced 16D5 variant TCBs, including 16D5 W96Y/D52E TCB, and of anti-drug antibodies.
  • Amino Acid Sequences of Exemplary Embodiments
      • 1) FolR binders useful in common light chain format, variable heavy chain
  • Description Sequence Seq ID No
    16A3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    1
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNYYAGVTPFDYWGQGTLVTVSS
    18D3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    2
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNYYTGGSSAFDYWGQGTLVTVS
    15H7 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    3
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNYYLFSTSFDYWGQGTLVTVSS
    15B6 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    4
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNYYIGIVPFDYWGQGTLVTVSS
    21D1 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    5
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNYYVGVSPFDYWGQGTLVTVSS
    16F12 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    6
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNFTVLRVPFDYWGQGTLVTVSS
    15A1 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    7
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARNYYIGVVTFDYWGQGTLVTVSS
    15A1_CDR1 SYYMH 8
    15A1_CDR2 IINPSGGSTSYAQKFQG
    9
    15A1_CDR3 NYYIGVVTFDY
    10
    19E5 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 11
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARGEWRRYTSFDYWGQGTLVTVSS
    19E5_CDR1 SYYMH 8
    19E5_CDR2 IINPSGGSTSYAQKFQG
    9
    19E5_CDR3 GEWRRYTSFDY 12
    19A4 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 13
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARGGWIRWEHFDYWGQGTLVTVSS
    19A4_CDR1 SYYMH 8
    19A4_CDR2 IINPSGGSTSYAQKFQG
    9
    19A4_CDR3 GGWIRWEHFDY 14
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 15
    WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED
    TAVYYCTTPWEWSWYDYWGQGTLVTVSS
    16D5_CDR1 NAWMS
    16
    16D5_CDR2 RIKSKTDGGTTDYAAPVKG 17
    16D5_CDR3 PWEWSWYDY 18
    15E12 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 19
    WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED
    TAVYYCTTPWEWSYFDYWGQGTLVTVSS
    15E12_CDR1 NAWMS
    16
    15E12_CDR2 RIKSKTDGGTTDYAAPVKG 17
    15E12_CDR3 PWEWSYFDY
    20
    21A5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 21
    WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED
    TAVYYCTTPWEWAWFDYWGQGTLVTVSS
    21A5_CDR1 NAWMS
    16
    21A5_CDR2 RIKSKTDGGTTDYAAPVKG 17
    21A5_CDR3 PWEWAWFDY 22
    21G8 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 23
    WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED
    TAVYYCTTPWEWAYFDYWGQGTLVTVSS
    21G8_CDR1 NAWMS
    16
    21G8_CDR2 RIKSKTDGGTTDYAAPVKG 17
    21G8_CDR3 PWEWAYFDY
    24
    19H3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE
    25
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARTGWSRWGYMDYWGQGTLVTVSS
    19H3_CDR1 SYYMH 8
    19H3_CDR2 IINPSGGSTSYAQKFQG
    9
    19H3_CDR3 TGWSRWGYMDY 26
    20G6 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 27
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARGEWIRYYHFDYWGQGTLVTVSS
    20G6_CDR1 SYYMH 8
    20G6_CDR2 IINPSGGSTSYAQKFQG
    9
    20G6_CDR3 GEWIRYYHFDY 28
    20H7 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 29
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARVGWYRWGYMDYWGQGTLVTVSS
    20H7_CDR1 SYYMH 8
    20H7_CDR2 IINPSGGSTSYAQKFQG
    9
    20H7_CDR3 VGWYRWGYMDY
    30
      • 2) CD3 binder common light chain (CLC)
  • Description Sequence Seq ID No
    common QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKP 31
    CD3 light GQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPED
    chain (VL) EAEYYCALWYSNLWVFGGGTKLTVL
    common GSSTGAVTTSNYAN 32
    CD3 light
    chain_CDR1
    common GTNKRAP 33
    CD3 light
    chain_CDR2
    common ALWYSNLWV 34
    CD3 light
    chain_CDR3
    common QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKP 35
    CD3 light GQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPED
    chain EAEYYCALWYSNLWVFGGGTKLTVLGQPKAAPSVTLFPPSSE
    (VLCL) ELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPS
    KQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVA
    PTECS
      • 3) CD3 binder, heavy chain
  • Seq ID
    Description Sequence No
    CD3 EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKG 36
    variable LEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSL
    heavy RAEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSS
    chain (VH)
    CD3 heavy TYAMN 37
    chain
    (VH)_CDR1
    CD3 heavy RIRSKYNNYATYYADSVKG 38
    chain
    (VH)_CDR2
    CD3 heavy HGNFGNSYVSWFAY 39
    chain
    (VH)_CDR3
    CD3 full EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKG 40
    heavy LEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSL
    chain RAEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSSASTKGPS
    (VHCH1) VFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH
    TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVD
    KKVEPKSC
    CD3 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSG 84
    constant ALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK
    heavy PSNTKVDKKVEPKSC
    chain CH1
      • 4) FolR binders useful for crossfab Format
  • Seq ID
    Description Sequence No
    11F8_VH QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLE 41
    WMGGIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTA
    VYYCARAVFYRAWYSFDYWGQGTTVTVSS
    11F8_VH_CDR1 SYAIS 42
    11F8_VH_CDR2 GIIPIFGTANYAQKFQG 43
    11F8_VH_CDR3 AVFYRAWYSFDY 44
    11F8_VL DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKL 45
    LIYDASSLESGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCQQYT
    SPPPT FGQGTKVEIK
    11F8_VL_CDR1 RASQSISSWLA
    46
    11F8_VL_CDR2 DASSLES 47
    11F8_VL_CDR3 QQYTSPPPT
    48
    36F2_VH QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 49
    WMGIINPSGGSTSYAQKFQGRVTMTHDTSTSTVYMELSSLRSEDTA
    VYYCARSFFTGFHLDYWGQGTLVTVSS
    36F2_VH_CDR1 SYYMH 8
    36F2_VH_CDR2 IINPSGGSTSYAQKFQG
    9
    36F2_VH_CDR3 SFFTGFHLDY
    50
    36F2_VL EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR 51
    LLIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQY
    TNEHYYT FGQGTKVEIK
    36F2_VL_CDR1 RASQSVSSSYLA
    52
    36F2_VL_CDR2 GASSRAT 53
    36F2_VL_CDR3 QQYTNEHYYT 54
    9D11_VH QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 55
    WMGIINPSGGPTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARGDFAWLDYWGQGTLVTVSS
    9D11_VH_CDR1 SYYMH 8
    9D11_VH_CDR2 IINPSGGPTSYAQKFQG
    56
    9D11_VH_CDR3 GDFAWLDY 57
    9D11_VL DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG 58
    QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY
    CMQASIMNRTFGQGTKVEIK
    9D11_VL_CDR1 RSSQSLLHSNGYNYLD 59
    9D11_VL_CDR2 LGSNRAS 60
    9D11_VL_CDR3 MQASIMNRT 61
    9D11_VL DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG 62
    N95S QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY
    CMQASIMSRTFGQGTKVEIK
    9D11_VL MQASIMSRT 63
    N95S_CDR3
    9D11_VL DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG 64
    N95Q QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY
    CMQASIMQRTFGQGTKVEIK
    9D11_VL MQASIMQRT 65
    N95Q_CDR3
    9D11_VL DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG 66
    T97A QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY
    CMQASIMNRAFGQGTKVEIK
    9D11_VL MQASIMNRA 67
    T97A
    9D11_VL DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG 68
    T97N QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY
    CMQASIMNRNFGQGTKVEIK
    9D11_VL MQASIMNRN 69
    T97N_CDR3
    5D9_VH QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 70
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARSYIDMDYWGQGTLVTVSS
    5D9_VH_CDR1 SYYMH 8
    5D9_VH_CDR2 IINPSGGSTSYAQKFQG
    9
    5D9_VH_CDR3 SYIDMDY 71
    5D9_VL EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR
    72
    LLIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQD
    NWSPT FGQGTKVEIK
    5D9_VL_CDR1 RASQSVSSSYLA
    52
    5D9_VL_CDR2 GASSRAT 53
    5D9_VL_CDR3 QQDNWS PT 73
    6B6_VH QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 74
    WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARSYVDMDYWGQGTLVTVSS
    6B6_VH_CDR1 SYYMH 8
    6B6_VH_CDR2 IINPSGGSTSYAQKFQG
    9
    6B6_VH_CDR3 SYVDMDY 75
    6B6_VL EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR 76
    LLIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQD
    IWSPT FGQGTKVEIK
    6B6_VL_CDR1 RASQSVSSSYLA
    52
    6B6_VL_CDR2 GASSRAT 53
    6B6_VL_CDR3 QQDIWSPT 77
    14E4_VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLE 78
    WVSAISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTA
    VYYCAKDSSYVEWYAFDYWGQGTLVTVSS
    14E4_VH_CDR1 SYAMS 79
    14E4_VH_CDR2 AISGSGGSTYYADSVKG 80
    14E4_VH_CDR3 DSSYVEWYAFDY 81
    14E4_VL EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR 82
    LLIYGASSRATGIPDRFSGSGSGTDSTLTISRLEPEDFAVYYCQQP
    TSSPITFG QGTKVEIK
    14E4_VL_CDR1 RASQSVSSSYLA
    52
    14E4_VL_CDR2 GASSRAT 53
    14E4_VL_CDR3 QQPTSSPIT 83
      • 5) CD3 binder useful in crossfab Format
  • Description Sequence Seq ID No
    CD3 heavy EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPG 36
    chain (VH) KGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQ
    MNSLRAEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSS
    CD3 heavy TYAMN 37
    chain
    (VH)_CDR1
    CD3 heavy RIRSKYNNYATYYADSVKG 38
    chain
    (VH)_CDR2
    CD3 heavy HGNFGNSYVSWFAY 39
    chain
    (VH)_CDR3
    CD3 light QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKP 31
    chain (VL) GQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPED
    EAEYYCALWYSNLWVFGGGTKLTVL
    CD3 light GSSTGAVTTSNYAN 32
    chain_CDR1
    CD3 light GTNKRAP 33
    chain_CDR2
    CD3 light ALWYSNLWV 34
    chain_CDR3
    pETR12940: QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKP 86
    crossed GQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPED
    common EAEYYCALWYSNLWVFGGGTKLTVLSSASTKGPSVFPLAPSS
    CD3 light KSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL
    chain QSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVE
    (VLCH1) PKSC
    Crossed EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPG 87
    CD3 heavy KGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQ
    chain MNSLRAEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSSA
    (VHCκ); SVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV
    e.g. in DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVY
    pCON1057 ACEVTHQGLSSPVTKSFNRGEC
    CD3-CH1 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWN 85
    SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN
    VNHKPSNTKVDKKVEPKSC
    CD3- VAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVD 88
    ckappa NALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYA
    CEVTHQGLSSPVTKSFNRGEC
      • 6)—Exemplary amino acid sequences of CD3-FolR bispecific antibodies 2+1 inverted crossmab format
  • Description Sequence Seq ID No
    VHCH1[9D11]_VHCL[CD3]_Fcknob_PGLALA QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 94
    pCON1057 WMGIINPSGGPTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARGDFAWLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGG
    TAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSS
    VVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDGGGGSGGGG
    SEVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGL
    EWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAE
    DTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSSASVAAPSVFIFP
    PSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTE
    QDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNR
    GECDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVV
    VDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVL
    HQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVYTLPPCR
    DELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSD
    GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    9D11_Fchole_PGLALA_HYRF QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 95
    WMGIINPSGGPTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA
    VYYCARGDFAWLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGG
    TAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSS
    VVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCP
    APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN
    WYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK
    VSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCA
    VKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDK
    SRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    9D11_LC DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG
    96
    pCON1063 QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY
    CMQASIMNRTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVV
    CLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTL
    TLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
    VLCH1[CD3] QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAF 86
    pETR12940 RGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCAL
    WYSNLWVFGGGTKLTVLSSASTKGPSVFPLAPSSKSTSGGTAALGC
    LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPS
    SSLGTQTYICNVNHKPSNTKVDKKVEPKSC
    CH1 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGAL 307
    TSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT
    KVDKKVEPKSCD
    VHCH1[36F2]_VHCL[CD3]_Fcknob_PGLALA QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 308
    pCON1056 WMGIINPSGGSTSYAQKFQGRVTMTHDTSTSTVYMELSSLRSEDTA
    VYYCARSFFTGFHLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS
    GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL
    SSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDGGGGSGG
    GGSEVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGK
    GLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLR
    AEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSSASVAAPSVFI
    FPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESV
    TEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF
    NRGECDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTC
    VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLT
    VLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVYTLPP
    CRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD
    SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSP
    GK
    36F2-Fchole QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 309
    PGLALA WMGIINPSGGSTSYAQKFQGRVTMTHDTSTSTVYMELSSLRSEDTA
    pCON1050 VYYCARSFFTGFHLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS
    GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL
    SSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPP
    CPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVK
    FNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYK
    CKVSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLS
    CAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTV
    DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    36F2 LC EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR 310
    pCON1062 LLIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQY
    TNEHYYTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLL
    NNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS
    KADYEKHKVYACEVTHXGLSSPVTKSFNRGEC
    CD3 VLCH1 QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAF 86
    pETR12940 RGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCAL
    WYSNLWVFGGGTKLTVLSSASTKGPSVFPLAPSSKSTSGGTAALGC
    LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPS
    SSLGTQTYICNVNHKPSNTKVDKKVEPKSC
      • 7) Exemplary amino acid sequences of CD3-FolR bispecific antibodies with common light chain
  • VHCH1[16D5]_VHCH1[CD3]_Fcknob EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 89
    pCON999 RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYC
    TTPWEWSWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC
    LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL
    GTQTYICNVNHKPSNTKVDKKVEPKSCDGGGGSGGGGSEVQLLESGGGL
    VQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVSRIRSKYNNYAT
    YYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYCVRHGNFGNSYV
    SWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF
    PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI
    CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPK
    DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN
    STYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREP
    QVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTP
    PVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS
    PGK
    VHCH1[16D5]_Fchole EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 90
    pCON983 RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYC
    TTPWEWSWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC
    LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL
    GTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFL
    FPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKP
    REEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAK
    GQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAVEWESNGQPEN
    NYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNRFTQ
    KSLSLSPGK
    CD3_common QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAFRGL 35
    light IGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCALWYSNLW
    chain VFGGGTKLTVLGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAV
    pETR13197 TVAWKADSSPVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSC
    QVTHEGSTVEKTVAPTECS
    VHCH1[CD3]_VHCH1[16D5]_Fcknob_PGLALA EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVS 91
    pETR13932 RIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYC
    VRHGNFGNSYVSWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGT
    AALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTV
    PSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDGGGGSGGGGSEVQLVE
    SGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIKSKT
    DGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPWEW
    SWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF
    PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI
    CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPK
    DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN
    STYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREP
    QVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTP
    PVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS
    PGK
    CD3_Fcknob_PGLALA EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVS 92
    pETR13917 RIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYC
    VRHGNFGNSYVSWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGT
    AALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTV
    PSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGG
    PSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHN
    AKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKT
    ISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESN
    GQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALH
    NHYTQKSLSLSPGK
    Fc_hole_PGLALA_HYRF DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE 93
    pETR10755 DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKE
    YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC
    AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR
    WQQGNVFSCSVMHEALHNRFTQKSLSLSPGK
    VHCL[CD3]_Fcknob_PGLALA EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVS 98
    pETR13378 RIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYC
    VRHGNFGNSYVSWFAYWGQGTLVTVSSASVAAPSVFIFPPSDEQLKSGT
    ASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSST
    LTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECDKTHTCPPCPAPE
    AAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGV
    EVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAP
    IEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVE
    WESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMH
    EALHNHYTQKSLSLSPGK
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 99
    inverted RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYC
    2 + 1 with TTPWEWSWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC
    N100A in LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL
    CDRH3 GTQTYICNVNHKPSNTKVDKKVEPKSCDGGGGSGGGGSEVQLLESGGGL
    pETR14096 VQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVSRIRSKYNNYAT
    YYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYCVRHGNFGASYV
    SWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF
    PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI
    CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPK
    DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN
    STYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREP
    QVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTP
    PVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS
    PGK
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 100
    inverted RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYC
    2 + 1 with TTPWEWSWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC
    S100aA in LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL
    CDR H3 GTQTYICNVNHKPSNTKVDKKVEPKSCDGGGGSGGGGSEVQLLESGGGL
    pETR14097 VQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVSRIRSKYNNYAT
    YYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYCVRHGNFGNAYV
    SWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF
    PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI
    CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPK
    DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN
    STYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREP
    QVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTP
    PVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS
    PGK
    CD3 light QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAFRGL 101
    chain fused IGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCALWYSNLW
    to CH1; VFGGGTKLTVLSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEP
    Fc_PGLALA; VTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNV
    pETR13862 NHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTL
    MISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTY
    RVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVY
    TLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL
    DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    16D5 VH EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 102
    fused to RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYC
    constant TTPWEWSWYDYWGQGTLVTVSSASVAAPSVFIFPPSDEQLKSGTASVVC
    kappa LLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSK
    chain; ADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
    pETR13859
    CD3 VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVS 103
    fused to RIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYC
    constant VRHGNFGNSYVSWFAYWGQGTLVTVSSASPKAAPSVTLFPPSSEELQAN
    lambda KATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYAASSY
    chain; LSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
    pETR13860
    IGHV1- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMG 104
    46*01 IINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR
    (X92343), GGSGGSFDYWGQGTLVTVSS
    plus JH4
    element
    IGHV1- QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 105
    69*06 GIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCAR
    (L22583), GGSGGSMDAWGQGTTVTVSS
    plus JH6
    element
    IGHV3- EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 106
    15*01 RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYC
