US20130058936A1 - Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use - Google Patents

Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use Download PDF

Info

Publication number
US20130058936A1
US20130058936A1 US13/591,024 US201213591024A US2013058936A1 US 20130058936 A1 US20130058936 A1 US 20130058936A1 US 201213591024 A US201213591024 A US 201213591024A US 2013058936 A1 US2013058936 A1 US 2013058936A1
Authority
US
United States
Prior art keywords
seq
fab fragment
antibody
antigen
bispecific antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/591,024
Other languages
English (en)
Inventor
Peter Bruenker
Tanja Fauti
Christiane Jaeger
Christian Klein
Pablo Umana
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.)
Roche Glycart AG
Original Assignee
Roche Glycart AG
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 Roche Glycart AG filed Critical Roche Glycart AG
Assigned to ROCHE GLYCART AG reassignment ROCHE GLYCART AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRUENKER, PETER, FAUTI, Tanja, JAEGER, CHRISTIANE, KLEIN, CHRISTIAN, UMANA, PABLO
Publication of US20130058936A1 publication Critical patent/US20130058936A1/en
Priority to US14/269,950 priority Critical patent/US11639397B2/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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

Definitions

  • the present invention contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Aug. 14, 2012, is named P4743_SequenceListing.txt and is 149,782 bytes in size.
  • the present invention relates to bispecific antibodies that specifically bind a T-cell activating antigen and a Tumor Antigen (TA), comprising a first Fab fragment and a second Fab fragment, wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody does not comprise a Fc domain; methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
  • TA Tumor Antigen
  • 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 undamaged.
  • One approach is to selectively induce an immune response against the tumor, which triggers the attack and subsequent destruction of tumor cells by immune effector cells such as natural killer (NK) cells or cytotoxic T lymphocytes (CTLs).
  • 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 which are able to bind to a surface antigen on cancer cells and to an activating invariant component of the T cell receptor (TCR) complex have become of interest in recent years. The simultaneous binding of the bispecific antibody to both of its targets forces a temporary interaction between cancer cell and T cell, causing activation of cytotoxic T cells and subsequent lysis of the tumor cell.
  • BiTE bispecific T cell engager
  • DART dual affinity retargeting
  • triomabs which are whole hybrid mouse/rat IgG molecules and also currently being evaluated in clinical trials, represent a larger sized format (reviewed in Seimetz et al., Cancer Treat Rev 36, 458-467 (2010)).
  • bispecific antibodies developed for T cell mediated cancer immunotherapy known so far have major drawbacks relating to their efficacy, toxicity and applicability.
  • Small constructs such as, for example, BiTE molecules—while being able to efficiently crosslink effector and target cells—have a very short serum half life requiring them to be administered to patients by continuous infusion.
  • IgG-like formats on the other hand—while having the great benefit of a long half life—suffer from toxicity associated with the native effector functions inherent to IgG molecules.
  • This immunogenic potential constitutes another unfavorable feature of IgG-like bispecific antibodies, for successful therapeutic development.
  • a major challenge in the general development of bispecific antibodies remains the production of bispecific antibody constructs at a clinically sufficient quantity and purity. The mispairing of antibody heavy and light chains of different specificities upon co-expression, decreases the yield of the correctly assembled construct and results in a number of non-functional side products.
  • the new bispecific antibodies can be easily produced with an increased yield due to a decreased amount of mispaired side-products, which show less aggregation than bispecific antibody fragments known in the art. Using the crossover approach correct LC association can be enforced without the need for the generation of a common light chain.
  • the new the new bispecific antibodies has a higher molecular weight compared to many conventional bispecific antibody fragments, thus preventing excessive kidney clearance and leading to an improved half-life in vivo.
  • the new bispecific antibodies are fully functional and have comparable or improved binding and activity as corresponding conventional bispecific antibodies.
  • the present invention provides bispecific antigen binding molecules designed for T cell activation and re-direction that combine good efficacy and produceability with low toxicity and favorable pharmacokinetic properties.
  • the present invention relates to bispecific antibodies that specifically bind a T-cell activating antigen and a Tumor Antigen (TA), comprising a first Fab fragment and a second Fab fragment, wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody does not comprise a Fc domain.
  • TA Tumor Antigen
  • the antibodies of the invention specifically bind to a Tumor Antigen on the surface of a tumor cell and at the same time bind to T-cell activating antigen.
  • the bispecific antibody is capable to elecit an immune response specifically at the site of the tumor, subsequently resulting in apoptosis of the target cell.
  • a bispecific antibody that specifically binds a T-cell activating antigen and a Tumor Antigen (TA)
  • TA Tumor Antigen
  • the first Fab fragment comprises at least one antigen binding site specific for a Tumor Antigen (TA); and the second Fab fragment comprises at least one antigen binding site specific for a T-cell activating antigen, wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody is devoid of a Fc domain.
  • the present invention relates to bispecific antibodies wherein the T-cell activating antigen is a CD3 T-Cell Co-Receptor (CD3) targeting antigen.
  • CD3 T-Cell Co-Receptor CD3 T-Cell Co-Receptor
  • a bispecific antibody that specifically binds CD3 T-Cell Co-Receptor (CD3) antigen and a Tumor Antigen (TA) comprising at least two fab fragments, wherein the first Fab fragment comprises at least one antigen binding site specific for a Tumor Antigen (TA); and the second Fab fragment comprises at least one antigen binding site specific for a CD3 T-Cell Co-Receptor (CD3) wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody is devoid of a Fc domain.
  • the first and second Fab fragments are connected via a peptide linker.
  • said peptide linker is a (G4S)2 linker.
  • said antibody additionally comprises a third Fab fragment.
  • said third Fab fragment comprises at least one antigen binding site specific for a Tumor Antigen.
  • the third Fab fragment is connected to the N or C-terminus of the light chain or the heavy chain of the first Fab fragment.
  • the third Fab fragment is connected to the N or C-terminus of the light chain or the heavy chain of the second Fab fragment.
  • the third Fab fragment is connected to the first or second Fab fragment via a peptide linker.
  • said peptide linker is a (G4S)2 linker.
  • the bispecific antibodies according to the invention are at least bivalent and can be trivalent or multivalent e.g. tetravalent or hexavalent.
  • said bispecific antibodies are bivalent (1+1 format) with one binding site each targeting a Tumor Antigen (TA) and a T-cell activating antigen, respectively.
  • said bispecific antibodies are trivalent (2+1 format) with two binding sites each targeting a Tumor Antigen (TA) and one binding site targeting a T-cell activating antigen, as detailed in the following section.
  • said a T-cell activating antigen is CD3.
  • the present invention relates to a pharmaceutical composition comprising a bispecific antibody of the present invention.
  • the present invention relates to a bispecific antibody of the present invention for the treatment of cancer.
  • use of the bispecific antibody as a medicament is provided.
  • Preferably said use is for the treatment of cancer.
  • the present invention relates to a nucleic acid sequence comprising a sequence encoding a heavy chain of a bispecific antibody of the present invention, a nucleic acid sequence comprising a sequence encoding a light chain of a bispecific antibody of the present invention, an expression vector comprising a nucleic acid sequence of the present invention and to a prokaryotic or eukaryotic host cell comprising a vector of the present invention.
  • a method of producing an antibody comprising culturing the host cell so that the antibody is produced is provided.
  • FIG. 1 Schematic illustration of exemplary bispecific antibody formats of the invention. a) Fab-Crossfab molecule C-terminal, b) Fab-Crossfab molecule N-terminal c) (Fab)2-Crossfab molecule C-terminal d) (Fab)2-Crossfab molecule N-terminal e) Fab-Crossfab-Fab molecule.
  • FIG. 2 Analysis of hu Fab(MCSP)-Crossfab(CD3) production and purification: SDS-Page: 4-12% Bis/Tris (NuPage [invitrogen]; coomassie stained): a) 1—Mark 12 (invitrogen), 2—hu Fab(MCSP)-Crossfab(CD3) non reduced; b) 1—Mark 12 (invitrogen), 2—hu Fab(MCSP)-Crossfab(CD3) reduced.
  • FIG. 3 Analysis Fab(MCSP)-Crossfab(CD3) production and purification. Analytical size exclusion chromatography, Chromatogram A280 (Superdex 200 10/300 GL [GE Healthcare]; 2 mM MOPS pH 7.3, 150 mM NaCl, 0.02% (w/v) NaCl; 50 ⁇ g sample were injected).
  • FIG. 4 Analysis of hu Fab(MCSP)-Fab(MCSP)-Crossfab(CD3) production and purification: SDS-Page: 4-12% Bis/Tris (NuPage [invitrogen]; coomassie stained): a) 1—Mark 12 (invitrogen), 2—hu Fab(MCSP)-Fab(MCSP)-Crossfab(CD3) non reduced; b) 1—Mark 12 (invitrogen), 2—hu Fab(MCSP)-Fab(MCSP)-Crossfab(CD3) reduced.
  • FIG. 5 Analysis of hu Fab(MCSP)-Fab(MCSP)-Crossfab(CD3) production and purification. Analytical size exclusion chromatography, Chromatogram A280 (Superdex 200 10/300 GL [GE Healthcare]; 2 mM MOPS pH 7.3, 150 mM NaCl, 0.02% (w/v) NaCl; 50 ⁇ g sample were injected).
  • FIG. 6 Analysis of hu Fab(MCSP)—Crossfab(CD3)-Fab(MCSP) production and purification.
  • SDS-Page 4-12% Bis/Tris (NuPage [invitrogen]; coomassie stained): a) 1—Mark 12 (invitrogen), 2—hu Fab(MCSP)—Crossfab(CD3)-Fab(MCSP) non reduced; b) 1—Mark 12 (invitrogen), 2—hu Fab(MCSP)-Crossfab(CD3)-Fab(MCSP) reduced.
  • FIG. 7 Analysis of hu Fab(MCSP)-Crossfab(CD3)-Fab(MCSP) production and purification. Analytical size exclusion chromatography, Chromatogram A280 (Superdex 200 10/300 GL [GE Healthcare]; 2 mM MOPS pH 7.3, 150 mM NaCl, 0.02% (w/v) NaCl; 50 ⁇ g sample were injected).
  • FIG. 8 Analysis of murine Crossfab(CD3)-Fab(MCSP)-Fab(MCSP) production and purification.
  • SDS-Page 4-12% Bis/Tris (NuPage [invitrogen]; coomassie stained): a) 1—Mark 12 (invitrogen), 2—murine Crossfab(CD3)-Fab(MCSP)-Fab(MCSP) non reduced; b) 1—Mark 12 (invitrogen), 2—murine Crossfab(CD3)-Fab(MCSP)-Fab(MCSP) reduced.
  • FIG. 9 Analysis of murine Crossfab(CD3)-Fab(MCSP)-Fab(MCSP) production and purification. Analytical size exclusion chromatography, Chromatogram A280 (Superdex 200 10/300 GL [GE Healthcare]; 2 mM MOPS pH 7.3, 150 mM NaCl, 0.02% (w/v) NaCl; 50 ⁇ g sample were injected).
  • the constructs with bivalent MCSP-targeting show comparable cytotoxic activity compared to the “(scFv)2” construct, whereas the “Fab-Crossfab” construct with monovalent MCSP binding is clearly less potent.
  • the “(Fab)2-Crossfab” induces apoptosis in target cells at least comparably good as the (scFv)2 molecule.
  • the effector to target cell ratio was 5:1.
  • the assay was analyzed after incubation for 23.5 hours at 37° C., 5% CO2.
  • the construct induces concentration-dependent, T cell-mediated apoptosis of human MCSP-expressing target cells.
  • the effector to target cell ratio was 5:1.
  • the assay was analyzed after incubation for 23.5 hours at 37° C., 5% CO2.
  • the construct induces T cell-mediated apoptosis of human MCSP-expressing target cells. There is only weak hyperactivation of T cells at this concentration of the construct.
  • the levels of granzyme B increased enormously upon activation of T cells in the presence of target cells.
  • the “(scFv)2” construct elevated the levels of TNF and IFNgamma, as well as granzyme B in the presence of target cells (A and B) a bit more compared to the other bispecific construct.
  • Th2 cytokines IL-10 and IL-4
  • IFNgamma induced by the “(Fab)2-Crossfab” construct in the absence of target cells.
  • FIG. 16 Surface expression level of the late activation marker CD25 on murine pan T cells, isolated from splenocytes.
  • E:T ratio is 10:1
  • Depicted is the expression level of the late activation marker CD25 on CD8+ T cells after 70 hours. Up-regulation of CD25 on CD8+ T cells with the (Fab)2-CrossFab construct occurs only in the presence of target cells.
  • FIG. 17 Analysis of Fab(CD33)-CrossFab (CD3) production and purification.
  • SDS-Page a) 3-8% Tris/Acetate (NuPage [invitrogen]; coomassie stained): a) 1—HiMark (invitrogen), 2—Fab(CD33)-CrossFab (CD3).non reduced; b) 4-12% Bis/Tris (NuPage [invitrogen]: 1—Mark 12 (invitrogen), 2—Fab(CD33)-CrossFab (CD3).reduced.
  • FIG. 18 Analysis of Fab(CD33)-CrossFab (CD3) production and purification. Analytical size exclusion chromatography, Chromatogram A280 (Superdex 200 10/300 GL [GE Healthcare]; 2 mM MOPS pH 7.3, 150 mM NaCl, 0.02% (w/v) NaCl; 50 ⁇ g sample were injected).
  • the “1+1 non-Fc” construct induces apoptosis in MV-3 target cells with a calculated EC50 of 25.4 pM, whereas the calculated EC50 for the “(scFv)2” reference molecule is 57 pM, showing a slight better potency of the “1+1 non-Fc” molecule in terms of EC50.
  • the CD69 median values are higher on CD8+ T cells compared to CD4+ T cells. There is a clear concentration-dependent increase in both, CD69 median values, as well percentage of CD69 positive cells for both constructs.
  • FIG. 21 Illustration of (scFv)2 reference molecule.
  • FIG. 22 Analysis of (scFv)2 (antiMCSP/anti huCD3e) production and purification.
  • SDS-Page 4-12% Bis/Tris (NuPage [invitrogen]; coomassie stained): 1—Mark 12 (invitrogen), 2—(scFv)2 (antiMCSP/anti huCD3e) reduced; 3-(scFv)2 (antiMCSP/anti huCD3e), non reduced.
  • FIG. 23 Analysis of (scFv)2 (antiMCSP/anti huCD3e) production and purification Analytical size exclusion chromatography, Chromatogram A280 (Superdex 75 10/300 GL [GE Healthcare]; 2 mM MOPS pH 7.3, 150 mM NaCl, 0.02% (w/v) NaCl; 50 ⁇ g sample ((scFv)2 (antiMCSP/anti huCD3e)) were injected).
  • “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.
  • acceptor human framework for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework “derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • a “human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest , Fifth Edition, NIH Publication 91-3242, Bethesda Md. (1991), vols. 1-3.
  • the subgroup is subgroup kappa I as in Kabat et al., supra.
  • the subgroup is subgroup III as in Kabat et al., supra.
  • 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.
  • CDRs complementarity determining regions
  • Exemplary hypervariable loops occur at amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3).
  • Exemplary CDRs CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3 occur at amino acid residues 24-34 of L1, 50-56 of L2, 89-97 of L3, 31-35B of H1, 50-65 of H2, and 95-102 of H3.
  • hypervariable regions HVRs
  • CDRs complementarity determining regions
  • 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.
  • CDRs generally comprise the amino acid residues that form the hypervariable loops.
  • CDRs also comprise “specificity determining residues,” or “SDRs,” which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a-CDRs.
  • Exemplary a-CDRs (a-CDR-L1, a-CDR-L2, a-CDR-L3, a-CDR-H1, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of L1, 50-55 of L2, 89-96 of L3, 31-35B of H1, 50-58 of H2, and 95-102 of H3.
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al., supra.
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • antibody includes the bispecific antibodies of the invention comprising at least two fab fragments but no Fc domain.
  • bispecific means that the antigen binding molecule is able to specifically bind to at least two distinct antigenic determinants. 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 “monovalent binding to an antigen” means that not more than one antigen comprised in the antibody specifically binds to that antigen.
  • a “human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NS0 or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell.
  • recombinant human antibodies have variable and constant regions in a rearranged form.
  • the recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation.
  • the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germ line VH and VL sequences, may not naturally exist within the human antibody germ line repertoire in vivo.
  • a “humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • a “humanized form” of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • humanized antibodies encompassed by the present invention are those in which the constant region has been additionally modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding.
  • FcR Fc receptor
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising a murine variable region and a human constant region are preferred. Other preferred forms of “chimeric antibodies” encompassed by the present invention are those in which the constant region has been modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding. Such chimeric antibodies are also referred to as “class-switched antibodies”.
  • Chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding immunoglobulin variable regions and DNA segments encoding immunoglobulin constant regions. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art. See e.g. Morrison, S. L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; U.S. Pat. Nos. 5,202,238 and 5,204,244.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • 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′-SH, F(ab′) 2 ; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • scFv antibodies are, e.g. described in Houston, J. S., Methods in Enzymol. 203 (1991) 46-96).
  • antibody fragments comprise single chain polypeptides having the characteristics of a VH domain, namely being able to assemble together with a VL domain, or of a VL domain, namely being able to assemble together with a VH domain to a functional antigen binding site and thereby providing the antigen binding property of full length antibodies.
  • Fab fragment refers to an antibody fragment comprising a light chain fragment comprising a VL domain and a constant domain of a light chain (CL), and a VH domain and a first constant domain (CH1) of a heavy chain.
  • the bispecific antibodies of the invention comprise at least two Fab fragments, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged. Due to the exchange of either the variable regions or the constant regions, said second Fab fragment is also referred to as “cross-Fab” fragment or “xFab” fragment or “crossover Fab” fragment.
  • crossover Fab molecule comprises a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1), and a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL).
  • VL variable region
  • CH1 heavy chain constant region
  • VH heavy chain variable region
  • CL light chain constant region
  • the crossover Fab molecule comprises a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL), and a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1).
  • This crossover Fab molecule is also referred to as CrossFab (CLCH1) .
  • said Fab fragments are connected via a peptide linker.
  • “connected” is meant that the Fab fragments are linked by peptide bonds, either directly or via one or more peptide linker.
  • peptide linker denotes a peptide with amino acid sequences, which is preferably of synthetic origin. These peptide linkers according to invention are used to connect one of the Fab fragments to the C- or N-terminus of the other Fab fragment to form a multispecific antibody according to the invention.
  • peptide linkers are peptides with an amino acid sequence with a length of at least 5 amino acids, preferably with a length of 5 to 100, more preferably of 10 to 50 amino acids.
  • linkers may comprise (a portion of) an immunoglobulin hinge region.
  • said peptide linker is (G 4 S) 2 (SEQ ID: NO 28).
  • peptide linkers suitable for connecting the Fab fragments for example, (G 4 S) 6 -GG (SEQ ID NO: 147) or (SG 3 ) 2 -(SEG 3 ) 4 -(SG 3 )-SG (SEQ ID NO: 148), or EPKSC(D)-(G 4 S) 2 (SEQ ID NOs 145 and 146).
  • an antigen binding domain refers to the part of an antigen binding molecule that comprises the area which specifically binds to and is complementary to part or all of an antigen. Where an antigen is large, an antigen binding molecule may only bind to a particular part of the antigen, which part is termed an epitope.
  • 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).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
  • antigen-binding site of an antibody when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the antigen-binding portion of an antibody comprises amino acid residues from the “complementary determining regions” or “CDRs”.
  • “Framework” or “FR” regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chain variable domains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • CDR3 of the heavy chain is the region which contributes most to antigen binding and defines the antibody's properties.
  • CDR and FR regions are determined according to the standard definition of Kabat et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991) and/or those residues from a “hypervariable loop”.
  • epitope includes any polypeptide determinant capable of specific binding to an antibody.
  • epitope determinant include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and or specific charge characteristics.
  • An epitope is a region of an antigen that is bound by an antibody.
  • Fc domain 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 Fc domain is composed of two identical protein fragments, derived from the second and third constant domains of the antibody's two heavy chains in IgG, IgA and IgD isotypes; IgM and IgE Fc domains contain three heavy chain constant domains (C H domains 2-4) in each polypeptide chain.
  • the bispecific antibodies of the invention are devoid of the Fc domain. “Devoid of the Fc domain” as used herein means that the bispecific antibodies of the invention do not comprise a CH2, CH3 or CH4 domain; i.e. the constant heavy chain consists solely of one or more CH1 domains.
  • Binding affinity refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
  • binding affinity refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • binding means 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)).
  • 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
  • the extent of binding of a bispecific antibody that specifically binds to a T-cell activating antigen and a Tumor Antigen (TA) to an unrelated protein is less than about 10% of the binding of the antibody to a T-cell activating antigen or a Tumor Antigen (TA) as measured, e.g., by a radioimmunoassay (RIA) or flow cytometry (FACS).
  • RIA radioimmunoassay
  • FACS flow cytometry
  • a bispecific antibody that specifically binds T-cell activating antigen and a Tumor Antigen has a dissociation constant (KD) 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).
  • KD dissociation constant
  • a bispecific antibody that specifically binds a T-cell activating antigen and a Tumor Antigen (TA) binds to an epitope of a T-cell activating antigen or a Tumor Antigen (TA) that is conserved among a T-cell activating antigen or a Tumor Antigen (TA) from different species.
  • an “affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
  • HVRs hypervariable regions
  • a bispecific antibody that specifically binds a T cell activating antigen and a Tumor Antigen refers to a bispecific antibody that is capable of binding a T cell activating antigen and a Tumor Antigen with sufficient affinity such that the antibody is useful in mediating a T-cell mediated immune response in or near cells expressing a Tumor Antigen.
  • the T cell activating antigen is the CD3 T-Cell Co-Receptor (CD3) antigen, particularly human or cynomolgus CD3, most particularly human CD3.
  • CD3 T-Cell Co-Receptor (CD3) antigen particularly human or cynomolgus CD3, most particularly human CD3.
  • the T cell activating antigen is the epsilon subunit of CD3.
  • the T cell activating antigen is the alpha or beta subunit of CD3.
  • the bispecific antibody that specifically binds a T cell activating antigen and a Tumor Antigen can compete with monoclonal antibody H2C (described in PCT publication no. WO2008/119567) for binding an epitope of CD3.
