US20210221908A1 - Bispecific antibodies against ceacam5 and cd47 - Google Patents

Bispecific antibodies against ceacam5 and cd47 Download PDF

Info

Publication number
US20210221908A1
US20210221908A1 US15/734,962 US201915734962A US2021221908A1 US 20210221908 A1 US20210221908 A1 US 20210221908A1 US 201915734962 A US201915734962 A US 201915734962A US 2021221908 A1 US2021221908 A1 US 2021221908A1
Authority
US
United States
Prior art keywords
bispecific antibody
seq
cancer
antibody
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/734,962
Other languages
English (en)
Inventor
Vanessa BUATOIS
Sara MAJOCCHI
Klaus Strein
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lamkap Bio Beta Ltd
Original Assignee
Lamkap Bio Beta Ltd
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 Lamkap Bio Beta Ltd filed Critical Lamkap Bio Beta Ltd
Publication of US20210221908A1 publication Critical patent/US20210221908A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K16/30Immunoglobulins [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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins 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
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • 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
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • C12N2015/8518Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic expressing industrially exogenous proteins, e.g. for pharmaceutical use, human insulin, blood factors, immunoglobulins, pseudoparticles

Definitions

  • the present invention relates to bispecific antibodies which bind to human carcinoembryonic antigen CEACAM5 (CEA) and human CD47 (CEAxCD47 bispecific antibodies).
  • CEA human carcinoembryonic antigen
  • CEAxCD47 bispecific antibodies bind to human carcinoembryonic antigen CEACAM5 (CEA) and human CD47 (CEAxCD47 bispecific antibodies).
  • the present invention relates to polynucleotides encoding such bispecific antibodies and vectors and host cells comprising such polynucleotides.
  • the invention further relates to methods for selecting and producing such antibodies and to methods of using such antibodies in the treatment of diseases.
  • the invention also relates to the therapeutic use of the CEAxCD47 bispecific antibodies in monotherapy and in combination therapy, especially with CEAxCD3 T-cell bispecific antibodies (TCB) and/or inhibitors of PD-1 or PD-L1.
  • TAB T-cell bispecific antibodies
  • CEA human CEA family contains 29 genes, of which 18 are expressed: 7 belonging to the CEA subgroup and 11 to the pregnancy-specific glycoprotein subgroup. Several CEA subgroup members are thought to possess cell adhesion properties. CEA is thought to have a role in innate immunity (Hammarstrom S., Semin Cancer Biol. 9(2):67-81 (1999)).
  • Carcinoembryonic antigen CEA, CEACAM5 or CD66e; UniProtKB-P06731
  • CEACAM carcinoembryonic antigen-related cell adhesion molecule
  • Tumoretrogen Gold and Freedman, J Exp. Med., 121:439-462, 1965; Berinstein N.
  • CEACAM6 (CD66c; UniProtKB-P40199) belongs also to the carcinoembryonic antigen (CEA) family. Multiple monoclonal antibodies have been raised against CEA for research purposes, as diagnostic tools, and for therapeutic purposes (see e.g. WO2012117002 (incorporated by reference in its entirety), see also Example 8 f)). Soluble CEA—in this application also called shed CEA or sCEA—is an established tumor marker. Levels in plasma of cancer patients can go in some cases over 1000 ng/ml, whereas plasma concentrations in healthy individuals are below 10 ng/ml (e.g. Sandler B.
  • the mouse monoclonal antibody PR1A3 was raised by fusion of NS1 (P3/NS 1/I-Ag-4-1) myeloma cells with spleen cells from mice immunized with normal colorectal epithelium Richman P. I. and Bodmer W. F., Int. J. Cancer, 39:317-328, 1987 describe mouse monoclonal antibody PR1A3.
  • Epitope mapping of PR1 A3 shows that the antibody targets the B3 domain and the GPI anchor of the CEA molecule (Durbin H. et al., Proc. Natl. Scad. Sci. USA, 91 :4313-4317, 1994).
  • the PR1A3 antibody binds mainly to the membrane- bound CEA, and not the soluble CEA form that can be found in the bloodstreams of cancer patients.
  • the epitope bound by PR1A3 is a conformational epitope, not a linear epitope (Stewart et al., Cancer Immunol Immunother, 47 (1999) 299-06).
  • Humanized PR1 A3 (hPR1 A3) antibodies are described e.g. by Conaghhan P. J., et al., Br. J. Cancer, 98 (2008)1217-1225 and WO2012117002 (incorporated by reference in its entirety).
  • a method for treating cancer by a combination of a human PD-1 axis antagonist and an anti-CEA/anti-CD3 bispecific antibody is mentioned in US20140242079 and WO2017118657 (each of which is incorporated by reference in its entirety) and clinical results have been published at ASCO conference 2017 (Tabernero et al, J Clin Oncol 35, 2017 (suppl;abstr 3002)).
  • a method of treating tumors by administering immune checkpoint antagonists binding two or more different targets of an immune checkpoint pathway, and a T cell-redirecting agent binding to CEA and a T cell surface antigen is mentioned in WO2015112534.
  • a conjugate consisting of a single domain anti-CEACAM6 antibody and urease is at present in clinical trials (NCT02309892; WO2016116907).
  • a class I antibody binding to CEACAM5, CEACAM6 and granulocytes is mentioned in US20110064653.
  • An anti CD3c antibody described in the state of the art is SP34 (Yang S J, The Journal of Immunology (1986) 137; 1097-1100). SP34 reacts with both primate and human CD3. SP34 is available from BD Biosciences.
  • a further anti CD3 antibody described in the state of the art is UCHT-1 (see WO2000041474).
  • a further anti CD3 antibody described in the state of the art is BC-3 (Fred Hutchinson Cancer Research Institute; used in Phase I/II trials of GvHD, Anasetti et al., Transplantation 54: 844 (1992)).
  • SP34 differs from UCHT-1 and BC-3 in that SP-34 recognizes an epitope present on solely the c chain of CD3 (see Salmeron et al., (1991) J. Immunol. 147: 3047) whereas UCHT-1 and BC-3 recognize an epitope contributed by both the ⁇ and ⁇ chains.
  • Anti CD3 antibodies are also described in WO2007042261, WO2008119565, WO2008119566, WO2008119567, WO2010037836, WO2010037837, WO2010037838, and U.S. Pat. No. 8,236,308 (each of which is incorporated by reference in its entirety).
  • a bispecific antibody comprising a binding part specific for CEA and a binding part specific for CD3 ⁇ is described in US20140242079A1 (incorporated by reference in its entirety).
  • Human CD47 (UniProtKB-Q08722 (CD47_HUMAN; IAP) is a transmembrane protein that binds the ligands thrombospondin-1 (TSP-1) and signal-regulatory protein alpha (SIRP ⁇ ; CD172a; UniProtKB P78324) and can act as a “don't eat me” signal to the immune system, especially for macrophages.
  • CD47 is involved in a range of cellular processes, including apoptosis, proliferation, adhesion, and migration. Furthermore, it plays a key role in immune and angiogenic responses. CD47 is overexpressed in different tumor cells.
  • Antibodies against CD47 are described in the state of the art and some are in clinical trials as therapeutic agents for tumor treating (Weiskopf K. European Journal of Cancer 76 (2017) 100-109; Huang Y et al., J Thorac Dis 2017;9(2):E168-E174. Antibodies of the IgG1 subclass that bind CD47 can result in the depletion of platelets and reduction of red blood cells RBC of hemoglobin in a Fc-dependent manner (see e.g. US20140140989).
  • WO2014087248 describe a bispecific antibody against CD19 and CD47.
  • a bispecific antibody against CD19 and CD47 comprising a common heavy chain of SEQ ID NO:5 and a variable light domain VL of SEQ ID NO:10 is described in WO2014087248 (incorporated by reference in its entirety).
  • Human FcRI (CD64) is restricted to monocytes/macrophages and dendritic cells (DCs) and, inducibly expressed on neutrophils and mast cells; hFc RIIA (CD32A) is expressed on all myeloid cells but not on lymphocytes; hFc RIM (CD32B) is highly expressed only on circulating B cells and basophils (L. Cassard, F. Joensson, S. Arnaud, M. Daeron, J.
  • hFc RIIC CD32C
  • hFc RIIIA CD16A
  • hFcRIIIB CD16B
  • hFc RIIA is the only activating IgG receptor constitutively expressed by mast cells, basophils, neutrophils and eosinophils (Bruhns P., Blood 119 (2012) 5640).
  • the biological activities of each subclass of IgG are poorly known.
  • IgG receptors (FcyRs) are strikingly numerous in humans. They comprise high-affinity and low-affinity receptors. Both high-affinity and low-affinity Fc ⁇ Rs bind IgG-immune complexes with a high avidity, but only high-affinity Fc ⁇ Rs bind monomeric IgG.
  • hFc ⁇ RI high-affinity IgG receptor in humans
  • hFc ⁇ RIIA high-affinity IgG receptor
  • IIB two families of low-affinity IgG receptors
  • hFc ⁇ RIIIA low-affinity IgG receptors
  • hFc ⁇ RIIA Fc ⁇ R associated activating receptors
  • hFc ⁇ RIIB single-domain activating receptors
  • hFc ⁇ RIIIB are GPI-anchored receptors whose function is uncertain (Bruhns P. Blood 113 (2009) 3716).
  • an antibody is produced as a population of glycoforms which share the same polypeptide backbone but have different oligosaccharides attached to the glycosylation sites.
  • the oligosaccharides normally found in the Fc region of serum IgG are of complex bi-antennary type (Wormald et al., Biochemistry 36: 130-38 (1997), with a low level of terminal sialic acid and bisecting N-acetylglucosamine (GlcNAc), and a variable degree of terminal galactosylation and core fucosylation.
  • Antibodies with a reduced fucose content in glycan moieties exhibit higher antibody dependent cellular cytotoxicity (ADCC) activity compared to a normally fucosylated antibody (Niwa R et al., Cancer Res, 64, 2127-33, 2004).
  • ADCC antibody dependent cellular cytotoxicity
  • the mechanism behind the enhanced ADCC of a low/no-fucose antibody is its increased affinity to Fc ⁇ RIIIa (CD16).
  • a cell line with knockout of both alleles for the gene responsible for fucose addition is described in U.S. Pat. Nos.
  • US20140242079 and WO2017055389 (each of which is incorporated by reference in its entirety) describe CEAxCD3 T-cell bispecific antibodies.
  • One antibody from US20140242079 and one from W02017055389 are both in clinical development (see clinicaltrials.gov; RO6958688 in NCT3866239 and RO7172508 in NCT03539484).
  • These T-cell bispecific antibodies bind to different epitopes of CEAxCD3 and have different tumor cell killing potency.
  • CEAxCD3 T-cell bispecific antibodies described in WO201705389 are by a factor of 10 to 100 or more potent than RO6958688/cibisatamab (CEA-TCB).
  • T-cell bispecific antibody aims to re-direct T-cells against tumor cells of advanced solid tumors
  • a therapeutic agent re-directing to the tumor cells other immune cells especially macrophages or macrophages and natural killer NK-cells.
  • This invention deals with bispecific antibodies re-directing macrophages and also NK-cells against CEA expressing solid tumors as a monotherapy or in combination with e.g. T-cell bispecific antibodies and/or PD-1/PD-L1 inhibiting antibodies.
  • CAR T-cells may have a simple explanation—the number of CAR T-cells penetrating the solid tumor and distributed in it are just not sufficient. This is certainly different in the majority of haematological malignancies; CAR T-cells can well access the tumor cells, explaining the difference of high efficacy in these malignancies compared to disappointing efficacy in solid tumors.
  • CAR T-cells may be heavily suppressed by the tumor microenvironment (TME) which is mostly strongly immune suppressive.
  • TEE tumor microenvironment
  • Monoclonal antibodies and also bispecific antibodies used in therapy can cause a variety of adverse effects.
  • An important toxicity issue is the cytokine-release syndrome (CRS), which was for example found in therapy with alemtuzumab, muromonab-CD3, rituximab, and CD19 x CD3 bispecific antibody blinatumomab.
  • CRS cytokine-release syndrome
  • anti-CD47 antibodies induce increased amounts of pro-inflammatory cytokines after anti-CD47 antibody mediated phagocytosis (see e.g. US20160144009).
  • Known adverse events of anti-CD47 monoclonal antibodies with wt IgG1 Fc are increased red blood cell RBC phagocytosis/lysis and platelet activation (see e.g. in FIGS. 8 and 10 RBC phagocytosis and platelet activation induced by the anti-CD47 antibody B6H12.2 carrying a wt IgG1 Fc).
  • the present invention provides bispecific antibodies specifically binding to human
  • CEACAM5 and human CD47 designated for the treatment of solid tumors.
  • These bispecific antibodies combine high efficacy with low toxicity, low immunogenicity and favourable pharmacokinetic properties.
  • the bispecific antibodies according to this invention induce their anti-tumor cells effects mainly via optimized ADCP (antibody dependent cellular phagocytosis) and ADCC (antibody dependent cellular cytotoxicity) due to involvement of immune cells especially macrophages and NK-cells.
  • the present invention also provides bispecific antibodies specifically binding to human CEACAM5 and human CD47 designated for the combination treatment with CEAxCD3 T-cell bispecific antibodies like RO6958688, RO7172508 and other CEAxCD3 T-cell bispecific antibodies e.g. as described below and showing strong phagocytosis of tumor cells like MKN-45 in the presence of human macrophages.
  • the invention relates to a bispecific antibody (further named also as “Mab CEAxCD47” or “CEAxCD47 bispecific antibody”) comprising a first binding part specifically binding to human CEACAM5 (further named also as “CEA”) and a second binding part specifically binding to human CD47 (further named also as “CD47”).
  • a bispecific antibody comprising a first binding part specifically binding to human CEACAM5 (further named also as “CEA”) and a second binding part specifically binding to human CD47 (further named also as “CD47”).
  • the invention relates to a bispecific antibody specifically binding to human CEACAM5 and human CD47 characterized in that the Fc region has been glycoengineered to have a reduced number of fucose residues as compared to the same but non-glycoengineered bispecific antibody.
  • the present invention provides a bispecific antibody, characterized in specifically binding to human CEACAM5 and CEACAM6 in the first binding part and to human CD47 in the second binding part.
  • the invention relates to a bispecific antibody CEAxCD47 specifically binding in a balanced manner to human CEACAM5 and human CEACAM6.
  • the bispecific antibody is characterized in binding to human recombinant CEACAM5 and CEACAM6, characterized in that the EC50 values of binding to CEACAM5 and CEACAM6 differing by less than a factor of 3 (balanced binding, binding in balanced manner, see table 5). Binding is measured in a streptavidin/biotin-based ELISA (see example 8f).
  • the present invention provides a bispecific antibody, specifically binding to human CEACAM5 and CEACAM6 in the first binding part and human CD47 in the second binding part, characterized in
  • the invention relates to a bispecific antibody specifically binding to human CEACAM5 and human CD47, the bispecific antibody comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in that the first binding part binds to the Ig-like V-type domain of CEACAM5 of amino acids 35-144.
  • the invention relates to a bispecific antibody specifically binding to human CEACAM5 and human CD47, the bispecific antibody comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in that said bispecific antibody competes with the anti-CEA antibody SM3E, comprising as VK and VH domains VK and VH of sequences SEQ ID NO:100 and 101, for binding to CEACAM5.
  • the invention relates to a bispecific antibody specifically binding to human CEASCAM5 and human CD47, the bispecific antibody comprising a first binding part, specifically binding to human CEASCAM5 and a second binding part, specifically binding to human CD47, characterized in that said bispecific antibody does not compete with anti-CEA antibodies SM3E, MEDI, comprising as VL and VH domains VL and VH of sequences SEQ ID NO:102 and 103, Labetuzumab (Lab), comprising as VK and VH domains VK and VH of sequences SEQ ID NO:110 and 111, SAR, comprising as VK and VH domains VK and VH of sequences SEQ ID NO:104 and 105, T86.66, comprising as VK and VH domains VK and VH of sequences SEQ ID NO:108 and 109, CH1A1A, comprising as VK and VH domains VK and VH of sequences SEQ ID NO:106 and 107 for binding to CEACA
  • the invention relates to a bispecific antibody specifically binding to human CEASCAM5 and human CD47, the bispecific antibody comprising a first binding part, specifically binding to human CEASCAM5 and a second binding part, specifically binding to human CD47, characterized in that the EC50 value of phagocytosis index curve of said bispecific antibody is in the range of 0.1 to 3 times of the E50 value of reference antibody K2AC22 under the same experimental conditions and in the presence or without of 1 mg/ml human IgG. In further embodiments the range is 0.2 to 3.0, 0.3 to 3.0, 0.5 to 2.5 or 1.0 to 2.5.
  • EC50 values of phagocytosis are measured as EC50 values of the phagocytosis index curve (imaging-based phagocytosis assay, see Example 9 and FIG. 12 and Table 3).
  • the invention relates to a bispecific antibody specifically binding to human CEACAM5 and human CD47, the bispecific antibody comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in
  • the bispecific antibody is characterized in comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in that:
  • the bispecific antibody is characterized in comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in that:
  • the bispecific antibody is characterized in comprising in the first binding part as light chain constant domain a human lambda type domain of SEQ ID NO:13
  • the bispecific antibody is characterized in comprising a first binding part, specifically binding to human CEASCAM5 and a second binding part, specifically binding to human CD47, characterized in that:
  • the bispecific antibody is characterized in comprising a first binding part, specifically binding to human CEASCAM5 and a second binding part, specifically binding to human CD47, characterized in that:
  • the bispecific antibody is characterized in being monovalent for the first binding part and monovalent for the second binding part.
  • the constant and variable framework region sequences are human.
  • the bispecific antibody is characterized in that each of the first and second binding part comprises an immunoglobulin heavy chain and an immunoglobulin light chain. In one embodiment the bispecific antibody is characterized in being of human IgG1 type. In one embodiment the bispecific antibody is a full-length antibody.
  • the bispecific antibody according to the invention is characterized in comprising a first binding part specifically binding to CEA, comprising a kappa light chain variable domain and a lambda light chain constant domain and a second binding part specifically binding to CD47, comprising a kappa light chain variable domain and a kappa light chain constant domain ( ⁇ bispecific antibody, ⁇ Body, type 1).
  • the bispecific antibody according to the invention is characterized in comprising a first binding part specific for CEA, comprising a lambda light chain variable domain and a lambda light chain constant domain and a second binding part specific for CD47, comprising a kappa light chain variable domain and a kappa light chain constant domain ( ⁇ bispecific antibody, ⁇ Body, type 2).
  • the bispecific antibody according to the invention is of fully human bispecific IgG (especially IgG1) format and in addition a ⁇ bispecific antibody of type 1 or type 2.
  • the bispecific antibody according to the invention is characterized in being a i bispecific antibody of type 1 or type 2 and comprising a common heavy chain (cHC).
  • cHC common heavy chain
  • the bispecific antibody is characterized in binding to human CD47 with a binding affinity of 100 nM to 600 nM, in one embodiment with a binding affinity of 100 nM to 500 nM.
  • the bispecific antibody is characterized in binding to MKN-45 cells with an EC50 value of 1 to 200 nM. In one embodiment the bispecific antibody is characterized in binding to MKN-45 cells with an EC50 value of 1 to 50 nM. In one embodiment the bispecific antibody is characterized in binding to MKN-45 cells with an EC50 value of 50 to 100 nM. In one embodiment the bispecific antibody is characterized in binding to MKN-45 cells with an EC50 value of 100 to 200 nM.
  • the bispecific antibody according to the invention is characterized in that the maximal achievable phagocytosis index for the phagocytosis of MKN-45 cells in the presence of human macrophages, by said bispecific antibody is not reduced by more than 20% in the presence of 5000 ng/ml soluble CEA compared to the phagocytosis index measured without soluble CEA.
  • the bispecific antibody according to the invention is characterized in that the EC50 for the phagocytosis index curve of MKN-45 cells in the presence of human macrophages, by said bispecific antibody is not shifted by more than a factor 4 towards higher concentrations in the presence of 200 ng/ml soluble CEA compared to the EC50 measured without soluble CEA and/or that the maximum of the phagocytosis index curve is not reduced by 10% or more, 15% or more, or 20% or more by addition of 200 ng/mL sCEA (see e.g. FIG. 20B ).
  • the bispecific antibody according to the invention is characterized in that the EC50 for the binding curve to MKN-45 cells of said bispecific antibody is not shifted by more than a factor 2 towards higher concentrations in the presence of 200 ng/ml soluble CEA compared to the EC50 measured without soluble CEA (see e.g. FIG. 20A ).
  • the bispecific antibody is characterized in that it does not cross-react with human CEACAM1.
  • the bispecific antibody is characterized in binding to human CEACAM6 expressed on recombinant CHO cells CHO-K1 (ATCC® CCL-61TM) with an EC50 value of 1 to 50 nM (CEACAM6 negative CHO cells are transfected with a vector containing cDNA of human CEACAM6 to get CEACAM6 protein expressed).
  • the bispecific antibody according to the invention is characterized in that a monoclonal antibody specifically binding to human CEACAM5 (further named also as MAB CEA), comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 in a concentration of 300 nM do not shift the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • a monoclonal antibody specifically binding to human CEACAM5 (further named also as MAB CEA)
  • MAB CEA monoclonal antibody specifically binding to human CEACAM5
  • MAB CEA monoclonal antibody specifically binding to human CEACAM5
  • the bispecific antibody according to the invention is characterized in that a bispecific antibody specifically binding to human CEASCAM5 and CD3c (further named also as CEA-TCB), comprising as heavy chains the heavy chains of SEQ ID NO:97 and 98 and as light chains the light chains of SEQ ID NO: 96 and 99 in a concentration of 300 nM does not shift the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • the bispecific antibody according to the invention and CEA-TCB are defined as “not competitive” and considered able to bind simultaneously to CEA without significantly interfering in binding to said CEA.
  • the bispecific antibody according to the invention is characterized that a bispecific antibody specifically binding to human CEASCAM5 and CD3c (further named also as CEA-TCB1), comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95 in a concentration of 30 nM does not shift the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • the bispecific antibody according to the invention and CEA-TCB1 are defined as “not competitive” and considered able to bind simultaneously to CEA without significantly interfering in binding to said CEA.
  • the bispecific antibody according to the invention and MAB CEA, CEA-TCB and/or CEA-TCB1 are defined as “not competitive” and considered able to bind simultaneously to CEA without significantly interfering in their binding to said CEA.
  • the bispecific antibody according to the invention is characterized in that a bispecific antibody specifically binding to human CEASCAM5 and CD3c (further named also as CEA-TCB1), comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95, in a concentration of 30 nM does not shift the EC50 of the phagocytosis index curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • a bispecific antibody specifically binding to human CEASCAM5 and CD3c (further named also as CEA-TCB1), comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95, in a concentration of 30 nM does not shift the EC50 of the phagocytosis index curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • the bispecific antibody according to the invention and CEA-TCB1 are defined as “not competitive” and considered able to bind simultaneously to CEA without significantly interfering in their binding to said CEA, and can therefore develop its effect on phagocytosis (CEAxCD47) undisturbed and also its effect on T-cell activation (CEAxTCB1) undisturbed, even if therapeutic levels of both drugs are simultaneously present in the tumor tissue (see FIG. 18 ).
  • the bispecific antibody according to the invention is characterized that a bispecific antibody specifically binding to human CEACAM5 and CD3c (further named also as CEA-TCB), comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 96 to 99 in a concentration of 300 nM does not shift the EC50 of the phagocytosis index curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • the bispecific antibody according to the invention and CEA-TCB are defined as “not competitive” and considered able to bind simultaneously to CEA without significantly interfering in their binding to said CEA and can therefore develop its effect on phagocytosis (CEAxCD47) undisturbed and also its effect on T-cell activation (CEA-TCB) undisturbed, even if therapeutic levels of both drugs are simultaneously present in the tumor tissue (see FIG. 18 ).
  • CEAxCD47 phagocytosis
  • CEA-TCB T-cell activation
  • sequences of SEQ ID NO 88 to 99 are according to US20140242079 respectively WO2017055389.
  • CEAxCD47 bispecific antibodies of the invention combined with CEAxCD3 bispecific antibodies like CEA-TCB and CEA-TCB1 show at least additive or even synergistic % killing of tumor cells in an assay containing e.g. MKN-45 tumor cells and human macrophages and T-cells derived from the same volunteer human donor (see FIGS. 19A and B).
  • the bispecific antibody is characterized in comprising a common heavy chain (cHC) as heavy chain of the first binding part and as heavy chain of the second binding part.
  • cHC common heavy chain
  • the bispecific antibody is characterized in that said common heavy chain of each binding part comprises as CDRs CDRH1 of SEQ ID NO:1, CDRH2 of SEQ ID NO:2 and CDRH3 of SEQ ID NO:3 or a CDRH1 of SEQ ID NO:25, CDRH2 of SEQ ID NO:26 and CDRH3 of SEQ ID NO:27.
  • the bispecific antibody is characterized in that said common heavy chain of each binding part comprises as common variable heavy domain (cVH) SEQ ID NO:4.
  • the bispecific antibody according to the invention is characterized in comprising a common heavy chain (cHC) selected of the group consisting of SEQ ID NO:5, SEQ ID NO:23, and SEQ ID NO:24.
  • cHC common heavy chain
  • the common heavy chain of SEQ ID NO:5 is encoded by the nucleic acid sequence shown in SEQ ID NO:6.