    (X92216), TTGGSGGSFDYWGQGTLVTVSS
    plus JH4
    element
    IGHV3- EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS 107
    23*01 AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK
    (M99660), GGSGGSFDYWGQGTLVTVSS
    plus JH4
    element
    IGHV4- QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIG 108
    59*01 YIYYSGSTNYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARG
    (AB019438), GSGGSFDYWGQGTLVTVSS
    plus JH4
    element
    IGHV5- EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 109
    51*01 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCAR
    (M99686), GGSGGSFDYWGQGTLVTVSS
    plus JH4
    element
    CD3 specific QTVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAFRGL 110
    antibody IGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCALWYSNLW
    based on VFGGGTKLTVL
    humanized
    CH2527
    light chain
    hVK1-39 DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIY 111
    (JK4 J- AASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPLTF
    element) GGGTKVEIK
    VL7_46-13 QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAFRGL 112
    (humanized IGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCALWYSNLW
    anti-CD3 VFGGGTKLTVL
    antibody
    light chain)
      • 8) Exemplary 16D5 variants with reduced affinity
        • a. Exemplary light chain variants with reduced affinity
  • Name Sequence Seq ID No
    K53A QTVVTQEPSLTVSPGGTVTLTC GSSTGAVTTSNYAN WVQQKPGQAPRGLIG G 113
    aa TNARAP GTPARFSGSLLGGKAALTLSGVQPEDEAEYYC ALWYSNLWV FGGGT
    KLTVL
    K53A_VL_CDR1 GSSTGAVTTSNYAN
    32
    K53A_VL_CDR2 GTNARAP 311
    K53A_VL_CDR3 ALWYSNLWV 34
    S93A QTVVTQEPSLTVSPGGTVTLTC GSSTGAVTTSNYAN WVQQKPGQAPRGLIG G 114
    aa TNKRAP GTPARFSGSLLGGKAALTLSGVQPEDEAEYYC ALWYANLWV FGGGT
    KLTVL
    S93A_VL_CDR1 GSSTGAVTTSNYAN
    32
    S93A_VL_CDR2 GTNKRAP 33
    S93A_VL_CDR3 ALWYANLWV 312
        • b. Exemplary heavy chain variants with reduced affinity
  • Name Sequence Seq ID No
    S35H EVQLVESGGGLVKPGGSLRLSCAASGFTFS NAWMH WVRQAPGKGLEWVG RIK 115
    aa SKTDGGTTDYAAPVKG RFTISRDDSKNTLYLQMNSLKTEDTAVYYCTT PWEW
    SWYDY WGQGTLVTVSSAS
    S35H_VH_CDR1 NAWMH 313
    S35H_VH_CDR2 RIKSKTDGGTTDYAAPVKG 17
    S35H_VH_CDR3 PWEWSWYDY 18
    G49S EVQLVESGGGLVKPGGSLRLSCAASGFTFS NAWMS WVRQAPGKGLEWVS RIK 116
    aa SKTDGGTTDYAAPVKG RFTISRDDSKNTLYLQMNSLKTEDTAVYYCTT PWEW
    SWYDY WGQGTLVTVSSAS
    G49S_VH_CDR1 NAWMS
    16
    G49S_VH_CDR2 RIKSKTDGGTTDYAAPVKG 17
    G49S_VH_CDR3 PWEWSWYDY 18
    R50S EVQLVESGGGLVKPGGSLRLSCAASGFTFS NAWMS WVRQAPGKGLEWVG SIK 117
    aa SKTDGGTTDYAAPVKG RFTISRDDSKNTLYLQMNSLKTEDTAVYYCTT PWEW
    SWYDY WGQGTLVTVSSAS
    R50S_VH_CDR1 NAWMS
    16
    R50S_VH_CDR2 SIKSKTDGGTTDYAAPVKG 314
    R50S_VH_CDR3 PWEWSWYDY 18
    W96Y EVQLVESGGGLVKPGGSLRLSCAASGFTFS NAWMS WVRQAPGKGLEWVG RIK 118
    aa SKTDGGTTDYAAPVKG RFTISRDDSKNTLYLQMNSLKTEDTAVYYCTT PYEW
    SWYDY WGQGTLVTVSSAS
    W96Y_VH_CDR1 NAWMS
    16
    W96Y_VH_CDR2 RIKSKTDGGTTDYAAPVKG 17
    W96Y_VH_CDR3 PYEWSWYDY 315
    W98Y EVQLVESGGGLVKPGGSLRLSCAASGFTFS NAWMS WVRQAPGKGLEWVG RIK 119
    aa SKTDGGTTDYAAPVKG RFTISRDDSKNTLYLQMNSLKTEDTAVYYCTT PWEY
    SWYDY WGQGTLVTVSSAS
    W98Y_VH_CDR1 NAWMS
    16
    W98Y_VH_CDR2 RIKSKTDGGTTDYAAPVKG 17
    W98Y_VH_CDR3 PWEYSWYDY 232
      • 9) Additional exemplary embodiments generated from a phage display library (CDRs underlined)
  • Name Sequence Seq ID No
    90D7 QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMG IIN 120
    aa PSGGSTSYAQKFQG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR NYTIVV
    SPFDY WGQGTLVTVSSAS
    90D7_VH_CDR1 SYYMH 8
    90D7_VH_CDR2 IINPSGGSTSYAQKFQG 9
    90D7_VH_CDR3 NYTIVVSPFDY 233
    90C1 QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMG IIN 121
    aa PSGGSTSYAQKFQG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR NYFIGS
    VAMDY WGQGTLVTVSSAS
    90C1_VH_CDR1 SYYMH 8
    90C1_VH_CDR2 IINPSGGSTSYAQKFQG 9
    90C1_VH_CDR3 NYFIGSVAMDY 234
    5E8 VH QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMG IIN 122
    aa PSGGSTSYAQKFQG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR GLTYSM
    DY WGQGTLVTVSSAS
    5E8_VH_CDR1 SYYMH 8
    5E8_VH_CDR2 IINPSGGSTSYAQKFQG 9
    5E8_VH_CDR3 GLTYSMDY 235
    5E8 VL DIVMTQSPLSLPVTPGEPASISC RSSQSLLHSNGYNYLD WYLQKPGQSPQLL 123
    aa IY LGSNRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQALQIPNT FG
    QGTKVEIKRT
    5E8_VL_CDR1 RSSQSLLHSNGYNYLD 59
    5E8_VL_CDR2 LGSNRAS 60
    5E8_VL_CDR3 MQALQIPNT 236
    12A4 VH EVQLLESGGGLVQPGGSLRLSCAASGFTFS SYAMS WVRQAPGKGLEWVS AIS 124
    aa GSGGSTYYADSVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK YAYALD
    Y WGQGTLVTVSSAS
    12A4_VH_CDR1 SYAMS 79
    12A4_VH_CDR2 AISGSGGSTYYADSVKG 80
    12A4_VH_CDR3 YAYALDY 237
    12A4 VL EIVLTQSPGTLSLSPGERATLSC RASQSVSSSYLA WYQQKPGQAPRLLIY GA 125
    aa SSRAT GIPDRFSGSGSGTDFTLTISRLEPEDFAVYY CQQHGSSST FGQGTKV
    EIKRT
    12A4_VL_CDR1 RASQSVSSSYLA 52
    12A4_VL_CDR2 GASSRAT 53
    12A4_VL_CDR3 QQHGSSST 238
    7A3 VH QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMG IIN 126
    aa PSGGSTSYAQKFQG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR GDFSAG
    RLMDY WGQGTLVTVSSAS
    7A3_VH_CDR1 SYYMH 8
    7A3_VH_CDR2 IINPSGGSTSYAQKFQG 9
    7A3_VH_CDR3 GDFSAGRLMDY 239
    7A3 VL DIVMTQSPLSLPVTPGEPASISC RSSQSLLHSNGYNYLD WYLQKPGQSPQLL 127
    aa IY LGSNRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQALQTPPIT F
    GQGTKVEIKRT
    7A3_VL_CDR1 RSSQSLLHSNGYNYLD 59
    7A3_VL_CDR2 LGSNRAS 60
    7A3_VL_CDR3 MQALQTPPIT 240
    6E10 VH QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMG IIN 128
    aa PSGGSTSYAQKFQG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR GDYNAF
    DY WGHGTLVTVSSAS
    6E10_VH_CDR1 SYYMH 8
    6E10_VH_CDR2 IINPSGGSTSYAQKFQG 9
    6E10_VH_CDR3 GDYNAFDY 241
    6E10 VL DIVMTQSPLSLPVTPGEPASISC RSSQSLLHSNGYNYLD WYLQKPGQSPQLL 129
    aa IY LGSNRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQAWHSPT FGQ
    GTKVEIKRT
    6E10_VL_CDR1 RSSQSLLHSNGYNYLD 59
    6E10_VL_CDR2 LGSNRAS 60
    6E10_VL_CDR3 MQAWHSPT 242
    12F9 VH QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMG IIN 130
    aa PSGGSTSYAQKFQG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR GATYTM
    DY WGQGTLVTVSSAS
    12F9_VH_CDR1 SYYMH 8
    12F9_VH_CDR2 IINPSGGSTSYAQKFQG 9
    12F9_VH_CDR3 GATYTMDY 243
    12F9 VL DIVMTQSPLSLPVTPGEPASISC RSSQSLLHSNGYNYLD WYLQKPGQSPQLL 131
    aa IY LGSNRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQALQTPIT FG
    QGTKVEIKRT
    12F9_VL_CDR1 RSSQSLLHSNGYNYLD 59
    12F9_VL_CDR2 LGSNRAS 60
    12F9_VL_CDR3 MQALQTPIT 244
      • 10) 9D11 Glyscosite variants: variable light chain of exemplary embodiments (CDRs underlined)
  • Variant Sequence Seq ID No
    N95S DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLL 132
    IYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQASIMSRTFG
    QGTKVEIK
    12F9_VL_CDR1 RSSQSLLHSNGYNYLD 59
    12F9_VL_CDR2 LGSNRAS 60
    12F9_VL_CDR3 MQASIMSRT 63
    N95Q DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLL 133
    IYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQASIMQRTFG
    QGTKVEIK
    N95Q_VL_CDR1 RSSQSLLHSNGYNYLD 59
    N95Q_VL_CDR2 LGSNRAS 60
    N95Q_VL_CDR3 MQASIMQRT 65
    T97A DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLL 134
    IYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQASIMNRA FG
    QGTKVEIK
    T97A_VL_CDR1 RSSQSLLHSNGYNYLD 59
    T97A_VL_CDR2 LGSNRAS 60
    T97A_VL_CDR3 MQASIMNRA 67
    T97N DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLL 135
    IYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQASIMNRN FG
    QGTKVEIK
    T97N_VL_CDR1 RSSQSLLHSNGYNYLD 59
    T97N_VL_CDR2 LGSNRAS 60
    T97N_VL_CDR3 MQASIMNRN 69
      • 11) Deamination Variants
  • Variant Sequence Seq ID No
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIK 248
    VH_D52dE SKTEGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPWEW
    SWYDYWGQGTLVTVSS
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIK 249
    VH_D52dQ SKTQGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPWEW
    SWYDYWGQGTLVTVSS
    CD3_VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVSRIR 250
    N100A SKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYCVRHGNF
    GASYVSWFAYWGQGTLVTVSS
    CD3_VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVSRIR 251
    S100aA SKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYCVRHGNF
    GNAYVSWFAYWGQGTLVTVSS
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIK 252
    [VHCH1]- SKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPWEW
    CD3[VHCH1- SWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEP
    N100A]- VTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK
    Fcknob_PGLALA PSNTKVDKKVEPKSCDGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASG
    FTFSTYAMNWVRQAPGKGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSK
    NTLYLQMNSLRAEDTAVYYCVRHGNFGASYVSWFAYWGQGTLVTVSSASTKG
    PSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL
    QSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTC
    PPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY
    VDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALGA
    PIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWE
    SNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHN
    HYTQKSLSLSPGK
    16D5- EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIK 253
    Fchole- SKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPWEW
    PGLALA SWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEP
    VTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK
    PSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTP
    EVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVL
    HQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKN
    QVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVD
    KSRWQQGNVFSCSVMHEALHNRFTQKSLSLSPGK
    CD3-CLC QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAFRGLIGG 254
    TNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCALWYSNLWVFGGGT
    KLTVLGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSS
    PVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKT
    VAPTECS
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIK 255
    [VHCH1]- SKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPWEW
    CD3[VHCH1- SWYDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEP
    S100aA]- VTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK
    Fcknob_PGLALA PSNTKVDKKVEPKSCDGGGGSGG
    GGSEVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVS
    RIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRAEDTAVYYCVRH
    GNFGNAYVSWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCL
    VKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQT
    YICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKD
    TLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR
    VVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVYTLPP
    CRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF
    LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    9D11 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIIN 256
    [VHCH1]- PSGGPTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGDFAWL
    CD3[VHCL- DYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV
    N100A]- SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSN
    Fcknob_PGLALA TKVDKKVEPKSCDGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGFTF
    STYAMNWVRQAPGKGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTL
    YLQMNSLRAEDTAVYYCVRHGNFGASYVSWFAYWGQGTLVTVSSASVAAPSV
    FIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQD
    SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECDKTHT
    CPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNW
    YVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALG
    APIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEW
    ESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALH
    NHYTQKSLSLSPGK
    9D11- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIIN 257
    Fchole PSGGPTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGDFAWL
    DYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV
    SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSN
    TKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVT
    CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD
    WLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVS
    LSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR
    WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    9D11_LC DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLL 258
    [N95Q] IYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQASIMQRTFG
    QGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVD
    NALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSS
    PVTKSFNRGEC
    CD3_VLCH1 QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQAFRGLIGG 259
    TNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCALWYSNLWVFGGGT
    KLTVLSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGA
    LTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKK
    VEPKSC
    9D11 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIIN 260
    [VHCH1]- PSGGPTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGDFAWL
    CD3[VHCH1- DYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV
    S100aA]- SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSN
    Fcknob_PGLALA TKVDKKVEPKSCDGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGFTF
    STYAMNWVRQAPGKGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTL
    YLQMNSLRAEDTAVYYCVRHGNFGNAYVSWFAYWGQGTLVTVSSASVAAPSV
    FIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQD
    SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECDKTHT
    CPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNW
    YVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALG
    APIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEW
    ESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALH
    NHYTQKSLSLSPGK
      • 12) Mov19 based TCBs of exemplary embodiments (CDRs underlined)
  • Name Sequence Seq ID No
    pETR11646 QVQLQQSGAELVKPGASVKISCKASGYSFT GYFMN WVKQSHGKSLEWIG RIH 136
    Mov19 PYDGDTFYNQNFKD KATLTVDKSSNTAHMELLSLTSEDFAVYYCTR YDGSRA
    VH-CH1- MDY WGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVT
    Fchole VSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPS
    PG/LALA NTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEV
    TCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQ
    DWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQV
    SLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKS
    RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    pETR11647 QVQLQQSGAELVKPGASVKISCKASGYSFT GYFMN WVKQSHGKSLEWIG RIH 137
    Mov19 PYDGDTFYNQNFKD KATLTVDKSSNTAHMELLSLTSEDFAVYYCTR YDGSRA
    VH-CH1- MDY WGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVT
    CD3 VH- VSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPS
    CL- NTKVDKKVEPKSCDGGGGSGGGGSEVQLVESGGGLVQPKGSLKLSCAASGFT
    Fcknob FNTYAMN WVRQAPGKGLEWVA RIRSKYNNYATYYADSVKD RFTISRDDSQSI
    PG/LALA LYLQMNNLKTEDTAMYYCVR HGNFGNSYVSWFAY WGQGTLVTVSAASVAAPS
    VFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQ
    DSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECDKTH
    TCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN
    WYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL
    GAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVE
    WESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL
    HNHYTQKSLSLSPGK
    pETR11644 DIELTQSPASLAVSLGQRAIISC KASQSVSFAGTSLMH WYHQKPGQQPKLLI 138
    Mov19 LC Y RASNLEA GVPTRFSGSGSKTDFTLNIHPVEEEDAATYYC QQSREYPYT FGG
    GTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDN
    ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSP
    VTKSFNRGEC
    Hu IgG1 DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPE 245
    Fc VKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVS
    NKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSD
    IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVM
    HEALHNHYTQKSLSLSPGK
      • 13) Additional FolR1 TCBs with intermediate affinity binders (CDRs according to Kabat, underlined):
  • Name Sequence Seq ID No
    16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIKSK 274
    variant TEGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPYEWSWYD
    W96Y/D52E YWGQGTLVTVSS
    VH
    W96Y/D52E_VH NAWMS 16
    CDR1
    W96Y/D52E_VH RIKSKTEGGTTDYAAPVKG 275
    CDR2
    W96Y/D52E_VH PYEWSWYDY 315
    CDR3
    16D5 QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKPGQA 31
    variant FRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYC
    W96Y/D52E ALWYSNLWVFGGGTKLTVL
    VL
    W96Y/D52E_CD3- EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIKSK 276
    VHCH1_Fc- TEGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPYEWSWYD
    knob_PGLALA YWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWN
    pETR14945 SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDK
    KVEPKSCDGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNW
    VRQAPGKGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSLRA
    EDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS
    GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPS
    SSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFP
    PKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
    TYRVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVYTLP
    PCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFL
    YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    W96Y/D52E_Fc- EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVGRIKSK 277
    hole_PGLALA_HYRF TEGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCTTPYEWSWYD
    pETR14946 YWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWN
    SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDK
    KVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVS
    HEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
    KVSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPS
    DIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMH
    EALHNRFTQKSLSLSPGK
    14B1 EVQLLESGGGLVQPGGSLRLSCAASGFTFS SYAMS WVRQAPGKGLEWVS AISGS 278
    VH GGSTYYADSVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR GDYRYRYFDY
    WGQGTLVTVSS
    14B1 SSELTQDPAVSVALGQTVRITC QGDSLRSYYAS WYQQKPGQAPVLVIY GKNNRP 279
    VL S GIPDRFSGSSSGNTASLTITGAQAEDEADYYC NSRESPPTGLVV FGGGTKLTV
    L
    14B1[EE]_CD3[VLCH1]_Fc- EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGS 280
    knob_PGLALA GGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGDYRYRYFDY
    pETR14976 WGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVEDYFPEPVTVSWNS
    GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDEK
    VEPKSCDGGGGSGGGGSQAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANW
    VQEKPGQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCA
    LWYSNLWVFGGGTKLTVLSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFP
    EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK
    PSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEV
    TCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDW
    LNGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWC
    LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN
    VFSCSVMHEALHNHYTQKSLSLSPGK
    14B1[EE]_Fc- EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGS 281
    hole_PGLALA GGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGDYRYRYFDY
    pETR14977 WGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVEDYFPEPVTVSWNS
    GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDEK
    VEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
    EDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK
    VSNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSD
    IAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHE
    ALHNHYTQKSLSLSPGK
    14B1 LC SSELTQDPAVSVALGQTVRITCQGDSLRSYYASWYQQKPGQAPVLVIYGKNNRP 282
    [KK] SGIPDRFSGSSSGNTASLTITGAQAEDEADYYCNSRESPPTGLVVFGGGTKLTV
    Constant LGQPKAAPSVTLFPPSSKKLQANKATLVCLISDFYPGAVTVAWKADSSPVKAGV
    lambda ETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
    pETR14979
    9C7 VH QVQLVQSGAEVKKPGASVKVSCKASGYTFT SYYMH WVRQAPGQGLEWMGI INPS 283
    GGSTSYAQKFOG RVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR GDWSYYMDY W
    GQGTLVTVSS
    9C7 VL DIVMTQSPLSLPVTPGEPASISC RSSQSLLHSNGYNYLD WYLQKPGQSPQLLIY 284
    LGSNRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MOARQTPT FGQGTKV
    EIK
    9C7[EE]_CD3[VLCH1]_Fc- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPS 285
    knob_PGLALA GGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGDWSYYMDYW
    pETR14974 GQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVEDYFPEPVTVSWNSG
    ALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDEKV
    EPKSCDGGGGSGGGGSQAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWV
    QEKPGQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPEDEAEYYCAL
    WYSNLWVFGGGTKLTVLSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE
    PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKP
    SNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVT
    CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWL
    NGKEYKCKVSNKALGAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCL
    VKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNV
    FSCSVMHEALHNHYTQKSLSLSPGK
    9C7[EE]_Fc- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPS 286
    hole_PGLALA GGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGDWSYYMDYW
    pETR14975 GQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVEDYFPEPVTVSWNSG
    ALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDEKV
    EPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
    DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKV
    SNKALGAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDI
    AVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEA
    LHNHYTQKSLSLSPGK
    9C7 LC DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLLIY 316
    [RK] LGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQARQTPTFGQGTKV
    pETR14980 EIKRTVAAPSVFIFPPSDRKLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGN
    SQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRG
    EC
      • 14) Antigen Sequences
  • Antigen Sequence Seq ID No
    hu MAQRMTTQLLLLLVWVAVVGEAQTRIAWARTELLNVCMNAKHHKEKPGPEDKL 139
    FolR1 HEQCRPWRKNACCSTNTSQEAHKDVSYLYRFNWNHCGEMAPACKRHFIQDTCL
    YECSPNLGPWIQQVDQSWRKERVLNVPLCKEDCEQWWEDCRTSYTCKSNWHKG
    WNWTSGFNKCAVGAACQPFHFYFPTPTVLCNEIWTHSYKVSNYSRGSGRCIQM
    WFDPAQGNPNEEVARFYAAAM SGAGPWAAWPFLLSLALMLLWLLS
    huFolR1 RIAWARTELLNVCMNAKHHKEKPGPEDKLHEQCRPWRKNACCSTNTSQEAHKD
    140
    ECD- VSYLYRFNWNHCGEMAPACKRHFIQDTCLYECSPNLGPWIQQVDQSWRKERVL
    AcTev- NVPLCKEDCEQWWEDCRTSYTCKSNWHKGWNWTSGFNKCAVGAACQPFHFYFP
    Fcknob- TPTVLCNEIWTHSYKVSNYSRGSGRCIQMWFDPAQGNPNEEVARFYAAAMVD E
    Avi tag QLYFQG GSPKSADKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC
    VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWL
    NGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWC
    LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQG
    NVFSCSVMHEALHNHYTQKSLSLSPGKSGGLNDIFEAQKIEWHE
    Fchole DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV 141
    KFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNK
    ALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAV
    EWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEAL
    HNRFTQKSLSLSPGK
    mu MAHLMTVQLLLLVMWMAECAQSRATRARTELLNVCMDAKHHKEKPGPEDNLHD 142
    FolR1 QCSPWKTNSCCSTNTSQEAHKDISYLYRFNWNHCGTMTSECKRHFIQDTCLYE
    CSPNLGPWIQQVDQSWRKERILDVPLCKEDCQQWWEDCQSSFTCKSNWHKGWN
    WSSGHNECPVGASCHPFTFYFPTSAALCEEIWSHSYKLSNYSRGSGRCIQMWF
    DPAQGNPNEEVARFYAEAMSGAGLHGTWPLLCSLSLVLLWVIS
    mu TRARTELLNVCMDAKHHKEKPGPEDNLHDQCSPWKTNSCCSTNTSQEAHKDIS 143
    FolR1 YLYRFNWNHCGTMTSECKRHFIQDTCLYECSPNLGPWIQQVDQSWRKERILDV
    ECD- PLCKEDCQQWWEDCQSSFTCKSNWHKGWNWSSGHNECPVGASCHPFTFYFPTS
