WO2021255146A1 - Anticorps se liant à cd3 et cea - Google Patents

Anticorps se liant à cd3 et cea Download PDF

Info

Publication number
WO2021255146A1
WO2021255146A1 PCT/EP2021/066354 EP2021066354W WO2021255146A1 WO 2021255146 A1 WO2021255146 A1 WO 2021255146A1 EP 2021066354 W EP2021066354 W EP 2021066354W WO 2021255146 A1 WO2021255146 A1 WO 2021255146A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antigen binding
domain
seq
amino acid
Prior art date
Application number
PCT/EP2021/066354
Other languages
English (en)
Inventor
Anne Freimoser-Grundschober
Maria Valeria GONZALEZ NICOLINI
Ralf Hosse
Alexander KNAUPP
Ekkehard Moessner
Wolfgang Richter
Halina TROCHANOWSKA
Pablo Umaña
Inja Waldhauer
Original Assignee
F. Hoffmann-La Roche Ag
Hoffmann-La Roche Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F. Hoffmann-La Roche Ag, Hoffmann-La Roche Inc. filed Critical F. Hoffmann-La Roche Ag
Publication of WO2021255146A1 publication Critical patent/WO2021255146A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [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
    • C07K16/3007Carcino-embryonic Antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/66Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a swap of domains, e.g. CH3-CH2, VH-CL or VL-CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention generally relates to antibodies that bind to CD3 and CEA, e.g. for activating T cells.
  • the present invention relates to polynucleotides encoding such antibodies, and vectors and host cells comprising such polynucleotides.
  • the invention further relates to methods for producing the antibodies, and to methods of using them in the treatment of disease.
  • CD3 (cluster of differentiation 3) is a protein complex composed of four subunits, the CD3y chain, the CD35 chain, and two CD3 chains. CD3 associates with the T-cell receptor and the z chain to generate an activation signal in T lymphocytes.
  • CD3 has been extensively explored as drug target. Monoclonal antibodies targeting CD3 have been used as immunosuppressant therapies in autoimmune diseases such as type I diabetes, or in the treatment of transplant rejection.
  • the CD3 antibody muromonab-CD3 (OKT3) was the first monoclonal antibody ever approved for clinical use in humans, in 1985.
  • CD3 antibodies A more recent application of CD3 antibodies is in the form ofbispecific antibodies, binding CD3 on the one hand and a target cell antigen such as carcinoembryonic antigen (CEA) on the other hand.
  • a target cell antigen such as carcinoembryonic antigen (CEA)
  • CEA carcinoembryonic antigen
  • an important requirement that antibodies have to fulfill is sufficient stability both in vitro (for storage of the drug) an in vivo (after administration to the patient). Modifications like asparagine deamidation are typical degradations for recombinant antibodies and can affect both in vitro stability and in vivo biological functions.
  • the present invention provides antibodies, including multispecific (e.g. bispecific) antibodies, that bind to CD3 and are resistant to degradation by e.g. asparagine deamidation and thus particularly stable as required for therapeutic purposes.
  • the (multispecific) antibodies provided further combine good efficacy and produceability with low toxicity and favorable pharmacokinetic properties.
  • the antibodies including multi specific antibodies, that bind to CD3, provided by the present invention, retain more than about 90% binding activity to CD3 after 2 weeks at pH 7.4, 37°C, relative to the binding activity after 2 weeks at pH 6, -80°C, as determined by surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises (a) a first antigen binding domain that binds to CD3, comprising a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; and (b) a second and optionally a third antigen binding domain that binds to CEA.
  • VH heavy chain variable region
  • HCDR heavy chain complementary determining region
  • VL light chain variable region
  • LCDR light chain complementarity determining region
  • the VH of the first antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 7, and/or the VL of the first antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 11.
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises (a) a first antigen binding domain that binds to CD3 comprising a VH sequence of SEQ ID NO: 7 and a VL sequence of SEQ ID NO: 11; and (b) a second and optionally a third antigen binding domain that binds to CEA.
  • the first, the second and/or, where present, the third antigen binding domain is a Fab molecule.
  • the antibody comprises an Fc domain composed of a first and a second subunit.
  • the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH or the constant domains CL and CHI, particularly the variable domains VL and VH, of the Fab light chain and the Fab heavy chain are replaced by each other.
  • the third antigen binding domain is a conventional Fab molecule.
  • the second and, where present, the third antigen binding domain is a Fab molecule wherein in the constant domain CL the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Rabat) and the amino acid at position 123 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Rabat), and in the constant domain CHI the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Rabat EU index) and the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Rabat EU index).
  • the first and the second antigen binding domain are fused to each other, optionally via a peptide linker.
  • the first and the second antigen binding domain are each a Fab molecule and either (i) the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N- terminus of the Fab heavy chain of the first antigen binding domain, or (ii) the first antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain.
  • the first, the second and, where present, the third antigen binding domain are each a Fab molecule and the antibody comprises an Fc domain composed of a first and a second subunit; and wherein either (i) the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain and the first antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain, or (ii) the first antigen binding domain is fused at the C- terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain and the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain; and the third antigen binding domain, where present, is fused at the C-terminus of the Fab heavy chain to the N-terminus of the second subunit
  • the Fc domain is an IgG, particularly an IgGi, Fc domain. In one aspect the Fc domain is a human Fc domain. In one aspect, the Fc comprises a modification promoting the association of the first and the second subunit of the Fc domain. In one aspect, the Fc domain comprises one or more amino acid substitution that reduces binding to an Fc receptor and/or effector function.
  • the third antigen binding domain comprises (i) a VH comprising a HCDR 1 of SEQ ID NO: 15, a HCDR 2 of SEQ ID NO: 16, and a HCDR 3 of SEQ ID NO: 17, and a VL comprising a LCDR 1 of SEQ ID NO: 19, a LCDR 2 of SEQ ID NO: 20 and a LCDR 3 of SEQ ID NO: 21; (ii) a VH comprising a HCDR 1 of SEQ ID NO: 28, a HCDR 2 of SEQ ID NO: 29, and a HCDR 3 of SEQ ID NO: 30, and a VL comprising a LCDR 1 of SEQ ID NO: 32, a LCDR 2 of SEQ ID NO: 33 and a LCDR 3 of SEQ ID NO: 34; or (iii) a VH comprising a HCDR 1 of SEQ ID NO: 36, a HCDR 2 of SEQ ID NO : 37, and a HCDR
  • the third antigen binding domain comprises (i) a VH comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 18, and/or a VL comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 22; (ii) a VH comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 31, and/or a VL comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 35; or (iii) a VH comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:
  • an isolated polynucleotide encoding an antibody of the invention, and a host cell comprising the isolated polynucleotide of the invention.
  • a method of producing an antibody that binds to CD3 and CEA comprising the steps of (a) culturing the host cell of the invention under conditions suitable for the expression of the antibody and optionally (b) recovering the antibody.
  • the invention also encompasses an antibody that binds to CD3 and CEA produced by the method of the invention.
  • the invention further provides a pharmaceutical composition comprising the antibody of the invention and a pharmaceutically acceptable carrier.
  • the invention provides an antibody or pharmaceutical composition according to the invention for use as a medicament.
  • an antibody or pharmaceutical composition according to the invention for use in the treatment of a disease is also provided.
  • the use of an antibody or pharmaceutical composition according to the invention in the manufacture of a medicament and the use of an antibody or pharmaceutical composition according to the invention in the manufacture of a medicament for the treatment of a disease.
  • the invention also provides a method of treating a disease in an individual, comprising administering to said individual an effective amount of the antibody or pharmaceutical composition according to the invention.
  • the disease is cancer.
  • Figure 1 Exemplary configurations of the (multispecific) antibodies of the invention.
  • A, D Illustration of the “1+1 CrossMab” molecule.
  • B, E Illustration of the “2+1 IgG Crossfab” molecule with alternative order of Crossfab and Fab components (“inverted”).
  • C, F Illustration of the “2+1 IgG Crossfab” molecule.
  • G, K Illustration of the “1+1 IgG Crossfab” molecule with alternative order of Crossfab and Fab components (“inverted”).
  • H, L Illustration of the “1+1 IgG Crossfab” molecule.
  • I, M Illustration of the “2+1 IgG Crossfab” molecule with two CrossFabs.
  • Crossfab molecules are depicted as comprising an exchange of VH and VL regions, but may - in aspects wherein no charge modifications are introduced in CHI and CL domains - alternatively comprise an exchange of the CHI and CL domains.
  • FIG. 3 Binding of original and optimized CD3 binders, CD3 0rig and CD3 opt , to Jurkat NFAT cells as measured by flow cytometry (IgG format). Antibodies bound to Jurkat NFAT cells were detected with a fluorescently labeled anti-human Fc specific secondary antibody.
  • FIG. 1 Schematic illustration of the CD3 activation assay used in Example 3.
  • FIG. 1 Jurkat NFAT activation with original and optimized CD3 binders, CD3 0rig and CD3 opt (IgG format).
  • Jurkat NFAT reporter cells were co-incubated with anti-PGLALA expressing CHO (CHO-PGLALA) cells in the presence of CD3 0 rig or CD3 opt IgG PGLALA, or CD3 opt IgG wt as negative control.
  • CD3 activation was quantified by measuring luminescence after 24 h.
  • Figure 6. (A) Schematic illustration of the T-cell bi specific antibody (TCB) molecules used in the Examples.
  • T-cell bi specific antibody (TCB) molecules used in the Examples.
  • B-E Components for the assembly of the TCB: light chain of anti-TYRP 1 Fab molecule with charge modifications in CHI and CL (B), light chain of anti-CD3 crossover Fab molecule (C), heavy chain with knob and PG LALA mutations in Fc region (D), heavy chain with hole and PG LALA mutations in Fc region (E).
  • FIG. 7 Binding of CE AC AM5 -T CB molecules, containing the optimized anti-CD3 antibodies CD3 0pt or the parental binder CD3 0rig , to CD3 -expressing T cells (A) and CEA-positive tumor cells (B), as measured by flow cytometry.
  • FIG. 8 Tumor cell lysis of CEA-positive tumor cells mediated by CE AC AM5 -TCB molecules, containing the optimized anti-CD3 antibody CD3 opt or the parental binder CD3 0rig , with PBMCs from a healthy donor. Tumor cell killing was measured by quantification ofLDH release after 24 hours (A) and 48 hours (B).
  • FIG. 9 Schematic illustration of the surface plasmon resonance (SPR) setup used in Example 8.
  • SPR surface plasmon resonance
  • the terms “first”, “second” or “third” with respect to antigen binding domains etc. are used for convenience of distinguishing when there is more than one of each type of moiety. Use of these terms is not intended to confer a specific order or orientation of the moiety unless explicitly so stated.
  • an antibody that binds to CD3 refers to an antibody that is capable of binding CD3 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting CD3.
  • the extent of binding of an anti-CD3 antibody to an unrelated, non-CD3 protein is less than about 10% of the binding of the antibody to CD3 as measured, e.g., by surface plasmon resonance (SPR).
  • an antibody that binds to CD3 has a dissociation constant (K D ) of ⁇ 1 mM, ⁇ 500 nM, ⁇ 200 nM, or ⁇ 100 nM.
  • an antibody is said to “specifically bind” to CD3 when the antibody has a K D of 1 mM or less, as measured, e.g., by SPR.
  • an anti-CD3 antibody binds to an epitope of CD3 that is conserved among CD3 from different species.
  • an antibody that binds to CEA refers to an antibody that is capable of binding CEA with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting CEA.
  • the extent of binding of an anti-CEA antibody to an unrelated, non-CEA protein is less than about 10% of the binding of the antibody to CEA as measured, e.g., by surface plasmon resonance (SPR).
  • an antibody that binds to CEA has a dissociation constant (K D ) of ⁇ 1 mM, ⁇ 500 nM, ⁇ 200 nM, or ⁇ 100 nM.
  • an antibody is said to “specifically bind” to CEA when the antibody has a K D of 1 pM or less, as measured, e.g., by SPR.
  • an anti-CEA antibody binds to an epitope of CEA that is conserved among CEA from different species.
  • 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 fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab’-SH, F(ab')2, diabodies, linear antibodies, single-chain antibody molecules (e.g. scFv and scFab), single-domain antibodies, and multispecific antibodies formed from antibody fragments.
  • full-length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure.
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e. the individual antibodies comprised in 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.
  • 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.
  • monoclonal antibodies 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.
  • 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, affinity chromatography, size exclusion chro matography) methods.
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC, affinity chromatography, size exclusion chro matography
  • the antibodies provided by the present invention are isolated antibodies.
  • 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.
  • a “humanized” antibody refers to a chimeric antibody comprising amino acid residues from non human CDRs 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 CDRs correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • Such variable domains are referred to herein as “humanized variable region”.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a “humanized form” of an antibody e.g. of a non-human antibody, refers to an antibody that has undergone humanization.
  • 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.
  • a human antibody is derived from a non-human transgenic mammal, for example a mouse, a rat, or a rabbit.
  • a human antibody is derived from a hybridoma cell line.
  • Antibodies or antibody fragments isolated from human antibody libraries are also considered human antibodies or human antibody fragments herein.
  • an antigen binding domain refers to the part of an antibody that comprises the area which binds to and is complementary to part or all of an antigen.
  • An antigen binding domain may be provided by, for example, one or more antibody variable domains (also called antibody variable regions).
  • an antigen binding domain comprises an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (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 complementarity determining regions (CDRs). See, e.g., Kindt et al., Kuby Immunology, 6 th ed., W.H. Freeman & Co., page 91 (2007).
  • a single VH or VL domain may be sufficient to confer antigen-binding specificity.
  • 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.
  • VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively.
  • Portolano et al. J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
  • Rabat numbering refers to the numbering system set forth by Rabat et al., Sequences of Proteins of Immunological Interest, 5 th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991).
  • the amino acid positions of all constant regions and domains of the heavy and light chain are numbered according to the Rabat numbering system described in Rabat, et al., Sequences ofProteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991), referred to as “numbering according to Rabat” or “Rabat numbering” herein.
  • Rabat numbering system (see pages 647-660 of Rabat, et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991)) is used for the light chain constant domain CL of kappa and lambda isotype and the Rabat EU index numbering system (see pages 661-723) is used for the heavy chain constant domains (CHI, hinge, CH2 and CH3), which is herein further clarified by referring to “numbering according to Rabat EU index” or “Rabat EU index numbering” in this case.
  • hypervariable region refers to each of the regions of an antibody variable domain which are hypervariable in sequence and which determine antigen binding specificity, for example “complementarity determining regions” (“CDRs”).
  • CDRs complementarity determining regions
  • antibodies comprise six CDRs; three in the VH (HCDR1, HCDR2, HCDR3), and three in the VL (LCDR1, LCDR2, LCDR3).
  • Exemplary CDRs herein include:
  • FR refers to variable domain residues other than complementarity determining regions (CDRs).
  • 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 order in VH (or VL): FR1-HCDR1(LCDR1)-FR2-HCDR2(LCDR2)-FR3-HCDR3(LCDR3)-FR4.
  • VH or VL
  • 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 aspects, the number of amino acid changes is 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 Rabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NGH Publication 91-3242, Bethesda MD (1991), vols. 1-3.
  • immunoglobulin molecule refers to a protein having the structure of a naturally occurring antibody.
  • immunoglobulins of the IgG class are heterotetrameric glycoproteins of about 150,000 daltons, composed of two light chains and two heavy chains that are disulfide-bonded. From N- to C -terminus, each heavy chain has a variable domain (VH), also called a variable heavy domain or a heavy chain variable region, followed by three constant domains (CHI, CH2, and CH3), also called a heavy chain constant region.
  • each light chain has a variable domain (VL), also called a variable light domain or a light chain variable region, followed by a constant light (CL) domain, also called a light chain constant region.
  • VL variable domain
  • CL constant light
  • the heavy chain of an immunoglobulin may be assigned to one of five types, called a (IgA), d (IgD), e (IgE), g (IgG), or m (IgM), some of which may be further divided into subtypes, e.g. gi (IgGi), g 2 (IgG2), j3 (IgG3), j4 (IgG4), on (IgAi) and on (IgA 2 ).
  • the light chain of an immunoglobulin may be assigned to one of two types, called kappa (K) and lambda (l), based on the amino acid sequence of its constant domain.
  • K kappa
  • l lambda
  • An immunoglobulin essentially consists of two Fab molecules and an Fc domain, linked via the immunoglobulin hinge region.
  • the “class” of an antibody or immunoglobulin refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, d, e, g, and m, respectively.
  • a “Fab molecule” refers to a protein consisting of the VH and CHI domain of the heavy chain (the “Fab heavy chain”) and the VL and CL domain of the light chain (the “Fab light chain”) of an immunoglobulin.
  • crossover Fab molecule also termed “Crossfab” is meant a Fab molecule wherein the variable domains or the constant domains of the Fab heavy and light chain are exchanged (i.e. replaced by each other), i.e. the crossover Fab molecule comprises a peptide chain composed of the light chain variable domain VL and the heavy chain constant domain 1 CHI (VL-CH1, in N- to C -terminal direction), and a peptide chain composed of the heavy chain variable domain VH and the light chain constant domain CL (VH-CL, in N- to C -terminal direction).
  • the peptide chain comprising the heavy chain constant domain 1 CHI is referred to herein as the “heavy chain” of the (crossover) Fab molecule.
  • the peptide chain comprising the heavy chain variable domain VH is referred to herein as the “heavy chain” of the (crossover) Fab molecule.
  • a “conventional” Fab molecule is meant a Fab molecule in its natural format, i.e. comprising a heavy chain composed of the heavy chain variable and constant domains (VH- CH1, in N- to C -terminal direction), and a light chain composed of the light chain variable and constant domains (VL-CL, in N- to C -terminal direction).
  • Fc domain or “Fc region” herein is used to define a C -terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • antibodies produced by host cells may undergo post-translational cleavage of one or more, particularly one or two, amino acids from the C -terminus of the heavy chain.
  • an antibody produced by a host cell by expression of a specific nucleic acid molecule encoding a full- length heavy chain may include the full-length heavy chain, or it may include a cleaved variant of the full-length heavy chain.
  • This may be the case where the final two C -terminal amino acids of the heavy chain are glycine (G446) and lysine (K447, numbering according to Kabat EU index). Therefore, the C -terminal lysine (Lys447), or the C-terminal glycine (Gly446) and lysine (Lys447), of the Fc region may or may not be present.
  • a heavy chain including an Fc region (subunit) as specified herein, comprised in an antibody according to the invention comprises an additional C-terminal glycine-lysine dipeptide (G446 and K447, numbering according to Kabat EU index).
  • a heavy chain including an Fc region (subunit) as specified herein, comprised in an antibody according to the invention comprises an additional C- terminal glycine residue (G446, numbering according to Kabat EU index).
  • a “subunit” of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association.
  • a subunit of an IgGFc domain comprises an IgG CH2 and an IgG CH3 constant domain.
  • fused is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.
  • multispecific means that the antibody is able to specifically bind to at least two distinct antigenic determinants.
  • a multispecific antibody can be, for example, a bispecific antibody.
  • a bispecific antibody comprises two antigen binding sites, each of which is specific for a different antigenic determinant.
  • the multispecific (e.g. bispecific) antibody is capable of simultaneously binding two antigenic determinants, particularly two antigenic determinants expressed on two distinct cells.
  • valent denotes the presence of a specified number of antigen binding sites in an antigen binding molecule.
  • monovalent binding to an antigen denotes the presence of one (and not more than one) antigen binding site specific for the antigen in the antigen binding molecule.
  • an “antigen binding site” refers to the site, i.e. one or more amino acid residues, of an antigen binding molecule which provides interaction with the antigen.
  • the antigen binding site of an antibody comprises amino acid residues from the complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • a native immunoglobulin molecule typically has two antigen binding sites, a Fab molecule typically has a single antigen binding site.
  • antigenic determinant refers to a site (e.g. a contiguous stretch of amino acids or a conformational configuration made up of different regions of non contiguous amino acids) on a polypeptide macromolecule to which an antigen binding domain binds, forming an antigen binding domain-antigen complex.
  • Useful antigenic determinants can be found, for example, on the surfaces of tumor cells, on the surfaces of virus-infected cells, on the surfaces of other diseased cells, on the surface of immune cells, free in blood serum, and/or in the extracellular matrix (ECM).
  • ECM extracellular matrix
  • the antigen is a human protein.
  • CD3 refers to any native CD3 from any vertebrate source, including mammals such as primates (e.g. humans), non-human primates (e.g. cynomolgus monkeys) and rodents (e.g. mice and rats), unless otherwise indicated.
  • the term encompasses “full-length,” unprocessed CD3 as well as any form of CD3 that results from processing in the cell.
  • the term also encompasses naturally occurring variants of CD3, e.g., splice variants or allelic variants.
  • CD3 is human CD3, particularly the epsilon subunit of human CD3 (CD3e).
  • the amino acid sequence of human CD3e is shown in SEQ ID NO: 46 (without signal peptide).
  • CD3 is cynomolgus (Macaca fascicularis) CD3, particularly cynomolgus CD3 .
  • the amino acid sequence of cynomolgus CD3e is shown in SEQ ID NO: 47 (without signal peptide). See also NCBI GenBank no. BAB71849.1.
  • the antibody of the invention binds to an epitope of CD3 that is conserved among the CD3 antigens from different species, particularly human and cynomolgus CD3. In preferred aspects, the antibody binds to human CD 3.
  • target cell antigen refers to an antigenic determinant presented on the surface of a target cell, for example a cell in a tumor such as a cancer cell or a cell of the tumor stroma (in that case a “tumor cell antigen”).
  • the target cell antigen is not CD3, and/or is expressed on a different cell than CD3.
  • the target cell antigen is CEA, particularly human CEA.
  • CEA stands for carcinoembyonic antigen (also known as Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5)) and refers to any native CEA from any vertebrate source, including mammals such as primates (e.g. humans), non-human primates (e.g. cynomolgus monkeys) and rodents (e.g. mice and rats), unless otherwise indicated.
  • the term encompasses “full-length,” unprocessed CEA as well as any form of CEA that results from processing in the cell.
  • the term also encompasses naturally occurring variants of CEA, e.g., splice variants or allelic variants.
  • CEA is human CEA.
  • the antibody of the invention binds to an epitope of CEA that is conserved among the CEA antigens from different species, particularly human and cynomolgus CEA. In preferred aspects, the antibody binds to human CEA.
  • Binding affinity refers to the strength of the sum total of non-covalent 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., an antibody and an antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (K D ). Affinity can be measured by well-established methods known in the art, including those described herein. A preferred method for measuring affinity is Surface Plasmon Resonance (SPR).
  • SPR Surface Plasmon Resonance