  • the bispecific antibody according to the invention is characterized in comprising as second binding part specific for CD47, a common heavy chain comprising as CDRs CDRH1 of SEQ ID NO:1, CDRH2 of SEQ ID NO:2 and CDRH3 of SEQ ID NO:3 and a light chain (LC) comprising as CDRs CDRL1 of SEQ ID NO:7, CDRL2 of Ala Ala Ser, included in SEQ ID NO:8, and CDRL3 of SEQ ID NO:9 , or a common heavy chain comprising as CDRs CDRH1 of SEQ ID NO:25, CDRH2 of SEQ ID NO:26 and CDRH3 of SEQ ID NO:27 and a light chain (LC) comprising as CDRs CDRL1 of SEQ ID NO:28, CDRL2 of SEQ ID NO:29, and CDRL3 of SEQ ID NO:30.
  • LC light chain
  • the bispecific antibody according to the invention is characterized in comprising as second binding part a heavy chain comprising as variable heavy domain (cVH) SEQ ID NO:4 and a variable light domain (VL) of SEQ ID NO:10.
  • the bispecific antibody according to the invention is characterized in comprising as second binding part a heavy chain (cHC) comprising of SEQ ID NO:5 and a light chain (CL) of SEQ ID NO:11. In one embodiment the bispecific antibody according to the invention is characterized in comprising as second binding part a heavy chain (cHC) comprising of SEQ ID NO:23 and a light chain (CL) of SEQ ID NO:11. In one embodiment the bispecific antibody according to the invention is characterized in comprising as second binding part a heavy chain (cHC) comprising of SEQ ID NO:24 and a light chain (CL) of SEQ ID NO:11. In one embodiment the light chain (LC) of SEQ ID NO:11 is encoded by the nucleic acid sequence shown in SEQ ID NO:12.
  • the bispecific antibody is characterized in specifically binding to CEA and comprising a light chain constant domain of SEQ ID NO:13.
  • the bispecific antibody according to the invention is characterized in inhibiting the interaction between CD47 on MKN-45 cells with an IC50 of 0.1 to 10 nM.
  • SIRP ⁇ SIRP ⁇ , CD172a; UniProtKB P78324
  • IC50 0.1 to 10 nM.
  • the bispecific antibody of the invention is characterized in a concentration dependent phagocytosis (ADCP) of CEA expressing tumor cell lines like MKN-45 cells by human macrophages at an EC50 of the bispecific antibody below 10 nM.
  • ADCP is measured according to the invention as phagocytosis index (EC50 or maximum) by imaging, usually with an E:T ratio of 1:3 (human macrophages;target cells (tumor cells); see e.g. FIGS. 12, 15, and 16 ). Results in FIG. 3B have been obtained with E:T of 1:1. Details of the assay are described in example 9.2.
  • ADCP phagocytosis
  • CEA phagocytosis
  • Flow Cytometry with an E:T ratio of e.g. 3:1 (human macrophages;target cells (tumor cells); see e.g. FIG. 3A ). Details of the assay are described in example 9 (1. Flow cytometry based ADCP assay).
  • the bispecific antibody is characterized in specifically binding to CEASCAM5 but is not competing for binding to CEASCAM5 on tumor cells like MKN-45 with MAB CEA, CEA-TCB and/or CEA-TCB1.
  • the bispecific antibody according to the invention is characterized in that the EC50 value for the binding to MKN-45 cells (EC50 between 1 and 200 nM) is increased by less than a factor of three by addition of MAB CEA or CEA-TCB at a concentration of 300 nM respectively by addition of CEA-TCB1 at a concentration of 30 nM (no competition).
  • the CEAxCD47 antibodies of the invention show a 100 or more times higher EC50 for RBC phagocytosis compared to the EC50 measured in the same assay (Example 15) with B6H12.2.
  • the CEAxCD47 antibodies of the invention do not show significant platelet activation in concentrations up to 200 ⁇ g/mL (see Example 15 and results mentioned in Example 15 for CEAxCD47 bispecific antibodies K2AC5 and K2AC22).
  • the present invention relates to a bispecific antibody according to the invention that has been glycoengineered to have an Fc region with modified oligosaccharides. It was surprisingly found, that such a glycoengineered bispecific antibody according to the invention is characterized in an at least 3 times lower EC50 value for the phagocytosis index curve measured by the imaging based assay) as the same not glycoengineered (parent) bispecific antibody if measured under the same experimental conditions. In one embodiment EC50 for the phagocytosis index is 5 to 10 times lower, or 10 to 30 times lower). In one embodiment, the Fc region has been modified to have a reduced number of fucose residues as compared to the same but non-glycoengineered bispecific antibody.
  • the Fc region has an increased proportion of bisected oligosaccharides as compared to the non-glycoengineered bispecific antibody.
  • the bisected oligosaccharides are predominantly bisected complex.
  • the glycoengineered antigen binding molecules of the invention have an increased proportion of bisected, nonfucosylated oligosaccharides in the Fc region of said bispecific antibody as compared to the non-glycoengineered bispecific antibody.
  • the bispecific antibodies of the invention may have an increased ratio of GIcNAc residues to fucose residues in the Fc region compared to the non-glycoengineered bispecific antibody.
  • the bisected, nonfucosylated oligosaccharides are predominantly in hybrid form.
  • the bisected, nonfucosylated oligosaccharides are predominantly complex type.
  • the bispecific antibody according to the invention is characterized in that 50% to 100% of the N-linked oligosaccharides in the Fc region are nonfucosylated.
  • the bispecific antibody is characterized in that 50% to 100% of the N-linked oligosaccharides in the Fc region are bisected.
  • the bispecific antibody is characterized that 80% to 100% of the N-linked oligosaccharides in the Fc region are bisected and nonfucosylated.
  • the bispecific antibody is characterized in that concentration/ADCC curve (decrease of EC50 or increase of maximum of ADCC (see FIGS. 13 and 14 ) induced by said glycoengineered antibody is increased by at least a factor of 1.2 compared to the ADCC induced by the same but non-glycoengineered bispecific antibody. In one embodiment ADCC is increased by a factor of 1.2 to 2.0.
  • the bispecific antibody is characterized in an at least 3 times lower EC50 value for the phagocytosis index curve measured by the imaging based assay as compared to the same but not glycoengineered (parent) bispecific antibody if measured under the same experimental conditions.
  • EC50 for the phagocytosis index is 5 to 10 times lower, or 10 to 30 times lower
  • the bispecific antibody is characterized in that the maximal phagocytosis index induced by said glycoengineered antibody and measured by flow cytometry is increased by at least a factor of 1.2 compared to the maximal phagocytosis index induced by the same but non-glycoengineered bispecific antibody. In one embodiment maximal phagocytosis index is increased by a factor of 1.2 to 2.0.
  • the bispecific antibody is characterized in that the maximal phagocytosis index induced by said glycoengineered antibody and measured by imaging is increased by at least a factor of 1.2 compared to maximal phagocytosis index induced by the same but non-glycoengineered bispecific antibody. In one embodiment maximal phagocytosis index is increased by a factor of 1.2 to 2.0.
  • the bispecific antibody according to the invention is characterized in comprising one, two or three amino acid substitutions in the Fc region (“Fc amino acid substitution”) selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E, S239D and G236A, and of triple-substitution S329D and I332E and G236A.
  • Fc amino acid substitution selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E, S239D and G236A, and of triple-substitution S329D and I332E and G236A.
  • the bispecific antibody according to the invention is characterized in comprising one, two or three amino acid substitutions in the Fc region selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E, S239D and G236A, and triple-substitution S329D and I332E and G236A and a Fc region which has been glycoengineered to have a reduced number of fucose residues as compared to the same but non-glycoengineered bispecific antibody.
  • the bispecific antibody comprising said substitutions in the Fc region is characterized in that concentration/ADCC curve (decrease of EC50 or increase of maximum of ADCC) induced by said amino acid substituted antibody is increased by at least a factor of 1.2 compared to the ADCC induced by said antibody comprising none of said amino acid substitutions in the Fc region. In one embodiment ADCC is increased by a factor of 1.2 to 2.0.
  • the bispecific antibody comprising said substitutions in the Fc region is characterized in an at least 3 times lower EC50 value for the phagocytosis index curve measured by the imaging based assay as compared to the same (parent) bispecific antibody comprising none of said amino acid substitutions in Fc region, if measured under the same experimental conditions.
  • EC50 for the phagocytosis index is 5 to 10 times lower, or 10 to 30 times lower
  • the bispecific antibody comprising said substitutions in the Fc region is characterized in that flow cytometry determined maximal phagocytosis (ADCP) induced by said amino acid substituted antibody is increased by at least a factor of 1.2 compared to the ADCP induced by said antibody comprising none of said amino acid substitutions in the Fc region. In one embodiment ADCP is increased by a factor of 1.2 to 2.0. In one embodiment the bispecific antibody comprising said substitutions in the Fc region is characterized in that by imaging determined maximal phagocytosis index induced by said amino acid substituted antibody is increased by at least a factor of 1.2 compared to the ADCP induced by said antibody comprising none of said amino acid substitutions in the Fc region. In one embodiment ADCP is increased by a factor of 1.2 to 2.0.
  • the bispecific antibody according to the invention is characterized in that 50% to 100%, 60% to 100%, 70% to 100% or 80% to 100% of the N-linked oligosaccharides in the Fc region are non-fucosylated. In one embodiment, the bispecific antibody according to the invention is characterized in 50% to 100%, 60% to 100%, 70% to 100% or 80% to 100% of the N-linked oligosaccharides in the Fc region are bisected. In one embodiment, the bispecific antibody according to the invention is characterized in that 50% to 100%, 60% to 100%, 70% to 100% or 80% to 100% of the N-linked oligosaccharides in the Fc region are bisected, nonfucosylated.
  • the glycoengineered bispecific antibody comprises increased effector functions compared to the non-glycoengineered bispecific antibody comprising as common heavy chain SEQ ID NO:5 (parent bispecific antibody, produced in a CHO K1 cell line CHO-K1 (ATCC® CCL-61TM at standard conditions as defined below).
  • the bispecific antibody according to the invention is characterized in that said glycoengineered bispecific antibody comprises one or more increased effector functions such as those from the group consisting of increased binding affinity to Fc ⁇ Rs, increased binding of macrophages (increased antibody dependent cellular phagocytosis; ADCP), increased binding of NK cells (increased antibody-mediated cellular cytotoxicity; ADCC), and increased binding to monocytes.
  • said glycoengineered bispecific antibody comprises one or more increased effector functions such as those from the group consisting of increased binding affinity to Fc ⁇ Rs, increased binding of macrophages (increased antibody dependent cellular phagocytosis; ADCP), increased binding of NK cells (increased antibody-mediated cellular cytotoxicity; ADCC), and increased binding to monocytes.
  • the concentration/phagocytosis index curve measured for the anti-CD47 monoclonal antibody hu5F9-G4 (tested in clinical trials since 2014, see e.g. clintrial.gov) is strongly reduced by the addition of huIgG added in physiological concentrations of 1 mg/mL to the assay (increase of EC50 and decrease of the maximum of the phagocytosis curve measured in imaging based assay, see e.g. FIG. 17 ).
  • CEAxCD47 antibodies of the invention show only a small shift below a factor of 3 of EC50 and no significant decrease of the maximum of the concentration/phagocytosis index curve if human IgG is added (see Table 4).
  • CEAxCD47 antibodies of the invention are characterized in that addition of 1 mg /mL of hu IgG to the imaging based phagocytosis assay causes a less than a factor of 0.9 reduction of the maximum of the concentration/phagocytosis index curve and/or a less than a factor of 3 shift of the EC50 towards higher concentrations (see Table 4)
  • a further embodiment of the invention is an isolated polynucleotide characterized in encoding a bispecific antibody according to the invention.
  • a further embodiment of the invention is an expression vector comprising the polynucleotide according to the invention.
  • a further embodiment of the invention is a host cell comprising the expression vector according to the invention.
  • a further embodiment of the invention is a method for the production of a bispecific antibody according to the invention, characterized in comprising:
  • the invention is characterized in comprising a method for producing a glycoengineered bispecific antibody according to the invention in a host cell, said method comprising:
  • the invention is characterized in comprising a method for producing a glycoengineered bispecific antibody in a host cell, said method comprising:
  • the invention is characterized in comprising a method for producing a Fc substituted bispecific antibody according to the invention in a host cell, said method comprising:
  • a further embodiment of the invention is a method of inducing cell lysis of a tumor cell comprising contacting the tumor cell with a bispecific antibody according to the invention.
  • the tumor cell is a human tumor cell, preferably in a patient.
  • a further embodiment of the invention is a method according to the invention, characterized in that the tumor cell is a colorectal cancer cell, NSCLC (non-small cell lung cancer) cell, gastric cancer cell, pancreatic cancer cell, breast cancer cell, or another tumor cell expressing CEA.
  • the tumor cell is a colorectal cancer cell, NSCLC (non-small cell lung cancer) cell, gastric cancer cell, pancreatic cancer cell, breast cancer cell, or another tumor cell expressing CEA.
  • a further embodiment of the invention is a method of treating a subject having a cancer that expresses CEA, the method comprising administering to the subject a therapeutically effective amount of a bispecific antibody according to the invention.
  • a further embodiment of the invention is a method of increasing survival time in a subject having a cancer that expresses CEA, said method comprising administering to said subject a therapeutically effective amount of a bispecific antibody according to the invention.
  • a further embodiment of the invention is a method according to the invention, characterized in that the cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer or breast.
  • NSCLC non-small cell lung cancer
  • gastric cancer pancreatic cancer or breast.
  • a further embodiment of the invention is a method according to the invention, characterized in that a bispecific antibody according to the invention is administered in combination with chemotherapy or radiation therapy to a human subject.
  • a further embodiment of the invention is a method of treating a subject having a cancer that expresses CEA, the method comprising administering to the subject a therapeutically effective amount of a bispecific antibody according to the invention, characterized in that the EC50 value of phagocytosis of said bispecific antibody is in the range of 0.1 to 3 times of the E50 value of reference antibody K2AC22 under the same experimental conditions and in the presence and/or without of 1 mg/ml human IgG. In further embodiments the range is 0.2 to 3.0, 0.3 to 3.0, 0.5 to 2.5 or 1.0 to 2.5.
  • the bispecific antibody is characterized in binding to human CD47 with a binding affinity of 100 nM to 600 nM, in one embodiment with a binding affinity of 100 nM to 500 nM.
  • a further embodiment of the invention is the use of a bispecific antibody according to the invention in a method of treating a subject having a cancer that expresses CEA, the method comprising administering to the subject a therapeutically effective amount of a bispecific antibody according to the invention, characterized in that the EC50 value of phagocytosis of said bispecific antibody is in the range of 0.1 to 3 times of the E50 value of reference antibody K2AC22 under the same experimental conditions and in the presence and/or without of 1 mg/ml human IgG. In further embodiments the range is 0.2 to 3.0, 0.3 to 3.0, 0.5 to 2.5 or 1.0 to 2.5.
  • the bispecific antibody is characterized in binding to human CD47 with a binding affinity of 100 nM to 600 nM, in one embodiment with a binding affinity of 100 nM to 500 nM.
  • ADCC and ADCP/phagocytosis index values of antibodies according to the invention are not or only to a low extend affected by human IgG in a concentration of 1 mg/ml (1 mg/ml or even higher human IgG is present in most patients), whereas for an anti-CD47 antibody of the state of the art (hu5F9-G4), ADCC and ADCP values are strongly reduced in the presence of 1 mg/mL human IgG.
  • a further embodiment of the invention is the use of the bispecific antibody according to the invention in the manufacture of a medicament for treating a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is the use of the bispecific antibody according to the invention in the manufacture of a medicament according to the invention, characterized in that the cancer is selected from the group consisting of colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer pancreatic cancer
  • breast cancer breast cancer
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination with a second bispecific antibody comprising a third binding part specifically binding to human CEASCAM5, and a fourth binding part specifically binding to human CD3 ⁇ in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination with a second bispecific antibody comprising a third binding part specifically binding to human CEASCAM5 and a fourth binding part specifically binding to an epitope of human CD3 ⁇ , said epitope comprising the amino acid sequence of SEQ ID NO:22 in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination with CEA-TCB and/or CEA/TCB1 in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination with a second bispecific antibody comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 and a fourth binding part specifically binding to an epitope of human CD3 ⁇ , said epitope comprising the amino acid sequence of SEQ ID NO:22 in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, characterized in not competing with said second bispecific antibody for use in simultaneous, separate, or sequential combination with said second bispecific antibody in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, characterized in not competing with CEA-TCB or CEA-TCB1 for use in simultaneous, separate, or sequential combination with said CEA-TCB or CEA-TCB1 in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, characterized in competing with CEA-TCB or CEA-TCB1 for use in simultaneous, separate, or sequential combination with said CEA-TCB or CEA-TCB1 in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination with a second bispecific antibody comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:88 and a light chain variable region of SEQ ID NO:89 and a fourth binding part specifically binding to human CD3c, comprising a heavy chain variable region of SEQ ID NO:90 and a light chain variable region of SEQ ID NO:91.
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use according to the invention, characterized in that the bispecific antibody according to the invention and the second bispecific antibody are administered to said subject alternately in 6 to 15 day intervals.
  • a further embodiment of the invention is a bispecific antibody according to the invention, for use according to the invention, characterized in that the bispecific antibody according to the invention and the second bispecific antibody are administered to said subject simultaneously in 6 to 15 day intervals.
  • a further embodiment of the invention is a first bispecific antibody according to the invention, comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, for use in simultaneous, separate, or sequential combination in the treatment of a subject having a cancer that expresses CEA, with a second bispecific antibody, comprising a third binding part specifically binding to human CEASCAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 and a fourth binding part specifically binding to an epitope of human CD3 ⁇ , comprising the amino acid sequence of SEQ ID NO:22, whereby said second bispecific antibody in a concentration of 300 nM does not shift the EC50 value of the phagocytosis index curve to MKN-45 cells of the bispecific antibody according to the invention by more than a factor of 3, in one embodiment towards higher concentrations.
  • a further embodiment of the invention is a first bispecific antibody according to the invention, comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, for use in simultaneous, separate, or sequential combination in the treatment of a subject having a cancer that expresses CEA, with a second bispecific antibody comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:88 and a light chain variable region of SEQ ID NO:89 and a fourth binding part specifically binding to human CD3c, comprising a heavy chain variable region of SEQ ID NO:90 and a light chain variable region of SEQ ID NO:91, whereby said second bispecific antibody in a concentration of 30 nM does not shift the EC50 of the binding curve to MKN-45 cells of the bispecific antibody according to the invention by more than a factor of 3, in one embodiment towards higher concentrations.
  • a further embodiment of the invention is a first bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination in the treatment of a subject having a cancer that expresses CEA, with CEA-TCB or CEA-TCB1, whereby said CEA-TCB in a concentration of 300 nM or CEA-TCB lin a concentration of 30 nM do not shift the EC50 of the binding curve to MKN-45 cells of the bispecific antibody according to the invention by more than a factor of 3, in one embodiment towards higher concentrations.
  • a further embodiment of the invention is a first bispecific antibody according to the invention, comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47 according to the invention, for use according to the invention, characterized in that said cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer.
  • NSCLC non-small cell lung cancer
  • a further embodiment of the invention is a composition comprising a bispecific antibody according to the invention, characterized in not competing with said second bispecific antibody as defined above for use in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a composition comprising a bispecific antibody according to the invention, characterized in not competing with a second bispecific antibody comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 and a fourth binding part specifically binding to an epitope of human CD3 ⁇ , comprising the amino acid sequence of SEQ ID NO:22, for use in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a composition comprising a bispecific antibody according to the invention, characterized in not competing with a second bispecific antibody comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:88 and a light chain variable region of SEQ ID NO:89 and a fourth binding part specifically binding to human CD3c, comprising a heavy chain variable region of SEQ ID NO:90 and a light chain variable region of SEQ ID NO:91, for use in the treatment of a subject having a cancer that expresses CEA.
  • a further embodiment of the invention is a composition comprising a bispecific antibody according to the invention, characterized in not competing with CEA-TCB and/or CEA-TCB1.
  • a further embodiment of the invention is a method for the treatment of a human patient diagnosed with a tumor (cancer), especially a solid tumor, especially a solid cancer that expresses CEA, especially colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer, comprising administering an effective amount of an bispecific antibody according to the invention and a second bispecific antibody as described above, against CEA and CD3 (in one embodiment CEA-TCB or CEA-TCB1), to the human patient, the method comprising subsequently:
  • administering to the patient a dose of 0.1 to 10 mg/kg, in a further embodiment of 0.5 to 10 mg/kg, in a further embodiment of 1 to 2 mg/kg of said second anti CEAxCD3 antibody, e.g. weekly over 4 to 12 weeks.
  • This “alternating” method is applied if the antibody of the invention and the second bispecific antibody are competitive.
  • the two bispecific antibodies can also be administered in a manner (“simultaneous manner”) that the patient experiences therapeutically effective plasma and tissue concentrations of both bispecific antibodies in parallel, e.g.
  • a dose of 0.1 to 10 mg/kg in a further embodiment of 0.5 to 10 mg/kg, in a further embodiment of 1 to 2 mg/kg of the CEA x CD3 bispecific antibody and 1 to 20 mg/kg of the CEA x CD47 bispecific antibody of this invention, followed by one or more of these combined administrations at a frequency of q1w or q2w or q3w or optionally q4w.
  • Q1w means administration once a week; q2w means administration every two weeks etc.
  • a further embodiment of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody according to the invention and a pharmaceutically acceptable recipient or carrier.
  • a further preferred embodiment of the invention is a pharmaceutical composition comprising an antibody according to the invention for use as a medicament.
  • a further preferred embodiment of the invention is a pharmaceutical composition comprising an antibody according to the invention for use as a medicament in the treatment of solid tumor disorders.
  • a further preferred embodiment of the invention is a pharmaceutical composition comprising an antibody according to the invention for use as a medicament in the treatment of colorectal cancer, NSCLC (non-small cell lung cancer), gastric cancer, pancreatic cancer or breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer gastric cancer
  • pancreatic cancer breast cancer
  • a further embodiment of the invention is a composition comprising a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination in the treatment of a subject having a cancer that expresses CEA, with a second bispecific antibody, comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 and a fourth binding part specifically binding to an epitope of human CD3c, comprising the amino acid sequence of SEQ ID NO:22, whereby said second bispecific antibody in a concentration of 300 nM does not shift the EC50 of the binding curve to MKN-45 cells of the bispecific antibody according to the invention by more than a factor of 3, in one embodiment towards higher concentrations.