  • the bispecific antibody that specifically binds a T cell activating antigen and a Tumor Antigen can compete with monoclonal antibody V9 (described in Rodrigues et al., Int J Cancer Suppl 7, 45-50 (1992) and U.S. Pat. No. 6,054,297) for binding an epitope of CD3.
  • the bispecific antibody that specifically binds a T cell activating antigen and a Tumor Antigen can compete with monoclonal antibody FN18 (described in Nooij et al., Eur J Immunol 19, 981-984 (1986)) for binding an epitope of CD3.
  • an “activating T cell antigen” as used herein refers to an antigenic determinant expressed on the surface of a T lymphocyte, particularly a cytotoxic T lymphocyte, which is capable of inducing T cell activation upon interaction with an antigen binding molecule. Specifically, interaction of an antigen binding molecule with an activating T cell antigen may induce T cell activation by triggering the signaling cascade of the T cell receptor complex. In a particular embodiment the activating T cell antigen is CD3.
  • 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.
  • CD3 T-Cell Co-Receptor refers to a protein complex and is composed of four distinct chains. In mammals, the complex contains a CD3 ⁇ chain, a CD3 ⁇ chain, and two CD3 ⁇ chains. These chains associate with a molecule known as the T cell receptor (TCR) and the ⁇ -chain to generate an activation signal in T lymphocytes.
  • TCR T cell receptor
  • CD3 T-Cell Co-Receptor (CD3)” includes any native CD3 from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated, preferably from a human source.
  • CD3 T-Cell Co-Receptor refers to human or cynomolgus CD3, particularly human CD3.
  • the T cell activating antigen is the epsilon subunit of CD3.
  • the T cell activating antigen is the alpha or beta subunit of CD3.
  • An exemplary sequence of human CD3 is given in SEQ ID NO.: 103.
  • Tumor Antigen refers to tumor-associated antigens as well as tumor-specific antigens, i.e. any immunogenic epitope (e.g., protein) expressed by a tumor cell.
  • the protein may be expressed by non tumor cells but be immunogenic only when expressed by a tumor cell.
  • the protein may be expressed by tumor cells, but not normal cells.
  • an anti-TA antibody of the invention binds to the extracellular domain of TA.
  • said Tumor Antigen is a human Tumor Antigen.
  • Tumor Antigens include but are not limited to Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP, UniProt Q6UVK1, NCBI Accession NP — 001888), Fibroblast Activation Protein (FAP, Uni Prot Q12884, Q86Z29, Q99998; NCBI Accession NP — 004451), Epidermal Growth Factor Receptor (EGFR, also known as ErbB1 and Her1, UniProt P00533; NCBI Accession NP — 958439, NP — 958440), Carcinoembryonic Antigen (CEA, also known as Carcinoembryonic antigen-related cell adhesion molecule 5 or CD66e; UniProt P06731, NCBI Accession NP — 004354) and CD33 (also known as gp76 or Sialic acid-binding Ig-like lectin 3 (Siglec-3), UniProt P20138, NCBI Accession NP — 00
  • the bispecific antibody of the invention comprises at least one antigen binding site that is specific for Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP).
  • MCSP Melanoma-associated Chondroitin Sulfate Proteoglycan
  • the bispecific antibody of the invention comprises at least one antigen binding site that is specific for CD33.
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Natural antibodies, for example, are monospecific. “Bispecific antibodies” according to the invention are antibodies which have two different antigen-binding specificities. Antibodies of the present invention are specific for two different antigens, i.e. a T-Cell activating antigen as first antigen and a Tumor Antigen as second antigen.
  • monospecific antibody denotes an antibody that has one or more binding sites each of which bind to the same epitope of the same antigen.
  • bispecific antibody denotes an antibody that has at least two binding sites each of which bind to different epitopes of the same antigen or a different antigen.
  • the antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites.
  • a bispecific antibody with binding specificities for a Tumor Antigen (TA) and a T-cell activating antigen.
  • TA Tumor Antigen
  • bispecific antibodies may bind to two different epitopes of TA.
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express TA.
  • bispecific antibodies as used within the current application denotes the presence of a specified number of binding sites in an antibody molecule.
  • the terms “bivalent”, “tetravalent”, and “hexavalent” denote the presence of two binding sites, four binding sites, and six binding sites, respectively, in an antibody molecule.
  • the bispecific antibodies according to the invention are at least “bivalent” and may be “trivalent” or “multivalent” (e.g.“tetravalent” or “hexavalent”).
  • Antibodies of the present invention have two or more binding sites and are bispecific. That is, the antibodies may be bispecific even in cases where there are more than two binding sites (i.e. that the antibody is trivalent or multivalent).
  • an “antibody that binds to the same epitope” as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more.
  • An exemplary competition assay is provided herein.
  • No substantial cross-reactivity means that a molecule (e.g., an antibody) does not recognize or specifically bind an antigen different from the actual target antigen of the molecule (e.g. an antigen closely related to the target antigen), particularly when compared to that target antigen.
  • an antibody may bind less than about 10% to less than about 5% to an antigen different from the actual target antigen, or may bind said antigen different from the actual target antigen at an amount selected from the group consisting of less than about 10%, 9%, 8% 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.2%, or 0.1%, preferably less than about 2%, 1%, or 0.5%, and most preferably less than about 0.2% or 0.1% antigen different from the actual target antigen.
  • 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:
  • an “isolated” antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding a bispecific antibody that specifically binds a T-Cell activating antigen and a Tumor Antigen (TA) refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • amino acid denotes the group of naturally occurring carboxy ⁇ -amino acids comprising alanine (three letter code: ala, one letter code: A), arginine (arg, R), asparagine (asn, N), aspartic acid (asp, D), cysteine (cys, C), glutamine (gln, Q), glutamic acid (glu, E), glycine (gly, G), histidine (his, H), isoleucine (ile, I), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y), and valine (val, V).
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • 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.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as “expression vectors.”
  • the expressions “cell”, “cell line”, and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transfectants” and “transfected cells” include the primary subject cell and cultures derived there from without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included.
  • 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.
  • naked antibody refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel.
  • the naked antibody may be present in a pharmaceutical formulation.
  • an “immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal
  • N-terminus denotes the last amino acid of the N-terminus
  • C-terminus denotes the last amino acid of the C-terminus
  • pharmaceutical formulation 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 formulation, 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 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.
  • antibodies of the invention are used to delay development of a disease or to slow the progression 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).
  • 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.
  • the individual or subject is a human.
  • an “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.
  • cancer refers to proliferative diseases, such as lymphomas, lymphocytic leukemias, lung cancer, non small cell lung (NSCL) cancer, bronchioloalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney
  • 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 invention is based, in part, on bispecific antibodies comprising a first antigen binding site specific for a T-cell activating antigen and a second antigen binding site specific for a Tumor Antigen (TA).
  • TA Tumor Antigen
  • the present invention relates to bispecific antibodies combining a T-cell activating antigen binding site with a second antigen binding site that targets a Tumor Antigen (TA).
  • the antibodies of the invention specifically bind to a Tumor Antigen on the surface of a tumor cell and at the same time bind to an antigen on the surface of cytotoxic T lymphocytes.
  • said antigen is a CD3 T-Cell Co-Receptor (CD3) antigen.
  • CD3 T-Cell Co-Receptor (CD3) antigen CD3 T-Cell Co-Receptor
  • the T cell activating bispecific antibody is capable of simultaneous binding to a tumor cell antigen, and an activating T cell antigen.
  • the T cell activating bispecific antibody is capable of crosslinking a T cell and a tumor cell by simultaneous binding to a tumor cell antigen and an activating T cell antigen.
  • such simultaneous binding results in lysis of the 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.
  • binding of the T cell activating bispecific antibody to the activating T cell antigen without simultaneous binding to the target cell antigen does not result in T cell activation.
  • the T cell activating bispecific antibody is capable of re-directing cytotoxic activity of a T cell to a target cell.
  • said re-direction is independent of MHC-mediated peptide antigen presentation by the target cell and/or specificity of the T cell.
  • a T cell according to any of the embodiments of the invention is a cytotoxic T cell.
  • the T cell is a CD4 + or a CD8 + T cell, particularly a CD8 + T cell.
  • bispecific antibodies are provided that specifically bind a T-cell activating antigen and a Tumor Antigen (TA), comprising a first Fab fragment and a second Fab fragment, wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody does not comprise a Fc domain.
  • TA Tumor Antigen
  • a bispecific antibody that specifically binds a T-cell activating antigen and a Tumor Antigen (TA)
  • TA Tumor Antigen
  • the first Fab fragment comprises at least one antigen binding site specific for a Tumor Antigen (TA); and the second Fab fragment comprises at least one antigen binding site specific for a T-cell activating antigen, wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody is devoid of a Fc domain.
  • the T cell activating antigen is the CD3 T-Cell Co-Receptor (CD3) antigen, particularly human or cynomolgus CD3, most particularly human CD3.
  • CD3 T-Cell Co-Receptor (CD3) antigen particularly human or cynomolgus CD3, most particularly human CD3.
  • the T cell activating antigen is the epsilon subunit of CD3.
  • the T cell activating antigen is the alpha or beta subunit of CD3.
  • a bispecific antibody that specifically binds CD3 T-Cell Co-Receptor (CD3) antigen and a Tumor Antigen (TA) is provided, comprising at least two fab fragments, wherein the first Fab fragment comprises at least one antigen binding site specific for a Tumor Antigen (TA); and the second Fab fragment comprises at least one antigen binding site specific for a CD3 T-Cell Co-Receptor (CD3) wherein either the variable regions or the constant regions of the second Fab heavy and light chain are exchanged; and wherein the bispecific antibody is devoid of a Fc domain.
  • CD3 T-Cell Co-Receptor CD3 T-Cell Co-Receptor
  • the first and second Fab fragments are connected via a peptide linker.
  • said peptide linker is a peptide with an amino acid sequence with a length of at least 5 amino acids, preferably with a length of 5 to 100, more preferably of 10 to 50 amino acids.
  • said peptide linker is (G 4 S) 2 . The peptide linker is used to connect the first and the second Fab fragment.
  • the first Fab fragment is connected to the C- or N-terminus of the second Fab fragment.
  • the first Fab fragment is connected to the N-terminus of the second Fab fragment.
  • the variable or the constant domains of the heavy and the light chains of the second Fab fragment are exchanged, different bispecific antibody molecules are possible when the first Fab fragment is connected to the N-terminus of the second Fab fragment.
  • variable domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (VHVL) ), and the C-terminus of the heavy or light chain of the first Fab fragment is connected to the N-terminus of the VLCH1 chain of the second Fab fragment.
  • the C-terminus heavy chain of the first Fab fragment is connected to the N-terminus of the VLCH1 chain of the second Fab fragment.
  • the bispecific antibody comprises three chains: a light chain (VLCL) of the first Fab fragment, the heavy chain of the first Fab fragment connected to the VLCH1 chain of the second Fab fragment via a peptide linker (VHCH1-linker-VLCH1) and a VHCL chain of the second Fab fragment.
  • VLCL light chain
  • VHCH1-linker-VLCH1 peptide linker
  • the constant domains of the second Fab fragment are exchanged (i.e the second Fab fragment is a CrossFab (CLCH1) ) and the C-terminus of the heavy or light chain of the first Fab fragment is connected to the N-terminus of the VHCL chain of the second Fab fragment.
  • the C-terminus of the heavy chain of the first Fab fragment is connected to the N-terminus of the VHCL chain of the second Fab fragment.
  • the bispecific antibody comprises three chains: a light chain (VLCL) of the first Fab fragment, the heavy chain of the first Fab fragment connected to the VHCL chain of the second Fab fragment via a peptide linker (VHCH1-linker-VHCL) and a VLCH1 chain of the second Fab fragment.
  • VLCL light chain
  • VHCH1-linker-VHCL peptide linker
  • the first Fab fragment is connected to the C-terminus of the second Fab fragment.
  • the variable or the constant domains of the heavy and the light chains of the second Fab fragment are exchanged different bispecific antibody molecules are possible when the first Fab fragment is connected to the C-terminus of the second Fab fragment.
  • variable domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (VHVL) ), and the CH1 domain of the second Fab fragment is connected to the N-terminus of the heavy or light chain of the first Fab fragment.
  • the CH1 domain of the second Fab fragment is connected to the N-terminus of the heavy chain of the first Fab fragment.
  • the bispecific antibody comprises three chains: a light chain (VLCL) of the first Fab fragment, the VLCH1 chain of the second Fab fragment connected to the heavy chain of the first Fab fragment via a peptide linker (VLCH1-linker-VHCH1) and a VHCL chain of the second Fab fragment.
  • the constant domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (CLCH1) ), and the CL domain of the second Fab fragment is connected to the N-terminus of the heavy of light chain of the first Fab fragment.
  • the CL domain of the second Fab fragment is connected to the N-terminus of the heavy chain of the first Fab fragment.
  • the bispecific antibody comprises three chains: a light chain (VLCL) of the first Fab fragment, the VHCL chain of the second Fab fragment connected to the heavy chain of the first Fab fragment via a peptide linker (VLCH1-linker-VHCH1) and a VLCH1 chain of the second Fab fragment.
  • the bispecific antibodies according to the invention are at least bivalent and can be trivalent or multivalent e.g. tetravalent or hexavalent.
  • said bispecific antibodies are bivalent (1+1 format) with one binding site each targeting a Tumor Antigen (TA) and a T-cell activating antigen, respectively.
  • said bispecific antibodies are trivalent (2+1 format) with two binding sites each targeting a Tumor Antigen (TA) and one binding site targeting a T-cell activating antigen, as detailed in the following section.
  • said antibody additionally comprises a third Fab fragment.
  • said third Fab fragment comprises at least one antigen binding site specific for a Tumor Antigen.
  • the antigen binding site of said third Fab fragment is specific for the same Tumor Antigen as the antigen binding site of the first Fab fragment.
  • the third Fab fragment is connected to the N or C-terminus of the first Fab fragment. In one embodiment the third Fab fragment is connected to the first Fab fragment via a peptide linker. Preferably said peptide linker is a (G4S)2 linker.
  • the third Fab fragment is connected to the N or C-terminus of the light chain or the heavy chain of the first Fab fragment. Depending on which terminus of the first Fab fragment is connected to the second Fab fragment (as detailed above), the third Fab fragment is connected on the opposite (free) terminus of the first fragment.
  • the bispecific antibody of the invention comprises three Fab fragments wherein said Fab fragments and said linker are connected in the following order from N-terminal to C-terminal direction: Fab fragment 3-linker-Fab fragment 1-linker-Fab fragment 2, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged.
  • the C-terminus of the third Fab fragment is connected to the N-terminus of the first Fab fragment.
  • the Fab fragments can be connected to each other via the heavy or the light chains.
  • the C-terminus of the heavy chain of the third Fab fragment is connected to the N-terminus of the heavy chain of the first Fab fragment via a peptide linker; and the C-terminus of the first Fab fragment is connected to the N-terminus of the second Fab fragment, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged.
  • variable or the constant domains of the heavy and the light chains of the second Fab fragment are exchanged different bispecific antibody molecules are possible.
  • variable domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (VHVL) ), and the chains of the three Fab fragments are connected in the following order from N-terminal to C-terminal direction: VHCH1-linker-VHCH1-linker-VLCH1.
  • the bispecific antibody comprises four chains: a light chain (VLCL) of the third Fab fragment, a light chain (VLCL) of the first Fab fragment, the heavy chain of the third fragment connected to the heavy chain of the first Fab fragment which itself is connected to the VLCH1 chain of the second Fab fragment via a peptide linker (VHCH1-linker-VHCH1-linker-VLCH1) and a VHCL chain of the second Fab fragment.
  • the constant domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (CLCH1) ), and the chains of the three Fab fragments are connected in the following order from N-terminal to C-terminal direction: VHCH1-linker-VHCH1-linker-VHCL.
  • CLCH1 CrossFab
  • the bispecific antibody comprises four chains: a light chain (VLCL) of the third Fab fragment, a light chain (VLCL) of the first Fab fragment, the heavy chain of the third fragment connected to the heavy chain of the first Fab fragment which itself is connected to the VHCL chain of the second Fab fragment via a peptide linker (VHCH1-linker-VHCH1-linker-VHCL) and a VLCH1 chain of the second Fab fragment.
  • the bispecific antibody of the invention comprises three Fab fragments wherein said Fab fragments and said linker are connected in the following order from N-terminal to C-terminal direction: Fab fragment 2-linker-Fab fragment 1-linker-Fab fragment 3, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged.
  • the N-terminus of the third Fab fragment is connected to the C-terminus of the first Fab fragment.
  • the Fab fragments can be connected to each other via the heavy or the light chains.
  • the N-terminus of the heavy chain of the third Fab fragment is connected to the C-terminus of the heavy chain of the first Fab fragment via a peptide linker; and the N-terminus of the first Fab fragment is connected to the C-terminus of the second Fab fragment, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged.
  • variable or the constant domains of the heavy and the light chains of the second Fab fragment are exchanged different bispecific antibody molecules are possible.
  • variable domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (VHVL) ), and the chains of the three Fab fragments are connected in the following order from N-terminal to C-terminal direction: VLCH1-linker-VHCH1-linker-VHCH1.
  • the bispecific antibody comprises four chains: a light chain (VLCL) of the third Fab fragment, a light chain (VLCL) of the first Fab fragment, the VLCH1 chain of the second Fab fragment connected to the heavy chain of the first fragment which itself is connected to the heavy chain of the first Fab fragment via a peptide linker (VLCH1-linker-VHCH1-linker-VHCH1) and a VHCL chain of the second Fab fragment.
  • the constant domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (CLCH1) ), and the chains of the three Fab fragments are connected in the following order from N-terminal to C-terminal direction: VHCL-linker-VHCH1-linker-VHCH1.
  • the bispecific antibody comprises four chains: a light chain (VLCL) of the third Fab fragment, a light chain (VLCL) of the first Fab fragment, the VHCL chain of the second Fab fragment connected to the heavy chain of the first fragment which itself is connected to the heavy chain of the first Fab fragment via a peptide linker (VHCL-linker-VHCH1-linker-VHCH1) and a VLCH1 chain of the second Fab fragment.
  • VLCL light chain
  • VLCL light chain
  • VLCL light chain of the first Fab fragment
  • VHCL chain of the second Fab fragment connected to the heavy chain of the first fragment which itself is connected to the heavy chain of the first Fab fragment via a peptide linker (VHCL-linker-VHCH1-linker-VHCH1) and a VLCH1 chain of the second Fab fragment.
  • the third Fab fragment is connected to N or C-terminus of the light chain or the heavy chain of the second Fab fragment.
  • the third Fab fragment is connected to the second Fab fragment via a peptide linker.
  • said peptide linker is a (G4S)2 linker.
  • the Fab fragments can be connected to each other via the heavy or the light chains.
  • the bispecific antibody of the invention comprises three Fab fragments wherein said Fab fragments and said linker are connected in the following order from N-terminal to C-terminal direction: Fab fragment 1-linker-Fab fragment 2-linker-Fab fragment 3, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged.
  • the N-terminus of the third Fab fragment is connected to the C-terminus of the second Fab fragment.
  • the C-terminus of the heavy chain of the third Fab fragment is connected to the N-terminus of the second Fab fragment via a peptide linker; and the N-terminus of the first Fab fragment is connected to the C-terminus of the second Fab fragment, wherein either the variable regions or the constant regions of the heavy and light chain of the second Fab fragment are exchanged.
  • variable or the constant domains of the heavy and the light chains of the second Fab fragment are exchanged different bispecific antibody molecules are possible.
  • variable domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (VHVL) ), and the chains of the three Fab fragments are connected in the following order from N-terminal to C-terminal direction: VHCH1-linker-VLCH1-linker-VHCH1.
  • the bispecific antibody comprises four chains: a light chain (VLCL) of the third Fab fragment, a light chain (VLCL) of the first Fab fragment, the heavy chain of the third fragment connected to to the N-terminus of the VLCH1 chain of the second Fab fragment, and the C-terminus of said VLCH1 chain connected to the N-terminus of the heavy chain of the first Fab fragment via a peptide linker (VHCH1-linker-VLCH1-linker-VHCH1) and a VHCL chain of the second Fab fragment.
  • the constant domains of the second Fab fragment are exchanged (i.e. the second Fab fragment is a CrossFab (CLCH1) ), and the chains of the three Fab fragments are connected in the following order from N-terminal to C-terminal direction: VHCH1-linker-VHCL-linker-VHCH1.
  • the bispecific antibody comprises four chains: a light chain (VLCL) of the third Fab fragment, a light chain (VLCL) of the first Fab fragment, the heavy chain of the third fragment connected to to the N-terminus of the VHCL chain of the second Fab fragment, and the C-terminus of said VHCL chain connected to the N-terminus of the heavy chain of the first Fab fragment via a peptide linker (VHCH1-linker-VHCL-linker-VHCH1) and a VLCH1 chain of the second Fab fragment.
  • the antigen binding site of said third Fab fragment is specific for the same Tumor Antigen as the antigen binding site of the first Fab fragment
  • the bispecific antibody of the invention comprises three Fab fragments connected via a peptide linker in the following order (either from N-terminal to C-terminal direction or from C-terminal to N-terminal direction): Fab (TA) -linker-Fab (TA) -linker-xFab (T-cell activating antigen) , wherein Fab (TA) denotes a Fab fragment with antigen binding site specific for a Tumor Antigen and xFab (T-cell activating antigen) denotes a Fab fragment with antigen binding site specific for a T-cell activating antigen, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