  • an “affinity matured” antibody refers to an antibody with one or more alterations in one or more complementary determining regions (CDRs), 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.
  • CDRs complementary determining regions
  • “Reduced binding”, for example reduced binding to an Fc receptor refers to a decrease in affinity for the respective interaction, as measured for example by SPR. For clarity, the term includes also reduction of the affinity to zero (or below the detection limit of the analytic method), i.e. complete abolishment of the interaction.
  • “increased binding” refers to an increase in binding affinity for the respective interaction.
  • T cell activation refers to one or more cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. Suitable assays to measure T cell activation are known in the art and described herein.
  • a “modification promoting the association of the first and the second subunit of the Fc domain” is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer.
  • a modification promoting association as used herein preferably includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the modifications are complementary to each other so as to promote association of the two Fc domain subunits.
  • a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their association sterically or electrostatically favorable, respectively.
  • (hetero)dimerization occurs between a polypeptide comprising the first Fc domain subunit and a polypeptide comprising the second Fc domain subunit, which may be non-identical in the sense that further components fused to each of the subunits (e.g. antigen binding domains) are not the same.
  • the modification promoting the association of the first and the second subunit of the Fc domain comprises an amino acid mutation in the Fc domain, specifically an amino acid substitution.
  • the modification promoting the association of the first and the second subunit of the Fc domain comprises a separate amino acid mutation, specifically an amino acid substitution, in each of the two subunits of the Fc domain.
  • effector functions refers to those biological activities attributable to the Fc region of an antibody, which vary with the antibody iso type. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor), and B cell activation.
  • CDC complement dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • cytokine secretion immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor),
  • an “activating Fc receptor” is an Fc receptor that following engagement by an Fc domain of an antibody elicits signaling events that stimulate the receptor-bearing cell to perform effector functions.
  • Human activating Fc receptors include FcyRIIIa (CD 16a), FcyRI (CD64), FcyRIIa (CD32), and FcaRI (CD89).
  • Antibody-dependent cell-mediated cytotoxicity is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells.
  • the target cells are cells to which antibodies or derivatives thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region.
  • reduced ADCC is defined as either a reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or an increase in the concentration of antibody in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC.
  • the reduction in ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered.
  • the reduction in ADCC mediated by an antibody comprising in its Fc domain an amino acid substitution that reduces ADCC is relative to the ADCC mediated by the same antibody without this amino acid substitution in the Fc domain.
  • Suitable assays to measure ADCC are well known in the art (see e.g. PCT publication no. WO 2006/082515 or PCT publication no. WO 2012/130831).
  • engine engineered, engineering
  • engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches.
  • amino acid mutation as used herein is meant to encompass amino acid substitutions, deletions, insertions, and modifications. Any combination of substitution, deletion, insertion, and modification can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., reduced binding to an Fc receptor, or increased association with another peptide.
  • Amino acid sequence deletions and insertions include amino- and/or carboxy- terminal deletions and insertions of amino acids.
  • Preferred amino acid mutations are amino acid substitutions.
  • non conservative amino acid substitutions i.e. replacing one amino acid with another amino acid having different structural and/or chemical properties, are particularly preferred.
  • Amino acid substitutions include replacement by non-naturally occurring amino acids or by naturally occurring amino acid derivatives of the twenty standard amino acids (e.g. 4-hydroxyproline, 3- methylhistidine, ornithine, homoserine, 5-hydroxylysine).
  • Amino acid mutations can be generated using genetic or chemical methods well known in the art. Genetic methods may include site- directed mutagenesis, PCR, gene synthesis and the like. It is contemplated that methods of altering the side chain group of an amino acid by methods other than genetic engineering, such as chemical modification, may also be useful. Various designations may be used herein to indicate the same amino acid mutation. For example, a substitution from proline at position 329 of the Fc domain to glycine can be indicated as 329G, G329, G329, P329G, or Pro329Gly.
  • 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, Clustal W, Megalign (DNASTAR) software or the FASTA program package.
  • the percent identity values can be 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 and is described in WO 2001/007611.
  • % amino acid sequence identity values are generated using the gg search program of the FASTA package version 36.3.8c or later with a BLOSUM50 comparison matrix.
  • the FASTA program package was authored by W. R. Pearson and D. J. Lipman (“Improved Tools for Biological Sequence Analysis”, PNAS 85 (1988) 2444- 2448), W. R. Pearson (“Effective protein sequence comparison” Meth. Enzymol. 266 (1996) 227- 258), and Pearson et. al.
  • nucleic acid molecule includes any compound and/or substance that comprises a polymer of nucleotides.
  • Each nucleotide is composed of a base, specifically a purine- or pyrimidine base (i.e. cytosine (C), guanine (G), adenine (A), thymine (T) or uracil (U)), a sugar (i.e. deoxyribose or ribose), and a phosphate group.
  • cytosine C
  • G guanine
  • A adenine
  • T thymine
  • U uracil
  • the nucleic acid molecule is described by the sequence of bases, whereby said bases represent the primary structure (linear structure) of a nucleic acid molecule.
  • nucleic acid molecule encompasses deoxyribonucleic acid (DNA) including e.g., complementary DNA (cDNA) and genomic DNA, ribonucleic acid (RNA), in particular messenger RNA (mRNA), synthetic forms of DNA or RNA, and mixed polymers comprising two or more of these molecules.
  • DNA deoxyribonucleic acid
  • cDNA complementary DNA
  • RNA ribonucleic acid
  • mRNA messenger RNA
  • the nucleic acid molecule may be linear or circular.
  • nucleic acid molecule includes both, sense and antisense strands, as well as single stranded and double stranded forms.
  • the herein described nucleic acid molecule can contain naturally occurring or non-naturally occurring nucleotides.
  • nucleic acid molecules also encompass DNA and RNA molecules which are suitable as a vector for direct expression of an antibody of the invention in vitro and/or in vivo , e.g., in a host or patient.
  • DNA e.g., cDNA
  • RNA e.g., mRNA
  • mRNA can be chemically modified to enhance the stability of the RNA vector and/or expression of the encoded molecule so that mRNA can be injected into a subject to generate the antibody in vivo (see e.g., Stadler et al. (2017) Nature Medicine 23:815-817, or EP 2 101 823 Bl).
  • nucleic acid molecule refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid molecule 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 polynucleotide (or nucleic acid) encoding an antibody refers to one or more polynucleotide molecules encoding antibody heavy and light chains (or fragments thereof), including such polynucleotide molecule(s) in a single vector or separate vectors, and such polynucleotide molecule(s) present at one or more locations in a host cell.
  • 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”.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a host cell is any type of cellular system that can be used to generate the antibodies of the present invention.
  • Host cells include cultured cells, e.g.
  • the host cell of the invention is a eukaryotic cell, particularly a mammalian cell. In one aspect, the host cell is not a cell within a human body.
  • composition or “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 composition would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical composition or 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 a disease in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • 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
  • 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.
  • 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 provides antibodies that bind CD3 and CEA.
  • the antibodies show superior stability, combined with other favorable properties for therapeutic application, e.g. with respect to efficacy and safety, pharmacokinetic s, as well as produceability.
  • Antibodies of the invention as useful, e.g., for the treatment of diseases such as cancer.
  • the invention provides antibodies that bind to CD3 and CEA. In one aspect, provided are isolated antibodies that bind to CD3 and CEA. In one aspect, the invention provides antibodies that specifically bind to CD3 and CEA. In certain aspects, the anti-CD3 / CEA antibodies retain more than about 90% binding activity to CD3 after 2 weeks at pH 7.4, 37°C, relative to the binding activity after 2 weeks at pH 6, -80°C, as determined by surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises (a) a first antigen binding domain that binds to CD3, comprising a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10.
  • the antibody is a humanized antibody.
  • the first antigen binding domain is a humanized antigen binding domain (i.e. an antigen binding domain of a humanized antibody).
  • the VH and/or the VL of the first antigen binding domain is a humanized variable region.
  • the VH and/or the VL of the first antigen binding domain comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • the VH of the first antigen binding domain comprises one or more heavy chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of the heavy chain variable region sequence of SEQ ID NO: 7.
  • the VH of the first antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 7.
  • the VH of the first antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 7. In one aspect, the VH of the first antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 7. In certain aspects, a VH sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CD3. In certain aspects, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 7.
  • the VH of the first antigen binding domain comprises the amino acid sequence of SEQ ID NO: 7.
  • the VH of the first antigen binding domain comprises the amino acid sequence of SEQ ID NO: 7, including post- translational modifications of that sequence.
  • the VL of the first antigen binding domain comprises one or more light chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of the light chain variable region sequence of SEQ ID NO: 11. In one aspect, the VL of the first antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 11. In one aspect, the VL of the first antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 11. In one aspect, the VL of the first antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 11.
  • a VL sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CD3.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 11.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VL of the first antigen binding domain comprises the amino acid sequence of SEQ ID NO: 11.
  • the VL of the first antigen binding domain comprises the amino acid sequence of SEQ ID NO: 11, including post-translational modifications of that sequence.
  • the VH of the first antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 7, and the VL of the first antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 11.
  • the VH of the first antigen binding domain comprises the amino acid sequence of SEQ ID NO: 7 and the VL of the first antigen binding domain comprises the amino acid sequence of SEQ ID NO: 11.
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises a first antigen binding domain that binds to CD3 comprising a VH comprising the amino acid sequence of SEQ ID NO: 7 and a VL comprising the amino acid sequence of SEQ ID NO: 11.
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises a first antigen binding domain that binds to CD3 comprising a VH sequence of SEQ ID NO: 7 and a VL sequence of SEQ ID NO: 11.
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises a first antigen binding domain that binds to CD3 comprising a VH comprising the heavy chain CDR sequences of the VH of SEQ ID NO: 7, and a VL comprising the light chain CDR sequences of the VL of SEQ ID NO: 11.
  • the first antigen binding domain comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences of the VH of SEQ ID NO: 7 and the LCDR1, LCDR2 and LCDR3 amino acid sequences of the VL of SEQ ID NO: 11.
  • the VH of the first antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 7 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VH of SEQ ID NO: 7.
  • the VH of the first antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 7 and a framework of at least 95% sequence identity to the framework sequence of the VH of SEQ ID NO: 7.
  • the VH of the first antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 7 and a framework of at least 98% sequence identity to the framework sequence of the VH of SEQ ID NO: 7.
  • the VL of the first antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 11 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VL of SEQ ID NO: 11.
  • the VL of the first antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 11 and a framework of at least 95% sequence identity to the framework sequence of the VL of SEQ ID NO: 11.
  • the VL of the first antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 11 and a framework of at least 98% sequence identity to the framework sequence of the VL of SEQ ID NO: 11.
  • the invention provides an antibody that binds to CD3 and CEA, wherein the antibody comprises a first antigen binding domain that binds to CD3 comprising a VH sequence as in any of the aspects provided above, and a VL sequence as in any of the aspects provided above.
  • the antibody comprises a human constant region.
  • the antibody is an immunoglobulin molecule comprising a human constant region, particularly an IgG class immunoglobulin molecule comprising a human CHI, CH2, CH3 and/or CL domain.
  • Exemplary sequences of human constant domains are given in SEQ ID NOs 53 and 54 (human kappa and lambda CL domains, respectively) and SEQ ID NO: 55 (human IgGl heavy chain constant domains CH1-CH2-CH3).
  • the antibody comprises a light chain constant region comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 54, particularly the amino acid sequence of SEQ ID NO: 53.
  • the antibody comprises a heavy chain constant region comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 55.
  • the heavy chain constant region may comprise amino acid mutations in the Fc domain as described herein.
  • the first antigen binding domain comprises a human constant region.
  • the first antigen binding moiety is a Fab molecule comprising a human constant region, particularly a human CHI and/or CL domain.
  • the first antigen binding domain comprises a light chain constant region comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 54, particularly the amino acid sequence of SEQ ID NO: 53.
  • the light chain constant region may comprise amino acid mutations as described herein under “charge modifications” and/or may comprise deletion or substitutions of one or more (particularly two) N-terminal amino acids if in a crossover Fab molecule.
  • the first antigen binding domain comprises a heavy chain constant region comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the CHI domain sequence comprised in the amino acid sequence of SEQ ID NO: 55.
  • the heavy chain constant region (specifically CHI domain) may comprise amino acid mutations as described herein under “charge modifications”.
  • the antibody is a monoclonal antibody.
  • the antibody is an IgG, particularly an IgGi, antibody. In one aspect, the antibody is a full-length antibody.
  • the antibody is an antibody fragment selected from the group of an Fv molecule, a scFv molecule, a Fab molecule, and a F(ab’)2 molecule; particularly a Fab molecule.
  • the antibody fragment is a diabody, a triabody or a tetrabody.
  • the first antigen binding domain is a Fab molecule.
  • the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH or the constant domains CL and CHI, particularly the variable domains VL and VH, of the Fab light chain and the Fab heavy chain are replaced by each other (i.e. the first antigen binding domain is a crossover Fab molecule).
  • the antibody according to any of the above aspects may incorporate any of the features, singly or in combination, as described in sections II. A. 1.-8. below.
  • the antibody comprises an Fc domain, particularly an IgG Fc domain, more particularly an IgGi Fc domain.
  • the Fc domain is a human Fc domain.
  • the Fc domain is a human IgGi Fc domain.
  • the Fc domain is composed of a first and a second subunit and may incorporate any of the features, singly or in combination, described hereinbelow in relation to Fc domain variants (section II. A. 8.).
  • the antibody comprises a second and optionally a third antigen binding domain which binds to CEA (i.e. the antibody is a multispecific antibody, as further described hereinbelow (section II. A. 7.).
  • an antibody provided herein is an antibody fragment.
  • the antibody fragment is a Fab, Fab’, Fab’-SH, or F(ab’)2 molecule, in particular a Fab molecule as described herein.
  • Fab molecule
  • “Fab’ molecule” differ from Fab molecules by the addition of residues at the carboxy terminus of the CHI domain including one or more cysteines from the antibody hinge region.
  • Fab’-SH are Fab’ molecules in which the cysteine residue(s) of the constant domains bear a free thiol group. Pepsin treatment yields an F(ab') 2 molecule that has two antigen binding sites (two Fab molecules) and a part of the Fc region.
  • the antibody fragment is a diabody, a triabody or a tetrabody.
  • “Diabodies” are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al, Nat. Med. 9:129-134 (2003); and Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat. Med. 9:129-134 (2003).
  • the antibody fragment is a single chain Fab molecule.
  • a “single chain Fab molecule” or “scFab” is a polypeptide consisting of an antibody heavy chain variable domain (VH), an antibody heavy chain constant domain 1 (CHI), an antibody light chain variable domain (VL), an antibody light chain constant domain (CL) and a linker, wherein said antibody domains and said linker have one of the following orders in N-terminal to C -terminal direction: a) VH-CH1- linker-VL-CL, b) VL-CL-linker-VH-CHl, c) VH-CL-linker-VL-CHl or d) VL-CH1 -linker- VH- CL.
  • said linker is a polypeptide of at least 30 amino acids, preferably between 32 and 50 amino acids.
  • Said single chain Fab molecules are stabilized via the natural disulfide bond between the CL domain and the CHI domain.
  • these single chain Fab molecules might be further stabilized by generation of interchain disulfide bonds via insertion of cysteine residues (e.g., position 44 in the variable heavy chain and position 100 in the variable light chain according to Kabat numbering).
  • the antibody fragment is single-chain variable fragment (scFv).
  • scFv single-chain variable fragment
  • a “single-chain variable fragment” or “scFv” is a fusion protein of the variable domains of the heavy (VH) and light chains (VL) of an antibody, connected by a linker.
  • the linker is a short polypeptide of 10 to 25 amino acids and is usually rich in glycine for flexibility, as well as serine or threonine for solubility, and can either connect the N-terminus of the VH with the C -terminus of the VL, or vice versa. This protein retains the specificity of the original antibody, despite removal of the constant regions and the introduction of the linker.
  • the antibody fragment is a single-domain antibody.
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U S. Patent No. 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as recombinant production by recombinant host cells (e.g., E. coll), as described herein.
  • recombinant host cells e.g., E. coll
  • an antibody provided herein 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 the CDRs (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • 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 CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the CDR 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.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the oligosaccharide attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997).
  • the oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fiicose attached to a GlcNAc in the “stem” of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants having a non-fucosylated oligosaccharide, i.e. an oligosaccharide structure that lacks fiicose attached (directly or indirectly) to an Fc region.
  • a non-fucosylated oligosaccharide also referred to as “afucosylated” oligosaccharide
  • Such non-fucosylated oligosaccharide particularly is an N-linked oligosaccharide which lacks a fiicose residue attached to the first GlcNAc in the stem of the biantennary oligosaccharide structure.
  • antibody variants having an increased proportion of non-fiicosylated oligosaccharides in the Fc region as compared to a native or parent antibody.
  • the proportion of non-fiicosylated oligosaccharides may be at least about 20%, at least about 40%, at least about 60%, at least about 80%, or even about 100% (i.e. no fucosylated oligosaccharides are present).
  • the percentage of non-fiicosylated oligosaccharides is the (average) amount of oligosaccharides lacking fucose residues, relative to the sum of all oligosaccharides attached to Asn 297 (e. g.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (EU numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies.
  • Such antibodies having an increased proportion of non- fiicosylated oligosaccharides in the Fc region may have improved FcyRIIIa receptor binding and/or improved effector function, in particular improved ADCC function.
  • Examples of cell lines capable of producing antibodies with reduced fucosylation include Lee 13 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986); US 2003/0157108; and WO 2004/056312, especially at Example 11), and knockout cell lines, such as alpha- 1,6-fiicosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane- Ohnuki et al. Biotech. Bioeng. 87:614-622 (2004); Kanda, Y. et al., Biotechnol.
  • antibody variants are provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc.
  • Such antibody variants may have reduced fucosylation and/or improved ADCC function as described above. Examples of such antibody variants are described, e.g., in Umana et al., Nat Biotechnol 17, 176-180 (1999); Ferrara et al., Biotechn Bioeng 93, 851-861 (2006); WO 99/54342; WO 2004/065540, WO 2003/011878.
  • Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087; WO 1998/58964; and WO 1999/22764.
  • cysteine engineered antibodies e.g., THIOMABTM antibodies
  • the substituted residues occur at accessible sites of the 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.
  • Cysteine engineered antibodies may be generated as described, e.g., inU.S. Patent No. 7,521,541, 8,30,930, 7,855,275, 9,000,130, or WO 2016040856.
  • an antibody provided herein may be further modified to contain additional non- proteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • 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.
  • 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.
  • the invention also provides immunoconjugates comprising an anti-CD3 / CEA antibody herein conjugated (chemically bonded) to one or more therapeutic agents such as cytotoxic agents, chemotherapeutic agents, 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.
  • therapeutic agents such as cytotoxic agents, chemotherapeutic agents, 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 of the therapeutic agents mentioned above.
  • ADC antibody-drug conjugate
  • the antibody is typically connected to one or more of the therapeutic agents using linkers.
  • an immunoconjugate comprises an antibody of the invention 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, exotoxin A chain (
  • an immunoconjugate comprises an antibody of the invention conjugated to a radioactive atom to form a radioconjugate.
  • a radioactive atom to form a radioconjugate.
  • 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 Tc 99m or I 123 , or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, MRI), such as I 123 , 1 131 , In 111 , F 19 , C 13 , N 15 , O 17 , gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of an antibody and cytotoxic agent may be made using a variety ofbifunctional 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 HC1), 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
  • a ricin immunotoxin can be prepared as described in Vitetta et al, Science 238:1098 (1987).
  • Carbon- 14-labeled 1- i sothiocyanatobenzy 1-3 - methyl di ethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody.
  • MX-DTPA 1- i sothiocyanatobenzy 1-3 - methyl di ethylene triaminepentaacetic acid
  • 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. Patent 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, IL., U.S.A).
  • cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS,
  • An antibody provided herein is a multispecific antibody, particularly a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different antigenic determinants (e.g., two different proteins, or two different epitopes on the same protein).
  • the multispecific antibody has three or more binding specificities.
  • one of the binding specificities is for CD3 and the other specificity is for CEA.
  • Multispecific antibodies may be prepared as full length antibodies or antibody fragments.
  • Techniques for making multispecific antibodies include, but are not limited to, recombinant co expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)) and “knob-in-hole” engineering (see, e.g., U S. Patent No. 5,731,168, and Atwell et al, J. Mol. Biol. 270:26 (1997)).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc- heterodimeric molecules (see, e.g., WO 2009/089004); cross-linking two or more antibodies or fragments (see, e.g., US Patent No.