  • a further embodiment of the invention is a composition comprising a bispecific antibody according to the invention, for use in simultaneous, separate, or sequential combination in the treatment of a subject having a cancer that expresses CEA, with a second bispecific antibody comprising a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:88 and a light chain variable region of SEQ ID NO:89 and a fourth binding part specifically binding to human CD3c, comprising a heavy chain variable region of SEQ ID NO:90 and a light chain variable region of SEQ ID NO:91, whereby said second bispecific antibody in a concentration of 30 nM does not shift the EC50 of the binding curve to MKN-45 cells of the bispecific antibody according to the invention by more than a factor of 3, towards higher concentrations.
  • a further embodiment of the invention is a composition according to the invention, characterized in that the cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer, or breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer gastric cancer
  • pancreatic cancer pancreatic cancer
  • breast cancer breast cancer
  • a further embodiment of the invention is the use of an antibody according to the invention for the manufacture of a pharmaceutical composition.
  • a further embodiment of the invention is the use of an antibody according to the invention and a pharmaceutically acceptable recipient or carrier for the manufacture of a pharmaceutical composition.
  • a further embodiment of the invention is the use of an antibody according to the invention for the manufacture of a medicament in the treatment of solid tumor disorders.
  • a further embodiment of the invention is the use of an antibody according to the invention in the treatment of colorectal cancer, NSCLC (non-small cell lung cancer), gastric cancer, pancreatic cancer or breast cancer.
  • Another aspect of the invention provides a method of inducing cell lysis of a tumor cell comprising contacting the tumor cell with the bispecific antibody of any of above described embodiments.
  • the tumor cell is a colorectal cancer cell, NSCLC (non-small cell lung cancer), gastric cancer cell, pancreatic cancer cell or breast cancer cell.
  • the cell lysis is induced by antibody dependent cellular phagocytosis and/or antibody dependent cellular cytotoxicity of the bispecific antibody.
  • Another aspect of the invention provides a method of treating a subject having a cancer that abnormally expresses CEA, the method comprising administering to the subject a therapeutically effective amount of the bispecific antibody of any of above described embodiments.
  • Another aspect of the invention provides a method of treating a subject having a cancer that abnormally expresses CEA, the method comprising administering to the subject a therapeutically effective amount of the bispecific antibody of any of above described embodiments in combination with a bispecific antibody binding to human CEA and human CD3.
  • the CEAxCD47 antibody and the CEAxCD3 antibody are competing they will compete for the CEA receptors on the surface of the tumor cell and the receptor occupancy and efficacy for each combination partner depends on their binding affinity and their plasma concentrations and is therefore difficult to predict and also variable over time if the concentrations of the two drugs have a different elimination half-life respectively clearance from the body. Therefore, competing CEAxCD3 and CEAxCD47 bispecific antibodies should be given sequentially (alternating).
  • CEAxCD3 and CEAxCD47 bispecific antibodies are not or only minimally competing they can be not only given sequentially but also in parallel (simultaneously) which may well be an advantage because tumor cell killing via engagement of T-cells by the CEAxCD3 bispecific antibody and at the same time via engagement of macrophages by the CEAxCD47 bispecific antibody is additive or may be even synergistic, which means efficacy is increased if both drugs are given in parallel.
  • Another aspect of the invention provides a method of increasing progression free survival and/or overall survival time in a subject having a cancer that abnormally expresses CEA, said method comprising administering to said subject a therapeutically effective amount of the bispecific antibody of any of above described embodiments.
  • the cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer or breast cancer or another cancer expressing CEA.
  • the bispecific antibody is administered in combination with chemotherapy or radiation therapy.
  • the subject is a patient suffering from colorectal cancer or lung cancer or gastric cancer or pancreatic cancer or breast cancer or another cancer expressing CEA.
  • Another aspect of the invention provides a method of treating a subject having a cancer that abnormally expresses CEA, the method comprising administering to the subject a therapeutically effective amount of the bispecific antibody of any of above described embodiments in combination with a bispecific antibody against human CEA and human CD3 epsilon.
  • Another aspect of the invention provides a method of increasing progression free survival time and/or overall survival time in a subject having a cancer that abnormally expresses CEA, said method comprising administering to said subject a therapeutically effective amount of the bispecific antibody of any of above described embodiments.
  • the cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer or breast cancer.
  • the bispecific antibody is administered in combination with chemotherapy or radiation therapy.
  • the subject is a cancer patient with colorectal cancer or lung cancer or gastric cancer or pancreatic cancer or breast cancer or another CEA expressing cancer.
  • the cancer is selected from the group consisting of colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer gastric cancer
  • pancreatic cancer breast cancer
  • FIG. 1 shows the general molecular format of the CEAxCD47 bispecific antibodies of this invention; fully human IgG1 structure undistinguishable from IgG monoclonal antibody and with no aa bridges to minimize immunogenicity and anti-drug antibody ADA formation; common heavy chain (see sequence list); kappa light chain (CL and VL) in the CD47 binding part (see sequence list); lambda CL in the CEA binding part (see sequence list) and lambda or kappa VL in the CEA binding part (A); Glycoengineering (low fucose) of the Fc or aa mutation(s) or both to increase ADCP, and ADCC (B).
  • FIG. 2 shows the binding of a TAAxCD47 bispecific antibody to tumor cells carrying on the surface the TAA as well as CD47. Binding of the TAAxCD47 with an EC50 between 1 and 50 nM (solid line, see also FIG. 11 for binding curves of various CEAxCD47 antibodies of the invention); the broken line shows exemplary and schematic a potential shift of the binding curve by e.g. soluble CEA, for concrete data see FIG. 20A showing e.g. the influence of 200 ng/mL soluble CEA on the binding curve to MKN-45 cells of CEAxCD47 bispecific antibodies of the invention like K2AC5 and 22.
  • soluble CEA for concrete data see FIG. 20A showing e.g. the influence of 200 ng/mL soluble CEA on the binding curve to MKN-45 cells of CEAxCD47 bispecific antibodies of the invention like K2AC5 and 22.
  • FIG. 3A shows the concentration dependent increase of phagocytosis (assessed by flow cytometry and expressed as % of phagocytosis) of human pancreatic cancer cell line HPAC (expresses CEA and other TAAs like mesothelin MSLN) with a TAAxCD47 bispecific antibody carrying the CD47 binding arm of the invention, the corresponding CD47 and TAA monovalent antibody as well as the high-affinity anti-TAA (MSLN) monoclonal antibody, Amatuximab. All antibodies bear wild-type IgG1 Fc portions. The bispecific antibody shows the highest phagocytosis.
  • FIG. 3B shows the concentration dependent increase of phagocytosis (assessed with the CellInsight assay and expressed as phagocytosis index) induced by various antibodies; curve 1: control hIgG1 not binding to TAA or CD47; curve 2: TAAxCD47 bispecific antibody with wild type hIgG1 Fc; curve 3: anti CD47 antibody B6H12.2 with wild type hIgG1; curve 4: TAAxCD47 bispecific antibody with DEA aa substitutions (S329D, I332E and G236A) in the hIgG1 Fc part. The strongest phagocytosis was achieved with TAAxCD47 with DEA mutated Fc.
  • FIG. 4 shows an example of ADCC dose response curves (assessed with the Cr51 + assay and expressed as % specific killing) for various TAA x CD47 bispecific antibodies (TAA is mesothelin MSLN), all carrying the same CD47 binding arm of this invention, using lung cancer NCI-H226 cancer cells as target cells (hu volunteer PBMC to tumor cells 50:1). All bispecific antibodies show stronger ADCC than the high-affinity anti-TAA monoclonal antibody, Amatuximab, an anti-MSLN mAb.
  • TAA is mesothelin MSLN
  • FIG. 5 shows ADCC dose response curves (assessed using the Cr51 + assay and expressed as % specific killing) for a TAAxCD47 bispecific antibody with wildtype Fc, the corresponding TAAxCD47 bispecific antibody with Fc carrying DEA mutations, a high-affinity anti-TAA monoclonal antibody (TAA in this figure is not CEA), and a human IgG1 control antibody; using lung cancer NCI-H226 cancer cells as target cells (effector to tumor cells 50:1). The strongest ADCC was observed with the bispecific antibody carrying the DEA mutations.
  • FIG. 6 shows concentration dependent blockade of soluble SIRPalpha binding to CD47 expressed on MKN-45 cells co-expressing CEA (the TAA) by a TAAxCD47 bispecific antibody (solid line) and the corresponding anti-CD47 monovalent antibody (dashed line). Higher blocking potency of the bispecific antibody is due to more potent binding to target cells and TAA co-engagement-dependent blockade of CD47.
  • FIG. 7A shows the release of cytokines IFN ⁇ and TNF ⁇ in whole human blood incubated with 200 ⁇ g/mL of the following antibodies: Erbitux as negative control, anti-CD52 antibody Campath as positive control, and three bispecific antibodies having identical antigen binding regions but different Fc regions (from left to right): wildtype human IgG1 Fc, IgG1 Fc with DEA mutations (S329D and I332E), IgG1 Fc with DE mutations.
  • biAb CD4710 and CD4710 refers to the same CD47xTAA bispecific antibody. IFN ⁇ and TNF ⁇ release observed with the bispecific antibody carrying DEA mutations is no higher than with the bispecific antibody with wildtype Fc.
  • FIG. 7B shows the release of cytokines IL-6 and IL-8 in whole human blood incubated with 200 ⁇ g/mL of the following antibodies: Erbitux as negative control, anti-CD52 antibody Campath as positive control, and three bispecific antibodies having identical antigen binding regions but different Fc regions (from left to right): wildtype human IgG1 Fc, IgG1 Fc with DEA mutations, IgG1 Fc with DE mutations.
  • biAb CD4710 and CD4710 refers to the same CD47xTAA bispecific antibody.
  • IL-6 and IL-8 release observed with the bispecific antibodies carrying DE or DEA mutations is no higher than with the bispecific antibody with wildtype Fc.
  • FIG. 8 shows concentration dependent phagocytosis of red blood cells (RBC), a major “antigen sink” for monoclonal anti-CD47 antibodies (every RBC expressing between 20, 000 and 25,000 CD47 molecules on the cell surface) induced by various antibodies: the anti-CD47 huIgG1 antibody B6H12.12 with wildtype Fc, a CD47xTAA bispecific antibody with wildtype Fc and the same CD47xTAA bispecific antibody with Fc carrying DEA mutations.
  • B6H12 shows RBC phagocytosis at concentrations of 10 to 100 ng/ml (approx.
  • TAAxCD47 bispecific antibody with wildtype huIgG1 Fc shows no RBC phagocytosis at concentrations up to 200 000 ng/ml (approx. 1350 nM);
  • the same bispecific antibody but with an Fc portion carrying DEA mutations shows increased phagocytosis of RBC as compared to wild-type Fc, at concentrations above 1000 ng/ml, (approx 7nM), which is still 2-2.5 logs higher than with the anti CD47 huIgG1 antibody B6H12.
  • FIG. 9 shows red blood cell counts and platelet counts in Non Human Primate (NHP; cynomolgus monkeys) that received four weekly iv infusions of either control hIgG1 antibody or TAAxCD47 bispecific antibody (30mg/kg for the first two weeks and 100 mg/kg for the last two weeks). No significant decrease in hematology counts was observed with the TAAxCD47 bispecific antibody in spite of high exposure (TAAxCD47 bispecific antibody plasma concentration at the end of the second dosing, at 30 mg/kg, was approx. 500 nM).
  • NHS Non Human Primate
  • FIG. 10 shows in vitro platelet activation (assessed by flow cytometry and expressed as %CD62P expression), induced by incubation of human whole blood with antibodies as indicated at different concentrations (from 0 to 200 ⁇ g/mL). Contrary to B6H12-hIgG1 which induces platelet activation at 2 ⁇ g/ml and higher, the TAAxCD47 bispecific antibody with wild type IgG1 Fc doesn't induce platelet activation even at the highest concentration tested (200 ⁇ g/mL).
  • CEAxCD47 bispecific antibodies K2AC5 and K2AC22 were also tested, versions with wtIgG1 Fc as well as afucosylated versions did not show significant platelet activation up to 20 ⁇ g/mL (see example 15).
  • FIGS. 11A and B show concentration dependent binding of CD47xCEA bispecific antibodies of the invention as compared to the corresponding anti-CD47 monovalent antibody. Binding to target cells (MKN-45) expressing CD47 and CEA was assessed by FACS.
  • FIG. 11A shows antibodies classified in bin 1, binding to MKN-45 cells inhibited by SM3E antibody by more than 80%;
  • FIG. 11B shows antibodies classified in bins 2, binding to MKN-45 cells not inhibited by anti-CEA antibodies SM3E, MEDI, T84.66, SAR, Lab, and CH1A1A (by less than 20%).
  • FIG. 12 shows the concentration dependent increase of phagocytosis (assessed by the Celllnsight assay and expressed as phagocytosis index) of MKN-45 cells induced by different CD47xCEA bispecific antibodies at different concentrations (K2AC5, K2AC22, K2AC23, K2AC25, K2AC26, K2AC27, K2AC28 and K2AC29) as compared to the corresponding anti-CD47 monovalent antibody. 1 mg/mL of hIgG (human immunoglobulin) is added in this experiment for each tested antibody.
  • hIgG human immunoglobulin
  • EC50 values established in this experiment are comprised between 0.2 and 20 ⁇ g/ml and the maximal phagocytosis index (at 10 ⁇ g/mL) ranges between 32.5% and 69% (see Table 3 in the Examples Chapter for summary of data for each individual CD47xCEA bispecific antibody tested).
  • FIGS. 13 and 14 show the concentration dependent increase of ADCC (assessed using the LDH release assay and expressed as % specific lysis of MKN45 cancer cells) induced by two selected CD47xCEA bispecific antibodies (K2AC5 and K2AC22) of the invention, and the corresponding CD47 monovalent antibody, either bearing a wild-type human IgG1 Fc part or an afucosylated Fc part.
  • the sequence-identical analogue of the anti-CD47 antibody Hu5F9-G4 (5F9 bearing a human IgG4 Fc portion, described in US20160333093) was run for comparison. The experiments were performed in the absence ( FIG. 13 ) or in the presence ( FIG. 14 ) of 1 mg/ml human IgG.
  • antibody versions with afucosylated Fc induce a higher lysis/killing activity as compared to the corresponding wild-type Fc-bearing versions (5-9 fold lower EC50 for afucosylated K2AC5 and K2AC22 bispecific antibodies as compared to the wt version).
  • FIG. 15 and FIG. 16 show the concentration dependent increase of phagocytosis (assessed with imaging based assay Celllnsight and expressed as phagocytosis index) induced by K2AC5 and K2AC22 CD47xCEA bispecific antibodies bearing either a wild-type human IgG1 Fc portion or an afucosylated Fc portion.
  • the corresponding CD47 monovalent antibody (with wild-type hIgG1 Fc) and the sequence-identical analogue of the anti-CD47 antibody Hu5F9-G4 (5F9) were run for comparison. The experiments were performed either in the absence ( FIG. 15 ) or in the presence ( FIG. 16 ) of 1 mg/ml human IgG.
  • bispecific antibody versions with afucosylated Fc show a higher phagocytic potency as compared to the corresponding wild-type Fc-bearing versions (3-10 fold lower EC50 for afucosylated K2AC5 and K2AC22 bispecific antibodies as compared to the wt version).
  • FIG. 17 shows the concentration dependent increase of phagocytosis (assessed with imaging based (Celllnsight) and expressed as phagocytosis index) induced by two selected CD47xCEA bispecific antibodies, i.e. K2AC5 and K2AC22, in presence or not of 1 mg/ml of human IgG.
  • the sequence-identical analogue of the anti-CD47 antibody Hu5F9-G4 (5F9 bearing a human IgG4 Fc portion, described in US20160333093) was run for comparison.
  • the addition of 1 mg/mL human IgG (this is even below the physiological plasma concentrations of IgG in men) sligthly impacts the potency (i.e. EC50) of the CD47xCEA bispecific antibodies while the activity driven by the mAb 5F9 is drastically impaired (EC50 and maximal phagocytosis).
  • FIG. 18 shows the effect of T-cell retargeting CEA-TCB1 (30nM) and CEA-TCB (300nM) bispecific antibodies (CEAxCD3 bispecific antibodies CEA-TCB: R06958688/cibisatamab, see for example Bacac et al Clin. Cancer Res., 22(13), 3286-97 (2016) and US20140242079; CEA-TCB1: from WO2017055389) on phagocytosis (assessed with imaging based assay (Celllnsight) and expressed as phagocytosis index) induced by the K2AC22 CEAxCD47 bispecific antibody.
  • CEAxCD3 bispecific antibodies CEA-TCB: R06958688/cibisatamab, see for example Bacac et al Clin. Cancer Res., 22(13), 3286-97 (2016) and US20140242079; CEA-TCB1: from WO2017055389
  • phagocytosis assessed with imaging
  • T cell retargeting bispecific antibodies the CD47xCEA bispecific antibody and the corresponding CD47 monovalent antibody were tested alone for comparison.
  • CEA-TCB nor CEA-TCB1 impairs the concentration dependent activity in phagocytosis of K2AC22.
  • FIG. 19 shows the killing (assessed by luminescence and expressed as % of killing) of MKN45 cancer cells in a mixed assay (with PBMCs and macrophages added, obtained from same human volunteer donor) by 2 selected CD47xCEA bispecific antibodies of the invention (K2AC5 ( FIG. 19A ) and K2AC22 ( FIG. 19B ) at two doses (i.e. 0.37 ⁇ g/mL or 1.1 ⁇ g/mL) alone, or in combination with CEA-TCB at 0.16 nM or 0.8 nM, and compared to the CEA-TCB alone or to an irrelevant hIgG1 control.
  • K2AC5 FIG. 19A
  • K2AC22 FIG. 19B
  • FIG. 20 shows the concentration dependent effects of the CEAxCD47 antibodies K2AC5 and 22 on binding (20A) and phagocytosis (20B) (assessed with imaging based assay (CellInsight) and expressed as phagocytosis index) in presence or not of 200 ng/mL of shed CEA. No significant influence of 200 ng/mL soluble CEA on the binding curves of both CEAxCD47 antibodies. No significant effect of soluble CEA on the maximal phagocytosis, EC50 are shifted by less than a factor of 4.
  • FIG. 21A shows the concentration dependent binding of CD47xCEA bispecific antibodies of the invention to MKN-45 cells as compared to the corresponding anti-CD47 monovalent antibody and an irrelevant hIgG1 control.
  • K2AC39 is a CD47xCEA bispecific antibody candidate cross-reactive to human CEACAM5 and human CEACAM6; while K2AC22 does not cross-react to CEACAM6. Binding to target cells (MKN-45) expressing CD47, CEACAM5 and CEACAM6 was assessed by FACS.
  • FIG. 21B shows the concentration dependent increase of phagocytosis (assessed by the imaging based assay (Celllnsight) and expressed as phagocytosis index) of MKN-45 cells induced by 2 different CD47xCEA bispecific antibodies (K2AC22 and K2AC39) as compared to the corresponding anti-CD47 monovalent antibody and an irrelevant hIgG1 control.
  • K2AC39 is CD47xCEA bispecific candidate cross-reactive to human CEACAM5 and human CEACAM6; while K2AC22 does not cross-react to CEACAM6. 1 mg/mL of human IgG is added in this experiment.
  • K2AC39 exhibits higher phagocytosis of MKN45 cells as compared to K2AC22.
  • antigenic determinant such as CEA, CD47 and CD3.
  • a binding part that binds membrane-bound human carcinoembryonic antigen (CEA, same as CEACAM5) or to CD47 specifically binds to CEA or CD47, more particularly to cell surface or membrane-bound CEA or CD47. Therefore, each binding part binds either to CEA or CD47.
  • CEA membrane-bound human carcinoembryonic antigen
  • CD47 specifically binds to CEA or CD47, more particularly to cell surface or membrane-bound CEA or CD47. Therefore, each binding part binds either to CEA or CD47.
  • specifically binding, specific for, binding to is meant that the binding is selective for the antigen and can be discriminated from unwanted or nonspecific interactions.
  • the extent of binding of an anti-target antibody to an unrelated, non-target protein is about 10-fold preferably >100-fold less than the binding of the antibody to said target as measured, e.g., by surface plasmon resonance (SPR) e.g.
  • SPR surface plasmon resonance
  • Biacore® enzyme-linked immunosorbent (ELISA) or flow cytometry (FACS).
  • Targets are the proteins discussed herein—e.g. CEA, CD47, and CD3c.
  • CEA binding part binds in addition to CEACAM6.
  • binding to CEA, CD47, binding to CEA, CD47, specific for CEA, CD47 refers in one embodiment to an antibody, e.g., bispecific antibody, that is capable of binding to the targets CEA and. CD47 with sufficient affinity such that the antibody is useful as a therapeutic agent in targeting tumor cells expressing CEA and CD47.
  • binding to MKN-45 cells with an EC50 value of refers to assay conditions whereby the bispecific antibody concentrations tested are between 0.1 and 1000 nM in the presence of anti-CD47 antibody B6H12.2 (ATCC® HB-9771TM, also named B6H12 herein) in a concentration of 300 nM.
  • the bispecific antibody according to the invention binds to cynomolgus CEACAM5 as well as human CEACAM5.
  • antibody refers to an antibody comprising two heavy chains and two light chains. In one embodiment the antibody is a full-length antibody.
  • antibody heavy chain refers to an antibody heavy chain, consisting of a variable region and a constant region as defined for a full-length antibody.
  • antibody light chain refers to an antibody light chain, consisting of a variable region and a constant region as defined for a full-length antibody.
  • full-length antibody denotes an antibody consisting of two “full-length antibody heavy chains” and two “full-length antibody light chains”.
  • a “full-length antibody heavy chain” is a polypeptide consisting in N-terminal to C- terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3), abbreviated as VH-CH1-HR-CH2-CH3.
  • a “full-length antibody light chain” is a polypeptide consisting in N-terminal to C- terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL), abbreviated as VL-CL.
  • the antibody light chain constant domain (CL) can be ⁇ (kappa) or ⁇ (lambda).
  • the two full-length antibody domains are linked together via inter-polypeptide disulphide bonds between the CL domain and the CH1 domain and between the hinge regions of the full-length antibody heavy chains.
  • Examples of typical full-length antibodies are natural antibodies like IgG (e.g. IgG 1 and IgG2), IgM, IgA, IgD, and IgE.
  • the full-length antibody according to the invention is in one embodiment of human IgG1 type, in one further embodiment comprising one or more amino acid substitutions in the Fc part as defined below and/or being glycoengineered at Asn297.
  • the full-length antibody according to the invention comprise two binding parts each formed by a pair of VH and VL, one binding to CEA and the other binding to CD47.
  • CDR complementarity determining region(s)
  • CDRs are also referred to as “hypervariable regions” and that term is used interchangeably herein with the term “CDR” in reference to the portions of the variable region that form the antigen binding regions. This particular region has been described by Kabat et al., U.S. Dept. of Health and Human Services, “Sequences of Proteins of Immunological Interest” (1983) and by Chothia et al., J. Mol. Biol.
  • FR-IMGT and CDR-IMGT regions of IG and TR is based on the “IMGT unique numbering for all IG and TR V-REGIONs of all species: interest for structure and evolution” (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003) and, for rearranged CDR3-IMGT and for FR4-IMGT, on the “IMGT unique numbering for V-DOMAIN and V-LIKE-DOMAIN” (Lefranc, M.-P. et al., Dev. Comp.
  • CDRL1 of SEQ ID NO:x
  • CDRL1 part of the referred variable light chain is of SEQ ID NO:x (comprising as CDRL1 a CDRL1 of SEQ ID NO:x). This is true also for the other CDRs.
  • Fc region refers to a C-terminal region of an IgG heavy chain; in case of an IgG1 antibody, the C-terminal region comprises —CH2—CH3 (see above).
  • the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to stretch from the amino acid residue at position Cys226 to the carboxyl-terminus.
  • Constant regions are well known in the state of the art and e.g. described by Kabat, E. A., (see e.g. Johnson, G., and Wu, T. T., Nucleic Acids Res.28 (2000) 214-218; Kabat, E. A., et al, Proc. Natl. Acad. Sci. USA 72 (1975) 2785-2788).
  • epitope includes any polypeptide determinant capable of specific binding to an antibody.
  • epitope includes chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and or specific charge characteristics.
  • An epitope is a region of a target that is bound by an antibody.
  • the bispecific antibody of the invention binds to the N-terminal domain of CEASCAM5 (Ig-like V-type domain of amino acids 35-144, UniProtKB-P06731). Binding location of the CEAxCD47 bispecific antibodies to CEASCAM5 is achieved via epitope binning.
  • antibodies are tested in a pairwise combinatorial manner, and antibodies that compete for the same binding region are grouped together into bins. Competition testing is performed herein with anti-CEA antibodies according to the state of the art and as described herein.
  • the bispecific antibody of the invention competes for binding to CEACAM5 with reference antibody SM3E (bin 1). In one embodiment the bispecific antibody of the invention does not compete for binding to CEACAM5 with reference antibodies SM3E, MEDI, T84.66, SAR, Lab, and CH1A1A (bin 2).
  • CEASCAM5 antibodies comprising the CEASCAM5 binding part of the CEAxCD47 bispecific antibody of the present invention are added at 0.2 ⁇ g/ml for 1 hour at room temperature. The plate is washed and the bound CEASCAM5 mAbs are detected.
  • the bispecific antibody of the invention binds to the B3 domain and the GPI anchor of CEACAMS. In one embodiment of the invention the antibody of the invention binds to the same epitope as an anti-CEA antibody (MAB CEA), which comprises a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21.
  • MAB CEA anti-CEA antibody
  • a common heavy chain refers to a polypeptide consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3), abbreviated as VH—CH1—HR—CH2—CH3.
  • VH antibody heavy chain variable domain
  • CH1 antibody constant heavy chain domain 1
  • HR antibody hinge region
  • CH2 antibody heavy chain constant domain 2
  • CH3 antibody heavy chain constant domain 3
  • Common heavy chains suitable for the bispecific antibodies according to the invention are heavy chains of an anti-CD47 antibody as described in WO2012023053, WO2013088259, WO2014087248, and WO2016156537 (each of which is incorporated by reference in its entirety).
  • the cHC of the bispecific antibody according to the invention comprises as light chain CDRs a CDRL1 of SEQ ID NO:1, a CDRL2 of SEQ ID NO:2, and a CDRL3 of SEQ ID NO:3 , and as heavy chain CDRs a CDRH1 of SEQ ID NO:25, a CDRH2 of SEQ ID NO:26 and a CDRH3 of SEQ ID NO:27.