    AcTev- AALCEEIWSHSYKLSNYSRGSGRCIQMWFDPAQGNPNEEVARFYAEAMVD EQL
    Fcknob- YFQG GSPKSADKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVV
    Avitag VDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG
    KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLV
    KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNV
    FSCSVMHEALHNHYTQKSLSLSPGKSGGLNDIFEAQKIEWHE
    cy FolR1 MAQRMTTQLLLLLVWVAVVGEAQTRTARARTELLNVCMNAKHHKEKPGPEDKL 144
    HEQCRPWKKNACCSTNTSQEAHKDVSYLYRFNWNHCGEMAPACKRHFIQDTCL
    YECSPNLGPWIQQVDQSWRKERVLNVPLCKEDCERWWEDCRTSYTCKSNWHKG
    WNWTSGFNKCPVGAACQPFHFYFPTPTVLCNEIWTYSYKVSNYSRGSGRCIQM
    WFDPAQGNPNEEVARFYAAAMSGAGPWAAWPLLLSLALTLLWLLS
    cy FolR1 RTARARTELLNVCMNAKHHKEKPGPEDKLHEQCRPWKKNACCSTNTSQEAHKD 145
    ECD- VSYLYRFNWNHCGEMAPACKRHFIQDTCLYECSPNLGPWIQQVDQSWRKERVL
    AcTev- NVPLCKEDCEQWWEDCRTSYTCKSNWHKGWNWTSGFNKCPVGAACQPFHFYFP
    Fcknob- TPTVLCNEIWTYSYKVSNYSRGSGRCIQMWFDPAQGNPNEEVARFYAAAMVD E
    Avi tag QLYFQG GSPKSADKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC
    VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWL
    NGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWC
    LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQG
    NVFSCSVMHEALHNHYTQKSLSLSPGKSGGLNDIFEAQKIEWHE
    hu MVWKWMPLLLLLVCVATMCSAQDRTDLLNVCMDAKHHKTKPGPEDKLHDQCSP 146
    FolR2 WKKNACCTASTSQELHKDTSRLYNFNWDHCGKMEPACKRHFIQDTCLYECSPN
    LGPWIQQVNQSWRKERFLDVPLCKEDCQRWWEDCHTSHTCKSNWHRGWDWTSG
    VNKCPAGALCRTFESYFPTPAALCEGLWSHSYKVSNYSRGSGRCIQMWFDSAQ
    GNPNEEVARFYAAAMHVNAGEMLHGTGGLLLSLALMLQLWLLG
    hu TMCSAQDRTDLLNVCMDAKHHKTKPGPEDKLHDQCSPWKKNACCTASTSQELH 147
    FolR2 KDTSRLYNFNWDHCGKMEPACKRHFIQDTCLYECSPNLGPWIQQVNQSWRKER
    ECD- FLDVPLCKEDCQRWWEDCHTSHTCKSNWHRGWDWTSGVNKCPAGALCRTFESY
    AcTev- FPTPAALCEGLWSHSYKVSNYSRGSGRCIQMWFDSAQGNPNEEVARFYAAAMH
    Fcknob- VVD EQLYFQG GSPKSADKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTP
    Avi tag EVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLH
    QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQV
    SLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
    WQQGNVFSCSVMHEALHNHYTQKSLSLSPGKSGGLNDIFEAQKIEWHE
    hu MAWQMMQLLLLALVTAAGSAQPRSARARTDLLNVCMNAKHHKTQPSPEDELYG 148
    FolR3 QCSPWKKNACCTASTSQELHKDTSRLYNFNWDHCGKMEPTCKRHFIQDSCLYE
    CSPNLGPWIRQVNQSWRKERILNVPLCKEDCERWWEDCRTSYTCKSNWHKGWN
    WTSGINECPAGALCSTFESYFPTPAALCEGLWSHSFKVSNYSRGSGRCIQMWF
    DSAQGNPNEEVAKFYAAAMNAGAPSRGIIDS
    hu SARARTDLLNVCMNAKHHKTQPSPEDELYGQCSPWKKNACCTASTSQELHKDT 149
    FolR3 SRLYNFNWDHCGKMEPTCKRHFIQDSCLYECSPNLGPWIRQVNQSWRKERILN
    ECD- VPLCKEDCERWWEDCRTSYTCKSNWHKGWNWTSGINECPAGALCSTFESYFPT
    AcTev- PAALCEGLWSHSFKVSNYSRGSGRCIQMWFDSAQGNPNEEVAKFYAAAMNAGA
    Fcknob- PSRGIIDSVD EQLYFQG GSPKSADKTHTCPPCPAPELLGGPSVFLFPPKPKDT
    Avi tag LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVV
    SVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRD
    ELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSK
    LTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKSGGLNDIFEAQKIEW
    HE
    hu CD3ε MQSGTHWRVLGLCLLSVGVWGQDGNEEMGGITQTPYKVSISGTTVILTCPQYP 150
    GSEILWQHNDKNIGGDEDDKNIGSDEDHLSLKEFSELEQSGYYVCYPRGSKPE
    DANFYLYLRARVCENCMEMDVMSVATIVIVDICITGGLLLLVYYWSKNRKAKA
    KPVTRGAGAGGRQRGQNKERPPPVPNPDYEPIRKGQRDLYSGLNQRRI
      • 15) Nucleotide sequences of exemplary embodiments
  • Seq
    ID
    Description Sequence No
    16A3 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 151
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTACTACGCTGGTGTTACTCCGTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    15A1 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 152
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTACTACATCGGTGTTGTTACTTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    18D3 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 153
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTACTACACTGGTGGTTCTTCTGCTT
    TCGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    19E5 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 154
    NTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCGGTGAATGGCGTCGTTACACTTCTTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    19A4 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 155
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCGGTGGTTGGATCCGTTGGGAACATTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    15H7 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 156
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTACTACCTGTTCTCTACTTCTTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    15B6 CAGGTGCAATTGGTTCAATCTGGTGCTGAGGTAAAAAAACCGGGCG 157
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTACTACATCGGTATCGTTCCGTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 158
    GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG
    ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC
    15E12 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 159
    GTTCCCNGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACCGAAGAC
    ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTACTTCG
    ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC
    21D1 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 160
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTACTACGTTGGTGTTTCTCCGTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    16F12 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 161
    NTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCNTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCAACTTCACTGTTCTGCGTGTTCCGTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    21A5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 162
    GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGGCTTGGTTCG
    ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC
    21G8 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 163
    GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACCGAAGAC
    ACCGCAGTCTACTACTGTACTACCCCTTGGGAATGGGCTTACTTCG
    ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC
    19H3 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 164
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCACTGGTTGGTCTCGTTGGGGTTACATGG
    ACTATTGGGGCCAAGGCACCCTCGTAACGGTTTCTTCT
    20G6 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 165
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCGGTGAATGGATCCGTTACTACCATTTCG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    20H7 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 166
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCGTTGGTTGGTACCGTTGGGGTTACATGG
    ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    11F8_VH CAGGTGCAATTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGT 167
    CCTCGGTGAAGGTCTCCTGCAAGGCCTCCGGAGGCACATTCAGCAG
    CTACGCTATAAGCTGGGTGCGACAGGCCCCTGGACAAGGGCTCGAG
    TGGATGGGAGGGATCATCCCTATCTTTGGTACAGCAAACTACGCAC
    AGAAGTTCCAGGGCAGGGTAACCATTACTGCAGACAAATCCACGAG
    CACAGCCTACATGGAGCTGAGCAGCCTGAGATCTGAGGACACCGCC
    GTGTATTACTGTGCGAGAGCTGTTTTCTACCGTGCTTGGTACTCTT
    TCGACTACTGGGGCCAAGGGACCACCGTGACCGTCTCCTCA
    11F8_VL GACATCCAGATGACCCAGTCTCCTTCCACCCTGTCTGCATCTGTAG 168
    GAGACCGTGTCACCATCACTTGCCGTGCCAGTCAGAGTATTAGTAG
    CTGGTTGGCCTGGTATCAGCAGAAACCAGGGAAAGCCCCTAAGCTC
    CTGATCTATGATGCCTCCAGTTTGGAAAGTGGGGTCCCATCACGTT
    TCAGCGGCAGTGGATCCGGGACAGAATTCACTCTCACCATCAGCAG
    CTTGCAGCCTGATGATTTTGCAACTTATTACTGCCAACAGTATACC
    AGCCCACCACCAACGTTTGGCCAGGGCACCAAAGTCGAGATCAAG
    36F2_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 169
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCATGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCTCTTTCTTCACTGGTTTCCATCTGGACT
    ATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    36F2_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 170
    GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG
    CAGCTACTTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG
    CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA
    GGTTCAGTGGCAGTGGATCCGGGACAGACTTCACTCTCACCATCAG
    CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGTAT
    ACCAACGAACATTATTATACGTTCGGCCAGGGGACCAAAGTGGAAA
    TCAAA
    9D11_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 171
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCCCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTGGACTATTGGG
    GTCAAGGCACCCTCGTAACGGTTTCTTCT
    9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 172
    GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA
    CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT
    CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG
    GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC
    GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC
    TGTATGCAGGCAAGCATTATGAACCGGACTTTTGGTCAAGGCACCA
    AGGTCGAAATTAAA
    9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 173
    N95S GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA
    CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT
    CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG
    GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC
    GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC
    TGTATGCAGGCAAGCATTATGAGCCGGACTTTTGGTCAAGGCACCA
    AGGTCGAAATTAAA
    9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 174
    N95Q GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA
    CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT
    CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG
    GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC
    GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC
    TGTATGCAGGCAAGCATTATGCAGCGGACTTTTGGTCAAGGCACCA
    AGGTCGAAATTAAA
    9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 175
    T97A GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA
    CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT
    CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG
    GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC
    GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC
    TGTATGCAGGCAAGCATTATGAACCGGGCTTTTGGTCAAGGCACCA
    AGGTCGAAATTAAA
    9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 176
    T97N GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA
    CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT
    CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG
    GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC
    GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC
    TGTATGCAGGCAAGCATTATGAACCGGAATTTTGGTCAAGGCACCA
    AGGTCGAAATTAAA
    5D9_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 177
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCTCTTACATCGACATGGACTATTGGGGTC
    AAGGCACCCTCGTAACGGTTTCTTCT
    5D9_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 178
    GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG
    CAGCTACTTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG
    CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA
    GGTTCAGTGGCAGTGGATCCGGGACAGACTTCACTCTCACCATCAG
    CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGGAT
    AACTGGAGCCCAACGTTCGGCCAGGGGACCAAAGTGGAAATCAAA
    6B6_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 179
    CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCTCTTACGTTGACATGGACTATTGGGGTC
    AAGGCACCCTCGTAACGGTTTCTTCT
    6B6_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 180
    GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG
    CAGCTACCTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG
    CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA
    GGTTCAGTGGCAGTGGATCCGGGACAGACTTCACTCTCACCATCAG
    CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGGAT
    ATTTGGAGCCCAACGTTCGGCCAGGGGACCAAAGTGGAAATCAAA
    14E4_VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGG 181
    GGTCCCTGAGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAG
    TTATGCCATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAG
    TGGGTCTCAGCTATTAGTGGTAGTGGTGGTAGCACATACTACGCAG
    ACTCCGTGAAGGGCCGGTTCACCATCTCCAGAGACAATTCCAAGAA
    CACGCTGTATCTGCAGATGAACAGCCTGAGAGCCGAGGACACGGCC
    GTATATTACTGTGCGAAAGACTCTTCTTACGTTGAATGGTACGCTT
    TCGACTACTGGGGCCAAGGAACCCTGGTCACCGTCTCGAGT
    14E4_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 182
    GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG
    CAGCTACTTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG
    CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA
    GGTTCAGTGGCAGTGGATCCGGGACAGACTCCACTCTCACCATCAG
    CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGCCA
    ACCAGCAGCCCAATTACGTTCGGCCAGGGGACCAAAGTGGAAATCA
    AA
    CD3 heavy GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCT 183
    chain GGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACC
    (VHCH1) TTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC
    AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAAC
    AACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTC
    ACCATCAGCCGGGACGACAGCAAGAACACCCTGTACCTGCAG
    ATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGT
    GTGCGGCACGGCAACTTCGGCAACAGCTATGTGTCTTGGTTT
    GCCTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCT
    AGTACCAAGGGCCCCAGCGTGTTCCCCCTGGCACCCAGCAGC
    AAGAGCACATCTGGCGGAACAGCCGCTCTGGGCTGTCTGGTG
    AAAGACTACTTCCCCGAGCCCGTGACCGTGTCTTGGAACTCT
    GGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCCGTGCTG
    CAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTG
    CCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTC
    AATCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAG
    CCCAAGAGCTGC
    Crossed GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 184
    CD3 heavy GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC
    chain CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA
    (VHCκ) TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT
    ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG
    CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC
    ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT
    ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT
    GTCAAGCGCTAGTGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCA
    TCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGC
    TGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGTGGA
    TAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAG
    GACTCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGT
    CTAAGGCTGATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCAC
    CCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGA
    GAGTGT
    Mutagenesis GCAGGCAAGCATTATGCAGCGGACTTTTGGTCAAGG 185
    primer
    GAB7734
    N95Q
    Mutagenesis CAGGCAAGCATTATGAGCCGGACTTTTGGTCAAGG 186
    primer
    GAB7735
    N95S
    Mutagenesis CATTATGAACCGGGCTTTTGGTCAAGGCACCAAGGTC 187
    primer
    GAB7736
    T97A
    Mutagenesis CATTATGAACCGGAATTTTGGTCAAGGCACCAAGGTC 188
    primer
    GAB7737
    T97N
    VHCH1[16D5]_VHCH1[CD3]_Fcknob_PGLALA GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 189
    pCON999 GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    (Inverted CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    TCB with TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    16D5 2 + 1: ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    pCON999 + CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    pCON983 + ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG
    pETR13197) ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC
    AAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACA
    AGCGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCC
    CCGAGCCCGTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGG
    CGTGCACACTTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCC
    CTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGA
    CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGA
    CAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGA
    GGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGG
    TGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTT
    CACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC
    AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACT
    ACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAG
    CCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTG
    CGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACT
    TCGGCAACAGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCAC
    CCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGCCCCAGCGTGTTC
    CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC
    TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC
    TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC
    GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG
    TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA
    TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG
    AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG
    CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA
    CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG
    GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG
    ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA
    GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC
    CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA
    AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG
    GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT
    GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT
    TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC
    GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC
    TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC
    AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA
    CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    VHCH1[16D5]_Fchole_PGLALA_HYRF GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 190
    pCON983 GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG
    ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC
    CAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACC
    AGCGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCC
    CCGAGCCCGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGG
    CGTGCACACCTTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGC
    CTGAGCAGCGTGGTCACCGTGCCTTCTAGCAGCCTGGGCACCCAGA
    CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGA
    CAAGAAGGTGGAGCCCAAGAGCTGCGACAAAACTCACACATGCCCA
    CCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCT
    TCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGA
    GGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTC
    AAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGA
    CAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAG
    CGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTAC
    AAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAA
    CCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTGCAC
    CCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTC
    TCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGT
    GGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCC
    CGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACC
    GTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCG
    TGATGCATGAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTC
    CCTGTCTCCGGGTAAA
    CD3_common CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCG 191
    light GCACCGTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCAC
    chain CAGCAACTACGCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTC
    pETR13197 AGAGGACTGATCGGCGGCACCAACAAGAGAGCCCCTGGCACCCCTG
    CCAGATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCCTGACACT
    GTCTGGCGCCCAGCCAGAAGATGAGGCCGAGTACTACTGCGCCCTG
    TGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACAG
    TCCTAGGTCAACCCAAGGCTGCCCCCAGCGTGACCCTGTTCCCCCC
    CAGCAGCGAGGAACTGCAGGCCAACAAGGCCACCCTGGTCTGCCTG
    ATCAGCGACTTCTACCCAGGCGCCGTGACCGTGGCCTGGAAGGCCG
    ACAGCAGCCCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAGCAA
    GCAGAGCAACAACAAGTACGCCGCCAGCAGCTACCTGAGCCTGACC
    CCCGAGCAGTGGAAGAGCCACAGGTCCTACAGCTGCCAGGTGACCC
    ACGAGGGCAGCACCGTGGAGAAAACCGTGGCCCCCACCGAGTGCAG
    C
    VHCH1[CD3]_VHCH1[16D5]_Fcknob_PGLALA GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 192
    pETR13932 GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC
    (Classical CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA
    TCB with TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT
    16D5; 2 + 1: ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG
    pETR13932 + CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC
    pCON983 + ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT
    pETR13197) ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT
    GTCATCTGCTAGCACAAAGGGCCCTAGCGTGTTCCCTCTGGCCCCC
    AGCAGCAAGAGCACAAGCGGCGGAACAGCCGCCCTGGGCTGCCTCG
    TGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCTTGGAACAGCGG
    AGCCCTGACAAGCGGCGTGCACACCTTCCCTGCCGTGCTGCAGAGC
    AGCGGCCTGTACTCCCTGAGCAGCGTGGTCACCGTGCCTAGCAGCA
    GCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAG
    CAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGC
    GGAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAATTGGTTGAAT
    CTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCCTGCGTCTGAGCTG
    CGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAGCTGGGTT
    CGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAGT
    CTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGG
    TCGTTTTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTG
    CAGATGAACTCTCTGAAAACTGAAGACACCGCAGTCTACTACTGTA
    CTACCCCGTGGGAATGGTCTTGGTACGATTATTGGGGCCAGGGCAC
    GCTGGTTACGGTGTCTAGCGCTAGTACCAAGGGCCCCAGCGTGTTC
    CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC
    TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC
    TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC
    GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG
    TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA
    TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG
    AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG
    CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA
    CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG
    GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG
    ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA
    GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC
    CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA
    AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG
    GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT
    GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT
    TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC
    GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC
    TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC
    AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA
    CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    VHCH1[CD3]_Fcknob_PGLALA GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 193
    pETR13719 GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC
    (16D5 IgG CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA
    format, 1 + 1: TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT
    pETR13719 + ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG
    pCON983 + CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC
    pETR13197) ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT
    ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT
    GTCATCTGCTAGCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCC
    TCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGG
    TCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGG
    CGCCCTGACCAGCGGCGTGCACACCTTCCCGGCTGTCCTACAGTCC
    TCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGCA
    GCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAG
    CAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGTGACAAA
    ACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGAC
    CGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGAT
    CTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCAC
    GAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGG
    TGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCAC
    GTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTG
    AATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCG
    CCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGA
    ACCACAGGTGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAG
    AACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCTTCTATCCCAGCG
    ACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTA
    CAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTC
    TACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACG
    TCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACAC
    GCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    Fc_hole_PGLALA_HYRF GACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAACTCCTGG 194
    pETR10755 GGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCT
    (16D5 Head- CATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTG
    to-tail, 1 + 1: AGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCG
    pCON999 + TGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAA
    pETR10755 + CAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGAC
    pETR13197) TGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCC
    TCCCAGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCC
    CCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAGCTG
    ACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATC
    CCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAA
    CAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC
    TTCCTCGTGAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGG
    GGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCG
    CTTCACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    VHCH1[9D11]_VHCL[CD3]_Fcknob_PGLALA CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 195
    pCON1057 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    (9D11 CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    inverted TGGATGGGCATCATTAACCCAAGCGGTGGCCCTACCTCCTACGCGC
    format, 2 + 1: AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    pCON1057 + TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    pCON1051 + GTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTGGACTATTGGG
    pCON1063 + GTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACAAAGGGCCC
    pETR12940) CAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACATCTGGCGGA
    ACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAGCCTG
    TGACCGTGTCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACAC
    CTTTCCAGCCGTGCTGCAGAGCAGCGGCCTGTACTCTCTGAGCAGC
    GTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGACCTACATCT
    GCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGACAAGAAGGT
    GGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGAGGCGGAGGA
    TCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTG
    GCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAG
    CACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTG
    GAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCT
    ACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGA
    CAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAG
    GACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACA
    GCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGAC
    CGTGTCAAGCGCTAGTGTGGCCGCTCCCTCCGTGTTTATCTTTCCC
    CCATCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTC
    TGCTGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGT
    GGATAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAA
    CAGGACTCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACAC
    TGTCTAAGGCTGATTATGAGAAACACAAAGTCTACGCCTGCGAAGT
    CACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGG
    GGAGAGTGTGACAAGACCCACACCTGTCCCCCTTGTCCTGCCCCTG
    AAGCTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAGCCCAA
    GGACACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTG
    GTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACG
    TGGACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGA
    GCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTG
    CACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCA
    ACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAA
    AGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGG
    GATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAG
    GCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCA
    GCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGAC
    GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGT
    GGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCT
    GCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    9D11_Fchole_PGLALA_HYRF CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 196
    pCON1051 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC
    CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA
    TGGATGGGCATCATTAACCCAAGCGGTGGCCCTACCTCCTACGCGC
    AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC
    TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA
    GTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTGGACTATTGGG
    GTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACCAAGGGCCC
    CTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGC
    ACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCCG
    TGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACAC
    CTTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGC
    GTGGTCACCGTGCCTTCTAGCAGCCTGGGCACCCAGACCTACATCT
    GCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAGAAGGT
    GGAGCCCAAGAGCTGCGACAAAACTCACACATGCCCACCGTGCCCA
    GCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAA
    AACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATG
    CGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAAC
    TGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGC
    GGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCAC
    CGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAG
    GTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCA
    AAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCC
    ATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTCTCGTGCGCA
    GTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCA
    ATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGA
    CTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGACAAG
    AGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATG
    AGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCC
    GGGTAAA
    9D11_LC GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 197
    pCON1063 GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA
    CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT
    CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG
    GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC
    GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC
    TGTATGCAGGCAAGCATTATGAACCGGACTTTTGGTCAAGGCACCA
    AGGTCGAAATTAAACGTACGGTGGCTGCACCATCTGTCTTCATCTT
    CCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTG
    TGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGA
    AGGTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCAC
    AGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTG
    ACGCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGCG
    AAGTCACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAA
    CAGGGGAGAGTGT
    VLCH1[CD3] CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCG 198
    pETR12940 GCACCGTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCAC
    CAGCAACTACGCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTC
    AGAGGACTGATCGGCGGCACCAACAAGAGAGCCCCTGGCACCCCTG
    CCAGATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCCTGACACT
    GTCTGGCGCCCAGCCAGAAGATGAGGCCGAGTACTACTGCGCCCTG
    TGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACAG
    TGCTGAGCAGCGCTTCCACCAAAGGCCCTTCCGTGTTTCCTCTGGC
    TCCTAGCTCCAAGTCCACCTCTGGAGGCACCGCTGCTCTCGGATGC
    CTCGTGAAGGATTATTTTCCTGAGCCTGTGACAGTGTCCTGGAATA
    GCGGAGCACTGACCTCTGGAGTGCATACTTTCCCCGCTGTGCTGCA
    GTCCTCTGGACTGTACAGCCTGAGCAGCGTGGTGACAGTGCCCAGC
    AGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGC
    CCAGCAACACCAAGGTGGACAAGAAGGTGGAACCCAAGTCTTGT
    VHCL[CD3]_Fcknob_PGLALA GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 199
    pETR13378 GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC
    (9D11 CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA
    CrossMab TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT
    format, 1 + 1: ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG
    pETR13378 + CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC
    pCON1051 + ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT
    pCON1063 + ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT
    pETR12940) GTCATCTGCTAGCGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCA
    TCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGC
    TGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGTGGA
    TAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAG
    GACTCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGT
    CTAAGGCTGATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCAC
    CCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGA
    GAGTGTGACAAGACCCACACCTGTCCCCCTTGTCCTGCCCCTGAAG
    CTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAGCCCAAGGA
    CACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTGGTG
    GATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACGTGG
    ACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGCA
    GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC
    CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA
    AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG
    GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT
    GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT
    TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC
    GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC
    TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC
    AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA
    CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 200
    inverted GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    2 + 1 with CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    N100A in TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    CDR H3 ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    pETR14096 + CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    (pETR14096 + ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG
    pCON983 + ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC
    pETR13197) AAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACA
    AGCGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCC
    CCGAGCCCGTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGG
    CGTGCACACTTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCC
    CTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGA
    CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGA
    CAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGA
    GGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGG
    TGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTT
    CACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC
    AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACT
    ACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAG
    CCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTG
    CGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACT
    TCGGCGCCAGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCAC
    CCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGCCCCAGCGTGTTC
    CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC
    TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC
    TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC
    GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG
    TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA
    TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG
    AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG
    CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA
    CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG
    GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG
    ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA
    GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC
    CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA
    AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG
    GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT
    GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT
    TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC
    GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC
    TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC
    AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA
    CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 201
    inverted GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    2 + 1 with CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    S100aA in TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    CDR H3 ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    pETR14097 CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    (pETR14097 + ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG
    pCON983 + ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC
    pETR13197) AAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACA
    AGCGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCC
    CCGAGCCCGTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGG
    CGTGCACACTTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCC
    CTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGA
    CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGA
    CAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGA
    GGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGG
    TGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTT
    CACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC
    AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACT
    ACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAG
    CCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTG
    CGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACT
    TCGGCAACGCCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCAC
    CCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGCCCCAGCGTGTTC
    CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC
    TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC
    TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC
    GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG
    TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA
    TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG
    AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG
    CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA
    CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG
    GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG
    ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA
    GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC
    CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA
    AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG
    GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT
    GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT
    TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC
    GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC
    TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC
    AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA
    CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    CD3 light CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCG 202
    chain fused GCACCGTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCAC
    to CH1; CAGCAACTACGCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTC
    Fc_PGLALA; AGAGGACTGATCGGCGGCACCAACAAGAGAGCCCCTGGCACCCCTG
    pETR13862 CCAGATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCCTGACACT
    (Kappa- GTCTGGCGCCCAGCCAGAAGATGAGGCCGAGTACTACTGCGCCCTG
    lambda TGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACAG
    antibody with TGCTGAGCAGCGCTAGCACCAAGGGCCCATCGGTCTTCCCCCTGGC
    CD3 common ACCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGC
    light chain CTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACT
    fused to CH1 + CAGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGGCTGTCCTACA
    Fc_PGLALA. GTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCC
    VHs fused to AGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGC
    kappa or CCAGCAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGTGA
    lambda CAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGG
    constant GGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCA
    chain TGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAG
    pETR13859 + CCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTG
    pETR13860 + GAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACA
    pETR13862) GCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTG
    GCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTC
    GGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCC
    GAGAACCA
    CAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACC
    AGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACAT
    CGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAG
    ACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACA
    GCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTT
    CTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAG
    AAGAGCCTCTCCCTGTCTCCGGGTAAA
    16D5 VH GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 203
    fused to GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA
    constant CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG
    kappa TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT
    chain; ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG
    pETR13859 CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC
    ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG
    ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCGT
    GGCCGCTCCCTCCGTGTTCATCTTCCCACCTTCCGACGAGCAGCTG
    AAGTCCGGCACCGCTTCTGTCGTGTGCCTGCTGAACAACTTCTACC
    CCCGCGAGGCCAAGGTGCAGTGGAAGGTGGACAACGCCCTGCAGTC
    CGGCAACAGCCAGGAATCCGTGACCGAGCAGGACTCCAAGGACAGC
    ACCTACTCCCTGTCCTCCACCCTGACCCTGTCCAAGGCCGACTACG
    AGAAGCACAAGGTGTACGCCTGCGAAGTGACCCACCAGGGCCTGTC
    TAGCCCCGTGACCAAGTCTTTCAACCGGGGCGAGTGC
    CD3 VH GAAGTGCAGCTGCTGGAATCCGGCGGAGGACTGGTGCAGCCTGGCG 204
    fused to GATCTCTGAGACTGTCTTGTGCCGCCTCCGGCTTCACCTTCTCCAC
    constant CTACGCCATGAACTGGGTGCGACAGGCTCCTGGCAAGGGCCTGGAA
    lambda TGGGTGTCCCGGATCAGATCCAAGTACAACAACTACGCCACCTACT
    chain; ACGCCGACTCCGTGAAGGGCCGGTTCACCATCTCTCGGGACGACTC
    pETR13860 CAAGAACACCCTGTACCTGCAGATGAACTCCCTGCGGGCCGAGGAC
    ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACTCCT
    ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT
    GTCATCTGCTAGCCCCAAGGCTGCCCCCAGCGTGACCCTGTTTCCC
    CCCAGCAGCGAGGAACTGCAGGCCAACAAGGCCACCCTGGTCTGCC
    TGATCAGCGACTTCTACCCAGGCGCCGTGACCGTGGCCTGGAAGGC
    CGACAGCAGCCCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAGC
    AAGCAGAGCAACAACAAGTACGCCGCCAGCAGCTACCTGAGCCTGA
    CCCCCGAGCAGTGGAAGAGCCACAGGTCCTACAGCTGCCAGGTGAC
    CCACGAGGGCAGCACC
    GTGGAGAAAACCGTGGCCCCCACCGAGTGCAGC
    VHCH1[36F2]_VHCL[CD3]_Fc CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 246
    knob_PGLALA GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    pCON1056 CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCATGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCTCTTTCTTCACT
    GGTTTCCATCTGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    GCTAGCACAAAGGGCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGC
    ACATCTGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCC
    GAGCCTGTGACCGTGTCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCAC
    ACCTTTCCAGCCGTGCTGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTG
    GTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTG
    AACCACAAGCCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAG
    AGCTGTGATGGCGGAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAGCTG
    CTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTCTGAGACTGAGC
    TGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAACTGGGTGCGC
    CAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTAC
    AACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATC
    AGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGG
    GCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAAC
    AGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGTG
    TCAAGCGCTAGTGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCATCCGAT
    GAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGCTGAACAATTTT
    TACCCTAGGGAAGCTAAAGTGCAGTGGAAAGTGGATAACGCACTGCAGTCC
    GGCAACTCCCAGGAATCTGTGACAGAACAGGACTCCAAGGACAGCACCTAC
    TCCCTGTCCTCCACCCTGACACTGTCTAAGGCTGATTATGAGAAACAC
    AAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAGCTCGCCCGTCACA
    AAGAGCTTCAACAGGGGAGAGTGTGACAAGACCCACACCTGTCCCCCTTGT
    CCTGCCCCTGAAGCTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAG
    CCCAAGGACACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTG
    GTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACGTGGAC
    GGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGCAGTACAAC
    AGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTG
    AATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCC
    ATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTG
    TACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTG
    TGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAG
    AGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGAC
    TCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGC
    AGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTG
    CACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    36F2-Fchole CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 247
    PGLALA GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    pCON1050 CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCATGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCTCTTTCTTCACT
    GGTTTCCATCTGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT
    GCTAGCACCAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGC
    ACCAGCGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCC
    GAGCCCGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCAC
    ACCTTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTG
    GTCACCGTGCCTTCTAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTG
    AACCACAAGCCCAGCAACACCAAGGTGGACAAGAAGGTGGAGCCCAAG
    AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCA
    GGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATG
    ATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAA
    GACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAAT
    GCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTC
    AGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAG
    TGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCC
    AAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCCATCC
    CGGGATGAGCTGACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGC
    TTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAG
    AACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTC
    CTCGTGAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTC
    TTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAG
    AAGAGCCTCTCCCTGTCTCCGGGTAAA
    36F2 LC GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAGGGGAA 97