  • Engineered antibodies with three or more antigen binding sites including for example, “Octopus antibodies”, or DVD-Ig are also included herein (see, e.g., WO 2001/77342 and WO 2008/024715).
  • Other examples of multispecific antibodies with three or more antigen binding sites can be found in WO 2010/115589, WO 2010/112193, WO 2010/136172, WO 2010/145792, and WO 2013/026831.
  • the multispecific antibody or antigen binding fragment thereof also includes a “Dual Acting FAb” or “DAF” comprising an antigen binding site that binds to CD3 as well as another different antigen, or two different epitopes of CD3 (see, e.g., US 2008/0069820 and WO 2015/095539).
  • Multi-specific antibodies may also be provided in an asymmetric form with a domain crossover in one or more binding arms of the same antigen specificity (so-called “CrossMab” technology), i.e. by exchanging the VH/VL domains (see e.g., WO 2009/080252 and WO 2015/150447), the CHI/CL domains (see e.g., WO 2009/080253) or the complete Fab arms (see e.g., WO 2009/080251, WO 2016/016299, also see Schaefer et al, PNAS, 108 (2011) 1187-1191, and Klein at al., MAbs 8 (2016) 1010-20).
  • Asymmetrical Fab arms can also be engineered by introducing charged or non-charged amino acid mutations into domain interfaces to direct correct Fab pairing. See e g., WO 2016/172485.
  • a particular type of multi specific antibodies are bispecific antibodies designed to simultaneously bind to a surface antigen on a target cell, e.g., a tumor cell, and to an activating, invariant component of the T cell receptor (TCR) complex, such as CD3, for retargeting of T cells to kill target cells.
  • a target cell e.g., a tumor cell
  • an activating, invariant component of the T cell receptor (TCR) complex such as CD3, for retargeting of T cells to kill target cells.
  • TCR T cell receptor
  • the antibody provided herein is a multispecific antibody, particularly a bispecific antibody, wherein one of the binding specificities is for CD3 and the other is for CEA as the target cell antigen.
  • bispecific antibody formats examples include, but are not limited to, the so-called “BiTE” (bispecific T cell engager) molecules wherein two scFv molecules are fused by a flexible linker (see, e.g., WO 2004/106381, WO 2005/061547, WO 2007/042261, and WO 2008/119567, Nagorsen and Bauerle, Exp Cell Res 317, 1255-1260 (2011)); diabodies (Holliger et al., Prot Eng 9, 299-305 (1996)) and derivatives thereof, such as tandem diabodies (“TandAb”; Kipriyanov et al., J Mol Biol 293, 41-56 (1999)); “DART” (dual affinity retargeting) molecules which are based on the diabody format but feature a C -terminal disulfide bridge for additional stabilization (Johnson et al., J Mol Biol 399, 436-449 (2010)), and so-called triomabs, which are
  • the invention provides an antibody that binds to CD3 and CEA, comprising a first antigen binding domain that binds to CD3, as described herein, and comprising a second and optionally a third antigen binding domain that binds to CEA.
  • the antigen binding domains comprised in the antibody are Fab molecules (i.e. antigen binding domains composed of a heavy and a light chain, each comprising a variable and a constant domain).
  • the first, the second and/or, where present, the third antigen binding domain is a Fab molecule.
  • said Fab molecule is human.
  • said Fab molecule is humanized.
  • said Fab molecule comprises human heavy and light chain constant domains.
  • at least one of the antigen binding domains is a crossover Fab molecule.
  • Such modification reduces mispairing of heavy and light chains from different Fab molecules, thereby improving the yield and purity of the (multispecific) antibody of the invention in recombinant production.
  • the variable domains of the Fab light chain and the Fab heavy chain (VL and VH, respectively) are exchanged. Even with this domain exchange, however, the preparation of the (multispecific) antibody may comprise certain side products due to a so-called Bence Jones-type interaction between mispaired heavy and light chains (see Schaefer et al, PNAS, 108 (2011) 11187-11191).
  • charged amino acids with opposite charges may be introduced at specific amino acid positions in the CHI and CL domains of either the Fab molecule binding to CD3, or the Fab molecule(s) binding to CEA, as further described herein.
  • Charge modifications are made either in the conventional Fab molecule(s) comprised in the (multispecific) antibody (such as shown e.g. in Figures 1 A-C, G- J), or in the VH/VL crossover Fab molecule(s) comprised in the (multispecific) antibody (such as shown e.g. in Figure 1 D-F, K-N) (but not in both).
  • the charge modifications are made in the conventional Fab molecule(s) comprised in the (multispecific) antibody (which in preferred aspects bind(s) to CEA).
  • the (multispecific) antibody is capable of simultaneous binding to CD3 and CEA.
  • the (multispecific) antibody is capable of crosslinking a T cell and a target cell by simultaneous binding to CD3 and CEA.
  • such simultaneous binding results in lysis of the target cell, particularly a CEA- expressing target cell such as a tumor cell.
  • such simultaneous binding results in activation of the T cell.
  • such simultaneous binding results in a cellular response ofa 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 (multispecific) antibody to CD3 without simultaneous binding to CEA does not result in T cell activation.
  • the (multispecific) 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 and/or specificity of the T cell.
  • a T cell according to any of the aspects of the invention is a cytotoxic T cell.
  • the T cell is a CD4 + or a CD8 + T cell, particularly a CD8 + T cell.
  • the (multispecific) antibody of the invention comprises at least one antigen binding domain (the first antigen binding domain) that binds to CD3.
  • CD3 is human CD3 (SEQ ID NO: 46) or cynomolgus CD3 (SEQ ID NO: 47) most particularly human CD3.
  • the first antigen binding domain is cross-reactive for (i.e. specifically binds to) human and cynomolgus CD3.
  • CD3 is the epsilon subunit of CD3 (CD3 epsilon).
  • the (multispecific) antibody comprises not more than one antigen binding domain that binds to CD3. In one aspect the (multispecific) antibody provides monovalent binding to CD3.
  • the antigen binding domain that binds to CD3 is an antibody fragment selected from the group of an Fv molecule, a scFv molecule, a Fab molecule, and a F(ab’)2 molecule.
  • the antigen binding domain that binds to CD3 is a Fab molecule.
  • the antigen binding domain that binds to CD3 is a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CHI and CL of the Fab heavy and light chains are exchanged / replaced by each other.
  • the antigen binding domain(s) that binds to CEA is preferably a conventional Fab molecule.
  • the antigen binding domain that binds to CD3 preferably is a crossover Fab molecule and the antigen binding domain that bind to CEA are conventional Fab molecules.
  • the antigen binding domain that binds to CD3 is a conventional Fab molecule.
  • the antigen binding domain(s) that binds CEA is a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CHI and CL of the Fab heavy and light chains are exchanged / replaced by each other.
  • the antigen binding domain that binds to CEA preferably is a crossover Fab molecule and the antigen binding domains that bind to CD3 are conventional Fab molecules.
  • the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH or the constant domains CL and CHI, particularly the variable domains VL and VH, of the Fab light chain and the Fab heavy chain are replaced by each other (i.e. according to such aspect, the first antigen binding domain is a crossover Fab molecule wherein the variable or constant domains of the Fab light chain and the Fab heavy chain are exchanged).
  • the second (and the third, if any) antigen binding domain is a conventional Fab molecule. In one aspect, not more than one antigen binding domain that binds to CD3 is present in the (multispecific) antibody (i.e. the antibody provides monovalent binding to CD3). b) Second (and third) antigen binding domain
  • the (multispecific) antibody of the invention comprises at least one antigen binding domain (the second and optionally the third antigen binding domain), particularly a Fab molecule, that binds to CEA.
  • the second antigen binding domain is able to direct the (multispecific) antibody to a target site, for example to a specific type of cell that expresses CEA.
  • the antigen binding domain that binds to CEA is an antibody fragment selected from the group of an Fv molecule, a scFv molecule, a Fab molecule, and a F(ab’)2 molecule.
  • the antigen binding domain that binds to CEA is a Fab molecule.
  • the (multispecific) antibody comprises two antigen binding domains, particularly Fab molecules, that bind to CEA.
  • all of these antigen binding domains are identical, i.e. they have the same molecular format (e.g. conventional or crossover Fab molecule) and comprise the same amino acid sequences including the same amino acid substitutions in the CHI and CL domain as described herein (if any).
  • the (multispecific) antibody comprises not more than two antigen binding domains, particularly Fab molecules, that bind to CEA.
  • the antigen binding domain(s) that bind to CEA is/are a conventional Fab molecule.
  • the antigen binding domain(s) that binds to CD3 is a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CHI and CL of the Fab heavy and light chains are exchanged / replaced by each other.
  • the antigen binding domain(s) that bind to CEA is/are a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CHI and CL of the Fab heavy and light chains are exchanged / replaced by each other.
  • the antigen binding domain(s) that binds to CD3 is a conventional Fab molecule.
  • the second (and, where present, third) antigen binding domain comprises a human constant region.
  • the second (and, where present, third) antigen binding domain is a Fab molecule comprising a human constant region, particularly a human CHI and/or CL domain.
  • Exemplary sequences of human constant domains are given in SEQ ID NOs 53 and 54 (human kappa and lambda CL domains, respectively) and SEQ ID NO: 55 (human IgGi heavy chain constant domains CH1-CH2-CH3).
  • the second (and, where present, third) antigen binding domain comprises a light chain constant region comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 54, particularly the amino acid sequence of SEQ ID NO: 53.
  • the light chain constant region may comprise amino acid mutations as described herein under “charge modifications” and/or may comprise deletion or substitutions of one or more (particularly two) N-terminal amino acids if in a crossover Fab molecule.
  • the second (and, where present, third) antigen binding domain comprises a heavy chain constant region comprising an amino acid sequence that is at least about 95%, 96%, 97%, 98%, 99% or 100% identical to the CHI domain sequence comprised in the amino acid sequence of SEQ ID NO: 55.
  • the heavy chain constant region (specifically CHI domain) may comprise amino acid mutations as described herein under “charge modifications”.
  • the second (and, where present, the third) antigen binding domain comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 15 , a HCDR 2 of SEQ ID NO : 16, and a HCDR 3 of SEQ ID NO: 17, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 19, a LCDR 2 of SEQ ID NO: 20 and a LCDR 3 of SEQ ID NO: 21.
  • VH heavy chain variable region
  • HCDR heavy chain complementary determining region
  • VL light chain variable region
  • the second (and, where present, third) antigen binding domain is (derived from) a humanized antibody. In one aspect, the second (and, where present, third) antigen binding domain is a humanized antigen binding domain (i.e. an antigen binding domain of a humanized antibody). In one aspect, the VH and/or the VL of the second (and, where present, third) antigen binding domain is a humanized variable region.
  • the VH and/or the VL of the second (and, where present, third) antigen binding domain comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • the VH of the second (and, where present, the third) antigen binding domain comprises one or more heavy chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of SEQ ID NO: 18. In one aspect, the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 18. In one aspect, the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 18.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 18.
  • a VH sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CEA.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 18.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 18.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 18, including post-translational modifications of that sequence.
  • the VL of the second (and, where present, the third) antigen binding domain comprises one or more light chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of SEQ ID NO: 22.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 22.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 22.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 22.
  • a VL sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CEA.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 22.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 22.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 22, including post-translational modifications of that sequence.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 18, and the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 22.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 18 and the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 22.
  • the second (and, where present, the third) antigen binding domain comprises a VH comprising the sequence of SEQ ID NO: 18 and a VL comprising the sequence of SEQ ID NO: 22.
  • the second (and, where present, the third) antigen binding domain comprises a VH sequence of SEQ ID NO: 18 and a VL sequence of SEQ ID NO: 22.
  • the second (and, where present, the third) antigen binding domain comprises a VH comprising the heavy chain CDR sequences of the VH of SEQ ID NO: 18, and a VL comprising the light chain CDR sequences of the VL of SEQ ID NO: 22.
  • the second (and, where present, the third) antigen binding domain comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences of the VH of SEQ ID NO: 18 and the LCDR1, LCDR2 and LCDR3 amino acid sequences of the VL of SEQ ID NO: 22.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 18 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VH of SEQ ID NO: 18.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 18 and a framework of at least 95% sequence identity to the framework sequence of the VH of SEQ ID NO: 18.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 18 and a framework of at least 98% sequence identity to the framework sequence of the VH of SEQ ID NO: 18.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 22 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VL of SEQ ID NO: 22.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 22 and a framework of at least 95% sequence identity to the framework sequence of the VL of SEQ ID NO: 22.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 22 and a framework of at least 98% sequence identity to the framework sequence of the VL of SEQ ID NO: 22.
  • the second (and, where present, the third) antigen binding domain comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 28, a HCDR 2 of SEQ ID NO: 29, and a HCDR 3 of SEQ ID NO: 30, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 32, a LCDR 2 of SEQ ID NO: 33 and a LCDR 3 of SEQ ID NO: 34.
  • VH heavy chain variable region
  • HCDR heavy chain complementary determining region
  • VL light chain variable region
  • the second (and, where present, third) antigen binding domain is (derived from) a humanized antibody. In one aspect, the second (and, where present, third) antigen binding domain is a humanized antigen binding domain (i.e. an antigen binding domain of a humanized antibody). In one aspect, the VH and/or the VL of the second (and, where present, third) antigen binding domain is a humanized variable region.
  • the VH and/or the VL of the second (and, where present, third) antigen binding domain comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • the VH of the second (and, where present, the third) antigen binding domain comprises one or more heavy chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of SEQ ID NO: 31.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 31.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 31.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 31.
  • a VH sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CEA.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 31.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 31.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 31, including post-translational modifications of that sequence.
  • the VL of the second (and, where present, the third) antigen binding domain comprises one or more light chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of SEQ ID NO: 35. In one aspect, the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 35. In one aspect, the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 35.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 35.
  • a VL sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CEA.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 35.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 35.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 35, including post-translational modifications of that sequence.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 31, and the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 35.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 31 and the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 35.
  • the second (and, where present, the third) antigen binding domain comprises a VH comprising the sequence of SEQ ID NO: 31 and a VL comprising the sequence of SEQ ID NO: 35.
  • the second (and, where present, the third) antigen binding domain comprises a VH sequence of SEQ ID NO: 31 and a VL sequence of SEQ ID NO: 35.
  • the second (and, where present, the third) antigen binding domain comprises a VH comprising the heavy chain CDR sequences of the VH of SEQ ID NO: 31, and a VL comprising the light chain CDR sequences of the VL of SEQ ID NO: 35.
  • the second (and, where present, the third) antigen binding domain comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences of the VH of SEQ ID NO: 31 and the LCDR1, LCDR2 and LCDR3 amino acid sequences of the VL of SEQ ID NO: 35.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 31 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VH of SEQ ID NO: 31.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 31 and a framework of at least 95% sequence identity to the framework sequence of the VH of SEQ ID NO: 31.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 31 and a framework of at least 98% sequence identity to the framework sequence of the VH of SEQ ID NO: 31.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 35 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VL of SEQ ID NO: 35.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 35 and a framework of at least 95% sequence identity to the framework sequence of the VL of SEQ ID NO: 35.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 35 and a framework of at least 98% sequence identity to the framework sequence of the VL of SEQ ID NO: 35.
  • the second (and, where present, the third) antigen binding domain comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 36, a HCDR 2 of SEQ ID NO: 37, and a HCDR 3 of SEQ ID NO: 38, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 40, a LCDR 2 of SEQ ID NO: 41 and a LCDR 3 of SEQ ID NO:
  • VH heavy chain variable region
  • HCDR heavy chain complementary determining region
  • VL light chain variable region
  • LCDR light chain complementarity determining region
  • the second (and, where present, third) antigen binding domain is (derived from) a humanized antibody.
  • the second (and, where present, third) antigen binding domain is a humanized antigen binding domain (i.e. an antigen binding domain of a humanized antibody).
  • the VH and/or the VL of the second (and, where present, third) antigen binding domain is a humanized variable region.
  • the VH and/or the VL of the second (and, where present, third) antigen binding domain comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • the VH of the second (and, where present, the third) antigen binding domain comprises one or more heavy chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of SEQ ID NO: 39.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 39.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 39.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 39.
  • a VH sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CEA.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 39.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 39.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 39, including post-translational modifications of that sequence.
  • the VL of the second (and, where present, the third) antigen binding domain comprises one or more light chain framework sequence (i.e. the FR1, FR2, FR3 and/or FR4 sequence) of SEQ ID NO: 43.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 43.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95% identical to the amino acid sequence of SEQ ID NO: 43.
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 98% identical to the amino acid sequence of SEQ ID NO: 43.
  • a VL sequence having at least 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody comprising that sequence retains the ability to bind to CEA.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the amino acid sequence of SEQ ID NO: 43.
  • substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs).
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 43.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 43, including post-translational modifications of that sequence.
  • the VH of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 39
  • the VL of the second (and, where present, the third) antigen binding domain comprises an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 43.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 39
  • the VL of the second (and, where present, the third) antigen binding domain comprises the amino acid sequence of SEQ ID NO: 43.
  • the second (and, where present, the third) antigen binding domain comprises a VH comprising the sequence of SEQ ID NO: 39 and a VL comprising the sequence of SEQ ID NO: 43.
  • the second (and, where present, the third) antigen binding domain comprises a VH sequence of SEQ ID NO: 39 and a VL sequence of SEQ ID NO: 43.
  • the second (and, where present, the third) antigen binding domain comprises a VH comprising the heavy chain CDR sequences of the VH of SEQ ID NO: 39, and a VL comprising the light chain CDR sequences of the VL of SEQ ID NO: 43.
  • the second (and, where present, the third) antigen binding domain comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences of the VH of SEQ ID NO: 39 and the LCDR1, LCDR2 and LCDR3 amino acid sequences of the VL of SEQ ID NO: 43.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 39 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VH of SEQ ID NO: 39.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 39 and a framework of at least 95% sequence identity to the framework sequence of the VH of SEQ ID NO: 39.
  • the VH of the second (and, where present, the third) antigen binding domain comprises the heavy chain CDR sequences of the VH of SEQ ID NO: 39 and a framework of at least 98% sequence identity to the framework sequence of the VH of SEQ ID NO: 39.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 43 and a framework of at least 95%, 96%, 97%, 98% or 99% sequence identity to the framework sequence of the VL of SEQ ID NO: 43.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 43 and a framework of at least 95% sequence identity to the framework sequence of the VL of SEQ ID NO: 43.
  • the VL of the second (and, where present, the third) antigen binding domain comprises the light chain CDR sequences of the VL of SEQ ID NO: 43 and a framework of at least 98% sequence identity to the framework sequence of the VL of SEQ ID NO: 43.
  • the (multispecific) antibody of the invention may comprise amino acid substitutions in Fab molecules comprised therein which are particularly efficient in reducing mispairing of light chains with non-matching heavy chains (Bence-Jones-type side products), which can occur in the production ofFab-based multispecific antibodies with a VH/VL exchange in one (or more, in case of molecules comprising more than two antigen-binding Fab molecules) of their binding arms (see also PCT publication no. WO 2015/150447, particularly the examples therein, incorporated herein by reference in its entirety).
  • the ratio of a desired (multispecific) antibody compared to undesired side products can be improved by the introduction of charged amino acids with opposite charges at specific amino acid positions in the CHI and CL domains (sometimes referred to herein as “charge modifications”).
  • the first and the second (and, where present, the third) antigen binding domain of the (multispecific) antibody are both Fab molecules, and in one of the antigen binding domains (particularly the first antigen binding domain) the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, i) in the constant domain CL of the second (and, where present, the third) antigen binding domain the amino acid at position 124 is substituted by a positively charged amino acid (numbering according to Kabat), and wherein in the constant domain CHI of the second (and, where present, the third) antigen binding domain the amino acid at position 147 or the amino acid at position 213 is substituted by a negatively charged amino acid (numbering according to Kabat EU index); or ii) in the constant domain CL of the first antigen binding domain the amino acid at position 124 is substituted by a positively charged amino acid (numbering according to Kabat), and wherein in the constant domain CHI of the first antigen binding domain the amino acid at position 124
  • the (multispecific) antibody does not comprise both modifications mentioned under i) and ii).