  • the antigen binding site of said third Fab fragment is specific for the same Tumor Antigen as the antigen binding site of the first Fab fragment
  • the bispecific antibody of the invention comprises three Fab fragments connected via a peptide linker in the following order (either from N-terminal to C-terminal direction or from C-terminal to N-terminal direction): Fab (TA) -linker-xFab (T-cell activating antigen) -linker-Fab (TA) , wherein Fab (TA) denotes a Fab fragment with antigen binding site specific for a Tumor Antigen and xFab (T-cell activating antigen) denotes a Fab fragment with antigen binding site specific for a T-cell activating antigen, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
  • the bispecific antibody comprises an antigen binding moiety that can compete with monoclonal antibody V9 for binding to an epitope of CD3. See for example Rodigues et al., Int J Cancer Suppl 7 (1992), 45-50; U.S. Pat. No. 6,054,297, incorporated herein by reference in its entirety.
  • the bispecific antibody comprises an antigen binding moiety that can compete with monoclonal antibody FN18 for binding to an epitope of CD3. See Nooij et al., Eur J Immunol 19 (1986), 981-984, incorporated herein by reference in its entirety.
  • the bispecific antibody comprises an antigen binding moiety that can compete with monoclonal antibody CH2527 (Sequence ID 157 and 158) or an affinity matured variant thereof for binding to an epitope of CD3.
  • the bispecific antibody comprises a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region comprises a CDR1 of SEQ ID. NO. 10 or SEQ ID. NO. 32, a CDR2 of SEQ ID. NO. 11 or SEQ ID. NO. 33, and a CDR3 of SEQ ID. NO. 12 or SEQ ID. NO. 34; and wherein the light chain variable region comprises a CDR1 of SEQ ID. NO. 7 or SEQ ID. NO. 29, a CDR2 of SEQ ID. NO. 8 or SEQ ID. NO. 30, and a CDR3 of SEQ ID. NO. 9 or SEQ ID. NO. 31.
  • the bispecific antibody comprises a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region comprises a CDR1 of SEQ ID. NO. 10, a CDR2 of SEQ ID. NO. 11, and a CDR3 of SEQ ID. NO. 12; and wherein the light chain variable region comprises a CDR1 of SEQ ID. NO. 7, a CDR2 of SEQ ID. NO. 8 and a CDR3 of SEQ ID. NO. 9.
  • the bispecific antibody comprises a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region comprises a CDR1 of SEQ ID. NO. 32, a CDR2 SEQ ID. NO. 33, and a CDR3 of SEQ ID. NO. 34; and wherein the light chain variable region comprises a CDR1 of SEQ ID. NO. 29, a CDR2 of SEQ ID. NO. 30, and a CDR3 of SEQ ID. NO. 31.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region sequence is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 20 or SEQ ID. NO. 36; wherein the light chain variable region sequence is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 19. or SEQ ID. NO 35, or variants thereof that retain functionality.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID. NO. 20; and a light chain variable region comprising an amino acid sequence of SEQ ID. NO. 19 or variants thereof that retain functionality.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID. NO. 36; and a light chain variable region comprising an amino acid sequence of SEQ ID. NO. 35 or variants thereof that retain functionality.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID. NO. 158; and a light chain variable region comprising an amino acid sequence of SEQ ID. NO. 157 or variants thereof that retain functionality.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region sequence is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein the heavy chain variable region sequence is an affinity matured variant of SEQ ID. NO. 158 and wherein the light chain variable region sequence is an affinity matured variant of SEQ ID. NO. 157.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 158 and/or SEQ ID. NO. 157 are exchanged.
  • the bispecific antibody comprises a light chain and a heavy chain of second Fab fragment specifically binding to CD3, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 22 or SEQ ID. NO. 38 or variants thereof that retain functionality.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 22 or SEQ ID. NO 38, and a light chain and a heavy chain of first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody comprises a light chain and a heavy chain of second Fab fragment specifically binding to CD3, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 22.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain and a heavy chain of first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 21 or SEQ ID. NO. 37.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 21 or SEQ ID. NO. 37, and a light chain and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 21.
  • the bispecific antibody comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 21, and a light chain and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • a bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, said heavy chain comprising a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22 or SEQ ID. NO. 38; and said light chain comprising a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21 or SEQ ID. NO. 37.
  • a bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, said heavy chain comprising a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22; and said light chain comprising a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21.
  • a bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, said heavy chain comprising a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22; and said light chain comprising a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21, and a light chain and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21, and a light chain and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 35 and a variable heavy chain of SEQ ID NO: 36, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 38, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 37.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 35 and a variable heavy chain of SEQ ID NO: 36, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 38, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 37, and a light chain and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 157 and a variable heavy chain of SEQ ID NO: 158, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 157 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 158 or an affinity matured variant thereof, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 158 and/or SEQ ID. NO. 157 are exchanged.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 157 and a variable heavy chain of SEQ ID NO: 158, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21, and a light chain and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • TA Tumor Antigen
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 157 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 158 or an affinity matured variant thereof, and a heavy chain constant region comprising the amino acid sequence of SEQ ID NO: 22, and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 21 and a heavy chain of a first Fab fragment specific for a Tumor Antigen (TA) comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 158 and/or SEQ ID. NO. 157 are exchanged.
  • the Tumor Antigen is selected from the group of Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP), Epidermal Growth Factor Receptor (EGFR), Carcinoembryonic Antigen (CEA), Fibroblast Activation Protein (FAP) and CD33.
  • MCSP Melanoma-associated Chondroitin Sulfate Proteoglycan
  • EGFR Epidermal Growth Factor Receptor
  • CEA Carcinoembryonic Antigen
  • FAP Fibroblast Activation Protein
  • CD33 CD33.
  • the Tumor Antigen is MCSP.
  • the T cell activating bispecific antibody comprises at least one antigen binding site that is specific for Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP).
  • the T cell activating bispecific antibody comprises at least one, typically two or more antigen binding moieties that can compete with monoclonal antibody M4-3 ML2 (Sequence ID 161 and 162) or an affinity matured variant thereof for binding to an epitope of MCSP.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the variable heavy chain comprises a CDR1 of SEQ ID. NO. 4, a CDR2 of SEQ ID. NO. 5, a CDR3 of SEQ ID. NO. 6; and the variable light chain comprises a CDR1 of SEQ ID. NO. 1, a CDR2 of SEQ ID. NO. 2, and a CDR3 of SEQ ID. NO. 3.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 14; and a light chain variable region is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 13.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 161; and a light chain variable region is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 162.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain variable region sequence is an affinity matured variant of SEQ ID. NO. 161 and wherein the light chain variable region sequence is an affinity matured variant of SEQ ID. NO. 162.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 16.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 16, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 15.
  • the bispecific antibody comprises a light chain and a heavy chain of a second antibody specifically binding to MCSP, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 15, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO: 15.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID NO: 14; and a light chain variable region comprising an amino acid sequence of SEQ ID NO: 13, and wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO:15.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 13 and a variable heavy chain of SEQ ID NO: 14.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID NO: 161; and a light chain variable region comprising an amino acid sequence of SEQ ID NO: 162, and wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO:15.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 161 and a variable heavy chain of SEQ ID NO: 162.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to MCSP, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID NO: 161 or an affinity matured variant thereof; and a light chain variable region comprising an amino acid sequence of SEQ ID NO: 162 or an affinity matured variant thereof, and wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO:15.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 161 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 162 or an affinity matured variant thereof.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 158 and a variable heavy chain of SEQ ID NO: 157; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 161 and a variable heavy chain of SEQ ID NO: 162.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 158 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 157 or an affinity matured variant thereof and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 161 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 162 or an affinity matured variant thereof.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of one or more of SEQ ID. NO. 157, SEQ ID. NO. 158, SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein the variable heavy chain comprises a CDR1 of SEQ ID. NO. 4, a CDR2 of SEQ ID. NO. 5, a CDR3 of SEQ ID. NO. 6; and the variable light chain comprises a CDR1 of SEQ ID. NO. 1, a CDR2 of SEQ ID. NO. 2, and a CDR3 of SEQ ID. NO. 3.
  • the bispecific antibody comprises a third Fab fragment, comprising a a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 14; and a light chain variable region is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 13.
  • the bispecific antibody comprises a third Fab fragment, comprising a a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 161; and a light chain variable region is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID. NO. 162.
  • the bispecific antibody comprises a third Fab fragment, comprising a a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain variable region sequence is an affinity matured variant of SEQ ID. NO. 161 and wherein the light chain variable region sequence is an affinity matured variant of SEQ ID. NO. 162.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 16.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein said heavy chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 16, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein, and a a light chain and a heavy chain of a first Fab fragment specific for MCSP comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 15.
  • the bispecific antibody comprises a light chain and a heavy chain of a second antibody specifically binding to MCSP, wherein said light chain comprises a constant region comprising the amino acid sequence of SEQ ID NO: 15, and a light chain and a heavy chain of a second Fab fragment specific for CD3, and a light chain and a heavy chain of a first Fab fragment specific for MCSP comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO: 15.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID NO: 14; and a light chain variable region comprising an amino acid sequence of SEQ ID NO: 13, and wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO:15.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain variable region comprises an amino acid sequence of SEQ ID NO: 161; and a light chain variable region comprising an amino acid sequence of SEQ ID NO: 162, and wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO:15.
  • the bispecific antibody comprises a third Fab fragment, comprising a light chain and a heavy chain specifically binding to MCSP, wherein the heavy chain variable region sequence is an affinity matured variant of SEQ ID. NO. 161 and wherein the light chain variable region sequence is an affinity matured variant of SEQ ID. NO. 162, and wherein the heavy chain constant region comprises an amino acid sequence of SEQ ID NO: 16; and a light chain constant region comprising an amino acid sequence of SEQ ID NO:15.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 13 and a variable heavy chain of SEQ ID NO: 14, and a light chain and a heavy chain of a third Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 13 and a variable heavy chain of SEQ ID NO: 14.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 and a variable heavy chain of SEQ ID NO: 161, and a light chain and a heavy chain of a third Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 and a variable heavy chain of SEQ ID NO: 161.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 19 and a variable heavy chain of SEQ ID NO: 20; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 161 or an affinity matured variant thereof, and a light chain and a heavy chain of a third Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 161 or an affinity matured variant thereof.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 157 and a variable heavy chain of SEQ ID NO: 158; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 and a variable heavy chain of SEQ ID NO: 161, and a light chain and a heavy chain of a third Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 and a variable heavy chain of SEQ ID NO: 161.
  • the bispecific antibody of the invention comprises a light chain and a heavy chain of a second Fab fragment specifically binding to CD3, comprising a variable light chain of SEQ ID NO: 157 and a variable heavy chain of SEQ ID NO: 158; and a light chain and a heavy chain of a first Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 161 or an affinity matured variant thereof, and a light chain and a heavy chain of a third Fab fragment specific for MCSP, comprising a variable light chain of SEQ ID NO: 162 or an affinity matured variant thereof and a variable heavy chain of SEQ ID NO: 161 or an affinity matured variant thereof.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 161 and/or SEQ ID. NO. 162 are exchanged.
  • said bispecific antibody comprises one or more amino acid sequences selected from the group of SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO. 41 and SEQ ID NO. 43.
  • said bispecific antibody comprises SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27.
  • the T cell activating bispecific antibody comprises at least one antigen binding site that is specific for Epidermal Growth Factor Receptor (EGFR). In another embodiment the T cell activating bispecific antibody comprises at least one, typically two or more antigen binding moieties that can compete with monoclonal antibody GA201 for binding to an epitope of EGFR. See PCT publication WO 2006/082515, incorporated herein by reference in its entirety.
  • the antigen binding site that is specific for EGFR comprises the heavy chain CDR1 of SEQ ID NO: 68, the heavy chain CDR2 of SEQ ID NO: 69, the heavy chain CDR3 of SEQ ID NO: 70, the light chain CDR1 of SEQ ID NO: 71, the light chain CDR2 of SEQ ID NO: 72, and the light chain CDR3 of SEQ ID NO: 73.
  • the antigen binding site that is specific for EGFR comprises a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 74 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 75, or variants thereof that retain functionality.
  • the bispecific antibody comprises a first Fab fragment comprising an antigen binding site that is specific for EGFR comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 74 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 75, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a first and a third Fab fragment comprising an antigen binding site that is specific for EGFR comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 74 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 75, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the T cell activating bispecific antibody comprises at least one antigen binding site that is specific for Fibroblast Activation Protein (FAP). In another embodiment the T cell activating bispecific antibody comprises at least one, typically two or more antigen binding moieties that can compete with monoclonal antibody 3F2 for binding to an epitope of FAP. See European patent application no. EP10172842.6, incorporated herein by reference in its entirety.
  • the antigen binding site that is specific for FAP comprises the heavy chain CDR1 of SEQ ID NO: 76, the heavy chain CDR2 of SEQ ID NO: 77, the heavy chain CDR3 of SEQ ID NO: 78, the light chain CDR1 of SEQ ID NO: 79, the light chain CDR2 of SEQ ID NO: 80, and the light chain CDR3 of SEQ ID NO: 81.
  • the antigen binding site that is specific for FAP comprises a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 82 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 83, or variants thereof that retain functionality.
  • the bispecific antibody comprises a first Fab fragment comprising an antigen binding site that is specific for FAP comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 82 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 83, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a first and a third Fab fragment comprising an antigen binding site that is specific for FAP comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 82 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 83, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the T cell activating bispecific antibody comprises at least one antigen binding site that is specific for Carcinoembryonic Antigen (CEA). In another embodiment the T cell activating bispecific antibody comprises at least one, typically two or more antigen binding moieties that can compete with monoclonal antibody CH1A1A for binding to an epitope of CEA. In one embodiment the T cell activating bispecific antibody comprises at least one, typically two or more antigen binding moieties that can compete with monoclonal antibody CH1A1A clone 98/99 (CH1A1 (98/99) ) for binding to an epitope of CEA. See PCT patent application number PCT/EP2010/062527, incorporated herein by reference in its entirety.
  • the antigen binding site that is specific for CEA comprises the heavy chain CDR1 of SEQ ID NO: 84, the heavy chain CDR2 of SEQ ID NO: 85, the heavy chain CDR3 of SEQ ID NO: 86, the light chain CDR1 of SEQ ID NO: 87, the light chain CDR2 of SEQ ID NO: 88, and the light chain CDR3 of SEQ ID NO: 89.
  • the antigen binding site that is specific for CEA comprises a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 90 or SEQ ID NO: 159 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 91 or SEQ ID NO: 160, or variants thereof that retain functionality.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to CEA, wherein the heavy chain variable region comprises an affinity matured variant of SEQ ID NO: 159 or thereof; and a light chain variable region comprising an affinity matured variant of SEQ ID NO: 160.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 159 and/or SEQ ID. NO. 160 are exchanged.
  • the bispecific antibody comprises a first Fab fragment comprising an antigen binding site that is specific for CEA comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 90 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 91, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a first Fab fragment comprising an antigen binding site that is specific for CEA comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 159 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 160, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the bispecific antibody comprises a light chain and a heavy chain of a first Fab fragment specifically binding to CEA, wherein the heavy chain variable region comprises an affinity matured variant of SEQ ID NO: 159; and a light chain variable region comprising an affinity matured variant of SEQ ID NO: 160 and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 159 and/or SEQ ID. NO. 160 are exchanged.
  • the bispecific antibody comprises a first and a third Fab fragment comprising an antigen binding site that is specific for CEA comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 90 or SEQ ID NO: 159 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 91 or SEQ ID NO:160, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 159 and/or SEQ ID. NO. 160 are exchanged.
  • the bispecific antibody comprises a first and a third Fab fragment comprising an antigen binding site that is specific for CEA wherein the heavy chain variable region comprises an affinity matured variant of SEQ ID NO: 159; and the light chain variable region comprising an affinity matured variant of SEQ ID NO: 160.
  • Affinity matured variants in this embodiment means that independently 1, 2, 3 or 4 amino acids of SEQ ID. NO. 159 and/or SEQ ID. NO. 160 are exchanged.
  • the T cell activating bispecific antibody comprises at least one antigen binding site that is specific for CD33.
  • the antigen binding site that is specific for CD33 comprises the heavy chain CDR1 of SEQ ID NO: 92, the heavy chain CDR2 of SEQ ID NO: 93, the heavy chain CDR3 of SEQ ID NO: 94, the light chain CDR1 of SEQ ID NO: 95, the light chain CDR2 of SEQ ID NO: 96, and the light chain CDR3 of SEQ ID NO: 97.
  • the antigen binding site that is specific for CD33 comprises a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 98 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 99, or variants thereof that retain functionality.
  • the bispecific antibody comprises a first Fab fragment comprising an antigen binding site that is specific for CD33 comprising a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 98 and a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 99, or variants thereof that retain functionality, and a light chain and a heavy chain of a second Fab fragment specific for CD3 comprising one or more amino acid sequences as defined in any of the embodiments described herein.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 100, SEQ ID NO: 101 and SEQ ID NO: 102.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 151, SEQ ID NO. 152 and SEQ ID NO. 153.
  • said bispecific antibody comprises one or more amino acid sequences selected from the group of SEQ ID NO: 100, SEQ ID NO: 101, SEQ ID NO: 151, SEQ ID NO. 152 and SEQ ID NO. 153.
  • the bispecific antibody is a humanized antibody, as detailed below.
  • the bispecific antibody is a human antibody, as detailed below.
  • the present invention relates to a pharmaceutical composition comprising a bispecific antibody of the present invention.
  • the present invention relates to a bispecific antibody of the present invention for the treatment of cancer.
  • use of the bispecific antibody as a medicament is provided.
  • Preferably said use is for the treatment of cancer.
  • the present invention relates to a nucleic acid sequence comprising a sequence encoding a heavy chain of a bispecific antibody of the present invention, a nucleic acid sequence comprising a sequence encoding a light chain of a bispecific antibody of the present invention, an expression vector comprising a nucleic acid sequence of the present invention and to a prokaryotic or eukaryotic host cell comprising a vector of the present invention.