    pCON1062 AGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAGCAGCTACTTA
    GCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGGCTCCTCATCTATGGA
    GCATCCAGCAGGGCCACTGGCATCCCnAGACAGGTTCAGTGGCAGTGGATC
    CGGGACAGACTTCACTCTCACCATCAGCAGACTGGAGCCTGAAGATTTTGC
    AGTGTATTACTGTCAGCAGTATACCAACGAACATTATTATACGTTCGGCCA
    GGGGACCAAAGTGGAAATCAAACGTACGGTGGCTGCACCATCTGTCTTCAT
    CTTCCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTGTG
    CCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGAAGGTGGA
    TAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCACAGAGCAGGACAG
    CAAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAAGCAGA
    CTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCANGGCCTGAG
    CTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGT
    CD3 VLCH1 CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACC 198
    pETR12940 GTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTAC
    GCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGC
    GGCACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTG
    CTGGGAGGAAAGGCCGCCCTGACACTGTCTGGCGCCCAGCCAGAAGATGAG
    GCCGAGTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGA
    GGCACCAAGCTGACAGTGCTGAGCAGCGCTTCCACCAAAGGCCCTTCCGTG
    TTTCCTCTGGCTCCTAGCTCCAAGTCCACCTCTGGAGGCACCGCTGCTCTC
    GGATGCCTCGTGAAGGATTATTTTCCTGAGCCTGTGACAGTGTCCTGGAAT
    AGCGGAGCACTGACCTCTGGAGTGCATACTTTCCCCGCTGTGCTGCAGTCC
    TCTGGACTGTACAGCCTGAGCAGCGTGGTGACAGTGCCCAGCAGCAGCCTG
    GGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAG
    GTGGACAAGAAGGTGGAACCCAAGTCTTGT
    Seq
    ID
    Name Sequence No
    K53A CAGACCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACC 205
    nt GTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTAC
    GCCAACTGGGTGCAGCAGAAGCCAGGCCAGGCTCCCAGAGGACTGATCGGC
    GGCACCAACGCCAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTG
    CTGGGAGGAAAGGCCGCCCTGACACTGTCTGGCGTGCAGCCTGAAGATGAG
    GCCGAGTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGA
    GGCACCAAGCTGACAGTCCTA
    S93A CAGACCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACC 206
    nt GTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTAC
    GCCAACTGGGTGCAGCAGAAGCCAGGCCAGGCTCCCAGAGGACTGATCGGC
    GGCACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTG
    CTGGGAGGAAAGGCCGCCCTGACACTGTCTGGCGTGCAGCCTGAAGATGAG
    GCCGAGTACTACTGCGCCCTGTGGTACGCCAACCTGTGGGTGTTCGGCGGA
    GGCACCAAGCTGACAGTCCTA
    S35H GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 207
    nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG
    CACTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATC
    AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC
    CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG
    AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG
    GAGTGGTCTTGGTACGACTATTGGGGCCAGGGCACCCTCGTGACCGTGTCC
    TCTGCTAGC
    G49S GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 208
    nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG
    AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGTCCCGGATC
    AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC
    CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG
    AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG
    GAGTGGTCTTGGTACGACTATTGGGGCCAGGGCACCCTCGTGACCGTGTCC
    TCTGCTAGC
    R50S GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 209
    nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG
    AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGATCTATC
    AAGAGCAAGACCGACGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC
    CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG
    AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG
    GAGTGGTCTTGGTACGACTATTGGGGCCAGGGCACCCTCGTGACCGTGTCC
    TCTGCTAGC
    W96Y GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 210
    nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG
    AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATC
    AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC
    CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG
    AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTAC
    GAGTGGTCTTGGTACGACTACTGGGGCCAGGGCACCCTCGTGACCGTGTCA
    TCT
    GCTAGC
    W98Y GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 211
    nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG
    AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATC
    AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC
    CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG
    AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG
    GAGTACTCTTGGTACGACTACTGGGGCCAGGGCACCCTCGTGACCGTGTCA
    TCT
    GCTAGC
    90D7 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 212
    nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCAACTACACTATC
    GTTGTTTCTCCGTTCGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCT
    TCTGCTAGC
    90C1 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 213
    nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCAACTACTTCATC
    GGTTCTGTTGCTATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCT
    TCTGCTAGC
    5E8 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 214
    nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTCTGACTTAC
    TCTATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGC
    5E8 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 215
    nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC
    TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG
    CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC
    GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA
    GAAGACGTGGGCGTTTATTACTGTATGCAGGCACTGCAGATTCCAAACACT
    TTTGGTCAAGGCACCAAGGTCGAAATTAAACGTACG
    12A4 VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCC 216
    nt CTGAGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAGTTATGCCATG
    AGCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCAGCTATT
    AGTGGTAGTGGTGGTAGCACATACTACGCAGACTCCGTGAAGGGCCGGTTC
    ACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAGATGAACAGC
    CTGAGAGCCGAGGACACGGCCGTATATTACTGTGCGAAATACGCTTACGCT
    CTGGACTACTGGGGCCAAGGAACCCTGGTCACCGTCTCGAGTGCTAGC
    12A4 VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAGGGGAA 217
    nt AGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAGCAGCTACTTA
    GCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGGCTCCTCATCTATGGA
    GCATCCAGCAGGGCCACTGGCATCCCAGACAGGTTCAGTGGCAGTGGATCC
    GGGACAGACTTCACTCTCACCATCAGCAGACTGGAGCCTGAAGATTTTGCA
    GTGTATTACTGTCAGCAGCATGGCAGCAGCAGCACGTTCGGCCAGGGGACC
    AAAGTGGAAATCAAACGTACG
    7A3 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 218
    nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCTCT
    GCTGGTCGTCTGATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCT
    TCTGCTAGC
    7A3 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 219
    nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC
    TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG
    CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC
    GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA
    GAAGACGTGGGCGTTTATTACTGTATGCAGGCACTGCAGACCCCACCAATT
    ACCTTTGGTCAAGGCACCAAGGTCGAAATTAAACGTACG
    6E10 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 220
    nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTACAAC
    GCTTTCGACTATTGGGGTCACGGCACCCTCGTAACGGTTTCTTCTGCTAGC
    6E10 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 221
    nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC
    TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG
    CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC
    GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA
    GAAGACGTGGGCGTTTATTACTGTATGCAGGCATGGCATAGCCCAACTTTT
    GGTCAAGGCACCAAGGTCGAAATTAAACGTACG
    12F9 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 222
    nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG
    CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT
    AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC
    ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC
    CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGCTACTTAC
    ACTATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGC
    12F9 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 223
    nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC
    TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG
    CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC
    GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA
    GAAGACGTGGGCGTTTATTACTGTATGCAGGCACTGCAGACCCCAATTACT
    TTTGGTCAAGGCACCAAGGTCGAAATTAAACGTACG
    pETR11646 CAGGTGCAGCTGCAGCAGTCTGGCGCCGAGCTCGTGAAACCTGGCGCCTCC 224
    Mov19 VH- GTGAAGATCAGCTGCAAGGCCAGCGGCTACAGCTTCACCGGCTACTTCATG
    CH1-Fchole AACTGGGTCAAGCAGAGCCACGGCAAGAGCCTGGAATGGATCGGCAGAATC
    PG/LALA CACCCCTACGACGGCGACACCTTCTACAACCAGAACTTCAAGGACAAGGCC
    ACCCTGACCGTGGACAAGAGCAGCAACACCGCCCACATGGAACTGCTGAGC
    CTGACCAGCGAGGACTTCGCCGTGTACTACTGCACCAGATACGACGGCAGC
    CGGGCCATGGATTATTGGGGCCAGGGCACCACCGTGACAGTGTCCAGCGCT
    AGCACCAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACC
    AGCGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAG
    CCCGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACACC
    TTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTGGTC
    ACCGTGCCTTCTAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAAC
    CACAAGCCCAGCAACACCAAGGTGGACAAGAAGGTGGAGCCCAAGAGCTGC
    GACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGA
    CCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCC
    CGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCT
    GAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAG
    ACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTC
    CTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAG
    GTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCC
    AAAGGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGAT
    GAGCTGACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTAT
    CCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAAC
    TACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTG
    AGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCA
    TGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTC
    TCCCTGTCTCCGGGTAAA
    pETR11647 CAGGTGCAGCTGCAGCAGTCTGGCGCCGAGCTCGTGAAACCTGGCGCCTCC 225
    Mov19VH- GTGAAGATCAGCTGCAAGGCCAGCGGCTACAGCTTCACCGGCTACTTCATG
    CH1-CD3 AACTGGGTCAAGCAGAGCCACGGCAAGAGCCTGGAATGGATCGGCAGAATC
    VH-CL- CACCCCTACGACGGCGACACCTTCTACAACCAGAACTTCAAGGACAAGGCC
    Fcknob ACCCTGACCGTGGACAAGAGCAGCAACACCGCCCACATGGAACTGCTGAGC
    PG/LALA CTGACCAGCGAGGACTTCGCCGTGTACTACTGCACCAGATACGACGGCAGC
    CGGGCCATGGATTATTGGGGCCAGGGCACCACCGTGACAGTGTCCAGCGCT
    AGCACAAAGGGCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACA
    TCTGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAG
    CCTGTGACCGTGTCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACACC
    TTTCCAGCCGTGCTGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTGGTC
    ACCGTGCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAAC
    CACAAGCCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGT
    GATGGCGGAGGAGGGTCCGGAGGCGGAGGATCCGAAGTGCAGCTGGTGGAA
    AGCGGCGGAGGCCTGGTGCAGCCTAAGGGCTCTCTGAAGCTGAGCTGTGCC
    GCCAGCGGCTTCACCTTCAACACCTACGCCATGAACTGGGTGCGCCAGGCC
    CCTGGCAAAGGCCTGGAATGGGTGGCCCGGATCAGAAGCAAGTACAACAAT
    TACGCCACCTACTACGCCGACAGCGTGAAGGACCGGTTCACCATCAGCCGG
    GACGACAGCCAGAGCATCCTGTACCTGCAGATGAACAACCTGAAAACCGAG
    GACACCGCCATGTACTACTGCGTGCGGCACGGCAACTTCGGCAACAGCTAT
    GTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCTGCT
    GCTAGCGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCATCCGATGAACAG
    CTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGCTGAACAATTTTTACCCT
    AGGGAAGCTAAAGTGCAGTGGAAAGTGGATAACGCACTGCAGTCCGGCAAC
    TCCCAGGAATCTGTGACAGAACAGGACTCCAAGGACAGCACCTACTCCCTG
    TCCTCCACCCTGACACTGTCTAAGGCTGATTATGAGAAACACAAAGTCTAC
    GCCTGCGAAGTCACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTC
    AACAGGGGAGAGTGTGACAAGACCCACACCTGTCCCCCTTGTCCTGCCCCT
    GAAGCTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAGCCCAAGGAC
    ACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTGGTGGATGTG
    TCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACGTGGACGGCGTGGAA
    GTGCACAACGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTAC
    CGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAG
    GAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAA
    ACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTG
    CCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTG
    GTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGG
    CAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC
    TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAG
    GGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTAC
    ACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    pETR11644 GACATCGAGCTGACCCAGAGCCCTGCCTCTCTGGCCGTGTCTCTGGGACAG 226
    Mov19 LC AGAGCCATCATCAGCTGCAAGGCCAGCCAGAGCGTGTCCTTTGCCGGCACC
    TCTCTGATGCACTGGTATCACCAGAAGCCCGGCCAGCAGCCCAAGCTGCTG
    ATCTACAGAGCCAGCAACCTGGAAGCCGGCGTGCCCACAAGATTTTCCGGC
    AGCGGCAGCAAGACCGACTTCACCCTGAACATCCACCCCGTGGAAGAAGAG
    GACGCCGCCACCTACTACTGCCAGCAGAGCAGAGAGTACCCCTACACCTTC
    GGCGGAGGCACCAAGCTGGAAATCAAGCGTACGGTGGCTGCACCATCTGTC
    TTCATCTTCCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTT
    GTGTGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGAAG
    GTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCACAGAGCAG
    GACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAA
    GCAGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGC
    CTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGT
    Seq
    ID
    Variant Sequence No
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 261
    VH_D52dE TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG
    CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG
    TCTAAAACTGAGGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT
    TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC
    TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG
    TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 262
    VH_D52dQ TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG
    CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG
    TCTAAAACTCAGGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT
    TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC
    TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG
    TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC
    CD3_VH GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTC 263
    N100A TGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAA
    CTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGA
    AGCAAGTACAACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGT
    TCACCATCAGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAG
    CCTGCGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTC
    GGCGCCAGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGA
    CCGTGTCAAGC
    CD3_VH GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTC 264
    S100aA TGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAA
    CTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGA
    AGCAAGTACAACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGT
    TCACCATCAGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAG
    CCTGCGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTC
    GGCAACGCCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGA
    CCGTGTCAAGC
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 265
    [VHCH1]- TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG
    CD3[VHCH1- CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG
    N100A]- TCTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT
    Fcknob_PGLALA TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC
    TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG
    TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTA
    GCACAAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACAAG
    CGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCC
    GTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGGCGTGCACACTTTCC
    CTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTCACCGT
    GCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAG
    CCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCG
    GAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGG
    CGGACTGGTGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGC
    TTCACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAG
    GCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTA
    CTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAG
    AACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGT
    ACTATTGTGTGCGGCACGGCAACTTCGGCGCCAGCTATGTGTCTTGGTTTGC
    CTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGC
    CCCAGCGTGTTCCCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAG
    CCGCTCTGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC
    TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCCGTGCTG
    CAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTGCCCTCTAGCT
    CCCTGGGAACACAGACATATATCTGTAATGTCAATCACAAGCCTTCCAACAC
    CAAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGCGACAAAACTCACACATGC
    CCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCC
    CCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATG
    CGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTAC
    GTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGT
    ACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTG
    GCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCC
    CCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGG
    TGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCT
    GTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAG
    AGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACT
    CCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTG
    GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAAC
    CACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    16D5- GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 266
    Fchole- TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG
    PGLALA CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG
    TCTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT
    TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC
    TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG
    TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTA
    GCACCAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAG
    CGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCC
    GTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACACCTTCC
    CCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTGGTCACCGT
    GCCTTCTAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAG
    CCCAGCAACACCAAGGTGGACAAGAAGGTGGAGCCCAAGAGCTGCGACAAAA
    CTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGT
    CTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCT
    GAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGT
    TCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCG
    GGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTG
    CACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAG
    CCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCG
    AGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAAC
    CAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATCGCCG
    TGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCC
    CGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGAC
    AAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGG
    CTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    CD3-CLC CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACCG 267
    TGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTACGC
    CAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGCGGC
    ACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTGCTGG
    GAGGAAAGGCCGCCCTGACACTGTCTGGCGCCCAGCCAGAAGATGAGGCCGA
    GTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACC
    AAGCTGACAGTCCTAGGTCAACCCAAGGCTGCCCCCAGCGTGACCCTGTTCC
    CCCCCAGCAGCGAGGAACTGCAGGCCAACAAGGCCACCCTGGTCTGCCTGAT
    CAGCGACTTCTACCCAGGCGCCGTGACCGTGGCCTGGAAGGCCGACAGCAGC
    CCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAGCAAGCAGAGCAACAACA
    AGTACGCCGCCAGCAGCTACCTGAGCCTGACCCCCGAGCAGTGGAAGAGCCA
    CAGGTCCTACAGCTGCCAGGTGACCCACGAGGGCAGCACCGTGGAGAAAACC
    GTGGCCCCCACCGAGTGCAGC
    16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 268
    [VHCH1]- TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG
    CD3[VHCH1- CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG
    S100aA]- TCTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT
    Fcknob_PGLALA TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC
    TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG
    TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTA
    GCACAAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACAAG
    CGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCC
    GTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGGCGTGCACACTTTCC
    CTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTCACCGT
    GCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAG
    CCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCG
    GAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGG
    CGGACTGGTGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGC
    TTCACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAG
    GCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTA
    CTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAG
    AACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGT
    ACTATTGTGTGCGGCACGGCAACTTCGGCAACGCCTATGTGTCTTGGTTTGC
    CTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGC
    CCCAGCGTGTTCCCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAG
    CCGCTCTGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC
    TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCCGTGCTG
    CAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTGCCCTCTAGCT
    CCCTGGGAACACAGACATATATCTGTAATGTCAATCACAAGCCTTCCAACAC
    CAAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGCGACAAAACTCACACATGC
    CCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCC
    CCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATG
    CGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTAC
    GTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGT
    ACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTG
    GCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCC
    CCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGG
    TGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCT
    GTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAG
    AGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACT
    CCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTG
    GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAAC
    CACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    9D11 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCG 269
    [VHCH1]- TTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCA
    CD3[VHCL- CTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAAC
    N100A]- CCAAGCGGTGGCCCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGA
    Fcknob_PGLALA TGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCG
    TTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTG
    GACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACAAAGG
    GCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACATCTGGCGGAAC
    AGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAGCCTGTGACCGTG
    TCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACACCTTTCCAGCCGTGC
    TGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTGGTCACCGTGCCTAGCAG
    CAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAAC
    ACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGT
    CCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGT
    GCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTC
    AGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAAT
    GGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACTACGCCGA
    CAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAGAACACCCTG
    TACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGTG
    TGCGGCACGGCAACTTCGGCGCCAGCTATGTGTCTTGGTTTGCCTACTGGGG
    CCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTGTGGCCGCTCCCTCCGTG
    TTTATCTTTCCCCCATCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCG
    TGTGTCTGCTGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGT
    GGATAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAGGAC
    TCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGTCTAAGGCTG
    ATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAG
    CTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGTGACAAGACCCACACC
    TGTCCCCCTTGTCCTGCCCCTGAAGCTGCTGGCGGCCCTTCTGTGTTCCTGT
    TCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCCGAAGTGAC
    CTGCGTGGTGGTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGG
    TACGTGGACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGC
    AGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGA
    CTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGC
    GCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCAC
    AGGTGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAG
    CCTGTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGG
    GAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGG
    ACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAG
    GTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC
    AACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    9D11- CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCG 270
    Fchole TTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCA
    CTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAAC
    CCAAGCGGTGGCCCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGA
    TGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCG
    TTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTG
    GACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACCAAGG
    GCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGCAC
    AGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCCGTGACCGTG
    TCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACACCTTCCCCGCCGTGC
    TGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTGGTCACCGTGCCTTCTAG
    CAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAAC
    ACCAAGGTGGACAAGAAGGTGGAGCCCAAGAGCTGCGACAAAACTCACACAT
    GCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTT
    CCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACA
    TGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGT
    ACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA
    GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGAC
    TGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCG
    CCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACA
    GGTGTGCACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGC
    CTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGG
    AGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGA
    CTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGACAAGAGCAGG
    TGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACA
    ACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    9D11_LC GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAAC 271
    [N95Q] CGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGCTA
    CAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTGCTG
    ATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGCGGCT
    CTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCAGAAGA
    CGTGGGCGTTTATTACTGTATGCAGGCAAGCATTATGCAGCGGACTTTTGGT
    CAAGGCACCAAGGTCGAAATTAAACGTACGGTGGCTGCACCATCTGTCTTCA
    TCTTCCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTGTG
    CCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGAAGGTGGAT
    AACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCACAGAGCAGGACAGCA
    AGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAAGCAGACTA
    CGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAGCTCG
    CCCGTCACAAAGAGCTTCAACAGGGGAGAGTGT
    CD3_VLCH1 CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACCG 272
    TGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTACGC
    CAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGCGGC
    ACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTGCTGG
    GAGGAAAGGCCGCCCTGACACTGTCTGGCGCCCAGCCAGAAGATGAGGCCGA
    GTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACC
    AAGCTGACAGTGCTGAGCAGCGCTTCCACCAAAGGCCCTTCCGTGTTTCCTC
    TGGCTCCTAGCTCCAAGTCCACCTCTGGAGGCACCGCTGCTCTCGGATGCCT
    CGTGAAGGATTATTTTCCTGAGCCTGTGACAGTGTCCTGGAATAGCGGAGCA
    CTGACCTCTGGAGTGCATACTTTCCCCGCTGTGCTGCAGTCCTCTGGACTGT
    ACAGCCTGAGCAGCGTGGTGACAGTGCCCAGCAGCAGCCTGGGCACCCAGAC
    CTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAGAAG
    GTGGAACCCAAGTCTTGT
    9D11 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCG 273
    [VHCH1]- TTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCA
    CD3[VHCH1- CTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAAC
    S100aA]- CCAAGCGGTGGCCCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGA
    Fcknob_PGLALA TGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCG
    TTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTG
    GACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACAAAGG
    GCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACATCTGGCGGAAC
    AGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAGCCTGTGACCGTG
    TCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACACCTTTCCAGCCGTGC
    TGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTGGTCACCGTGCCTAGCAG
    CAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAAC
    ACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGT
    CCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGT
    GCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTC
    AGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAAT
    GGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACTACGCCGA
    CAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAGAACACCCTG
    TACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGTG
    TGCGGCACGGCAACTTCGGCAACGCCTATGTGTCTTGGTTTGCCTACTGGGG
    CCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTGTGGCCGCTCCCTCCGTG
    TTTATCTTTCCCCCATCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCG
    TGTGTCTGCTGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGT
    GGATAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAGGAC
    TCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGTCTAAGGCTG
    ATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAG
    CTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGTGACAAGACCCACACC
    TGTCCCCCTTGTCCTGCCCCTGAAGCTGCTGGCGGCCCTTCTGTGTTCCTGT
    TCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCCGAAGTGAC
    CTGCGTGGTGGTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGG
    TACGTGGACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGC
    AGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGA
    CTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGC
    GCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCAC
    AGGTGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAG
    CCTGTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGG
    GAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGG
    ACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAG
    GTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC
    AACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    Seq
    ID
    Name Sequence No
    16D5 GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCTCTG 287
    variant AGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATGAGCTGG
    W96Y/D52E GTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATCAAGAGCAAG
    VH ACCGAGGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGCCGGTTCACCATC
    AGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGAAAACC
    GAGGACACCGCCGTGTACTACTGCACCACCCCCTACGAGTGGTCTTGGTACGAC
    TACTGGGGCCAGGGCACCCTCGTGACCGTGTCATCT
    W96Y/D52E_CD3- GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCTCTG 288
    VHCH1_Fc- AGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATGAGCTGG
    knob_PGLALA GTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATCAAGAGCAAG
    pETR14945 ACCGAGGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGCCGGTTCACCATC
    AGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGAAAACC
    GAGGACACCGCCGTGTACTACTGCACCACCCCCTACGAGTGGTCTTGGTACGAC
    TACTGGGGCCAGGGCACCCTCGTGACCGTGTCATCTGCTAGCACAAAGGGCCCT
    AGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACAAGCGGCGGAACAGCCGCC
    CTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCTTGGAAC
    AGCGGAGCCCTGACAAGCGGCGTGCACACCTTCCCTGCCGTGCTGCAGAGCAGC
    GGCCTGTACTCCCTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACC
    CAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGACAAG
    AAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGAGGCGGAGGATCC
    GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTCTG
    AGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAACTGG
    GTGCGCCAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAG
    TACAACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATC
    AGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCC
    GAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCTAT
    GTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCT
    AGTACCAAGGGCCCCAGCGTGTTCCCCCTGGCACCCAGCAGCAAGAGCACATCT
    GGCGGAACAGCCGCTCTGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTG
    ACCGTGTCTTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC
    GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTGCCCTCT
    AGCTCCCTGGGAACACAGACATATATCTGTAATGTCAATCACAAGCCTTCCAAC
    ACCAAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGCGACAAAACTCACACATGC
    CCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCCCC
    CCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTG
    GTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGAC
    GGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGC
    ACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGC
    AAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAA
    ACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCC
    CCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAA
    GGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAG
    AACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTC
    TACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCA
    TGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCC
    CTGTCTCCGGGTAAA
    W96Y/D52E_Fc- GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCTCTG 289
    hole_PGLALA_HYRF AGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATGAGCTGG
    pETR14946 GTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATCAAGAGCAAG
    ACCGAGGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGCCGGTTCACCATC
    AGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGAAAACC
    GAGGACACCGCCGTGTACTACTGCACCACCCCCTACGAGTGGTCTTGGTACGAC
    TACTGGGGCCAGGGCACCCTCGTGACCGTGTCATCTGCTAGCACCAAGGGCCCC
    TCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGCACAGCCGCT
    CTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCCGTGACCGTGTCCTGGAAC
    AGCGGAGCCCTGACCTCCGGCGTGCACACCTTCCCCGCCGTGCTGCAGAGTTCT
    GGCCTGTATAGCCTGAGCAGCGTGGTCACCGTGCCTTCTAGCAGCCTGGGCACC
    CAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAG
    AAGGTGGAGCCCAAGAGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCA
    CCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGAC
    ACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGC
    CACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCAT
    AATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTC
    AGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGC
    AAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCC
    AAAGGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAG
    CTGACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGC
    GACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACC
    ACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACC
    GTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCAT
    GAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    14B1 GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCCCTG 290
    VH AGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAGTTATGCCATGAGCTGG
    GTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCAGCTATTAGTGGTAGT
    GGTGGTAGCACATACTACGCAGACTCCGTGAAGGGCCGGTTCACCATCTCCAGA
    GACAATTCCAAGAACACGCTGTATCTGCAGATGAACAGCCTGAGAGCCGAGGAC
    ACGGCCGTATATTACTGTGCGCGTGGTGACTACCGTTACCGTTACTTCGACTAC
    TGGGGCCAAGGAACCCTGGTCACCGTCTCGAGT
    14B1 TCTTCTGAACTGACTCAAGATCCAGCTGTTAGCGTGGCTCTGGGTCAGACTGTA 291
    VL CGTATCACCTGCCAAGGCGATTCTCTGCGCTCCTACTACGCAAGCTGGTACCAG
    CAGAAACCGGGTCAGGCCCCAGTTCTGGTGATTTACGGCAAAAACAACCGTCCG
    TCTGGGATCCCGGACCGTTTCTCCGGCAGCTCTTCCGGTAACACGGCGAGCCTC
    ACCATCACTGGCGCTCAAGCAGAAGACGAGGCCGACTATTACTGTAACTCTCGG
    GAAAGCCCACCAACCGGCCTGGTTGTCTTCGGTGGCGGTACCAAGCTGACCGTC
    CTA
    14B1[EE]_CD3[VLCH1]_Fc- GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCCCTG 292
    knob_PGLALA AGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAGTTATGCCATGAGCTGG
    pETR14976 GTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCAGCTATTAGTGGTAGT
    GGTGGTAGCACATACTACGCAGACTCCGTGAAGGGCCGGTTCACCATCTCCAGA
    GACAATTCCAAGAACACGCTGTATCTGCAGATGAACAGCCTGAGAGCCGAGGAC
    ACGGCCGTATATTACTGTGCGCGTGGTGACTACCGTTACCGTTACTTCGACTAC
    TGGGGCCAAGGAACCCTGGTCACCGTCTCGAGTGCTAGCACCAAGGGCCCCTCC
    GTGTTTCCTCTGGCCCCTTCCAGCAAGTCCACCTCTGGCGGAACTGCCGCTCTG
    GGCTGCCTGGTGGAAGATTACTTCCCCGAGCCCGTGACCGTGTCCTGGAATTCT
    GGCGCTCTGACCTCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGC
    CTGTACTCCCTGTCCTCCGTCGTGACAGTGCCCTCCAGCTCTCTGGGCACCCAG
    ACCTACATCTGCAACGTGAACCACAAGCCCTCCAACACCAAGGTGGACGAGAAG
    GTGGAACCCAAGTCCTGCGACGGTGGCGGAGGTTCCGGAGGCGGAGGATCCCAG
    GCTGTCGTGACCCAGGAACCCTCCCTGACAGTGTCTCCTGGCGGCACCGTGACC
    CTGACCTGTGGATCTTCTACCGGCGCTGTGACCACCTCCAACTACGCCAATTGG
    GTGCAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGCGGCACCAACAAG
    AGAGCCCCTGGCACCCCTGCCAGATTCTCCGGTTCTCTGCTGGGCGGCAAGGCT
    GCCCTGACTCTGTCTGGTGCTCAGCCTGAGGACGAGGCCGAGTACTACTGCGCC
    CTGTGGTACTCCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACCGTGCTG
    TCCAGCGCTTCCACCAAGGGACCCAGTGTGTTCCCCCTGGCCCCCAGCTCCAAG
    TCTACATCCGGTGGCACAGCTGCCCTGGGATGTCTCGTGAAGGACTACTTTCCT
    GAGCCTGTGACAGTGTCTTGGAACAGCGGAGCCCTGACCAGCGGAGTGCACACA
    TTCCCTGCAGTGCTGCAGAGCAGCGGCCTGTATAGCCTGAGCAGCGTCGTGACC
    GTGCCTTCCTCTAGCCTGGGAACACAGACATATATCTGTAATGTGAATCATAAG
    CCCAGTAATACCAAAGTGGATAAGAAAGTGGAACCTAAGAGCTGCGATAAGACC
    CACACCTGTCCCCCCTGCCCTGCTCCTGAAGCTGCTGGTGGCCCTAGCGTGTTC
    CTGTTCCCCCCAAAGCCCAAGGACACCCTGATGATCTCCCGGACCCCCGAAGTG
    ACCTGCGTGGTGGTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGG
    TACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAG
    TACAACTCCACCTACCGGGTGGTGTCCGTGCTGACAGTGCTGCACCAGGACTGG
    CTGAACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGGGCGCTCCC
    ATCGAAAAGACCATCTCCAAGGCCAAGGGCCAGCCCCGGGAACCCCAGGTGTAC
    ACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGC
    CTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGG
    CAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCC
    TTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAAC
    GTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAG
    AGCCTCTCCCTGTCTCCGGGTAAA
    14B1[EE]_Fc- GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCCCTG 293
    hole_PGLALA AGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAGTTATGCCATGAGCTGG
    pETR14977 GTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCAGCTATTAGTGGTAGT
    GGTGGTAGCACATACTACGCAGACTCCGTGAAGGGCCGGTTCACCATCTCCAGA
    GACAATTCCAAGAACACGCTGTATCTGCAGATGAACAGCCTGAGAGCCGAGGAC
    ACGGCCGTATATTACTGTGCGCGTGGTGACTACCGTTACCGTTACTTCGACTAC
    TGGGGCCAAGGAACCCTGGTCACCGTCTCGAGTGCTAGCACCAAGGGCCCCTCC
    GTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGCACAGCCGCTCTG
    GGCTGCCTGGTCGAGGACTACTTCCCCGAGCCCGTGACCGTGTCCTGGAACAGC
    GGAGCCCTGACCTCCGGCGTGCACACCTTCCCCGCCGTGCTGCAGAGTTCTGGC
    CTGTATAGCCTGAGCAGCGTGGTCACCGTGCCTTCTAGCAGCCTGGGCACCCAG
    ACCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACGAGAAG
    GTGGAGCCCAAGAGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCT
    GAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACC
    CTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCAC
    GAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAAT
    GCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGC
    GTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAG
    GTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAA
    GGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAGCTG
    ACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGCGAC
    ATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACG
    CCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTG
    GACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAG
    GCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    14B1 LC TCTTCTGAACTGACTCAAGATCCAGCTGTTAGCGTGGCTCTGGGTCAGACTGTA 294
    [KK] CGTATCACCTGCCAAGGCGATTCTCTGCGCTCCTACTACGCAAGCTGGTACCAG
    Constant CAGAAACCGGGTCAGGCCCCAGTTCTGGTGATTTACGGCAAAAACAACCGTCCG
    lambda TCTGGGATCCCGGACCGTTTCTCCGGCAGCTCTTCCGGTAACACGGCGAGCCTC
    pETR14979 ACCATCACTGGCGCTCAAGCAGAAGACGAGGCCGACTATTACTGTAACTCTCGG
    GAAAGCCCACCAACCGGCCTGGTTGTCTTCGGTGGCGGTACCAAGCTGACCGTC
    CTAGGTCAACCCAAGGCTGCCCCCAGCGTGACCCTGTTCCCCCCCAGCAGCAAG
    AAACTGCAGGCCAACAAGGCCACCCTGGTCTGCCTGATCAGCGACTTCTACCCA
    GGCGCCGTGACCGTGGCCTGGAAGGCCGACAGCAGCCCCGTGAAGGCCGGCGTG
    GAGACCACCACCCCCAGCAAGCAGAGCAACAACAAGTACGCCGCCAGCAGCTAC
    CTGAGCCTGACCCCCGAGCAGTGGAAGAGCCACAGGTCCTACAGCTGCCAGGTG
    ACCCACGAGGGCAGCACCGTGGAGAAAACCGTGGCCCCCACCGAGTGCAGC
    9C7 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCGTT 295
    AAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCACTGG
    GTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAACCCAAGC
    GGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGATGACCCGT
    GACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGAT
    ACTGCAGTGTACTACTGTGCACGCGGTGACTGGTCTTACTACATGGACTATTGG
    GGTCAAGGCACCCTCGTAACGGTTTCTTCT
    9C7 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAACCG 296
    GCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGCTACAAC
    TATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTGCTGATCTAC
    CTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGCGGCTCTGGATCC
    GGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTT
    TATTACTGTATGCAGGCACGGCAGACCCCAACTTTTGGTCAAGGCACCAAGGTC
    GAAATTAAA
    9C7[EE]_CD3[VLCH1]_Fc- CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCGTT 297
    knob_PGLALA AAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCACTGG
    pETR14974 GTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAACCCAAGC
    GGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGATGACCCGT
    GACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGAT
    ACTGCAGTGTACTACTGTGCACGCGGTGACTGGTCTTACTACATGGACTATTGG
    GGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACCAAGGGCCCCTCCGTG
    TTTCCTCTGGCCCCTTCCAGCAAGTCCACCTCTGGCGGAACTGCCGCTCTGGGC
    TGCCTGGTGGAAGATTACTTCCCCGAGCCCGTGACCGTGTCCTGGAATTCTGGC
    GCTCTGACCTCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTG
    TACTCCCTGTCCTCCGTCGTGACAGTGCCCTCCAGCTCTCTGGGCACCCAGACC
    TACATCTGCAACGTGAACCACAAGCCCTCCAACACCAAGGTGGACGAGAAGGTG
    GAACCCAAGTCCTGCGACGGTGGCGGAGGTTCCGGAGGCGGAGGATCCCAGGCT
    GTCGTGACCCAGGAACCCTCCCTGACAGTGTCTCCTGGCGGCACCGTGACCCTG
    ACCTGTGGATCTTCTACCGGCGCTGTGACCACCTCCAACTACGCCAATTGGGTG
    CAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGCGGCACCAACAAGAGA
    GCCCCTGGCACCCCTGCCAGATTCTCCGGTTCTCTGCTGGGCGGCAAGGCTGCC
    CTGACTCTGTCTGGTGCTCAGCCTGAGGACGAGGCCGAGTACTACTGCGCCCTG
    TGGTACTCCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACCGTGCTGTCC
    AGCGCTTCCACCAAGGGACCCAGTGTGTTCCCCCTGGCCCCCAGCTCCAAGTCT
    ACATCCGGTGGCACAGCTGCCCTGGGATGTCTCGTGAAGGACTACTTTCCTGAG
    CCTGTGACAGTGTCTTGGAACAGCGGAGCCCTGACCAGCGGAGTGCACACATTC
    CCTGCAGTGCTGCAGAGCAGCGGCCTGTATAGCCTGAGCAGCGTCGTGACCGTG
    CCTTCCTCTAGCCTGGGAACACAGACATATATCTGTAATGTGAATCATAAGCCC
    AGTAATACCAAAGTGGATAAGAAAGTGGAACCTAAGAGCTGCGATAAGACCCAC
    ACCTGTCCCCCCTGCCCTGCTCCTGAAGCTGCTGGTGGCCCTAGCGTGTTCCTG
    TTCCCCCCAAAGCCCAAGGACACCCTGATGATCTCCCGGACCCCCGAAGTGACC
    TGCGTGGTGGTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTAC
    GTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTAC
    AACTCCACCTACCGGGTGGTGTCCGTGCTGACAGTGCTGCACCAGGACTGGCTG
    AACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGGGCGCTCCCATC
    GAAAAGACCATCTCCAAGGCCAAGGGCCAGCCCCGGGAACCCCAGGTGTACACC
    CTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTG
    GTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAG
    CCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC
    TTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTC
    TTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGC
    CTCTCCCTGTCTCCGGGTAAA
    9C7[EE]_Fc- CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCGTT 298
    hole_PGLALA AAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCACTGG
    pETR14975 GTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAACCCAAGC
    GGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGATGACCCGT
    GACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGAT
    ACTGCAGTGTACTACTGTGCACGCGGTGACTGGTCTTACTACATGGACTATTGG
    GGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACCAAGGGCCCCTCCGTG
    TTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGCACAGCCGCTCTGGGC
    TGCCTGGTCGAGGACTACTTCCCCGAGCCCGTGACCGTGTCCTGGAACAGCGGA
    GCCCTGACCTCCGGCGTGCACACCTTCCCCGCCGTGCTGCAGAGTTCTGGCCTG
    TATAGCCTGAGCAGCGTGGTCACCGTGCCTTCTAGCAGCCTGGGCACCCAGACC
    TACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACGAGAAGGTG
    GAGCCCAAGAGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAA
    GCTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTC
    ATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAA
    GACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCC
    AAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTC
    CTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTC
    TCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGG
    CAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAGCTGACC
    AAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATC
    GCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCT
    CCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGAC
    AAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCT
    CTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
    9C7 LC GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAACCG 299
    [RK] GCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGCTACAAC
    pETR14980 TATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTGCTGATCTAC
    CTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGCGGCTCTGGATCC
    GGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTT
    TATTACTGTATGCAGGCACGGCAGACCCCAACTTTTGGTCAAGGCACCAAGGTC
    GAAATTAAACGTACGGTGGCTGCACCATCTGTCTTCATCTTCCCGCCATCTGAT
    CGGAAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTAT
    CCCAGAGAGGCCAAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCGGGTAAC
    TCCCAGGAGAGTGTCACAGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGC
    AGCACCCTGACGCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGC
    GAAGTCACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGA
    GAGTGT
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.