  • the constant domains CL and CHI of the antigen binding domain having the VH/VL exchange are not replaced by each other (i.e. remain unexchanged).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CHI of the second (and, where present, the third) antigen binding domain the amino acid at position 147 or the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index); or ii) in the constant domain CL of the first antigen binding domain the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CHI of the first antigen binding domain the amino acid at position 147 or the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat) (numbering according to Kabat).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CHI of the second (and, where present, the third) antigen binding domain the amino acid at position 147 or the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CHI of the second (and, where present, the third) antigen binding domain the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) and the amino acid at position 123 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CHI of the second (and, where present, the third) antigen binding domain the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted by lysine (K) (numbering according to Kabat) and the amino acid at position 123 is substituted by lysine (K) (numbering according to Kabat)
  • the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted by lysine (K) (numbering according to Kabat) and the amino acid at position 123 is substituted by arginine (R) (numbering according to Kabat), and in the constant domain CHI of the second (and, where present, the third) antigen binding domain the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Kabat EU index).
  • the constant domain CL of the second (and, where present, the third) antigen binding domain is of kappa isotype.
  • the amino acid substitutions according to the above aspects may be made in the constant domain CL and the constant domain CHI of the first antigen binding domain instead of in the constant domain CL and the constant domain CHI of the second (and, where present, the third) antigen binding domain.
  • the constant domain CL of the first antigen binding domain is of kappa isotype.
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Rabat), and in the constant domain CHI of the first antigen binding domain the amino acid at position 147 or the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Rabat EU index).
  • the amino acid at position 124 is substituted independently by lysine (R), arginine (R) or histidine (H) (numbering according to Rabat), and in the constant domain CHI of the first antigen binding domain the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Rabat EU index).
  • the amino acid at position 124 is substituted independently by lysine (R), arginine (R) or histidine (H) (numbering according to Rabat) and the amino acid at position 123 is substituted independently by lysine (R), arginine (R) or histidine (H) (numbering according to Rabat), and in the constant domain CHI of the first antigen binding domain the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Rabat EU index) and the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Rabat EU index).
  • the amino acid at position 124 is substituted by lysine (R) (numbering according to Rabat) and the amino acid at position 123 is substituted by lysine (R) (numbering according to Rabat), and in the constant domain CHI of the first antigen binding domain the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Rabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Rabat EU index).
  • the amino acid at position 124 is substituted by lysine (R) (numbering according to Rabat) and the amino acid at position 123 is substituted by arginine (R) (numbering according to Rabat), and in the constant domain CHI of the first antigen binding domain the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Rabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Rabat EU index).
  • the (multispecific) antibody of the invention comprises
  • the (multispecific) antibody according to the present invention can have a variety of configurations. Exemplary configurations are depicted in Figure 1.
  • the antigen binding domains comprised in the (multispecific) antibody are Fab molecules.
  • the first, second, third etc. antigen binding domain may be referred to herein as first, second, third etc. Fab molecule, respectively.
  • the first and the second antigen binding domain of the (multispecific) antibody are fused to each other, optionally via a peptide linker.
  • the first and the second antigen binding domain are each a Fab molecule.
  • the first antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain.
  • the second antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain.
  • the first antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain or (ii) the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain, additionally the Fab light chain of the first antigen binding domain and the Fab light chain of the second antigen binding domain may be fused to each other, optionally via a peptide linker.
  • a (multispecific) antibody with a single antigen binding domain capable of specific binding to a second antigen, e.g. a target cell antigen such as CEA, (for example as shown in Figure 1A, D, G, H, K, L) is useful, particularly in cases where internalization of the second antigen is to be expected following binding of a high affinity antigen binding domain.
  • a target cell antigen such as CEA
  • the presence of more than one antigen binding domain specific for the second antigen may enhance internalization of the second antigen, thereby reducing its availability.
  • a (multispecific) antibody comprising two or more antigen binding domains (such as Fab molecules) specific for a second antigen, e.g. a target cell antigen such as CEA (see examples shown in Figure IB, 1C, IE, IF, II, 1J, 1M or IN), for example to optimize targeting to the target site or to allow crosslinking of target cell antigens.
  • a target cell antigen such as CEA
  • the (multispecific) antibody according to the present invention comprises a third antigen binding domain.
  • the third antigen binding domain binds to CEA. In one aspect, the third antigen binding domain is a Fab molecule.
  • the third antigen domain is identical to the second antigen binding domain.
  • the third and the second antigen binding domain are each a Fab molecule and the third antigen binding domain is identical to the second antigen binding domain.
  • the second and the third antigen binding domain comprise the same heavy and light chain amino acid sequences and have the same arrangement of domains (i.e. conventional or crossover).
  • the third antigen binding domain comprises the same amino acid substitutions, if any, as the second antigen binding domain.
  • charge modifications will be made in the constant domain CL and the constant domain CHI of each of the second antigen binding domain and the third antigen binding domain.
  • said amino acid substitutions may be made in the constant domain CL and the constant domain CHI of the first antigen binding domain (which in preferred aspects is also a Fab molecule), but not in the constant domain CL and the constant domain CHI of the second antigen binding domain and the third antigen binding domain.
  • the third antigen binding domain preferably is a conventional Fab molecule.
  • the second and the third antigen binding domains are crossover Fab molecules (and the first antigen binding domain is a conventional Fab molecule) are, however, also contemplated.
  • the second and the third antigen binding domains are each a conventional Fab molecule
  • the first antigen binding domain is a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CL and CHI of the Fab heavy and light chains are exchanged / replaced by each other.
  • the second and the third antigen binding domains are each a crossover Fab molecule and the first antigen binding domain is a conventional Fab molecule.
  • the first antigen domain binds to CD3, and the second and third antigen binding domain bind to CEA.
  • the (multispecific) antibody of the invention comprises an Fc domain composed of a first and a second subunit.
  • the first and the second subunit of the Fc domain are capable of stable association.
  • the (multispecific) antibody according to the invention can have different configurations, i.e. the first, second (and optionally third) antigen binding domain may be fused to each other and to the Fc domain in different ways.
  • the components may be fused to each other directly or, preferably, via one or more suitable peptide linkers. Where fusion of a Fab molecule is to the N-terminus of a subunit of the Fc domain, it is typically via an immunoglobulin hinge region.
  • the first and the second antigen binding domain are each a Fab molecule and the first antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • the second antigen binding domain may be fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain or to the N-terminus of the other one of the subunits of the Fc domain.
  • the second antigen binding domain is a conventional Fab molecule
  • the first antigen binding domain is a crossover Fab molecule as described herein, i.e.
  • the second antigen binding domain is a crossover Fab molecule and the first antigen binding domain is a conventional Fab molecule.
  • the first and the second antigen binding domain are each a Fab molecule, the first antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain, and the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain.
  • the (multispecific) antibody essentially consists of the first and the second Fab molecule, the Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first Fab molecule, and the first Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • Such a configuration is schematically depicted in Figures 1G and IK (with the first antigen binding domain in these examples being a VH/VL crossover Fab molecule).
  • the Fab light chain of the first Fab molecule and the Fab light chain of the second Fab molecule may additionally be fused to each other.
  • the first and the second antigen binding domain are each a Fab molecule and the first and the second antigen binding domain are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain.
  • the (multispecific) antibody essentially consists of the first and the second Fab molecule, the Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first and the second Fab molecule are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain.
  • first antigen binding domain being a VH/VL crossover Fab molecule and the second antigen binding domain being a conventional Fab molecule.
  • the first and the second Fab molecule may be fused to the Fc domain directly or through a peptide linker.
  • the first and the second Fab molecule are each fused to the Fc domain through an immunoglobulin hinge region.
  • the immunoglobulin hinge region is a human IgGi hinge region, particularly where the Fc domain is an IgGi Fc domain.
  • the first and the second antigen binding domain are each a Fab molecule and the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • the first antigen binding domain may be fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain or (as described above) to the N-terminus of the other one of the subunits of the Fc domain.
  • said second antigen binding domain is a conventional Fab molecule
  • the first antigen binding domain is a crossover Fab molecule as described herein, i.e.
  • said second antigen binding domain is a crossover Fab molecule and the first antigen binding domain is a conventional Fab molecule.
  • the first and the second antigen binding domain are each a Fab molecule
  • the second antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain
  • the first antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain.
  • the (multispecific) antibody essentially consists of the first and the second Fab molecule, the Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second Fab molecule, and the second Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.
  • Such a configuration is schematically depicted in Figures 1H and 1L (in these examples with the first antigen binding domain being a VH/VL crossover Fab molecule and the second antigen binding domain being a conventional Fab molecule).
  • the Fab light chain of the first Fab molecule and the Fab light chain of the second Fab molecule may additionally be fused to each other.
  • a third antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.
  • said second and third antigen binding domains are each a conventional Fab molecule
  • the first antigen binding domain is a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CL and CHI of the Fab heavy and light chains are exchanged / replaced by each other.
  • said second and third antigen binding domains are each a crossover Fab molecule and the first antigen binding domain is a conventional Fab molecule.
  • the first and the third antigen binding domain are each fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the second antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first Fab molecule.
  • the (multispecific) antibody essentially consists of the first, the second and the third Fab molecule, the Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first Fab molecule, and the first Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain, and wherein the third Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the second subunit of the Fc domain.
  • Such a configuration is schematically depicted in Figure IB and IE (in these examples with the first antigen binding domain being a VH/VL crossover Fab molecule, and the second and the third antigen binding domain being a conventional Fab molecule), and Figure 1 J and IN (in these examples with the first antigen binding domain being a conventional Fab molecule, and the second and the third antigen binding domain being a VH/VL crossover Fab molecule).
  • the first and the third Fab molecule may be fused to the Fc domain directly or through a peptide linker.
  • the first and the third Fab molecule are each fused to the Fc domain through an immunoglobulin hinge region.
  • the immunoglobulin hinge region is a human IgGi hinge region, particularly where the Fc domain is an IgGi Fc domain.
  • the Fab light chain of the first Fab molecule and the Fab light chain of the second Fab molecule may additionally be fused to each other.
  • the second and the third antigen binding domain are each fused at the C- terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the first antigen binding domain is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain.
  • the (multispecific) antibody essentially consists of the first, the second and the third Fab molecule, the Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second Fab molecule, and the second Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain, and wherein the third Fab molecule is fused at the C -terminus of the Fab heavy chain to the N-terminus of the second subunit of the Fc domain.
  • Such a configuration is schematically depicted in Figure 1C and IF (in these examples with the first antigen binding domain being a VH/VL crossover Fab molecule, and the second and the third antigen binding domain being a conventional Fab molecule) and in Figure II and 1M (in these examples with the first antigen binding domain being a conventional Fab molecule, and the second and the third antigen binding domain being a VH/VL crossover Fab molecule).
  • the second and the third Fab molecule may be fused to the Fc domain directly or through a peptide linker.
  • the second and the third Fab molecule are each fused to the Fc domain through an immunoglobulin hinge region.
  • the immunoglobulin hinge region is a human IgGi hinge region, particularly where the Fc domain is an IgGi Fc domain.
  • the Fab light chain of the first Fab molecule and the Fab light chain of the second Fab molecule may additionally be fused to each other.
  • the two Fab molecules, the hinge regions and the Fc domain essentially form an immunoglobulin molecule.
  • the immunoglobulin molecule is an IgG class immunoglobulin.
  • the immunoglobulin is an IgGi subclass immunoglobulin.
  • the immunoglobulin is an IgG 4 subclass immunoglobulin.
  • the immunoglobulin is a human immunoglobulin.
  • the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
  • the immunoglobulin comprises a human constant region, particularly a human Fc region.
  • the Fab light chain of the first Fab molecule and the Fab light chain of the second Fab molecule are fused to each other, optionally via a peptide linker.
  • the Fab light chain of the first Fab molecule may be fused at its C-terminus to the N-terminus of the Fab light chain of the second Fab molecule, or the Fab light chain of the second Fab molecule may be fused at its C-terminus to the N-terminus of the Fab light chain of the first Fab molecule. Fusion of the Fab light chains of the first and the second Fab molecule further reduces mispairing of unmatched Fab heavy and light chains, and also reduces the number of plasmids needed for expression of some of the (multispecific) antibody of the invention.
  • the antigen binding domains may be fused to the Fc domain or to each other directly or through a peptide linker, comprising one or more amino acids, typically about 2-20 amino acids.
  • Peptide linkers are known in the art and are described herein. Suitable, non-immunogenic peptide linkers include, for example, (G4S) n , (SG4)n, (G4S) n , G4(SG4)n or (G4S) n Gs peptide linkers “n” is generally an integer from 1 to 10, typically from 2 to 4.
  • said peptide linker has a length of at least 5 amino acids, in one aspect a length of 5 to 100, in a further aspect of 10 to 50 amino acids.
  • said peptide linker is (G 4 S) 2 .
  • said peptide linker is G4SG5.
  • a particularly suitable peptide linker for fusing the Fab light chains of the first and the second Fab molecule to each other is (G 4 S) 2 .
  • An exemplary peptide linker suitable for connecting the Fab heavy chains of the first and the second Fab fragments comprises the sequence (D)-(G 4 S) 2 (SEQ ID NOs 49 and 50).
  • Another suitable such linker comprises the sequence (D)-G 4 SGs (SEQ ID NOs 51 and 52).
  • linkers may comprise (a portion of) an immunoglobulin hinge region. Particularly where a Fab molecule is fused to the N-terminus of an Fc domain subunit, it may be fused via an immunoglobulin hinge region or a portion thereof, with or without an additional peptide linker.
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e.
  • the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with an Fc domain subunit (VL (i) -CHf (i) -CH2-CH3(-CH4)), and a polypeptide wherein the Fab heavy chain of the second Fab molecule shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (2) -CH1 (2) -CH2-CH3(-CH4)).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VH (i) -CL (i) ) and the Fab light chain polypeptide of the second Fab molecule (VLp ) - CL ( 2 ) ).
  • the polypeptides are covalently linked, e.g., by a disulfide bond.
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e.
  • the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (i) -CL (i) -CH2-CH3(-CH4)), and a polypeptide wherein the Fab heavy chain of the second Fab molecule shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (2) -CH1 (2) -CH2-CH3(-CH4)).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (VL (i) -CHl (i) ) and the Fab light chain polypeptide of the second Fab molecule (VLp ) - CL ( 2 ) ).
  • the polypeptides are covalently linked, e.g., by a disulfide bond.
  • the (multispecific) antibody comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with an Fc domain subunit (VL (i) -CHl (i) -VH (2) -CHl (2) -CH2-CH3(-CH4)).
  • VL (i) -CHl (i) -VH (2) -CHl (2) -CH2-CH3(-CH4) an Fc domain subunit
  • the (multispecific) antibody comprises a polypeptide wherein the Fab heavy chain of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain variable region of the first Fab molecule which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (2) -CHl (2) -VL (i) -CHl (i) -CH2- CH3(-CH4)).
  • VH (2) -CHl (2) -VL (i) -CHl (i) -CH2- CH3(-CH4) an Fc domain subunit
  • the (multispecific) antibody further comprises a crossover Fab light chain polypeptide of the first Fab molecule, wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VH (i) -CL (i) ), and the Fab light chain polypeptide of the second Fab molecule (VL ( 2 ) -CL ( 2 ) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule which in turn shares a carboxy-terminal peptide bond with the Fab light chain polypeptide of the second Fab molecule (VH (i) -CL (i) -VL ( 2 ) -CL ( 2 ) ), or a polypeptide wherein the Fab light chain polypeptide of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the first Fab molecule which in turn shares a carboxy- terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VL ⁇ ) - CL ( 2 ) -VH (i) -CL (i) ), as appropriate.
  • the (multispecific) antibody may further comprise (i) an Fc domain subunit polypeptide (CH2-CH3 (-CH4)), or (ii) a polypeptide wherein the Fab heavy chain of a third Fab molecule shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (3) -CH1 (3) -CH2-CH3 (-CH4)) and the Fab light chain polypeptide of a third Fab molecule (VL ( 3 ) -CL ( 3 ) ).
  • the polypeptides are covalently linked, e.g., by a disulfide bond.
  • the (multispecific) antibody comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (i) -CL (i) -VH (2) -CHl (2) -CH2-CH3(-CH4)).
  • VH (i) -CL (i) -VH (2) -CHl (2) -CH2-CH3(-CH4) an Fc domain subunit
  • the (multispecific) antibody comprises a polypeptide wherein the Fab heavy chain of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the first Fab molecule which in turn shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (2) -CHl (2) -VH (i) -CL (i) -CH2- CH3(-CH4)).
  • VH (2) -CHl (2) -VH (i) -CL (i) -CH2- CH3(-CH4) an Fc domain subunit
  • the (multispecific) antibody further comprises a crossover Fab light chain polypeptide of the first Fab molecule, wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (VL (i) -CHl (i) ), and the Fab light chain polypeptide of the second Fab molecule (VL ( 2 ) -CL ( 2 ) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule which in turn shares a carboxy-terminal peptide bond with the Fab light chain polypeptide of the second Fab molecule (VL (i) -CHl (i) -VL ( 2 ) -CL ( 2 ) ), or a polypeptide wherein the Fab light chain polypeptide of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the first Fab molecule which in turn shares a carboxy- terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VL ⁇ ) - CL ( 2 ) -VH (i) -CL (i) ), as appropriate.
  • the (multispecific) antibody may further comprise (i) an Fc domain subunit polypeptide (CH2-CH3 (-CH4)), or (ii) a polypeptide wherein the Fab heavy chain of a third Fab molecule shares a carboxy-terminal peptide bond with an Fc domain subunit (VH (3) -CH1 (3) -CH2-CH3 (-CH4)) and the Fab light chain polypeptide of a third Fab molecule (VL ( 3 ) -CL ( 3 ) ).
  • the polypeptides are covalently linked, e.g., by a disulfide bond.
  • the (multispecific) antibody does not comprise an Fc domain.
  • said second and, if present, third antigen binding domains are each a conventional Fab molecule, and the first antigen binding domain is a crossover Fab molecule as described herein, i.e. a Fab molecule wherein the variable domains VH and VL or the constant domains CL and CH1 of the Fab heavy and light chains are exchanged / replaced by each other.
  • said second and, if present, third antigen binding domains are each a crossover Fab molecule and the first antigen binding domain is a conventional Fab molecule.
  • the (multispecific) antibody essentially consists of the first and the second antigen binding domain, and optionally one or more peptide linkers, wherein the first and the second antigen binding domain are both Fab molecules and the second antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain.
  • Figures lO and IS depicted in Figures lO and IS (in these examples with the first antigen binding domain being a VH/VL crossover Fab molecule and the second antigen binding domain being a conventional Fab molecule).
  • the (multispecific) antibody essentially consists of the first and the second antigen binding domain, and optionally one or more peptide linkers, wherein the first and the second antigen binding domain are both Fab molecules and the first antigen binding domain is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain.
  • Figures IP and IT depicted in Figures IP and IT (in these examples with the firs antigen binding domain being a VH/VL crossover Fab molecule and the second antigen binding domain being a conventional Fab molecule).
  • the second Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first Fab molecule
  • the (multispecific) antibody further comprises a third antigen binding domain, particularly a third Fab molecule, wherein said third Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second Fab molecule.
  • the (multispecific) antibody essentially consists of the first, the second and the third Fab molecule, and optionally one or more peptide linkers, wherein the second Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first Fab molecule, and the third Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second Fab molecule.
  • Such a configuration is schematically depicted in Figures IQ and 1U (in these examples with the first antigen binding domain being a VH/VL crossover Fab molecule and the second and the third antigen binding domain each being a conventional Fab molecule), or Figures IX and 1Z (in these examples with the first antigen binding domain being a conventional Fab molecule and the second and the third antigen binding domain each being a VH/VL crossover Fab molecule).
  • the first Fab molecule is fused at the C-terminus of the Fab heavy chain to the N- terminus of the Fab heavy chain of the second Fab molecule
  • the (multispecific) antibody further comprises a third antigen binding domain, particularly a third Fab molecule, wherein said third Fab molecule is fused at the N-terminus of the Fab heavy chain to the C-terminus of the Fab heavy chain of the second Fab molecule.
  • the (multispecific) antibody essentially consists of the first, the second and the third Fab molecule, and optionally one or more peptide linkers, wherein the first Fab molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second Fab molecule, and the third Fab molecule is fused at the N-terminus of the Fab heavy chain to the C-terminus of the Fab heavy chain of the second Fab molecule.
  • Such a configuration is schematically depicted in Figures 1R and IV (in these examples with the first antigen binding domain being a VH/VL crossover Fab molecule and the second and the third antigen binding domain each being a conventional Fab molecule), or Figures 1W and 1Y (in these examples with the first antigen binding domain being a conventional Fab molecule and the second and the third antigen binding domain each being a VH/VL crossover Fab molecule).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain variable region of the first Fab molecule, which in turn shares a carboxy- terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region) (VH ( 2 ) -CHl ( 2 ) -VL (i) -CHl (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VH (i) -CL (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL ( 2 ) -CL ( 2 ) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule (VL (i) -CHl (i) -VH ( 2 ) -CHl ( 2 ) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VH (i) -CL (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the first Fab molecule, which in turn shares a carboxy- terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constnt region is replaced by a light chain constant region) (VH (2) -CHl (2) -VH (i) -CL (i) .