  • the cHC of the bispecific antibody according to the invention comprises as heavy chain variable region VH a VH region of SEQ ID NO:4.
  • the cHC of the bispecific antibody according to the invention is of SEQ ID NO:5.
  • the antibody according to the invention is a i bispecific antibody comprising a cHC ( ⁇ Body).
  • the ⁇ Body format allows the affinity purification of bispecific antibodies which are undistinguishable from a standard IgG molecule and with characteristics that are undistinguishable from a standard monoclonal antibody (see e.g. WO2013088259, WO2012023053), promising no or low immunogenicity potential in patients.
  • Bispecific antibodies of the invention comprising a common heavy chain
  • WO2012023053 incorporated by reference in its entirety
  • the methods described in WO2012023053 generate bispecific antibodies that are identical in structure to a human immunoglobulin.
  • This type of molecule is composed of two copies of a unique heavy chain polypeptide, a first light chain variable region fused to a constant Kappa domain and second light chain variable region fused to a constant Lambda domain.
  • One binding site displays specificity to CEA and the other site displays specificity to CD47, wherein to each the heavy and the respective light chain contribute.
  • the light chain variable regions can be of the Lambda or Kappa family and are preferably fused to a Lambda and Kappa constant domains, respectively.
  • bispecific antibodies of the invention by fusing a Kappa light chain variable domain to a constant Lambda domain for a first specificity or fusing a Lambda light chain variable domain to a constant Kappa domain for the second specificity.
  • the other light chain is then always fully kappa (VL and CL) or fully lambda).
  • the bispecific antibodies described in WO 2012023053 are “ ⁇ Bodies”.
  • This ⁇ -Body format allows the affinity purification of a bispecific antibody that is undistinguishable from a standard IgG molecule with characteristics that are undistinguishable from a standard monoclonal antibody and, therefore, favourable as compared to previous formats including e.g. amino acid bridges or other unnatural elements.
  • An essential step of the method is the identification of two antibody Fv regions (each composed by a variable light domain and variable heavy domain) having different antigen specificities that share the same heavy chain variable domain.
  • Numerous methods have been described for the generation of monoclonal antibodies and fragments thereof. (see, e.g., Antibodies: A Laboratory Manual, Harlow E, and Lane D, 1988, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.).
  • Fully human antibodies are antibody molecules in which the sequence of both the light chain and the heavy chain, including the CDRs 1 and 2, arise from human genes.
  • the CDR3 region can be of human origin or designed by synthetic means. Such antibodies are termed “human antibodies”, or “fully human antibodies”.
  • Human monoclonal antibodies can be prepared by using the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al., 1983 Immunol Today 4: 72); and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al., 1985 In: Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • Human monoclonal antibodies may be utilized and may be produced by using human hybridomas (see Cote, et al., 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see Cole, et al., supra).
  • CEA, CEACAM5 refers to human carcinoembryonic antigen (CEA, CEACAM-5 or CD66e; UniProtKB-P06731) which is a cell surface glycoprotein and a tumor-associated antigen (Gold and Freedman, J Exp. Med., 121:439-462, 1965; Berinstein N L, J Clin Oncol., 20:2197-2207, 2002).
  • CEACAM6 refers to human CEACAM6 (CD66c; UniProtKB-P40199), which is also a member of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family.
  • CEACAM1 refers to human CEACAM1 (UniProtKB-P13688 (CEAM1_HUMAN) which is also a member of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family. Further information and information on other members of the CEA family can be found under http://www.uniprot.org.
  • MAB CEA refers to a monoclonal antibody specifically binding to human CEASCAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21.
  • MAB CEA1 refers to a monoclonal antibody specifically binding to human CEASCAM5, comprising a heavy chain variable region of SEQ ID NO:88 and a light chain variable region of SEQ ID NO:89.
  • the bispecific antibody according to the invention is competitive with MAB CEA, MAB CEA1, CEA-TCB, or CEA-TCB1; in a further embodiment the bispecific antibody according to the invention is not competitive with MAB CEA, MAB CEA1, CEA-TCB, or CEA-TCB1.
  • MAB CEA and said variable chains are described in US20140242079 (SEQ ID NO:21 and 27 of US20140242079 (incorporated by reference in its entirety)).
  • a bispecific anti-CEA x anti-CD3 ⁇ antibody (CEA-TCB) comprising the VH and VL of MAB CEA is described in Bacac et al Clin. Cancer Res., 22(13), 3286-97 (2016)).
  • bispecific CEAxCD3 Mab comprising the VH and VL of MAB CEA1 (CEA-TCB1) is described in WO2017055389 as molecule B “2+1 IgG CrossFab, inverted” with charge modifications (VH/VL exchange in CD3 binder, charge modification in CEA binder, humanized CEA binder) (see FIG. 3B and SEQ ID NOs 34, 36-38 of WO2017055389 (incorporated by reference in its entirety)).
  • bispecific CEA x CD3 antibody refers to antibody CEA-TCB or antibody CEA-TCB1.
  • CD47 is a multi-pass membrane protein and comprises three extracellular domains (amino acids 19-141, 198-207, and 257-268; see UniProtKB-Q08722).
  • binding affinity to CD47 is measured by SPR.
  • binding of the bispecific antibody according to the invention to CD47 occurs via one or more of said extracellular domains.
  • the bispecific antibodies according to the invention inhibit the interaction between human CD47 and human SIRP ⁇ .
  • binding to CEA refers in the context of the bispecific antibodies according to the invention to specificity for CEASCAM5 on the surface of a cell. Binding to CEA (CEACAMS) on cells is preferably measured with gastric adenocarcinoma MKN-45 cells comprising 200.000 to 600.000 CEA copies per cell. The concentration of the antibody according to the invention is varied in an appropriate range in regard to a resulting EC50 value for binding to MKN-45 cells as defined above.
  • the bispecific antibodies according to the invention are specifically binding to such cell membrane-bound CEACAM5 and do not or only minimally bind in a further embodiment to soluble CEACAM5, in concentrations like found in the blood/plasma of patients, i.e. soluble CEA in such concentrations, does not or only minimally influence the efficacy of a bispecific antibody of the invention. This is measured by influence of soluble CEA on the phagocytosis of MKN-45 cells by the bispecific antibodies of this invention as described.
  • membrane-bound human CEA refers to human carcinoembryonic antigen (CEA) that is bound to a membrane-portion of a cell or to the surface of a cell, in particular, the surface of a tumor cell.
  • CEA human carcinoembryonic antigen
  • the term “membrane-bound human CEA” may, in certain circumstances, refer to CEA which is not bound to the membrane of a cell, but which has been constructed so as to preserve the membrane bound CEA epitope to which the antibody according to the invention binds.
  • non-binding to soluble CEACAM5 refer in the context of the bispecific antibodies according to the invention that such antibodies do not show relevant binding to soluble CEACAM5, particularly when compared to membrane-bound CEACAM5.
  • non-binding can be indirectly determined by low influence of the soluble CEA on the phagocytosis activity of the bispecific antibody in a phagocytosis assay with MKN-45 cells as described below, preferably imaging based measurement of phagocytosis index, see Example 9).
  • Addition of 200 ng/ml soluble CEA will not shift this EC50 by more than a factor of 3, in one embodiment towards higher concentrations.
  • no or minimal influence of soluble CEA on binding of a bispecific antibody of this invention to CEA on cells is expected by the influence of the soluble CEA on the binding curve measured by flow cytometry (such a binding curve is shown in FIG. 20B ).
  • Addition of 200 ng/ml soluble CEA to the flow cytometry assay will not shift the binding curve respectively the EC50 by more than a factor of 3, in one embodiment towards higher concentrations
  • soluble CEA, shed CEA, sCEA refers to CEACAM5 that is not bound to or is cleaved from a cell membrane or cell surface (e.g., a tumor cell surface). Soluble CEA can, for example, be found in the blood stream of a subject with cancer. When CEA is shed from the cell membrane, it is assumed that the GPI anchor is disrupted, and CEACAM5 undergoes a conformational change that can prevent or at least weaken the binding of soluble CEA to the antibody according to the invention.
  • the terms “cross-reactivity against CEACAM6, specifically binding to CEACAM6, binding to CEACAM6, CEACAM6 binding part” refer in the context of the bispecific antibodies according to the invention that the bispecific antibody according to the invention recognizes specifically CEACAM5 and CEACAM6 on the surface (membrane) of a cell.
  • the bispecific antibodies according to the invention are specifically binding to membrane-bound CEACAM6, when compared to binding to membrane-bound CEA.
  • the ratio of the occupancy of CEACAM5 to CEACAM6 receptors on a cell surface by a given bispecific antibody of the invention is dependent on the binding affinities to CEACAM5 respectively CEACAM6 and can be easily calculated if these binding affinities have been measured, e.g. by SPR.
  • an antibody that specifically binds to CEACAM5 does not bind to carcinoembryonic antigen-related cell adhesion proteins such as, CEACAM1, CEACAM3, CEACAM4, CEACAM6, CEACAM7 and CEACAM8. In certain embodiments, an antibody that specifically binds to CEACAM5 also binds to CEACAM6 at similar EC50.
  • the terms “no substantial cross-reactivity against CEACAM1 and/or CEACAM3, CEACAM4, CEACAM6, CEACAM7 and CEACAM8, non-binding to said CEACAM” refer in the context of the bispecific antibodies according to the invention that such antibodies do not show any relevant binding to said membrane-bound CEACAM at therapeutic plasma concentrations (1 to 1000 nM), when compared to membrane-bound CEACAM5.
  • Non-binding to CEACAM1 and/ or CEACAM5 can be determined by flow cytometry based measurement of the binding curve to recombinant CHO cells expressing said CEACAM and to CEACAM3 and/ or CEACAM4, and CEACAM7 by measurement of the binding curve to recombinant PEAK cells expressing said CEACAM or by an ELISA assay measuring the binding to the recombinant CEACAM proteins.
  • the terms “does not bind, no binding to” a compound mentioned herein e.g. human IgG
  • OD values for such unrelated compounds will be about equal to that of the limit of detection.
  • CEA-TCB refers to a bispecific antibody binding to CEA and CD3 as described in US20140242079 (incorporated by reference in its entirety) as SEQ ID NO:1, 2, 21, and 22.
  • SEQ ID NO:96 The amino acid sequences of CEA-TCB are also described as SEQ ID NO:96 to 99 of the present invention.
  • CEA-TCB1 refers to molecule B in the “2+1 IgG CrossFab, inverted” format with charge modifications (VH/VL exchange in CD3 binder, charge modification in CEA binder, humanized CEA binder); FIG. 3B , SEQ ID NOs 34, 36-38 of WO2017055389(incorporated by reference in its entirety)).
  • the amino acid sequences of CEA-TCB1 are described as SEQ ID NO:92 to 95 of the present invention.
  • Further CEAxCD3 Mabs are described in WO2007071426, WO2013012414, WO2015112534, WO2017118675, US20140242079 and WO2017055389 (each of which is incorporated by reference in its entirety).
  • a further CEAxCD3 Mab is RO6958688 (see e.g. Bacac et al Clin. Cancer Res., 22(13), 3286-97 (2016).
  • said CEAxCD3 Mab is competitive and/or binds to the same epitope of human CEASCAM5 as MAB CEA.
  • said CEAxCD3 Mab is competitive and/or binds to the same epitope of human CEASCAM5 as MAB CEA1.
  • CD3 Mab antibody against CD3 refers to human CD3c (UniProtKB-P07766 (CD3E HUMAN).
  • the term “antibody against CD3c, anti CD3c antibody” relates to an antibody specifically binding to CD3c.
  • the antibody against CD3c is specifically binding to the same epitope as anti-CD3 antibody SP34 (BD Biosciences Catalog No.565983).
  • the antibody against CD3c is specifically binding to an epitope of human CD3c, comprising the amino acid sequence of SEQ ID NO:22.
  • the antibody against CD3c is specifically binding to human CD3c and comprises a heavy chain variable region of SEQ ID NO:90 and a light chain variable region of SEQ ID NO:91.
  • CEA-TCB and/or CEA-TCB1 for binding on CEA as presented on MKN-45 cells Therefore CEA-TCB in a concentration of 300 nM (CEA-TCB) or 30 nM (CEA-TCB1) do not shift the EC50 of the phagocytosis index curve of said the bispecific antibody of the invention for MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • 300 nM are a concentration measured in patient plasma at therapeutically effective doses of CEA-TCB ((J. Tabernero et. al., J. Clin. Oncol. 35, 2017 (suppl. Abstr. 3002)).
  • CEA-TCB1 is in preclinical investigations approx. 10 to 100 times more potent than CEA-TCB (binding affinity, tumor cell lysis, WO2017055389), therefore the shift of the EC50 is tested at 30 nM.
  • Non-competition means that EC50 is shifted by less than a factor of 3, in one embodiment to towards higher concentrations, if 300 nM of MAB CEA or CEA-TCB are added to the assay. 300 nM are a concentration in the range of therapeutically active doses/plasma-concentrations of CEA x CD3 bispecific antibody (CEA-TCB) (J.Tabernero et. al., J. Clin. Oncol. 35, 2017 (suppl. Abstr. 3002)).
  • Non-competition by MAB CEA1 or CEA-TCB1 means that EC50 is shifted by less than a factor of 3 if 30 nM of MAB CEA1 respectively CEA-TCB1 are added to the assay.
  • Non-competition means that EC50 is changed by less than a factor of 3 if 300 nM of MAB CEA, or CEA-TCB are added to the assay. 300 nM are a concentration in the range of therapeutically active doses/plasma-concentrations of CEA x CD3 bispecific antibody (CEA-TCB) (J. Tabernero et. al., J. Clin. Oncol. 35, 2017 (suppl. Abstr. 3002)).
  • Non-competition by MAB CEA1 or CEA-TCB1 means that EC50 is changed by less than a factor of 3 if 30 nM of MAB CEA1 respectively CEA-TCB1 are added to the assay.
  • noncompetitive means that a second antibody (MAB CEA, MAB CEA1 or a bispecific antibody against CEAxCD3c, like CEA-TCB or CEA-TCB1) in a concentration of 300 nM (MAB-CEA, CEA-TCB) or 30 nM (MAB CEA1, CEA-TCB1) does not shift the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • the term “competitive” means that a second antibody (MAB CEA, MAB CEA1 or bispecific antibody against CEAxCD3c, like CEA-TCB or CEA-TCB1) in a concentration of 300 nM respectively 30 nM (MAB CEA1 or CEA-TCB1) shifts the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, preferably by more than a factor of 5 towards higher concentrations.
  • a second antibody MAB CEA, MAB CEA1 or bispecific antibody against CEAxCD3c, like CEA-TCB or CEA-TCB1
  • MAB CEA1 or CEA-TCB1 shifts the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, preferably by more than a factor of 5 towards higher concentrations.
  • CDR complementarity determining region
  • Kabat numbering refers to the numbering system set forth by Kabat et al., U.S. Dept. of Health and Human Services, “Sequence of Proteins of Immunological Interest” (1983). Unless otherwise specified, references to the numbering of specific amino acid residue positions in bispecific antibody according to the invention are according to the Kabat numbering system.
  • ADCP antibody-dependent cell-mediated phagocytosis
  • phagocytosis As used herein “phagocytosis, EC50 value of phagocytosis, maximum of phagocytosis, phagocytosis index” according to the invention refer to phagocytosis measured with MKN-45 cells by “imaging”.
  • An appropriate imaging method with incubation at an effector (macrophages):target (tumor) cell ratio of e.g. 1:1 or 1:3 and with the “phagocytosis index” as readout (Imaging determined ADCP′′) is described in Example 9.
  • FIG. 3B shows the maximal achievable phagocytosis index as determined in tested concentration range of 0.1 or even lower to approx. 500 nM of bispecific TAA x CD47 antibodies.
  • phagocytosis of said bispecific antibody means phagocytosis caused/induced by said antibody.
  • Antibody K2AC22 comprises a first binding part, specifically binding to human CEASCAM5 and a second binding part, specifically binding to human CD47, whereby the first binding part comprises a heavy chain of SEQ ID NO:5 and a light chain of SEQ ID NO:65, and that the second binding part comprises a heavy chain of SEQ ID NO:5 and a light chain of SEQ ID NO:11.
  • Antibody K2AC5 comprises a first binding part, specifically binding to human CEASCAM5 and a second binding part, specifically binding to human CD47, whereby the first binding part comprises a heavy chain of SEQ ID NO:5 and a light chain of SEQ ID NO:64, and that the second binding part comprises a heavy chain of SEQ ID NO:5 and a light chain of SEQ ID NO:11.
  • Antibodies K2AC10, K2AC13 K2AC18, K2AC23, K2AC25, K2AC26, K2AC27, K2AC28, K2AC29 comprise the same heavy chains and second binding part light chain, but differ in the first binding part light chain (see table 1, sequence list).
  • phagocytosis can also be measured by a flow cytometry based method as % phagocytosis (see Example 9 and FIG. 3A for dependency of % phagocytosis from the concentrations of monovalent TAA and CD47 antibodies and TAA x CD47 bispecific antibody) and at a ratio of e.g. 3 human macrophages to 1 target/tumor-cell (“flow cytometry determined ADCP”).
  • human IgG, hIgG refers to a commercially available clinical-grade homogeneous preparation of human immunoglobulin IgG (from company Bio-rad.com) that does not bind specifically to CD47 and CEACAMS.
  • Antibodies produced in CHO cells typically have complex biantennary structures with very low or no bisecting-N-acetylglucosamine (bisecting GlcNAc) and high levels of core fucosylation.
  • Bisecting GlcNAc bisecting GlcNAc
  • Overexpression of N-acetylglucosaminyltransferase III has been used to increase the fraction of bisecting GlcNAc that resides on antibodies to improve antibody-dependent cellular cytotoxicity (ADCC).
  • RNAi and gene deletion technologies have also been used to decrease or eliminate the fucose on antibodies to dramatically increase ADCC activity (Davis J. et al.; Biotechnol. Bioeng. 2001;74:288-294; Saba J A, et al.; Anal. Biochem.
  • the bispecific antibody according to the invention is glycoengineered.
  • the glycoengineered bispecific antibody according to the invention has increased ADCC and/or ADCP activity (decreased EC50 and/or higher maximum of phagocytosis index) compared to the bispecific antibody comprising an Fc part included in SEQ ID NO:5 (parent antibody), comprising glycosylation according to a production in a CHO K1 cell line (ATCC® CCL-61TM) at standard conditions (1000m1 vessel, temperature 37° C., pH 7.0, impeller speed 80 rpm, minimum dissolved oxygen 30%; cultivation time 14 days).
  • the increase in ADCC is by a factor of 1.2 to 2.0 or even at least 2.0 as compared to said parent antibody see e.g. FIGS. 13 and 14 ).
  • the increase in ADCP is by a factor of at least 3 or even 5 or more as compared to said parent antibody (see e.g. FIGS. 15 and 16 ).
  • polypeptide having GnTIII activity refers to polypeptides that are able to catalyze the addition of a N-acetylglucosamine (GlcNAc) residue in ⁇ 1-4 linkage to the ⁇ -linked mannoside of the trimannosyl core of N-linked oligosaccharides, eg. ⁇ -1,4-mannosyl-glycoprotein4- ⁇ -N-acetylglucosaminyl-transferase (EC 2.4.1.144).
  • GlcNAc N-acetylglucosamine
  • FUT8 refers to ⁇ 1,6-fucosyltransferase (EC:2.4.1.68).
  • effector function Fc-mediated cellular cytotoxicity refers to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody.
  • antibody effector functions include, but are not limited to, Fc receptor binding affinity, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune-complex-mediated antigen uptake by antigen-presenting cells, down-regulation of cell surface receptors, etc.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • cytokine secretion immune-complex-mediated antigen uptake by antigen-presenting cells, down-regulation of cell surface receptors, etc.
  • immune mechanism is leading to the lysis of “targeted cells” by “human immune effector cells.”
  • glycoengineered antibody refers to a bispecific antibody according to the invention which comprises a reduced amount of fucosylated and/or bisecting oligosaccharides attached to the Fc region of said antibody, usually at amino acid Asn297, compared to a parent antibody.
  • parent antibody, parent bispecific antibody in the context of glycoengineering refers to a bispecific antibody according to the invention which comprises the same amino acid composition as the glycoengineered antibody but is non-glycoengineered.
  • the parent antibody and the glycoengineered antibody are produced in the same host cell, but in the first case in the host cell without glycoengineering, and in the second case in the same host cell but engineered by targeted disruption of the FUT8 gene or engineered by expressing a polynucleotide encoding a polypeptide having GnTIII activity under standard conditions (see above).
  • human immune effector cells refers to a population of leukocytes that display Fc receptors on their surfaces, through which they bind to the Fc-region of antigen binding molecules or of Fc-fusion proteins and perform effector functions.
  • a population may include, but is not limited to, peripheral blood mononuclear cells (PBMC) and/or natural killer (NK) cells and/or macrophages.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer
  • the term “increased Fc-mediated cellular cytotoxicity” is defined as either an increase in the number of “targeted cells” that are lysed in a given time, at a given concentration of the bispecific antibody of the invention in the medium surrounding the target cells, by the mechanism of Fc-mediated cellular cytotoxicity defined above, and/or a reduction in the concentration of the bispecific antibody of the invention, in the medium surrounding the target cells, required to achieve the lysis of a given number of “targeted cells” in a given time, by the mechanism of Fc-mediated cellular cytotoxicity.
  • the increase in Fc-mediated cellular cytotoxicity is relative to the cellular cytotoxicity mediated by the same bispecific antibody of the invention produced by the same type of host cells, using the same standard conditions, but that has not been produced by host cells engineered to have an altered pattern of glycosylation (e.g., to express the glycosyltransferase, GnTIII, or other glycosyltransferases or FUT8 disruption) by the methods described herein.
  • expression vector refers to one or more vectors which comprise the heavy and light chains of the antibody according to the invention in an appropriate manner as known from the state of the art.
  • host cells engineered by targeted disruption of the FUT8 gene refers to host cells capable of expressing an antibody according to the invention and being in addition glycoengineered by targeted disruption of the FUT8 gene as described e.g. in U.S. Pat. Nos. 8,067,232, 7,425,446, 6,946,292 (each of which is incorporated by reference in its entirety), and Yamane-Ohnuki N. et al., Biotech. Bioeng.; 87 (2004) 614-622.
  • An antibody according to the invention expressed in such host cell comprises a Fc region comprising complex N-glycoside-linked sugar chains bound to the Fc region, which comprise a reducing end which contains an N-acetylglucosamine, wherein the sugar chains do not contain fucose bound to the 6 position of N-acetylglucosamine in the reducing end of the sugar chains.
  • the present invention is further directed to a method for the production of a bispecific antibody according to the present invention characterized in comprising nonfucosylation of 50% to 100%, 60% to 100%, 70% to 100%, 80% to 100%, or 90% to 100%, that are produced by a host cell, comprising expressing in said host cell a nucleic acid encoding a bispecific antibody of the invention and a nucleic acid encoding a polypeptide with a glycosyltransferase activity, or a vector comprising such nucleic acids.
  • Genes with glycosyltransferase activity include (1,4)-N-acetylglucosaminyltransferase III (GnTIII), a-mannosidase II (Manll), (1,4)-galactosyltransferase (GalT), (1,2)-N-acetylglucosaminyltransferase I (GnTI), and ⁇ (1,2)-N-acetylglucosaminyltransferase II (GnTII).
  • a combination of genes with glycosyltransferase activity is expressed in the host cell (e.g., GnTIII and Man II).
  • the method also encompasses expression of one or more polynucleotide(s) encoding the bispecific antibody in a host cell in which a glycosyltransferase gene has been disrupted or otherwise deactivated (e.g., a host cell in which the activity of the gene encoding al-6 core fucosyltransferase has been knocked out).
  • the bispecific antibodies of the present invention can be produced in a host cell that further expresses a polynucleotide encoding a polypeptide having GnTIII activity to modify the glycosylation pattern.
  • the polypeptide having GnTIII activity is a fusion polypeptide comprising the Golgi localization domain of a Golgi resident polypeptide.
  • the expression of the bispecific antibodies of the present invention in a host cell that expresses a polynucleotide encoding a polypeptide having GnTIII activity results in bispecific antibodies with increased Fc receptor binding affinity and increased effector function.
  • the present invention is further directed to a method for the production of a bispecific antibody according to the present invention characterized in comprising non-fucosylation of 50% to 100%, 60% to 100%, 70% to 100%, 80% to 100%, or 90% to 100%, that are produced by a host cell, comprising expressing in said host cell a nucleic acid encoding a bispecific antibody of the invention and a disrupted FUT8 gene.
  • the bispecific antibodies with altered glycosylation produced by the host cells of the invention exhibit increased Fc receptor binding affinity and/or increased effector function as a result of the modification of the host cell (e.g., by expression of a glycosyltransferase gene).
  • the increased Fc receptor binding affinity is increased binding to a Fc ⁇ activating receptor, such as the Fc ⁇ RIIIa receptor.
  • the percentage of nonfucosylated oligosaccharides is 50% to 100%, specifically 60% to 100%, 70% to 100%, and more specifically, 80% to 100%.
  • the nonfucosylated oligosaccharides may be of the hybrid or complex type.
  • the bispecific antibody produced by the methods of the invention has an increased proportion of bisected oligosaccharides in the Fc region as a result of the modification of its oligosaccharides by the methods of the present invention.
  • the percentage of bisected oligosaccharides is 50% to 100%, specifically 50%, 60% to 70%, and more specifically, 80%.
  • the bispecific antibody produced by the host cells and methods of the invention has an increased proportion of bisected, nonfucosylated oligosaccharides in the Fc region.