Claims (39)

1-123. (canceled)
124. An antigen-binding molecule comprising a Folate Receptor 1 (FolR1) antigen-binding moiety that binds to FolR1, wherein the FolR1 antigen-binding moiety comprises at least one heavy chain complementarity determining region (CDR) comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 18 and at least one light chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34.
125. The antigen-binding molecule of claim 124, wherein the FolR1 antigen-binding moiety comprises:
(i) a complementarity-determining region heavy chain 1 (CDR-H1) comprising the amino acid sequence of SEQ ID NO: 16;
(ii) a complementarity-determining region heavy chain 2 (CDR-H2) comprising the amino acid sequence of SEQ ID NO: 17;
(iii) a complementarity-determining region heavy chain 3 (CDR-H3) comprising the amino acid sequence of SEQ ID NO: 18;
(iv) a complementarity-determining region light chain 1 (CDR-L1) comprising the amino acid sequence of SEQ ID NO: 32;
(v) a complementarity-determining region light chain 2 (CDR-L2) comprising the amino acid sequence of SEQ ID NO: 33; and
(vi) a complementarity-determining region light chain 3 (CDR-L3) comprising the amino acid sequence of SEQ ID NO: 34.
126. The antigen-binding molecule of claim 125, wherein the FolR1 antigen-binding moiety comprises a variable heavy chain comprising the amino acid sequence of SEQ ID NO: 15 and a variable light chain comprising the amino acid sequence of SEQ ID NO: 31.
127. An antigen-binding molecule comprising a FolR1 antigen-binding moiety that binds to FolR1, wherein the FolR1 antigen-binding moiety comprises at least one heavy chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 56, and SEQ ID NO: 57 and at least one light chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 59, SEQ ID NO: 60, and SEQ ID NO: 65.
128. The antigen-binding molecule of claim 127, wherein the FolR1 antigen-binding moiety comprises:
(i) a CDR-H1 comprising the amino acid sequence of SEQ ID NO: 8;
(ii) a CDR-H2 comprising the amino acid sequence of SEQ ID NO: 56;
(iii) a CDR-H3 comprising the amino acid sequence of SEQ ID NO: 57;
(iv) a CDR-L1 comprising the amino acid sequence of SEQ ID NO: 59;
(v) a CDR-L2 comprising the amino acid sequence of SEQ ID NO: 60; and
(vi) a CDR-L3 comprising the amino acid sequence of SEQ ID NO: 65.
129. The antigen-binding molecule of claim 128, wherein the FolR1 antigen-binding moiety comprises a variable heavy chain comprising the amino acid sequence of SEQ ID NO: 55 and a variable light chain comprising the amino acid sequence of SEQ ID NO: 64.
130. An antigen-binding molecule comprising a FolR1 antigen-binding moiety that binds to FolR1, wherein the antigen-binding moiety comprises at least one heavy chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 275, and SEQ ID NO: 315 and at least one light chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34.
131. The antigen-binding molecule of claim 130, wherein the FolR1 antigen-binding moiety comprises:
(i) a CDR-H1 comprising the amino acid sequence of SEQ ID NO: 16;
(ii) a CDR-H2 comprising the amino acid sequence of SEQ ID NO: 275;
(iii) a CDR-H3 comprising the amino acid sequence of SEQ ID NO: 315;
(iv) a CDR-L1 comprising the amino acid sequence of SEQ ID NO: 32;
(v) a CDR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and
(vi) a CDR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
132. The antigen-binding molecule of claim 131, wherein the FolR1 antigen-binding moiety comprises a variable heavy chain comprising the amino acid sequence of SEQ ID NO: 274 and a variable light chain comprising the amino acid sequence of SEQ ID NO: 31.
133. An antigen-binding molecule comprising a FolR1 antigen-binding moiety that binds to FolR1, wherein the antigen-binding moiety comprises at least one heavy chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, and SEQ ID NO: 50 and at least one light chain CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 52, SEQ ID NO: 53, and SEQ ID NO: 54.
134. The antigen-binding molecule of claim 133, wherein the FolR1 antigen-binding moiety comprises:
(i) a CDR-H1 comprising the amino acid sequence of SEQ ID NO: 8;
(ii) a CDR-H2 comprising the amino acid sequence of SEQ ID NO: 9;
(iii) a CDR-H3 comprising the amino acid sequence of SEQ ID NO: 50;
(iv) a CDR-L1 comprising the amino acid sequence of SEQ ID NO: 52;
(v) a CDR-L2 comprising the amino acid sequence of SEQ ID NO: 53; and
(vi) a CDR-L3 comprising the amino acid sequence of SEQ ID NO: 54.
135. The antigen-binding molecule of claim 134, wherein the FolR1 antigen-binding moiety comprises a variable heavy chain comprising the amino acid sequence of SEQ ID NO: 49 and a variable light chain comprising the amino acid sequence of SEQ ID NO: 51.
136. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the FolR1 antigen-binding moiety binds to human FolR1 and cynomolgus monkey FolR1.
137. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the FolR1 antigen-binding moiety binds to FolR1 expressed on a human tumor cell.
138. The antigen-binding molecule of claim 137, wherein the FolR1 antigen-binding moiety binds to a conformational epitope of human FolR1 expressed on the human tumor cell.
139. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the FolR1 antigen-binding moiety binds to a FolR1 polypeptide comprising amino acids 25 to 234 of human FolR1 (SEQ ID NO: 227).
140. The antigen-binding molecule of claim 139, wherein the FolR1 antigen-binding moiety:
(a) binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO: 230;
(b) binds to a FolR1 polypeptide comprising the amino acid sequence of SEQ ID NO: 231;
(c) does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NO: 228; and/or
(d) does not bind to a FolR polypeptide comprising the amino acid sequence of SEQ ID NO: 229.
141. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the FolR1 antigen-binding moiety does not bind to human Folate Receptor 2 (FolR2) or to human Folate Receptor 3 (FolR3).
142. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the FolR1 antigen-binding moiety binds to human FolR1 with a monovalent binding KD of at least about 1,000 nM.
143. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the antigen-binding molecule additionally comprises an Fc domain composed of a first subunit and a second subunit capable of stable association.
144. The antigen-binding molecule of claim 143, wherein the Fc domain is an IgG class immunoglobulin Fc domain.
145. The antigen-binding molecule of claim 144, wherein the antigen-binding molecule is bivalent for FolR1.
146. The antigen-binding molecule of any one of claims 124, 127, 130, and 133, wherein the antigen-binding molecule is a bispecific antigen-binding molecule.
147. The antigen-binding molecule of claim 146, wherein the bispecific antigen-binding molecule is a T cell activating bispecific antigen-binding molecule.
148. The antigen-binding molecule of claim 147, wherein the T cell activating bispecific antigen-binding molecule further comprises a CD3 antigen-binding moiety that binds to CD3.
149. The antigen-binding molecule of claim 148, wherein the FolR1 antigen-binding moiety and the CD3 antigen-binding moiety share a common light chain.
150. The antigen-binding molecule of claim 149, wherein the common light chain comprises the amino acid sequence of SEQ ID NO: 35.
151. The antigen-binding molecule of claim 148, wherein the CD3 antigen-binding moiety is a crossover Fab molecule, wherein either the variable or the constant regions of the Fab light chain and the Fab heavy chain are exchanged.
152. An affinity variant of the antigen-binding molecule of claim 147, wherein the affinity variant has a lower affinity to FolR1 as compared to the parental T cell activating bispecific antigen-binding molecule, and wherein the affinity variant is capable of inducing T cell-mediated lysis of a FolR1+ cancer cell.
153. An isolated polynucleotide encoding the antigen-binding molecule of any one of claims 124, 127, 130, and 133.
154. An expression vector comprising the isolated polynucleotide of claim 153.
155. A host cell comprising the isolated polynucleotide of claim 153.
156. A method of producing an antigen-binding molecule capable of specific binding to FolR1, the method comprising the steps of (a) culturing a host cell comprising an isolated polynucleotide encoding the antigen-binding molecule of any one of claims 124, 127, 130, and 133 under conditions suitable for the expression of the antigen-binding molecule and (b) recovering the antigen-binding molecule.
157. An antigen-binding molecule produced by the method of claim 156.
158. A pharmaceutical composition comprising the antigen-binding molecule of any one of claims 124, 127, 130, and 133 and a pharmaceutical acceptable carrier.
159. A method of treating a disease in an individual, wherein the method comprises administering to said individual a therapeutically effective amount of a composition comprising the antigen-binding molecule of any one of claims 124, 127, 130, and 133 in a pharmaceutically acceptable form.
160. The method of claim 159, wherein the disease is cancer.
161. A method of inducing lysis of a FolR1+ target cell, comprising contacting a target cell with the antigen-binding molecule of claim 148 in the presence of a CD3+ T cell.
US16/016,209 2014-11-20 2018-06-22 T cell activating bispecific antigen binding molecules Abandoned US20190031784A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/016,209 US20190031784A1 (en) 2014-11-20 2018-06-22 T cell activating bispecific antigen binding molecules
US17/580,248 US20220220224A1 (en) 2014-11-20 2022-01-20 T cell activating bispecific antigen binding molecules