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (VL (i) -CHl (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e. the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule (VH (i) -CL (i) -VH (2) -CHl (2) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (VL (i) -CHl (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab light chain variable region of the first Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e.
  • the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region) (VH (3) -CHl (3) -VH (2) -CHl (2) -VL (i) - CHl (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VH (i) -CL (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody further comprises the Fab light chain polypeptide of a third Fab molecule (VL (3) -CL (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the first Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e.
  • the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region) (VH (3) -CHl (3) -VH (2) -CHl (2) -VH (i) - CL (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (VL (i) -CHl (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody further comprises the Fab light chain polypeptide of a third Fab molecule (VL (3) -CL (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (i.e.
  • the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of a third Fab molecule (VL (i) -CHl (i) -VH (2) -CHl (2) -VH (3) - CH1 ( 3 ) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (VH (i) -CL (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody further comprises the Fab light chain polypeptide of a third Fab molecule (VL (3) -CL (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the first Fab molecule (i.e.
  • the first Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain of a third Fab molecule (VH (i) -CL (i) -VH (2) -CHl (2) -VH (3) - CH1 ( 3 ) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the first Fab molecule (VL (i) -CHl (i) ) and the Fab light chain polypeptide of the second Fab molecule (VL (2) -CL (2) ).
  • the (multispecific) antibody further comprises the Fab light chain polypeptide of a third Fab molecule (VL (3) -CL (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain variable region of the second Fab molecule, which in turn shares a carboxy- terminal peptide bond with the Fab heavy chain constant region of the second Fab molecule (i.e.
  • the second Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with the Fab light chain variable region of a third Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of a third Fab molecule (i.e. the third Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region) (VH (i) -CHl (i) -VL (2) -CHl (2) -VL (3) - CH1 ( 3 ) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the second Fab molecule (VH (2) -CL (2) ) and the Fab light chain polypeptide of the first Fab molecule (VL (i) -CL (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of a third Fab molecule (VH (3) -CL (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain of the first Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the second Fab molecule, which in turn shares a carboxy- terminal peptide bond with the Fab light chain constant region of the second Fab molecule (i.e.
  • the second Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of a third Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab light chain constant region of a third Fab molecule (i.e. the third Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region) (VH (i) -CHl (i) -VH (2) -CL (2) -VH (3) - CL (3) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the second Fab molecule shares a carboxy -terminal peptide bond with the Fab heavy chain constant region of the second Fab molecule (VL (2) -CH1 (2) ) and the Fab light chain polypeptide of the first Fab molecule (VL (i) -CL (i) ).
  • the (multi specific) antibody further comprises a polypeptide wherein the Fab light chain variable region of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of a third Fab molecule (VL (3) -CH1 (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab light chain variable region of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of a third Fab molecule (i.e. the third Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy-terminal peptide bond with the Fab light chain variable region of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the second Fab molecule (i.e.
  • the second Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain variable region is replaced by a light chain variable region), which in turn shares a carboxy- terminal peptide bond with the Fab heavy chain of the first Fab molecule (VL (3) -CH1 (3) -VL (2) - CHl (2) -VH (i) -CHl (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of the second Fab molecule (VH ( 2 ) -CL ( 2 ) ) and the Fab light chain polypeptide of the first Fab molecule (VL (i) -CL (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab heavy chain variable region of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of a third Fab molecule (VH (3) -CL (3) ).
  • the (multispecific) antibody according to the invention comprises a polypeptide wherein the Fab heavy chain variable region of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab light chain constant region of a third Fab molecule (i.e. the third Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy-terminal peptide bond with the Fab heavy chain variable region of the second Fab molecule, which in turn shares a carboxy-terminal peptide bond with the Fab light chain constant region of the second Fab molecule (i.e.
  • the second Fab molecule comprises a crossover Fab heavy chain, wherein the heavy chain constant region is replaced by a light chain constant region), which in turn shares a carboxy- terminal peptide bond with the Fab heavy chain of the first Fab molecule (VH (3) -CL (3) -VH (2) -CL (2) - VH (i) -CHl (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of the second Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of the second Fab molecule (VL (2) -CH1 (2) ) and the Fab light chain polypeptide of the first Fab molecule (VL (i) -CL (i) ).
  • the (multispecific) antibody further comprises a polypeptide wherein the Fab light chain variable region of a third Fab molecule shares a carboxy-terminal peptide bond with the Fab heavy chain constant region of a third Fab molecule (VL (3) -CH1 (3) ).
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH or the constant domains CL and CHI of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second antigen binding domain that binds to CEA, wherein the second antigen binding domain is a (conventional) Fab molecule; c) an Fc domain composed
  • the first antigen binding domain under a) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain under b), and the second antigen binding domain under b) is fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain under c), or
  • the second antigen binding domain under b) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain under a), and the first antigen binding domain under a) is fused at the C -terminus of the Fab heavy chain to the N- terminus of one of the subunits of the Fc domain under c).
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH or the constant domains CL and CHI of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule
  • the first antigen binding domain under a) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain under b), and the second antigen binding domain under b) and the third antigen binding domain under b) are each fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain under c), or
  • the second antigen binding domain under b) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain under a), and the first antigen binding domain under a) and the third antigen binding domain under b) are each fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain under c).
  • the invention provides a (multi specific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH or the constant domains CL and CHI of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second antigen binding domain that binds to CEA, wherein the second antigen binding domain is a (conventional) Fab molecule; c) an Fc domain composed
  • the amino acid substitutions (“charge modifications”) described herein may either be in the CHI and CL domains of the second and (if present) the third antigen binding domain/Fab molecule, or in the CHI and CL domains of the first antigen binding domain/Fab molecule. Preferably, they are in the CHI and CL domains of the second and (if present) the third antigen binding domain/Fab molecule.
  • amino acid substitutions as described herein are made in the second (and, if present, the third) antigen binding domain/Fab molecule, no such amino acid substitutions are made in the first antigen binding domain/Fab molecule.
  • amino acid substitutions as described herein are made in the first antigen binding domain/Fab molecule, no such amino acid substitutions are made in the second (and, if present, the third) antigen binding domain/Fab molecule.
  • Amino acid substitutions are preferably made in (multispecific) antibodies comprising a Fab molecule wherein the variable domains VL and VHl of the Fab light chain and the Fab heavy chain are replaced by each other.
  • the constant domain CL of the second (and, if present, the third) antigen binding domain/Fab molecule is of kappa isotype.
  • the constant domain CL of the first antigen binding domain/Fab molecule is of kappa isotype.
  • the constant domain CL of the second (and, if present, the third) antigen binding domain/Fab molecule and the constant domain CL of the first antigen binding domain/Fab molecule are of kappa isotype.
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second antigen binding domain that binds to CEA, wherein the second antigen binding domain is a (conventional) Fab molecule; c) an Fc domain composed of a first and a second subset of S
  • the first antigen binding domain under a) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain under b), and the second antigen binding domain under b) is fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain under c), or
  • the second antigen binding domain under b) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain under a), and the first antigen binding domain under a) is fused at the C -terminus of the Fab heavy chain to the N- terminus of one of the subunits of the Fc domain under c).
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second and a third antigen binding domain that bind to CEA, wherein the second and third antigen binding domain are each a (conventional) Fab molecule; and c) an Fc domain composed
  • the first antigen binding domain under a) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding domain under b), and the second antigen binding domain under b) and the third antigen binding domain under b) are each fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain under c), or
  • the second antigen binding domain under b) is fused at the C -terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding domain under a), and the first antigen binding domain under a) and the third antigen binding domain under b) are each fused at the C -terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain under c).
  • the invention provides a (multi specific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second antigen binding domain that binds to CEA, wherein the second antigen binding domain is a (conventional) Fab molecule; c) an Fc domain composed of a first and a second subset of S
  • components of the (multispecific) antibody may be fused directly or through various linkers, particularly peptide linkers comprising one or more amino acids, typically about 2-20 amino acids, that are described herein or are known in the art.
  • Suitable, non-immunogenic peptide linkers include, for example, (G4S)n, (SG4)n, (G4S) n , G4(SG4)n or (G4S)nGs peptide linkers, wherein n is generally an integer from 1 to 10, typically from 2 to 4.
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule, and comprise a heavy chain variable region (VH) comprising
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 11; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule, and comprise a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 18 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 22; c) an Fc domain composed of a first and a second subunit; wherein in the constant domain CL of the second and the third antigen binding domain under b) the amino acid at position
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule, and comprise a heavy chain variable region
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 11; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule, and comprise a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 35; c) an Fc domain composed of a first and a second subunit; wherein in the constant domain CL of the second and the third antigen binding domain under b) the amino acid at
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region (VH) comprising a heavy chain complementary determining region (HCDR) 1 of SEQ ID NO: 2, a HCDR 2 of SEQ ID NO: 3, and a HCDR 3 of SEQ ID NO: 5, and a light chain variable region (VL) comprising a light chain complementarity determining region (LCDR) 1 of SEQ ID NO: 8, a LCDR 2 of SEQ ID NO: 9 and a LCDR 3 of SEQ ID NO: 10; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule, and comprise a heavy chain variable region
  • the invention provides a (multispecific) antibody comprising a) a first antigen binding domain that binds to CD3, wherein the first antigen binding domain is a Fab molecule wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, and comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 11; b) a second and a third antigen binding domain that bind to CEA, wherein the second and the third antigen binding domain are each a (conventional) Fab molecule, and comprise a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 39 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 43; c) an Fc domain composed of a first and a second subunit; wherein in the constant domain CL of the second and the third antigen binding domain under b) the amino acid at
  • the threonine residue at position 366 in the first subunit of the Fc domain is replaced with a tryptophan residue (T366W), and in the second subunit of the Fc domain the tyrosine residue at position 407 is replaced with a valine residue (Y407V) and optionally the threonine residue at position 366 is replaced with a serine residue (T366S) and the leucine residue at position 368 is replaced with an alanine residue (L368A) (numberings according to Kabat EU index).
  • the serine residue at position 354 is replaced with a cysteine residue (S354C) or the glutamic acid residue at position 356 is replaced with a cysteine residue (E356C) (particularly the serine residue at position 354 is replaced with a cysteine residue), and in the second subunit of the Fc domain additionally the tyrosine residue at position 349 is replaced by a cysteine residue (Y349C) (numberings according to Kabat EU index).
  • the leucine residue at position 234 is replaced with an alanine residue (L234A)
  • the leucine residue at position 235 is replaced with an alanine residue (L235A)
  • the proline residue at position 329 is replaced by a glycine residue (P329G) (numbering according to Kabat EU index).
  • the Fc domain is a human IgGi Fc domain.
  • the (multispecific) antibody comprises a polypeptide comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 23, a polypeptide comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 24, a polypeptide (particularly two polypeptides) comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 25, and a polypeptide comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 27.
  • the (multispecific) antibody comprises a polypeptide comprising the amino acid sequence of SEQ ID NO: 23, a polypeptide comprising the amino acid sequence of SEQ ID NO: 24, a polypeptide (particularly two polypeptides) comprising the amino acid sequence of SEQ ID NO: 25 and a polypeptide comprising the amino acid sequence of SEQ ID NO: 27.
  • the invention provides a (multispecific) antibody that binds to CD3 and CEA, comprising a polypeptide comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 23, a polypeptide comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 24, a polypeptide (particularly two polypeptides) comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 25, and a polypeptide comprising an amino acid sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 27.
  • the invention provides a (multispecific) antibody that binds to CD3 and CEA, comprising a polypeptide comprising the amino acid sequence of SEQ ID NO: 23, a polypeptide comprising the amino acid sequence of SEQ ID NO: 24, a polypeptide (particularly two polypeptides) comprising the amino acid sequence of SEQ ID NO: 25 and a polypeptide comprising the amino acid sequence of SEQ ID NO: 27.
  • the (multispecific) antibody of the invention comprises an Fc domain composed of a first and a second subunit.
  • the Fc domain of the (multispecific) antibody consists of a pair of polypeptide chains comprising heavy chain domains of an immunoglobulin molecule.
  • the Fc domain of an immunoglobulin G (IgG) molecule is a dimer, each subunit of which comprises the CH2 and CH3 IgG heavy chain constant domains.
  • the two subunits of the Fc domain are capable of stable association with each other.
  • the (multispecific) antibody of the invention comprises not more than one Fc domain.
  • the Fc domain of the (multispecific) antibody is an IgG Fc domain.
  • the Fc domain is an IgGi Fc domain.
  • the Fc domain is an IgG* Fc domain.
  • the Fc domain is an IgG 4 Fc domain comprising an amino acid substitution at position S228 (Kabat EU index numbering), particularly the amino acid substitution S228P. This amino acid substitution reduces in vivo Fab arm exchange of IgGt antibodies (see Stubenrauch et al, Drug Metabolism and Disposition 38, 84-91 (2010)).
  • the Fc domain is a human Fc domain.
  • the Fc domain is a human IgGi Fc domain.
  • An exemplary sequence of a human IgGi Fc region is given in SEQ ID NO: 48. a) Fc domain modifications promoting heterodimerization
  • (Multispecific) antibodies according to the invention comprise different antigen binding domains, which may be fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised in two non-identical polypeptide chains. Recombinant co-expression of these polypeptides and subsequent dimerization leads to several possible combinations of the two polypeptides. To improve the yield and purity of (multispecific) antibodies in recombinant production, it will thus be advantageous to introduce in the Fc domain of the (multispecific) antibody a modification promoting the association of the desired polypeptides.
  • the Fc domain of the (multispecific) antibody according to the invention comprises a modification promoting the association of the first and the second subunit of the Fc domain.
  • the site of most extensive protein-protein interaction between the two subunits of a human IgG Fc domain is in the CH3 domain of the Fc domain.
  • said modification is in the CH3 domain of the Fc domain.
  • the CH3 domain of the first subunit of the Fc domain and the CH3 domain of the second subunit of the Fc domain are both engineered in a complementary manner so that each CH3 domain (or the heavy chain comprising it) can no longer homodimerize with itself but is forced to heterodimerize with the complementarily engineered other CH3 domain (so that the first and second CH3 domain heterodimerize and no homdimers between the two first or the two second CH3 domains are formed).
  • These different approaches for improved heavy chain heterodimerization are contemplated as different alternatives in combination with the heavy-light chain modifications (e.g.
  • said modification promoting the association of the first and the second subunit of the Fc domain is a so-called “knob-into-hole” modification, comprising a “knob” modification in one of the two subunits of the Fc domain and a “hole” modification in the other one of the two subunits of the Fc domain.
  • the method involves introducing a protuberance (“knob”) at the interface of a first polypeptide and a corresponding cavity (“hole”) in the interface of a second polypeptide, such that the protuberance can be positioned in the cavity so as to promote hetero dimer formation and hinder homodimer formation.
  • Protuberances are constructed by replacing small amino acid side chains from the interface of the first polypeptide with larger side chains (e.g. tyrosine or tryptophan).
  • Compensatory cavities of identical or similar size to the protuberances are created in the interface of the second polypeptide by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine).
  • an amino acid residue in the CH3 domain of the first subunit of the Fc domain of the (multispecific) antibody an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.
  • amino acid residue having a larger side chain volume is selected from the group consisting of arginine (R), phenylalanine (F), tyrosine (Y), and tryptophan (W).
  • amino acid residue having a smaller side chain volume is selected from the group consisting of alanine (A), serine (S), threonine (T), and valine (V).
  • the protuberance and cavity can be made by altering the nucleic acid encoding the polypeptides, e.g. by site-specific mutagenesis, or by peptide synthesis.
  • the threonine residue at position 366 is replaced with a tryptophan residue (T366W)
  • the CH3 domain of the second subunit of the Fc domain the “hole” subunit
  • the tyrosine residue at position 407 is replaced with a valine residue (Y407V).
  • the threonine residue at position 366 is replaced with a serine residue (T366S) and the leucine residue at position 368 is replaced with an alanine residue (L368A) (numberings according to Kabat EU index).
  • the serine residue at position 354 is replaced with a cysteine residue (S354C) or the glutamic acid residue at position 356 is replaced with a cysteine residue (E356C) (particularly the serine residue at position 354 is replaced with a cysteine residue), and in the second subunit of the Fc domain additionally the tyrosine residue at position 349 is replaced by a cysteine residue (Y349C) (numberings according to Kabat EU index). Introduction of these two cysteine residues results in formation of a disulfide bridge between the two subunits of the Fc domain, further stabilizing the dimer (Carter, J Immunol Methods 248, 7-15 (2001)).
  • the first subunit of the Fc domain comprises the amino acid substitutions S354C and T366W
  • the second subunit of the Fc domain comprises the amino acid substitutions Y349C, T366S, L368A and Y407V (numbering according to Kabat EU index).
  • the antigen binding domain that binds to CD3 is fused (optionally via the second antigen binding domain, which binds to CEA, and/or a peptide linker) to the first subunit of the Fc domain (comprising the “knob” modification).
  • fusion of the antigen binding domain that binds CD3 to the knob-containing subunit of the Fc domain will (further) minimize the generation of antibodies comprising two antigen binding domains that bind to CD3 (steric clash of two knob-containing polypeptides).
  • CH3 -modification for enforcing the heterodimerization is contemplated as alternatives according to the invention and are described e.g. in WO 96/27011, WO 98/050431, EP 1870459, WO 2007/110205, WO 2007/147901, WO 2009/089004, WO 2010/129304, WO 2011/90754, WO 2011/143545, WO 2012/058768, WO 2013/157954, WO 2013/096291.
  • the heterodimerization approach described in EP 1870459 is used alternatively.
  • This approach is based on the introduction of charged amino acids with opposite charges at specific amino acid positions in the CH3/CH3 domain interface between the two subunits of the Fc domain.
  • a particular aspect for the (multispecific) antibody of the invention are amino acid mutations R409D; K370E in one of the two CH3 domains (of the Fc domain) and amino acid mutations D399K; E357K in the other one of the CH3 domains of the Fc domain (numbering according to Kabat EU index).
  • the (multispecific) antibody of the invention comprises amino acid mutation T366W in the CH3 domain of the first subunit of the Fc domain and amino acid mutations T366S, L368A, Y407V in the CH3 domain of the second subunit of the Fc domain, and additionally amino acid mutations R409D; K370E in the CH3 domain of the first subunit of the Fc domain and amino acid mutations D399K; E357K in the CH3 domain of the second subunit of the Fc domain (numberings according to Kabat EU index).
  • the (multispecific) antibody of the invention comprises amino acid mutations S354C, T366W in the CH3 domain of the first subunit of the Fc domain and amino acid mutations Y349C, T366S, L368A, Y407V in the CH3 domain of the second subunit of the Fc domain, or said (multispecific) antibody comprises amino acid mutations Y349C, T366W in the CH3 domain of the first subunit of the Fc domain and amino acid mutations S354C, T366S, L368A, Y407V in the CH3 domains of the second subunit of the Fc domain and additionally amino acid mutations R409D; K370E in the CH3 domain of the first subunit of the Fc domain and amino acid mutations D399K; E357K in the CH3 domain of the second subunit of the Fc domain (all numberings according to Kabat EU index).
  • a first CH3 domain comprises amino acid mutation T366K and a second CH3 domain comprises amino acid mutation L351D (numberings according to Kabat EU index).
  • the first CH3 domain comprises further amino acid mutation L351K.
  • the second CH3 domain comprises further an amino acid mutation selected from Y349E, Y349D and L368E (particularly L368E) (numberings according to Kabat EU index).
  • the heterodimerization approach described in WO 2012/058768 is used alternatively.
  • a first CH3 domain comprises amino acid mutations L351Y, Y407A and a second CH3 domain comprises amino acid mutations T366A, K409F.
  • the second CH3 domain comprises a further amino acid mutation at position T411, D399, S400, F405, N390, or K392, e.g.
  • T411N, T411R, T411Q, T411K, T411D, T411E or T411W b) D399R, D399W, D399Y or D399K
  • S400E, S400D, S400R, or S400K d) F405I, F405M, F405T, F405S, F405V or F405W, e) N390R, N390K or N390D, f) K392V, K392M, K392R, K392L, K392F or K392E (numberings according to Kabat EU index).
  • a first CH3 domain comprises amino acid mutations L351Y, Y407A and a second CH3 domain comprises amino acid mutations T366V, K409F.
  • a first CH3 domain comprises amino acid mutation Y407A and a second CH3 domain comprises amino acid mutations T366A, K409F.