  • the bisected, nonfucosylated oligosaccharides may be either hybrid or complex.
  • the term “host cell” covers any kind of cellular system which can be engineered to generate the bispecific antibodies of the present invention.
  • the host cell is engineered to allow the production of an antigen binding molecule with modified glycoforms.
  • the host cells have been further manipulated to express increased levels of one or more polypeptides having GnTIII activity.
  • Host cells include cultured cells, e.g., mammalian cultured cells, such as CHO cells (see above), BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
  • Host cells for the production of glycoengineered bispecific antibodies of the present invention have been described e.g. in U.S. Pat. No. 6,602,684, US20040241817, US20030175884; and WO 2004065540.
  • the bispecific antibodies of the present invention can alternatively be glycoengineered to have reduced fucose residues in the Fc region according to the techniques disclosed in US2003/0157108, EP1176195, W02003084570, WO2003085119 and US2003/0115614, US2004/093621, US2004/110282, US2004/110704, US2004/132140 (each of which is incorporated by reference in its entirety).
  • Glycoengineered bispecific antibodies of the invention may also be produced in expression systems that produce modified glycoproteins, such as those described in WO2003/056914, WO2004/057002, and WO2004/024927(each of which is incorporated by reference in its entirety).
  • the antibody according to the invention comprises one or two or three amino acid substitutions in the Fc region (“Fc amino acid substitution”) selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E (“DE substitution”), S239D and G236A, and triple-substitution S329D and I332E and G236A (“DEA substitution”); (Richards J O, et al., Mol. Cancer Ther. 7 (2008) 2517-2527). Due to different counting of a heavy chain, these amino acid numbers can be different for +/ ⁇ one, two or three amino acids, but with the same shift for all three.
  • Fc amino acid substitution selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E (“DE substitution”), S239D and G
  • SEQ ID: 5 In case of the heavy chain of SEQ ID NO:5 there is a one amino acid shift and S329D and I332E and G236A therefore denotes S328D and I33 1E and G235A.
  • SEQ ID: NO:5 with DE substitution is shown in SEQ ID NO:23 and SEQ ID: NO:5 with DEA substitution is shown in SEQ ID NO:24.
  • ADCC and/or ADCP activity of the bispecific antibody can be increased by such amino acid modification of the Fc part.
  • parent antibody, parent bispecific antibody in the context of Fc substitution refers to a bispecific antibody according to the invention which comprises the same amino acid composition as the Fc substituted antibody, but without said substitution(s).
  • parent antibody and the Fc substituted antibody are produced—as in the case of glycoengineered antibodies—in the same host cell under the same conditions, but in the first case in the host cell without Fc substitution, and in the second case in the same host cell but with such Fc substitution(s).
  • a useful host cell line is e.g. CHO-K1.
  • parent antibody in the context of a bispecific antibody according to the invention which comprises Fc substitution and is glycoengineered, such parent antibody therefore is the respective bispecific antibody which comprises the same amino acid composition as the Fc substituted antibody, but without said substitution(s) and is not glycoengineered.
  • ADCC and/or ADCP activity of the bispecific antibody is increased by amino acid substitution of the Fc part in combination with glycoengineering of the Fc part compared ADCC and/or ADCP activity of the respective parent antibody.
  • the invention comprises therefore in one embodiment a bispecific antibody specifically binding to human CEASCAM5 and human CD47, characterized in comprising one or two or three amino acid substitutions in the Fc region (“Fc amino acid substitution”) selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E, of triple-substitutions S329D and I332E and G236A and comprising non-fucosylation of the Fc part of 50% to 100%, 60% to 100%, 70% to 100%, 80% to 100%, or 90% to 100%.
  • Fc amino acid substitution selected from the group consisting of mono-substitutions S239D, I332E, G236A, of bi-substitutions I332E and G236A, S239D and I332E, of triple-substitutions S329D and I332E and G236A and comprising non-fucosylation of the Fc
  • Example 9 describes assays used for the determination of ADCC activity and also of ADCP activity
  • ADCC can be measured by an in vitro ADCC assay as follows:
  • ADCC is defined as either an increase in the maximum percentage of specific lysis observed within the bispecific antibody concentration range tested above, and/or a reduction in the concentration of bispecific antibody required to achieve one half of the maximum percentage of specific lysis (EC50) observed within the bispecific antibody concentration range tested above.
  • the increase in ADCC is relative to the ADCC, measured with the above assay, mediated by the same bispecific antibody, produced by the same type of host cells, using the same standard production, purification, formulation and storage methods, but that has not been produced by host cells engineered to overexpress GnTIII or by host cells engineered by targeted disruption of the FUT8 gene (“parent antibody”).
  • the increase in ADCC is relative to the ADCC measured with the parent bispecific antibody not carrying the substitution(s).
  • the increase in ADCC is relative to the ADCC measured with the parent non glycoengineered, bispecific antibody not carrying the sub stitution(s).
  • the CEACAM x CD47 bispecific antibodies according to the invention are optimized for treatment of solid tumors mainly by macrophages mediated phagocytosis of the tumor cells, either in monotherapy or in combination therapy especially together with a CEAxCD3 T-cell bispecific antibody like CEA-TCB or CEA-TCB1 and/or PD-1 axis antagonist.
  • the antibody according to the invention and the CEAxCD3 T-cell bispecific antibody can be administered as described below.
  • the disease resp. solid tumor is a cancer that expresses or even overexpresses CEA, including but not limited to the group of colorectal tumors, non-small cell lung tumors, gastric tumors, pancreatic tumors and breast tumors.
  • the tumor is a colorectal tumor. All therapeutic applications methods of use, uses, combinations, etc. described herein are especially embodiments for the treatment of these tumors/diseases.
  • the inventors recognize that the antibodies according to the invention show low or no ADA formation potential respectively loss of exposure due to neutralizing ADA respectively loss of efficacy.
  • the invention provides a method of treating carcinomas (cancer, tumors, for example, human carcinomas), especially CEA expressing tumors, in vivo.
  • This method comprises administering to a subject a pharmaceutically effective amount of a composition containing a bispecific antibody of the invention.
  • subject is meant a human subject, in one embodiment a patient suffering from cancer/tumor/carcinoma.
  • CEA expression in various tumor entities is generally very high, especially in colorectal carcinoma, pancreatic adenocarcinoma, gastric cancer, non-small cell lung cancer, breast cancer, head and neck carcinoma, uterine and bladder cancers among others.
  • CEA is mainly expressed in a polarized pattern on the apical surface of the cells. This polarized expression pattern limits the accessibility by anti-CEA mono or bispecific antibodies which are administered systemically and therefore potential toxicity. Together with the low affinity CD47 binding of the antibody of the invention this leads to no or limited phagocytosis of such normal cells by the antibody of the invention. This polarized expression pattern gets lost in the cells of gastrointestinal and other malignant tumors.
  • CEA is expressed equally over the whole cell surface of the cancer cells that means cancer cells are much better accessible to an antibody of the invention than normal, healthy cells and can be selectively killed by the CEAxCD47 bispecific antibodies of the invention respectively by the combinations mentioned above.
  • the bispecific antibodies of this invention can be used in monotherapy for the treatment of advanced solid tumors, in one embodiment CEA expressing tumors.
  • a bispecific antibody according to the invention is used in combination with a CEAxCD3 Mab in simultaneous, separate, or sequential combination.
  • a bispecific antibody according to the invention is used in combination with a CEAxCD3 Mab and/or a PD-1 axis antagonist in simultaneous, separate, or sequential combination.
  • a bispecific antibody according to the invention is used in combination with a PD-1 axis antagonist in simultaneous, separate, or sequential combination.
  • Such PD-1 axis antagonists are described e.g. in W02017118675. Such combinations attack the solid cancer by macrophages and T-cells.
  • CEA-TCB and CEA-TCB1 Two CEAxCD3 Mabs are in clinical development (CEA-TCB and CEA-TCB1; see clinicaltrials.gov; R06958688 in NCT3866239 and R07172508 in NCT03539484).
  • MEDI-565 was in clinical development but no active clinical trial could be identified in clinicaltrials.gov.
  • antibody CEA-TCB or CEA-TCB1 is used as bispecific antibody against CEA and CD3, antibody CEA-TCB or CEA-TCB1 is used.
  • the binder to CEA used in CEA-TCB has been derived from anti-CEA antibody
  • CEA-TCB has a low nM binding affinity to CEA and shows efficacy in high doses (between 40 and 600 mg per dose and patient; (see e.g. J.Tabernero et. al., J. Clin. Oncol. 35, 2017 (suppl. Abstr. 3002)). At these doses nearly all CEA targets on the cell surfaces are occupied by the CEA-TCB.
  • Combination of CEA-TCB or CEA-TCB1 and CEAxCD47 generates therapeutic plasma levels of both drugs at the same time and achieves best results (additive or even synergistic), if both drugs are non-competitive for the CEA antigen.
  • the terms “combination, simultaneous, separate, or sequential combination” of a an antibody according to the invention and a second bispecific antibody, binding to human CEA and human CD3c refer to any administration of the two antibodies (or three antibodies in case of the combination of an antibody of the invention, a CEAxCD3 Mab and a PD-1 axis antagonist), either separately or together, where the two or three antibodies are administered as part of an appropriate dose regimen designed to obtain the benefit of the combination therapy, for example in separate, sequential, simultaneous, concurrent, chronologically staggered or alternating administration.
  • the two or three antibodies can be administered either as part of the same pharmaceutical composition or in separate pharmaceutical compositions.
  • the antibody according to the invention can be administered prior to, at the same time as, or subsequent to the administration of the second bispecific antibody, or in some combination thereof.
  • the second bispecific antibody can be administered prior to, at the same time as, or subsequent to, each administration of the antibody of the invention or some combination thereof, or at different intervals in relation to the treatment with the antibody of the invention, or in a single dose prior to, at any time during, or subsequent to the course of treatment with the antibody of the invention.
  • the antibody according to the invention and the second bispecific antibody are administered in alternating administration, in one embodiment in intervals of 6 to 15 days between administration of the antibody of the invention and the second antibody. In such alternating administration the first dose can be the antibody of the invention or the second antibody.
  • PD-1 axis antagonist refers to an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • Anti-PD-1 antibodies are e.g. pembrolizumab (Keytruda®, MK-3475), nivolumab, pidilizumab, lambrolizumab, MEDI-0680, PDR001, and REGN2810.
  • Anti-PD-1 antibodies are described e.g. in 5 WO200815671, WO2013173223, WO2015026634, U.S. Pat. Nos.
  • Anti-PD-L1 antibodies are e.g. atezolizumab, MDX-1 105, durvalumab and avelumab.
  • Anti-PD-L1 antibodies are e.g. described in WO2015026634, WO2013/019906, WO2010077634, U.S. Pat. No. 8,383,796, WO2010077634, WO2007005874, and WO2016007235 (each of which is incorporated by reference in its entirety).
  • both compounds may be present in one single dosage form or in separate dosage forms, for example in two different or identical dosage forms.
  • both antibodies if desired by the physician, can be administered simultaneously. If the antibody of the invention and the second antibody are competing in regard to CEASCAM5, in one embodiment both antibodies are administered in alternating administration.
  • the antibody of the invention will typically be administered to the patient in a dose regimen that provides for the most effective treatment of the cancer (from both efficacy and safety perspectives) for which the patient is being treated, as known in the art.
  • tumor cells are attacked at the same time by T-cells and macrophages, to achieve full therapeutic potential of this approach, CEA-CD3 and CEAxCD47 bispecific antibody have to be non-competitive regarding binding to CEA on cell surface.
  • the amount of the antibody administered and the timing of the administration of the antibody of the invention can depend on the type (e.g. gender, age, weight) and condition of the patient being treated, the severity of the disease or condition being treated, and on the route of administration.
  • the antibody of the invention and the second antibody can be administered to a patient in doses ranging from 0.1 to 100 mg/kg of body weight per day or per week in single or divided doses, or by continuous infusion.
  • each of the antibodies of the invention and the second antibody is administered to a patient in doses ranging from 0.1 to 20 mg/kg.
  • dosage levels below the lower limit of the aforesaid range may be adequate, while in other cases still larger doses may be employed without causing any harmful side effect.
  • half-life of the antibody refers to the half-life of said antibody as measured in a usual pharmacokinetic assay, e.g. as described in example 17.
  • An antibody according to the invention and the second bispecific antibody against CEA and CD3 have elimination half-life of 3-14 days.
  • the invention is also directed to use of the bispecific antibody according to the invention in the treatment of disease, particularly cell proliferation disorders wherein CEA is expressed, particularly wherein CEA is abnormally expressed (e.g., overexpressed or expressed in a different pattern on the cell surface) compared to normal tissue of the same cell type.
  • diseases include, but are not limited to colorectal cancer, NSCLC (non-small cell lung cancer), gastric cancer, pancreatic cancer and breast cancer.
  • CEA expression levels may be determined by methods known in the art (e.g., via immunohistochemistry assay, immunofluorescence assay, immunoenzyme assay, ELISA, flow cytometry, radioimmunoassay etc.).
  • bispecific antibodies of the present invention can be used for targeting cells in vivo or in vitro that expresses CEA.
  • the bispecific antibodies of the invention are particularly useful in the prevention of tumor formation, eradication of tumors and inhibition of tumor growth or metastasis via the induction of ADCP and ADCC of tumor cells.
  • the bispecific antibodies of the invention can be used to treat any tumor expressing CEA.
  • Particular malignancies that can be treated with the bispecific antibodies of the invention include, but are not limited to, colorectal cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer and breast cancer.
  • the bispecific antibodies of the invention are administered to a mammal, preferably a human, in a pharmaceutically acceptable dosage form such as those discussed below, including those that may be administered to a human intravenously as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intra-cerebrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • the bispecific antibodies of the invention also are suitably administered by intra tumoral, peritumoral, intralesional, or perilesional routes, to exert local as well as systemic therapeutic effects.
  • bispecific antibodies of the invention For the treatment of disease, the appropriate dosage of bispecific antibodies of the invention will depend on the type of disease to be treated, the severity and course of the disease, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the bispecific antibody of the invention is suitably administered to the patient at one time or over a series of treatments.
  • the present invention provides a method for selectively killing tumor cells expressing CEA.
  • This method comprises interaction of the bispecific antibodies of the invention with said tumor cells.
  • These tumor cells may be from a human carcinoma including colorectal carcinoma, non-small cell lung carcinoma (NSCLC), gastric carcinoma, pancreatic carcinoma and breast carcinoma.
  • NSCLC non-small cell lung carcinoma
  • gastric carcinoma pancreatic carcinoma
  • breast carcinoma breast carcinoma
  • the invention is directed to the use of the bispecific antibodies of the invention for the manufacture of a medicament for treating a disease related to abnormal CEA expression.
  • the disease is a cancer that expresses or even overexpresses CEA, including but not limited to colorectal tumor, non-small cell lung tumor, gastric tumor, pancreatic tumor and breast tumor.
  • the tumor is a colorectal tumor.
  • compositions Compositions, Formulations, Dosages, and Routes of Administration
  • the present invention is directed to pharmaceutical compositions comprising the bispecific antibodies of the present invention and a pharmaceutically acceptable carrier.
  • the present invention is further directed to the use of such pharmaceutical compositions in the method of treatment of disease, such as cancer, or in the manufacture of a medicament for the treatment of disease, such as cancer.
  • the present invention is directed to a method for the treatment of disease, and more particularly, for the treatment of cancer, the method comprising administering a therapeutically effective amount of the pharmaceutical composition of the invention.
  • the present invention encompasses pharmaceutical compositions, combinations and methods for treating human carcinomas, tumors, as defined above.
  • the invention includes pharmaceutical compositions for use in the treatment of human carcinomas comprising a pharmaceutically effective amount of an antibody of the present invention and a pharmaceutically acceptable carrier.
  • the bispecific antibody compositions of the invention can be administered using conventional modes of administration including, but not limited to, intravenous, intraperitoneal, oral, intralymphatic or direct intratumoral administration. Intravenous administration or subcutaneous administration are preferred.
  • therapeutic formulations containing the bispecific antibodies of the invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or liquid formulations.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed.
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • the most effective mode of administration and dosage regimen for the pharmaceutical compositions of this invention depends upon the severity and course of the disease, the patient's condition and response to treatment and the judgment of the treating physician. Accordingly, the dosages of the compositions may be flat doses or may be adapted to the individual patient, e.g. the body weight. Nevertheless, an effective dose of the compositions of this invention will generally be in a range from 0.1 to 20 mg/kg.
  • the bispecific antibodies of this invention have a molecular weight in a magnitude of 150 kD per Mol. They carry in one embodiment a Fc part.
  • the elimination half-life in patients is in a range of 3 to 14 days. This half-life allows for, but not limited to administration once a day, once a week, or once every two weeks.
  • bispecific antibodies of the present invention and their respective compositions may be in a variety of dosage forms which include, but are not limited to, liquid solutions or suspensions, tablets, pills, powders, suppositories, polymeric microcapsules or microvesicles, liposomes, and injectable or infusible solutions.
  • dosage forms include, but are not limited to, liquid solutions or suspensions, tablets, pills, powders, suppositories, polymeric microcapsules or microvesicles, liposomes, and injectable or infusible solutions.
  • the preferred form depends upon the mode of administration and the therapeutic application.
  • composition comprising a bispecific antibody of the present invention will be formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disease or disorder being treated, the particular mammal being treated, the clinic condition of the individual patient, the cause of the disease or disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a bispecific antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a bispecific antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such
  • a bispecific antibody comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47.
  • bispecific antibody according to embodiment 1 characterized in that the Fc region has been glycoengineered to have a reduced number of fucose residues as compared to the same but non-glycoengineered bispecific antibody.
  • the bispecific antibody according to embodiment 1 or 2 characterized in that the first binding part binds to the Ig-like V-type domain of CEACAM5 of amino acids 35-144.
  • bispecific antibody according to any one of embodiments 1 to 3, characterized in that said bispecific antibody competes with antibody SM3E for binding to CEACAM5.
  • bispecific antibody according to any one of embodiments 1 to 3, characterized in that said bispecific antibody does not compete with antibodies SM3E, MEDI, LAB, SAR, T86.66, CH1A1A.
  • bispecific antibody according to any one of embodiments 1 to 5, characterized in that the EC50 value of phagocytosis of said bispecific antibody is in the range of 0.1 to 10 times of the E50 value of reference antibody K2AC22 under the same experimental conditions and in the presence or without of 1 mg/ml human IgG.
  • bispecific antibody according to any one of embodiments 1 to 6, characterized in that in presence of 1 mg/ml human IgG maximum of phagocytosis index measured in imaging based assay is not decreased by more than 30% in comparison to phagocytosis without human IgG under the same experimental conditions.
  • bispecific antibody according to any one of embodiments 1 to 7, characterized in being monovalent for the first binding part and monovalent for the second binding part.
  • each of the first and second binding part comprises an immunoglobulin heavy chain and an immunoglobulin light chain.
  • bispecific antibody according to any one of embodiments 1 to 9, characterized in being of human IgG1 type.
  • bispecific antibody according to any one of embodiments 1 to 10, characterized in that the constant and variable framework region sequences are human or of human origin.
  • bispecific antibody according to any one of embodiments 1 to 11, characterized in that the bispecific antibody is a full-length antibody.
  • bispecific antibody according to any one of embodiments 1 to 12, characterized in comprising a first binding part specifically binding to human CEACAM5, comprising a lambda light chain variable domain and a lambda light chain constant domain and a second binding part specifically binding to human CD47, comprising a kappa light chain variable domain and a kappa light chain constant domain.
  • the bispecific antibody according to any one of embodiments 13 or 14, characterized in comprising a common heavy chain.
  • bispecific antibody according to any one of embodiments 1 to 16, characterized in comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in
  • bispecific antibody according to any one of embodiments 1 to 17, characterized in comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in
  • bispecific antibody according to any one of embodiments 1 to 18, characterized in comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, characterized in
  • bispecific antibody according to any one of embodiments 1 to 21, characterized in binding to MKN-45 cells with an EC50 value of 1 to 200 nM.
  • bispecific antibody according to any one of embodiments 1 to 24, characterized in binding to human recombinant CEASCAM5 and CEACAM6, whereby the EC50 values of binding to recombinant CEASCAM5 and CEACAM6 differing by less than a factor of 3.
  • bispecific antibody according to any one of embodiments 1 to 27, characterized that a bispecific antibody specifically binding to human CEACAM5 and CD3 ⁇ (further named also as CEA-TCB1), comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95 in a concentration of 30 nM does not shift the EC50 of the binding curve of the bispecific antibody of the invention to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations . . .
  • bispecific antibody according to any one of embodiments 29 to 31, characterized that 80% to 100% of the N-linked oligosaccharides in the Fc region are bisected and nonfucosylated.
  • bispecific antibody according to any one of embodiments 29 to 32, characterized in that EC50 value of ADCC and/or ADCC maximum induced by said antibody is increased by a factor of 1.2 to 2.0 or a factor of at least 2.0 and/or EC50 value of the phagocytosis index curve is decreased by at least a factor of 1.2 to 2.0 or a factor of at least 2.0 compared to the maximum of ADCC and/or EC50 value induced by the respective parent bispecific antibody.
  • bispecific antibody according to any one of embodiments 29 to 33, characterized in that by imaging determined maximum of the phagocytosis index induced by said antibody is increased by at least a factor of 3 and/or EC50 value of the phagocytosis index curve is decreased by at least a factor of 3 or a factor of at least 5 compared to the maximum of the phagocytosis index respectively the EC50 value induced by the respective parent bispecific antibody.
  • a vector comprising the polynucleotide according to embodiment 37.
  • a host cell comprising the vector according to embodiment 38.
  • composition comprising a bispecific antibody according to any one of embodiments 1 to 34 and a pharmaceutically acceptable carrier.
  • a method of inducing cell lysis of a tumor cell comprising contacting the tumor cell with a bispecific antibody according to any one of embodiments 1 to 34.
  • tumor cell is a colorectal cancer cell, NSCLC (non-small cell lung cancer), gastric cancer cell, pancreatic cancer cell, breast cancer cell, or another tumor cell expressing human CEACAM5.
  • NSCLC non-small cell lung cancer
  • gastric cancer cell gastric cancer cell
  • pancreatic cancer cell pancreatic cancer cell
  • breast cancer cell or another tumor cell expressing human CEACAM5.
  • a method of treating a subject having a cancer that expresses CEA comprising administering to the subject a therapeutically effective amount of a bispecific antibody according to any one of embodiments 1 to 34.