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP14194147 2014-11-20
EP14194147.6 2014-11-20
US14/947,657 US20160208019A1 (en) 2014-11-20 2015-11-20 T cell activating bispecific antigen binding molecules
US16/016,209 US20190031784A1 (en) 2014-11-20 2018-06-22 T cell activating bispecific antigen binding molecules

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/947,657 Continuation US20160208019A1 (en) 2014-11-20 2015-11-20 T cell activating bispecific antigen binding molecules

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/580,248 Division US20220220224A1 (en) 2014-11-20 2022-01-20 T cell activating bispecific antigen binding molecules

Publications (1)

Publication Number Publication Date
US20190031784A1 true US20190031784A1 (en) 2019-01-31

Family

ID=52100988

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/947,657 Abandoned US20160208019A1 (en) 2014-11-20 2015-11-20 T cell activating bispecific antigen binding molecules
US16/016,209 Abandoned US20190031784A1 (en) 2014-11-20 2018-06-22 T cell activating bispecific antigen binding molecules
US17/580,248 Pending US20220220224A1 (en) 2014-11-20 2022-01-20 T cell activating bispecific antigen binding molecules

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/947,657 Abandoned US20160208019A1 (en) 2014-11-20 2015-11-20 T cell activating bispecific antigen binding molecules

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/580,248 Pending US20220220224A1 (en) 2014-11-20 2022-01-20 T cell activating bispecific antigen binding molecules

Country Status (33)

Country Link
US (3) US20160208019A1 (en)
EP (1) EP3221356B1 (en)
JP (2) JP7146395B2 (en)
KR (1) KR102648966B1 (en)
CN (1) CN107074955B (en)
AR (1) AR102732A1 (en)
AU (1) AU2015348595B2 (en)
BR (1) BR112017007086A2 (en)
CA (1) CA2960929A1 (en)
CL (1) CL2017000954A1 (en)
CO (1) CO2017003048A2 (en)
CR (1) CR20170203A (en)
DK (1) DK3221356T3 (en)
EA (1) EA201791121A1 (en)
ES (1) ES2829829T3 (en)
HR (1) HRP20201747T1 (en)
HU (1) HUE051715T2 (en)
IL (1) IL251292A0 (en)
LT (1) LT3221356T (en)
MA (1) MA40972B1 (en)
MX (1) MX2017006571A (en)
MY (1) MY184889A (en)
PE (1) PE20170585A1 (en)
PH (1) PH12017500939A1 (en)
PL (1) PL3221356T3 (en)
PT (1) PT3221356T (en)
RS (1) RS61010B1 (en)
SG (1) SG11201702976TA (en)
SI (1) SI3221356T1 (en)
TW (2) TWI756164B (en)
UA (1) UA122676C2 (en)
WO (1) WO2016079076A1 (en)
ZA (1) ZA201701843B (en)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
ES2549638T3 (en) 2011-02-28 2015-10-30 F. Hoffmann-La Roche Ag Antigen binding proteins
CN103403025B (en) 2011-02-28 2016-10-12 弗·哈夫曼-拉罗切有限公司 Monovalent antigen binding protein
KR101870555B1 (en) 2011-08-23 2018-06-22 로슈 글리카트 아게 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
WO2014056783A1 (en) 2012-10-08 2014-04-17 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
RU2015140915A (en) 2013-02-26 2017-04-03 Роше Гликарт Аг BSPECIFIC ANTI-BINDING MOLECULES ACTIVATING T-CELLS
MY192312A (en) 2013-02-26 2022-08-17 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
MX2016003593A (en) * 2013-10-11 2016-06-02 Hoffmann La Roche Multispecific domain exchanged common variable light chain antibodies.
PE20170263A1 (en) 2014-08-04 2017-03-30 Hoffmann La Roche T-CELL ACTIVATING ANTIGEN-BINDING BI-SPECIFIC MOLECULES
BR112017010324A2 (en) 2014-11-20 2018-05-15 F. Hoffmann-La Roche Ag method for treating or slowing cancer progression in an individual, molecules, methods for enhancing immune function in an individual and for selecting a patient for treatment, kits, pharmaceutical composition and uses of a combination of one molecule
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
CN107849137B (en) 2015-10-02 2021-11-26 豪夫迈·罗氏有限公司 Bispecific anti-CEAXCD 3T cell activating antigen binding molecules
JP7325186B2 (en) 2015-12-09 2023-08-14 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Type II anti-CD20 antibody for reducing the formation of anti-drug antibodies
MX2018008347A (en) 2016-01-08 2018-12-06 Hoffmann La Roche Methods of treating cea-positive cancers using pd-1 axis binding antagonists and anti-cea/anti-cd3 bispecific antibodies.
FI3433280T3 (en) 2016-03-22 2023-06-06 Hoffmann La Roche Protease-activated t cell bispecific molecules
EP3519437B1 (en) * 2016-09-30 2021-09-08 F. Hoffmann-La Roche AG Bispecific antibodies against p95her2
AU2017355401A1 (en) * 2016-11-02 2019-05-02 Jounce Therapeutics, Inc. Antibodies to PD-1 and uses thereof
WO2018108759A1 (en) * 2016-12-13 2018-06-21 F. Hoffmann-La Roche Ag Method to determine the presence of a target antigen in a tumor sample
CN110087682B (en) * 2016-12-19 2023-12-15 豪夫迈·罗氏有限公司 Combination therapy with targeted 4-1BB (CD 137) agonists
KR102346336B1 (en) 2017-04-05 2022-01-04 에프. 호프만-라 로슈 아게 Bispecific antibodies that specifically bind to PD1 and LAG3
JP2020527332A (en) * 2017-06-12 2020-09-10 ブルーフィン バイオメディシン, インコーポレイテッド Anti-IL1RAP antibody and antibody drug conjugate
CA3069842A1 (en) * 2017-07-14 2019-01-17 Immatics Biotechnologies Gmbh Improved dual specificity polypeptide molecule
CA3079036A1 (en) 2017-11-01 2019-05-09 F. Hoffmann-La Roche Ag Combination therapy with targeted ox40 agonists
EP3502140A1 (en) 2017-12-21 2019-06-26 F. Hoffmann-La Roche AG Combination therapy of tumor targeted icos agonists with t-cell bispecific molecules
KR20200104364A (en) * 2017-12-27 2020-09-03 테네오바이오, 인코포레이티드 CD3-delta/epsilon heterodimer specific antibody
AU2019218959A1 (en) 2018-02-08 2020-09-03 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
CN115925952A (en) * 2018-03-13 2023-04-07 东莞凡恩世生物医药有限公司 Anti-folate receptor 1 antibodies and uses thereof
CN112041336A (en) * 2018-03-13 2020-12-04 脉管生物生长有限公司 Bispecific antibodies against MOSPD2 and T cell or NK cell specific molecules
JP2021531825A (en) * 2018-07-09 2021-11-25 マルチチュード インコーポレーテッドMultitude Inc. Antibodies specific for folic acid receptor alpha
EP3830129A4 (en) * 2018-07-30 2022-05-04 Invenra, Inc. Multispecific treg binding molecules
JP6935383B2 (en) * 2018-09-27 2021-09-15 公益財団法人実験動物中央研究所 Immunodeficient mouse
CN116121302A (en) * 2018-09-27 2023-05-16 公益财团法人实验动物中央研究所 Immunodeficient mice
MY198034A (en) 2018-12-21 2023-07-27 Hoffmann La Roche Tumor-targeted agonistic cd28 antigen binding molecules
TWI829831B (en) 2018-12-21 2024-01-21 瑞士商赫孚孟拉羅股份公司 Antibodies binding to cd3
EP4023230A4 (en) * 2019-06-05 2023-11-15 Chugai Seiyaku Kabushiki Kaisha Antibody cleavage site-binding molecule
EP3986924A1 (en) 2019-06-19 2022-04-27 F. Hoffmann-La Roche AG Method for the generation of a multivalent, multispecific antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
MX2021015540A (en) * 2019-06-19 2022-02-10 Hoffmann La Roche Method for the generation of a trivalent antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization.
WO2020260326A1 (en) 2019-06-27 2020-12-30 F. Hoffmann-La Roche Ag Novel icos antibodies and tumor-targeted antigen binding molecules comprising them
JP2022540611A (en) * 2019-07-11 2022-09-16 メモリアル スローン ケタリング キャンサー センター Antibodies targeting DLL3 and uses thereof
CN111018969A (en) * 2019-12-27 2020-04-17 上海药明生物技术有限公司 Method for purifying bispecific antibody by light chain select combined chromatography
AR121706A1 (en) 2020-04-01 2022-06-29 Hoffmann La Roche OX40 AND FAP-TARGETED BSPECIFIC ANTIGEN-BINDING MOLECULES
WO2021255146A1 (en) * 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and cea
WO2021255143A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and folr1
AU2021291011A1 (en) * 2020-06-19 2023-01-05 F. Hoffmann-La Roche Ag Antibodies binding to CD3 and CD19
AU2021291407A1 (en) * 2020-06-19 2022-09-29 F. Hoffmann-La Roche Ag Antibodies binding to CD3
US20230374162A1 (en) * 2020-10-07 2023-11-23 Amgen Inc. Rational selection of building blocks for the assembly of multispecific antibodies
TW202233684A (en) * 2020-11-18 2022-09-01 美商泰尼歐生物公司 Heavy chain antibodies binding to folate receptor alpha
CN112794911B (en) * 2021-04-14 2021-08-03 上海偌妥生物科技有限公司 Humanized anti-folate receptor 1 antibody and application thereof
WO2023079493A1 (en) * 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
WO2023161457A1 (en) 2022-02-27 2023-08-31 Evobright Gmbh Bispecific antibodies against cd277 and a tumor-antigen

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2011010169A (en) 2009-04-07 2011-10-11 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies.
CN103748114B (en) * 2011-08-23 2017-07-21 罗切格利卡特公司 T cell activation bispecific antigen binding molecules
NZ726258A (en) * 2012-08-31 2019-07-26 Immunogen Inc Antibodies and uses thereof to detect folate receptor 1
ES2691794T3 (en) * 2012-12-07 2018-11-28 Kyowa Hakko Kirin Co., Ltd. Anti-FOLR1 antibody
JP6233933B2 (en) 2012-12-25 2017-11-22 国立大学法人 鹿児島大学 Antibodies that recognize folate receptors α and β
RU2015140915A (en) * 2013-02-26 2017-04-03 Роше Гликарт Аг BSPECIFIC ANTI-BINDING MOLECULES ACTIVATING T-CELLS
EP3424952A1 (en) * 2013-03-15 2019-01-09 Amgen, Inc Heterodimeric bispecific antibodies
US20160009824A1 (en) * 2013-03-15 2016-01-14 Merck Patent Gmbh Tetravalent bispecific antibodies
UA118028C2 (en) * 2013-04-03 2018-11-12 Рош Глікарт Аг Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use
EP3733710A1 (en) * 2013-09-25 2020-11-04 Amgen, Inc Hetrodimeric v-c-fc-v-c antibody

Also Published As

Publication number Publication date
HRP20201747T1 (en) 2020-12-25
US20220220224A1 (en) 2022-07-14
CA2960929A1 (en) 2016-05-26
JP2017536121A (en) 2017-12-07
PT3221356T (en) 2020-10-29
JP2021058189A (en) 2021-04-15
ES2829829T3 (en) 2021-06-02
CO2017003048A2 (en) 2017-07-11
CL2017000954A1 (en) 2017-12-29
CR20170203A (en) 2017-06-29
KR20170081188A (en) 2017-07-11
SG11201702976TA (en) 2017-05-30
PH12017500939A1 (en) 2017-11-27
WO2016079076A1 (en) 2016-05-26
MX2017006571A (en) 2017-09-29
CN107074955A (en) 2017-08-18
JP7146395B2 (en) 2022-10-04
AR102732A1 (en) 2017-03-22
RS61010B1 (en) 2020-11-30
UA122676C2 (en) 2020-12-28
IL251292A0 (en) 2017-05-29
HUE051715T2 (en) 2021-03-29
MY184889A (en) 2021-04-29
AU2015348595B2 (en) 2021-06-10
DK3221356T3 (en) 2020-11-02
MA40972B1 (en) 2020-11-30
LT3221356T (en) 2020-11-25
US20160208019A1 (en) 2016-07-21
EP3221356B1 (en) 2020-09-02
PL3221356T3 (en) 2021-01-11
PE20170585A1 (en) 2017-05-11
BR112017007086A2 (en) 2018-01-16
TW201632556A (en) 2016-09-16
KR102648966B1 (en) 2024-03-19
AU2015348595A1 (en) 2017-04-20
EP3221356A1 (en) 2017-09-27
ZA201701843B (en) 2018-05-30
EA201791121A1 (en) 2018-04-30
SI3221356T1 (en) 2021-01-29
CN107074955B (en) 2021-06-22
TWI756164B (en) 2022-03-01

Similar Documents

Publication Publication Date Title
US20220220224A1 (en) T cell activating bispecific antigen binding molecules
US20220213224A1 (en) T cell activating bispecific antigen binding molecules
US20230076791A1 (en) Bispecific t cell activating antigen binding molecules
US20140242080A1 (en) Bispecific t cell activating antigen binding molecules
US20180282410A1 (en) Anti-cd3xrob04 bispecific t cell activating antigen binding molecules

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

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION