US20210292418A1 - Antibodies - Google Patents

Antibodies Download PDF

Info

Publication number
US20210292418A1
US20210292418A1 US17/204,604 US202117204604A US2021292418A1 US 20210292418 A1 US20210292418 A1 US 20210292418A1 US 202117204604 A US202117204604 A US 202117204604A US 2021292418 A1 US2021292418 A1 US 2021292418A1
Authority
US
United States
Prior art keywords
region
seq
antibody
amino acid
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.)
Pending
Application number
US17/204,604
Other languages
English (en)
Inventor
Louise KOOPMAN
Patrick Engelberts
Dennis VERZIJL
Edward N. Van Den Brink
Rik RADEMAKER
Sieto BOSGRA
Frederikke L. EGEROD
David Satijn
Esther C. W. BREIJ
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.)
Genmab AS
Original Assignee
Genmab AS
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 Genmab AS filed Critical Genmab AS
Publication of US20210292418A1 publication Critical patent/US20210292418A1/en
Priority to US17/494,545 priority Critical patent/US11261254B1/en
Assigned to GENMAB A/S reassignment GENMAB A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RADEMAKER, Rik, ENGELBERTS, PATRICK, SATIJN, DAVID, BOSGRA, Sieto, VERZIJL, Dennis, KOOPMAN, Louise, EGEROD, Frederikke L., BREIJ, Esther C. W., VAN DEN BRINK, EDWARD N.
Priority to US17/695,550 priority patent/US20220324980A1/en
Pending 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
    • C07K16/2803Immunoglobulins [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
    • C07K16/2827Immunoglobulins [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 B7 molecules, e.g. CD80, CD86
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies binding to B7H4, in particular to bispecific antibodies binding to B7H4 and CD3.
  • the invention further provides pharmaceutical compositions comprising the antibodies and use of the antibodies for therapeutic and diagnostic procedures, in particular in cancer therapy.
  • B7H4 (B7-H4, V-set domain containing T cell activation inhibitor 1 or VTCN1) is a member of the B7 family of proteins, which family comprises cell-surface protein ligands that bind to receptors on lymphocytes.
  • the B7 family plays an important role in the regulation of immune responses.
  • B7H4 negatively regulates T cell-mediated immune responses by inhibiting T cell activation, proliferation, cytokine production and cytotoxic activity (Prasad et al., 2003, Immunity 18: 863-873).
  • B7H4 is a type I transmembrane protein that includes a short intracellular domain, a hydrophobic transmembrane domain, and an extracellular domain with an IgV- and an IgC-like domain with four conserved cysteine residues and seven sites for N-linked glycosylation. (Sica et al., 2003, Immunity 18: 849-861). To date, no receptor for B7H4 has been identified.
  • B7H4 expression is very limited, whereas B7H4 expression is found on tumor cells in numerous cancer tissues (Kaur and Janakiram, 2019, ESMO Open 4:e000554).
  • B7H4 expression is correlated with advanced stages of cancer, poor prognosis, and decreased overall patient survival.
  • B7H4 binding antibodies are in development for cancer therapy.
  • FPA150 is an afucosylated human antibody that relieves the B7H4-mediated suppression of T cell activation and exhibits antibody dependent cellular cytotoxicity (ADCC) activity (Wainberg et al., 2019, Annals of Oncology 30, Suppl. 5, v489 (1198P). It is currently in early clinical trials as a monotherapy or in combination with pembrolizumab in advanced solid tumors.
  • Fab scFv single-chain variable fragments
  • the antigen-binding regions of such antibodies comprise at least human framework regions, such as e.g FR1, FR2, FR3 and FR4. Most preferred is that all framework regions are human.
  • Such antigen-binding regions are humanized and/or human antigen-binding regions.
  • These antibodies are useful in the treatment of conditions wherein specific targeting and T cell mediated killing of B7H4 expressing cells is desired, e.g. in conditions such as cancer.
  • such an antibody is suitable for human use, e.g. in a medical treatment.
  • Cancers that may be suitable for treatment are solid tumors.
  • Said B7H4 expression, and T cell mediated killing, e.g. in cancer cells may range in accordance with the invention from relatively low expression of B7H4, such as in MCF-7 cells, to relatively high expression of B7H4, such as in SK-BR3 cells, as shown e.g. in example 12.
  • B7H4 expression, and T cell mediated killing may range in accordance with the invention from relatively low expression of B7H4, such as in MCF-7 cells, to relatively high expression of B7H4, such as in SK-BR3 cells, as shown e.g. in example 12.
  • More preferably such bispecific antibodies have substitutions within the constant region that renders the Fc region, if present, inert.
  • a bispecific antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region that is capable of binding to CD3, such as human CD3 ⁇ (epsilon), wherein the antigen-binding region capable of binding to human B7H4 comprises a variable heavy chain (VH) region comprising the CDR1, CDR2 and CDR3 regions of SEQ ID NO. 25, 29 or 31, and a variable light chain region comprising the CDR1, CDR2 and CDR3 of SEQ ID NO.
  • VH variable heavy chain
  • the antigen-binding region that is capable of binding to CD3 comprises a heavy chain variable region (VH) comprising the CDR1, CDR2, and CDR3 sequences of 18, 19 and 21 respectively; and, a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 23, GTN and 24, respectively.
  • VH heavy chain variable region
  • VL light chain variable region
  • nucleic acids such as DNA or RNA
  • methods of producing the antibodies, or components thereof, as defined herein are provided encoding antibodies as defined herein, as well as methods of producing the antibodies, or components thereof, as defined herein.
  • said antibodies, or nucleic acids, in accordance with the invention are for use in a medical treatment.
  • FIGS. 1A-1D Determination of B7H4 domain involved in binding using B7H4-B7H3 chimeric molecules.
  • the B7H4 domain specificity of the B7H4 antibodies was determined using a panel of cells transfected to express human B7H4 ( FIG. 1A ), human B7H4-B7H3 chimeric molecules B7H3-IgV/B7H4-IgC ( FIG. 1B ) or B7H4-IgV/B7H3-IgC ( FIG. 1C ), or human B7H3 ( FIG. 1D ). Binding was determined by flow cytometry.
  • FIG. 2 Binding of B7H4 antibodies to B7H4, B7H3 or B7H4-B7H3 chimeric molecules. Binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR, bsIgG1-huCD3-H101G-FEALxB7H4-C2-FEAR, bsIgG1-huCD3-H101G-FEALxB7H4-C3-FEAR, bsIgG1-huCD3-H101G-FEALxB7H4-C4-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C5-FEAR to HEK cells transiently transfected to express human B7H4 or the B7H4-B7H3 chimeric molecules B7H3-IgV/B7H4-IgC or B7H4-
  • FIGS. 3A-3C Binding of B7H4 antibodies to B7H4 variants with alanine mutations in the ECD. Binding was expressed as fold change compared to a reference antibody. Fold change was defined as Log 10(Normalized gMFI[ala mutant]/Normalized gMFI[wt]). Residues where the Fold Change in binding was lower than mean Fold Change ⁇ 1.5 ⁇ SD were considered ‘loss of binding mutants’. Residues with a positive Fold Change in binding are loss of binding residues for the reference antibody. Numbers below the x-axis refer to amino acid positions.
  • FIG. 3A Results for C1-N52S, with C2 as reference antibody.
  • FIG. 3B Results for C2, with C1-N52S as reference antibody.
  • FIG. 3C Results for C3, with C2 as reference antibody.
  • FIG. 4 Binding of B7H4 antibody and CD3xB7H4 bispecific antibody to human and cynomolgus monkey B7H4. Binding of IgG1-B7H4-C1-N52S-FEAR (I) and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR (II) to HEK-293F cells transiently transfected with human B7H4 or cynomolgus monkey B7H4 was determined by flow cytometry. Non-transfected HEK-293F cells (III) were used as negative control; for these binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR is shown.
  • IgG1-B7H4-C1-N52S-FEAR IgG1-B7H4-C1-N52S-FEAR
  • II Ig
  • FIG. 5 Binding of B7H4 antibody and CD3xB7H4 bispecific antibody to B7H4 from rabbit, rat, mouse, dog and pig. Binding of IgG1-B7H4-C1-N52S-FEAR (I) and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR (II) to HEK-293F cells transiently transfected with B7H4 from rabbit, rat, mouse, dog or pig was determined by flow cytometry. Non-transfected HEK-293F cells (III) were used as negative control; for these binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR is shown.
  • IgG1-B7H4-C1-N52S-FEAR IgG1-B7H4-C1-N52S-FEAR
  • II IgG
  • FIG. 6 Binding of B7H4 antibodies to HEK-293F cells transiently transfected with B7H4 from different species. Binding of IgG1-B7H4-C1-N52S-FEAR (I), IgG1-B7H4-C3-FEAR (II), IgG1-B7H4-C2-FEAR (III),IgG1-B7H4-C4-FEAR (IV), and IgG1-B7H4-C5-FEAR (V) to HEK-293F cells transfected with B7H4 from human, cynomolgus monkey, mouse, rat or pig, or to untransfected HEK-293F cells, was determined by flow cytometry. IgG1-b12 was used as non-binding control antibody (not shown).
  • FIG. 7 Binding of IgG1-B7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR to MCF-7 and MDA-MB-468 cells. Binding was determined by flow cytometry. I: IgG1-B7H4-C1-N52S-FEAR. II: bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR. IgG1-b12 (III) and bsIgG1-huCD3-H101G-FEALxb12-FEAR (IV) were used as non-binding control antibodies.
  • FIG. 8 Binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR to NIH-OVCAR-3, HCC1954 and HeLa cells. Binding was determined by flow cytometry. I: bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR. BsIgG1-huCD3-H101G-FEALxb12-FEAR (II) was used as non-binding control antibody.
  • FIG. 9 Binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR to SK-BR3 and MDA-MB-486 cells. Binding was determined by flow cytometry. I: bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR. II: bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR. bsIgG1-huCD3-FEALxb12-FEAR (III) and bsIgG1-huCD3-H101G-FEALxb12-FEAR (IV) were used as non-binding control antibodies.
  • FIG. 10 Binding of various B7H4 antibodies in homodimer and bsAb format to MDA-MB-486 and HCC1954 cells. Binding of IgG1-B7H4-C1-N52S-FEAR (I homodimer), IgG1-B7H4-C2-FEAR (II homodimer), IgG1-B7H4-C3-FEAR (III homodimer), IgG1-B7H4-C4-FEAR (IV homodimer), IgG1-B7H4-C5-FEAR (V homodimer), bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR (I bsAb), bsIgG1-huCD3-FEALxB7H4-C2-FEAR [MDA-MB-468] or bsIgG1-huCD3-H101G-FEALxB7H
  • VI bsIgG1-huCD3-H101G-FEALxb12-FEAR VI bsAb
  • IgG1-b12-K409R VI homodimer
  • FIG. 11 Induction of T cell mediated cytotoxicity of SK-BR3 cells in vitro by CD3xB7H4 bispecific antibodies using purified T cells as effector cells at varying effector to target ratios (E:T).
  • bsIgG1-huCD3-FEALxb12-FEAR was used as non-binding control antibody.
  • I bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR;
  • II bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR;
  • III bsIgG1-huCD3-FEALxb12-FEAR.
  • FIG. 12 Induction of T cell mediated cytotoxicity in various tumor cell lines in vitro in the presence of CD3xB7H4 bispecific antibodies with different CD3 arms.
  • bsIgG1-huCD3-FEALxb12-FEAR was used as non-binding control antibody.
  • I bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR;
  • II bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR;
  • III bsIgG1-huCD3-FEALxb12-FEAR
  • IV bsIgG1-huCD3-H101G-FEALxb12-FEAR.
  • FIGS. 13A and 13B B7H4 expression levels and IC50 of T cell-mediated tumor cell killing.
  • FIG. 13B IC50 of T cell-mediated tumor cell killing in the presence of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR (I) or bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR (II) for the different tumor cell lines. Each dot represents an experiment performed with an individual T cell donor (4-6 donors per cell line), horizontal lines indicate median. Cell lines are ranked according to B7H4 expression level.
  • FIGS. 14A and 14B T cell activation by B7H4 bispecific antibodies in T cell-tumor cell co-cultures.
  • FIG. 14A T cell activation (% of CD69 on CD8+ cells) in the presence of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR (I) or bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR (II) for various B7H4-positive tumor cell lines, determined by flow cytometry.
  • FIG. 14B EC50 of T cell activation, using T cells derived from 3-5 donors, for each of the target cell lines. Each dot represents an experiment performed with an individual T cell donor; horizontal lines indicate geometric mean.
  • FIG. 15 IFN ⁇ in the supernatant of T cell-tumor cell co-cultures at EC50, EC90 and EC99 for bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR using T cells from 3-4 donors, determined by a multiplex U-plex assay. Shown are individual measurements (dots), geometric means (horizontal lines) and standard deviation (error bars).
  • I bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR.
  • II bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR.
  • FIGS. 16A and 16B IL-6 and MCP-1 levels in the plasma of cynomolgus monkeys treated with single dose IV infusion of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR ( FIG. 16A ) or bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR ( FIG. 16B ).
  • FIG. 17 Mean plasma concentration-time profiles following a single IV infusion of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR or bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR.
  • I bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR.
  • II bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR.
  • FIG. 18 B7H4 mRNA expression levels in a range of primary solid tumors. B7H4 mRNA levels were extracted from the Omicsoft TCGA database and visualized using Oncoland software. Indications are ranked according to median of the B7H4 mRNA expression.
  • THYM thymoma
  • UVM uveal melanoma
  • PCPG pheochromocytoma and paraganglioma
  • ACC adrenocortical carcinoma
  • MESO mesothelioma
  • SKCM skin cutaneous melanoma
  • READ rectum adenocarcinoma
  • COAD colon adenocarcinoma
  • GMB glioblastoma multiforme
  • SARC sarcoma
  • LIHC liver hepatocellular carcinoma
  • LGG brain lower grade glioma
  • KIRC kidney renal clear cell carcinoma
  • TGCT testicular germ cell tumors
  • KICH kidney chromophobe
  • STAD stomach adenocarcinoma
  • THCA thyroid carcinoma
  • HNSC head and neck squamous cell carcinoma
  • PRAD prostate adenocarcinoma
  • LUAD lung adenocarcinoma
  • antibody as used herein is intended to refer to an immunoglobulin molecule, a fragment of an immunoglobulin molecule, or a derivative of either thereof, which has the ability to specifically bind to an antigen under typical physiological and/or tumor-specific conditions with a half-life of significant periods of time, such as at least about 30 minutes, at least about 45 minutes, at least about one hour, at least about two hours, at least about four hours, at least about 8 hours, at least about 12 hours, at least about 24 hours or more, at least about 48 hours or more, at least about 3, 4, 5, 6, 7 or more days, etc., or any other relevant functionally-defined period (such as a time sufficient to induce, promote, enhance, and/or modulate a physiological response associated with antibody binding to the antigen and/or time sufficient for the antibody to be internalized).
  • significant periods of time such as at least about 30 minutes, at least about 45 minutes, at least about one hour, at least about two hours, at least about four hours, at least about 8 hours, at least about 12 hours, at least about 24
  • An antibody comprises a binding region (or binding domain which may be used herein, both having the same meaning) which can interact with an antigen, a binding region comprising variable regions of both heavy and light chains of an immunoglobulin molecule, or the like.
  • Antibodies can comprise constant regions of the antibodies (Abs) which may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (such as effector cells) and components of the complement system such as C1q, the first component in the classical pathway of complement activation.
  • the term “antibody” includes a monoclonal antibody (mAb), an antibody-like polypeptide, a chimeric antibody, a humanized antibody, as well as an ‘antibody fragment’ or a ‘fragment thereof’ retaining the ability to specifically bind to the antigen (antigen-binding fragment) provided by any known technique, such as enzymatic cleavage, peptide synthesis, and recombinant DNA technology.
  • mAb monoclonal antibody
  • an antibody-like polypeptide a chimeric antibody
  • humanized antibody as well as an ‘antibody fragment’ or a ‘fragment thereof’ retaining the ability to specifically bind to the antigen (antigen-binding fragment) provided by any known technique, such as enzymatic cleavage, peptide synthesis, and recombinant DNA technology.
  • antibody includes bispecific antibodies and/or antibodies having further modifications, e.g. antibody-drug conjugates thereof.
  • An antibody as defined according to the invention can possess any isotype unless the disclosure herein is otherwise limited.