  • a method of increasing survival time in a subject having a cancer that expresses CEA comprising administering to said subject a therapeutically effective amount of a bispecific antibody according to any one of embodiments 1 to 34.
  • the method according to embodiment 43 or 44 characterized in that the cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer or breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer pancreatic cancer or breast cancer.
  • bispecific antibody according to any one of embodiments 1 to 34 for use in the manufacture of a medicament for treating a subject having a cancer that expresses CEA.
  • the bispecific antibody for use according to embodiment 47 characterized in that the cancer is selected from the group consisting of colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer gastric cancer
  • pancreatic cancer breast cancer
  • the bispecific antibody according to any one of embodiments 1 to 34 for use in a method of treating a subject having a cancer that expresses CEA comprising administering to the subject a therapeutically effective amount of a said bispecific antibody, characterized in that the EC50 value of phagocytosis of said bispecific antibody is in the range of 0.1 to 10 times of the E50 value of reference antibody K2AC22, which comprises a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, whereby the first binding part comprises a heavy chain of SEQ ID NO:5 and a light chain of SEQ ID NO:65, and that the second binding part comprises a heavy chain of SEQ ID NO:5 and a light chain of SEQ ID NO:11, under the same experimental conditions and in the presence and/or without of 1 mg/ml human IgG.
  • a first bispecific antibody comprising a first binding part, specifically binding to human CEACAM5 and a second binding part, specifically binding to human CD47, for use in simultaneous, separate, or sequential combination with a second bispecific antibody comprising a third binding part specifically binding to human CEASCAM5 and a fourth binding part specifically binding to human CD3 ⁇ , in the treatment of a human subject having a cancer that expresses CEA.
  • the first bispecific antibody for use according to embodiment 50 characterized in that said fourth binding part of the second bispecific antibody binds to an epitope of human CD3 ⁇ which comprises the amino acid sequence of SEQ ID NO:22.
  • the first bispecific antibody for use according to embodiment 50 or 51 characterized in that said second antibody comprises as heavy and light chains the chains of SEQ ID NO:96 to 99 or comprises as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95 and that said second antibody in a concentration of 300 nM or 30 nM does not shift the EC50 of the binding curve of said first bispecific antibody to MKN-45 cells by more than a factor of 3, in one embodiment towards higher concentrations.
  • bispecific antibody for use in simultaneous, separate, or sequential combination with a second bispecific antibody comprising as heavy and light chains the chains of SEQ ID NO:96 to 99 or comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95 in the treatment of a subject having a cancer that expresses CEA.
  • bispecific antibody according to any one of embodiments 1 to 34 and 50 to 53, characterized in not competing with a second bispecific antibody comprising as heavy and light chains the chains of SEQ ID NO:96 to 99 or comprising as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95 for use in simultaneous, separate, or sequential combination with said second bispecific antibody in the treatment of a subject having a cancer that expresses human CEACAMS.
  • the bispecific antibody for use according to any one of embodiments 50 to 55 characterized in that said cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer pancreatic cancer
  • breast cancer breast cancer
  • bispecific antibody for use according to any one of embodiments 50 to 56 characterized in that the bispecific antibody according to any one of embodiments 1 to 34 and the second bispecific antibody are administered to said subject alternately or simultaneously in 6 to 15 day intervals.
  • a composition comprising a bispecific antibody according to any one of embodiments 1 to 34, characterized in not cross reacting with a second bispecific antibody comprising a) a third binding part specifically binding to human CEASCAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 and a fourth binding part specifically binding to, an epitope of human CD3c, comprising the amino acid sequence of SEQ ID NO:22, or b) as heavy and light chains the chains of SEQ ID NO:96 to 99 or as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95, for use in the treatment of a subject having a cancer that expresses human CEACAM5.
  • a composition comprising a bispecific antibody according to any one of embodiments 1 to 34, for use in simultaneous, separate, or sequential combination in the treatment of a subject having a cancer that expresses human CEASCAM5, with a second bispecific antibody comprising a) a third binding part specifically binding to human CEACAM5, comprising a heavy chain variable region of SEQ ID NO:20 and a light chain variable region of SEQ ID NO:21 and a fourth binding part specifically binding to, an epitope of human CD3 ⁇ , comprising the amino acid sequence of SEQ ID NO:22, orb) as heavy and light chains the chains of SEQ ID NO:96 to 99 or as heavy and light chains the chains of amino acid sequences SEQ ID NO: 92 to 95, whereby said second bispecific antibody in a concentration of 300 nM respectively 30 nM (SEQ ID NO: 92 to 95) does not shift the EC50 of the phagocytosis index curve of MKN-45 cells of the bispecific antibody according to any one of the embodiments 1 to 34
  • composition according to embodiment 58 or 59 characterized in that the cancer is colorectal cancer, non-small cell lung cancer (NSCLC), gastric cancer, pancreatic cancer and breast cancer.
  • NSCLC non-small cell lung cancer
  • gastric cancer gastric cancer
  • pancreatic cancer breast cancer
  • bispecific antibody according to any one of embodiments 61 or 62, characterized in that said bispecific antibody is administered in combination with chemotherapy or radiation therapy to a human subject.
  • NSCLC non-small cell lung cancer
  • gastric cancer pancreatic cancer or breast cancer.
  • the sequence corresponding to the extracellular domain of human CD47 is amplified from human cDNA by polymerase chain reaction (PCR) using specific oligonucleotides.
  • the amplification product is gel-purified and cloned into the pEAK8 mammalian expression vector (Edge Biosystems, Gaithersburg, Md.).
  • the vector is further modified to introduce an AvitagTM (Avidity, Denver Colo.) and a hexa-histidine tag at the C-terminus allowing for single site biotinylation of the protein and purification by IMAC (Immobilized Metal Ion Affinity Chromatography), respectively.
  • the constructs are verified by DNA sequencing.
  • the plasmid is then transfected into mammalian cells using a liposome-based transfection reagent such as Lipofectamine2000 (Thermofisher Scientific).
  • the transfection step requires only small quantities of DNA and cells, typically 2 ⁇ 10 5 cells and 2 ⁇ g of plasmid DNA per well and the transfection carried out in a 6-well plate.
  • different mammalian cell lines can be used, in the examples given below, transformed human embryo kidney monolayer epithelial cells (PEAK cells) are transfected. These cells stably express the EBNA-1 gene, further supporting the episomal replication process, are semi-adherent and can be grown under standard cell culture conditions (5% CO 2 ; 37° C. in DMEM medium supplemented with 10% fetal calf serum). After 24 h, cells are placed under selective conditions by adding medium containing 0.5-2 ⁇ g/mL puromycin: cells harboring the episomal vector are resistant to this antibiotic.
  • the CELLineTM is a two-compartment bioreactor that can be used in a standard cell culture incubator.
  • the smaller compartment (15 ml) contains the cells and is separated from a larger (one liter) medium containing compartment by a semi-permeable membrane with a cut-off size of 10 kDa (Bruce et al. 2002, McDonald et al. 2005).
  • This system allows for the diffusion of nutrients, gazes and metabolic waste products, while retaining cells and secreted proteins in the smaller compartment.
  • the culture is maintained for 7-10 days before harvest of the supernatant. As the medium contains serum, the cells maintain good viability and several production runs can be generated using the same cells and containers.
  • the cell culture supernatants are clarified by centrifugation.
  • the supernatant is then supplemented with 100 mM imidazole and loaded on Ni-NTA affinity chromatography resin (Qiagen).
  • Ni-NTA affinity chromatography resin Qiagen
  • the relatively high concentration of imidazole minimizes binding of contaminants to the resin.
  • proteins are eluted at a flow rate of 2 mL/min using a 30 mL imidazole gradient (20-400 mM imidazole) on an AKTA Prime chromatography system (Amersham Pharmacia Biotech).
  • the elution gradient further improves the purity of the recombinant protein but can be replaced by a step elution approach if a chromatography system is not available.
  • the eluted fractions can be analyzed by SDS-PAGE or ELISA to determine their content in recombinant protein.
  • the fractions of interest are pooled and desalted on Amicon 10 KD columns (Millipore) equilibrated with phosphate buffered saline or another appropriate buffer.
  • the desalted proteins can then be quantified using various techniques and their purity analyzed by SDS-PAGE.
  • Recombinant CD47 is biotinylated in vitro using biotin ligase (Avidity, Denver Colo.) according to manufacturer's instructions. After desalting the biotinylation level is evaluated by pull-down assays using streptavidin magnetic beads and SDS-PAGE analysis.
  • CEASCAM5 The sequence corresponding to the complete extracellular domain (ECD) and A3-B3 domains of CEASCAM5 were synthesized by Eurofins and Twist Bioscience. These synthetic genes were subcloned into the pEAK8 mammalian expression vector (Edge Biosystems, Gaithersburg, Md.). The vectors were modified to introduce an AvitagTM (Avidity, Denver Colo.) and either a hexa-histidine tag, a human FC region or a mouse FC region at the C-terminus. Constructs were verified by DNA sequencing.
  • AvitagTM Asvidity, Denver Colo.
  • Vectors encoding for the full-length version of human CEACAM 1, 3, 4, 5, 6, 7, 8, 18, 19, 20, 21 and cynomolgus CEACAM5 were also generated for expression at the cell surface of PEAK and/or CHO cells.
  • the soluble, full-length human CEACAM16 was also similarly cloned.
  • scFv phage libraries (10 12 Pfu) are blocked with PBS containing 3% (w/v) skimmed milk for one hour at room temperature on a rotary mixer.
  • Blocked phage is deselected on streptavidin magnetic beads (DynabeadsTM M-280) for one hour at room temperature on a rotary mixer.
  • Deselected phage is incubated with 100 nM of either biotinylated human CEACAM5 or the A3-B3 domain captured on streptavidin magnetic beads for two hours at room temperature on a rotary mixer. Beads are captured using a magnetic stand followed by five washes with PBS/0.1% Tween 20 and two washes with PBS.
  • Phage is eluted with 100 nM TEA for 30 minutes at room temperature on a rotary mixer. Eluted phage and beads are neutralized with Tris-HCl 1M pH 7.4 and directly added to 10 ml of exponentially growing TG1 cells and incubated for one hour at 37° C. with slow shaking (90 rpm). An aliquot of the infected TG1 is serial diluted to titer the selection output. The remaining infected TG1 are spun at 3800 rpm for 10 minutes and resuspended in 2 ml 2xTY and spread on 2xTYAG (2xTY medium containing 100 ⁇ g/ml ampicillin and 2% glucose) agar Bioassay plates.
  • 50 ⁇ l of cell suspension obtained from previous selection rounds are added to 50 ml of 2 ⁇ TYAG and grown at 37° C. with agitation (240 rpm) until an OD600 of 0.3 to 0.5 is reached.
  • the culture is then super-infected with 1.2 ⁇ 10 11 M13K07 helper phage and incubated for one hour at 37° C. (90 rpm).
  • the medium is changed by centrifuging the cells at 3800 rpm for 10 minutes, removing the medium and resuspending the pellet in 50 ml of 2 ⁇ TYAK (2 ⁇ TY medium containing 100 ⁇ g/ml ampicillin; 50 ⁇ g/ml kanamycin).
  • the culture is then grown overnight at 30° C. (240 rpm). The next day, the phage containing supernatant is used for the next round of selection.
  • Phage containing supernatants are blocked with PBS containing 3% (w/v) skimmed milk for one hour at room temperature on a rotary mixer. Blocked phage is then deselected for one hour on MKN45 CEACAMS KO that do not express human CEACAMS. Deselected phage is incubated with 2 ⁇ 10 7 MKN45 cells expressing CEASCAM5 (blocked in PBS 3% BSA 0.1% NaN 3 ) for two hours at room temperature with gentle shaking. Cells are pelleted and washed six times with PBS. Bound phage is eluted with 76 mM citric acid and shaking for 10 minutes.
  • Individual clones are inoculated into a deep-well microtiter plate containing 0.9 ml per well of 2 ⁇ TYAG medium (2 ⁇ TY medium contaning 100 ⁇ g/ml ampicillin, 0.1% glucose) and grown at 37° C. for 5-6 hours (240 rpm). 100 ⁇ l per well of 0.2 mM IPTG in 2 ⁇ TY medium are then added to give a final concentration of 0.02 mM IPTG. The plate is incubated overnight at 30° C. with shaking at 240 rpm. The deep-well plate is centrifuged at 3200 rpm for 10 minutes at 4° C. and the supernatant carefully removed.
  • the pellets are resuspended in 150 ⁇ l TES buffer (50 mM Tris-HCl (pH 8), 1 mM EDTA (pH 8), 20% sucrose, complemented with Complete protease inhibitor, Roche).
  • a hypotonic shock is produced by adding 150 ⁇ l of diluted TES buffer (1:5 TES:water dilution) and incubation on ice for 30 minutes. The plate is centrifuged at 4000 rpm for 10 minutes at 4° C. to pellet cells and debris. The supernatants are carefully transferred into another microtiter plate and kept on ice for immediate testing in functional assays or binding assays.
  • scFv for binding to CEACAM5 is tested in a homogenous assay using CellInsightTM technology.
  • the following reagents are mixed in each well of a 384 clear bottom well plate (Corning): 30 ⁇ l of a streptavidin polystyrene bead suspension (Polysciences; 3000 beads/well) coated with either biotinylated CEACAM5, biotinylated domain A3-B3 or biotinylated NusA for a control protein; 60 ⁇ l of blocked scFv periplasmic preparation; 10 ⁇ l of detection buffer (PBS containing mouse anti-c-myc antibody at 5 ⁇ g/ml; anti-mouse Fc AlexaFluor® 647 diluted 1:200).
  • detection buffer PBS containing mouse anti-c-myc antibody at 5 ⁇ g/ml
  • the 384-well plate After mixing at 600 rpm for 5 minutes, the 384-well plate is incubated at room temperature and read after 2 hours on a CellInsightTM CX5 High-Content Screening platform (ThermoFisher Scientific). Clones expressing scFv giving a specific signal for CEACAM5 and not NusA are selected for further analysis or sequencing.
  • CEACAM1, CEACAM6 and other CEACAMs Binding to CEACAM1, CEACAM6 and other CEACAMs can be measured in the same manner.
  • Single clones are inoculated into a 96-deep-well microtiter plate containing 1 ml LBAG medium (LB medium with 100 ⁇ g/ml ampicillin and 2% glucose) per well and grown overnight at 37° C., 240 rpm. DNA is extracted using the Zyppy-96 Plamis Miniprep kit (Zymo Research) and sequenced.
  • LBAG medium LB medium with 100 ⁇ g/ml ampicillin and 2% glucose
  • scFv candidates with the desired binding properties are reformatted into IgG and expressed by transient transfection into PEAK cells.
  • the VH and VL sequences of selected scFv are amplified with specific oligonucleotides and cloned into an expression vector containing the heavy and light chain constant regions and the constructions are verified by sequencing.
  • the expression vectors are transfected into mammalian cells using Lipofectamine 2000 (Thermofisher Scientific) according to manufacturer's instructions. Briefly, 3.5 ⁇ 10 6 PEAK cells are cultured in T75 flasks in 25 ml culture media containing fetal bovine serum.
  • Transfected cells are cultured for 5-6 days at 37° C., IgG production is quantified by OctetRED96 instrument.
  • the supernatant is harvested for IgG purification on FcXL affinity resin (Thermofisher Scientific) according to manufacturer's instructions. Briefly, supernatants from transfected cells are incubated overnight at 4° C. with an appropriate amount of FcXL resin. After resin wash by PBS, samples are loaded on Amicon Pro column and the IgG consequently eluted in 50 mM Glycine pH3.5. The eluted IgG fraction is then dialyzed by Amicon 50 kDa against Histidine NaCl pH6.0 buffer and the IgG content is quantified by absorption at 280 nm. Purity and IgG integrity are verified by Agilent Bioanalyzer manufacturer (Agilent Technologies, Santa Clara, Calif., USA).
  • CEACAM5 monoclonal antibodies can be shown by flow cytometry using PEAK and/or CHO cells transfected with different members of the CEACAM family.
  • Vectors encoding the full-length version of human CEACAM 1, 3, 4, 5, 6, 7, 8, 18, 19, 20 and 21 and 20 are used to express these proteins at the surface of PEAK and/or CHO cells as described in Example 2.
  • Non-transfected PEAK and/or CHO cells are used as negative control. Cells are harvested, counted, checked for viability and resuspended at 3 ⁇ 10 6 cells/ml in FACS buffer (PBS 2% BSA, 0.1% NaN 3 ).
  • CEACAM1, CEACAM6 and other CEACAMs can be characterized.
  • purified Mabs are incubated with cells expressing one of the CEACAM family proteins at a final concentration of 10 ⁇ g/ml for 30 minutes. After two washes, bound antibodies are detected using a Cy-5 conjugated anti-human Fc secondary antibody (BD biosciences).
  • An antibody according to the invention is found as non-binding to said CEACAM, if no bound antibody is detected by the PE-conjugated anti-human IgG Fc secondary antibody.
  • CEACAM5 monoclonal antibodies of the present invention can be tested by flow cytometry using PEAK CHO cells transfected with a vector expressing full-length cynomolgus CEACAM5.
  • Flow cytometry allows detecting if the said antibody is binding to the PEAK CHO cells expressing cynoCEACAM5 or if the said antibody is not binding to the PEAK CHO cells respectively non-binding to said CEACAM.
  • the simultaneous expression of one heavy chain and two lights chain in the same cell can lead to the assembly of three different antibodies. Simultaneous expression can be achieved in different ways such as that the transfection of multiple vectors expressing one of the chains to be co-expressed or by using vectors that drive multiple gene expression.
  • the vector encoding the different anti- CEACAM5 antibodies are co-transfected with another vector expressing the heavy and light chain of anti-CD47 antibody Ka3 (SEQ ID NO:5 and 11), an anti-CD47 antibody bearing the same common heavy chain and that is described in US 2014/0303354.
  • the two light chains are cloned into the vector pNovi KM, that is previously generated to allow for the co-expression of one heavy chain, one Kappa light chain and one Lambda light chain as described in US 2012/0184716 and WO 2012/023053, each of which is hereby incorporated by reference in its entirety.
  • the expression of the three genes is driven by human cytomegalovirus promoters (hCMV) and the vector also contains a glutamine synthetase gene (GS) that enables the selection and establishment of stable cell lines.
  • hCMV human cytomegalovirus promoters
  • GS glutamine synthetase gene
  • the common VH and the VL genes of the anti- CEACAM5 IgG and of the anti-CD47 IgG are cloned in the vector pNovi ⁇ H ⁇ , for transient expression in mammalian cells. Peak cells are cultured in appropriate Flask with suitable cells number and culture medium volume (containing fetal bovine serum). Plasmid DNA is transfected into the cells using Lipofectamine 2000) according to manufacturer's instructions. Antibody concentration in the supernatant of transfected cells is measured during the production using OctetRED96. According to antibody concentration, supernatants are harvested 5 to 7 days after transfection and clarified by centrifugation at 1300 g for 10 min. The purification process is composed of three affinity steps.
  • the FcXL affinity matrix (Thermofisher Scientific) is washed with PBS and then added in the clarified supernatant. After incubation overnight at +4° C., supernatants are centrifuged at 2000 g for 10 min, flow through is stored and resin washed twice with PBS. Then, the resin is transferred on Amicon Pro columns and a solution containing 50 mM glycine at pH 3.0 is used for elution. Several elution fractions are generated, pooled and desalted against PBS using 50 kDa AmiconTM Ultra Centrifugal filter units (Merck KGaA, Darmstadt, Germany).
  • the elueted product containing total human IgGs from the supernatant, is quantified using a Nanodrop spectrophotometer (NanoDrop Technologies, Wilmington, Del.) and incubated for 15 min at RT and 20 rpm with the appropriate volume of Kappa select affinity matrix (GE Healthcare). Incubation, resin recovery, elution and desalting steps are performed as described previously. The last affinity purification step is performed using the lambda Fab select affinity matrix (GE Healthcare) applying the same process as for the two previous purifications. The final product is quantified using the Nanodrop. Purified bispecific antibodies are analyzed by electrophoresis in denaturing and reducing conditions.
  • the Agilent 2100 Bioanalyzer is used with the Protein 80 kit as described by the manufacturer (Agilent Technologies, Santa Clara, Calif., USA). 4 ⁇ L of purified samples are mixed with sample buffer supplemented with dithiothreitol (DTT; Sigma Aldrich, St. Louis, Mo.). Samples are heated at 95° C. for 5 min and then loaded on the chip. All samples are tested for endotoxin contamination using the Limulus Amebocyte Lysate test (LAL; Charles River Laboratories, Wilmington, Mass.).
  • a dual targeting i antibody composed of one arm binding with high affinity to CEASCAM5 or to CEASCAM5 and CEACAM6, and a second arm binding with lower affinity to CD47-but sufficient to inhibit CD47/SIRP ⁇ upon CEASCAM5 or CEACAM5 and CEACAM6 co-engagement with CD47 should allow preferential inhibition of CD47 in cancer versus normal cells.
  • the binding affinity of the antibodies according to the invention to human CD47 can be evaluated by surface plasmon resonance technology using a Biacore T200 instrument.
  • the biotinylated human CD47 soluble recombinant protein can be captured on a streptavidin coated sensor chip (Series S Sensor Chip SA). Then a concentration series of the test antibody can be injected over the surface, with regeneration of the surface between each injection.
  • the binding affinity measured in repeated determinations was between 400 and 500 nM.
  • the CD47 binding arm of this antibody is the same as the CD47 binding arm of the CEAxCD47 bispecific antibodies of this invention. According to the knowledge of the inventors that same experiments performed with the CEAxCD47 bispecific antibodies of the invention will provide similar results within the standard deviation of such experiments.
  • FIG. 6 shows results with a TAAxCD47 bispecific antibody (TAA is not CEA) containing the same CD47 binding arm than the bispecific antibodies of this invention in comparison to the corresponding monovalent anti-CD47 antibody.
  • the detection of bound SIRP ⁇ cell-based assay monitoring the interaction of soluble SIRP ⁇ with human CD47 expressed at the surface of MKN45 is used for the detection of the blocking activity.
  • Dose-response experiments with bispecific antibodies according to the invention allow determination of an IC50 value.
  • MKN45 cancer cells expressing both CD47 and CEACAM5, are stained with CFSE violet to allow the imaging system (CX5) to detect the cells.
  • CX5 imaging system
  • 3,000 stained MKN45 cells per well are seeded in a 384 optical well plate (Costar) and incubated for 50 minutes with increased concentrations of bispecific antibodies of the invention (1.9 pM to 333 nM, in quadruplicates). Then, a fixe concentration of SIRP ⁇ —mouseFc premixed with anti-mouse IgG-Fc AF647 coupled antibody (Jackson Immunoresearch diluted 1:2000) is added at 50 ng/mL final.
  • Table 2 shows the potency of several CEAxCD47 bispecific antibodies at inhibiting CD47/SIRP ⁇ binding displaying a range of IC50, from 0.22 nM to 7 nM.
  • Epitope binning is a competitive immunoassay used to characterize the binding of antibodies according to the invention or e.g. the binding of the related anti-CEA (target protein) antibodies of the first binding part.
  • a competitive blocking profile of an antibody binding to the target protein is created against antibodies also binding to this target protein and for which the binding epitope has already been established/published. Competition to one of these reference antibodies indicate that the antibody has the same or a closely located epitope and they are “binned” together.
  • CEACAMS mAbs which are part of the bispecific antibodies of the present invention to compete with CEASCAM5 reference antibodies is tested by ELISA on recombinant human CEACAMS with the following reference antibodies carrying a mouse Fc region: SM3E, sequences of mAb derived from SM3E described in patent US20050147614A1, mAb produced using standard methods; MEDI, mAb derived from MEDI-565 described in patent WO2016036678A1; SAR, mAb derived from Mab2_VLg5VHg2 described in patent EP3199552A1; CH1A1A, mAb derived from CH1A1A-2F1 described in patent US20120251529 and by Klein et al in Oncoimmunology, 2017 Jan.
  • SM3E sequences of mAb derived from SM3E described in patent US20050147614A1, mAb produced using standard methods
  • MEDI mAb derived from MEDI-565 described
  • SM3E binds e.g. more to the N-terminal, cell membrane distal part of CEA, MEDI to the middle part and CH1A1A binds close to the membrane.
  • Biotinylated human CEACAM5 is coated at 0.5 ⁇ g/ml in a Streptavidin-coated 96-well plate and incubated with 10 ⁇ g/ml of the reference mAbs or an irrelevant mAb carrying a mouse Fc region for 1 hour.
  • the CEACAM5 mAbs (as bivalent monoclonal anti-CEA antibodies and not as respective CEAxCD47 bispecific antibodies) are added at 0.2 ⁇ g/ml for 1 hour at room temperature.
  • the plate is washed and the bound CEACAM5 mAbs are detected with an anti-human IgG(Fc)-HRP (Jackson ImmunoResearch). After washing, the plate is revealed with Amplex Red reagent.
  • the fluorescence signal is measured on a Synergy HT plate reader (Biotek).
  • Bin 1 means that the respective antibody competes with SM3E for binding to CEACAMS.
  • Bin 2 means, that the respective antibody does not compete with any of the tool antibodies mentioned above.
  • the competition experiments were for all of the CEAxCD47 bispecific antibodies listed in Table 2 performed with the respective anti-CEA bivalent monoclonal antibodies. In case binding of such a monoclonal antibody to CEASCAM5 was reduced by the respective tool antibody by 80% or more, it was concluded that the CEAxCD47 bispecific antibody is classified to bind competitively with the tool antibody.
  • a CEAxCD47 antibody is identified as non-competitive with a tool antibody in case binding of the respective anti-CEA bivalent mAb to CEASCAM5 is reduced by 20% or less if the results with and w/o addition of a tool antibody are compared.
  • Bin 1 K2AC13, K2AC18, K2AC23, K2AC27, K2AC29