  • a method of producing an antibody comprising culturing the host cell so that the antibody is produced is provided.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, and SEQ ID NO: 67.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 107, SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, and SEQ ID NO: 112.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 113, SEQ ID NO: 114, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 117, SEQ ID NO: 118, SEQ ID NO: 119, and SEQ ID NO: 120.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 121, SEQ ID NO: 122, SEQ ID NO: 123, SEQ ID NO: 124, SEQ ID NO: 125, SEQ ID NO: 126, SEQ ID NO: 127, and SEQ ID NO: 128.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 129, SEQ ID NO: 130, SEQ ID NO: 131, SEQ ID NO: 132, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 135, and SEQ ID NO: 136.
  • the T cell activating bispecific antibody comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 105, SEQ ID NO: 106, SEQ ID NO: 137, SEQ ID NO: 138, SEQ ID NO: 139, SEQ ID NO: 140, SEQ ID NO: 141, SEQ ID NO: 142, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 154, SEQ ID NO: 155 and SEQ ID NO: 156.
  • a bispecific antibody according to any of the above embodiments may incorporate any of the features, singly or in combination, as described in Sections 1-5 below:
  • the affinity of the T cell activating bispecific antibody for 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
  • BIAcore instrument GE Healthcare
  • receptors or target proteins such as may be obtained by recombinant expression.
  • binding of T cell activating bispecific antibodies 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 bispecific antibody provided herein has a dissociation constant (KD) 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).
  • KD dissociation constant
  • KD is measured using surface plasmon resonance assays using a BIACORE®-2000 or a BIACORE®-3000 (BIAcore, Inc., Piscataway, N.J.) at 25° C. with immobilized antigen CM5 chips at ⁇ 10 response units (RU).
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N′-(3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml ( ⁇ 0.2 ⁇ M) before injection at a flow rate of 5 ⁇ l/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20TM) surfactant (PBST) at 25° C. at a flow rate of approximately 25 ⁇ l/min.
  • TWEEN-20TM polysorbate 20
  • Association rates (ka or k on ) and dissociation rates (kd or k off ) are calculated using a simple one-to-one Langmuir binding model (BIACORE® Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams.
  • the equilibrium dissociation constant (KD) is calculated as the ratio k off /k on . See, e.g., Chen et al., J. Mol. Biol. 293:865-881 (1999).
  • a bispecific antibody provided herein is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)).
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a “class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the HVR residues are derived
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the “best-fit” method (see, e.g., Sims et al. J. Immunol. 151:2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol., 151:2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci.
  • a bispecific antibody provided herein is a human antibody.
  • Human antibodies 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 antibodies may 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.
  • Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal's chromosomes.
  • the endogenous immunoglobulin loci have generally been inactivated.
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol., 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications , pp. 51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al., Proc. Natl. Acad. Sci. USA, 103:3557-3562 (2006).
  • Additional methods include those described, for example, in U.S. Pat. No. 7,189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas).
  • Human hybridoma technology Trioma technology
  • Vollmers and Brandlein, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3):185-91 (2005).
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Bispecific antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in Hoogenboom et al. in Methods in Molecular Biology 178:1-37 (O'Brien et al., ed., Human Press, Totowa, N.J., 2001) and further described, e.g., in the McCafferty et al., Nature 348:552-554; Clackson et al., Nature 352: 624-628 (1991); Marks et al., J. Mol. Biol.
  • repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al., Ann. Rev. Immunol., 12: 433-455 (1994).
  • Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • scFv single-chain Fv
  • Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas.
  • naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol., 227: 381-388 (1992).
  • Patent publications describing human antibody phage libraries include, for example: U.S. Pat. No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • amino acid sequence variants of the bispecific antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the bispecific antibody.
  • Amino acid sequence variants of a bispecific antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the bispecific antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody variants having one or more amino acid substitutions are provided.
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • Conservative substitutions are shown in Table 1 under the heading of “conservative substitutions.” More substantial changes are provided in Table 1 under the heading of “exemplary substitutions,” and as further described below in reference to amino acid side chain classes.
  • Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding or decreased immunogenicity.
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
  • Alterations may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR “hotspots,” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • HVR “hotspots” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom
  • affinity maturation diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis).
  • a secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity.
  • Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • Such alterations may be outside of HVR “hotspots” or SDRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • a useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called “alanine scanning mutagenesis” as described by Cunningham and Wells (1989) Science, 244:1081-1085.
  • a residue or group of target residues e.g., charged residues such as arg, asp, his, lys, and glu
  • a neutral or negatively charged amino acid e.g., alanine or polyalanine
  • Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions.
  • a crystal structure of an antigen-antibody complex to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution.
  • Variants may be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • cysteine engineered bispecific antibodies e.g., “thioMAbs”
  • one or more residues of a bispecific antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the bispecific antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain and A118 (EU numbering) of the heavy chain.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Pat. No. 7,521,541.
  • a bispecific antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the bispecific antibody include but are not limited to water soluble polymers.
  • Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1,3-dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof.
  • PEG polyethylene glycol
  • copolymers of ethylene glycol/propylene glycol carboxymethylcellulose
  • dextran polyvinyl alcohol
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of a bispecific antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • T cell activating bispecific antibodies 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 antibody (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. (1989); and Ausubel et al., C URRENT P ROTOCOLS IN M OLECULAR B IOLOGY , 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 antibody (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 antibody (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 â-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. promoter 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 antibody 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.
  • TPA tissue plasminogen activator
  • 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 antibody may be included within or at the ends of the T cell activating bispecific antibody (fragment) encoding polynucleotide.
  • a host cell comprising one or more polynucleotides of the invention is provided.
  • 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.
  • a host cell comprises (e.g.
  • the term “host cell” refers to any kind of cellular system which can be engineered to generate the T cell activating bispecific antibodies of the invention or fragments thereof.
  • Host cells suitable for replicating and for supporting expression of T cell activating bispecific antibodies 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 antibody for clinical applications.
  • Suitable host cells include prokaryotic microorganisms, such as E.
  • 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.
  • 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.
  • Vertebrate cells may also be used as hosts.
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • 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 BBL 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). 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.
  • a method of producing a T cell activating bispecific antibody according to the invention comprises culturing a host cell comprising a polynucleotide encoding the T cell activating bispecific antibody, as provided herein, under conditions suitable for expression of the T cell activating bispecific antigen binding molecule, and recovering the T cell activating bispecific antibody from the host cell (or host cell culture medium).
  • the components of the T cell activating bispecific antibody are genetically fused to each other.
  • T cell activating bispecific antibody 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 antibodies 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 antibodies 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 to McCafferty).
  • any animal species of antibody, antibody fragment, antigen binding domain or variable region can be used in the T cell activating bispecific antibodies 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 antibody 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 bispecific antibodies of 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 antibody 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)).
  • ELISA enzyme-linked immunosorbent assay
  • 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.
  • epitope e.g. a linear or a conformational epitope
  • 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.).
  • immobilized antigen e.g.
  • CD3 is incubated in a solution comprising a first labeled antibody that binds to the antigen (e.g. V9 antibody) 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.
  • 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.
  • T cell activating bispecific antibodies 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.
  • an antibody, ligand, receptor or antigen can be used to which the T cell activating bispecific antibody binds.
  • 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 antibody essentially as described in the Examples.
  • the purity of the T cell activating bispecific antibodies can be determined by any of a variety of well known analytical methods including gel electrophoresis, high pressure liquid chromatography, and the like.
  • Bispecific antibodies 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.
  • a bispecific antibody of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, Western blot, etc.
  • competition assays may be used to identify an antibody that competes with a specific anti-TA antibody or an antibody specific for aT-cell activating antigen for binding to the Tumor Antigen (TA) or a T-cell activating antigen respectively.
  • TA Tumor Antigen
  • a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by a specific anti-TA antibody or an antibody specific for aT-cell activating antigen.
  • epitope e.g., a linear or a conformational epitope
  • 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.).
  • assays are provided for identifying bispecific antibodies that bind to a T-cell activating antigen and a Tumor Antigen (TA) thereof having biological activity.
  • Biological activity may include, e.g., lysis of targeted cells or induction of apoptosis.
  • Antibodies having such biological activity in vivo and/or in vitro are also provided.
  • a bispecific antibody of the invention is tested for such biological activity.
  • Assays for detecting cell lysis e.g. by measurement of LDH release
  • apoptosis e.g. using the TUNEL assay
  • the invention also provides immunoconjugates comprising a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody is conjugated to one or more drugs, including but not limited to a maytansinoid (see U.S. Pat. Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 B1); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF) (see U.S. Pat. Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S. Pat. Nos.
  • ADC antibody-drug conjugate
  • drugs including but not limited to a maytansinoid (see U.S. Pat. Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 B1); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and
  • an immunoconjugate comprises a bispecific antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxi
  • an immunoconjugate comprises a bispecific antibody as described herein conjugated to a radioactive atom to form a radioconjugate.
  • a variety of radioactive isotopes are available for the production of radioconjugates. Examples include At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • the radioconjugate When used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or I123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123 again, iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of a bispecific antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCl), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis(p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238:1098 (1987).
  • Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.
  • the linker may be a “cleavable linker” facilitating release of a cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52:127-131 (1992); U.S. Pat. No. 5,208,020) may be used.
  • the immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, Ill., U.S.A).
  • cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC
  • any of the bispecific antibodies that bind to a T-cell activating antigen and a Tumor Antigen (TA) provided herein is useful for detecting the presence of a T-cell activating antigen and/or a Tumor Antigen (TA) in a biological sample.
  • TA Tumor Antigen
  • the term “detecting” as used herein encompasses quantitative or qualitative detection.
  • a biological sample comprises a cell or tissue.
  • a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of a T-cell activating antigen 3 and/or a Tumor Antigen (TA) in a biological sample is provided.
  • the method comprises contacting the biological sample with a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) as described herein under conditions permissive for binding of the bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) to a T-cell activating antigen and/or a Tumor Antigen (TA), and detecting whether a complex is formed between the bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) and a T-cell activating antigen and/or a Tumor Antigen (TA).
  • TA Tumor Antigen
  • Such method may be an in vitro or in vivo method.
  • a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen is used to select subjects eligible for therapy with a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA), e.g. where a Tumor Antigen (TA) is a biomarker for selection of patients.
  • Exemplary disorders that may be diagnosed using an antibody of the invention include cancer.
  • labeled bispecific antibodies that bind to a T-cell activating antigen and a Tumor Antigen (TA) are provided.
  • Labels include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction.
  • Exemplary labels include, but are not limited to, the radioisotopes 32 P, 14 C, 125 I, 3 H, and 131 I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (U.S. Pat. No.
  • luciferin 2,3-dihydrophthalazinediones
  • horseradish peroxidase HRP
  • alkaline phosphatase alkaline phosphatase
  • ⁇ -galactosidase glucoamylase
  • lysozyme saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase
  • heterocyclic oxidases such as uricase and xanthine oxidase, coupled with an enzyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP, lactoperoxidase, or microperoxidase, biotin/avidin, spin labels, bacteriophage labels, stable free radicals, and the like.
  • compositions of a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) as described herein are prepared by mixing such bispecific antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers ( Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • TA Tumor Antigen
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and 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, arg
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rHuPH20 HYLENEX®, Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody formulations are described in U.S. Pat. No. 6,267,958.
  • Aqueous antibody formulations include those described in U.S. Pat. No. 6,171,586 and WO2006/044908, the latter formulations including a histidine-acetate buffer.
  • the formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • 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. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • any of the bispecific antibodies that bind to a T-cell activating antigen and a Tumor Antigen (TA) provided herein may be used in therapeutic methods.
  • a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) for use as a medicament is provided.
  • a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) use in treating cancer is provided.
  • a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) for use in a method of treatment is provided.
  • the invention provides a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) for use in a method of treating an individual having cancer comprising administering to the individual an effective amount of the bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA).
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • An “individual” according to any of the above embodiments is preferably a human.
  • the invention provides for the use of a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) in the manufacture or preparation of a medicament.
  • the medicament is for treatment of cancer.
  • the medicament is for use in a method of treating cancer comprising administering to an individual having cancer an effective amount of the medicament.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • An “individual” according to any of the above embodiments may be a human.
  • the invention provides a method for treating cancer.
  • the method comprises administering to an individual having cancer an effective amount of a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA).
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below.
  • An “individual” according to any of the above embodiments may be a human.
  • the invention provides pharmaceutical formulations comprising any of the bispecific antibodies that bind to a T-cell activating antigen and a Tumor Antigen (TA) provided herein, e.g., for use in any of the above therapeutic methods.
  • a pharmaceutical formulation comprises any of the bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA) provided herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation comprises any of the bispecific antibodies that bind to a T-cell activating antigen and a Tumor Antigen (TA) provided herein and at least one additional therapeutic agent, e.g., as described below.
  • bispecific antibodies of the invention can be used either alone or in combination with other agents in a therapy.
  • a bispecific antibody of the invention may be co-administered with at least one additional therapeutic agent.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Bispecific antibodies of the invention can also be used in combination with radiation therapy.
  • a bispecific antibody of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • Bispecific antibodies 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.
  • the bispecific antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These 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.
  • a bispecific antibody 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 type of antibody, the severity and course of the disease, whether the bispecific antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the bispecific antibody, and the discretion of the attending physician.
  • the antibody 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 bispecific antibody 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. 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 bispecific antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 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 bispecific antibody).
  • 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.
  • any of the above formulations or therapeutic methods may be carried out using an immunoconjugate of the invention in place of or in addition to a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA).
  • TA Tumor Antigen
  • 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 bispecific antibody 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 bispecific antibody 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
  • any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to a bispecific antibody that binds to a T-cell activating antigen and a Tumor Antigen (TA).
  • TA Tumor Antigen
  • 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 is driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS.
  • each vector contains an EBV OriP sequence.
  • the molecule is produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using a calcium phosphate-transfection. Exponentially growing HEK293-EBNA cells are transfected by the calcium phosphate method. Alternatively, HEK293-EBNA cells growing in suspension are transfected by polyethylenimine. The cells are transfected with the corresponding expression vectors in a 1:1:1 ratio (“vector CH1-VH-CK-VH”: “vector light chain”: “vector light chain CH1-VL”).
  • transfection using calcium phosphate cells are grown as adherent monolayer cultures in T-flasks using DMEM culture medium supplemented with 10% (v/v) FCS, and are transfected when they are between 50 and 80% confluent.
  • DMEM culture medium supplemented with 10% (v/v) FCS
  • For the transfection of a T150 flask 15 million cells are seeded 24 hours before transfection in 25 ml DMEM culture medium supplemented with FCS (at 10% v/v final), and cells are placed at 37° C. in an incubator with a 5% CO2 atmosphere overnight.
  • a solution of DNA, CaCl2 and water is prepared by mixing 94 ⁇ g total plasmid vector DNA divided in the corresponding ratio, water to a final volume of 469 ⁇ l and 469 ⁇ l of a 1 M CaCl2 solution.
  • 938 ⁇ l of a 50 mM HEPES, 280 mM NaCl, 1.5 mM Na2HPO4 solution at pH 7.05 are added, mixed immediately for 10 s and left to stand at room temperature for 20 s.
  • the suspension is diluted with 10 ml of DMEM supplemented with 2% (v/v) FCS, and added to the T150 in place of the existing medium.
  • transfection medium 13 ml of transfection medium are added.
  • the cells are incubated at 37° C., 5% CO2 for about 17 to 20 hours, then medium is replaced with 25 ml DMEM, 10% FCS.
  • the conditioned culture medium is harvested approx. 7 days post-media exchange by centrifugation for 15 min at 210 ⁇ g, the solution is sterile filtered (0.22 ⁇ m filter) and sodium azide in a final concentration of 0.01% (w/v) is added, and kept at 4° C.
  • 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 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. After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature.
  • 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 (Lonza) 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 ⁇ m filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C.
  • the secreted protein is purified from cell culture supernatants by affinity chromatography using Protein A and Protein G affinity chromatography, followed by a size exclusion chromatographic step.
  • an additional wash step is necessary to wash only the HiTrap Protein G HP column using at least 8 column volume 20 mM sodium phosphate, 20 mM sodium citrate, pH 7.5.