  • an antibody may be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term “antibody” include (i) a Fab′ or Fab fragment, a monovalent fragment consisting of the light chain variable domain (VL), heavy chain variable domain (VH), light chain constant region (CL) and heavy chain constant region domain 1 (CH1) domains, or a monovalent antibody as described in WO 2007/059782; (ii) F(ab′) 2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) an Fd fragment consisting essentially of the VH and CH1 domains; (iv) an Fv fragment consisting essentially of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment Ward et al., Nature 341, 544-546 (1989), which consists essentially of a VH domain and is also called domain antibody Hol
  • VL and VH are coded for by separate genes, they may be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain antibodies or single chain Fv (scFv), see for instance Revets et al; Expert Opin Biol Ther.
  • An antibody can be produced in and collected from different in vitro or ex vivo expression or production systems, for example from recombinantly modified host cells, from hybridomas or systems that use cellular extracts supporting in vitro transcription and/or translation of nucleic acid sequences encoding the antibody. It is to be understood that a multitude of different antibodies, the antibodies being as defined in the context of the present invention, can be provided by producing each antibody separately in a production system as mentioned above and thereafter mixing the antibodies, or by producing several antibodies in the same production system.
  • immunoglobulin heavy chain or “heavy chain of an immunoglobulin” as used herein is intended to refer to one of the heavy chains of an immunoglobulin.
  • a heavy chain is typically comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region (abbreviated herein as CH) which defines the isotype of the immunoglobulin.
  • the heavy chain constant region typically is comprised of three domains, CH1, CH2, and CH3.
  • immunoglobulin as used herein is intended to refer to a class of structurally related glycoproteins consisting of two pairs of polypeptide chains, one pair of light (L) low molecular weight chains and one pair of heavy (H) chains, all four potentially inter-connected by disulfide bonds.
  • the structure of immunoglobulins has been well characterized (see for instance Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)). Within the structure of the immunoglobulin, the two heavy chains are inter-connected via disulfide bonds in the so-called “hinge region”.
  • each light chain is typically comprised of several regions; a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region typically is comprised of one domain, CL.
  • the VH and VL regions may be further subdivided into regions of hypervariability (or hypervariable regions which may be hypervariable in sequence and/or form of structurally defined loops), also termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • half molecule When used herein, the terms “half molecule”, “Fab-arm” and “arm” refer to one heavy chain-light chain pair.
  • a bispecific antibody is described to comprise a half-molecule antibody “derived from” a first antibody, and a half-molecule antibody “derived from” a second antibody, the term “derived from” indicates that the bispecific antibody was generated by recombining, by any known method, said half-molecules from each of said first and second antibodies into the resulting bispecific antibody.
  • recombining is not intended to be limited by any particular method of recombining and thus includes all of the methods for producing bispecific antibodies described herein below, including for example recombining by half-molecule exchange, as well as recombining at nucleic acid level and/or through co-expression of two half-molecules in the same cells.
  • antigen-binding region refers to a region of an antibody which is capable of binding to the antigen.
  • the antigen can be any molecule, such as a polypeptide.
  • Antigens may e.g. be presented on a cell, bacterium, or virion.
  • the terms “antigen” and “target” may, unless contradicted by the context, be used interchangeably in the context of the present invention.
  • the terms “antigen-binding region” and “antigen-binding site” may, unless contradicted by the context, be used interchangeably in the context of the present invention.
  • blocking binding or “blocking the binding of an antibody” or “cross-blocking binding” or “cross-blocks binding” refers to the situation where one antibody bound to a specific antigen prevents binding of the second antibody to the same antigen and vice versa. In the absence of the other antibody, each antibody has the ability to bind to the antigen as determined by a significant binding response, whereas one of the antibodies lacks a binding response when the other antibody is present.
  • the ability of one antibody to block the binding of another antibody may be determined by biolayer interferometry in a classical sandwich epitope binning assay format, for instance as described in Example 5 in the present application and by Abdiche et al.
  • block binding and “blocking the binding of an antibody” and “cross-blocking binding” and “cross-blocks binding” may, unless contradicted by the context, be used interchangeably in the context of the present invention.
  • An antibody that is said to blocks binding of another antibody, may also be said to compete with the other antibody for binding to the target.
  • K D (M), as used herein, refers to the equilibrium dissociation constant of a particular antibody-antigen interaction, and is obtained by dividing k d by k a . K D can also be referred to as “binding affinity”.
  • k d (sec ⁇ 1 ), as used herein, refers to the dissociation rate constant of a particular antibody-antigen interaction. Said value is also referred to as the k off value or off-rate.
  • k a (M ⁇ 1 ⁇ sec ⁇ 1 ), as used herein, refers to the association rate constant of a particular antibody-antigen interaction. Said value is also referred to as the k on value or on-rate.
  • binding refers to the binding of an antibody to a predetermined antigen or target, typically with a binding affinity corresponding to a K D of 1E ⁇ 6 M or less, e.g. 5E ⁇ 7 M or less, 1E ⁇ 7 M or less, such as 5E ⁇ 8 M or less, such as 1E ⁇ 8 M or less, such as 5E ⁇ 9 M or less, or such as 1E ⁇ 9 M or less, when determined by biolayer interferometry using the antibody as the ligand and the antigen as the analyte and binds to the predetermined antigen with an affinity corresponding to a K D that is at least ten-fold lower, such as at least 100-fold lower, for instance at least 1,000-fold lower, such as at least 10,000-fold lower, for instance at least 100,000-fold lower than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen.
  • a non-specific antigen
  • B7H4 refers to a protein entitled B7H4, which is also referred to as: B7-H4; V-set domain containing T cell activation inhibitor 1; or VTCN1.
  • B7H4 is a member of the B7 family of proteins, which family comprises cell-surface protein ligands that bind to receptors on lymphocytes.
  • B7H4 is a type I transmembrane protein that includes a short intracellular domain, a hydrophobic transmembrane domain, and an extracellular domain with an IgV- and an IgC-like domain with four conserved cysteine residues and seven sites for N-linked glycosylation. (Sica et al., 2003, Immunity 18: 849-861).
  • B7H4 proteins are known from various species, such as human ( Homo sapiens ) B7H4 (Uniprot accession no. Q7Z7D3), cynomolgus monkey ( Macaca fascicularis ) B7H4 transcript 1 (Uniprot accession no. A0A2K5U6P5), dog ( Canis familiaris ) B7H4 (Uniprot accession no. F1P8R9), rabbit ( Oryctolagus cuniculus ) B7H4 (Uniprot accession no. G1TQE8), rat ( Rattus norvegicus ) B7H4 (Uniprot accession no.
  • CD3 refers to the human Cluster of Differentiation 3 protein which is part of the T-cell co-receptor protein complex and is composed of four distinct chains. CD3 is found in various species, and thus, the term “CD3” may not be limited to human CD3, unless contradicted by context.
  • the complex contains a CD3 ⁇ (gamma) chain (human CD3 ⁇ chain UniProtKB/Swiss-Prot No P09693, or cynomolgus monkey CD3 ⁇ UniProtKB/Swiss-Prot No Q95LI7), a CD3 ⁇ (delta) chain (human CD3 ⁇ UniProtKB/Swiss-Prot No P04234, or cynomolgus monkey CD3 ⁇ UniProtKB/Swiss-Prot No Q95LI8), two CD3 ⁇ (epsilon) chains (human CD3 ⁇ : UniProtKB/Swiss-Prot No P07766, of which a sequence herein is incorporated as SEQ ID NO: 13, in which amino acid residues 1-22 represent a signal peptide and amino acid residues 23-207 represent the mature CD3 ⁇ polypeptide; cynomolgus monkey CD3 ⁇ UniProtKB/Swiss-Prot No Q95L15; or rhesus
  • antibody binding region refers to a region of the antigen, which comprises the epitope to which the antibody binds.
  • An antibody binding region may be determined by epitope binning using biolayer interferometry, by alanine scan, or by domain shuffle assays (using antigen constructs in which regions of the antigen are exchanged with that of another species and determining whether the antibody still binds to the antigen or not).
  • the amino acids within the antibody binding region that are involved in the interaction with the antibody may be determined by hydrogen/deuterium exchange mass spectrometry and/or by crystallography of the antibody bound to its antigen.
  • epitope means an antigenic determinant which is specifically bound by an antibody.
  • Epitopes usually consist of surface groupings of molecules such as amino acids, sugar side chains or a combination thereof and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • the epitope may comprise amino acid residues which are directly involved in the binding, and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked or covered by the antibody when it is bound to the antigen (in other words, the amino acid residue is within or closely adjacent to the footprint of the specific antibody).
  • monoclonal antibody refers to a preparation of antibody molecules of single molecular composition and typically displays a single binding specificity and affinity for a particular epitope.
  • a monoclonal antibody can be typically made by identical cells that are all clones of a unique parent cell, such as for example hybridomas, stable cell lines or the like.
  • human monoclonal antibody refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobulin sequences.
  • the human monoclonal antibodies may be produced by a hybridoma which includes a B cell obtained from a transgenic or transchromosomal nonhuman animal, such as a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene, fused to an immortalized cell.
  • Human monoclonal antibodies may be derived from human B cells or plasma cells.
  • Monoclonal antibodies may also be produced from recombinantly modified host cells, or systems that use cellular extracts supporting in vitro transcription and/or translation of nucleic acid sequences encoding the antibody.
  • isotype refers to the immunoglobulin class (for instance IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM) or any allotypes thereof, such as IgG1m(za) and IgG1m(f)) that is encoded by heavy chain constant region genes. Further, each heavy chain isotype can be combined with either a kappa ( ⁇ ) or lambda ( ⁇ ) light chain.
  • full-length antibody when used herein, refers to an antibody (e.g., a parent or variant antibody) comprising one pair of a heavy and light chain or two different pairs of heavy and light chains, each pair containing heavy and light chain constant and variable domains such as normally found in a heavy chain-light chain pair of a wild-type antibody of that isotype.
  • the heavy and light chain constant and variable domains may in particular contain amino acid substitutions that modify and/or improve functional properties of the antibody when compared to the full length parent or wild-type antibody.
  • a full-length antibody according to the present invention may be produced by a method comprising the steps of (i) cloning the CDR sequences into one or more suitable vectors comprising complete heavy and light chain sequences, and (ii) expressing the obtained suitable vectors with the heavy and light chain sequences in suitable expression systems. It is within the knowledge of the skilled person to produce a full-length antibody when starting out from either CDR sequences or full variable region sequences. Thus, the skilled person knows how to generate a full-length antibody in accordance with the present invention.
  • humanized antibody refers to a genetically engineered non-human antibody, which contains human antibody constant domains and non-human variable domains modified to contain a high level of sequence homology to human variable domains. This can be achieved by grafting of non-human antibody complementarity-determining regions (CDRs), which together form the antigen binding site, onto a homologous human acceptor framework region (FR) (see i.a. WO92/22653 and EP0629240). In order to fully reconstitute the binding affinity and specificity of the parental antibody, substitution of framework residues from the parental antibody (i.e. the non-human antibody) into the human framework regions (back-mutations) may be required.
  • CDRs complementarity-determining regions
  • FR homologous human acceptor framework region
  • a humanized antibody may comprise non-human CDR sequences, primarily human framework regions optionally comprising one or more amino acid back-mutations to the non-human amino acid sequence, and fully human constant regions.
  • additional amino acid modifications which are not necessarily back-mutations, may be applied to obtain a humanized antibody with preferred characteristics, such as particular useful affinity and biochemical properties, e.g. to include modifications to avoid deamidation, provide an “inert Fc region”, and/or improve manufacturing.
  • human antibody is intended to include antibodies having variable and framework regions derived from human germline immunoglobulin sequences and a constant domain derived from a human immunoglobulin constant domain.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations, insertions or deletions introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • a “human antibody” can incorporate VH and VL sequences that have been generated from human germline immunoglobulin sequences in a human, in a transgenic animal such as described in the examples herein, a HIS mouse, or the like. Such VH and VL sequences are considered human VH and VL sequences, which have been e.g. fused to constant domains derived from a human immunoglobulin constant domain.
  • human antibodies can be engineered antibodies.
  • a “human antibody” may have been subjected to further engineering, e.g. include modifications to avoid deamidation, provide an “inert Fc region”, enable bispecific antibody generation and/or improve manufacturing.
  • a human antibody may also be produced in non-human cells, e.g. in CHO cells or the like.
  • the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another non-human species, such as a mouse, have been grafted onto human framework sequences.
  • Fc region refers to a region comprising, in the direction from the N- to C-terminal ends of the two heavy chains of the antibody, at least a hinge region, a CH2 region and a CH3 region.
  • An Fc region of the antibody may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (such as effector cells) and components of the complement system.
  • hinge region refers to the hinge region of an immunoglobulin heavy chain.
  • the hinge region of a human IgG1 antibody corresponds to amino acids 216-230 according to the Eu numbering as set forth in Kabat Kabat, E. A. et al., Sequences of proteins of immunological interest. 5th Edition—US Department of Health and Human Services, NIH publication No. 91-3242, pp 662, 680, 689 (1991).
  • the hinge region may also be any of the other subtypes as described herein.
  • CH1 region refers to the CH1 region of an immunoglobulin heavy chain.
  • the CH1 region of a human IgG1 antibody corresponds to amino acids 118-215 according to the Eu numbering as set forth in Kabat (ibid).
  • the CH1 region may also be any of the other subtypes as described herein.
  • CH2 region refers to the CH2 region of an immunoglobulin heavy chain.
  • the CH2 region of a human IgG1 antibody corresponds to amino acids 231-340 according to the Eu numbering as set forth in Kabat (ibid).
  • the CH2 region may also be any of the other subtypes as described herein.
  • CH3 region refers to the CH3 region of an immunoglobulin heavy chain.
  • the CH3 region of a human IgG1 antibody corresponds to amino acids 341-447 according to the Eu numbering as set forth in Kabat (ibid).
  • the CH3 region may also be any of the other subtypes as described herein.
  • Fc-mediated effector functions is intended to refer to functions that are a consequence of binding a polypeptide or antibody to its target or antigen on a cell membrane wherein the Fc-mediated effector function is attributable to the Fc region of the polypeptide or antibody.
  • Fc-mediated effector functions include (i) C1q binding, (ii) complement activation, (iii) complement-dependent cytotoxicity (CDC), (iv) antibody-dependent cell-mediated cytotoxity (ADCC), (v) Fc-gamma receptor (FcgR)-binding, (vi) antibody-dependent, Fc ⁇ R-mediated antigen crosslinking, (vii) antibody-dependent cellular phagocytosis (ADCP), (viii) complement-dependent cellular cytotoxicity (CDCC), (ix) complement-enhanced cytotoxicity, (x) binding to complement receptor of an opsonized antibody mediated by the antibody, (xi) opsonisation, and (xii) a combination of any of (i) to (xi).
  • inertness refers to an Fc region which is at least not able to bind any Fc ⁇ R, induce Fc-mediated cross-linking of Fc ⁇ Rs, or induce Fc ⁇ R-mediated cross-linking of target antigens via two Fc regions of individual antibodies, or is not able to bind C1q.
  • An example thereof is FEA substitutions within the constant domain as described herein.
  • the inertness of an Fc region of an antibody may be tested using the antibody in a monospecific or bispecific format.
  • full-length when used in the context of an antibody indicates that the antibody is not a fragment, but contains all of the domains corresponding with the particular isotype such as normally found for that isotype in nature, e.g. the VH, CH1, CH2, CH3, hinge, VL and CL domains for an IgG1 antibody.
  • bispecific antibody refers to an antibody molecule that can interact with an antigen, with only one antigen-binding domain (e.g. one Fab arm).
  • monovalent antibody binding refers to the binding of the bispecific antibody to one antigen with only one antigen-binding domain (e.g. one Fab arm).
  • the term “monospecific antibody” in the context of the present invention refers to an antibody that has binding specificity to one antigen, one epitope only.
  • the antibody may be a monospecific, monovalent antibody (i.e. carrying only one antigen-binding region) or a monospecific, bivalent antibody (e.g. an antibody with two identical antigen-binding regions).
  • bispecific antibody refers to an antibody having two antigen-binding domains that bind different epitopes, e.g. two non-identical pairs of VH and VL regions, two non-identical Fab-arms or two Fab-arms with non-identical CDR regions.
  • bispecific antibodies have specificity for at least two different epitopes. Such epitopes may be on the same or different antigens or targets. If the epitopes are on different antigens, such antigens may be on the same cell or different cells, cell types or structures, such as extracellular matrix or vesicles and soluble protein. A bispecific antibody may thus be capable of crosslinking multiple antigens, e.g. two different cells.
  • bivalent antibody refers to an antibody that has two antigen-binding regions, which bind to two of the same epitopes on two of the same antigens or binds to two different epitopes on the same or different antigen(s).
  • a bivalent antibody may be a monospecific antibody or a bispecific antibody.
  • amino acid and “amino acid residue” may herein be used interchangeably, and are not to be understood limiting.