  • the second CH3 domain further comprises amino acid mutations K392E, T411E, D399R and S400R (numberings according to Kabat EU index).
  • the heterodimerization approach described in WO 2011/143545 is used alternatively, e.g. with the amino acid modification at a position selected from the group consisting of 368 and 409 (numbering according to Kabat EU index).
  • a first CH3 domain comprises amino acid mutation T366W and a second CH3 domain comprises amino acid mutation Y407A.
  • a first CH3 domain comprises amino acid mutation T366Y and a second CH3 domain comprises amino acid mutation Y407T (numberings according to Kabat EU index).
  • the (multispecific) antibody or its Fc domain is of IgG2 subclass and the heterodimerization approach described in WO 2010/129304 is used alternatively.
  • a modification promoting association of the first and the second subunit of the Fc domain comprises a modification mediating electrostatic steering effects, e.g. as described in PCT publication WO 2009/089004.
  • this method involves replacement of one or more amino acid residues at the interface of the two Fc domain subunits by charged amino acid residues so that homodimer formation becomes electrostatically unfavorable but heterodimerization electrostatically favorable.
  • a first CH3 domain comprises amino acid substitution of K392 or N392 with a negatively charged amino acid (e.g.
  • a second CH3 domain comprises amino acid substitution of D399, E356, D356, or E357 with a positively charged amino acid (e.g. lysine (K) or arginine (R), particularly D399K, E356K, D356K, or E357K, and more particularly D399K and E356K).
  • the first CH3 domain further comprises amino acid substitution of K409 or R409 with a negatively charged amino acid (e.g. glutamic acid (E), or aspartic acid (D), particularly K409D or R409D).
  • the first CH3 domain further or alternatively comprises amino acid substitution of K439 and/or K370 with a negatively charged amino acid (e.g. glutamic acid (E), or aspartic acid (D)) (all numberings according to Kabat EU index).
  • a negatively charged amino acid e.g. glutamic acid (E), or aspartic acid (D)
  • E glutamic acid
  • D aspartic acid
  • a first CH3 domain comprises amino acid mutations K253E, D282K, and K322D and a second CH3 domain comprises amino acid mutations D239K, E240K, and K292D (numberings according to Kabat EU index).
  • the heterodimerization approach described in WO 2007/110205 can be used alternatively.
  • the first subunit of the Fc domain comprises amino acid substitutions K392D and K409D
  • the second subunit of the Fc domain comprises amino acid substitutions D356K and D399K (numbering according to Kabat EU index).
  • the Fc domain confers to the (multispecific) antibody favorable pharmacokinetic properties, including a long serum half-life which contributes to good accumulation in the target tissue and a favorable tissue-blood distribution ratio. At the same time it may, however, lead to undesirable targeting of the (multispecific) antibody to cells expressing Fc receptors rather than to the preferred antigen-bearing cells. Moreover, the co-activation of Fc receptor signaling pathways may lead to cytokine release which, in combination with the T cell activating properties and the long half-life of the (multispecific) antibody, results in excessive activation of cytokine receptors and severe side effects upon systemic administration. Activation of (Fc receptor-bearing) immune cells other than T cells may even reduce efficacy of the (multispecific) antibody due to the potential destruction of T cells e.g. by NK cells.
  • the Fc domain of the (multispecific) antibody according to the invention exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgGi Fc domain.
  • the Fc domain (or the (multispecific) antibody comprising said Fc domain) exhibits less than 50%, particularly less than 20%, more particularly less than 10% and most particularly less than 5% of the binding affinity to an Fc receptor, as compared to a native IgGi Fc domain (or a (multispecific) antibody comprising a native IgGi Fc domain), and/or less than 50%, particularly less than 20%, more particularly less than 10% and most particularly less than 5% of the effector function, as compared to a native IgGi Fc domain domain (or a (multispecific) antibody comprising a native IgGi Fc domain).
  • the Fc domain domain does not substantially bind to an Fc receptor and/or induce effector function.
  • the Fc receptor is an Fey receptor.
  • the Fc receptor is a human Fc receptor.
  • the Fc receptor is an activating Fc receptor.
  • the Fc receptor is an activating human Fey receptor, more specifically human FcyRIIIa, FcyRI or FcyRIIa, most specifically human FcyRIIIa.
  • the effector function is one or more selected from the group of CDC, ADCC, ADCP, and cytokine secretion. In a preferred aspect, the effector function is ADCC.
  • the Fc domain domain exhibits substantially similar binding affinity to neonatal Fc receptor (FcRn), as compared to a native IgGi Fc domain domain.
  • FcRn neonatal Fc receptor
  • Substantially similar binding to FcRn is achieved when the Fc domain (or the (multispecific) antibody comprising said Fc domain) exhibits greater than about 70%, particularly greater than about 80%, more particularly greater than about 90% of the binding affinity of a native IgGi Fc domain (or the (multispecific) antibody comprising a native IgGi Fc domain) to FcRn.
  • the Fc domain is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain.
  • the Fc domain of the (multispecific) antibody comprises one or more amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function. Typically, the same one or more amino acid mutation is present in each of the two subunits of the Fc domain.
  • the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor. In one aspect, the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor by at least 2-fold, at least 5 -fold, or at least 10-fold.
  • the combination of these amino acid mutations may reduce the binding affinity of the Fc domain to an Fc receptor by at least 10-fold, at least 20-fold, or even at least 50-fold.
  • the (multispecific) antibody comprising an engineered Fc domain exhibits less than 20%, particularly less than 10%, more particularly less than 5% of the binding affinity to anFc receptor as compared to a (multispecific) antibody comprising a non-engineered Fc domain.
  • the Fc receptor is an Fey receptor.
  • the Fc receptor is a human Fc receptor.
  • the Fc receptor is an activating Fc receptor.
  • the Fc receptor is an activating human Fey receptor, more specifically human FcyRIIIa, FcyRI or FcyRIIa, most specifically human FcyRIIIa.
  • binding to each of these receptors is reduced.
  • binding affinity to a complement component, specifically binding affinity to Clq is also reduced.
  • binding affinity to neonatal Fc receptor (FcRn) is not reduced. Substantially similar binding to FcRn, i.e.
  • the Fc domain (or the (multispecific) antibody comprising said Fc domain) exhibits greater than about 70% of the binding affinity of a non-engineered form of the Fc domain (or the (multispecific) antibody comprising said non-engineered form of the Fc domain) to FcRn.
  • the Fc domain, or (multispecific) antibodies of the invention comprising said Fc domain may exhibit greater than about 80% and even greater than about 90% of such affinity.
  • the Fc domain of the (multispecific) antibody is engineered to have reduced effector function, as compared to a non-engineered Fc domain.
  • the reduced effector function can include, but is not limited to, one or more of the following: reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen-presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced crosslinking of target-bound antibodies, reduced dendritic cell maturation, or reduced T cell priming.
  • CDC complement dependent cytotoxicity
  • ADCC reduced antibody-dependent cell-mediated cytotoxicity
  • ADCP reduced antibody-dependent cellular phagocytosis
  • reduced immune complex-mediated antigen uptake by antigen-presenting cells reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing
  • the reduced effector function is one or more selected from the group of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In a preferred aspect, the reduced effector function is reduced ADCC. In one aspect the reduced ADCC is less than 20% of the ADCC induced by a non- engineered Fc domain (or a (multispecific) antibody comprising a non-engineered Fc domain).
  • the amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function is an amino acid substitution.
  • the Fc domain comprises an amino acid substitution at a position selected from the group of E233, L234, L235, N297, P331 and P329 (numberings according to Kabat EU index).
  • the Fc domain comprises an amino acid substitution at a position selected from the group of L234, L235 and P329 (numberings according to Kabat EU index).
  • the Fc domain comprises the amino acid substitutions L234A and L235A (numberings according to Kabat EU index).
  • the Fc domain is an IgGi Fc domain, particularly a human IgGi Fc domain.
  • the Fc domain comprises an amino acid substitution at position P329.
  • the amino acid substitution is P329A or P329G, particularly P329G (numberings according to Kabat EU index).
  • the Fc domain comprises an amino acid substitution at position P329 and a further amino acid substitution at a position selected from E233, L234, L235, N297 and P331 (numberings according to Kabat EU index).
  • the further amino acid substitution is E233P, L234A, L235A, L235E, N297A, N297D or P331S.
  • the Fc domain comprises amino acid substitutions at positions P329, L234 and L235 (numberings according to Kabat EU index).
  • the Fc domain comprises the amino acid mutations L234A, L235A and P329G (“P329G LALA”, “PGLALA” or “LALAPG”).
  • each subunit of the Fc domain comprises the amino acid substitutions L234A, L235A and P329G (Kabat EU index numbering), i.e.
  • the leucine residue at position 234 is replaced with an alanine residue (L234A)
  • the leucine residue at position 235 is replaced with an alanine residue (L235A)
  • the proline residue at position 329 is replaced by a glycine residue (P329G) (numbering according to Kabat EU index).
  • the Fc domain is an IgGi Fc domain, particularly a human IgGi Fc domain.
  • the “P329G LALA” combination of amino acid substitutions almost completely abolishes Fey receptor (as well as complement) binding of a human IgGi Fc domain, as described in PCT publication no. WO 2012/130831, which is incorporated herein by reference in its entirety.
  • WO 2012/130831 also describes methods of preparing such mutant Fc domains and methods for determining its properties such as Fc receptor binding or effector functions.
  • the Fc domain of the (multispecific) antibodies of the invention is an IgG 4 Fc domain, particularly a human IgG 4 Fc domain.
  • the IgG 4 Fc domain comprises an amino acid substitution at position S228, specifically the amino acid substitution S228P (numberings according to Kabat EU index).
  • the IgG 4 Fc domain comprises an amino acid substitution at position L235, specifically the amino acid substitution L235E (numberings according to Kabat EU index).
  • the IgGi Fc domain comprises an amino acid substitution at position P329, specifically the amino acid substitution P329G (numberings according to Kabat EU index).
  • the IgGi Fc domain comprises amino acid substitutions at positions S228, L235 and P329, specifically amino acid substitutions S228P, L235E and P329G (numberings according to Kabat EU index).
  • Such IgGi Fc domain mutants and their Fey receptor binding properties are described in PCT publication no. WO 2012/130831, incorporated herein by reference in its entirety.
  • the Fc domain exhibiting reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgGi Fc domain is a human IgGi Fc domain comprising the amino acid substitutions L234A, L235A and optionally P329G, or a human IgG 4 Fc domain comprising the amino acid substitutions S228P, L235E and optionally P329G (numberings according to Kabat EU index).
  • the Fc domain comprises an amino acid mutation at position N297, particularly an amino acid substitution replacing asparagine by alanine (N297A) or aspartic acid (N297D) (numberings according to Kabat EU index).
  • Fc domains with reduced Fc receptor binding and/or effector function also include those with substitution of one or more of Fc domain residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056) (numberings according to Kabat EU index).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called “DANA” Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
  • Mutant Fc domains can be prepared by amino acid deletion, substitution, insertion or modification using genetic or chemical methods well known in the art. Genetic methods may include site- specific mutagenesis of the encoding DNA sequence, PCR, gene synthesis, and the like. The correct nucleotide changes can be verified for example by sequencing.
  • Binding to Fc receptors can be easily determined e.g. by ELISA, or by Surface Plasmon Resonance (SPR) using standard instrumentation such as a BIAcore instrument (GE Healthcare), and Fc receptors such as may be obtained by recombinant expression.
  • binding affinity of Fc domains or (multispecific) antibodies comprising an Fc domain for Fc receptors may be evaluated using cell lines known to express particular Fc receptors, such as human NK cells expressing Fcyllla receptor.
  • Effector function of an Fc domain, or a (multispecific) antibody comprising an Fc domain can be measured by methods known in the art.
  • Examples of in vitro assays to assess ADCC activity of a molecule of interest are described in U S. Patent No. 5,500,362; Hell strom et al. Proc Natl Acad Sci USA 83, 7059-7063 (1986) and Hellstrom et al., Proc Natl Acad Sci USA 82, 1499-1502 (1985); U S. Patent No. 5,821,337; Bruggemann et al., J Exp Med 166, 1351-1361 (1987).
  • non-radioactive assays may be employed (see, for example, ACTITM non radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA); and CytoTox 96 ® non-radioactive cytotoxicity assay (Promega, Madison, WI)).
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo , e.g. in a animal model such as that disclosed in Clynes et al., Proc Natl Acad Sci USA 95, 652- 656 (1998).
  • binding of the Fc domain to a complement component, specifically to Clq is reduced.
  • said reduced effector function includes reduced CDC.
  • Clq binding assays may be carried out to determine whether the Fc domain, or the (multispecific) antibody comprising the Fc domain, is able to bind Clq and hence has CDC activity. See e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J Immunol Methods 202, 163 (1996); Cragg et al., Blood 101, 1045-1052 (2003); and Cragg and Glennie, Blood 103, 2738-2743 (2004)).
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S B. et al, Int 7. Immunol. 18(12): 1759- 1769 (2006); WO 2013/120929).
  • the invention further provides an isolated polynucleotide encoding an antibody of the invention.
  • Said isolated polynucleotide may be a single polynucleotide or a plurality of polynucleotides.
  • polynucleotides encoding (multispecific) antibodies of the invention may be expressed as a single polynucleotide that encodes the entire antibody or as multiple (e.g., two or more) polynucleotides that are co-expressed.
  • Polypeptides encoded by polynucleotides that are co expressed may associate through, e.g., disulfide bonds or other means to form a functional antibody.
  • the light chain portion of an antibody may be encoded by a separate polynucleotide from the portion of the antibody comprising the heavy chain of the antibody. When co-expressed, the heavy chain polypeptides will associate with the light chain polypeptides to form the antibody.
  • the portion of the antibody comprising one of the two Fc domain subunits and optionally (part of) one or more Fab molecules could be encoded by a separate polynucleotide from the portion of the antibody comprising the other of the two Fc domain subunits and optionally (part of) a Fab molecule.
  • the Fc domain subunits When co-expressed, the Fc domain subunits will associate to form the Fc domain.
  • the isolated polynucleotide encodes the entire antibody molecule according to the invention as described herein. In other aspects, the isolated polynucleotide encodes a polypeptide comprised in the antibody according to the invention as described herein.
  • RNA for example, in the form of messenger RNA (mRNA).
  • mRNA messenger RNA
  • RNA of the present invention may be single stranded or double stranded.
  • 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
  • one or more polynucleotide encoding the antibody, e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • Such polynucleotide may be readily isolated and sequenced using conventional procedures.
  • a vector, particularly an expression vector, comprising the polynucleotide (i.a. a single polynucleotide or a plurality of polynucleotides) of the invention is provided.
  • 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 antibody (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 antibody 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 cytomegaloviruse s (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 b-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as inducible promoters (e.g. promoters inducible by 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.
  • DNA encoding a signal sequence may be placed upstream of the nucleic acid encoding an 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 (TP A) or mouse b-glucuronidase.
  • DNA encoding a short protein sequence that could be used to facilitate later purification (e.g. a histidine tag) or assist in labeling the antibody may be included within or at the ends of the antibody (fragment) encoding polynucleotide.
  • a host cell comprising a polynucleotide (i.e. a single polynucleotide or a plurality of polynucleotides) of the invention.
  • a host cell comprising a vector of the invention.
  • the polynucleotides and vectors may incorporate any of the features, singly or in combination, described herein in relation to polynucleotides and vectors, respectively.
  • a host cell comprises (e.g. has been transformed or transfected with) one or more vector comprising one or more polynucleotide that encodes (part of) an antibody of the invention.
  • the term "host cell” refers to any kind of cellular system which can be engineered to generate the antibody of the invention or fragments thereof.
  • Host cells suitable for replicating and for supporting expression of 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 antibody for clinical applications.
  • Suitable host cells include prokaryotic microorganisms, such as E. coli, or various eukaryotic cells, such as Chinese hamster ovary cells (CHO), insect cells, or the like.
  • polypeptides may be produced in bacteria in particular when glycosylation is not needed.
  • the polypeptide may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been “humanized”, resulting in the production of a polypeptide with a partially or fully human glycosylation pattern. See Gerngross, Nat Biotech 22, 1409-1414 (2004), and Li et al, Nat Biotech 24, 210-215 (2006).
  • Suitable host cells for the expression of (glycosylated) polypeptides are also derived from multicellular organisms (invertebrates and vertebrates).
  • invertebrate cells examples 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. US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts. For example, 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 3 A
  • 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 dhfir 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
  • dhfir CHO cells Urlaub et al., Proc Natl Acad Sci USA 77, 4216 (1980)
  • myeloma cell lines such as YO, NS0, P3X63 and Sp2/0.
  • 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, particularly 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).
  • the host cell is not a cell within a human body. 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 an antibody according to the invention comprises culturing a host cell comprising a polynucleotide encoding the antibody, as provided herein, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • the components of the (multispecific) antibody of the invention may be genetically fused to each other.
  • the (multispecific) 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 (multispecific) antibodies are 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.
  • 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.
  • affinity chromatography purification an antibody, ligand, receptor or antigen can be used to which the antibody binds.
  • a matrix with protein A or protein G may be used for affinity chromatography purification of antibodies of the invention.
  • Sequential Protein A or G affinity chromatography and size exclusion chromatography can be used to isolate an antibody essentially as described in the Examples.
  • the purity of the antibody can be determined by any of a variety of well-known analytical methods including gel electrophoresis, high pressure liquid chromatography, and the like.
  • 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.
  • the binding (affinity) of the antibody to an Fc receptor or a target antigen can be determined for example 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 BIAcore instrument
  • receptors or target proteins such as may be obtained by recombinant expression.
  • binding of antibodies to different receptors or target antigens may be evaluated using cell lines expressing the particular receptor or target antigen, for example by flow cytometry (FACS).
  • FACS flow cytometry
  • the binding activity to CD3 is determined by SPR as follows:
  • the CD3 antigen used is a heterodimer of CD3 delta and CD3 epsilon ectodomains fused to a human Fc domain with knob-into-hole modifications and a C -terminal Avi-tag (see SEQ ID NOs 44 and 45).
  • CD3 antigen is injected at a concentration of 10 pg/ml for 120 s and dissociation is monitored at a flow rate of 5 m ⁇ /min for about 120 s.
  • the chip surface is regenerated by two consecutive injections of 10 mM glycine pH 2.1 for about 60 s each. Bulk refractive index differences are corrected by subtracting blank injections and by subtracting the response obtained from the blank control flow cell. For evaluation, the binding response is taken 5 seconds after injection end.
  • the CD3 binding is divided by the anti-Fab response (the signal (RU) obtained upon capture of the CD3 antibody on the immobilized anti-Fab antibody).
  • the binding activity to CD3 of an antibody after a certain treatment, relative to the binding activity to CD3 of the antibody after a different treatment is calculated by referencing the binding activity of a sample of the antibody after the certain treatment to the binding activity of a corresponding sample of the antibody after the different treatment.
  • Biological activity of the (multispecific) antibodies of the invention can be measured by various assays as described in the Examples.
  • Biological activities may for example include the induction of proliferation of T cells, the induction of signaling in T cells, the induction of expression of activation markers in T cells, the induction of cytokine secretion by T cells, the induction of lysis of target cells such as tumor cells, and the induction of tumor regression and/or the improvement of survival.
  • the invention provides pharmaceutical compositions comprising any of the antibodies provided herein, e.g., for use in any of the below therapeutic methods.
  • a pharmaceutical composition comprises an antibody according to the invention and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprises an antibody according to the invention and at least one additional therapeutic agent, e.g., as described below.
  • an antibody of the invention in a form suitable for administration in vivo , the method comprising (a) obtaining an antibody according to the invention, and (b) formulating the antibody with at least one pharmaceutically acceptable carrier, whereby a preparation of antibody is formulated for administration in vivo.
  • compositions of the present invention comprise an effective amount of antibody dissolved or dispersed in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to molecular entities and compositions that are generally non-toxic to recipients at the dosages and concentrations employed, i.e. do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate.
  • the preparation of a pharmaceutical composition that contains an antibody and optionally an additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference.
  • compositions are lyophilized formulations or aqueous solutions.
  • pharmaceutically acceptable carrier includes any and all solvents, buffers, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g.
  • antibacterial agents antifungal agents
  • isotonic agents absorption delaying agents, salts, preservatives, antioxidants, proteins, drugs, drug stabilizers, polymers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329, incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the pharmaceutical compositions is contemplated.
  • An 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.
  • Parenteral compositions include those designed for administration by injection, e.g. subcutaneous, intradermal, intralesional, intravenous, intraarterial intramuscular, intrathecal or intraperitoneal injection.
  • the antibodies of the invention may be formulated in aqueous solutions, particularly in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • the solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the antibodies may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • Sterile injectable solutions are prepared by incorporating the antibodies of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated below, as required. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and/or the other ingredients.
  • a sterile vehicle which contains the basic dispersion medium and/or the other ingredients.
  • the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered liquid medium thereof.
  • the liquid medium should be suitably buffered if necessary and the liquid diluent first rendered isotonic prior to injection with sufficient saline or glucose.
  • the composition must be stable under the conditions of manufacture and storage, and preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Suitable pharmaceutically acceptable carriers include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids
  • Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl cleats or triglycerides, or liposomes.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin micro spheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin micro spheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide, which matrices are in the form of shaped articles, e.g. films, or micro capsules.