  • the target protein is eluted from HiTrap Protein G HP column using a step gradient with 7 column volume 8.8 mM formic acid, pH 3.0. Protein solution is neutralized by adding 1/10 of 0.5 M sodium phosphate, pH 8.0. Target protein is concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 25 mM potassium phosphate, 125 mM sodium chloride, 100 mM glycine solution of pH 6.7.
  • the protein concentration of purified protein samples is 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 antibodies are analyzed by SDS-PAGE in the presence and absence of a reducing agent (5 mM 1,4-dithiotreitol) and staining with Coomassie (SimpleBlueTM SafeStain from Invitrogen).
  • a reducing agent 5 mM 1,4-dithiotreitol
  • Coomassie SimpleBlueTM SafeStain from Invitrogen.
  • the NuPAGE® Pre-Cast gel system (Invitrogen, USA) is used according to the manufacturer's instruction (4-12% Tris-Acetate gels or 4-12% Bis-Tris).
  • the aggregate content of antibody samples is analyzed using a Superdex 200 10/300GL analytical size-exclusion column (GE Healthcare, Sweden) in 2 mM MOPS, 150 mM NaCl, 0.02% (w/v) NaN3, pH 7.3 running buffer at 25° C.
  • This molecule is further referred to as Fab (MCSP)-Crossfab (CD3) or hu Fab (MCSP)-Crossfab (CD3).
  • 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 is driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS.
  • each vector contains an EBV OriP sequence.
  • the molecule is produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using a calcium phosphate-transfection. Exponentially growing HEK293-EBNA cells are transfected by the calcium phosphate method. Alternatively, HEK293-EBNA cells growing in suspension are transfected by polyethylenimine. The cells are transfected with the corresponding expression vectors in a 1:2:1 ratio (“vector CH1-VH-CH1-VH-CK-VH”: “vector light chain”: “vector light chain CH1-VL”).
  • transfection using calcium phosphate cells are grown as adherent monolayer cultures in T-flasks using DMEM culture medium supplemented with 10% (v/v) FCS, and are transfected when they are between 50 and 80% confluent.
  • DMEM culture medium supplemented with 10% (v/v) FCS
  • For the transfection of a T150 flask 15 million cells are seeded 24 hours before transfection in 25 ml DMEM culture medium supplemented with FCS (at 10% v/v final), and cells are placed at 37° C. in an incubator with a 5% CO2 atmosphere overnight.
  • a solution of DNA, CaCl2 and water is prepared by mixing 94 ⁇ g total plasmid vector DNA divided in the corresponding ratio, water to a final volume of 469 ⁇ l and 469 ⁇ l of a 1 M CaCl2 solution.
  • 938 ⁇ l of a 50 mM HEPES, 280 mM NaCl, 1.5 mM Na2HPO4 solution at pH 7.05 are added, mixed immediately for 10 s and left to stand at room temperature for 20 s.
  • the suspension is diluted with 10 ml of DMEM supplemented with 2% (v/v) FCS, and added to the T150 in place of the existing medium.
  • transfection medium 13 ml of transfection medium are added.
  • the cells are incubated at 37° C., 5% CO2 for about 17 to 20 hours, then medium is replaced with 25 ml DMEM, 10% FCS.
  • the conditioned culture medium is harvested approx. 7 days post-media exchange by centrifugation for 15 min at 210 ⁇ g, the solution is sterile filtered (0.22 ⁇ m filter) and sodium azide in a final concentration of 0.01% (w/v) is added, and kept at 4° C.
  • HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium.
  • HEK293 EBNA cells For the production in 500 ml shake flask 400 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 200 ⁇ g DNA. 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.
  • the secreted protein is purified from cell culture supernatants by affinity chromatography using Protein A and Protein G affinity chromatography, followed by a size exclusion chromatographic step.
  • the target protein is eluted from HiTrap Protein G HP column using a step gradient with 7 column volume 8.8 mM formic acid, pH 3.0. Protein solution is neutralized by adding 1/10 of 0.5 M sodium phosphate, pH 8.0. Target protein is concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 25 mM potassium phosphate, 125 mM sodium chloride, 100 mM glycine solution of pH 6.7.
  • the protein concentration of purified protein samples is 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 antibodies are analyzed by SDS-PAGE in the presence and absence of a reducing agent (5 mM 1,4-dithiotreitol) and staining with Coomassie (SimpleBlueTM SafeStain from Invitrogen).
  • a reducing agent 5 mM 1,4-dithiotreitol
  • Coomassie SimpleBlueTM SafeStain from Invitrogen.
  • the NuPAGE® Pre-Cast gel system (Invitrogen, USA) is used according to the manufacturer's instruction (4-12% Tris-Acetate gels or 4-12% Bis-Tris).
  • the aggregate content of antibody samples is analyzed using a Superdex 200 10/300GL analytical size-exclusion column (GE Healthcare, Sweden) in 2 mM MOPS, 150 mM NaCl, 0.02% (w/v) NaN 3 , pH 7.3 running buffer at 25° C. and compared with prior art antibody fragment (scFv)2 (results see table below).
  • HMW High Molecular Weight
  • FIGS. 4 and 5 This molecule is further referred to as Fab (MCSP)-Fab (MCSP)-Crossfab (CD3) or hu Fab (MCSP)-Fab (MCSP)-Crossfab (CD3).
  • MCSP Fab-Fab
  • CD3 hu Fab
  • MCSP MCSP-Fab
  • CD3 hu Fab
  • VHCH1(MCSP)-VLCH1(CD3 V9 )-VHCH1(MCSP) SEQ ID NO:27
  • FIGS. 6 and 7 This molecule is further referred to as Fab (MCSP)-Fab (MCSP)-Crossfab (CD3) or hu Fab (MCSP)-Fab (MCSP)-Crossfab (CD3).
  • MCSP Fab
  • MCSP Fab
  • CD3 hu Fab
  • MCSP MCSP-Fab
  • This molecule is further referred to as murine Crossfab (CD3)-Fab (MCSP)
  • 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 is driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS.
  • each vector contains an EBV OriP sequence.
  • the molecule is produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using a calcium phosphate-transfection. Exponentially growing HEK293-EBNA cells are transfected by the calcium phosphate method. Alternatively, HEK293-EBNA cells growing in suspension are transfected by polyethylenimine. The cells are transfected with the corresponding expression vectors in a 1:1:1 ratio (“vector CH1-VH-CK-VH”: “vector light chain”: “vector light chain CH1-VL”).
  • transfection using calcium phosphate cells are grown as adherent monolayer cultures in T-flasks using DMEM culture medium supplemented with 10% (v/v) FCS, and are transfected when they are between 50 and 80% confluent.
  • DMEM culture medium supplemented with 10% (v/v) FCS
  • For the transfection of a T150 flask 15 million cells are seeded 24 hours before transfection in 25 ml DMEM culture medium supplemented with FCS (at 10% v/v final), and cells are placed at 37° C. in an incubator with a 5% CO2 atmosphere overnight.
  • a solution of DNA, CaCl2 and water is prepared by mixing 94 ⁇ g total plasmid vector DNA divided in the corresponding ratio, water to a final volume of 469 ⁇ l and 469 ⁇ l of a 1 M CaCl2 solution.
  • 938 ⁇ l of a 50 mM HEPES, 280 mM NaCl, 1.5 mM Na2HPO4 solution at pH 7.05 are added, mixed immediately for 10 s and left to stand at room temperature for 20 s.
  • the suspension is diluted with 10 ml of DMEM supplemented with 2% (v/v) FCS, and added to the T150 in place of the existing medium.
  • transfection medium 13 ml of transfection medium are added.
  • the cells are incubated at 37° C., 5% CO2 for about 17 to 20 hours, then medium is replaced with 25 ml DMEM, 10% FCS.
  • the conditioned culture medium is harvested approx. 7 days post-media exchange by centrifugation for 15 min at 210 ⁇ g, the solution is sterile filtered (0.22 ⁇ m filter) and sodium azide in a final concentration of 0.01% (w/v) is added, and kept at 4° C.
  • 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 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. After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature.
  • 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 (LONZA) 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 ⁇ m filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C.
  • LONZA 7% Feed 1
  • the secreted protein is purified from cell culture supernatants by affinity chromatography using Protein A and ProteinG affinity chromatography, followed by a size exclusion chromatographic step.
  • the target protein is eluted from HiTrap ProteinG HP column using a step gradient with 7 column volume 8.8 mM formic acid, pH 3.0. Protein solution is neutralized by adding 1/10 of 0.5M sodium phosphate, pH 8.0. Target protein is concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 25 mM potassium phosphate, 125 mM sodium chloride, 100 mM glycine solution of pH 6.7.
  • the protein concentration of purified protein samples is 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 antibodies are analyzed by SDS-PAGE in the presence and absence of a reducing agent (5 mM 1,4-dithiotreitol) and staining with Coomassie (SimpleBlueTM SafeStain from Invitrogen).
  • a reducing agent 5 mM 1,4-dithiotreitol
  • Coomassie SimpleBlueTM SafeStain from Invitrogen.
  • the NuPAGE® Pre-Cast gel system (Invitrogen, USA) is used according to the manufacturer's instruction (4-12% Tris-Acetate gels or 4-12% Bis-Tris).
  • the aggregate content of antibody samples is analyzed using a Superdex 200 10/300GL analytical size-exclusion column (GE Healthcare, Sweden) in 2 mM MOPS, 150 mM NaCl, 0.02% (w/v) NaN3, pH 7.3 running buffer at 25° C.
  • This molecule is further referred to as Fab(CD33)-CrossFab (CD3) or hu Fab(CD33)-CrossFab (CD3).
  • variable region of heavy and light chain DNA sequences have been subcloned in frame into the respective recipient mammalian expression vector.
  • the antibody expression is driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS.
  • each vector contains an EBV OriP sequence.
  • the molecule is produced by transfecting HEK293-EBNA cells with the mammalian expression vector using polyethylenimine.
  • 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 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. After addition of 540 ⁇ l PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature.
  • 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 (LONZA) are 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 ⁇ m filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C.
  • LONZA 7% Feed 1
  • the secreted protein is purified from cell culture supernatants by affinity chromatography using Immobilized Metal Ion Affinity Chromatography (IMAC), followed by a size exclusion chromatographic step.
  • IMAC Immobilized Metal Ion Affinity Chromatography
  • Prior first purification step disturbing components from the supernatant are removed by diafiltration using the tangential flow filtration system Sarcojet (Sartorius) equipped with a 5.000 MWCO membrane (Sartocon Slice Cassette, Hydrosart; Sartorius).
  • Supernatant is concentrated to 210 ml and subsequently diluted in 1 l 20 mM sodium phosphate, 500 mM sodium chloride, pH 6.5.
  • the protein solution is concentrated again to 210 ml. This process is repeated twice to ensure a complete buffer exchange.
  • Target protein is eluted in 2 column volume 20 mM sodium phosphate, 500 mM sodium chloride, 125 mM imidazole, pH 6.5. Column is washed subsequently with 20 mM sodium phosphate, 500 mM sodium chloride, 250 mM imidazole, pH 6.5.
  • Target protein is concentrated prior loading on a HiLoad Superdex 75 column (GE Healthcare) equilibrated with 25 mM KH 2 PO 4 , 125 mM NaCl, 200 mM Arginine, pH 6.7. Yields, aggregate content after the first purification step and final monomer content is shown in the table above. Comparison of the aggregate content after the first purification step indicates the superior stability of the Fab-Crossfab construct in contrast to the (scFv)2.
  • the protein concentration of purified protein samples is 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 antibodies are analyzed by SDS-PAGE in the presence and absence of a reducing agent (5 mM 1,4-dithiotreitol) and staining with Coomassie (SimpleBlueTM SafeStain from Invitrogen).
  • a reducing agent 5 mM 1,4-dithiotreitol
  • Coomassie SimpleBlueTM SafeStain from Invitrogen.
  • the NuPAGE® Pre-Cast gel system (Invitrogen, USA) is used according to the manufacturer's instruction (4-12% Tris-Acetate gels or 4-12% Bis-Tris).
  • the aggregate content of antibody samples is analyzed using a Superdex 75 10/300GL analytical size-exclusion column (GE Healthcare, Sweden) in 2 mM MOPS, 150 mM NaCl, 0.02% (w/v) NaN3, pH 7.3 running buffer at 25° C.
  • FIG. 21 A schematic drawing of the (scFv)2 molecule is shown in FIG. 21 .
  • PBMCs Peripheral blood mononuclar cells
  • 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 ⁇ l cold buffer per 10 Mio cells (PBS with 0.5% BSA, 2 mM EDTA—sterile filtered) and incubated with 10 ⁇ l Biotin-Antibody Cocktail per 10 Mio cells for 10 min at 4° C.
  • 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).
  • 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 manufacturers' instructions.
  • a GentleMACS C-tube Miltenyi Biotech #130-093-237
  • MACS buffer PBS+0.5% BSA+2 mM EDTA
  • the cell suspension was passed through a pre-separation filter to get rid-off remaining undissociated tissue particles. After centrifugation at 400 g for 4 minutes at 4° C., ACK Lysis Buffer was added to to lyse red blood cells (incubation for 5 minutes 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 manufacturers' instructions. The resulting T cell population was counted automatically (ViCell) and used immediately for further assays.
  • Bispecific constructs targeting CD3 on human, or mouse T cells and human on tumor cells are analyzed by a LDH release assay regarding their potential to induce T cell-mediated apoptosis of target cells.
  • target cells human Colo-38, human MDA-MB-435, human melanoma MV-3 or murine B16/F10-huMCSP Fluc 2 clone 48 cells, all expressing human MCSP
  • MCSP Cell Dissociation Buffer
  • MCSP is trypsin-sensitive
  • trypsin and then plated the day before
  • washed and resuspendend in the appropriate cell culture medium see detailed description of the different figures.
  • 20 000-30 000 cells per well are plated in a round-bottom 96-well-plate and the respective antibody dilution was added as indicated (triplicates). Effector cells were added to obtain a final E:T ratio of 5:1 (for human pan T cells), 10:1 (for human PBMCs).
  • PHA-M a mixture of isolectins, isolated from Phaseolus vulgaris , was used as a mitogenic stimulus to induce human or cynomolgus T cell activation.
  • a 5% solution of “rat T-Stim with ConA” (BD #354115) was used as a positive control for T cell activation.
  • LDH release of apoptotic/necrotic target cells into the supernatant is measured with the LDH detection kit (Roche Applied Science, #11 644 793 001), according to the manufacturer's instructions.
  • MCSP Purified Fab
  • CD3 Fab
  • MCSP Fab
  • MCSP Fab
  • CD3 Fab
  • MCSP Fab
  • CD3 Fab
  • scFv antiMCSP/anti huCD3e
  • huMCSP-expressing MDA-MB-435 human melanoma target cells are harvested with Cell Dissociation Buffer, washed and resuspendend in AIM-V medium (Invitrogen #12055-091). 30 000 cells per well were plated in a round-bottom 96-well-plate and the respective antibody dilution was added at the indicated concentrations. All constructs and controls were adjusted to the same molarity.
  • Human pan T effector cells were added to obtain a final E:T ratio of 5:1.
  • 1 ⁇ g/ml PHA-M (Sigma #L8902) was used.
  • LDH release of apoptotic/necrotic target cells into the supernatant was measured with the LDH detection kit (Roche Applied Science, #11 644 793 001), according to the manufacturer's instructions.
  • the constructs with bivalent MCSP-targeting show comparable cytotoxic activity compared to the (scFv)2 (antiMCSP/anti huCD3e) construct, whereas the Fab (MCSP)-Crossfab (CD3) construct with monovalent MCSP binding is clearly less potent.
  • MCSP Purified Fab
  • MCSP Fab
  • CD3 Chromatin-binding protein
  • scFv antiMCSP/anti huCD3e
  • huMCSP-expressing MDA-MB-435 human melanoma target cells are harvested with Cell Dissociation Buffer, washed and resuspendend in AIM-V medium (Invitrogen #12055-091). 30 000 cells per well were plated in a round-bottom 96-well-plate and the respective antibody dilution was added at the indicated concentrations. All constructs and controls were adjusted to the same molarity.
  • Human pan T effector cells were added to obtain a final E:T ratio of 5:1.
  • 5 ⁇ g/ml PHA-M (Sigma #L8902) was used.
  • LDH release of apoptotic/necrotic target cells into the supernatant was measured with the LDH detection kit (Roche Applied Science, #11 644 793 001), according to the manufacturer's instructions.
  • the Fab (MCSP)-Fab (MCSP)-Crossfab (CD3) induces apoptosis in target cells at least comparably good as the (scFv)2 (antiMCSP/anti huCD3e) molecule.
  • MCSP Purified Fab
  • MCSP Fab
  • CD3 Chromatin-binding protein
  • scFv antiMCSP/anti huCD3e
  • huMCSP-expressing MV-3 human melanoma target cells are harvested with trypsin on the day before the LDH release assay was started. Cell were washed and resuspendend in the appropriate cell culture medium. 30 000 cells per well were plated in a round-bottom 96-well-plate. The next day, the supernatant was discarded and 100 ⁇ l/well AIM-V medium (Invitrogen #12055-091), as well as the respective antibody dilution were added at the indicated concentrations. All constructs and controls were adjusted to the same molarity.
  • Human PBMC effector cells were added to obtain a final E:T ratio of 10:1.
  • 5 ⁇ g/ml PHA-M (Sigma #L8902) was used.
  • LDH release of apoptotic/necrotic target cells into the supernatant was measured with the LDH detection kit (Roche Applied Science, #11 644 793 001), according to the manufacturer's instructions.
  • the Fab (MCSP)-Fab (MCSP)-Crossfab (CD3) induces apoptosis in target cells at least comparably good as the (scFv)2 (antiMCSP/anti huCD3e) molecule.
  • huMCSP-expressing B16/F10-huMCSP Fluc2 clone 48 tumor target cells are harvested with Cell Dissociation Buffer, washed and resuspendend in RPMI1640 medium, including 1 ⁇ NEAA, 10 mM Hepes, 50 ⁇ m 2-b-ME and 1 mM sodium pyruvate.
  • Murine pan T effector cells isolated from splenocytes (C57BL/6 mice) were added to obtain a final E:T ratio of 10:1.
  • LDH release of apoptotic/necrotic target cells into the supernatant was measured with the LDH detection kit (Roche Applied Science, #11 644 793 001), according to the manufacturer's instructions.
  • the bispecific construct induces concentration-dependent LDH release from target cells, comparable to the positive control with “T Cell Stim with ConA”.
  • huMCSP-expressing B16/F10-huMCSP Fluc2 clone 48 tumor target cells are harvested with Cell Dissociation Buffer, washed and resuspendend in RPMI1640 medium, including 1 ⁇ NEAA, 10 mM Hepes, 50 ⁇ M 2-b-ME and 1 mM sodium pyruvate.
  • Murine pan T effector cells isolated from splenocytes (C57BL/6 mice) were added to obtain a final E:T ratio of 10:1. To assess the level of hyperactivation of murine T cells in the absence of target cells, control wells with 50 nM bispecific construct and T cells were plated accordingly.
  • LDH release of apoptotic/necrotic target cells into the supernatant was measured with the LDH detection kit (Roche Applied Science, #11 644 793 001), according to the manufacturer's instructions.
  • the bispecific construct induces strong LDH release from target cells.
  • LDH reflecting hyperactivation of T cells
  • untreated murine T cells co-incubated with target cells.
  • None of the control IgGs induces LDH release of target cells.
  • human PBMCs were isolated from Buffy Coats and 0.3 Mio cells per well were plated into a round-bottom 96-well plate.
  • 280 ⁇ l whole blood from a healthy donor were plated per well of a deep-well 96-well plate.
  • Tumor target cells e.g. MDA-MB-435 cells for CD3-MCSP-bispecific constructs
  • Bispecific constructs and controls were added as indicated. After an incubation of up to 24 h at 37° C., 5% CO2, the assay plate was centrifuged for 5 min at 350 g and the supernatant was transferred into a new deep-well 96-well-plate for the subsequent analysis.
  • the CBA analysis was performed according to manufacturers' instructions for FACS Cantoll, using the combination of the following CBA Flex Sets: human granzyme B (BD 560304), human IFN- ⁇ Flex Set (BD 558269), human TNF Flex Set (BD 558273), human IL-10 Flex Set (BD 558274), human IL-6 Flex Set (BD 558276), human IL-4 Flex Set (BD 558272).
  • human granzyme B BD 560304
  • human IFN- ⁇ Flex Set BD 558269
  • human TNF Flex Set BD 558273
  • human IL-10 Flex Set BD 558274
  • human IL-6 Flex Set BD 558276
  • human IL-4 Flex Set BD 558272
  • the CBA analysis was performed according to manufacturers' instructions for FACS Cantoll, using the combination of the following CBA Flex Sets: human granzyme B (BD 560304), human IFN- ⁇ Flex Set (BD 558269), human TNF Flex Set (BD 558273), human IL-10 Flex Set (BD 558274), human IL-6 Flex Set (BD 558276), human IL-4 Flex Set (BD 558272).
  • human granzyme B BD 560304
  • human IFN- ⁇ Flex Set BD 558269
  • human TNF Flex Set BD 558273
  • human IL-10 Flex Set BD 558274
  • human IL-6 Flex Set BD 558276
  • human IL-4 Flex Set BD 558272
  • FIG. 15 depicts different cytokine levels, that were measured in the supernatant of whole blood after treatment with 1 nM of different CD3-MCSP bispecific constructs (Fab (MCSP)-Fab (MCSP)-Crossfab (CD3) and the (scFv)2 (antiMCSP/anti huCD3e)) in the presence (A, B) or absence (C,D) of Colo-38 tumor cells for 24 hours.
  • Fab MCSP
  • MCSP CD3-MCSP bispecific constructs
  • CD3 CD3-MCSP bispecific constructs
  • scFv)2 antiMCSP/anti huCD3e
  • the levels of granzyme B increased enormously upon activation of T cells in the presence of target cells.
  • the (scFv)2 (antiMCSP/anti huCD3e) construct elevated the levels of TNF and IFNgamma, as well as granzyme B in the presence of target cells (A and B) a bit more compared to the other bispecific construct.
  • Th2 cytokines IL-10 and IL-4
  • the purified huMCSP-muCD3-targeting bispecific molecule as murine Crossfab (CD3)-Fab (MCSP)-Fab (MCSP) was tested by flow cytometry for its potential to up-regulate the late activation marker CD25 on CD8+ T cells in the presence of human MCSP-expressing tumor cells.
  • MCSP-positive B16/F10-huMCSP Fluc2 clone 48 tumor cells were harvested with Cell Dissociation buffer, counted and checked for viability.
  • Cells were adjusted to 0.3 ⁇ 10 6 (viable) cells per ml in RPMI1640 medium (including 1 ⁇ NEAA, 10 mM Hepes, 50 ⁇ m 2-b-ME, 1 mM sodium pyruvate), 100 ⁇ l of this cell suspension were pipetted per well into a round-bottom 96-well plate (as indicated). 50 ⁇ l of the (diluted) bispecific construct was added to the cell-containing wells to obtain a final concentration of 50 nM.
  • Human murine T effector cells were isolated from splenocytes (C57BL/6 mice) and adjusted to 3 ⁇ 10 6 (viable) cells per ml in AIM-V medium. 50 ⁇ l of this cell suspension was added per well of the assay plate (see above) to obtain a final E:T ratio of 10:1. To analyze, if the bispecific construct is able to activate T cells only in the presence of target cells, expressing huMCSP, wells were included that contained 50 nM of the respective bispecific molecule, as well as T effector, but no target cells.
  • cells were centrifuged (5 min, 350 ⁇ g) and washed twice with 150 ⁇ l/well PBS, including 0.1% BSA.
  • CD8a rat IgG2a; clone 53-6.7; BioLegend #100712
  • CD25 rat IgG2b; clone 3C7; BD #553075
  • FIG. 16 shows that the as murine Crossfab (CD3)-Fab (MCSP)-Fab (MCSP) construct induces up-regulation of CD25 in the presence of target cells only.
  • primary human PBMCs isolated as described above were incubated with the indicated concentrations of bispecific constructs for at least 24 h in the presence or absence of tumor antigen-positive target cells.
  • ⁇ ективное PBMCs 0.3 million primary human PBMCs were plated per well of a flat-bottom 96-well plate, containing the huMCSP-positive target cells (MV-3 tumor cells) or medium.
  • the final effector to target cell (E:T) ratio was 10:1.
  • the cells were incubated with the indicated concentration of the CD3-MCSP bispecific constructs (Fab (MCSP)-Crossfab (CD3); designated as “1+1 non-Fc”, and the (scFv)2 (antiMCSP/anti huCD3e) reference molecule (designated as“(scFv)2”) for the indicated incubation times at 37° C., 5% CO2.
  • the effector cells were stained for CD8, and the early activation marker CD69 or the late activation marker CD25 and analyzed by FACS Cantoll.
  • FIG. 20 shows the result of this experiment.
US13/591,024 2011-08-23 2012-08-21 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use Abandoned US20130058936A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/269,950 US11639397B2 (en) 2011-08-23 2014-05-05 Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP11178410 2011-08-23
EP11178410.4 2011-08-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/269,950 Continuation US11639397B2 (en) 2011-08-23 2014-05-05 Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use