  • Amino acids are organic compounds containing amine (—NH 2 ) and carboxyl (—COOH) functional groups, along with a side chain (R group) specific to each amino acid.
  • amino acids may be classified based on structure and chemical characteristics. Thus, classes of amino acids may be reflected in one or both of the following tables:
  • substitution of one amino acid for another may be classified as a conservative or non-conservative substitution.
  • a “conservative substitution” is a substitution of one amino acid with another amino acid having similar structural and/or chemical characteristics, such substitution of one amino acid residue for another amino acid residue of the same class as defined in any of the two tables above: for example, leucine may be substituted with isoleucine as they are both aliphatic, branched hydrophobes. Similarly, aspartic acid may be substituted with glutamic acid since they are both small, negatively charged residues.
  • Xaa or X may typically represent any of the 20 naturally occurring amino acids.
  • naturally occurring refers to any one of the following amino acid residues; glycine, alanine, valine, leucine, isoleucine, serine, threonine, lysine, arginine, histidine, aspartic acid, asparagine, glutamic acid, glutamine, proline, tryptophan, phenylalanine, tyrosine, methionine, and cysteine.
  • the notation “K409R” or “Lys409Arg” means, that the antibody comprises a substitution of Lysine with Arginine in amino acid position 409.
  • Substitution of an amino acid at a given position to any other amino acid is referred to as: Original amino acid—position; or e.g. “K409”.
  • the original amino acid(s) and/or substituted amino acid(s) may comprise more than one, but not all amino acid(s)
  • the more than one amino acid may be separated by “,” or “/”.
  • Lysine with Arginine, Alanine, or Phenylalanine in position 409 is: “Lys409Arg,Ala,Phe” or “Lys409Arg/Ala/Phe” or “K409R,A,F” or “K409R/A/F” or “K409 to R, A, or F”. Such designation may be used interchangeably in the context of the invention but have the same meaning and purpose.
  • a substitution embraces a substitution into any one or the other nineteen natural amino acids, or into other amino acids, such as non-natural amino acids.
  • a substitution of amino acid K in position 409 includes each of the following substitutions: 409A, 409C, 409D, 409E, 409F, 409G, 409H, 409I, 409L, 409M, 409N, 409O, 409R, 409S, 409T, 409V, 409W, 409P, and 409Y.
  • This is, by the way, equivalent to the designation 409X, wherein the X designates any amino acid other than the original amino acid.
  • substitutions may also be designated K409A, K409C, etc. or K409A,C, etc. or K409A/C/etc. The same applies by analogy to each and every position mentioned herein, to specifically include herein any one of such substitutions.
  • the antibody according to the invention may also comprise a deletion of an amino acid residue.
  • Such deletion may be denoted “del”, and includes, e.g., writing as K409del.
  • the Lysine in position 409 has been deleted from the amino acid sequence.
  • host cell is intended to refer to a cell into which a nucleic acid such as an expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell, but may also include the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • Recombinant host cells include, for example, transfectomas, such as CHO cells, HEK-293 cells, Expi293F cells, PER.C6 cells, NS0 cells, and lymphocytic cells, and prokaryotic cells such as E. coli and other eukaryotic hosts such as plant cells and fungi.
  • transfectoma includes recombinant eukaryotic host cells expressing the antibody or a target antigen, such as CHO cells, PER.C6 cells, NS0 cells, HEK-293 cells, Expi293F cells, plant cells, or fungi, including yeast cells.
  • sequence identity between two amino acid sequences is determined over the length of the referenced sequence using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 5.0.0 or later.
  • the parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix.
  • the output of Needle labeled “longest identity” is used as the percent identity and is calculated as follows:
  • Suitable variants typically exhibit at least about 45%, such as at least about 55%, at least about 65%, at least about 75%, at least about 85%, at least about 90%, at least about 95%, or more (e.g., about 99%) similarity to the parent or referenced sequence.
  • internalized refers to a biological process in which molecules such as the antibody according to the present invention, are engulfed by the cell membrane and drawn into the interior of the cell. Internalization may also be referred to as “endocytosis”.
  • an antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein said antigen-binding regions comprise heavy and light chain variable regions, wherein said antigen-binding regions are human variable regions and/or humanized variable regions.
  • one antigen-binding region may comprise human heavy and light chain variable regions, and the other antigen-binding region may comprise humanized heavy and light chain variable regions.
  • both antigen-binding region may comprise human heavy and light chain variable regions, or both antigen-binding regions may comprise humanized heavy and light chain variable regions.
  • an antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein said antigen-binding regions comprise heavy and light chain variable regions, wherein said heavy and light chain variable regions comprise human framework regions.
  • An antibody in accordance with the invention as described herein comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, may also be referred to herein e.g. as a B7H4xCD3 antibody.
  • Such antibodies are preferably bispecific antibodies.
  • Such an antibody as described above are in a further embodiment capable of binding cancer cells and T-cells, such as e.g. described in the examples.
  • Cancer cells that may be selected are cancer cells that express human B7H4 and/or are cancer cells that are of a solid tumor.
  • Such an antibody preferably is capable of inducing T-cell mediated cell killing of the cancer cells.
  • Capable of binding is understood to comprise, as shown in the examples, that in a binding assay, an antibody binds to its target, as shown by e.g. typical binding curves such as shown in FIGS. 3 and 4 herein, or by determining binding affinity, using e.g. biolayer interferometry, as shown in examples 3 and 4.
  • An antigen-binding region not capable of binding to a specified target has e.g. an undectable binding affinity to its target, e.g. having a response of ⁇ 0.05 nm at the highest concentration used in a typical biolayer interferometry assay such as shown in example 3.
  • the skilled person is well aware how to determine whether or not an antigen-binding region is capable of binding to its target.
  • the present invention provides bispecific CD3xB7H4 antibodies which efficiently promote T cell-mediated killing of B7H4-expressing tumor cells.
  • particular antigen-binding regions can be selected from the set of antibodies or antigen-binding regions provided by the present invention.
  • Many different formats and uses of bispecific antibodies are known in the art, and were reviewed by Kontermann; Drug Discov Today, 2015 July; 20(7):838-47 and; MAbs, 2012 March-April; 4(2):182-97.
  • a bispecific antibody according to the present invention may not be limited to any particular bispecific format or method of producing it.
  • the bispecific antibody of the present invention is a diabody, a cross-body, or a bispecific antibody obtained via a controlled Fab-arm exchange (such as described in WO2011131746 (Genmab)).
  • bispecific antibodies include but are not limited to (i) IgG-like molecules with complementary CH3 domains to force heterodimerization; (ii) recombinant IgG-like dual targeting molecules, wherein the two sides of the molecule each contain the Fab fragment or part of the Fab fragment of at least two different antibodies; (iii) IgG fusion molecules, wherein full length IgG antibodies are fused to extra Fab fragment or parts of Fab fragment; (iv) Fc fusion molecules, wherein single chain Fv molecules or stabilized diabodies are fused to heavy-chain constant-domains, Fc-regions or parts thereof; (v) Fab fusion molecules, wherein different Fab-fragments are fused together, fused to heavy-chain constant-domains, Fc-regions or parts thereof; and (vi) ScFv- and diabody-based and heavy chain antibodies (e.g., domain antibodies, nanobodies) wherein different single chain Fv molecules or different diabodies or different heavy-chain antibodies
  • IgG-like molecules with complementary CH3 domain molecules include but are not limited to the Triomab/Quadroma molecules (Trion Pharma/Fresenius Biotech; Roche, WO2011069104), the so-called Knobs-into-Holes molecules (Genentech, WO9850431), CrossMAbs (Roche, WO2011117329) and the electrostatically-matched molecules (Amgen, EP1870459 and WO2009089004; Chugai, US201000155133; Oncomed, WO2010129304), the LUZ-Y molecules (Genentech, Wranik et al. J. Biol. Chem.
  • IgG-like dual targeting molecules examples include but are not limited to Dual Targeting (DT)-Ig molecules (WO2009058383), Two-in-one Antibody (Genentech; Bostrom, et al 2009. Science 323, 1610-1614.), Cross-linked Mabs (Karmanos Cancer Center), mAb2 (F-Star, WO2008003116), Zybody molecules (Zyngenia; LaFleur et al. MAbs. 2013 March-April; 5(2):208-18), approaches with common light chain (Crucell/Merus, U.S. Pat. No.
  • IgG fusion molecules include but are not limited to Dual Variable Domain (DVD)-Ig molecules (Abbott, U.S. Pat. No. 7,612,181), Dual domain double head antibodies (Unilever; Sanofi Aventis, WO20100226923), IgG-like Bispecific molecules (ImClone/Eli Lilly, Lewis et al. Nat Biotechnol. 2014 February; 32(2):191-8), Ts2Ab (MedImmune/AZ; Dimasi et al. J Mol Biol. 2009 Oct.
  • DVD Dual Variable Domain
  • U.S. Pat. No. 7,612,181 Dual domain double head antibodies
  • IgG-like Bispecific molecules ImClone/Eli Lilly, Lewis et al. Nat Biotechnol. 2014 February; 32(2):191-8
  • Ts2Ab MedImmune/AZ; Dimasi et al. J Mol Biol. 2009 Oct.
  • BsAb molecules Zymogenetics, WO2010111625), HERCULES molecules (Biogen Idec, US007951918), scFv fusion molecules (Novartis), scFv fusion molecules (Changzhou Adam Biotech Inc, CN 102250246) and TvAb molecules (Roche, WO2012025525, WO2012025530).
  • Fc fusion molecules include but are not limited to ScFv/Fc Fusions (Pearce et al., Biochem Mol Biol Int. 1997 September; 42(6):1179-88), SCORPION molecules (Emergent BioSolutions/Trubion, Blankenship J W, et al. AACR 100 th Annual meeting 2009 (Abstract #5465); Zymogenetics/BMS, WO2010111625), Dual Affinity Retargeting Technology (Fc-DART) molecules (MacroGenics, WO2008157379, WO2010080538) and Dual(ScFv)2-Fab molecules (National Research Center for Antibody Medicine—China).
  • Fab fusion bispecific antibodies include but are not limited to F(ab)2 molecules (Medarex/AMGEN; Deo et al J Immunol. 1998 Feb. 15; 160(4):1677-86.), Dual-Action or Bis-Fab molecules (Genentech, Bostrom, et al 2009. Science 323, 1610-1614.), Dock-and-Lock (DNL) molecules (ImmunoMedics, WO2003074569, WO2005004809), Bivalent Bispecific molecules (Biotecnol, Schoonjans, J Immunol. 2000 Dec. 15; 165(12):7050-7.) and Fab-Fv molecules (UCB-Celltech, WO 2009040562 A1).
  • ScFv-, diabody-based and domain antibodies include but are not limited to Bispecific T Cell Engager (BiTE) molecules (Micromet, WO2005061547), Tandem Diabody molecules (TandAb) (Affimed) Le Gall et al., Protein Eng Des Sel. 2004 April; 17(4):357-66.), Dual Affinity Retargeting Technology (DART) molecules (MacroGenics, WO2008157379, WO2010080538), Single-chain Diabody molecules (Lawrence, FEBS Lett. 1998 Apr.
  • BiTE Bispecific T Cell Engager
  • TandAb Tandem Diabody molecules
  • DART Dual Affinity Retargeting Technology
  • Single-chain Diabody molecules Single-chain Diabody molecules
  • TCR-like Antibodies AIT, ReceptorLogics
  • Human Serum Albumin ScFv Fusion Merrimack, WO2010059315
  • COMBODY molecules Epigen Biotech, Zhu et al. Immunol Cell Biol. 2010 August; 88(6):667-75.
  • dual targeting nanobodies Ablynx, Hmila et al., FASEB J. 2010
  • dual targeting heavy chain only domain antibodies
  • the bispecific antibody of the invention can be of any isotype.
  • Exemplary isotypes include but are not limited to either of the human IgG1, IgG2, IgG3, and IgG4 isotypes.
  • bispecific antibodies may be selected to be of the human IgG1 isotype, as shown in the examples. Either of the human light chain constant regions, kappa or lambda, may be used.
  • both heavy chains of an antibody of the present invention are of the IgG1 isotype, for instance an IgG1, ⁇ .
  • the two heavy chains of a bispecific antibody are of the IgG1 and IgG4 isotypes, respectively.
  • bispecific antibodies may be selected to be of the human IgG1 isotype, as shown in the examples.
  • the heavy chain and Fc sequences thereof of the selected isotype may be modified in the hinge and/or CH3 region as described herein to enable the generation of bispecific antibodies and introduce inertness.
  • the bispecific antibody of the invention comprises an Fc-region comprising a first heavy chain with a first Fc sequence comprising a first CH3 region, and a second heavy chain with a second Fc sequence comprising a second CH3 region, wherein the sequences of the first and second CH3 regions are different and are such that the heterodimeric interaction between said first and second CH3 regions is stronger than each of the homodimeric interactions of said first and second CH3 regions. More details on these interactions and how they can be achieved are provided in WO2011131746 and WO2013060867 (Genmab), which are hereby incorporated by reference.
  • a stable bispecific CD3xB7H4 antibody can be obtained at high yield on the basis of one B7H4 antibody and one CD3 antibody, each composed of two identical heavy chains and two identical light chains, each antibody containing only a few, fairly conservative, (asymmetrical) mutations in the CH3 regions.
  • Asymmetrical mutations mean that the sequences of said first and second CH3 regions contain one or more amino acid substitutions at non-identical positions.
  • the invention provides an antibody according to the invention comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3. Furthermore, the invention provides an antibody according to the invention comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region capable of binding CD3, is capable of binding human CD3 ⁇ (epsilon), such as human CD3 ⁇ (epsilon) as specified in SEQ ID NO: 13. Such antigen-binding region is capable of binding human CD3 ⁇ (epsilon), as presented on a T cell, such as a primary human T cell.
  • a T cell such as a primary human T cell.
  • Said antibody according to the invention may be an antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises
  • CDR1, CDR2 and CDR3 regions can be identified from variable heavy and light chain regions using methods known in the art.
  • the CDR regions from said variable heavy and light chain regions can be annotated according to IMGT (see Lefranc M P. et al., Nucleic Acids Research, 27, 209-212, 1999] and Brochet X. Nucl. Acids Res. 36, W503-508 (2008)).
  • IMGT see Lefranc M P. et al., Nucleic Acids Research, 27, 209-212, 1999] and Brochet X. Nucl. Acids Res. 36, W503-508 (2008).
  • antibodies comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises
  • antibodies comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises
  • antigen-binding regions that are capable of binding human CD3 have been described i.a. in WO2015001085, and WO2017009442. Further antigen-binding regions that are capable of binding human CD3 are disclosed and described in WO2015001085 and WO2017009442, which can be further contemplated and serve as the basis for generating antibodies in accordance with the current invention, which are incorporated by reference herein.
  • the said antibody in accordance with the invention may bind with an equilibrium dissociation constant K D between the antigen-binding region that binds to human CD3, and human CD3 is within the range of 1-1000 nM.
  • the said antibody in accordance with the invention may bind with a equilibrium dissociation constant K D between the antigen-binding region that binds to human CD3, and human CD3 is within the range of 1-100 nM, such as within the range of 5-100 nM, within the range of 10-100 nM, within the range of 1-80 nM, within the range of 1-60 nM within the range of 1-40 nM, within the range of 1-20 nM, within the range of 5-80 nM, within the range of 5-60 nM, within the range of 5-40 nM, within the range of 5-20 nM, within the range of 10-80 nM, within the range of 10-60 nM, within the range of 10-40 nM, or such as within the range of 10-20 nM.
  • An exemplary and suitable antigen-binding region comprises a heavy chain variable region (VH) of SEQ ID NO: 16 and a light chain variable region (VL) regions of SEQ ID NO: 22.
  • VH heavy chain variable region
  • VL light chain variable region
  • said antibody has a lower binding affinity for human CD3 ⁇ than an antibody having an antigen-binding region comprising a VH sequence as set forth in SEQ ID NO: 16, and a VL sequence as set forth in SEQ ID NO: 22, preferably wherein said affinity is at least 5-fold lower, such as at least 10-fold lower, e.g. at least 20-fold lower, at least 30 fold lower, at least 40 fold lower, at least 45 fold lower or such as at least 50-fold lower.
  • said antibody may bind with an equilibrium dissociation constant K D between the antigen-binding region that binds to human CD3, and human CD3 antigen-binding which is within the range of 200-1000 nM, such as within the range of 300-1000 nM, within the range of 400-1000 nM, within the range of 500-1000 nM, within the range of 300-900 nM within the range of 400-900 nM, within the range of 400-700 nM, within the range of 500-900 nM, within the range of 500-800 nM, within the range of 500-700 nM, within the range of 600-1000 nM, within the range of 600-900 nM, within the range of 600-800 nM, or such as within the range of 600-700 nM.