  • prolonged absorption of an injectable composition can be brought about by the use in the compositions of agents delaying absorption, such as, for example, aluminum monostearate, gelatin or combinations thereof.
  • the antibodies may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the antibodies may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions comprising the antibodies of the invention may be manufactured by means of conventional mixing, dissolving, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the proteins into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the antibodies may be formulated into a composition in a free acid or base, neutral or salt form.
  • Pharmaceutically acceptable salts are salts that substantially retain the biological activity of the free acid or base. These include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
  • Antibodies of the invention may be used as immunotherapeutic agents, for example in the treatment of cancers.
  • 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.
  • antibodies of the invention for use as a medicament are provided.
  • antibodies of the invention for use in treating a disease are provided.
  • antibodies of the invention for use in a method of treatment are provided.
  • the invention provides an antibody of the invention for use in the treatment of a disease in an individual in need thereof.
  • the invention provides an antibody for use in a method of treating an individual having a disease comprising administering to the individual an effective amount of the antibody.
  • the disease is a proliferative disorder.
  • the disease is cancer, particularly a CEA-expressing cancer.
  • the cancer is selected from the group consisting of colorectal cancer, pancreatic cancer, gastric cancer, non-small cell lung cancer and breast cancer.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer.
  • the invention provides an antibody of the invention for use in inducing lysis of a target cell, particularly a tumor cell.
  • the invention provides an antibody of the invention for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the antibody to induce lysis of a target cell.
  • An “individual” according to any of the above aspects is a mammal, preferably a human.
  • the invention provides for the use of an antibody of the invention in the manufacture or preparation of a medicament.
  • the medicament is for the treatment of a disease in an individual in need thereof.
  • the medicament is for use in a method of treating a disease comprising administering to an individual having the disease an effective amount of the medicament.
  • the disease is a proliferative disorder.
  • the disease is cancer, particularly a CEA-expressing cancer.
  • the cancer is selected from the group consisting of colorectal cancer, pancreatic cancer, gastric cancer, non small cell lung cancer and breast cancer.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., an anti cancer agent if the disease to be treated is cancer.
  • the medicament is for inducing lysis of a target cell, particularly a tumor cell.
  • the medicament is for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the medicament to induce lysis of a target cell.
  • An “individual” according to any of the above aspects may be a mammal, preferably a human.
  • the invention provides a method for treating a disease.
  • the method comprises administering to an individual having such disease an effective amount of an antibody of the invention.
  • a composition is administered to said individual, comprising the antibody of the invention in a pharmaceutically acceptable form.
  • the disease is a proliferative disorder.
  • the disease is cancer, particularly a CEA-expressing cancer.
  • the cancer is selected from the group consisting of colorectal cancer, pancreatic cancer, gastric cancer, non-small cell lung cancer and breast cancer.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer.
  • An “individual” according to any of the above aspects may be a mammal, preferably a human.
  • the invention provides a method for inducing lysis of a target cell, particularly a CEA-expressing cell such as a tumor cell.
  • the method comprises contacting a target cell with an antibody of the invention in the presence of a T cell, particularly a cytotoxic T cell.
  • a method for inducing lysis of a target cell, particularly a CEA-expressing cell such as a tumor cell, in an individual is provided.
  • the method comprises administering to the individual an effective amount of an antibody of the invention to induce lysis of a target cell.
  • an “individual” is a human.
  • the disease to be treated is a proliferative disorder, particularly cancer.
  • Non limiting examples of cancers include bladder cancer, brain cancer, head and neck cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, uterine cancer, cervical cancer, endometrial cancer, esophageal cancer, colon cancer, colorectal cancer, rectal cancer, gastric cancer, prostate cancer, blood cancer, skin cancer, squamous cell carcinoma, bone cancer, and kidney cancer.
  • cancers located in the: abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic region, and urogenital system. Also included are pre-cancerous conditions or lesions and cancer metastases.
  • the cancer is a cancer expressing CEA.
  • the cancer is selected from the group consisting of colorectal cancer, pancreatic cancer, gastric cancer, non-small cell lung cancer and breast cancer.
  • the cancer is colorectal cancer.
  • an amount of antibody that provides a physiological change is considered an "effective amount".
  • the subject, patient, or individual in need of treatment is typically a mammal, more specifically a human.
  • an effective amount of an antibody of the invention is administered to an individual for the treatment of disease.
  • an 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 route of administration, the body weight of the patient, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous or concurrent therapeutic interventions, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 pg/kgto 15 mg/kg (e.g. 0.1 mg/kg - 10 mg/kg) of 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 pg/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.005 mg/kg to about 10 mg/kg.
  • a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10 milligram/kg body weight, about 50 milligram/kg body weight, about 100 milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram/kg body weight to about 500 milligram/kg body weight, etc. can be administered, based on the numbers described above.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the 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.
  • the antibodies of the invention will generally be used in an amount effective to achieve the intended purpose.
  • the antibodies of the invention, or pharmaceutical compositions thereof are administered or applied in an effective amount.
  • an effective dose can be estimated initially from in vitro assays, such as cell culture assays.
  • a dose can then be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. Dosage amount and interval may be adjusted individually to provide plasma levels of the antibodies which are sufficient to maintain therapeutic effect. Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/ day. Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
  • an effective dose of the antibodies of the invention will generally provide therapeutic benefit without causing substantial toxicity.
  • Toxicity and therapeutic efficacy of an antibody can be determined by standard pharmaceutical procedures in cell culture or experimental animals.
  • Cell culture assays and animal studies can be used to determine the LD50 (the dose lethal to 50% of a population) and the ED50 (the dose therapeutically effective in 50% of a population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50.
  • Antibodies that exhibit large therapeutic indices are preferred.
  • the antibody according to the present invention exhibits a high therapeutic index.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans.
  • the dosage lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al, 1975, in: The Pharmacological Basis of Therapeutics, Ch. 1, p. 1, incorporated herein by reference in its entirety).
  • the attending physician for patients treated with antibodies of the invention would know how and when to terminate, interrupt, or adjust administration due to toxicity, organ dysfunction, and the like. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.
  • the antibodies of the invention may be administered in combination with one or more other agents in therapy.
  • an antibody of the invention may be co-administered with at least one additional therapeutic agent.
  • therapeutic agent encompasses any agent administered to treat a symptom or disease in an individual in need of such treatment.
  • additional therapeutic agent may comprise any active ingredients suitable for the particular disease being treated, preferably those with complementary activities that do not adversely affect each other.
  • an additional therapeutic agent is an immunomodulatory agent, a cytostatic agent, an inhibitor of cell adhesion, a cytotoxic agent, an activator of cell apoptosis, or an agent that increases the sensitivity of cells to apoptotic inducers.
  • the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.
  • the additional therapeutic agent is one or more selected from the group of fluorouracil, capecitabine, irinotecan, oxaliplatin, bevacizumab, cetuximab, panitumumab, aflibercept, ramucirumab, trifluridine/tipiracil, and regorafenib.
  • Such other agents are suitably present in combination in amounts that are effective for the purpose intended.
  • the effective amount of such other agents depends on the amount of antibody used, the type of disorder or treatment, and other factors discussed above.
  • the antibodies are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate compositions), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Antibodies of the invention may also be used in combination with radiation therapy.
  • 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 an 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 an 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 aspect of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bac
  • any of the antibodies provided herein is useful for detecting the presence of its target (e.g. CD3 or CEA) in a biological sample.
  • detecting encompasses quantitative or qualitative detection.
  • a biological sample comprises a cell or tissue, such as prostate tissue.
  • an antibody according to the invention for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of CD3 or CEA in a biological sample is provided.
  • the method comprises contacting the biological sample with an antibody of the present invention under conditions permissive for binding of the antibody to CD3 or CEA, and detecting whether a complex is formed between the antibody and CD3 or CEA.
  • Such method may be an in vitro or in vivo method.
  • an antibody of the invention is used to select subjects eligible for therapy with an antibody that binds CD3 and/or CEA, e.g. where CD3 and/or CEA is a biomarker for selection of patients.
  • Exemplary disorders that may be diagnosed using an antibody of the invention include cancer, particularly colorectal cancer, pancreatic cancer, gastric cancer, non-small cell lung cancer or breast cancer.
  • an antibody according to the present invention is provided, wherein the antibody is labelled.
  • 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 1, 3 H, and 133 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. Patent No.
  • luciferin 2,3- dihydrophthalazinediones
  • horseradish peroxidase HRP
  • alkaline phosphatase b-galactosidase
  • glucoamylase lysozyme
  • saccharide oxidases e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase
  • heterocyclic oxidases such as uricase and xanthine oxidase
  • 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.
  • CD3 binder Starting from a previously described (see e.g. WO 2014/131712, incorporated herein by reference) CD3 binder, termed “CD3 0 rig” herein and comprising the VH and VL sequences of SEQ ID NOs 6 and 11, respectively, we aimed at optimizing properties of this binder by removal of two asparagine deamidation sequence motifs at Kabat positions 97 and 100 of the heavy chain CDR3.
  • This library was put on a filamentous phage via fusion to minor coat protein p3 (Marks et al. (1991) J Mol Biol 222, 581-597) and selected for binding to recombinant CD3 .
  • CD3 opt The selected clone, termed “CD3 opt ” herein and comprising the VH and VL sequences of SEQ ID NOs 7 and 11, respectively, was further evaluated and converted into bispecific format as described in the following.
  • Binding to recombinant CD3 was determined by surface plasmon resonance (SPR) for the optimized CD3 binder “CD3 opt ” and the original CD3 binder “CD3 0rig ”, both in human IgGi format with P329G L234A L235A (“PGLALA”, EU numbering) mutations in the Fc region (SEQ ID NOs 12 and 14 (CD3 orig ) and SEQ ID NOs 13 and 14 (CD3 opt )).
  • SPR surface plasmon resonance
  • RAC Relative Active Concentration
  • CD3 antigen (see below) was injected at a concentration of 10 pg/ml for 120 s and dissociation was monitored at a flow rate of 5 m ⁇ /min for 120 s.
  • the chip surface was regenerated by two consecutive injections of 10 rnM glycine pH 2.1 for 60 s each. Bulk refractive index differences were corrected by subtracting blank injections and by subtracting the response obtained from the blank control flow cell. For evaluation, the binding response was taken 5 seconds after injection end. To normalize the binding signal, the CD3 binding was divided by the anti-Fab response (the signal (RU) obtained upon capture of the CD3 antibody on the immobilized anti-Fab antibody). The relative active concentration was calculated by referencing each temperature stressed sample to the corresponding, non-stressed sample.
  • the antigen used was a heterodimer of CD3 delta and CD3 epsilon ectodomains fused to a human Fc domain with knob-into-hole modifications and a C -terminal Avi-tag (see SEQ ID NOs 44 and
  • Binding to CD3 on the human reporter T-cell line Jurkat NFAT was determined by FACS for the optimized CD3 binder “CD3 opt ” and the original CD3 binder “CD3 0rig ”, both in human IgGi format with P329G L234A L235A (“PGLALA”, EU numbering) mutations in the Fc region (SEQ ID NOs 12 and 14 (CD3 orig ) and SEQ ID NOs 13 and 14 (CD3 opt )).
  • Jurkat-NFAT reporter cells are a human acute lymphatic leukemia reporter cell line with a NFAT promoter, expressing human CD3.
  • the cells were cultured in RPMI1640, 2g/l glucose, 2 g/1 NaHCCE, 10% FCS, 25 mM HEPES, 2 mM L-glutamine, 1 x NEAA, 1 x sodium-pyruvate at 0.1-0.5 mio cells per ml.
  • a final concentration of 200 pg per ml hygromycin B was added whenever cells were passaged.
  • Jurkat NFAT cells were harvested, washed with PBS and resuspended in FACS buffer.
  • the antibody staining was performed in a 96-well round bottom plate. Therefore 100 ⁇ 00 to 200 ⁇ 00 cells were seeded per well.
  • the plate was centrifuged for 4 min at 400 x g and the supernatant was removed.
  • the test antibodies were diluted in FACS buffer and 20 m ⁇ of the antibody solution were added to the cells for 30 min at 4°C.
  • the cells were washed twice with FACS buffer before addition of the diluted secondary antibody (PE- conjugated AffmiPure F(ab’)2 Fragment goat anti-human IgG Fcg Fragment Specific; Jackson ImmunoResearch #109-116-170). After 30 min incubation at 4°C unbound secondary antibody was washed away. Before measurement the cells were resuspended in 200 m ⁇ FACS buffer and then analyzed by flow cytometry using a BD Canto II device.
  • PE- conjugated AffmiPure F(ab’)2 Fragment goat anti-human IgG Fcg Fragment Specific Jackson ImmunoResearch #109-116-170.
  • the optimized CD3 binder “CD3 opt ” and the original CD3 binder “CD3 0 rig” bound comparably well to CD3 on Jurkat cells.
  • the functional activity of the optimized CD3 binder “CD3 opt ” was tested in a Jurkat reporter cell assay and compared to the activity of the original CD3 binder “CD3 0rig ”.
  • anti-PGLALA expressing CHO cells were co-incubated with Jurkat NFAT reporter cells in the presence of increasing concentrations of CD3 opt human IgGi PGLALA or CD3 o rig human IgGi PGLALA.
  • Activation of CD3 on the Jurkat NFAT reporter cells upon T cell cross-linking induces the production of luciferase and luminescence can be measured as an activation marker.
  • CD3 0 rig human IgGi wt was included as negative control which cannot bind to anti-PGLALA expressing CHO cells and therefore cannot be crosslinked on Jurkat NFAT cells.
  • a schematic illustration of the assay is provided in Figure 4.
  • Anti-PGLALA expressing CHO cells are CHO-K1 cells engineered to express on their surface an antibody that specifically binds human IgGi Fc(PGLALA) (see WO 2017/072210, incorporated herein by reference). These cells were cultured in DMEM/F12 medium containing 5% FCS + 1% GluMax. The Jurkat NFAT reporter cells are as described in Example 2.
  • the NFAT promoter Upon simultaneous binding of the CD3 huIgGi PGLALA to anti-PGLALA expressed on CHO and CD3 expressed on Jurkat-NFAT reporter cells, the NFAT promoter is activated and leads to expression of active firefly luciferase.
  • the intensity of luminescence signal (obtained upon addition of luciferase substrate) is proportional to the intensity of CD3 activation and signaling.
  • Jurkat-NFAT reporter cells grow in suspension and were cultured in RPMI1640, 2g/l glucose, 2 g/1 NaHC03, 10 % FCS, 25 mM HEPES, 2 mM L-glutamin, 1 x NEAA, 1 x sodium-pyruvate at 0.1-0.5 mio cells per ml, 200 pg per ml hygromycin.
  • CHO cells were harvested and viability determined using ViCell. 30 000 target cells/well were plated in a flat-bottom, white- walled 96-well-plate (Greiner bio-one #655098) in 100 m ⁇ medium and 50 m 1/well of diluted antibodies or medium (for controls) were added to the CHO cells.
  • Jurkat-NFAT reporter cells were harvested and viability assessed using ViCell.
  • Cells were resuspended at 1.2 mio cells/ml in cell culture medium without hygromycin B and added to CHO cells at 60 000 cells/well (50 m 1/well) to obtain a final effector-to-target (E:T) ratio of 2:1 and a final volume of 200 m ⁇ per well.
  • E:T effector-to-target
  • 4 m ⁇ of Glo Sensor Promega #E1291 was added to each well (2% of final volume).
  • Cells were incubated for 24 h at 37°C in a humidified incubator. At the end of incubation time, luminescence was detected using TEC AN Spark 10M.
  • the optimized CD3 binder CD3 opt had a similar activity on Jurkat NFAT cells upon crosslinking as CD3 0 rig.
  • Example 4 Generation of T-cell bispecific antibody comprising optimized CD3 binder
  • the optimized CD3 binder identified in Example 1 (“CD3 opt ”, SEQ ID NOs 7 (VH) and 11 (VL)) was used to generate T-cell bispecific antibodies (TCBs) targeting CD3 and CEA (CEACAM5) (“CEACAM5-TCB”), using an anti-CEA antibody (SEQ ID NOs 15-22) as CEA binding moiety.
  • FIG. 6 A A schematic illustration of the TCB molecule is provided in Figure 6 A, and its full sequences are given in SEQ ID NOs 23, 24, 25 and 27.
  • variable region of heavy and light chain DNA sequences were subcloned in frame with either the constant heavy chain or the constant light chain pre-inserted into the respective recipient mammalian expression vectors as shown in Figure 6 B-E.
  • knob-into-hole mutations were introduced in the constant region of the antibody heavy chains (T366W/S354C and T366S/L368A/Y407V/ Y349C, respectively).
  • P329G, L234A and L235A mutations were introduced in the constant region of the antibody heavy chains to abrogate binding to Fey receptors.
  • the TCB was prepared by Evitria (Switzerland) using their proprietary vector system with conventional (non-PCR based) cloning techniques and using suspension-adapted CHO K1 cells (originally received from ATCC and adapted to serum-free growth in suspension culture at Evitria).
  • Evitria used its proprietary, animal-component free and serum-free media (eviGrow and eviMake2) and its proprietary transfection reagent (eviFect).
  • the cells were transfected with the corresponding expression vectors in a 1 : 1 :2: 1 (“vector knob heavy chain” “‘vector hole heavy chain” “‘vector CD3 light chain” “‘vector CEA light chain”). Supernatant was harvested by centrifugation and subsequent filtration (0.2 pm filter).
  • Protein was purified from the harvested supernatant by standard methods.
  • Fc containing protein was purified from filtered cell culture supernatants by Protein A-affmity chromatography (equilibration buffer: 20 mM sodium citrate, 20 mM sodium phosphate, pH 7.5; elution buffer: 20 mM sodium citrate, pH 3.0). Elution was achieved at pH 3.0 followed by immediate pH neutralization of the sample.
  • the protein was concentrated by centrifugation (Millipore Amicon® ULTRA- 15, #UFC903096), and aggregated protein was separated from monomeric protein by size exclusion chro matography in 20 mM histidine, 140 mM sodium chloride, pH 6.0.
  • the concentration of purified protein were determined by measuring the absorption at 280 nm using the mass extinction coefficient calculated on the basis of the amino acid sequence according to Pace, et al, Protein Science, 1995, 4, 2411-1423. Purity and molecular weight of the proteins were analyzed by CE-SDS in the presence and absence of a reducing agent using a LabChipGXII (Perkin Elmer).
  • Determination of the aggregate content was performed by HPLC chromatography at 25°C using analytical size-exclusion column (TSKgel G3000 SW XL or UP-SW3000) equilibrated in running buffer (25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrocloride, pH 6.7 or 200 mM KH2PO4, 250 mM KC1 pH 6.2, respectively).
  • Results from the biochemical and biophysical analysis of the prepared TCB molecule are given in Table 1.
  • the TCB molecules could be produced in good quality.
  • Example 5 Binding of CE AC AM5-T CB molecules with optimized anti-CD3 antibodies to human CEACAM5- and human CD3-expressing cells
  • CEACAM5-TCB molecules prepared in Example 4 were tested on CEACAM5 (CEA)-positive tumor cells (LS-180 cells, ECACC #87021202) and CD3 -expressing immortalized T lymphocyte cells (GloResponse Jurkat NFAT-RE-luc2P; Promega, #CS176501). Briefly, adherent LS-180 cells were detached, using Cell Dissociation Buffer (Gibco), counted, checked for viability and re-suspended at 2 million cells per ml in FACS buffer (PBS with 0.1% BSA). Likewise, Jurkat suspension cells were harvested and plated for subsequent stainings.
  • the staining was fixed for 20 min at 4°C in the dark, using 150 m ⁇ of 2% PFA in FACS buffer per well. Fluorescence was analyzed by FACS using a FACS Fortessa (Software FACS Diva). Binding curves were obtained using GraphPadPrism6.
  • Example 6 Tumor cell lysis, induced by CEACAM5-TCB molecules with optimized anti- CD3 antibodies
  • Tumor cell lysis mediated by the CE AC AM5 -T CB molecules prepared in Example 4 was assessed in presence of CEACAM5-expressing LS-180 human tumor cells (ECACC #87021202).
  • Human PBMCs were used as effectors and tumor cell lysis was detected after 24 h and 48 h in presence of the bispecific antibody. Briefly, adherent target cells were harvested with trypsin-EDTA (Life Technologies, #25300096), washed, and plated at density of 25 000 cells/well using flat-bottom 96-well.
  • PBMCs Peripheral blood mononuclear cells
  • the mixture was centrifuged (400 x g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350 x g, 10 minutes).
  • the resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 2% FCS and 1% L-alanyl-L-glutamine (Biochrom, #K0302) at 37°C, in a humidified incubator until the assay was started.
  • the antibody was added at the indicated concentrations (range of 0.3 pM - 20 nM in triplicates).
  • PBMCs were added to target cells to obtain a final E:T ratio of 10:1.
  • Target cell killing was assessed after 24 h and 48 h of incubation at 37°C, 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/ necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001).
  • the EC50 values of tumor cell lysis were calculated using GraphPadPrism6, see Table 2
  • Example 7 Determination of thermal stability of CEACAM5-TCB molecules with optimized anti-CD3 antibodies
  • Example 8 Functional characterization of CEACAM5-TCB molecules with optimized anti- CD3 antibodies by surface plasmon resonance (SPR)
  • CEACAM5 TCB molecules prepared in Example 4 were captured on a Cl sensorchip (GE Healthcare) surface with immobilized anti-Fc(P329G) IgG (an antibody that specifically binds human IgGi Fc(P329G); “anti-PG antibody” - see WO 2017/072210, incorporated herein by reference).
  • the experimental setup is schematically depicted in Figure 9. Capture IgG was coupled to the sensorchip surface by direct immobilization of around 400 resonance units (RU) using the standard amine coupling kit (GE Healthcare Life Sciences).
  • TCB molecules were captured for 40 s at 25 nM with a flow rate of 10 m ⁇ /min.
  • Human and cynomolgus CD3 stalk-F c(knob)- Avi/ CD35 stalk-Fc(hole) (CD3 /5, see SEQ ID NOs 44 and 45 (human) and SEQ ID NOs 56 and 57 (cynomolgus)) were passed at a concentration of 0.5 - 50 nM with a flow rate of 30 m ⁇ /min through the flow cells for 240 s. The dissociation was monitored for 800 s.
  • Example 9 Characterization of CE AC AM5-T CB molecules with optimized anti-CD3 antibodies by surface plasmon resonance (SPR) after stress
  • CE AC AM5 -T CB molecules prepared in Example 4 were incubated for 14 days at 37°C, pH 7.4 and at 40°C, pH 6 and further analyzed by SPR for their binding capability to human CD3 /5.
  • Samples stored at -80°C pH 6 were used as reference.
  • the reference samples and the samples stressed at 40°C were in 20 mM His, 140 mM NaCl, pH 6.0, and the samples stressed at 37°C in PBS, pH 7.4, all at a concentration of 1.0 mg/ml. After the stress period (14 days) samples in PBS were dialyzed back to 20 mM His, 140 mM NaCl, pH 6.0 for further analysis.
  • TCBs with a concentration of 2 pg/ml were injected for 30 s at a flow rate of 5 m ⁇ /min, and dissociation was monitored for 120 s.
  • the surface was regenerated by injecting 10 mM glycine pH 1.5 for 60 s. Bulk refractive index differences were corrected by subtracting blank injections and by subtracting the response obtained from a blank control flow cell. For evaluation, the binding response 5 seconds after injection end was taken. To normalize the binding signal, the CD3 binding was divided by the anti-huIgG response (the signal (RU) obtained upon capture of the TCB on the immobilized anti-huIgG antibody). The relative binding activity was calculated by referencing each temperature stressed sample to the corresponding, non- stressed sample.
  • the CE AC AM5 -T CB comprising the optimized CD3 binder CD3 opt shows an improved binding upon stress to CD3 /5, as compared to the TCB comprising CD3 0 rig.