Publications (1)

Publication Number Publication Date
US20130058936A1 true US20130058936A1 (en) 2013-03-07

Family

ID=46704675

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/591,024 Abandoned US20130058936A1 (en) 2011-08-23 2012-08-21 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
US14/269,950 Active US11639397B2 (en) 2011-08-23 2014-05-05 Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/269,950 Active US11639397B2 (en) 2011-08-23 2014-05-05 Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use

Country Status (11)

Country Link
US (2) US20130058936A1 (de)
EP (1) EP2747781B1 (de)
JP (2) JP6159724B2 (de)
KR (2) KR101870555B1 (de)
CN (2) CN103889452B (de)
BR (1) BR112014004168A2 (de)
CA (1) CA2844540C (de)
HK (1) HK1249529A1 (de)
MX (1) MX356947B (de)
RU (1) RU2605390C2 (de)
WO (1) WO2013026839A1 (de)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9309306B2 (en) 2011-08-23 2016-04-12 Roche Glycart Ag Anti-MCSP antibodies
WO2016138491A1 (en) * 2015-02-27 2016-09-01 Icell Gene Therapeutics Llc Chimeric antigen receptors (cars) targeting hematologic malignancies, compositions and methods of use thereof
WO2016164920A1 (en) * 2015-04-09 2016-10-13 Cornell University Gene therapy to prevent reactions to allergens
WO2016168766A1 (en) * 2015-04-15 2016-10-20 The California Institute For Biomedical Research Optimized chimeric receptor t cell switches and uses thereof
WO2017075045A3 (en) * 2015-10-30 2017-06-08 Mayo Foundation For Medical Education And Research Antibodies to b7-h1
WO2017127499A1 (en) 2016-01-22 2017-07-27 Janssen Biotech, Inc. Anti-ror1 antibodies, ror1 x cd3 bispecific antibodies, and methods of using the same
WO2017180555A1 (en) * 2016-04-11 2017-10-19 The Trustees Of Columbia University In The City Of New York Humanized anti-rage antibody
US20170327580A1 (en) * 2014-12-03 2017-11-16 Engmab Ag Bispecific antibodies against cd3 epsilon and bcma for use in treatment of diseases
US9879088B2 (en) 2012-08-14 2018-01-30 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
CN107921127A (zh) * 2015-05-22 2018-04-17 纪念斯隆-凯特琳癌症中心 对于prame肽具有特异性的t细胞受体样抗体
US10087250B2 (en) 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
US10111954B2 (en) 2012-08-14 2018-10-30 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10155815B2 (en) 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10183992B2 (en) 2012-08-14 2019-01-22 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
US10245321B2 (en) 2012-08-14 2019-04-02 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10316104B2 (en) 2011-04-29 2019-06-11 Roche Glycart Ag Immunoconjugates
US10323099B2 (en) * 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
US10611840B2 (en) 2014-08-04 2020-04-07 Hoffman-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US10882918B2 (en) 2016-09-30 2021-01-05 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
US11173179B2 (en) 2015-06-25 2021-11-16 Icell Gene Therapeutics Llc Chimeric antigen receptor (CAR) targeting multiple antigens, compositions and methods of use thereof
US11242390B2 (en) 2016-03-22 2022-02-08 Hoffmann-La Roche Inc. Protease-activated T cell bispecific molecules
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
US11655452B2 (en) 2015-06-25 2023-05-23 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods of use thereof
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
US11820819B2 (en) 2016-06-24 2023-11-21 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods thereof
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use