  • K D equilibrium dissociation constant
  • An exemplary and suitable antigen-binding region comprises a heavy chain variable region (VH) of SEQ ID NO: 16 or of SEQ ID NO. 17, and, a light chain variable region (VL) regions of SEQ ID NO: 22.
  • VH heavy chain variable region
  • VL light chain variable region
  • binding affinity can be determined by biolayer interferometry, optionally as set forth in Example 4 herein.
  • the antibody according to the invention having a binding affinity to human CD3 as defined herein may have the binding affinity determined using biolayer interferometry comprising the steps of:
  • binding affinity may be determined using an antibody such as a monospecific, bivalent antibody, such as an antibody which is a full length IgG1.
  • the antibody according to the invention is an antibody, wherein
  • the antibody according to the invention is an antibody, wherein the antigen-binding region that binds to CD3 comprises in the heavy chain variable (VH) region as defined herein comprises a substitution selected from the group consisting of: T31M, T31P, N57E, H101G, H101N, G105P, S110A, S110G, Y114M, Y114R, Y114V.
  • VH heavy chain variable
  • the antibody according to the invention is an antibody wherein the antigen-binding region that binds to CD3 comprises a heavy chain variable region as defined herein having at the amino acid position 31 an M or P, or at the amino acid position 57 an E, or at the amino acid position 101 a G or N, or at the amino acid 105 a P, or at the amino acid position 110 and A or G, or at the amino acid position 114 an M, R or V, said positions corresponding with the amino acid position numbering of the heavy chain variable (VH) region having the sequence set forth in SEQ ID NO: 16.
  • the antibody according to the invention is an antibody wherein the CDR1, CDR2 and CDR3 of the heavy chain variable (VH) region of the antigen-binding region that binds to CD3 as defined herein comprises, in total, at the most 1, 2, 3, 4 or 5 amino acid substitutions, when compared with the CDR1, CDR2 and CDR3 of the sequences of SEQ ID NO: 16, said amino acid substitutions comprising preferably amino acid substitutions as defined above.
  • VH heavy chain variable
  • the invention provides an antibody according to the invention comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein said human B7H4 is human B7H4 of SEQ ID NO. 1.
  • said antibody in accordance with the invention comprises an antigen-binding region capable of binding to human CD3 ⁇ (epsilon) as specified in SEQ ID NO: 13, and an antigen-binding region capable of binding human B7H4 of SEQ ID NO. 1.
  • the antibody according to the invention is an antibody wherein said antigen-binding region capable of binding to human B7H4 is capable of binding to the extracellular domain of human B7H4.
  • said B7H4 is expressed on a cell, more preferably a human cell.
  • the antibody according to the invention is an antibody wherein said antigen-binding region capable of binding to human B7H4 is capable of binding to the IgC-like constant region of human B7H4.
  • the antibody according to the invention is an antibody wherein said antigen-binding region capable of binding to human B7H4 is capable of binding to B7H3-IgV/B7H4-IgC.
  • B7H3-IgV/B7H4-IgC represents a fusion between human B7H3 and B7H4, wherein the B7H3 IgV-like domain is fused with the B7H4 IgC-like domain, corresponding with SEQ ID NO. 11.
  • the antibody according to the invention is an antibody wherein said antigen-binding region capable of binding to human B7H4 is not capable of binding to B7H4-IgV/B7H3-IgC.
  • B7H4-IgV/B7H3-IgC represents a fusion between human B7H3 and B7H4, wherein the B7H4 IgV-like domain is fused with the B7H3 IgC-like domain, corresponding with SEQ ID NO. 10.
  • Said B7H4-IgV/B7H3-IgC being expressed by a cell such as described in the example 7 herein.
  • Suitable antigen-binding regions capable of binding to human B7H4, that are contemplated according to the invention as described herein comprise:
  • CDR1, CDR2 and CDR3 regions can be identified from variable heavy and light chain regions using methods known in the art.
  • the CDR regions from said variable heavy and light chain regions can be annotated according to IMGT (see Lefranc M P. et al., Nucleic Acids Research, 27, 209-212, 1999] and Brochet X. Nucl. Acids Res. 36, W503-508 (2008)).
  • suitable antigen-binding regions capable of binding to human B7H4 that are contemplated according to the invention as described herein comprise:
  • antigen-binding regions capable of binding to human B7H4 that are contemplated according to the invention as described herein comprise:
  • said antigen-binding regions that binds to B7H4 comprise heavy and light chain variable regions (VH) having at least 90%, at least 95%, at least 97%, or at least 99% amino acid sequence identity with:
  • the antibody according to the invention may have an antigen-binding region capable of binding to B7H4 having a binding affinity to human B7H4 that corresponds to a K D value of 5E-7 M or less, such as 1E-7 M or less, such as with a binding affinity corresponding to a K D value which is within the range of 5E-7 to 2E-10 M, such as within the range of 2E-7 to 1E-10 M or 1E-7 to 5E-9 M.
  • binding affinity can be determined by biolayer interferometry, optionally as set forth in Example 3 herein.
  • the antibody according to the invention having a binding affinity to human B7H4 as defined herein may have the binding affinity determined using biolayer interferometry comprising the steps of:
  • binding affinity may be determined using an antibody such as a monospecific, bivalent antibody, such as an antibody which is a full length IgG1.
  • an antibody in accordance with the invention comprising an antigen region capable of binding to human B7H4, wherein said antigen-binding region is capable of crossblocking:
  • said antibody in accordance with the invention comprises an antigen region capable of binding to human B7H4, said antigen-binding region capable of crossblocking
  • cross-blocking or the ability of an antibody according to the invention to block binding of another antibody to B7H4, is defined as the ability of a first antibody bound to B7H4 to block binding of a second antibody to the B7H4 bound to the first antibody.
  • Crossblocking can be determined using an assay as described in example 5. Such crossblocking can also be determined e.g. in a procedure comprising the steps of:
  • the first antibody is considered to cross-block the second antibody.
  • the skilled person will be familiar with suitable technologies for determining the ability of an antibody to crossblock the binding of another antibody to its target, the present application discloses procedures suitable for determining blocking of binding and displacement.
  • crossblocking as described herein is determined as described in Example 5.
  • the antibody in accordance with the invention having an antigen-binding region capable of binding to human B7H4 complying with a crossblocking feature as described above, wherein said an antigen-binding region capable of binding to human B7H4 is capable of binding to B7H3-IgV/B7H4-IgC (SEQ ID NO. 11), and optionally is not capable of binding to B7H4-IgV/B7H3-IgC (SEQ ID NO. 10).
  • the present disclosure further provides an antibody according to the invention comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises
  • an antibody according to the invention may be an antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises
  • the present disclosure further provides an antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region capable of binding to CD3 comprises:
  • the present disclosure further provides an antibody comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region capable of binding to CD3 comprises:
  • antibodies comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises:
  • antibodies comprising an antigen-binding region capable of binding to human B7H4 and an antigen-binding region capable of binding to human CD3, wherein the antigen-binding region that binds to CD3 comprises:
  • said antigen binding region capable of binding to human B7H4 is comprised in an heavy chain and a light chain, said heavy chain comprising said VH region and an IgG1 heavy chain constant region and said light chain comprising said VL region and a kappa light chain constant region; and wherein said antigen binding region capable of binding to human CD3 is comprised in a heavy chain and a light chain, said heavy chain comprising said VH region and an IgG1 heavy chain constant region and said light chain comprising said VL region and a lambda light chain constant region. More preferably, in such a bispecific antibody, one IgG1 heavy chain constant region is as defined in SEQ ID NO.
  • each antigen-binding region of an antibody generally comprise a heavy chain variable region (VH) and a light chain variable region (VL), and each of the variable regions comprises three CDR sequences, CDR1, CDR2 and CDR3, respectively, and may comprise four framework sequences, FR1, FR2, FR3 and FR4, respectively.
  • Each antigen-binding region of an antibody may generally comprise a heavy chain variable region (VH) and a light chain variable region (VL), and each of the variable regions comprises three CDR sequences, CDR1, CDR2 and CDR3, respectively, and may comprise four human framework sequences, FR1, FR2, FR3 and FR4, respectively.
  • This structure is preferably also found in the antibodies according to the present invention.
  • the antibodies according to the invention may comprise two heavy chain constant regions (CH), and two light chain constant regions (CL). Examples of constant regions are provided i.a. in SEQ ID NOs. 57-64.
  • the antibody according to the invention comprises a first and a second heavy chain, such as a first and second heavy chain each comprising at least a hinge region, a CH2 and CH3 region.
  • a first and second heavy chain each comprising at least a hinge region, a CH2 and CH3 region.
  • Stable, heterodimeric antibodies can be obtained at high yield for instance by so-called Fab-arm exchange as provided in WO 2008/119353 and WO 2011/131746, on the basis of two homodimeric starting proteins containing only a few, asymmetrical mutations in the CH3 regions.
  • the antibody comprises a first heavy chain wherein at least one of the amino acids at the positions corresponding to positions selected from the group consisting of T366, L368, K370, D399, F405, Y407 and K409 in a human IgG1 heavy chain has been substituted, and a second heavy chain wherein at least one of the amino acids in the positions corresponding to a position selected from the group consisting of T366, L368, K370, D399, F405, Y407, and K409 in a human IgG1 heavy chain has been substituted, wherein said substitutions of said first and said second heavy chains are not in the same positions, and wherein the amino acid positions are numbered according to Eu numbering.
  • constant domains having such a substitution are provided i.a. in SEQ ID NO. 58 and 62, which can be compared with SEQ ID NO. 57, which does not have such a substitution.
  • amino acid corresponding to positions refers to an amino acid position number in a human IgG1 heavy chain. Corresponding amino acid positions in other immunoglobulins may be found by alignment with human IgG1. Unless otherwise stated or contradicted by context, the amino acids of the constant region sequences are herein numbered according to the EU-index of numbering (described in Kabat, E. A. et al., 1991, Sequences of proteins of immunological interest. 5th Edition—US Department of Health and Human Services, NIH publication No. 91-3242, pp 662, 680, 689).
  • an amino acid or segment in one sequence that “corresponds to” an amino acid or segment in another sequence is one that aligns with the other amino acid or segment using a standard sequence alignment program such as ALIGN, ClustalW or similar, typically at default settings and has at least 50%, at least 80%, at least 90%, or at least 95% identity to a human IgG1 heavy chain. It is considered well-known in the art how to align a sequence or segment in a sequence and thereby determine the corresponding position in a sequence to an amino acid position according to the present invention.
  • the invention provides an antibody, wherein the amino acid in the position corresponding to K409 in a human IgG1 heavy chain is R in said first heavy chain, and the amino acid in the position corresponding to F405 in a human IgG1 heavy chain is L in said second heavy chain, or vice versa.
  • the antibody according to the present invention comprises, in addition to the antigen-binding regions, comprises an Fc region with Fc sequences of the two heavy chains.
  • the first and second Fc sequence may each be of any isotype, including any human isotype, such as an IgG1, IgG2, IgG3, IgG4, IgE, IgD, IgM, or IgA isotype or a mixed isotype.
  • the Fc region is a human IgG1, IgG2, IgG3, IgG4 isotype or a mixed isotype, such as a human IgG1 isotype.
  • it is preferred that the antibody according to the invention is a full-length antibody, most preferably it is of the IgG1 type.
  • Antibodies according to the present invention may comprise modifications in the Fc region to render the antibody an inert, or non-activating, antibody.
  • one or both heavy chains may be modified so that the antibody induces Fc-mediated effector function to a lesser extent relative to an antibody which is identical, except for comprising non-modified first and second heavy chains.
  • the Fc-mediated effector function may be measured by determining Fc-mediated CD69 expression on T cells (i.e. CD69 expression as a result of CD3 antibody-mediated, Fc ⁇ receptor-dependent CD3 crosslinking), by binding to Fc ⁇ receptors, by binding to C1q, or by induction of Fc-mediated cross-linking of Fc ⁇ Rs.
  • the heavy chain constant sequences may be modified so that the Fc-mediated CD69 expression is reduced by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 99% or 100% when compared to a wild-type (unmodified) antibody, wherein said Fc-mediated CD69 expression is determined in a PBMC-based functional assay, e.g. as described in Example 3 of WO2015001085.
  • Modifications of the heavy and light chain constant sequences may also result in reduced binding of C1q to said antibody. As compared to an unmodified antibody the reduction may be by at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, or 100% and the C1q binding may be determined by ELISA.
  • the Fc region which may be modified so that said antibody mediates reduced Fc-mediated T-cell proliferation compared to an unmodified antibody by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 99% or 100%, wherein said T-cell proliferation is measured in a PBMC-based functional assay.
  • amino acid positions that may be modified, e.g. in an IgG1 isotype antibody, include positions L234 and L235.
  • the antibody according to the invention may comprises a first and a second heavy chain, and wherein in both the first and the second heavy chain, the amino acid residues at the positions corresponding to positions L234 and L235 in a human IgG1 heavy chain according to Eu numbering are F and E, respectively. It is understood that in addition to modifications of amino acid positions L234 and L235, further positions may be modified.
  • the antibody according to the invention may comprise a first and a second heavy chain, wherein in both the first and the second heavy chain, the amino acid residue at the position corresponding to position D265 in a human IgG1 heavy chain according to Eu numbering is A.
  • Further embodiments of the invention provide antibodies wherein, in at least one, such as in both, of said first and second heavy chains the amino acids in the positions corresponding to positions L234, L235, and D265 in a human IgG1 heavy chain, are F, E, and A, respectively.
  • antibodies which have the combination of three amino acid substitutions L234F, L235E and D265A and in addition the K409R or the F405L mutation disclosed herein above may be termed with the suffix “FEAR” or “FEAL”, respectively.
  • an amino acid sequence of a wild type IgG1 heavy chain constant region is identified herein as SEQ ID NO: 57.
  • the antibody of the invention may comprise an IgG1 heavy chain constant region carrying the F405L substitution and may have the amino acid sequence set forth in SEQ ID NO: 58 and/or an IgG1 heavy chain constant region carrying the K409R substitution and may have the amino acid sequence set forth in SEQ ID NO: 62.
  • An amino acid sequence of an IgG1 heavy chain constant region carrying the L234F, L235E and D265A substitutions is identified herein as SEQ ID NO: 59.
  • An amino acid sequence of an IgG1 heavy chain constant region carrying the L234F, L235E, D265A and F405L substitutions is identified herein as SEQ ID NO: 60.
  • An amino acid sequence of an IgG1 heavy chain constant region carrying the L234F, L235E, D265A and K409R substitutions is identified herein as SEQ ID NO: 61.
  • the constant region sequences listed in SEQ ID NOs. 57-62 list a terminal lysine (K), such sequences were used in the example section herein.
  • the origin of this lysine is a naturally occurring sequence found in humans from which these Fc regions are derived.
  • this terminal lysine can be cleaved off by proteolysis by endogenous carboxypeptidase(s), resulting in a constant region having the same sequence but lacking the C-terminal lysine.
  • the DNA encoding this terminal lysine can be omitted from the sequence such that antibodies are produced without the lysine.
  • Antibodies produced from nucleic acid sequences that either do, or do not encode a terminal lysine are substantially identical in sequence and in function since the degree of processing of the terminal lysine is typically high when e.g. using antibodies produced in CHO-based production systems (Dick, L. W. et al. Biotechnol. Bioeng. 2008; 100: 1132-1143).
  • antibodies in accordance with the invention can be generated without encoding or having a terminal lysine such as listed herein. For manufacturing purposes, antibodies can thus be generated without having a terminal lysine.
  • the present invention further provides an antibody, wherein
  • the invention provides an antibody, wherein
  • the antibody comprises a kappa ( ⁇ ) light chain.
  • the sequence of particular embodiments of the invention concerning bispecific antibodies, the kappa light chain comprises the CDR1, -2 and -3 sequences of a B7H4 antibody light chain as disclosed above.
  • the antibody according to any one of the preceding claims wherein said antibody comprises a lambda (A) light chain.
  • the lambda light chain comprises the CDR1, -2 and -3 sequences of a CD3 antibody light chain as disclosed above, in particular a the CDR1, -2 and -3 sequences of a CD3 antibody having reduced affinity for CD3 as disclosed above.
  • the amino acid sequence of a kappa light chain constant region is included herein as SEQ ID NO: 63 and the amino acid sequence of a lambda light chain constant region is included herein as SEQ ID NO: 64.
  • the antibody comprises a lambda (A) light chain and a kappa ( ⁇ ) light chain; e.g. an antibody with a heavy chain and a lambda light chain which comprise the binding region capable of binding to CD3, and a heavy chain and a kappa light chain which comprise the binding region capable of binding to B7H4.