  • Bin 2 K2AC10, K2AC25, K2AC28 K2AC26
  • Biotinylated recombinant human CEACAM5 or CEACAM6 proteins are captured at 0.5 ⁇ g/mL in a streptavidin coated 96-well microplate.
  • the plate is washed and monoclonal anti-CEA bivalent antibodies of the present invention are added as a broad concentration-range (e.g. from 5 ⁇ 10 4 to 1 ⁇ g/mL) and incubated during 1 hr.
  • the plate is washed and bound antibodies are detected with an anti-human IgG(Fc)-HRP (Jackson ImmunoResearch). After washing, the plate is revealed with Amplex Red reagent (Molecular Probes). The fluorescence signal is measured on a Synergy HT plate reader (Biotek).
  • results obtained for the monoclonal antibody AC39 are contained in table 5; this antibody shows balanced CEASCAM5 and CEACAM6 binding, that means EC50 for binding to CEASCAM5 and CEACAM6 are similar (range of the ratio of the EC50 for CEASCAM5 binding to CEACAM6 binding of balanced antibodies from 0.33 to 3). Antibodies with a ratio outside this range are considered as not balanced.
  • TAAxCD47 ⁇ antibodies The ability of dual targeting TAAxCD47 ⁇ antibodies to co-engage CD47 and TAA results in a significant increase in the affinity of binding to TAA-positive cells as compared to TAA-negative cells and in TAA -dependent neutralization of the CD47-SIRP ⁇ interaction. This, in turn, could translate into efficient and selective cancer cell killing mediated by CEAxCD47 ⁇ antibodies.
  • FIG. 3A results as demonstrated from ADCP experiments (flow cytometry based assay) shown in FIG. 3A demonstrate higher ADCP of bispecific TAAxCD47 antibody compared to the corresponding monovalent TAA as well as the monovalent CD47 antibody.
  • FIG. 4 is showing higher ADCC (Cr51 based assay) of four bispecific TAAxCD47 antibodies (TAA is mesothelin MSLN) compared to the high affinity anti-TAA monoclonal antibody Amatuximab (TAA is MSLN) (lung cancer NCI-H226 tumor cells carrying MSLN are used.
  • Healthy PBMC were activated overnight at 37° C. with RPMI/10% heat inactivated FCS supplemented with 10 ng/mL of recombinant hIL-2.
  • targets cells i.e. cancer cells expressing the TAA
  • 100 ⁇ Ci Cr51 Perkin Elmer, 37° C., 1h
  • cells were opsonized with test antibodies (30 min, 37° C.).
  • Cr51-loaded cancer cells were then mixed with PBMC cells to obtain the final 80:1 or 50:1 ratio between effector (PBMC) and target cells (TAA-expressing cells).
  • the cell mixture was incubated for 4 h at 37° C. before being centrifuged for 10 min at 1500 rpm.
  • FIG. 5 shows the results for ADCC (Cr 51 based assay) of comparison experiment between the wt IgG1 Fc version versus the additionally DEA mutated Fc version of a CD47xTAA bispecfic antibody (TAA not CEA) carrying the same CD47 arm as the CEAxCD47 antibodies of the invention. Also, the results for a high affinity anti-TAA bivalent mAb are shown. Highest ADCC of the TAAxCD47 bispecific antibody carrying IgG1 Fc with DEA mutations, followed by CEAxCD47 biAb with IgG1 Fc and by the bivalent mAb with the wt IgG1 Fc.
  • ADCC of the CEAxCD47 bispecific antibodies was tested in the following assay:
  • Healthy PBMC were activated overnight at 37° C. with RPMI/10% heat inactivated FCS supplemented with 10 ng/mL of recombinant hIL-2.
  • target cells e.g. MKN45 cancer cells
  • the PBMCs and the opsonized target cells are co-incubated at a ratio effector/target 50/1 in round bottom plates for 6 hours at 37° c in a cell culture incubator. After this incubation, supernatants are transferred into optical flat bottom plate and the LDH release is quantified with a commercial kit from Roche by measuring OD with a microplate reader.
  • the % of specific lysis is calculated with the following formula:
  • FIG. 13 shows the results of comparison experiments between bispecific antibodies of the invention with their glycoengineered forms and with antibody 5F9.
  • FIG. 14 shows the results of comparison experiments between bispecific antibodies of the invention with their glycoengineered forms and with antibody 5F9 in the presence of 1 mg/ml human Immunoglobulin (IgG).
  • IgG human Immunoglobulin
  • FIG. 3A shows results obtained with this FACS based assay for a TAAxCD47 antibody carrying the CD47 binding arm also used in the CEAxCD47 antibodies.
  • the imaging-based method which makes use of the Celllnsight CX5 High Content Screening Platform, the phagocytosis index, defined as the average number of target cells engulfed by 100 macrophages, is determined.
  • FIG. 3B shows results obtained with a TAAxCD47 bispecific antibody (TAA not CEA) carrying the CD47 binding arm of the CEAxCD47 antibodies.
  • TAAxCD47 bispecific antibody TAA not CEA
  • FIG. 12 shows results obtained with CEAxCD47 bispecific antibodies of the invention.
  • FIG. 15 shows the results of comparison experiments between bispecific antibodies of the invention with their glycoengineered forms and with anti-CD47 antibody 5F9.
  • FIG. 16 shows the results of comparison experiments between bispecific antibodies of the invention with their glycoengineered forms and with antibody 5F9 in the presence of 1 mg/ml human Immunoglobulin (IgG) as usually present in patients.
  • FIG. 17 shows the results of comparison experiments between bispecific antibodies of the invention in presence or not of 1 mg/ml human Immunoglobulin (hulgG, 1 mg/mL or even higher are present in patients).
  • FIG. 18 shows the concentration/phagocytosis index curves of K2AC22 in presence or not of 30 nM of CEA-TCB1 or 300 nM CEA-TCB.
  • Phagocytosis Assays 1. Imaging Assay Based on CellInsight CX5 High Content Screening Platform and 2. Flow Cytometry Based Assay
  • PBMCs Human peripheral blood mononuclear cells
  • M-CSF human macrophage colony-stimulating factor
  • Non-adherent cells are subsequently eliminated in the differentiation phase (day+1) by exchanging the cell culture medium, and adherent cells representing macrophages are detached using cell dissociation buffer (Sigma-Aldrich) and washed in complete medium the day of use (day8 or day9) for ADCP experiment based on cytometry.
  • cell dissociation buffer Sigma-Aldrich
  • Macrophages (stained with calcein red orange) adhering to microplate wells are co-incubated with Calcein AM-labeled target tumor cells at an effector : target cells ratio of 1:3 for 2.5 hours at 37 degree C. in the presence of different concentrations of the to be tested antibody.
  • supernatants are replaced by complete culture medium and the microplates are imaged with the CelllnsightTM CX5 High Content Screening Platform. 1500 macrophages are acquired and analyzed per well. Phagocytosis is evidenced as double-positive events (macrophage +target tumor cell) and the phagocytosis indexes are calculated by the CellInsightTM manufacturers' software.
  • phagocytosis index values shown in tables 3 and 4 are obtained with MKN-45 cells expressing CEA and with an effector cell to target/tumor cell ratio of 1:3.
  • the data in FIG. 3B are obtained with NCI-N87 cells carrying the TAA (which is not CEA) and with an effector to target/tumor cell ratio of 1:1.
  • TAAxCD47 bispecific antibody TAA not CEA
  • ADCP phagocytosis
  • ADCP can also be measured by a method as described as follows:
  • the macrophages are co-incubated with CSFE-labeled target tumor cells (e.g. MKN-45, LS174T or HPAC tumor cells) at an effector : target cells ratio of e.g. 3:1 for 2.5 hours at 37 degree C. in the presence of different concentrations of to be tested antibody.
  • target tumor cells e.g. MKN-45, LS174T or HPAC tumor cells
  • target cells ratio of e.g. 3:1 for 2.5 hours at 37 degree C. in the presence of different concentrations of to be tested antibody At the end of the incubation period, biotinylated anti-human CD14 antibody and Strep-Cy5 are added to label the macrophages.
  • the cells are then washed and subjected to flow cytometry analysis. Phagocytosis is evidenced by double-positive events CD14+and CFSE+.
  • Percentage of phagocytosis is presented as the ratio between CD14+/CSFE+double positive events and total target cells multiplied by 100.
  • the data in FIG. 3A are obtained with HPAC cells carrying the TAA (which is not CEA) and with an effector to target/tumor cell ratio of 1:1.
  • Flow cytometry based assay has been used.
  • the data in FIG. 3B are obtained with NCI-N87 cells carrying the TAA (which is not CEA) and with an effector to target/tumor cell ratio of 1:1. Imaging based assay was used.
  • Example 10 Binding of CEAxCD47 and CEAxCD3 to MNK-45 Cells; Competition of Binding with CEAxCD3 a) The Binding of CD47xCEACAM5 Bispecific Antibody is Tested on e.g. CEA-Expressing Human Gastric Adenocarcinoma Cells (MKN-45, DSMZ ACC 409).
  • Cells are harvested, counted, checked for viability and resuspended at 3 ⁇ 10 6 cells/ml in FACS buffer (PBS 2% BSA, 0.1% NaN3). 100 ⁇ l of the cell suspension are distributed in V-bottom 96-well plates (3 ⁇ 10 5 cells/well). The supernatant is removed by centrifugation 3 minutes at 4° C., 1300 rpm. Increasing concentrations of the antibody according to the invention are then added into the wells and incubated for 15 minutes at 4° C. Cells are washed twice with cold FACS buffer and re-incubated for further 15 minutes at 4° C.
  • FACS buffer PBS 2% BSA, 0.1% NaN3
  • FIG. 11 shows the binding curves of several CEAxCD47 bispecific antibodies to MKN-45 cells.
  • CD47xCEACAM5 bispecific antibody according to the invention and CEAxCD3 T-cell bispecific antibodies like CEA-TCB or CEA-TCB1 the binding of the CEACAM5xCD47 to MKN-45 cells can be determined as described above, but with and w/o addition of the CEAxCD3 T-cell bispecific antibody to study if a CEAxCD3 T-cell bispecific antibody as combination partner for the CEAxCD47 bispecific antibodies of this invention is competitive for binding to CEA or not.
  • CHO pool (one for K2AC5 and one for K2AC22) is inoculated at a viable cell concentration of 0.3 ⁇ 10 6 cells/mL in a Thomson erlen device with a working volume of 700 mL or 100 mL for the production of fucosylated and afucosylated antibodies, respectively. All the pools are operated in a 15 days duration fed-batch mode using CDACF medium CDCHO and an adapted feeding regime.
  • bolus of 200 ⁇ M fucose inhibitor (1,3,4-Tri-O-acetyl-2-deoxy-2-fluoro-L-fucose) are added at day 0, 5, 8 and 11 during the fed batch process based on afucosylation strategy described by Rillahan et al. Nature Chem. Biol. 2012 July; 8(7):661-8 and based on EP2282773.
  • Harvest of the K2AC5 and K2AC22 pools supernatants containing fucosylated or afucosylated antibodies is performed after 15 days of Fed batch culture.
  • Harvests of CHO pools supernatants are clarified using the Sartoclear Dynamics® Lab V Cell Harvesting Sartorius system (see supplier instructions).
  • Purification of fucosylated and afucosylated K2AC5 and K2AC22 bispecific antibodies is a three affinity step purification process. Before starting purification, antibody concentration in the supernatant of K2AC5 and K2AC 22 pools is measured using OctetRED96 in order to use columns with appropriate volume of affinity matrix. Each clarified CHO pool supernatant containing fucosylated or afucosylated bispecific antibodies, is loaded onto a MabSelect SuRe (MSS) column (GE Healthcare) without prior adjustment, to remove a major part of cell culture contaminants. The MSS eluate is then treated by low pH hold to inactivate viruses, and neutralized at pH 6 with Tris 1M pH9.
  • MSS MabSelect SuRe
  • the MSS eluate's is then loaded onto the LambdaFabSelect (LFS) column (GE Healthcare) to remove monospecific ⁇ (mono ⁇ ).
  • the LFS eluate is then pH adjusted at pH 6.
  • the LFS is loaded onto the Capto L (CL) column (GE Healthcare) to remove monospecific ⁇ (mono ⁇ ).
  • the CL Eluate is pH adjusted before storage.
  • the final material is then concentrated and diafiltered into the final formulation buffer, its concentration adjusted using the Nanodrop. Fucosylated and afucosylated K2AC5 and K2AC22 bispecific antibodies are aliquoted and stored at ⁇ 80° C. until delivery.
  • Purified bispecific antibodies are analyzed for sizing by electrophoresis in denaturing and reducing conditions with the Agilent 2100 Bioanalyzer using the Protein 80 kit as described by the manufacturer (Agilent Technologies, Santa Clara, Calif, USA). Aggregation level is assessed by size exclusion chromatography (SEC-UPLC) using the ACQUITY UPLC H-Class Bio System (Waters). Charge variant analysis of purified bispecific antibodies is achieved by isoelectric focusing technique (IEF) using the Multiphor II Electrophoresis System (GE Healthcare).
  • the relative distribution of N-linked complex biantennary glycoforms of fucosylated and afucosylated K2AC5 and K2AC22 antibodies is determined using the throughput microchip-CE method on the LabChip GXII Touch (Perkin Elmer). All antibodies are tested for endotoxin contamination using the Limulus Amebocyte Lysate test (LAL; Charles River Laboratories, Wilmington, Mass). Glycoengineered K2AC5 shows an afucosylation of 79.68% and glycoengineered K2AC22 shows an afucosylation of 89.13%.
  • afucosylated bispecific antibodies according to the invention can be produced also according to the method as follows:
  • RNA is isolated from CHO/DG44 cells using the RNeasy® Mini Kit (Qiagen, Hilden, Germany) and reverse transcribed with oligo-dT using a Superscript first-strand synthesis system for reverse transcript-polymerase chain reaction (RT-PCR) (Invitrogen, Carlsbad, Calif.).
  • a Chinese hamster FUT8 cDNA is amplified from single-stranded CHO/DG44 cell cDNAs by PCR using primers 5V-GTCTGAAGCATTATGTGTTGAAGC-3V (SEQ ID NO:14) and 5V-GTGAGTACATTCATTGTACTGTG-3V (SEQ ID NO:15), designed from the murine FUT8 cDNA (Hayashi, 2000; DNA Seq 11:91-96).
  • the targeted disruption of the FUT8 gene in CHO/DG44 cells is carried out using two replacement vectors, pKOFUT8Neo and pKOFUT8Puro.
  • the 9.0-kb fragment of the FUT8 gene including the first coding exon is isolated by screening the CHO-K1 cell E-genomic library (Stratagene, La Jolla, Calif.) with the Chinese hamster FUT8 cDNA as a probe to establish the targeting constructs.
  • a 234-bp segment containing the translation initiation site is replaced with the neomycin-resistance gene (Neor) cassette or the puromycin-resistance gene (Puror) cassette from plasmid pKOSelectNeo or pKOSelectPuro (Lexicon, Tex.), respectively, flanked by loxP sites.
  • the diphtheria toxin gene (DT) cassette from plasmid pKOSelectDT (Lexicon) is inserted at the 5V homologous region.
  • the resulting targeting constructs included the 1.5-kb 5V homologous sequence and the 5.3-kb 3V homologous sequence. Before transfection, the targeting constructs are linearized at a unique SalI site.
  • Subconfluent CHO/DG44 cells (1.6 106) are electroporated with 4 Ag of linearized pKOFUT8Neo at 350 V and 250 AF using a Bio-Rad GenePulser® II. After electroporation, transfectants are selected with 600 Ag/mL G418 (Nacalai Tesque, Kyoto, Japan). Genomic PCR is performed in 96-well plates by the modified microextraction method reported previously (Ramirez-Solis et al., 1992; Anal Biochem 201:331-335.) using the following primers:
  • Homologous recombinants are identified by the 1.7-kb fragment obtained using genomic PCR and confirmed by Southern blot analysis using the 221-bp fragment amplified with the following primers:
  • the hemizygous clone is subject to a second round of homologous recombination using linearized pKOFUT8Puro and drug selection with 15 Ag/mL puromycin (Sigma-Aldrich, St. Louis, Mo.) as described earlier.
  • the identified homozygous disruptants are electroporated with the Cre-recombinase expression vector pBS185 (Invitrogen) to remove drug-resistance gene cassettes from both FUT8 alleles.
  • FUT8(-) cell lines are electroporated with an expression vector encoding an bispecific antibody according to the invention and selected in media lacking hypoxanthine and thymidine.
  • the confluent transfectants are cultured in Ex-Cell® 301 Medium (JRH Biosciences, Lenexa, Kans.) for 1 week.
  • the antibody is purified from culture supernatants using MabSelectTM (Amersham Biosciences, Piscataway, N.J.). Further purification steps can be anion/cation exchange chromatography, size exclusion chromatography and especially purification using kappa respectively lambda selective resins as described above.
  • the anti-tumor activity of a bispecific antibody according to the invention can be evaluated in Xenograft models, e.g. by the following model: 1 or 2 ⁇ 10 6 CEA positive tumor cells like MKN-45 or LS174T cells are implanted subcutaneously in NOD/SCID mice. Tumor volumes are measured 3 times per week. After the tumor graft reached approx. 0.1cm 3 , mice are randomized into groups (e.g. 4 to 6 mice per group) and the antibody treatment is initiated. This experiment could e.g. compare the effect of the bispecific antibody according to the invention and positive control Mabs, e.g. the CD47 Mab B6H12.2 Antibody is injected e.g. i.v. every week until the end of the experiment (d25). Antibodies are administered at e.g. 50 to 1200 ⁇ g per mouse per injection.
  • Combinations of a bispecific antibody of this invention with a CEAxCD3 bispecific antibody can be tested in an appropriate model.
  • Models, in which the combination of an antibody according to the invention together with MAB CEA, MAB CEA1 or CEA-TCB CEA-TCB1 can be tested are e.g. described by Bacac et al (Clin. Cancer Res., 22(13);3286-97;2016) and are also used, especially for combination studies of CEACAM5xCD47 and CEA-TCB or CEA-TCB1.
  • Example 14 Cytokine Release Tested in Whole Blood and PBMCs from Healthy Human Donors Human Blood
  • an in vitro cytokine release assay can be performed using whole blood (WB CRA) with minimal dilution by the test antibodies (95% v/v blood) in aqueous presentation.
  • This assay format is considered to mimic more closely the in vivo environment, containing factors at physiological concentrations that may influence mechanisms of cytokine release.
  • this format is thought to be poorly predictive of T cell-mediated cytokine release (e.g., anti-CD28).
  • the assay can be also performed using peripheral blood mononuclear cells (PBMCs) from healthy human donors and with an immobilized mAb (Solid Phase, SP) presentation to assess T cell-mediated cytokine release (PBMC SP CRA).
  • PBMC SP CRA peripheral blood mononuclear cells
  • This assay format simulates cross-linking and high density presentation of mAbs, which may occur in vivo (e.g. clustering of the target via the interaction of the Fc part of the antibody with Fcy receptors on other immune cells or the cross-linking of mAbs by anti-drug antibodies).
  • This format is predictive of T cell-mediated cytokine release.
  • Negative controls as well as specific positive controls for each assay format can be tested in parallel to a TAAxCD47 antibody like a CD47xCEA bispecific antibody. see FIGS. 7A and B.
  • Example 15 Antibody Binding to Erythrocytes, Phagocytosis of Erythrocytes, and Platelet Activation and Aggregation
  • human whole blood samples collected from healthy donors in citrate can be mixed with 3 ⁇ g/mL of AF488-coupled CEA x CD47 bispecific antibodies of this invention, B6H12.2 or isotype control and surface staining antibodies (PE-Cy7 anti-hCD45 and PE anti-hCD41a, for platelets only) for 30 min at 4° C.
  • whole blood is divided in two samples: 5 ⁇ L are diluted and washed in PBS for erythrocyte analysis while 150 ⁇ L are incubated with erythrocyte lysing solution and washed for platelet analysis. Samples are acquired on a CytoFLEX instrument and analyzed with the FlowJo software to determine MFI values.
  • human red blood cells can be isolated from human whole blood by centrifugation at 300 ⁇ g, washed twice in PBS, labeled with CFSE-(Carboxyfluorescein succinimidyl ester) and pre-incubated with the test antibody for 1 hour at 37° C. before the addition of macrophages.
  • Labeled RBCs can be cultured with human macrophages in the presence of an antibody according to the invention or control (non-binding IgG1 antibody) for one hour at a target-to-effector ratio of 200:1. After culture, cells are stained with anti-CD14-APC and analyzed by flow cytometry.
  • Phagocytosis was quantitated as the percent of CD14+ events (macrophages) that are also CFSE+ and had therefore engulfed at least one RBC (events are gated on singlets). Phagocytosis and FACS analysis is done as described in example 9, except that the erythrocytes were lysed with FACS lysing solution after macrophage staining.
  • FIG. 8 shows that RBC phagocytosis for the IgG1 anti-CD47 monoclonal antibody B6H12.2 is much more potent than for an IgG1 TAA x CD47 (not CEAxCD47) bispecific antibodies containing the CD47 binding arm of the CEA x CD47 bispecific antibodies of this invention.
  • TAA x CD47 bispecific antibody with wildtype Fc showed no phagocytosis in the tested concentration range, if Fc carries the aa mutations DEA (S329D and I332E and G236A), phagocytosis is detected but at higher concentrations as for B6H12.2 antibody.
  • TAAxCD47 and CEAxCD47 bispecific antibodies were measured by the upregulation of surface marker CD62P. Briefly, 5 ⁇ L of whole blood is incubated with 10 ⁇ L of each sample (prepared at 2 ⁇ ) for 15 minutes at room temperature. Each tested antibody is added at different concentrations (0, 0.02, 0.2, 2, 20 and 200 ⁇ g/mL). Adenosine diphosphate (ADP) and anti-CD9 (ALB6), included as positive control reagents known to induce platelet activation, are added at a concentration of 10 ⁇ M and 10 ⁇ g/mL, respectively.
  • ADP Adenosine diphosphate
  • ARB6 anti-CD9
  • FIG. 10 shows results obtained in blood from seven volunteer donors. It was found that neither the anti-TAA monoclonal antibody nor the TAAxCD47 bispecific antibody induced relevant platelet activation (both with wt IgG1 Fc). In contrast the anti-CD47 antibody B6H12.2 with wt IgG1 Fc induced platelet activation and also the TAAxCD47 biAb with Fc carrying DEA mutations showed platelet activation.
  • CEAxCD47 antibodies K2AC5 and K2AC22 have been studied in the concentration range as shown in FIG. 10 for platelet activation in whole blood.
  • TAAxCD47 In the blood of 6 from 7 donors no significant platelet activation was seen, like shown for TAAxCD47 in FIG. 10 .
  • One donor showed already with positive control agents uncommon platelet activation and then also some platelet activation with K2AC5 and 22. Results of this one donor were disregarded due to uncommon platelet activation.
  • the potential for aggregation in the presence of CD47/CEA bispecific antibody could be assessed on platelet rich plasma (PRP).
  • PRP platelet rich plasma
  • PRP is challenged with ADP at 10 ⁇ M and 5 ⁇ M or with the test articles at 200, 100, 20, 25, and 12.5 ⁇ g/mL, as well as with saline or the isotype control.
  • Platelet aggregation can be evaluated throughout platelet stimulation (i.e. 10 min) with a Thrombo-aggregometer TA 4V under constant stirring.
  • Thrombosoft 1.6 software SD Innovation, Frouard, France) can be used for analysis of the data.
  • cynomolgus monkey cross-reactive antibodies could be tested in vivo in Cynomolgus Monkeys for any effect on hematology parameters (including RBC and platelets).
  • An antibody according to the invention is e.g. given to cynomolgus monkeys per intravenous route, at doses up to 100 mg/kg, on a weekly basis.
  • Hematology parameters including red blood cell and platelet counts, are monitored over time and compared to control values in monkeys (pre-dose values). Hematology parameters are determined by routine methods.
  • results in FIG. 9 have been obtained with an IgG1 TAA x CD47 (not CEAxCD47) bispecific antibody containing the CD47 binding arm of the CEA x CD47 bispecific antibodies according to this invention.
  • IgG4 anti-CD47 antibody hu5F9-G4 Jie Liu et al (Open access article, PLOS ONE 10(9) September 2015) showing dose dependent decrease of hemoglobin starting already at single doses around 1 mg/kg.
  • IgG4 format was used to minimize effects on red blood cells and platelets, compared to IgG1 format. Despite of this measure even at already rather low doses of 1 mg/kg and less, dose dependent reductions of e.g. hemoglobin are observed in cynomolgus monkeys.
  • Blood withdrawals are scheduled according to the experimental protocol at multiple time points, e,g, 0.25, 1, 4, 8, 24, 48, 72, 96, 120, 168, 240, 336, 504 (day 22), 672 (day 29), 840 (day 36), 1008 (day 43), 1176 (day 50) and 1344 h (day 57) after the intravenous administration of the bispecific antibody.
  • Blood samples of approximately 2 mL per animal and time-point are collected. Concentrations of the antibodies were either measured in serum or in plasma. An ELISA test is developed and validated to measure the concentrations. Each sample is measured in duplicates.
  • PK parameters like Cmax, clearance, elimination half-life, area under the curve etc. can be determined by using industry standard software (Phoenix WinNonlin; non-compartmental analysis).
  • MKN45 cells used as target cells are stained with calcein AM.
  • concentrations of tested antibodies are incubated or not with a fixed dose (200ng/mL) of commercial shed CEA (BioRad). After this incubation the stained MKN-45 are opsonized for 20 minutes at room temperature with the antibodies previously mixed with shed CEA. Then macrophages (stained with calcein red orange) adhering to microplate wells are co-incubated with the opsonized labeled target tumor cells at an effector : target cells ratio of 1:3 for 2.5 hours at 37° C.
  • the ADCP is performed in a presence of 1 g/mL of human IgG.
  • Example 19 ADCP Mediated by Bispecific Antibodies in Presence of CEA-TCB and CEA-TCB1
  • Calcein AM-labeled MKN45 cells used as target cells are pre incubated or not with a fixed dose of CEA-TCB (300 nM) or CEA-TCB1 (30 nM) for 20 min at RT. After this incubation different concentrations of tested antibody are added in appropriate well for 20 min. Then macrophages (stained with calcein red orange) adhering to microplate wells are co-incubated with the opsonized labeled target tumor cells at an effector:target cells ratio of 1:3 for 2.5 hours at 37 ° C.
  • the ADCP is performed in a presence of 1 mg/mL of human hIgG.
  • FIG. 18 shows that neither CEA-TCB nor CEA-TCB1 added decreases phagocytosis induced by K2AC22. Surprisingly phagocytosis of K2AC22 was even slightly increased by the addition of 30 nM CEA-TCB1.
  • PBMCs Human peripheral blood mononuclear cells
  • a CEAxCD3 T-cell bispecific antibody at certain concentrations together with certain concentrations of of aCEAxCD47 bispecific antibody.
  • Opsonized targets were added to the plates containing macrophages and autologous PBMCs; and the plates were incubated at 37° C. for 48 h. After 48 h, half of the well medium was removed and a solution of 2 ⁇ Luciferin was added to the plates to obtain a final concentration of 150 ⁇ g/mL. After 5 minutes incubation at RT, plates were read using a Synergy NEO. Percentage of viability was calculated dividing the luminescence value (minus background) by the control containing only target cells and multiplying by 100. Percentage of killing was then extrapolated by subtracting the percentage of viability to 100.
  • FIGS. 19A and B show results obtained with combinations of CEA-TCB and K2AC5 and K2AC22 at various concentrations.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US15/734,962 2018-06-03 2019-05-31 Bispecific antibodies against ceacam5 and cd47 Abandoned US20210221908A1 (en)