Families Citing this family (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG194510A1 (en) 2011-04-22 2013-12-30 Emergent Product Dev Seattle Prostate-specific membrane antigen binding proteins and related compositionsand methods
RU2617970C2 (ru) * 2011-08-23 2017-04-28 Рош Гликарт Аг Антитела, не содержащие fc-фрагмента, включающие два fab-фрагмента, и способы их применения
MX2015000426A (es) 2012-07-13 2015-07-14 Univ Pennsylvania Incremento de actividad de celulas t car mediante cointroduccion de un anticuerpo biespecifico.
TWI682941B (zh) 2013-02-01 2020-01-21 美商再生元醫藥公司 含嵌合恆定區之抗體
JP6636803B2 (ja) 2013-02-05 2020-01-29 エンクマフ エスアーエールエル Bcmaに対する抗体の選択のための方法
EP2789630A1 (de) 2013-04-09 2014-10-15 EngMab AG Bispezifische Antikörper gegen CD3e und ROR1
HRP20220553T1 (hr) 2013-07-25 2022-06-10 Cytomx Therapeutics Inc. Polispecifična protutijela, polispecifična protutijela koja se mogu aktivirati i postupci uporabe navedenih protutijela
CN104560894B (zh) * 2013-10-16 2017-07-14 北京中科永邦生物科技有限公司 编码分泌介导效应细胞杀伤靶细胞的双特异分子的框架及病毒
CN104707136A (zh) * 2013-12-17 2015-06-17 江苏铼泰医药生物技术有限公司 新型双功能抗体偶联物及其制法和用途
TWI701042B (zh) 2014-03-19 2020-08-11 美商再生元醫藥公司 用於腫瘤治療之方法及抗體組成物
TWI718992B (zh) 2014-07-21 2021-02-21 瑞士商諾華公司 使用cll-1嵌合抗原受體治療癌症
CN106687483B (zh) 2014-07-21 2020-12-04 诺华股份有限公司 使用人源化抗-bcma嵌合抗原受体治疗癌症
WO2016014974A2 (en) 2014-07-25 2016-01-28 Cytomx Therapeutics, Inc. Anti-cd3 antibodies, activatable anti-cd3 antibodies, multispecific anti-cd3 antibodies, multispecific activatable anti-cd3 antibodies, and methods of using the same
EP2982692A1 (de) * 2014-08-04 2016-02-10 EngMab AG Bispezifische Antikörper gegen CD3-Epsilon und BCMA
ES2850325T3 (es) * 2014-10-09 2021-08-27 Engmab Sarl Anticuerpos biespecíficos contra CD3epsilon y ROR1
WO2016081490A1 (en) 2014-11-17 2016-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using cd3xcd20 bispecific antibody
AU2016242866B2 (en) 2015-03-30 2021-06-03 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to FC gamma receptors
CN106467576B (zh) * 2015-08-18 2020-04-07 中国科学院微生物研究所 一种抗体融合蛋白及其制备方法与应用
EP3352760A4 (de) 2015-09-21 2019-03-06 Aptevo Research and Development LLC Cd3-bindende polypeptide
IL258036B (en) * 2015-09-23 2022-09-01 Regeneron Pharma Optimized bispecific antibodies against class 3 determinants and their uses
BR112018002570A2 (pt) * 2015-10-02 2018-10-16 Hoffmann La Roche molécula de ligação ao antígeno biespecífica, anticorpo biespecífico, polinucleotídeos, anticorpo que se liga especificamente ao ox40, composição farmacêutica e método para inibir o crescimento de células tumorais em um indivíduo
CN106661117B (zh) * 2015-12-30 2020-11-17 深圳先进技术研究院 IgG杂合型抗TNFα和IL-17A双特异性抗体
CN109073635A (zh) * 2016-01-25 2018-12-21 豪夫迈·罗氏有限公司 用于测定t细胞依赖性双特异性抗体的方法
CN110072554A (zh) * 2016-05-05 2019-07-30 宾夕法尼亚大学理事会 用于抗铜绿假单胞菌的dna抗体构建体
BR112019018767A2 (pt) 2017-04-03 2020-05-05 Hoffmann La Roche anticorpos, molécula de ligação ao antígeno biespecífica, um ou mais polinucleotídeos isolados, um ou mais vetores, célula hospedeira, método para produzir um anticorpo, composição farmacêutica, usos, método para tratar uma doença em um indivíduo e invenção
CN111148763B (zh) 2017-07-20 2023-11-03 Aptevo研究与开发公司 结合于5t4和4-1bb的抗原结合蛋白以及相关组合物和方法
JP7438106B2 (ja) 2017-10-14 2024-02-26 シートムエックス セラピューティクス,インコーポレイテッド 抗体、活性化可能抗体、二重特異性抗体、および二重特異性活性化可能抗体ならびにその使用方法
US11952422B2 (en) 2017-12-05 2024-04-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule comprising altered antibody variable region binding CD3 and CD137
MX2020006119A (es) 2017-12-21 2020-08-24 Hoffmann La Roche Anticuerpos de union a hla-a2/wt1.
US20210002669A1 (en) 2017-12-22 2021-01-07 Genentech, Inc. Targeted integration of nucleic acids
WO2019149716A1 (en) 2018-01-31 2019-08-08 F. Hoffmann-La Roche Ag Bispecific antibodies comprising an antigen-binding site binding to lag3
CN111630063A (zh) 2018-01-31 2020-09-04 豪夫迈·罗氏有限公司 稳定化的免疫球蛋白结构域
TWI829667B (zh) 2018-02-09 2024-01-21 瑞士商赫孚孟拉羅股份公司 結合gprc5d之抗體
CN112119099A (zh) 2018-03-02 2020-12-22 Cdr-生物科技股份有限公司 三特异性抗原结合蛋白
AU2019245243A1 (en) 2018-03-29 2020-09-03 Genentech, Inc Modulating lactogenic activity in mammalian cells
AR115052A1 (es) * 2018-04-18 2020-11-25 Hoffmann La Roche Anticuerpos multiespecíficos y utilización de los mismos
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
EP3844189A1 (de) 2018-08-31 2021-07-07 Regeneron Pharmaceuticals, Inc. Dosierstrategie zur abschwächung des cytokinfreisetzungssyndroms für bispezifische cd3/c20-antikörper
JPWO2020071554A1 (ja) * 2018-10-05 2021-09-09 北海道公立大学法人 札幌医科大学 がん幹細胞特異的抗体
CA3118816A1 (en) * 2018-11-07 2020-05-14 Crispr Therapeutics Ag Anti-cd33 immune cell cancer therapy
TWI829831B (zh) 2018-12-21 2024-01-21 瑞士商赫孚孟拉羅股份公司 結合cd3之抗體
CA3123448A1 (en) 2018-12-21 2020-06-25 Genentech, Inc. Targeted integration of nucleic acids
WO2020132574A1 (en) * 2018-12-21 2020-06-25 CentryMed Pharmaceutical Inc. Protease cleavable bispecific antibodies and uses thereof
CN111378045B (zh) * 2018-12-28 2022-08-02 长春金赛药业有限责任公司 二价双特异性抗体及其制备方法、编码基因、宿主细胞、组合物
CN112175087B (zh) * 2019-07-02 2022-05-13 深圳市迈加瑞生物技术有限公司 一种抗CD4和TGFβ的双特异性抗体、其药物组合及其用途
JP2022543551A (ja) 2019-07-31 2022-10-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Gprc5dに結合する抗体
JP2022543553A (ja) 2019-07-31 2022-10-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Gprc5dに結合する抗体
US20210130492A1 (en) 2019-09-18 2021-05-06 Genentech, Inc. Anti-klk7 antibodies, anti-klk5 antibodies, multispecific anti-klk5/klk7 antibodies, and methods of use
PE20221282A1 (es) 2019-12-18 2022-09-05 Hoffmann La Roche Anticuerpos que se unen a hla-a2/mage-a4
KR20220118527A (ko) 2019-12-23 2022-08-25 제넨테크, 인크. 아포지질단백질 l1-특이적 항체 및 이용 방법
US11179473B2 (en) 2020-02-21 2021-11-23 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
JP2023518841A (ja) 2020-03-26 2023-05-08 ジェネンテック, インコーポレイテッド 宿主細胞タンパク質が減少した修飾哺乳動物細胞
WO2021200898A1 (en) 2020-03-31 2021-10-07 Chugai Seiyaku Kabushiki Kaisha Dll3-targeting multispecific antigen-binding molecules and uses thereof
MX2022013173A (es) 2020-04-24 2022-11-30 Hoffmann La Roche Modulacion de enzimas y vias con compuestos de sulfhidrilo y sus derivados.
US20230340081A1 (en) 2020-06-08 2023-10-26 Hoffmann-La Roche Inc. Anti-hbv antibodies and methods of use
AU2021291407A1 (en) 2020-06-19 2022-09-29 F. Hoffmann-La Roche Ag Antibodies binding to CD3
WO2021255143A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and folr1
WO2021255146A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and cea
KR20230027043A (ko) 2020-06-24 2023-02-27 제넨테크, 인크. 핵산의 표적화 통합
WO2021262783A1 (en) 2020-06-24 2021-12-30 Genentech, Inc. Apoptosis resistant cell lines
AU2021300362A1 (en) 2020-07-01 2023-02-23 ARS Pharmaceuticals, Inc. Anti-ASGR1 antibody conjugates and uses thereof
WO2022007807A1 (zh) * 2020-07-07 2022-01-13 百奥泰生物制药股份有限公司 双特异性抗体及其应用
AU2021308653A1 (en) 2020-07-17 2023-02-16 Genentech, Inc. Anti-Notch2 antibodies and methods of use
CN116322763A (zh) 2020-08-27 2023-06-23 学校法人顺天堂 抗切断型突变calr-cd3双特异性抗体及医药组合物
KR20230056766A (ko) 2020-08-28 2023-04-27 제넨테크, 인크. 숙주 세포 단백질의 CRISPR/Cas9 다중 녹아웃
EP4228704A1 (de) * 2020-10-18 2023-08-23 Ardeagen Corporation Anti-cspg4-bindemittel, konjugate davon und verfahren zur verwendung davon
KR20230120665A (ko) 2020-12-17 2023-08-17 에프. 호프만-라 로슈 아게 항-hla-g 항체 및 이의 용도
JP2024509695A (ja) 2021-02-03 2024-03-05 ジェネンテック, インコーポレイテッド 多重特異性結合タンパク質分解プラットフォームおよび使用方法
AR125074A1 (es) 2021-03-12 2023-06-07 Genentech Inc Anticuerpos anti-klk7, anticuerpos anti-klk5, anticuerpos multiespecíficos anti-klk5 / klk7 y métodos de uso
TW202305122A (zh) 2021-04-19 2023-02-01 美商建南德克公司 經修飾之哺乳動物細胞
KR20240010469A (ko) 2021-05-21 2024-01-23 제넨테크, 인크. 관심 재조합 생성물의 생성을 위한 변형된 세포
WO2023287663A1 (en) 2021-07-13 2023-01-19 Genentech, Inc. Multi-variate model for predicting cytokine release syndrome
WO2023001884A1 (en) 2021-07-22 2023-01-26 F. Hoffmann-La Roche Ag Heterodimeric fc domain antibodies
WO2023012147A1 (en) 2021-08-03 2023-02-09 F. Hoffmann-La Roche Ag Bispecific antibodies and methods of use
AR127887A1 (es) 2021-12-10 2024-03-06 Hoffmann La Roche Anticuerpos que se unen a cd3 y plap
US20230322958A1 (en) 2022-01-19 2023-10-12 Genentech, Inc. Anti-Notch2 Antibodies and Conjugates and Methods of Use
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024079015A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and imids
WO2024079009A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and proteasome inhibitors
WO2024079010A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and cd38 antibodies
WO2024081918A1 (en) 2022-10-14 2024-04-18 Talem Therapeutics Llc Anti-trkb/cd3 antibodies and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991003493A1 (en) * 1989-08-29 1991-03-21 The University Of Southampton Bi-or trispecific (fab)3 or (fab)4 conjugates
US20060160184A1 (en) * 2003-05-30 2006-07-20 Merus Biopharmaceuticals, B.V. I.O. Fab library for the preparation of anti VEGF and anti rabies virus fabs
US20070269369A1 (en) * 2005-08-12 2007-11-22 Gegg Colin V Modified Fc molecules
US20090022738A1 (en) * 2003-10-16 2009-01-22 Micromet Ag Multispecific deimmunized CD3-binders
US20100055034A1 (en) * 2006-09-15 2010-03-04 Dompe Pha.Ma S.P.A. Human Anti-Folate Receptor Alpha Antibodies and Antibody Fragments for the Radioimmunotherapy of Ovarian Carcinoma