  • A lambda
  • kappa
  • said antigen binding region capable of binding to human B7H4 is comprised in a heavy chain and a light chain, said heavy chain comprising said VH region and an IgG1 heavy chain constant region and said light chain comprising said VL region and a kappa light chain constant region; and said antigen binding region capable of binding to human CD3 is comprised in a heavy chain and a light chain, said heavy chain comprising said VH region and an IgG1 heavy chain constant region and said light chain comprising said VL region and a lambda light chain constant region. More preferably, in said bispecific antibody, one IgG1 heavy chain constant region is as defined in SEQ ID NO.
  • IgG1 heavy chain constant regions as defined in SEQ ID NO. 60 and 61 may have their terminal lysines deleted.
  • Antibodies such as bispecific antibodies, as described herein that can bind to human CD3 and human B7H4 can advantageously target T cells to human B7H4 expressing cancer cells, thereby inducing T-cell mediated killing of said cancer cells.
  • T cells to human B7H4 expressing cancer cells, thereby inducing T-cell mediated killing of said cancer cells.
  • the antibody in accordance with the invention is devoid of, or has reduced Fc-mediated effector function, and furthermore, the antibody:
  • the antibody in accordance with the invention may be devoid of, or has reduced Fc-mediated effector function, and, furthermore capable of inducing T-cell mediated cytotoxicity antibody, wherein cytoxicity is assessed in an in vitro IC50 assay comprising:
  • purified T-cells may also be provided in step i).
  • PBMCs peripheral blood mononuclear cells
  • the antibody may have an IC50 in the range of 0.001-2 microgram/ml, wherein the IC50 is determined in an in vitro cytotoxicity assay comprising the steps of:
  • the antibody may have an IC50 in the range of 0.001-5 microgram/ml, wherein the IC50 is determined in an in vitro cytotoxicity assay comprising the steps of:
  • the antibody in accordance with the invention may have an IC50 in the range of 0.001-5 microgram/ml. In one embodiment, the antibody in accordance with the invention may have an IC50 in the range of 0.001-2 microgram/ml. In another embodiment, the antibody in accordance with the invention may have an IC50 is in the range of 0.001-0.03 microgram/ml. In still a further embodiment, the IC50 may be in the range of 0.05-2 microgram/ml. In yet another further embodiment, the IC50 may be in the range of 0.05-5 microgram/ml. Said IC50 may be determined using a method such as described in Example 12.
  • the ability of the antibody in accordance with the invention to mediate T cell activation is determined in an in vitro assay comprising the steps of:
  • Exemplary cytokines that can be e.g. detected are e.g. IFN- ⁇ , such as e.g. described in example 13.
  • B7H4-expressing tumor cells are human B7H4-expressing tumors, such as primary tumors, or tumor cell lines selected from the group consisting of MCF-7, MDA-MB-468, SK-BR3, NIH-OVCAR-3, and HCC1954.
  • an antibody comprising an antigen-binding region capable of binding to human B7H4, wherein said antigen-binding region capable of binding to human B7H4 comprises:
  • Such antibodies do not necessarily comprise an antigen-binding region that binds to CD3. Such antibodies may be useful, e.g. in kits and assays for detecting B7H4. Such antibodies may also be useful in the treatment of cancer. Hence, such an antibody may be monospecific antibody binding to B7H4. Such an antibody may be a bivalent antibody.
  • such an antibody is an antibody comprising a heavy chain constant region which is a human IgG1 constant region.
  • a heavy chain constant region such as listed in SEQ ID NO. 57-62.
  • a preferred light chain constant region is a kappa light chain, such as listed in SEQ ID NO. 63.
  • the antibody provided herein may bind to an epitope or antibody binding region on human B7H4 comprising one or more of the amino acid residues S151, V157, D158, Y159, E164, L166, W173, P175, P177, V179, W181, F199, M208, V210, T222, Y223, V240, E242 and I245; the numbering of each amino acid residue referring to its position in SEQ ID NO: 1.
  • the antibody provided herein may bind to an epitope or antibody binding region on human B7H4 comprising one or more of the amino acid residues V157, D158, Y159, E164, L166; the numbering of each amino acid residue referring to its position in SEQ ID NO: 1.
  • the antibody provided herein may bind to an epitope or antibody binding region on human B7H4 comprising the amino acid residues S151, V157, D158, Y159, E164, L166, W173, P175, P177, V179, W181, F199, M208, V210, T222, Y223, V240, E242 and I245; the numbering of each amino acid residue referring to its position in SEQ ID NO: 1.
  • the antibody provided herein may bind to an epitope or antibody binding region on human B7H4 comprising the amino acid residues V157, D158, Y159, E164, L166; the numbering of each amino acid residue referring to its position in SEQ ID NO: 1.
  • any one or more of these amino acid residues i.e. S151, V157, D158, Y159, E164, L166, W173, P175, P177, V179, W181, F199, M208, V210, T222, Y223, V240, E242 and I245 is/are directly involved in binding of the antibody, such as by way of non-covalent interactions; e.g with amino acid residues within the CDR sequences of the antibody.
  • amino acid residues comprised by said epitope or antibody binding region and optionally the one or more additional amino acid residues which are indirectly involved in binding may be identified by alanine scanning of human B7H4 having the amino acid sequence set forth in SEQ ID NO: 1 or the extracellular domain sequence of SEQ ID NO: 1.
  • the alanine scanning may in particular be performed as set forth or essentially as set forth in Example 7 herein.
  • alanine scanning may be performed by a procedure comprising the steps of:
  • an antibody may also be a bispecific antibody comprising in addition to an antigen-binding region capable of binding to B7H4 another antigen-binding region.
  • another antigen-binding region may be an antigen-binding region capable of binding to human CD3.
  • antigen-binding region capable of binding to human CD3 may be antigen-binding regions capable of binding to CD3 as described and disclosed herein.
  • said antigen binding region capable of binding to human B7H4 is comprised in an heavy chain and a light chain, said heavy chain comprising said VH region and an IgG1 heavy chain constant region and said light chain comprising said VL region and a kappa light chain constant region; and wherein said antigen binding region capable of binding to human CD3 is comprised in a heavy chain and a light chain, said heavy chain comprising said VH region and an IgG1 heavy chain constant region and said light chain comprising said VL region and a lambda light chain constant region. More preferably, in such a bispecific antibody, one IgG1 heavy chain constant region is as defined in SEQ ID NO.
  • a highly preferred bispecific antibody in accordance with the invention is as described and used in the example section, and is referred to as BsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR.
  • a bispecific antibody capable of binding human CD3 and human B7H4 comprising:
  • human IgG1 heavy chain constant regions as defined herein may encompass substitutions as defined herein (e.g. FEAR/FEAL), or the like. It is also understood that the human IgG1 heavy chain constant region may have its terminal lysine (K) deleted.
  • a bispecific antibody capable of binding human CD3 and human B7H4 comprising:
  • the human IgG1 heavy chain constant region may have its terminal lysine (K) deleted.
  • a bispecific antibody capable of binding human CD3 and human B7H4 comprising:
  • a bispecific antibody capable of binding human CD3 and human B7H4 comprising:
  • Another strategy to promote formation of heterodimers over homodimers is a “knob-into-hole” strategy in which a protuberance is introduced on a first heavy-chain polypeptide and a corresponding cavity in a second heavy-chain polypeptide, such that the protuberance can be positioned in the cavity at the interface of these two heavy chains so as to promote heterodimer formation and hinder homodimer formation.
  • “Protuberances” are constructed by replacing small amino-acid side-chains from the interface of the first polypeptide with larger side chains.
  • Compensatory “cavities” of identical or similar size to the protuberances are created in the interface of the second polypeptide by replacing large amino-acid side-chains with smaller ones (U.S. Pat. No.
  • EP1870459 Choi
  • WO2009089004 Amgen
  • EP1870459 Choi
  • WO2009089004 Amgen
  • one or more residues that make up the CH3-CH3 interface in both CH3 domains are replaced with a charged amino acid such that homodimer formation is electrostatically unfavorable and heterodimerization is electrostatically favorable.
  • WO2007110205 Merck
  • bispecific antibodies Another in vitro method for producing bispecific antibodies has been described in WO2008119353 (Genmab), wherein a bispecific antibody is formed by “Fab-arm” or “half-molecule” exchange (swapping of a heavy chain and attached light chain) between two monospecific IgG4- or IgG4-like antibodies upon incubation under reducing conditions.
  • the resulting product is a bispecific antibody having two Fab arms which may comprise different sequences.
  • a preferred method for preparing the bispecific CD3xB7H4 antibodies of the present invention includes methods described in WO2011131746 and WO13060867 (Genmab) comprising the following steps:
  • the said first antibody together with said second antibody are incubated under reducing conditions sufficient to allow the cysteines in the hinge region to undergo disulfide-bond isomerization, wherein the heterodimeric interaction between said first and second antibodies in the resulting heterodimeric antibody is such that no Fab-arm exchange occurs at 0.5 mM GSH after 24 hours at 37° C.
  • step c) the heavy-chain disulfide bonds in the hinge regions of the parent antibodies are reduced and the resulting cysteines are then able to form inter heavy-chain disulfide bond with cysteine residues of another parent antibody molecule (originally with a different specificity).
  • the reducing conditions in step c) comprise the addition of a reducing agent, e.g.
  • step c) comprises restoring the conditions to become non-reducing or less reducing, for example by removal of a reducing agent, e.g. by desalting.
  • any of the CD3 and B7H4 antibodies described herein may be used.
  • the CD3 and B7H4 antibodies, respectively may be chosen so as to obtain a bispecific CD3xB7H4 antibody as described herein.
  • said first and/or second antibodies are full-length antibodies.
  • the Fc regions of the first and second antibodies may be of any isotype, including, but not limited to, IgG1, IgG2, IgG3 or IgG4.
  • the Fc regions of both said first and said second antibodies are of the IgG1 isotype.
  • one of the Fc regions of said antibodies is of the IgG1 isotype and the other of the IgG4 isotype.
  • the resulting bispecific antibody comprises an Fc region of an IgG1 and an Fc region of IgG4 and may thus have interesting intermediate properties with respect to activation of effector functions.
  • one of the antibody starting proteins has been engineered to not bind Protein A, thus allowing to separate the heterodimeric protein from said homodimeric starting protein by passing the product over a protein A column.
  • the sequences of the first and second CH3 regions of the homodimeric starting antibodies are different and are such that the heterodimeric interaction between said first and second CH3 regions is stronger than each of the homodimeric interactions of said first and second CH3 regions. More details on these interactions and how they can be achieved are provided in WO2011131746 and WO2013060867 (Genmab), which are hereby incorporated by reference in their entirety.
  • a stable bispecific CD3xB7H4 antibody can be obtained at high yield using the above method of the invention on the basis of two homodimeric starting antibodies which bind CD3 and B7H4, respectively, and contain only a few, fairly conservative, asymmetrical mutations in the CH3 regions.
  • Asymmetrical mutations mean that the sequences of said first and second CH3 regions contain amino acid substitutions at non-identical positions.
  • the bispecific antibodies of the invention may also be obtained by co-expression of constructs encoding the first and second polypeptides in a single cell.
  • the invention relates to a method for producing a bispecific antibody, said method comprising the following steps:
  • the present invention also relates to a recombinant eukaryotic or prokaryotic host cell which produces a bispecific antibody of the present invention.
  • Suitable expression vectors including promoters, enhancers, etc., and suitable host cells for the production of antibodies are well-known in the art.
  • suitable host cells include yeast, bacterial and mammalian cells, such as CHO or HEK cells.
  • a method for producing an antibody capable of binding to both B7H4 and CD3 in accordance with the invention comprising the steps of:
  • the steps of providing an antibody capable of binding to B7H4 and/or CD3, may comprise the steps of
  • the invention furthermore provides for
  • nucleic acids comprising:
  • the nucleic acid, or one or more nucleic acids, as defined herein can be RNA or DNA.
  • the nucleic acid, or one or more nucleic acids, as defined herein may be for use in expression in mammalian cells.
  • the invention provides for a cell or cells, comprising a nucleic acid, or comprising one or more nucleic acids, as defined herein.
  • the nucleic acid in the context of the present invention may be an expression vector, which may be any suitable vector, including chromosomal, non-chromosomal, and synthetic nucleic acid vectors (a nucleic acid sequence comprising a suitable set of expression control elements).
  • suitable vector including chromosomal, non-chromosomal, and synthetic nucleic acid vectors (a nucleic acid sequence comprising a suitable set of expression control elements).
  • suitable vectors include derivatives of SV40, bacterial plasmids, phage DNA, baculovirus, yeast plasmids, vectors derived from combinations of plasmids and phage DNA, and viral nucleic acid (RNA or DNA) vectors.
  • a B7H4 or a CD3 antibody-encoding nucleic acid is comprised in a naked DNA or RNA vector, including, for example, a linear expression element (as described in for instance Sykes and Johnston, Nat Biotech 17, 355 59 (1997)), a compacted nucleic acid vector (as described in for instance U.S. Pat. No.
  • a plasmid vector such as pBR322, pUC 19/18, or pUC 118/119, a “midge” minimally-sized nucleic acid vector (as described in for instance Schakowski et al., Mol Ther 3, 793 800 (2001)), or as a precipitated nucleic acid vector construct, such as a CaP04-precipitated construct (as described in for instance WO200046147, Benvenisty and Reshef, PNAS USA 83, 9551 55 (1986), Wigler et al., Cell 14, 725 (1978), and Coraro and Pearson, Somatic Cell Genetics 7, 603 (1981)).
  • Such nucleic acid vectors and the usage thereof are well known in the art (see for instance U.S. Pat. Nos. 5,589,466 and 5,973,972).
  • the vector is suitable for expression of the B7H4 antibody and/or the CD3 antibody in a bacterial cell.
  • expression vectors such as BlueScript (Stratagene), pIN vectors (Van Heeke & Schuster, J Biol Chem 264, 5503 5509 (1989), pET vectors (Novagen, Madison Wis.) and the like).
  • An expression vector may also or alternatively be a vector suitable for expression in a yeast system. Any vector suitable for expression in a yeast system may be employed. Suitable vectors include, for example, vectors comprising constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH (reviewed in: F. Ausubel et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley InterScience New York (1987), and Grant et al., Methods in Enzymol 153, 516 544 (1987)).
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH
  • a nucleic acid and/or expression vector may also comprises a nucleic acid sequence encoding a secretion/localization sequence, which can target a polypeptide, such as a nascent polypeptide chain, to the periplasmic space or into cell culture media. Such sequences are known in the art, and include secretion leader or signal peptides.
  • the nucleic acid and/or expression vector may comprise any suitable elements facilitating expression, i.e. transcription and/or translation of the nucleic acid such that the components of the (bispecific) antibodies are expressed.
  • the nucleic acid and/or vector be associated with any suitable promoter, enhancer, and other expression-facilitating elements.
  • Such elements include strong expression promoters (e.g., human CMV IE promoter/enhancer as well as RSV, SV40, SL3 3, MMTV, and HIV LTR promoters), effective poly (A) termination sequences, an origin of replication for plasmid product in E. coli , an antibiotic resistance gene as selectable marker, and/or a convenient cloning site (e.g., a polylinker). Nucleic acids may also comprise an inducible promoter as opposed to a constitutive promoter such as CMV IE.
  • strong expression promoters e.g., human CMV IE promoter/enhancer as well as RSV, SV40, SL3 3, MMTV, and HIV LTR promoters
  • effective poly (A) termination sequences e.g., an origin of replication for plasmid product in E. coli
  • an antibiotic resistance gene as selectable marker
  • Nucleic acids may also comprise an inducible promoter as opposed to a constitutive promoter such as
  • the B7H4 and/or CD3 antibody-encoding expression vector may be positioned in and/or delivered to a cell.
  • the invention relates to a host cell comprising the nucleic acid or vector as defined herein.
  • the cell may be of human origin, such as a human embryonic kidney (HEK) cell, such as a HEK/Expi cell, or can be of rodent origin, such as a Chinese hamster ovary cell, such as a CHO/N50 cell.
  • the invention provides for a composition comprising an antibody as defined herein.
  • a composition is a pharmaceutical composition, i.e. the antibody is comprised in a pharmaceutically acceptable carrier.
  • the pharmaceutical composition of the present invention may contain a bispecific antibody of the present invention targeting both B7H4 and CD3.
  • the pharmaceutical composition may also comprise an antibody targeting B7H4.
  • the pharmaceutical composition may also comprise a combination of antibodies, including an antibody targeting B7H4 and/or a bispecific antibody in accordance with the present invention.