Abstract

La présente invention concerne de manière générale des anticorps qui se lient à CD3 et CEA, par exemple pour activer des lymphocytes T. De plus, la présente invention concerne des polynucléotides codant pour de tels anticorps, et des vecteurs et des cellules hôtes comprenant de tels polynucléotides. L'invention concerne en outre des procédés de production des anticorps et des procédés d'utilisation de ceux-ci dans le traitement d'une maladie.
PCT/EP2021/066354 2020-06-19 2021-06-17 Anticorps se liant à cd3 et cea WO2021255146A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20181076.9 2020-06-19
EP20181076 2020-06-19

Publications (1)

Publication Number Publication Date
WO2021255146A1 true WO2021255146A1 (fr) 2021-12-23

Family

ID=71111328

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/066354 WO2021255146A1 (fr) 2020-06-19 2021-06-17 Anticorps se liant à cd3 et cea

Country Status (1)

Country Link
WO (1) WO2021255146A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11591397B2 (en) 2018-12-21 2023-02-28 Hoffmann-La Roche Inc. Bispecific antibody molecules binding to CD3 and EGFRvIII
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19

Citations (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1993001161A1 (fr) 1991-07-11 1993-01-21 Pfizer Limited Procede de preparation d'intermediaires de sertraline
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
WO1994011026A2 (fr) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Application therapeutique d'anticorps chimeriques et radio-marques contre l'antigene a differentiation restreinte des lymphocytes b humains pour le traitement du lymphome des cellules b
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
WO1996027011A1 (fr) 1995-03-01 1996-09-06 Genentech, Inc. Procede d'obtention de polypeptides heteromultimeriques
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1997030087A1 (fr) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation d'anticorps glycosyles
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1998050431A2 (fr) 1997-05-02 1998-11-12 Genentech, Inc. Procede de preparation d'anticorps multispecifiques presentant des composants heteromultimeres
WO1998058964A1 (fr) 1997-06-24 1998-12-30 Genentech, Inc. Procedes et compositions concernant des glycoproteines galactosylees
WO1999022764A1 (fr) 1997-10-31 1999-05-14 Genentech, Inc. Compositions renfermant des glycoformes de glycoproteine et methodes afferentes
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
WO1999054342A1 (fr) 1998-04-20 1999-10-28 Pablo Umana Modification par glycosylation d'anticorps aux fins d'amelioration de la cytotoxicite cellulaire dependant des anticorps
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
WO2001007611A2 (fr) 1999-07-26 2001-02-01 Genentech, Inc. Nouveaux polynucleotides et technique d'utilisation de ceux-ci
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
WO2001077342A1 (fr) 2000-04-11 2001-10-18 Genentech, Inc. Anticorps multivalents et leurs utilisations
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
WO2003011878A2 (fr) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Variants de glycosylation d'anticorps presentant une cytotoxicite cellulaire accrue dependante des anticorps
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
WO2003085107A1 (fr) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cellules à génome modifié
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
WO2004056312A2 (fr) 2002-12-16 2004-07-08 Genentech, Inc. Variants d'immunoglobuline et utilisations
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2004065540A2 (fr) 2003-01-22 2004-08-05 Glycart Biotechnology Ag Constructions hybrides et leur utilisation pour produire des anticorps presentant une affinite de liaison accrue pour le recepteur fc et fonction d'effecteur
WO2004106381A1 (fr) 2003-05-31 2004-12-09 Micromet Ag Compositions pharmaceutiques comprenant des constructions d'anticorps anti-cd3, anti-cd19 bispecifiques pour le traitement de troubles associes aux lymphocytes b
US20040259150A1 (en) 2002-04-09 2004-12-23 Kyowa Hakko Kogyo Co., Ltd. Method of enhancing of binding activity of antibody composition to Fcgamma receptor IIIa
US20050031613A1 (en) 2002-04-09 2005-02-10 Kazuyasu Nakamura Therapeutic agent for patients having human FcgammaRIIIa
WO2005061547A2 (fr) 2003-12-22 2005-07-07 Micromet Ag Anticorps bispecifiques
WO2005100402A1 (fr) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anticorps anti-p-selectine
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
WO2006029879A2 (fr) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anticorps anti-ox40l
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO2006082515A2 (fr) 2005-02-07 2006-08-10 Glycart Biotechnology Ag Molecules de liaison d'antigenes se liant au recepteur egfr, vecteurs codant pour ces molecules et leurs applications
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2007042261A2 (fr) 2005-10-11 2007-04-19 Micromet Ag Compositions comportant des anticorps specifiques d'especes croisees et leurs utilisations
WO2007110205A2 (fr) 2006-03-24 2007-10-04 Merck Patent Gmbh Domaines de proteine heterodimerique d'ingenierie
EP1870459A1 (fr) 2005-03-31 2007-12-26 Chugai Seiyaku Kabushiki Kaisha Procede pour la production de polypeptide au moyen de la regulation d'un ensemble
WO2007147901A1 (fr) 2006-06-22 2007-12-27 Novo Nordisk A/S Production d'anticorps bispécifiques
WO2008024715A2 (fr) 2006-08-21 2008-02-28 Welczer Avelyn Legal Represent Traitement d'amygdalite
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008119567A2 (fr) 2007-04-03 2008-10-09 Micromet Ag Domaine de liaison spécifique d'espèces croisées
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
WO2009080251A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009080253A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009080252A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
WO2010112193A1 (fr) 2009-04-02 2010-10-07 Roche Glycart Ag Anticorps multispécifiques renfermant des anticorps de longueur entière et des fragments fab à chaîne unique
WO2010115589A1 (fr) 2009-04-07 2010-10-14 Roche Glycart Ag Anticorps trivalents bispécifiques
WO2010129304A2 (fr) 2009-04-27 2010-11-11 Oncomed Pharmaceuticals, Inc. Procédé de fabrication de molécules hétéromultimères
WO2010136172A1 (fr) 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Anticorps tri- ou tétraspécifiques
WO2010145792A1 (fr) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Protéines bispécifiques se liant à un antigène
WO2011034605A2 (fr) 2009-09-16 2011-03-24 Genentech, Inc. Complexes protéiques contenant une super-hélice et/ou une attache et leurs utilisations
WO2011090762A1 (fr) 2009-12-29 2011-07-28 Emergent Product Development Seattle, Llc Protéines de liaison hétérodimères et utilisations de celles-ci
WO2011143545A1 (fr) 2010-05-14 2011-11-17 Rinat Neuroscience Corporation Protéines hétérodimériques et leurs procédés de production et de purification
WO2012058768A1 (fr) 2010-11-05 2012-05-10 Zymeworks Inc. Conception d'anticorps hétérodimérique stable ayant des mutations dans le domaine fc
WO2012130831A1 (fr) 2011-03-29 2012-10-04 Roche Glycart Ag Variants de fc d'anticorps
WO2013026833A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t.
WO2013026831A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Molécules bispécifiques de liaison à un antigène
WO2013026839A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Anticorps bispécifiques spécifiques pour les antigènes d'activation des lymphocytes t et un antigène tumoral et procédés d'utiliation correspondants
WO2013096291A2 (fr) 2011-12-20 2013-06-27 Medimmune, Llc Polypeptides modifiés pour des échafaudages d'anticorps bispécifiques
WO2013120929A1 (fr) 2012-02-15 2013-08-22 F. Hoffmann-La Roche Ag Chromatographie d'affinité faisant appel à des récepteurs fc
WO2013157954A1 (fr) 2012-04-20 2013-10-24 Merus B.V. Procédés et moyens de production de molécules de type ig
WO2014131712A1 (fr) 2013-02-26 2014-09-04 Roche Glycart Ag Molécules de liaison à l'antigène bispécifiques activant des lymphocytes t
US9000130B2 (en) 2010-06-08 2015-04-07 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2015095539A1 (fr) 2013-12-20 2015-06-25 Genentech, Inc. Anticorps à double spécificité
WO2015150447A1 (fr) 2014-04-02 2015-10-08 F. Hoffmann-La Roche Ag Anticorps multispécifiques
WO2016016299A1 (fr) 2014-07-29 2016-02-04 F. Hoffmann-La Roche Ag Anticorps multispécifiques
WO2016020309A1 (fr) 2014-08-04 2016-02-11 F. Hoffmann-La Roche Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t
WO2016040856A2 (fr) 2014-09-12 2016-03-17 Genentech, Inc. Anticorps et conjugués modifiés génétiquement avec de la cystéine
WO2016079076A1 (fr) * 2014-11-20 2016-05-26 F. Hoffmann-La Roche Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t ciblant folr1 et cd3
WO2016172485A2 (fr) 2015-04-24 2016-10-27 Genentech, Inc. Protéines multispécifiques de liaison à l'antigène
EP2101823B1 (fr) 2007-01-09 2016-11-23 CureVac AG Anticorps code par un arn
WO2017055389A1 (fr) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t anti-ceaxcd3
WO2017072210A1 (fr) 2015-10-29 2017-05-04 F. Hoffmann-La Roche Ag Anticorps anti-région fc variante et procédés d'utilisation
WO2020127619A1 (fr) * 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Anticorps se liant à cd3
WO2020127618A1 (fr) * 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Molécules de liaison à l'antigène cd28 agonistes de ciblage de tumeurs

Patent Citations (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. 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
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993001161A1 (fr) 1991-07-11 1993-01-21 Pfizer Limited Procede de preparation d'intermediaires de sertraline
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
WO1994011026A2 (fr) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Application therapeutique d'anticorps chimeriques et radio-marques contre l'antigene a differentiation restreinte des lymphocytes b humains pour le traitement du lymphome des cellules b
WO1996027011A1 (fr) 1995-03-01 1996-09-06 Genentech, Inc. Procede d'obtention de polypeptides heteromultimeriques
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US7695936B2 (en) 1995-03-01 2010-04-13 Genentech, Inc. Knobs and holes heteromeric polypeptides
WO1997030087A1 (fr) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation d'anticorps glycosyles
WO1998050431A2 (fr) 1997-05-02 1998-11-12 Genentech, Inc. Procede de preparation d'anticorps multispecifiques presentant des composants heteromultimeres
WO1998058964A1 (fr) 1997-06-24 1998-12-30 Genentech, Inc. Procedes et compositions concernant des glycoproteines galactosylees
WO1999022764A1 (fr) 1997-10-31 1999-05-14 Genentech, Inc. Compositions renfermant des glycoformes de glycoproteine et methodes afferentes
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO1999054342A1 (fr) 1998-04-20 1999-10-28 Pablo Umana Modification par glycosylation d'anticorps aux fins d'amelioration de la cytotoxicite cellulaire dependant des anticorps
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
WO2001007611A2 (fr) 1999-07-26 2001-02-01 Genentech, Inc. Nouveaux polynucleotides et technique d'utilisation de ceux-ci
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2001077342A1 (fr) 2000-04-11 2001-10-18 Genentech, Inc. Anticorps multivalents et leurs utilisations
WO2003011878A2 (fr) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Variants de glycosylation d'anticorps presentant une cytotoxicite cellulaire accrue dependante des anticorps
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
US20040259150A1 (en) 2002-04-09 2004-12-23 Kyowa Hakko Kogyo Co., Ltd. Method of enhancing of binding activity of antibody composition to Fcgamma receptor IIIa
US20050031613A1 (en) 2002-04-09 2005-02-10 Kazuyasu Nakamura Therapeutic agent for patients having human FcgammaRIIIa
WO2003085107A1 (fr) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cellules à génome modifié
WO2004056312A2 (fr) 2002-12-16 2004-07-08 Genentech, Inc. Variants d'immunoglobuline et utilisations
WO2004065540A2 (fr) 2003-01-22 2004-08-05 Glycart Biotechnology Ag Constructions hybrides et leur utilisation pour produire des anticorps presentant une affinite de liaison accrue pour le recepteur fc et fonction d'effecteur
WO2004106381A1 (fr) 2003-05-31 2004-12-09 Micromet Ag Compositions pharmaceutiques comprenant des constructions d'anticorps anti-cd3, anti-cd19 bispecifiques pour le traitement de troubles associes aux lymphocytes b
WO2005061547A2 (fr) 2003-12-22 2005-07-07 Micromet Ag Anticorps bispecifiques
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
WO2005100402A1 (fr) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anticorps anti-p-selectine
WO2006029879A2 (fr) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anticorps anti-ox40l
US7855275B2 (en) 2004-09-23 2010-12-21 Genentech, Inc. Cysteine engineered antibodies and conjugates
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2006082515A2 (fr) 2005-02-07 2006-08-10 Glycart Biotechnology Ag Molecules de liaison d'antigenes se liant au recepteur egfr, vecteurs codant pour ces molecules et leurs applications
EP1870459A1 (fr) 2005-03-31 2007-12-26 Chugai Seiyaku Kabushiki Kaisha Procede pour la production de polypeptide au moyen de la regulation d'un ensemble
WO2007042261A2 (fr) 2005-10-11 2007-04-19 Micromet Ag Compositions comportant des anticorps specifiques d'especes croisees et leurs utilisations
WO2007110205A2 (fr) 2006-03-24 2007-10-04 Merck Patent Gmbh Domaines de proteine heterodimerique d'ingenierie
WO2007147901A1 (fr) 2006-06-22 2007-12-27 Novo Nordisk A/S Production d'anticorps bispécifiques
WO2008024715A2 (fr) 2006-08-21 2008-02-28 Welczer Avelyn Legal Represent Traitement d'amygdalite
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
EP2101823B1 (fr) 2007-01-09 2016-11-23 CureVac AG Anticorps code par un arn
WO2008119567A2 (fr) 2007-04-03 2008-10-09 Micromet Ag Domaine de liaison spécifique d'espèces croisées
WO2009080251A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009080253A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009080252A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
WO2010112193A1 (fr) 2009-04-02 2010-10-07 Roche Glycart Ag Anticorps multispécifiques renfermant des anticorps de longueur entière et des fragments fab à chaîne unique
WO2010115589A1 (fr) 2009-04-07 2010-10-14 Roche Glycart Ag Anticorps trivalents bispécifiques
WO2010129304A2 (fr) 2009-04-27 2010-11-11 Oncomed Pharmaceuticals, Inc. Procédé de fabrication de molécules hétéromultimères
WO2010136172A1 (fr) 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Anticorps tri- ou tétraspécifiques
WO2010145792A1 (fr) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Protéines bispécifiques se liant à un antigène
WO2011034605A2 (fr) 2009-09-16 2011-03-24 Genentech, Inc. Complexes protéiques contenant une super-hélice et/ou une attache et leurs utilisations
WO2011090762A1 (fr) 2009-12-29 2011-07-28 Emergent Product Development Seattle, Llc Protéines de liaison hétérodimères et utilisations de celles-ci
WO2011090754A1 (fr) 2009-12-29 2011-07-28 Emergent Product Development Seattle, Llc Hétérodimères polypeptidiques et leurs utilisations
WO2011143545A1 (fr) 2010-05-14 2011-11-17 Rinat Neuroscience Corporation Protéines hétérodimériques et leurs procédés de production et de purification
US9000130B2 (en) 2010-06-08 2015-04-07 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2012058768A1 (fr) 2010-11-05 2012-05-10 Zymeworks Inc. Conception d'anticorps hétérodimérique stable ayant des mutations dans le domaine fc
WO2012130831A1 (fr) 2011-03-29 2012-10-04 Roche Glycart Ag Variants de fc d'anticorps
WO2013026831A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Molécules bispécifiques de liaison à un antigène
WO2013026833A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t.
WO2013026839A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Anticorps bispécifiques spécifiques pour les antigènes d'activation des lymphocytes t et un antigène tumoral et procédés d'utiliation correspondants
WO2013096291A2 (fr) 2011-12-20 2013-06-27 Medimmune, Llc Polypeptides modifiés pour des échafaudages d'anticorps bispécifiques
WO2013120929A1 (fr) 2012-02-15 2013-08-22 F. Hoffmann-La Roche Ag Chromatographie d'affinité faisant appel à des récepteurs fc
WO2013157954A1 (fr) 2012-04-20 2013-10-24 Merus B.V. Procédés et moyens de production de molécules de type ig
WO2013157953A1 (fr) 2012-04-20 2013-10-24 Merus B.V. Procédés et moyens de production de molécules de type ig
WO2014131712A1 (fr) 2013-02-26 2014-09-04 Roche Glycart Ag Molécules de liaison à l'antigène bispécifiques activant des lymphocytes t
WO2015095539A1 (fr) 2013-12-20 2015-06-25 Genentech, Inc. Anticorps à double spécificité
WO2015150447A1 (fr) 2014-04-02 2015-10-08 F. Hoffmann-La Roche Ag Anticorps multispécifiques
WO2016016299A1 (fr) 2014-07-29 2016-02-04 F. Hoffmann-La Roche Ag Anticorps multispécifiques
WO2016020309A1 (fr) 2014-08-04 2016-02-11 F. Hoffmann-La Roche Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t
WO2016040856A2 (fr) 2014-09-12 2016-03-17 Genentech, Inc. Anticorps et conjugués modifiés génétiquement avec de la cystéine
WO2016079076A1 (fr) * 2014-11-20 2016-05-26 F. Hoffmann-La Roche Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t ciblant folr1 et cd3
WO2016172485A2 (fr) 2015-04-24 2016-10-27 Genentech, Inc. Protéines multispécifiques de liaison à l'antigène
WO2017055389A1 (fr) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t anti-ceaxcd3
WO2017072210A1 (fr) 2015-10-29 2017-05-04 F. Hoffmann-La Roche Ag Anticorps anti-région fc variante et procédés d'utilisation
WO2020127619A1 (fr) * 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Anticorps se liant à cd3
WO2020127618A1 (fr) * 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Molécules de liaison à l'antigène cd28 agonistes de ciblage de tumeurs

Non-Patent Citations (72)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. BAB71849.1
"NCBI", Database accession no. NP _004354.2
"Remington's Pharmaceutical Sciences", 1990, MACK PRINTING COMPANY, pages: 1289 - 1329
"UniProt", Database accession no. P06731
ALMAGROFRANSSON, FRONT. BIOSCI., vol. 13, 2008, pages 1619 - 1633
ATWELL ET AL., J. MOL. BIOL., vol. 270, 1997, pages 26
BACA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684
BACAC ET AL., ONCOIMMUNOLOGY, vol. 5, no. 8, 2016, pages e1203498
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
BRUGGEMANN ET AL., J EXP MED, vol. 166, 1987, pages 1351 - 1361
CARTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 4285
CARTER, J IMMUNOL METH, vol. 248, 2001, pages 7 - 15
CARTER, J IMMUNOL METHODS, vol. 248, 2001, pages 7 - 15
CHARI ET AL., CANCER RES, vol. 52, 1992, pages 127 - 131
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLARKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLYNES ET AL., PROC NATL ACAD SCI USA, vol. 95, 1998, pages 652 - 656
CRAGG ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
CRAGGGLENNIE, BLOOD, vol. 103, 2004, pages 2738 - 2743
DALL'ACQUA ET AL., METHODS, vol. 36, 2005, pages 61 - 68
FERRARA ET AL., BIOTECHN BIOENG, vol. 93, 2006, pages 851 - 861
FINGL ET AL.: "The Pharmacological Basis of Therapeutics", 1975, pages: 1
FLATMAN ET AL., J. CHROMATOGR. B, vol. 848, 2007, pages 79 - 87
GAZZANO-SANTORO ET AL., J IMMUNOL METHODS, vol. 202, 1996, pages 163
GERNGROSS, NAT BIOTECH, vol. 22, 2004, pages 1409 - 1414
GRAHAM ET AL., J GEN VIROL, vol. 36, 1977, pages 59
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368 - 315
HELLSTROM ET AL., PROC NATL ACAD SCI USA, vol. 82, 1985, pages 1499 - 1502
HELLSTROM ET AL., PROC NATL ACAD SCI USA, vol. 83, 1986, pages 7059 - 7063
HOLLIGER ET AL., PROT ENG, vol. 9, 1996, pages 617 - 621
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOLLINGERHUDSON, NATURE BIOTECHNOLOGY, vol. 23, 2005, pages 1126 - 1136
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
JOHNSON ET AL., J MOL BIOL, vol. 399, 2010, pages 436 - 449
KANDA, Y ET AL., BIOTECHNOL. BIOENG., vol. 94, no. 4, 2006, pages 680 - 688
KINDT ET AL.: "Kuby Immunology", vol. 91, 2007, W.H. FREEMAN & CO.
KIPRIYANOV ET AL., J MOL BIOL, vol. 293, 1999, pages 41 - 56
KLEIN, MABS, vol. 8, 2016, pages 1010 - 20
KLIMKA ET AL., BR. J. CANCER, vol. 83, 2000, pages 252 - 260
KOSTELNY ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
LI ET AL., NAT BIOTECH, vol. 24, 2006, pages 210 - 215
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 745
MANIATIS ET AL.: "CURRENT PROTOCOLS IN MOLECULAR BIOLOGY", 1989, GREENE PUBLISHING ASSOCIATES AND WILEY INTERSCIENCE
MARKS ET AL., J MOL BIOI, vol. 222, 1991, pages 581 - 597
MATHER ET AL., ANNALS N.Y. ACAD SCI, vol. 383, 1982, pages 44 - 68
MATHER, BIOL REPROD, vol. 23, 1980, pages 243 - 251
MILSTEINCUELLO, NATURE, vol. 305, 1983, pages 537
NAGORSENBAUERLE, EXP CELL RES, vol. 317, 2011, pages 1255 - 1260
PACE ET AL., PROTEIN SCIENCE, vol. 4, 1995, pages 2411 - 1423
PADLAN, MOL. IMMUNOL., vol. 28, 1991, pages 489 - 498
PEARSON, GENOMICS, vol. 46, 1997, pages 24 - 36
PETKOVA, S.B. ET AL., INT'L. IMMUNOL., vol. 18, no. 12, 2006, pages 1759 - 1769
PHARMACOL REVIEW, vol. 68, 2016, pages 3 - 19
PORTOLANO ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623 - 887
QUEEN ET AL., PROC. NAT'LACAD. SCI. USA, vol. 86, 1989, pages 10029 - 10033
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
RIPKA ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
ROSOK ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 22611 - 22618
SCHAEFER ET AL., PNAS, vol. 108, 2011, pages 11187 - 11191
SEIMETZ ET AL., CANCER TREAT REV, vol. 36, 2010, pages 458 - 467
SPIESS ET AL., MOL IMMUNOL, vol. 67, 2015, pages 95 - 106
STADLER ET AL., NATURE MEDICINE, vol. 23, 2017, pages 815 - 817
STUBENRAUCH ET AL., DRUG METABOLISM AND DISPOSITION, vol. 38, 2010, pages 84 - 91
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
UMANA ET AL., NAT BIOTECHNOL, vol. 17, 1999, pages 176 - 180
URLAUB ET AL., PROC NATL ACAD SCI USA, vol. 77, 1980, pages 4216
VITETTA ET AL., SCIENCE, vol. 238, 1987, pages 1098
W. R. PEARSON: "Effective protein sequence comparison", METH. ENZYMOL., vol. 266, 1996, pages 227 - 258
W. R. PEARSOND. J. LIPMAN: "Improved Tools for Biological Sequence Analysis", PNAS, vol. 85, 1988, pages 2444 - 2448
WRIGHT ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614 - 622
YAZAKIWU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 255 - 268

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11591397B2 (en) 2018-12-21 2023-02-28 Hoffmann-La Roche Inc. Bispecific antibody molecules binding to CD3 and EGFRvIII
US11672858B2 (en) 2018-12-21 2023-06-13 Hoffmann-La Roche Inc. Bispecific antibody molecules binding to CD3 and TYRP-1
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19

Similar Documents

Publication Publication Date Title
AU2019410075B2 (en) Antibodies binding to CD3
US11827711B2 (en) Antibodies binding to NKG2D
US20210230278A1 (en) Antibodies binding to HLA-A2/MAGE-A4
US20230416411A1 (en) Antibodies binding to cd3 and folr1
US11780920B2 (en) Antibodies binding to CD3 and CD19
US20200190212A1 (en) Antibodies binding to STEAP-1
US20220010015A1 (en) Antibodies binding to cd3
WO2021255146A1 (fr) Anticorps se liant à cd3 et cea
US20240018240A1 (en) Antibodies binding to cd3 and plap

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21732327

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21732327

Country of ref document: EP

Kind code of ref document: A1