Family Cites Families (179)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2388385B1 (fr) 1977-04-18 1982-01-08 Hitachi Metals Ltd Piece d'ornement fixee par des aimants permanents
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
KR0184860B1 (ko) 1988-11-11 1999-04-01 메디칼 리써어치 카운실 단일영역 리간드와 이를 포함하는 수용체 및 이들의 제조방법과 이용(법)
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
CA2026147C (en) 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ES2113940T3 (es) 1990-12-03 1998-05-16 Genentech Inc Metodo de enriquecimiento para variantes de proteinas con propiedades de union alteradas.
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
ES2206447T3 (es) 1991-06-14 2004-05-16 Genentech, Inc. Anticuerpo humanizado para heregulina.
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
ES2136092T3 (es) 1991-09-23 1999-11-16 Medical Res Council Procedimientos para la produccion de anticuerpos humanizados.
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
AU675929B2 (en) 1992-02-06 1997-02-27 Curis, Inc. Biosynthetic binding protein for cancer marker
GB9221657D0 (en) 1992-10-15 1992-11-25 Scotgen Ltd Recombinant bispecific antibodies
CA2149329C (en) 1992-11-13 2008-07-15 Darrell R. Anderson Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5798100A (en) 1994-07-06 1998-08-25 Immunomedics, Inc. Multi-stage cascade boosting vaccine
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
CA2222231A1 (en) 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US7727761B2 (en) 1995-08-01 2010-06-01 Vegenics Limited Vascular endothelial growth factor C (VEGF-C) protein and gene, mutants thereof, and uses thereof
EP0861091B1 (de) * 1995-10-13 2003-08-20 THE GOVERNMENT OF THE UNITED STATES OF AMERICA as represented by the Secretary of the Department of Health and Human Services Immunotoxin enthaltend ein disulfid-stabilisiertes antikörperfragment
DE69830901T2 (de) 1997-05-02 2006-05-24 Genentech Inc., San Francisco ein verfahren zur herstellung multispezifischer antikörper die heteromultimere und gemeinsame komponenten besitzen
US6089968A (en) 1997-05-28 2000-07-18 Acraloc Corporation Loin knife assembly for automatic loin puller apparatus
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US6610833B1 (en) 1997-11-24 2003-08-26 The Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
ATE531812T1 (de) 1997-12-05 2011-11-15 Scripps Research Inst Humanisierung von nager-antikörpern
EP1049787B1 (de) 1998-01-23 2004-11-24 Vlaams Interuniversitair Instituut voor Biotechnologie Mehrzweck-antikörperderivate
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
AU782626B2 (en) 1999-10-04 2005-08-18 Medicago Inc. Method for regulating transcription of foreign genes
AU784983B2 (en) 1999-12-15 2006-08-17 Genentech Inc. Shotgun scanning, a combinatorial method for mapping functional protein epitopes
AU767394C (en) 1999-12-29 2005-04-21 Immunogen, Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
ES2528794T3 (es) * 2000-04-11 2015-02-12 Genentech, Inc. Anticuerpos multivalentes y usos de los mismos
US6773883B2 (en) 2000-07-31 2004-08-10 The Brigham & Women's Hospital, Inc. Prognostic classification of endometrial cancer
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
JP3523245B1 (ja) 2000-11-30 2004-04-26 メダレックス,インコーポレーテッド ヒト抗体作製用トランスジェニック染色体導入齧歯動物
US7432063B2 (en) 2002-02-14 2008-10-07 Kalobios Pharmaceuticals, Inc. Methods for affinity maturation
EP1513879B1 (de) 2002-06-03 2018-08-22 Genentech, Inc. Bibliotheken synthetischer antikörperphagen
AU2004205631A1 (en) 2003-01-16 2004-08-05 Genentech, Inc. Synthetic antibody phage libraries
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
EA036531B1 (ru) 2003-11-05 2020-11-19 Роше Гликарт Аг Гуманизированное антитело типа ii к cd20 (варианты), фармацевтическая композиция, содержащая эти варианты антитела, и их применение
KR101520209B1 (ko) 2003-11-06 2015-05-13 시애틀 지네틱스, 인크. 리간드에 접합될 수 있는 모노메틸발린 화합물
US7235641B2 (en) * 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
MXPA06011199A (es) 2004-03-31 2007-04-16 Genentech Inc Anticuerpos anti-tgf-beta humanizados.
US7785903B2 (en) 2004-04-09 2010-08-31 Genentech, Inc. Variable domain library and uses
US7728114B2 (en) 2004-06-03 2010-06-01 Novimmune S.A. Anti-CD3 antibodies and methods of use thereof
EP1791565B1 (de) 2004-09-23 2016-04-20 Genentech, Inc. Zystein-modifizierte Antikörper und Konjugate
JO3000B1 (ar) 2004-10-20 2016-09-05 Genentech Inc مركبات أجسام مضادة .
NZ556286A (en) 2005-02-07 2010-11-26 Glycart Biotechnology Ag Antigen binding molecules that bind EGFR, vectors encoding same, and uses thereof
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
AU2006232310B9 (en) * 2005-04-06 2011-07-21 Ibc Pharmaceuticals, Inc. Improved stably tethered structures of defined compositions with multiple functions or binding specificities
WO2006114115A1 (de) 2005-04-26 2006-11-02 Trion Pharma Gmbh Kombination von antikörpern mit glukokortikoiden zur behandlung von krebs
EP1888649A2 (de) 2005-05-09 2008-02-20 GlycArt Biotechnology AG Antigen-bindungsmoleküle mit modifizierten fc-regionen und veränderte bindung an fc-rezeptoren
JP2009504571A (ja) * 2005-07-25 2009-02-05 トルービオン ファーマスーティカルズ インコーポレイテッド タンパク質解離のための組成物および方法
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
RU2515108C2 (ru) 2005-08-19 2014-05-10 Эббви Инк Иммуноглобулин с двойными вариабельными доменами и его применения
AU2006301492B2 (en) 2005-10-11 2011-06-09 Amgen Research (Munich) Gmbh Compositions comprising cross-species-specific antibodies and uses thereof
ES2577292T3 (es) 2005-11-07 2016-07-14 Genentech, Inc. Polipéptidos de unión con secuencias hipervariables de VH/VL diversificadas y consenso
US20070237764A1 (en) 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
SG153825A1 (en) 2005-12-16 2009-07-29 Ibc Pharmaceuticals Inc Multivalent immunoglobulin-based bioactive assemblies
AU2007229698B9 (en) 2006-03-24 2012-11-08 Merck Patent Gmbh Engineered heterodimeric protein domains
CA2651567A1 (en) 2006-05-09 2007-11-22 Genentech, Inc. Binding polypeptides with optimized scaffolds
US20070274985A1 (en) 2006-05-26 2007-11-29 Stefan Dubel Antibody
JP2009539413A (ja) 2006-06-12 2009-11-19 トゥルビオン・ファーマシューティカルズ・インコーポレーテッド エフェクター機能を有する単鎖多価結合タンパク質
JP2009541275A (ja) 2006-06-22 2009-11-26 ノボ・ノルデイスク・エー/エス 二重特異性抗体の生産
MX2009010611A (es) 2007-04-03 2010-03-26 Micromet Ag Enlazadores biespecificos, especificos para especies.
CN109456410B (zh) 2007-04-03 2022-01-28 安进研发(慕尼黑)股份有限公司 跨物种特异性CD3-ε结合结构域
LT2520590T (lt) 2007-04-03 2018-09-10 Amgen Research (Munich) Gmbh Susijusioms rūšims specifinis rišantis domenas
CN100592373C (zh) 2007-05-25 2010-02-24 群康科技(深圳)有限公司 液晶显示面板驱动装置及其驱动方法
JP2010535032A (ja) * 2007-07-31 2010-11-18 メディミューン,エルエルシー 多重特異性エピトープ結合性タンパク質およびその用途
JP5490714B2 (ja) 2007-11-28 2014-05-14 メディミューン,エルエルシー タンパク質製剤
US8227577B2 (en) 2007-12-21 2012-07-24 Hoffman-La Roche Inc. Bivalent, bispecific antibodies
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US8242247B2 (en) 2007-12-21 2012-08-14 Hoffmann-La Roche Inc. Bivalent, bispecific antibodies
PT2235064E (pt) 2008-01-07 2016-03-01 Amgen Inc Método de preparação de moléculas heterodiméricas de fc de anticorpos utilizando efeitos de indução eletrostática
CA2726087A1 (en) * 2008-06-03 2009-12-10 Tariq Ghayur Dual variable domain immunoglobulins and uses thereof
US20100093563A1 (en) 2008-09-22 2010-04-15 Robert Anthony Williamson Methods and vectors for display of molecules and displayed molecules and collections
DK2356153T3 (en) * 2008-10-01 2016-07-04 Amgen Res (Munich) Gmbh Bispecific single CHAIN ​​PSMAXCD3 ANTIBODY THAT ARE SPECIFICALLY TO ALL SPECIES
CA2756244A1 (en) 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
EP2417156B1 (de) 2009-04-07 2015-02-11 Roche Glycart AG Trivalente, bispezifische antikörper
MX2011010169A (es) * 2009-04-07 2011-10-11 Roche Glycart Ag Anticuerpos biespecificos anti-erbb-1/anti-c-met.
CN102459346B (zh) 2009-04-27 2016-10-26 昂考梅德药品有限公司 制造异源多聚体分子的方法
WO2010136172A1 (en) * 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Tri- or tetraspecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US8703132B2 (en) * 2009-06-18 2014-04-22 Hoffmann-La Roche, Inc. Bispecific, tetravalent antigen binding proteins
BR112012003983A2 (pt) 2009-08-31 2021-09-14 Roche Glycart Ag Molecula de ligação abm moleculas de ligação de antigenos abm variante polipeptideo isolado molecula de ligação de antigeneos humanizada anticorpo polinucleotideo isolado composição vetor celula hospedeira metodo de produção de abm metodo de indução da lise celular de tumor, metodo de diagnostico de doença em pacientes que possuem um cancer metodo de aumento de tempo de sobrevivencia em pacientes que possuem um cancer metodo de indução em pacientes de regressão de um motor uso abm e invenção
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
SI2519543T1 (sl) 2009-12-29 2016-08-31 Emergent Product Development Seattle, Llc Beljakovine, ki se vežejo s heterodimeri in njihova uporaba
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
US9527926B2 (en) 2010-05-14 2016-12-27 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
SG187746A1 (en) 2010-08-13 2013-03-28 Roche Glycart Ag Anti-fap antibodies and methods of use
AU2011325833C1 (en) 2010-11-05 2017-07-13 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
AR084053A1 (es) 2010-11-30 2013-04-17 Chugai Pharmaceutical Co Ltd Agente terapeutico que induce citotoxicidad
PE20140303A1 (es) 2011-02-10 2014-03-22 Roche Glycart Ag Polipeptidos interleuquina-2 mutantes
PT2691417T (pt) 2011-03-29 2018-10-31 Roche Glycart Ag Variantes de fc de anticorpos
US20140170148A1 (en) 2011-04-20 2014-06-19 Genmab A/S Bispecific antibodies against her2
EA201892619A1 (ru) 2011-04-29 2019-04-30 Роше Гликарт Аг Иммуноконъюгаты, содержащие мутантные полипептиды интерлейкина-2
CN103842383B (zh) 2011-05-16 2017-11-03 健能隆医药技术(上海)有限公司 多特异性fab融合蛋白及其使用方法
SG195072A1 (en) 2011-05-21 2013-12-30 Macrogenics Inc Cd3-binding molecules capable of binding to human and non-human cd3
US20130078250A1 (en) 2011-08-23 2013-03-28 Oliver Ast Bispecific t cell activating antigen binding molecules
RU2617970C2 (ru) 2011-08-23 2017-04-28 Рош Гликарт Аг Антитела, не содержащие fc-фрагмента, включающие два fab-фрагмента, и способы их применения
CN103748114B (zh) 2011-08-23 2017-07-21 罗切格利卡特公司 T细胞活化性双特异性抗原结合分子
SI2748202T1 (sl) 2011-08-23 2018-10-30 Roche Glycart Ag Bispecifične molekule, ki se vežejo na antigen
KR101870555B1 (ko) 2011-08-23 2018-06-22 로슈 글리카트 아게 T 세포 활성화 항원 및 종양 항원에 대해 특이적인 이중특이적 항체 및 이의 사용 방법
EP2578230A1 (de) 2011-10-04 2013-04-10 Trion Pharma Gmbh Entfernung von Tumorzellen aus einer intraoperativen autologen Blutsammlung
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
CA2859667C (en) 2011-12-20 2022-05-24 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
EP2806887A4 (de) 2012-01-26 2015-11-04 Ibc Pharmaceuticals Inc Abzielung auf interferon-lambda mit antikörpern zur verstärkung von antitumor- und antiviralen wirkungen
EP2809682B1 (de) 2012-02-03 2020-04-08 F.Hoffmann-La Roche Ag Bispezifische antikörpermoleküle mit antigen-transfizierten t-zellen und deren verwendung in der medizin
MX360109B (es) 2012-04-20 2018-10-23 Merus Nv Metodos y medios para la produccion de moleculas de tipo ig.
DK2857420T3 (da) 2012-05-30 2020-11-23 Chugai Pharmaceutical Co Ltd Målvævsspecifikt antigenbindende molekyle
US20150203591A1 (en) 2012-08-02 2015-07-23 Regeneron Pharmaceuticals, Inc. Mutivalent antigen-binding proteins
SG10201800535XA (en) 2012-08-07 2018-02-27 Roche Glycart Ag Composition comprising two antibodies engineered to have reduced and increased effector function
US9315567B2 (en) 2012-08-14 2016-04-19 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
JOP20200236A1 (ar) 2012-09-21 2017-06-16 Regeneron Pharma الأجسام المضادة لمضاد cd3 وجزيئات ربط الأنتيجين ثنائية التحديد التي تربط cd3 وcd20 واستخداماتها
WO2014056783A1 (en) 2012-10-08 2014-04-17 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
UY35148A (es) 2012-11-21 2014-05-30 Amgen Inc Immunoglobulinas heterodiméricas
AU2014205086B2 (en) 2013-01-14 2019-04-18 Xencor, Inc. Novel heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
JP6636803B2 (ja) 2013-02-05 2020-01-29 エンクマフ エスアーエールエル Bcmaに対する抗体の選択のための方法
JO3529B1 (ar) 2013-02-08 2020-07-05 Amgen Res Munich Gmbh مضاد التصاق خلايا الدم البيض من أجل التخفيف من الاثار السلبية الممكنة الناتجة عن مجالات ارتباط cd3- المحدد
RU2015140915A (ru) 2013-02-26 2017-04-03 Роше Гликарт Аг Биспецифические антигенсвязывающие молекулы, активирующие т-клетки
MY192312A (en) 2013-02-26 2022-08-17 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
EP2961770A1 (de) 2013-02-26 2016-01-06 Roche Glycart AG Bispezifische antigenbindende moleküle zur t-zellen-aktivierung
WO2014153164A1 (en) 2013-03-14 2014-09-25 The California Institute For Biomedical Research Targeting agent antibody conjugates and uses thereof
SG11201506499YA (en) 2013-03-15 2015-09-29 Glaxosmithkline Ip No 2 Ltd Low concentration antibody formulations
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
UA118028C2 (uk) 2013-04-03 2018-11-12 Рош Глікарт Аг Біспецифічне антитіло, специфічне щодо fap і dr5, антитіло, специфічне щодо dr5, і спосіб їх застосування
EP2789630A1 (de) 2013-04-09 2014-10-15 EngMab AG Bispezifische Antikörper gegen CD3e und ROR1
SG11201509361TA (en) 2013-05-28 2015-12-30 Numab Ag Novel antibodies
CA2920192A1 (en) 2013-09-27 2015-04-02 Immunomedics, Inc. Anti-trop-2 antibody-drug conjugates and uses thereof
DK3083689T3 (da) 2013-12-17 2020-08-03 Genentech Inc Anti-CD3-antistoffer og fremgangsmåder til anvendelse
EP3083692B1 (de) 2013-12-17 2020-02-19 F.Hoffmann-La Roche Ag Verfahren zur behandlung von krebs mit pd-1-achsenbindenden antagonisten und taxanen
UA117289C2 (uk) 2014-04-02 2018-07-10 Ф. Хоффманн-Ля Рош Аг Мультиспецифічне антитіло
EP2982692A1 (de) 2014-08-04 2016-02-10 EngMab AG Bispezifische Antikörper gegen CD3-Epsilon und BCMA
PE20170263A1 (es) 2014-08-04 2017-03-30 Hoffmann La Roche Moleculas biespecificas de union a antigeno activadoras de celulas t
WO2016019969A1 (en) 2014-08-08 2016-02-11 Ludwig-Maximilians-Universität München Subcutaneously administered bispecific antibodies for use in the treatment of cancer
WO2016030350A1 (en) 2014-08-29 2016-03-03 F. Hoffmann-La Roche Ag Combination therapy of tumor-targeted il-2 variant immunocytokines and antibodies against human pd-l1
WO2016036678A1 (en) 2014-09-02 2016-03-10 Medimmune, Llc Formulations of bispecific antibodies
MA40764A (fr) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd Agent thérapeutique induisant une cytotoxicité
AU2015329966A1 (en) 2014-10-09 2017-04-27 Engmab Sàrl Bispecific antibodies against CD3epsilon and ROR1 for use in the treatment of ovarian cancer
ES2850325T3 (es) 2014-10-09 2021-08-27 Engmab Sarl Anticuerpos biespecíficos contra CD3epsilon y ROR1
EP3023437A1 (de) 2014-11-20 2016-05-25 EngMab AG Bispezifische Antikörper gegen CD3epsilon und BCMA
MX2017006571A (es) 2014-11-20 2017-09-29 Hoffmann La Roche Moleculas de union a antigeno biespecificas activadoras de celulas t.
PL3221357T3 (pl) 2014-11-20 2020-11-02 F. Hoffmann-La Roche Ag Wspólne łańcuchy lekkie i sposoby zastosowania
EP3029068A1 (de) 2014-12-03 2016-06-08 EngMab AG Bispezifische Antikörper gegen CD3-Epsilon-BCMA und zur Verwendung bei der Behandlung von Krankheiten
JP2018520642A (ja) 2015-05-01 2018-08-02 ジェネンテック, インコーポレイテッド マスク抗cd3抗体及びその使用方法
TWI731861B (zh) 2015-06-16 2021-07-01 美商建南德克公司 FcRH5之人源化及親和力成熟抗體及使用方法
SI3331910T1 (sl) 2015-08-03 2020-07-31 Engmab Sarl Monoklonska protitelesa proti humani antigen dozorevanja limfocitov B (BCMA)
JP6937746B2 (ja) 2015-10-02 2021-09-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト 二重特異性抗cd19×cd3t細胞活性化抗原結合分子
US20170096485A1 (en) 2015-10-02 2017-04-06 Hoffmann-La Roche Inc. Bispecific t cell activating antigen binding molecules
WO2017055318A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Cd33xcd3 bispecific t cell activating antigen binding molecules
CN107849137B (zh) 2015-10-02 2021-11-26 豪夫迈·罗氏有限公司 双特异性抗ceaxcd3 t细胞活化性抗原结合分子
WO2017055388A2 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific t cell activating antigen binding molecules
JP7325186B2 (ja) 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 抗薬物抗体の形成を減少させるためのii型抗cd20抗体

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991003493A1 (en) * 1989-08-29 1991-03-21 The University Of Southampton Bi-or trispecific (fab)3 or (fab)4 conjugates
US20060160184A1 (en) * 2003-05-30 2006-07-20 Merus Biopharmaceuticals, B.V. I.O. Fab library for the preparation of anti VEGF and anti rabies virus fabs
US20090022738A1 (en) * 2003-10-16 2009-01-22 Micromet Ag Multispecific deimmunized CD3-binders
US20070269369A1 (en) * 2005-08-12 2007-11-22 Gegg Colin V Modified Fc molecules
US20100055034A1 (en) * 2006-09-15 2010-03-04 Dompe Pha.Ma S.P.A. Human Anti-Folate Receptor Alpha Antibodies and Antibody Fragments for the Radioimmunotherapy of Ovarian Carcinoma

Cited By (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10927163B2 (en) 2007-12-21 2021-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10316104B2 (en) 2011-04-29 2019-06-11 Roche Glycart Ag Immunoconjugates
US11130822B2 (en) 2011-04-29 2021-09-28 Roche Glycart Ag Immunoconjugates
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
US9309306B2 (en) 2011-08-23 2016-04-12 Roche Glycart Ag Anti-MCSP antibodies
US9879088B2 (en) 2012-08-14 2018-01-30 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US10111954B2 (en) 2012-08-14 2018-10-30 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
US10308688B2 (en) 2012-08-14 2019-06-04 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
US10239938B2 (en) 2012-08-14 2019-03-26 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
US10245321B2 (en) 2012-08-14 2019-04-02 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
US10183992B2 (en) 2012-08-14 2019-01-22 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
US10087250B2 (en) 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
US10155815B2 (en) 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10781258B2 (en) 2013-02-26 2020-09-22 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10781257B2 (en) 2013-02-26 2020-09-22 Roche GlyeArt AG Bispecific T cell activating antigen binding molecules
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US11136393B2 (en) 2013-10-01 2021-10-05 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of Bim
US10323099B2 (en) * 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer
US10611840B2 (en) 2014-08-04 2020-04-07 Hoffman-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10611841B2 (en) 2014-08-04 2020-04-07 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11117965B2 (en) 2014-08-04 2021-09-14 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US11613587B2 (en) 2014-11-20 2023-03-28 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US20170327580A1 (en) * 2014-12-03 2017-11-16 Engmab Ag Bispecific antibodies against cd3 epsilon and bcma for use in treatment of diseases
WO2016138491A1 (en) * 2015-02-27 2016-09-01 Icell Gene Therapeutics Llc Chimeric antigen receptors (cars) targeting hematologic malignancies, compositions and methods of use thereof
US10273280B2 (en) 2015-02-27 2019-04-30 Icell Gene Therapeutics Llc Chimeric antigen receptors (CARs), targeting hematologic malignancies, compositions and methods of use thereof
WO2016164920A1 (en) * 2015-04-09 2016-10-13 Cornell University Gene therapy to prevent reactions to allergens
US10293059B2 (en) 2015-04-09 2019-05-21 Cornell University Gene therapy to prevent reactions to allergens
WO2016168766A1 (en) * 2015-04-15 2016-10-20 The California Institute For Biomedical Research Optimized chimeric receptor t cell switches and uses thereof
CN107921127A (zh) * 2015-05-22 2018-04-17 纪念斯隆-凯特琳癌症中心 对于prame肽具有特异性的t细胞受体样抗体
US11384144B2 (en) * 2015-05-22 2022-07-12 Memorial Sloan-Kettering Cancer Center T cell receptor-like antibodies specific for a PRAME peptide
US11655452B2 (en) 2015-06-25 2023-05-23 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods of use thereof
US11173179B2 (en) 2015-06-25 2021-11-16 Icell Gene Therapeutics Llc Chimeric antigen receptor (CAR) targeting multiple antigens, compositions and methods of use thereof
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
WO2017075045A3 (en) * 2015-10-30 2017-06-08 Mayo Foundation For Medical Education And Research Antibodies to b7-h1
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
WO2017127499A1 (en) 2016-01-22 2017-07-27 Janssen Biotech, Inc. Anti-ror1 antibodies, ror1 x cd3 bispecific antibodies, and methods of using the same
US11242390B2 (en) 2016-03-22 2022-02-08 Hoffmann-La Roche Inc. Protease-activated T cell bispecific molecules
WO2017180555A1 (en) * 2016-04-11 2017-10-19 The Trustees Of Columbia University In The City Of New York Humanized anti-rage antibody
US10550184B2 (en) 2016-04-11 2020-02-04 The Trustees Of Columbia University In The City Of New York Humanized anti-rage antibody
US11820819B2 (en) 2016-06-24 2023-11-21 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods thereof
US10882918B2 (en) 2016-09-30 2021-01-05 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
US11850262B2 (en) 2017-02-28 2023-12-26 Purdue Research Foundation Compositions and methods for CAR T cell therapy
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19

Also Published As

Publication number Publication date
EP2747781A1 (de) 2014-07-02
CN103889452A (zh) 2014-06-25
CN103889452B (zh) 2017-11-03
CN107586340B (zh) 2022-01-21
BR112014004168A2 (pt) 2017-12-12
MX356947B (es) 2018-06-20
EP2747781B1 (de) 2017-11-15
RU2014109551A (ru) 2015-09-27
KR20140041856A (ko) 2014-04-04
MX2014002097A (es) 2014-05-28
KR20160138583A (ko) 2016-12-05
KR101870555B1 (ko) 2018-06-22
KR101681818B1 (ko) 2016-12-01
CN107586340A (zh) 2018-01-16
US20150274845A1 (en) 2015-10-01
CA2844540C (en) 2018-10-16
US11639397B2 (en) 2023-05-02
HK1249529A1 (zh) 2018-11-02
CA2844540A1 (en) 2013-02-28
WO2013026839A1 (en) 2013-02-28
JP2014529600A (ja) 2014-11-13
JP6159724B2 (ja) 2017-07-05
RU2605390C2 (ru) 2016-12-20
JP2017137329A (ja) 2017-08-10

Similar Documents

Publication Publication Date Title
US11639397B2 (en) Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
US10087250B2 (en) Fc-free antibodies comprising two fab-fragments and methods of use
EP2748200B1 (de) Fc-freie antikörper mit zwei fab-fragmenten sowie verwendungsverfahren dafür
US20220363755A1 (en) Anti-tim3 antibodies and methods of use
JP6738285B2 (ja) ヒト及びカニクイザルcd3イプシロンに結合する抗体
CN105829347B (zh) 双特异性her2抗体及使用方法
JP6470384B2 (ja) 抗ビオチン抗体および使用方法
JP6549278B2 (ja) 抗テオフィリン抗体および使用方法
CN104755500B (zh) 结合HER3 β-发夹的HER3抗原结合蛋白
JP7248761B2 (ja) 抗brdu抗体および使用方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: ROCHE GLYCART AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRUENKER, PETER;FAUTI, TANJA;JAEGER, CHRISTIANE;AND OTHERS;REEL/FRAME:029319/0530

Effective date: 20120824

STCB Information on status: application discontinuation

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