  • a pharmaceutical composition may be formulated in accordance with conventional techniques such as those disclosed in Remington: The Science and Practice of Pharmacy, 19th Edition, Gennaro, Ed., Mack Publishing Co., Easton, Pa., 1995.
  • a pharmaceutical composition of the present invention may e.g. include diluents, fillers, salts, buffers, detergents (e.g., a nonionic detergent, such as Tween-20 or Tween-80), stabilizers (e.g., sugars or protein-free amino acids), preservatives, tissue fixatives, solubilizers, and/or other materials suitable for inclusion in a pharmaceutical composition.
  • detergents e.g., a nonionic detergent, such as Tween-20 or Tween-80
  • stabilizers e.g., sugars or protein-free amino acids
  • preservatives e.g., tissue fixatives, solubilizers, and/or other materials suitable for inclusion in a pharmaceutical composition.
  • the antibody, composition, or pharmaceutical composition in accordance with the invention is preferably for use as a medicament.
  • the antibody, composition, or pharmaceutical composition in accordance with the invention is preferably for use in the treatment of disease.
  • Bispecific antibodies of the invention may be used for a number of purposes.
  • the bispecific antibodies of the invention may be used for the treatment of various forms of cancer, including metastatic cancer and refractory cancer.
  • the cancer may be of the solid tumor type.
  • bispecific antibodies according to the invention may be useful in therapeutic settings in which specific targeting and T cell-mediated killing of cells that express B7H4 is desired.
  • the present invention provides a method for treating a cancer in a subject, which method comprises administration of a therapeutically effective amount of a bispecific B7H4xCD3 antibody of the present invention.
  • the present invention provides a method for treating a disorder involving cells expressing B7H4, in a subject, which method comprises administration of a therapeutically effective amount of a bispecific antibody of the present invention.
  • the present invention provides a method for treating a cancer in a subject, which method comprises administration of a therapeutically effective amount of an antibody capable of binding to human B7H4 of the present invention.
  • the present invention provides a method for treating a disorder involving cells expressing B7H4, in a subject, which method comprises administration of a therapeutically effective amount of a monospecific antibody of the present invention that is capable of binding to human B7H4.
  • suitable diseases that can be contemplated in methods and uses in accordance with the invention are cancer.
  • Said cancer most preferably is characterized by expression of B7H4.
  • Expression of B7H4 in a cancer can easily be determined using methods known in the art, such as PCR, immunostaining, or FACS analysis, i.e. detecting expression of B7H4 transcript and/or protein.
  • the antibodies as described herein that are capable of binding to human B7H4 may be used e.g. in immunostaining and/or FACS analysis or the like.
  • Cancers that can express B7H4 include Breast cancer, Uterine/endometrial cancer, Uterine carcinosarcoma cancer, Ovarian cancer, Cervical cancer, Non-small cell lung cancer (squamous cell carcinoma and adenocarcinoma), Head and neck squamous cell carcinoma, Bladder cancer, esophageal cancer, cholangiocarcinoma, Pancreatic cancer, Stomach cancer, Renal cancer and Prostate cancer.
  • Cancers that can express B7H4 include cancers such as cancers of the stomach, cholangiocarcinoma, bladder cancer, non small cell lung cancer (in particular squamous NSCLC), pancreatic cancer, cervical cancer, head and neck cancer, breast cancer (including triple negative breast cancer), ovarian cancer and uterine cancer.
  • cancers selected from uterine carcinosarcoma (UCS), bladder urothelial carcinoma (BLCA), pancreatic adenocarcinoma (PAAD), lung squamous cell carcinoma (LUSC), breast invasive carcinoma (BRCA), uterine corpus endometrial carcinoma (UCEC), ovarian serous cystadenocarcinoma (OV) and cholangiocarcinoma (CHOL).
  • UCS uterine carcinosarcoma
  • BLCA bladder urothelial carcinoma
  • PAAD pancreatic adenocarcinoma
  • LUSC lung squamous cell carcinoma
  • BRCA breast invasive carcinoma
  • UCEC uterine corpus endometrial carcinoma
  • OV ovarian serous cystadenocarcinoma
  • CHOL cholangiocarcinoma
  • a patient being diagnosed with cancer may be subjected to an assessment of B7H4 expression in the cancer cells, and when B7H4 is detected, which may be in the range from low to high, such a patient may be selected for treatment with an antibody in accordance with the invention.
  • Patients diagnosed with having cancer of the stomach, cholangiocarcinoma, bladder cancer, non small cell lung cancer (in particular squamous NSCLC), pancreatic cancer, cervical cancer, head and neck cancer, breast cancer (including triple negative breast cancer), ovarian cancer or uterine cancer may be subjected to such test.
  • a patient being diagnosed with having uterine carcinosarcoma (UCS), bladder urothelial carcinoma (BLCA), pancreatic adenocarcinoma (PAAD), lung squamous cell carcinoma (LUSC), breast invasive carcinoma (BRCA), uterine corpus endometrial carcinoma (UCEC), ovarian serous cystadenocarcinoma (OV) or cholangiocarcinoma (CHOL), may be subjected to such test.
  • UCS uterine carcinosarcoma
  • BLCA bladder urothelial carcinoma
  • PAAD pancreatic adenocarcinoma
  • LUSC lung squamous cell carcinoma
  • BRCA breast invasive carcinoma
  • UCEC uterine corpus endometrial carcinoma
  • OV ovarian serous cystadenocarcinoma
  • CHOL cholangiocarcinoma
  • the invention further provides a kit-of-parts comprising an antibody as disclosed above, such as a kit for use as a companion diagnostic/for identifying within a population of patients, those patients which have a propensity to respond to treatment with an antibody as defined herein above or an immunoconjugate or antibody-drug conjugate (ADC) as defined herein above, or for predicting efficacy or anti-tumor activity of said antibody or immunoconjugate or ADC when used in treatment of a patient, the kit comprising an antibody as defined above; and instructions for use of said kit.
  • kit-of-parts comprising an antibody as disclosed above, such as a kit for use as a companion diagnostic/for identifying within a population of patients, those patients which have a propensity to respond to treatment with an antibody as defined herein above or an immunoconjugate or antibody-drug conjugate (ADC) as defined herein above, or for predicting efficacy or anti-tumor activity of said antibody or immunoconjugate or ADC when used in treatment of a
  • kits-of-parts such as a kit for use as a companion diagnostic/for identifying within a population of patients those patients which have a propensity to respond to treatment with an antibody as defined in any one of claims 1 to 55 , comprising an antibody as defined in any one of claims 1 to 55 ; and instructions for use of said kit.
  • the invention relates to a diagnostic composition comprising a bispecific CD3xB7H4 antibody as defined herein, or a B7H4 antibody as defined herein, and to its use.
  • the invention relates to a kit for detecting cross-linking between CD3- and B7H4 expressing cells, in a sample derived from a patient, comprising
  • the present invention provides a kit for diagnosis of cancer comprising a container comprising a bispecific CD3xB7H4 antibody, and one or more reagents for detecting cross-linking of B7H4 expressing cells and CD3 expressing cells.
  • Reagents may include, for example, fluorescent tags, enzymatic tags, or other detectable tags.
  • the reagents may also include secondary or tertiary antibodies or reagents for enzymatic reactions, wherein the enzymatic reactions produce a product that may be visualized.
  • the invention relates to a method for detecting whether cross-linking between CD3- and B7H4-expressing cells occurs in a sample derived from a patient, upon administration of a bispecific antibody according to any one of the embodiments as disclosed herein, comprising the steps of:
  • B7H4 (Uniprot accession no. Q7Z7D3), cynomolgus monkey ( Macaca fascicularis ) B7H4 transcript 1 (Uniprot accession no. A0A2K5U6P5), dog ( Canis familiaris ) B7H4 (Uniprot accession no. F1P8R9), rabbit ( Oryctolagus cuniculus ) B7H4 (Uniprot accession no. G1TQE8), rat ( Rattus norvegicus ) B7H4 (Uniprot accession no.
  • GCCGCCACC Kozak, M., Gene 1999; 234(2):187-208.
  • pSB a mammalian expression vector containing Sleeping Beauty inverted terminal repeats flanking an expression cassette consisting of a CMV promoter and HSV-TK polyA signal.
  • FreestyleTM 293-F (a HEK-293 subclone adapted to suspension growth and chemically defined Freestyle medium [HEK-293F]) cells were obtained from Invitrogen (cat. no. R790-07) and transfected with the constructs described supra, using 293fectin (Invitrogen, cat. no. 12347-019) according to the manufacturer's instructions.
  • B7H4ECD-FcHisC was expressed using the Expi293F expression platform (Thermo Fisher Scientific, Waltham, Mass., USA, cat. no. A14527) essentially as described by the manufacturer.
  • the His-tag enables purification with immobilized metal affinity chromatography Ni-NTA.
  • the His-tagged protein binds strongly to the column material, while other proteins present in the culture supernatant do not bind or bind weakly compared to the His-tagged proteins and elute in the flow-through.
  • the column was washed in order to remove weakly bound proteins.
  • the strongly bound His-tagged proteins were then eluted with a buffer containing imidazole, which competes with the binding of His to Ni 2+ .
  • the eluent was removed by buffer exchange on a desalting column.
  • OmniRat® animals transgenic rats expressing a diversified repertoire of antibodies with fully human idiotypes; Ligand Pharmaceuticals Inc., San Diego, USA
  • adjuvant system Sigma-Aldrich, St. Louis, Mo., USA, cat. no. S6322
  • CFA Complete Freund Adjuvant (1 s
  • RNA from hybridomas producing B7H4 specific antibody was extracted and 5′-RACE-complementary DNA (cDNA) was prepared from 100 ng total RNA, using the SMART RACE cDNA Amplification kit (Clontech), according to the manufacturer's instructions.
  • VH and VL coding regions were amplified by PCR and cloned directly, in frame, in the p33G1f, p33Kappa and p33Lambda expression vectors (pcDNA3.3 based vectors with codon optimized human IgG1m(f), Kappa and Lambda constant domains respectively), by ligation independent cloning (Aslanidis, C. and P. J. de Jong, Nucleic Acids Res 1990; 18(20): 6069-74). The variable domains from these expression vectors were sequenced and CDRs were annotated according to IMGT definitions (Lefranc M P.
  • Clones with a correct Open Reading Frame were expressed and tested for binding to the antigen.
  • ORF Open Reading Frame
  • the sequences of variable regions of heavy and light chain were gene synthesized and cloned into an expression vector including a human IgG1 heavy chain containing the following amino acid mutations: L234F, L235E, D265A and K409R (FEAR) wherein the amino acid position number is according to Eu numbering (correspond to SEQ ID NO 60), and into expression vectors including human kappa or lambda light chain.
  • a variant with point mutation in the variable domains was generated to remove a cysteine residue, which potentially could generate undesired disulphide bridge formation, or to replace an Asparagine to Serine or germline residue to remove a potential N-linked glycosylation site.
  • a variant with an N52S substitution was made corresponding with a substitution in CDR2 (see TABLE 1, SEQ ID NOs. 25 and 29), and a further variant can have an N52Q substitution (SEQ ID NO. 31).
  • B7H4 antibodies in sera of immunized animals, or hybridoma and transfectoma culture supernatant was determined in a homogeneous binding assay. Samples were analyzed for binding of antibodies to HEK-293F cells transiently transfected with the constructs made to express full length B7H4 variants expressing human B7H4, cynomolgus monkey B7H4 or murine B7H4, or HEK-293F wild-type cells (negative control). Samples were added to the cells to allow antibody binding to B7H4.
  • test and control antibodies Serial dilutions of test and control antibodies (range 0.003 to 3 ⁇ g/mL in 2-fold dilution steps) were prepared and 2 ⁇ l antibody dilution was added to 5 ⁇ l of the cell/conjugate mixture in 1536 well plates (Greiner, cat. no. 789866). Plates were incubated at room temperature for 9 hours, and after which fluorescence intensity was determined using an ImageXpress Velos Laser Scanning Cytometer (Molecular Devices, LLC, Sunnyvale, Calif., USA) and total fluorescence was used as read-out. Samples were stated positive when counts were higher than 50 and counts x fluorescence was at least three times higher than the negative control.
  • WO2009073533 (referenced therein as SEQ ID No 2 and 7), corresponding herein to B7H4-C4 and relevant sequences of the variable domains are listed herein in TABLE 1 and include SEQ ID NO. 50 and 54; and US20190085080A1 corresponding herein to B7H4-C5 and relevant sequences of the variable domains are listed herein in TABLE 1 and include SEQ ID NO. 65 and 69.
  • the corresponding VH and VL antibody variable domain encoding sequences were synthesized and cloned into pcDNA3.3 based vectors with codon optimized human IgG1m(f) and Kappa or Lambda constant domains, or variants thereof, to produce monospecific and bispecific antibodies.
  • antibody IgG1-B7H4-CX -FEAL When reference is made to antibody IgG1-B7H4-CX -FEAL, this represents an antibody having the B7H4-CX variable regions, being of the IgG1 isotype, and having amino acid substitutions L234F, L235E, D265A and F409R in the constant region of the heavy chain.
  • the antibody b12 an HIV-1 gp120 specific antibody (Barbas, C F. J Mol Biol. 1993 Apr. 5; 230(3):812-23) was used in some examples as a negative control IgG1, or as the non-binding control Fab-arm of a control bispecific.
  • the codon optimized antibody encoding sequences for this control antibody were synthesized and cloned into pcDNA3.3 based vectors with codon optimized human IgG1m(f) and Kappa constant domains, or variants thereof.
  • the sequence of the variable heavy chain (VH) region and the sequence of the variable light chain (VL) region are included herein as SEQ ID NOs.: 14 and 15, respectively.
  • IgG1-huCD3-H1L1 (of which the variable heavy and light chain region sequences are listed herein in SEQ ID NO: 16 and 22) is described in Example 1 of WO2015/001085.
  • IgG1-huCD3-H1L1 is referred to herein as ‘IgG1-huCD3’.
  • Antibody IgG1-huCD3-H1L1-FEAL is a variant hereof with three amino acid substitutions in the Fc domain (L234F, L235E, D265A), in addition to an amino acid substitution that allows the generation of bispecific antibodies through controlled Fab-arm exchange (F405L), as described herein below. It has been shown that such mutations did not have effect on target binding of the antibodies in which they are introduced (see e.g. US 2015/0337049 and Engelberts et al., 2020, EBioMedicine 52: 102625).
  • IgG1-huCD3-H1L1-H101G (of which the variable heavy chain and light chain region sequences are listed as SEQ ID NO: 17 and 22 herein) is described in Example 2 of WO2017/009442.
  • IgG1-huCD3-H1L1-H101G will be referred to as ‘IgG1-huCD3-H101G’.
  • This variant comprises a substitution H101G in the variable heavy chain region sequence (compare SEQ ID NO.16 and 17), and has the same light chain as IgG1-huCD3-H1L1.
  • Antibody IgG1-huCD3-H101G-FEAL is a variant hereof with amino acid substitutions L234F, L235E, D265A and F405L.
  • Target binding affinity of B7H4 antibodies was determined by label-free biolayer interferometry (BLI) on an Octet HTX instrument (FortéBio). Experiments were carried out while shaking at 1,000 RPM at 30° C. Initially, the affinity of IgG1-B7H4-C1-N52S-FEAR, IgG1-B7H4-C2-FEAR, IgG1-B7H4-C3-FEAR, and IgG1-B7H4-C4-FEAR for human and mouse B7H4 was determined using BLI. Anti-Human IgG Fc Capture (AHC) biosensors (FortéBio, cat. no.
  • AHC sensors were regenerated by exposure to 10 mM glycine buffer pH 1.7 for 5 s, followed by neutralization in Sample Diluent for 5 s; both steps were repeated twice. Subsequently sensors were loaded again with antibody for the next cycle of kinetics measurements.
  • Data were acquired using Data Acquisition Software v9.0.0.49d (FortéBio) and analyzed with Data Analysis Software v9.0.0.12 (FortéBio). Data traces were corrected per antibody by subtraction of the reference sensor. The Y-axis was aligned to the last 10 s of the baseline, Interstep Correction alignment to dissociation and Savitzky-Golay filtering were applied. Data traces with a response ⁇ 0.05 nm were excluded from analysis. The data was fitted with the 1:1 Global Full fit model using a window of interest for the association and dissociation times set at 300 s and 200 s respectively.
  • Data were acquired using Data Acquisition Software v12.0.1.8 (FortéBio) and analyzed with Data Analysis Software v12.0.1.2 (FortéBio).
  • the data was fitted with the 1:1 Global Full Fit model using a window of interest for the association time of 200 s and a window of interest for the dissociation time of 200 s, except for IgG1-B7H4-C2-FEAR for which a 1,000 s dissociation time was used.
  • the dissociation time was chosen based upon R 2 value, visual inspection of the curve and at least 5% signal decay during the dissociation step.
  • Data traces generated with antigen concentrations higher than 100 nM were excluded from analysis for antibodies with an affinity below 50 nM.
  • the affinity of for cynomolgus monkey B7H4 was determined by BLI.
  • Amine Reactive 2 nd Generation (AR2G) biosensors (FortéBio, cat. no. 18-5092) were activated by reaction with 20 mM EDC (N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride) (FortéBio, cat. no. 18-1033) and 10 mM s-NHS (N-hydroxysulfosuccinimide sodium salt) (FortéBio, cat. no. 18-1067) for 300 s.
  • the activated sensors were loaded with 10 ⁇ g/mL recombinant hIgG1 Fc-tagged cynomolgus monkey B7H4 (Creative BioMart, cat. no.
  • VTCN1-1517R in 10 mM Sodium Acetate pH 4.0 (FortéBio, cat. no. 18-1068) for 600 s and quenched with 1 M ethanolamine pH 8.5 (FortéBio, cat. no. 18-1071) for 300 s.
  • the association (100 s) and dissociation (1,000 s) of functionally monovalent B7H4 binding by CD3xB7H4 bispecific antibodies was determined using a concentration range of 0.23-15 ⁇ g/mL (1.56-100 nM) with two-fold dilution steps in Sample Diluent.
  • a molecular mass of 150 kDa of the antibodies was used for calculations.
  • For each antibody a reference sensor was used, which was incubated with Sample Diluent instead of antibody.
  • Data were acquired using Data Acquisition Software v9.0.0.49d (FortéBio) and analyzed with Data Analysis Software v9.0.0.12 (FortéBio). Data traces were corrected per antibody by subtraction of the reference sensor. The Y-axis was aligned to the last 10 s of the baseline, Interstep Correction alignment to dissociation and Savitzky-Golay filtering were applied. Data traces with a response ⁇ 0.05 nm were excluded from analysis. The data was fitted with the 1:1 Global Full fit model using a window of interest for the association and dissociation times set at 100 s and 200 s respectively.
  • the data was fitted with the 1:1 Global Full fit model using a window of interest for the association time and dissociation time of 200 s.
  • the dissociation time was chosen based upon R 2 value, visual inspection of the curve and at least 5% signal decay during the dissociation step.
  • Data traces generated with antibody concentrations higher than 200 nM were excluded from analysis for antibodies with an affinity below 50 nM. All results were determined with an R 2 of at least 0.98.
  • K D (M) refers to the equilibrium dissociation constant of the antibody-antigen interaction, and is obtained by dividing k d by k a .
  • k d (sec ⁇ 1 ) refers to the dissociation rate constant of the antibody-antigen interaction. This is sometimes also referred to as the k off value or off-rate.
  • k a (M ⁇ 1 ⁇ sec ⁇ 1 ) refers to the association rate constant of the antibody-antigen interaction. This is sometimes also referred to as the k on value or on-rate.
  • Tables 4 and 5 show the results of the first and the second experiment in which the association rate constant k a (1/Ms), dissociation rate constant k d (1/s) and equilibrium dissociation constant K D (M) of the indicated antibodies for human B7H4 were determined by biolayer interferometry.
  • Tables 6 and 7 show the results of two experiments in which the k a (1/Ms), k d (1/s), and K D (M) of the indicated antibodies for mouse B7H4 were determined by biolayer interferometry.
  • Tables 8 and 9 show the results of two experiments in which the k a (1/Ms), k d (1/s), and K D (M) of the indicated antibodies for cynomolgus monkey B7H4 were determined by biolayer interferometry.
  • Binding affinities of IgG1-huCD3-FEAL and IgG1-huCD3-H101G-FEAL were determined as described in Example 7 of WO2017/009442.
  • binding affinities of selected CD3 antibodies in an IgG1-huCD3-FEAL format for recombinant soluble CD3 ⁇ (CD3E27-GSKa) (mature protein of SEQ ID NO: 13) were determined using biolayer interferometry on a ForteBio Octet HTX (ForteBio).
  • Anti-human Fc capture biosensors (ForteBio, cat. no. 18-5060) were loaded for 600 s with hIgG (1 ⁇ g/mL).
  • CD3E27-GSKa concentration range of 27.11 ⁇ g/mL-0.04 ⁇ g/mL (1000 nM-1.4 nM) with three-fold dilution steps (sample diluent, ForteBio, cat. no. 18-5028).
  • sample diluent ForteBio, cat. no. 18-5028.
  • the theoretical molecular mass of CD3E27-GSKa based on the amino acid sequence was used, i.e. 27.11 kDa. Experiments were carried out while shaking at 1000 rpm and at 30° C. Each antibody was tested in at least two independent experiments.
  • Table 10 shows the association rate constant k a (1/Ms), dissociation rate constant k d (1/s) and equilibrium dissociation constant K D (M) for recombinant CD3 ⁇ determined by biolayer interferometry.
  • IgG1-huCD3-FEAL showed a relatively high (K D : 15 nM) binding affinity to recombinant CD3 ⁇ compared to IgG1-huCD3-H101G-FEAL (K D : 683 nM).
  • Antibody cross-block analysis inpitope binning in classical Sandwich format was performed by BLI on an Octet HTX instrument (FortéBio).
  • Amine Reactive 2 nd Generation (AR2G) biosensors were activated for 300 s with a solution of 20 mM EDC (N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride) (Sigma-Aldrich, cat. no. 03449) and 10 mM s-NHS (N-Hydroxysulfosuccinimide sodium salt) (Sigma-Aldrich, cat. no. 56485).
  • the activated AR2G sensors were loaded with 20 ⁇ g/mL first antibody in 10 mM Sodium Acetate pH 6.0 (FortéBio, cat. no.
  • the cross-block experiment was repeated to also include IgG1-B7H4-C5-FEAR and was performed as described above, with minor adaptations.
  • the experiment was carried out while shaking at 1,000 RPM and at 22° C. Data were acquired using Data Acquisition Software v12.0.1.8 (ForteBio) and analyzed with Data Analysis HT Software v12.0.1.55 (ForteBio). In general, responses >0.1 nm were considered non-cross-blocking antibodies, while responses ⁇ 0.1 nm were considered to be blocking antibody pairs.
  • Cross-block of antibodies is indicated by bold font, and non-blocking antibody combinations are unmarked (transparent background), showing that IgG1-B7H4-C1-N52S-FEAR, IgG1-B7H4-C3-FEAR, and IgG1-B7H4-C5-FEAR are cross-blocking with each other and not with IgG1-B7H4-C4-FEAR and IgG1-B7H4-C2-FEAR, and vice versa.
  • Bispecific antibodies were generated in vitro using the DuoBody® platform technology, i.e. 2-MEA-induced Fab-arm exchange as described in WO2011147986, WO2011131746 and WO2013060867 (Genmab) and Labrijn et al. (Labrijn et al., PNAS 2013, 110: 5145-50; Gramer et al., MAbs 2013, 5: 962-973).
  • IgG1 molecules carrying specific point mutations in the CH3 domain were generated: in one parental IgG1 antibody the F405L mutation (i.e. the CD3 antibodies in this application), in the other parental IgG1 antibody the K409R mutation (i.e. the B7H4 or control, HIV-1 gp120-specific, antibodies in this application).
  • the parental IgG1 antibodies included substitutions L234F, L235E, D265A (FEA).
  • the two parental antibodies were mixed in equal mass amounts in PBS buffer (Phosphate Buffered Saline; 8.7 mM HPO 4 2 ⁇ , 1.8 mM H 2 PO 4 ⁇ , 163.9 mM Na + , 140.3 mM Cl ⁇ , pH 7.4).
  • PBS buffer Phosphate Buffered Saline; 8.7 mM HPO 4 2 ⁇ , 1.8 mM H 2 PO 4 ⁇ , 163.9 mM Na + , 140.3 mM Cl ⁇ , pH 7.4
  • 2-mercaptoethylamine-HCl (2-MEA) was added to a final concentration of 75 mM and the reaction mixture was incubated at 31° C. for 5 h.
  • the 2-MEA was removed by dialysis into PBS buffer using 10 kDa molecular-weight cutoff Slide-A-Lyzer carriages (Thermo Fisher Scientific) according to the manufacturer's protocol in order to allow re-oxidation of the inter-chain disulfide bonds and formation of intact bispecific antibodies.
  • IgG1-B7H4-C1-FEAR having the VH and VL sequences set forth in SEQ ID NO: 25 and SEQ ID NO: 33.
  • IgG1-B7H4-C1-N52S-FEAR having the VH and VL sequences set forth in SEQ ID NO: 29 and SEQ ID NO: 33.
  • IgG1-B7H4-C2-FEAR having the VH and VL sequences set forth in SEQ ID NO: 43 and SEQ ID NO: 47).
  • IgG1-B7H4-C3-FEAR having the VH and VL sequences set forth in SEQ ID NO: 36 and SEQ ID NO: 40).
  • IgG1-B7H4-C4-FEAR having the VH and VL sequences set forth in SEQ ID NO: 50 and SEQ ID NO: 54).
  • IgG1-B7H4-C5-FEAR having the VH and VL sequences set forth in SEQ ID NO: 65 and SEQ ID NO: 69).
  • the annotation IgG1 indicates that full length antibodies of the IgG1 isotype were made, and the FEAR annotation indicates that the heavy chain constant regions contains amino acid substitutions L234F, L235E, D265A and K409R and the light chain constant regions were of the kappa type (SEQ ID NO. 61 and 63, respectively).
  • IgG1-huCD3-FEAL having the VH and VL sequences set forth in SEQ ID NO: 16 and SEQ ID NO: 22.
  • IgG1-huCD3-H101G-FEAL having the VH and VL sequences set forth in SEQ ID NO: 17 and SEQ ID NO: 22).
  • the annotation IgG1 indicates that full length antibodies of the IgG1 isotype were made, and the FEAL annotation indicates that the heavy chain constant regions contains amino acid substitutions L234F, L235E, D265A and F405L and the light chain constant regions were of the lambda type (SEQ ID NO. 60 and 64, respectively).
  • IgG1-b12-K409R having the VH and VL sequences set forth in SEQ ID NO: 14 and SEQ ID NO: 15).
  • the annotation IgG1 indicates that full length antibodies of the IgG1 isotype were made, and the K409R annotation indicates that the heavy chain constant regions contains amino acid substitution K409R and the light chain constant regions were of the kappa type (SEQ ID NO. 62 and 63, respectively).
  • the CD3 and B7H4 antibodies described above were combined to generate a bispecific antibody, having one antigen-binding region capable of binding human CD3 and the other antigen-binding region capable of binding B7H4, providing a bispecific antibody of the isotype IgG1, which is annotated as bsIgG1.
  • the B7H4 domain specificity of the B7H4 antibodies was determined using a panel of cells transfected to express human B7H4, human B7H3 (a structurally comparable protein with sufficient amino acid sequence difference in the extracellular domain) or two different human B7H4-B7H3 chimeric molecules. Expression constructs were prepared encoding human B7H4, human B7H3 (Uniprot accession no.
  • HEK cells were transiently transfected to express these constructs.
  • FIG. 1 shows that the IgC domain of B7H4 is involved in binding of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-FEALxB7H4-C4-FEAR, both the IgC and IgV domain of B7H4 are involved in binding of bsIgG1-huCD3-FEALxB7H4-C3-FEAR, and at least the IgV domain of B7H4 is involved in binding of bsIgG1-huCD3-FEALxB7H4-C2-FEAR.
  • variable domains For the C2 antibody from which the variable domains were used to created bsIgG1-huCD3-FEALxB7H4-C2-FEAR, it has been described that it binds to the IgV domain; the data in FIG. 1 indicates that the IgC domain is also involved in binding (WO2014159835 and Leong et al 2015, Mol. Pharmaceutics 12, 1717-1729).
  • FIG. 2 shows the dose-response curves, showing that the IgC domain of B7H4 is involved in binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR, in line with the findings of the alanine scanning library experiments.
  • the IgV domain is involved in the binding of bsIgG1-huCD3-H101G-FEALxB7H4-C2-FEAR, bsIgG1-huCD3-H101G-FEALxB7H4-C4-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C5-FEAR, whereas both the IgC and IgV domain appear involved in the binding of bsIgG1-huCD3-H101GFEALxB7H4-C3-FEAR.
  • a human B7H4 (Uniprot Q7Z7D3-1) single residue alanine library was synthesized (GeneArt) in which all amino acid residues in the extracellular domain of human B7H4 were individually mutated to alanines except for positions containing alanines or cysteines. Cysteines were not mutated to minimize the chance of structural disruption of the antigen.
  • the library was cloned in the pMAC expression vector containing a CMV/TK-polyA expression cassette, an Amp resistance gene and a pBR322 replication origin.
  • the antibodies C1-N52S, C2 and C3 were generated as recombinant monovalent antibodies as described in WO2007059782 with a mNeonGreen tag.
  • the wild type B7H4 and alanine mutants were expressed individually in FreeStyle HEK293 cells according to the manufacturer's instructions (Thermo Scientific).
  • One day post transfection the cells were harvested. Approximately 50,000 cells were incubated with 20 ⁇ L mNeoGreen labeled antibody of interest. Cells were incubated for 1 hour at room temperature. Subsequently, 150 ⁇ L FACS buffer was added and cells were washed twice with FACS buffer. Cells were resuspended in 30 ⁇ L fresh FACS buffer and analyzed by flow cytometry using an iQue Screener (Intellicyt Corporation, USA).
  • the average antibody binding per cell was determined as the geometric mean of the fluorescence intensity (gMFI) for the ungated cell population.
  • the gMFI is influenced by the affinity of the antibody for the B7H4 mutant and the expression level of the B7H4 mutant per cell. Since specific alanine mutations can impact the surface expression level of the mutant B7H4, and to correct for expression differences for each B7H4 mutant in general, data were normalized against the binding intensity of a non-cross blocking B7H4 specific reference antibody, using the following equation:
  • FIG. 3 shows the Fold Change in binding of the B7H4 antibodies to B7H4 variants with ala mutations in the ECD, with the amino acid residues where the Fold Change in binding was lower than the mean Fold Change ⁇ 1.5 ⁇ SD annotated.
  • the Fold Change is indicated in FIG. 3 as Z-score. The results indicate that:
  • binding of bispecific CD3xB7H4 antibodies and monospecific B7H4 antibodies to HEK-293F cells transiently transfected with human B7H4 or with cynomolgus monkey ( Macaca fascicularis ) B7H4 was analyzed by flow cytometry. Non-transfected HEK-293F cells were used as negative control; these cells were (also) confirmed not to express CD3.
  • Cells (3 ⁇ 10 4 cells/well) were incubated in polystyrene 96-well round-bottom plates (Greiner bio-one, cat. no. 650180) with serial dilutions of antibodies (ranging from 0.000458 to 30 ⁇ g/mL in 4-fold dilution steps) in 100 ⁇ L PBS/0.1% BSA/0.02% azide (staining buffer) at 4° C. for 30 min. Experiments were performed in technical duplicate. After washing twice in staining buffer, cells were incubated in 50 ⁇ L secondary antibody at 4° C. for 30 min.
  • R-Phycoerythrin (PE)-conjugated goat-anti-human IgG F(ab′) 2 (1:500 in FACS buffer; Jackson ImmunoResearch Laboratories, Inc., West Grove, Pa., cat. no. 109-116-098), was used. Cells were washed twice in staining buffer, re-suspended in 30 ⁇ L FACS buffer containing Topro-3 (1:10,000 dilution) and analyzed on an iQue Screener (Intellicyt Corporation, USA). Binding curves were analyzed using non-linear regression (sigmoidal dose-response with variable slope) using Graph Pad Prism V7.02 software (GraphPad Software, San Diego, Calif., USA).
  • FIG. 4 shows that both IgG1-B7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR bound to cells expressing human B7H4 or cynomolgus monkey B7H4.
  • FIG. 5 shows that IgG1-B7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR bound to B7H4 from dog, rabbit, rat and mouse to varying degrees; for each the apparent affinity (EC50) of the bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR was lower than that of IgG1-B7H4-C1-N52S-FEAR.
  • EC50 apparent affinity
  • the EC50s for binding to human and cynomolgus monkey B7H4 of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR and IgG1-B7H4-C1-N52S-FEAR were in a similar range.
  • FIG. 6 shows that binding to HEK cells transfected with human and cynomolgus B7H4 was similar for the tested antibodies. Similar results were obtained with cells expressing rabbit and dog B7H4.