Applications Claiming Priority (17)

Application Number Priority Date Filing Date Title
EP18175658.6 2018-06-03
EP18175657.8 2018-06-03
EP18175655.2 2018-06-03
EP18175656.0 2018-06-03
EP18175658 2018-06-03
EP18175655 2018-06-03
EP18175656 2018-06-03
EP18175657 2018-06-03
EP18188788.6 2018-08-13
EP18188792 2018-08-13
EP18188790.2 2018-08-13
EP18188792.8 2018-08-13
EP18188790 2018-08-13
EP18188788 2018-08-13
EP18190983.9 2018-08-27
EP18190983 2018-08-27
PCT/IB2019/054559 WO2019234576A1 (fr) 2018-06-03 2019-05-31 Anticorps bispécifiques dirigés contre ceacam5 et cd47

Publications (1)

Publication Number Publication Date
US20210221908A1 true US20210221908A1 (en) 2021-07-22

Family

ID=66752156

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/734,962 Abandoned US20210221908A1 (en) 2018-06-03 2019-05-31 Bispecific antibodies against ceacam5 and cd47
US16/428,539 Active 2040-11-22 US11555071B2 (en) 2018-06-03 2019-05-31 Bispecific antibodies against CEACAM5 and CD47
US18/063,477 Pending US20240084001A1 (en) 2018-06-03 2022-12-08 Bispecific antibodies against ceacam5 and cd47

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/428,539 Active 2040-11-22 US11555071B2 (en) 2018-06-03 2019-05-31 Bispecific antibodies against CEACAM5 and CD47
US18/063,477 Pending US20240084001A1 (en) 2018-06-03 2022-12-08 Bispecific antibodies against ceacam5 and cd47

Country Status (9)

Country Link
US (3) US20210221908A1 (fr)
EP (1) EP3802599B1 (fr)
JP (1) JP7403479B2 (fr)
KR (1) KR20210029158A (fr)
CN (1) CN112566938A (fr)
AU (1) AU2019281358A1 (fr)
CA (1) CA3102398A1 (fr)
MX (1) MX2020013122A (fr)
WO (1) WO2019234576A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11555071B2 (en) * 2018-06-03 2023-01-17 Lamkap Bio Beta Ltd. Bispecific antibodies against CEACAM5 and CD47

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3831849A1 (fr) * 2019-12-02 2021-06-09 LamKap Bio beta AG Anticorps bispécifiques contre ceacam5 et cd47
WO2021124073A1 (fr) * 2019-12-17 2021-06-24 Pfizer Inc. Anticorps spécifiques pour cd47, pd-l1, et leurs utilisations
CN111018969A (zh) * 2019-12-27 2020-04-17 上海药明生物技术有限公司 采用轻链select联合层析色谱纯化双特异性抗体的方法
PE20231386A1 (es) 2020-12-18 2023-09-12 Lamkap Bio Beta Ltd Anticuerpos biespecificos contra ceacam5 y cd47
KR20220125394A (ko) 2021-03-05 2022-09-14 주식회사 엘지에너지솔루션 전지 팩 및 이를 포함하는 디바이스
CA3212599A1 (fr) * 2021-03-22 2022-09-29 Novimmune S.A. Anticorps bispecifiques ciblant cd47 et pd-l1 et leurs methodes d'utilisation
CN115819596A (zh) * 2021-09-17 2023-03-21 优迈生物科技(连云港)有限公司 一种靶向人ceacam5/6的抗体、制备方法和应用
WO2023070353A1 (fr) * 2021-10-27 2023-05-04 Adagene Pte. Ltd. Anticorps anti-cd47 et leurs procédés d'utilisation
US20240002544A1 (en) * 2022-03-07 2024-01-04 Novimmune Sa Cd28 bispecific antibodies for targeted t cell activation
WO2023242351A1 (fr) 2022-06-16 2023-12-21 Lamkap Bio Beta Ag Polythérapie d'anticorps bispécifiques dirigés contre ceacam5 et cd47 et anticorps bispécifiques dirigés contre ceacam5 et cd3

Family Cites Families (107)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US20020142000A1 (en) 1999-01-15 2002-10-03 Digan Mary Ellen Anti-CD3 immunotoxins and therapeutic uses therefor
ES2568899T3 (es) 1999-04-09 2016-05-05 Kyowa Hakko Kirin Co., Ltd. Procedimiento para controlar la actividad de una molécula inmunofuncional
WO2002030077A1 (fr) 2000-10-05 2002-04-11 Matsushita Electric Industrial Co., Ltd. Procede d'etablissement du niveau de decision et recepteur de donnees
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US20020165360A1 (en) 2000-11-30 2002-11-07 Junghans Richard P. Chimeric effector cell receptors against carcinoembryonic antigen
HUP0700103A3 (en) 2001-08-03 2012-09-28 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
CN100423777C (zh) 2001-10-25 2008-10-08 杰南技术公司 糖蛋白组合物
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
EP2359685B1 (fr) 2001-12-27 2013-12-04 GlycoFi, Inc. Procede d'ingenierie de structures de sucres typiques des mammiferes
US7657380B2 (en) 2003-12-04 2010-02-02 Xencor, Inc. Methods of generating variant antibodies with increased host string content
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US20050031613A1 (en) 2002-04-09 2005-02-10 Kazuyasu Nakamura Therapeutic agent for patients having human FcgammaRIIIa
AU2003236020B2 (en) 2002-04-09 2009-03-19 Kyowa Hakko Kirin Co., Ltd. Cell with depression or deletion of the activity of protein participating in GDP-fucose transport
CN1930288B (zh) 2002-04-09 2012-08-08 协和发酵麒麟株式会社 基因组被修饰的细胞
WO2003085118A1 (fr) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Procede de production de composition anticorps
EP1498491A4 (fr) 2002-04-09 2006-12-13 Kyowa Hakko Kogyo Kk Procede d'amelioration de l'activite d'une composition d'anticorps de liaison avec le recepteur fc gamma iiia
IL149820A0 (en) 2002-05-23 2002-11-10 Curetech Ltd Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
US7232888B2 (en) 2002-07-01 2007-06-19 Massachusetts Institute Of Technology Antibodies against tumor surface antigens
DE60333201D1 (de) 2002-09-12 2010-08-12 Greenovation Biotech Gmbh Verfahren zur herstellung von proteinen
CA2549932C (fr) 2002-12-20 2013-08-20 Greenovation Biotech Gmbh Ameliorations apportees ou associees a la production de proteines
AU2003288675B2 (en) 2002-12-23 2010-07-22 Medimmune Limited Antibodies against PD-1 and uses therefor
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8367374B2 (en) 2003-01-22 2013-02-05 Roche Glycart Ag Fusion constructs and use of same to produce antibodies with increased Fc receptor binding affinity and effector function
WO2005018669A1 (fr) 2003-08-18 2005-03-03 Macrogenics, Inc. Anticorps specifiques des proteines fc?riib et procedes d'utilisation associes
AU2004266159A1 (en) 2003-08-22 2005-03-03 Biogen Idec Ma Inc. Improved antibodies having altered effector function and methods for making the same
WO2005063815A2 (fr) 2003-11-12 2005-07-14 Biogen Idec Ma Inc. Variants de polypeptides de liaison au recepteur fc$g(g) et procede apparentes
US20050266425A1 (en) * 2003-12-31 2005-12-01 Vaccinex, Inc. Methods for producing and identifying multispecific antibodies
WO2005092925A2 (fr) 2004-03-24 2005-10-06 Xencor, Inc. Variantes d'immunoglobuline a l'exterieur de la region fc
WO2005110474A2 (fr) 2004-05-10 2005-11-24 Macrogenics, Inc. ANTICORPS SPÉCIFIQUES FcϜRIIB HUMANISÉS ET MÉTHODES D’UTILISATION
WO2006085967A2 (fr) 2004-07-09 2006-08-17 Xencor, Inc. Anticorps monoclonaux optimises anti-cd20 a variants fc
WO2006019447A1 (fr) 2004-07-15 2006-02-23 Xencor, Inc. Variantes genetiques de fc optimisees
JP2008510008A (ja) 2004-08-16 2008-04-03 メディミューン,インコーポレーテッド 抗体依存性細胞性細胞傷害活性が増強されたインテグリンアンタゴニスト
WO2006047350A2 (fr) 2004-10-21 2006-05-04 Xencor, Inc. Variants d'immunoglobuline igg a fonction effectrice optimisee
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
CA2602663A1 (fr) 2005-03-31 2006-10-05 Xencor, Inc. Variants fc presentant des proprietes optimisees
CA2606102C (fr) 2005-04-26 2014-09-30 Medimmune, Inc. Modulation de la fonction d'effecteur d'anticorps par ingenierie de domaine "charniere"
CN109485727A (zh) 2005-05-09 2019-03-19 小野药品工业株式会社 程序性死亡-1(pd-1)的人单克隆抗体及使用抗pd-1抗体来治疗癌症的方法
MX2007015942A (es) 2005-07-01 2008-03-07 Medarex Inc Anticuerpos monoclonales humanos para ligandos 1 (pd-l1) de muerte programada.
US7557190B2 (en) 2005-07-08 2009-07-07 Xencor, Inc. Optimized proteins that target Ep-CAM
SI1919503T1 (sl) 2005-08-10 2015-02-27 Macrogenics, Inc. Identifikacija in inĺ˝eniring protiteles z variantnimi fc regijami in postopki za njih uporabo
EP1931709B1 (fr) 2005-10-03 2016-12-07 Xencor, Inc. Variants de fc dotés de propriétés de liaison aux récepteurs fc optimisées
CA2625998C (fr) 2005-10-06 2015-12-01 Xencor, Inc. Anticorps anti-cd30 optimises
JP5686953B2 (ja) 2005-10-11 2015-03-18 アムゲン リサーチ (ミュンヘン) ゲーエムベーハー 交差種特異的(cross−species−specific)抗体を含む組成物および該組成物の使用
DK1945666T3 (da) 2005-10-21 2013-07-01 Genzyme Corp Antistoffer med forøget antistofafhængig cellulær cytotoksicitetsaktivitet, fremgangsmåder til fremstilling af disse og anvendelse heraf
NZ569204A (en) 2005-12-21 2012-03-30 Micromet Ag Pharmaceutical compositions with resistance to soluble CEA - comprising antibodies that bind to CD3 and CEA
AU2007226752A1 (en) 2006-03-10 2007-09-20 Macrogenics, Inc. Identification and engineering of antibodies with variant heavy chains and methods of using same
IL177158A0 (en) 2006-07-30 2008-01-20 Assaf Klar Method for designing and implementing improved longitudinal flexibility multilayered pipeline
WO2008022152A2 (fr) 2006-08-14 2008-02-21 Xencor, Inc. Anticorps optimisés ciblant cd19
AU2007299843B2 (en) 2006-09-18 2012-03-08 Xencor, Inc Optimized antibodies that target HM1.24
WO2008121160A2 (fr) 2006-11-21 2008-10-09 Xencor, Inc. Anticorps optimisés qui ciblent cd5
US8652466B2 (en) 2006-12-08 2014-02-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
WO2008091798A2 (fr) 2007-01-22 2008-07-31 Xencor, Inc. Anticorps de ca9 optimises et methodes d'utilisation associees
AU2008207898B2 (en) 2007-01-23 2012-05-03 Xencor, Inc Optimized CD40 antibodies and methods of using the same
WO2008092117A2 (fr) 2007-01-25 2008-07-31 Xencor, Inc. Nouvelles insertions, délétions et substitutions d'immunoglobulines
WO2008098115A2 (fr) 2007-02-07 2008-08-14 Xencor, Inc. Anti-corps igf-1r optimisés et procédés utilisant ceux-ci
WO2008119566A2 (fr) 2007-04-03 2008-10-09 Micromet Ag Éléments de liaison bispécifiques spécifiques d'espèces croisées
NZ580755A (en) 2007-04-03 2012-05-25 Micromet Ag Cross-species-specific cd3-epsilon binding domain
BRPI0809594A2 (pt) 2007-04-03 2019-08-27 Micromet Ag polipeptídeo, seqüência de ácido nucléico, vetor, hospedeiro, processo para a produção de um polipeptídeo, composição farmacêutica, uso de um polipeptídeo, método para prevenção, tratamento ou melhora de uma doença, em um indivíduo com necessidade do mesmo, kit, método para a identificação de um polipeptídeo(s)
EP3392273A1 (fr) 2007-05-30 2018-10-24 Xencor, Inc. Procédés et compositions pour inhiber des cellules exprimant cd32b
DK2170959T3 (da) 2007-06-18 2014-01-13 Merck Sharp & Dohme Antistoffer mod human programmeret dødsreceptor pd-1
ES2639857T3 (es) 2008-02-11 2017-10-30 Cure Tech Ltd. Anticuerpos monoclonales para el tratamiento del tumor
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US8163551B2 (en) 2008-05-02 2012-04-24 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
AU2009288289B2 (en) 2008-08-25 2012-11-08 Amplimmune, Inc. PD-1 antagonists and methods of use thereof
CA2734908A1 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Compositions d'antagonistes de pd-1 et methodes d'utilisation associees
ES2742419T3 (es) 2008-09-17 2020-02-14 Xencor Inc Nuevas composiciones y métodos para tratar trastornos mediados por IgE
SI2352763T2 (sl) 2008-10-01 2022-11-30 Amgen Research (Munich) Gmbh Bispecifična enoverižna protitelesa s specifičnostjo za tarčne antigene z visoko molekulsko maso
SI2356153T1 (sl) 2008-10-01 2016-07-29 Amgen Research (Munich) Gmbh Bispecifično enoverižno protitelo, specifično za križane vrste
EP2352765B1 (fr) 2008-10-01 2018-01-03 Amgen Research (Munich) GmbH Anticorps monocaténaire bispécifique à domaine unique, spécifique d'espèces croisées
TWI686405B (zh) 2008-12-09 2020-03-01 建南德克公司 抗pd-l1抗體及其於增進t細胞功能之用途
CA2774260C (fr) 2009-09-16 2018-10-09 Immunomedics, Inc. Anticorps anti-cea de classe i et leurs utilisations
JP5997154B2 (ja) 2010-08-16 2016-09-28 ノビミューン エスアー 多重特異性多価抗体の生成方法
UA115030C2 (uk) 2011-03-02 2017-09-11 Рош Глікарт Аг Антитіло, яке зв'язується зі зв'язаним з мембраною карциноембріональним антигеном (сеа)
US20140242081A1 (en) 2011-07-18 2014-08-28 Micromet Ag Dosing regimens for treatment of cea-expressing cancers
KR102049817B1 (ko) 2011-08-01 2019-12-02 제넨테크, 인크. Pd-1 축 결합 길항제 및 mek 억제제를 사용하는 암 치료 방법
PT2768857T (pt) 2011-10-19 2020-01-27 Novimmune Sa Métodos para purificar anticorpos
US20140140989A1 (en) 2012-02-06 2014-05-22 Inhibrx Llc Non-Platelet Depleting and Non-Red Blood Cell Depleting CD47 Antibodies and Methods of Use Thereof
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
RU2015117393A (ru) * 2012-10-08 2016-12-10 Роше Гликарт Аг Лишенные fc антитела, содержащие два Fab-фрагмента, и способы их применения
ES2625742T3 (es) 2012-11-20 2017-07-20 Sanofi Anticuerpos anti-CEACAM5 y usos de los mismos
ES2784631T3 (es) * 2012-12-03 2020-09-29 Novimmune Sa Anticuerpos anti-CD47 y métodos de uso de los mismos
EP2945969A1 (fr) 2013-01-15 2015-11-25 Xencor, Inc. Elimination rapide de complexes antigéniques à l'aide de nouveaux anticorps
MA38308B1 (fr) 2013-02-26 2022-09-30 Hoffmann La Roche Molécules de liaison à l'antigène bispécifiques activant des lymphocytes t
CA2910466A1 (fr) 2013-04-29 2014-11-06 The Board Of Trustees Of The Leland Stanford Junior University Utilisation d'agents anti-cd47 pour ameliorer l'immunisation
RU2705795C2 (ru) 2013-08-20 2019-11-12 Мерк Шарп И Доум Корп. Лечение рака комбинацией антагониста pd-1 и динациклиба
ES2728668T3 (es) 2014-01-08 2019-10-28 Univ Leland Stanford Junior Terapia dirigida para el cáncer de pulmón de células pequeñas
CA2936244A1 (fr) 2014-01-21 2015-07-30 Medimmune, Llc Compositions et procedes pour moduler et reorienter des reponses immunitaires
EP3166974A1 (fr) 2014-07-11 2017-05-17 Genentech, Inc. Anticorps anti-pd-l1 et leurs utilisations
WO2016036678A1 (fr) 2014-09-02 2016-03-10 Medimmune, Llc Formulations d'anticorps bispécifiques
TWI595006B (zh) * 2014-12-09 2017-08-11 禮納特神經系統科學公司 抗pd-1抗體類和使用彼等之方法
KR102318994B1 (ko) 2015-01-23 2021-10-29 헬릭스 바이오파마 코포레이션 치료학적 목적을 위한 항체-우레아제 접합체
RU2739163C2 (ru) * 2015-03-23 2020-12-21 Байер Фарма Акциенгезельшафт Анти-сеасам6 антитела и их применения
CA2981204C (fr) 2015-03-31 2023-09-19 Novimmune Sa Procede d'optimisation de l'assemblage et de la production de complexes proteiques hetero-multimeres
DE102015212888A1 (de) 2015-07-09 2017-01-12 BSH Hausgeräte GmbH Geschirrspülmaschine mit einer Trocknungseinrichtung
CN108350048B (zh) * 2015-08-07 2024-02-09 Alx肿瘤生物技术公司 具有SIRP-α结构域或其变体的构建体
CN107849137B (zh) 2015-10-02 2021-11-26 豪夫迈·罗氏有限公司 双特异性抗ceaxcd3 t细胞活化性抗原结合分子
EP3165536A1 (fr) 2015-11-09 2017-05-10 Ludwig-Maximilians-Universität München Molécule de combinaison de ciblage de tumeur spécifique et d'inhibition de point de contrôle immunitaire local
GB2546072B (en) 2016-01-05 2019-07-10 Bja Trading Ltd Attachment assembly and method of using same
IL259588B2 (en) 2016-01-08 2023-09-01 Hoffmann La Roche Methods for the treatment of cea-positive cancer using pd-1 spindle-binding antagonists and bispecific antibodies against anti-cea and anti-cd3
CN114716552B (zh) * 2016-01-11 2024-05-24 四十七公司 人源化、小鼠或嵌合抗cd47单克隆抗体
US20200255515A1 (en) 2016-05-09 2020-08-13 Celgene Corporation Cd47 antibodies and methods of use thereof
CN107556386A (zh) * 2016-06-30 2018-01-09 中国科学院深圳先进技术研究院 抗EGFRvIII和CD3特异性双靶向抗体、含双靶向抗体表达盒的微环DNA及应用
GB201611530D0 (en) * 2016-07-01 2016-08-17 Alligator Bioscience Ab Novel polypeptides
WO2018098384A1 (fr) * 2016-11-22 2018-05-31 Regents Of The University Of California Modulation par ségrégation pour immunothérapie
CA3102398A1 (fr) * 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Anticorps bispecifiques diriges contre ceacam5 et cd47
PE20231386A1 (es) * 2020-12-18 2023-09-12 Lamkap Bio Beta Ltd Anticuerpos biespecificos contra ceacam5 y cd47

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11555071B2 (en) * 2018-06-03 2023-01-17 Lamkap Bio Beta Ltd. Bispecific antibodies against CEACAM5 and CD47

Also Published As

Publication number Publication date
CN112566938A (zh) 2021-03-26
US11555071B2 (en) 2023-01-17
EP3802599C0 (fr) 2023-12-20
US20200123252A1 (en) 2020-04-23
EP3802599B1 (fr) 2023-12-20
AU2019281358A1 (en) 2021-01-07
EP3802599A1 (fr) 2021-04-14
CA3102398A1 (fr) 2019-12-12
JP2021525544A (ja) 2021-09-27
WO2019234576A1 (fr) 2019-12-12
KR20210029158A (ko) 2021-03-15
JP7403479B2 (ja) 2023-12-22
MX2020013122A (es) 2021-04-13
US20240084001A1 (en) 2024-03-14

Similar Documents

Publication Publication Date Title
US11555071B2 (en) Bispecific antibodies against CEACAM5 and CD47
CA2959171C (fr) Agents de liaison cd123 et leurs utilisations
US20190270826A1 (en) Anti-il1rap antibodies, bispecific antigen binding molecules that bind il1rap and cd3, and uses thereof
US10407501B2 (en) Humanized or chimeric CD3 antibodies
EP3831849A1 (fr) Anticorps bispécifiques contre ceacam5 et cd47
CA3101271A1 (fr) Anticorps anti-cd3 et leurs utilisations
US11753481B2 (en) Bispecific antibodies against CEACAM5 and CD47
CA3150265A1 (fr) Anticorps bispecifiques diriges contre ceacam5 et cd3
CN113924118A (zh) 在低ph下具有增强的结合特异性的伊匹单抗变体
EP4190813A1 (fr) Anticorps contre le cd47 et son application
RU2798246C2 (ru) Биспецифические антитела против ceacam5 и cd47
CN114641500B (zh) 使用抗ox40抗体与抗tim3抗体的组合治疗癌症的方法
WO2023242351A1 (fr) Polythérapie d'anticorps bispécifiques dirigés contre ceacam5 et cd47 et anticorps bispécifiques dirigés contre ceacam5 et cd3
WO2023052541A1 (fr) Combinaison d'un anticorps d'activation anti-btn3a et d'un agoniste il-2 en vue d'une utilisation en thérapie
TW202327649A (zh) Pvrig/tigit結合蛋白聯合免疫檢查點抑制劑用於治療癌症

Legal Events

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

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

STCB Information on status: application discontinuation

Free format text: ABANDONED -- INCOMPLETE APPLICATION (PRE-EXAMINATION)