  • binding of gG1-B7H4-C1-N52S-FEAR and IgG1-B7H4-C3-FEAR to rat B7H4 appeared lower relative to IgG1-B7H4-C4-FEAR, IgG1-B7H4-C2-FEAR, and IgG1-B7H4-C5-FEAR.
  • IgG1-B7H4-C4-FEAR, IgG1-B7H4-C2-FEAR and IgG1-B7H4-C5-FEAR bound to pig B7H4, binding of IgG1-B7H4-C1-052S-FEAR was very weak and only apparent at the highest antibody concentration tested. Binding of IgG1-B7H4-C3-FEAR to pig B7H4 was undetectable.
  • NIH-OVCAR-3 ovarian adenocarcinoma; ATCC, cat. no. HTB-161
  • HCC1954 breast ductal carcinoma; ATCC, cat. no. CRL-23378
  • Solid tumor cell lines typically do not express CD3.
  • tumor cell line HeLa that showed no detectable B7H4 expression (cervix adenocarcinoma; ATCC, cat. no. CCL-2) was used. Binding was analyzed by flow cytometry as described above.
  • FIG. 7 shows that IgG1-B7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR showed comparable dose-dependent binding to MCF-7 and MDA-MB-468 cells, with comparable maximum binding levels.
  • FIG. 8 shows dose-dependent binding of bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR to NIH-OVCAR-3 and HCC1954 cells, and lack of detectable binding to a non-B7H4 expressing cell line, HeLa.
  • FIG. 9 shows that bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR showed comparable dose-dependent binding to these cells, with comparable maximum binding levels.
  • FIG. 10 shows dose-dependent binding of the C1-N52S, C2, C3, C4, and C5 B7H4 antibodies in homodimer or bispecific antibody format to MDA-MB-468 and HCC1954 cells.
  • the antibodies based on C4 and C5 showed most efficient binding, the antibodies based on C1-N52S and C2 showed intermediate binding efficiency, and the antibodies based on C3 showed the lowest binding efficiency.
  • Maximum binding was comparable between the antibodies based on C1-N52S, C2, C4 and C5, but lower for the antibodies based on C3.
  • CD3 EF450 labeled; eBioscience, cat. no. 48-0037-42
  • CD45 BV786 labeled; Biolegend, cat. no. 304048
  • CD14 PE-Cy7 labeled; BD Biosciences, cat. no. 557742
  • CD86 PerCP-Cy5.5 labeled; Biolegend, cat. no. 305420
  • CD163 APC-Cy7 labeled; Biolegend, cat. no. 333622
  • EpCAM EpCAM
  • FAB9601N specific antibodies, at 4° C. for 30 min. After washing cells were resuspended in staining buffer and analyzed using a FACS Fortessa (BD Biosciences). Single cells were gated based on scatter FSC/SSC and live cells were identified by exclusion of FVS-BV510 positive cells. Tumor cells were identified as EpCAM positive cells.
  • an in vitro cytotoxicity assay was performed using B7H4-positive tumor cell lines as target cells and purified T cells as effector cells, with varying effector to target cell (E:T) ratios.
  • T cells were obtained from healthy human donor buffy coats (Sanquin, Amsterdam, The Netherlands) and isolated using the RosetteSepTM human T cell enrichment cocktail (Stemcell Technologies, France, cat. no. 15061) according to the manufacturer's instructions.
  • SK-BR3 cells (16,000 cells/well) were seeded into flat bottom 96-well plates (Greiner-bio-one, The Netherlands, cat. no. 655180) and left to adhere for 4 hours at 37° C.
  • T cells were added to tumor cells at an effector to target (E:T) ratio of 2:1, 4:1 or 8:1.
  • phenylarsine oxide As a positive control for cytotoxicity, cells were incubated with 16 ⁇ g/mL phenylarsine oxide (PAO; Sigma-Aldrich, cat. no. P3075; dissolved in dimethylsulfoxide [DMSO; Sigma-Adrich, cat. no. D2438]). AlamarBlue fluorescence, as a measure of metabolic activity of the tumor cell cultures and thus of viable tumor cells, was measured at 615 nm (OD615) on an EnVision plate reader (PerkinElmer). The absorbance of PAO-treated tumor cell samples was set as 0% viability and the absorbance of untreated tumor cell samples was set as 100% viability. The ‘percentage viable cells’ was calculated as follows:
  • % viable cells ([absorbance sample ⁇ absorbance PAO-treated target cells]/[absorbance untreated target cells ⁇ absorbance PAO-treated target cells]) ⁇ 100.
  • Dose-response curves and IC50 values were generated using non-linear regression analysis (sigmoidal dose-response with variable slope) using Graph Pad Prism V7.02 software (GraphPad Software, San Diego, Calif., USA).
  • FIG. 11 shows that T cell mediated cytotoxicity was observed at all E:T ratio's, with maximal tumor cell killing (less than 10% viable tumor cells) observed at an E:T ratio of 8:1.
  • Example 12 Induction of Cytotoxicity In Vitro in Various Tumor Cell Lines by CD3xB7H4 Bispecific Antibodies and Correlation with B7H4 Expression Level
  • the T cell-mediated kill of bispecific antibodies bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR of various B7H4 expressing tumor cell lines was determined in an in vitro cytotoxicity assay as described above, using an E:T ratio of 8:1.
  • the following cell lines were used: MCF-7, MDA-MB-486, SK-BR3, NIH-OVCAR-3, HCC1954, and NCI-H1650. From each incubation, 150 ⁇ L supernatants containing T cells was transferred to U-bottom 96 Well culture plates (CellStar, cat. no. 650180) prior to washing and alamarBlue incubation (to determine T cell activation and cytokine release, as described below)
  • B7H4 was quantified by quantitative flow cytometry (Human IgG calibrator, BioCytex) according to the manufacturer's instructions, using bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR to detect B7H4.
  • FIG. 12 shows both bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR induced dose-dependent T cell mediated cytotoxicity in MCF-7, MDA-MB-486, SK-BR3, NIH-OVCAR-3 and HCC1954 cells in vitro.
  • FIG. 13A No significant relation between tumor cell lysis and the level of B7H4 expression was observed for either bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR or bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR ( FIG. 13B ).
  • FIG. 13A No significant relation between tumor cell lysis and the level of B7H4 expression was observed for either bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR or bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR
  • FIG. 13B shows the IC50 of T cell-mediated kill, using T cells derived from 4-6 donors, in the presence of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR or bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR for each cell line, with the cell lines arranged from lowest to highest level of B7H4 expression. This means that T cell mediated killing can occur over a wide range of B7H4 expression levels.
  • Table 13 summarizes results across a panel of 5 cell lines and 4 donors.
  • IC50 range (4 donors each cell line) ( ⁇ g/ml) CD3- H101GxB7H4 CD3x67H4 cell line lowest highest lowest highest MCF7 0.55 1.29 0.012 0.025 OVCAR3 0.09 1.629 0.003 0.012 NCI-H16650 1.67 5.07 N.D. N.D. MDA-MB-468 0.08 0.16 0.001 0.004 HCC1954 0.06 0.22 0.001 0.008 SK-BR3 0.09 0.22 0.002 0.016
  • bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR also induced dose-dependent T-cell mediated cytotoxicity of the tested NCI-H1650 NSCLC cell line.
  • Example 13 Induction of T Cell Activation and Cytokine Production In Vitro by CD3xB7H4 Bispecific Antibodies in the Presence of B7H4-Positive Tumor Cells
  • T cells were stained for T cell markers CD3 (1:200; eBioscience, clone OKT3, conjugated to eFluor450), CD4 (1:50; eBioscience, clone OKT4, conjugated to APC-eFluor780), CD8 (1:100; Biolegend, clone RPA-T8, conjugated to AF700) and T cell activation markers CD69 (1:50; BD Biosciences, clone AB2439, conjugated to APC), CD25 (1:50; eBioscience, clone BC96, conjugated to PE-Cy7) and CD279/PD1 (1:50; Biolegend, clone EH12.2H7, conjugated to BV605).
  • CD3 1:200; eBioscience, clone OKT3, conjugated to eFluor450
  • CD4 (1:50; eBioscience, clone OKT4, conjugated to APC-eFluor780
  • Dose-response curves, EC50, EC90 and EC99 values were calculated using non-linear regression analysis (sigmoidal dose-response with variable slope) using GraphPad Prism V7.02 software (GraphPad Software, San Diego, Calif., USA).
  • FIG. 14A shows T cell activation in the presence of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR or bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR for the B7H4-positive tumor cell lines, as defined by the expression of activation markers CD69 on CD8+ T cells (determined by flow cytometry).
  • FIG. 14B shows the EC50 of T cell activation, using T cells derived from 3-4 donors, for each of the tumor cell lines.
  • T cell activation induced by bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR generally occurred at higher concentrations than that induced by bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR ( FIG. 14A ).
  • the EC50 of T cell activation for both bispecific antibodies was variable between target cell line used and between donors ( FIG. 14B ).
  • cytokine production was assessed in supernatants of the tumor cell-T cell cultures by Mesoscale Discovery U-plex multiplex ELISA.
  • IL-4, IL-6 and IL-13 were modulated at much lower levels ( ⁇ 500 pg/ml), while IL-1beta, IL-2, IL-10, IL-12p70, and TNFalpha levels were generally below 50 pg/ml.
  • IFN-gamma changes were robustly and consistently detected and IFN-gamma is one of the core cytokines elevated in serum of patients with cytokine release syndrome, the data for this cytokine is represented.
  • FIG. 15 shows the levels of IFN-gamma in the supernatant of T cell-tumor cell co-cultures at antibody concentrations that induced T cell mediated cytotoxicity in 50%, 90% and 99% of tumor cells (EC50, EC90, EC99, resp) in the presence of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR, using T cells from at least 3 donors analyzed per cell line. Cytokine production levels varied per donor and per target tumor cell line.
  • the non-clinical safety profile of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR was evaluated in non-human primates (cynomolgus monkeys, Macaca fascicularis , originating from Mauritius) at Citoxlab, France.
  • Cynomolgus monkeys were considered the only relevant species for non-clinical safety studies based on the species-specificity of the CD3 arms of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR and bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR, and furthermore due to similar binding of the B7H4 arm to human and cynomolgus B7H4 and further pharmacological findings. These studies were conducted in compliance with animal health regulations (Council Directive No. 2010/63/EU of 22 Sep. 2010 and French decret No. 2013-118 of 1 Feb. 2013 on the protection of animals used for scientific purposes).
  • the aim of the studies were to determine the potential toxicity and toxicokinetics of the CD3xB7H4 bispecific antibodies. Here only the results of the toxicokinetics and the determination of cytokine levels in plasma are described.
  • the animals were treated with a single dose of 0.1, 1, 3 or 10 mg/kg bsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR or bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR (one female animal per dose) by intravenous (IV) infusion.
  • IV intravenous
  • the day of infusion was indicated as Day 1 in the study.
  • Blood samples were obtained twice before dosing and 0.5h, 2h 4h, 12h, 24h and 48h after dosing for evaluation of the toxicokinetic profile and plasma cytokine levels, and additionally 168, 336 and 504 hours after dosing for toxicokinetics.
  • Plasma samples were analyzed for cytokine levels (IL-1 ⁇ , IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, TNF, IL-12p70, IL-15 and CCL2/MCP1) using Luminex xMAP technology.
  • BsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR administration to cynomolgus monkey produced only minor changes in plasma cytokine levels, which were considered unrelated to test compound, whereas administration of bsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR resulted in dose-dependent increase of IL-6 and MCP-1 levels, as shown in FIG. 16 .
  • the lower cytokine levels produced after treatment with bispecific BsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR, as compared with BsIgG1-huCD3-FEALxB7H4-C1-N52S-FEAR antibody, may offer an advantage in a clinical setting.
  • Plasma concentrations of CD3xB7H4 bispecifics were determined using a generic IgG PK ECLIA method. Toxicokinetic parameters were estimated using Certara Phoenix WinNonlin pharmacokinetic software version 8.1 using a non-compartmental approach consistent with the intravenous infusion injection route of administration.
  • FIG. 17 shows that the toxicokinetic profiles of both CD3xB7H4 bispecific antibodies were highly comparable up to 7 days post-dose, with both showing dose-related plasma exposure.
  • a pharmacokinetic modeling exercise was undertaken to assess whether the projected clinical dose range required by the BsIgG1-huCD3-H101G-FEALxB7H4-C1-N52S-FEAR variant with lower CD3 affinity would be unsustainably high.
  • a PK model was used that was informed by observations in cynomolgus monkey. The clinical dose range was derived that is expected to give rise to one-week average plasma exposure equal to the EC50 to EC90 for T cell mediated cell kill as observed in vitro.
  • B7H4 mRNA levels were extracted from the Omicsoft TCGA database and visualized using Oncoland software (Qiagen, USA).
  • FIG. 18 shows the B7H4 mRNA expression levels in a range of primary solid tumors, ranked according to median of the expression.
  • mRNA expression was found in a wide range of cancer indication and varied within each indication, with highest median expression found in uterine carcinosarcoma (UCS), bladder urothelial carcinoma (BLCA), pancreatic adenocarcinoma (PAAD), lung squamous cell carcinoma (LUSC), breast invasive carcinoma (BRCA), uterine corpus endometrial carcinoma (UCEC), ovarian serous cystadenocarcinoma (OV) and cholangiocarcinoma (CHOL).
  • UCS uterine carcinosarcoma
  • BLCA bladder urothelial carcinoma
  • PAAD pancreatic adenocarcinoma
  • LUSC lung squamous cell carcinoma
  • BRCA breast invasive carcinoma
  • UCEC ovarian serous cystadenocarcinoma
  • OV cholangiocarcinoma
  • IHC immunohistochemistry
  • B7H4 IHC was performed using a commercial rabbit anti-human B7-H4 monoclonal antibody (clone D1M8I, #14572, Cell Signaling Technologies) at optimal dilution (1:25; final concentration 2.6 ⁇ g/mL) for 30 min (RT) on a LabVision autostainer platform. Subsequently, sections were incubated with anti-rabbit IgG polymer (EnvisionTM FLEX+ rabbit (DAKO, S2022), washed and incubated with DAKO Liquid DAB+ Substrate chromogen system (DAKO, K3468). Hematoxylin (DAKO, S3301) was used to detect nucleated cells.
  • Cytokeratin (to determine the tumor region of interest, ROI) IHC was performed with mouse anti-cytokeratin antibody mix (clones AE1/AE3) on Ventana Benchmark using OptiView detection. Cytokeratin was visualized with DAB and nuclei counterstained with hematoxylin using default Ventana reagents. Stained TMA sections were digitized at 20 ⁇ magnification on a AxioScan (Zeiss). Initially, manual scoring was performed to determine the average B7H4 staining intensity (negative-low-medium-high) and the percentage of tumor cores with >10% B7H4-positive tumor cells.
  • the tumor ROI was defined using cytokeratin mask on TMA sections adjacent to those stained for B7H4.
  • B7H4 staining intensity in the tumor ROI was quantified (negative, weak (1), moderate (2) or string (3) and the percentage of B7H4 percentage positive tumor cells (range 0-100%) was determined using HALO image analysis software. For each indication, the percentage of tumor cores with >10% B7H4-positive tumor cells was determined.
  • Table 14 shows B7H4 protein expression determined by IHC analysis of BioMax TMAs. No to very low B7H4 expression was seen in colon, prostate, kidney, and small cell lung cancer samples. In samples from the other indications the B7H4 expression varied, with increasing B7H4 expression found in stomach cancer, pancreatic cancer, cholangiocarcinoma, oesophageal cancer, bladder cancer, non-small cell lung cancer (in particular squamous NSCLC), cervical cancer, head and neck cancer, breast cancer (triple negative breast cancer [TNBC] and non-TNBC), ovarian cancer, and uterine cancer.
  • stomach cancer pancreatic cancer
  • cholangiocarcinoma cholangiocarcinoma
  • oesophageal cancer bladder cancer
  • non-small cell lung cancer in particular squamous NSCLC
  • cervical cancer head and neck cancer
  • breast cancer triple negative breast cancer [TNBC] and non-TNBC
  • ovarian cancer and uterine cancer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US17/204,604 2020-03-18 2021-03-17 Antibodies Pending US20210292418A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/494,545 US11261254B1 (en) 2020-03-18 2021-10-05 Antibodies
US17/695,550 US20220324980A1 (en) 2020-03-18 2022-03-15 Antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20164059.6 2020-03-18
EP20164059 2020-03-18

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17/494,545 Division US11261254B1 (en) 2020-03-18 2021-10-05 Antibodies
US17/695,550 Division US20220324980A1 (en) 2020-03-18 2022-03-15 Antibodies

Publications (1)

Publication Number Publication Date
US20210292418A1 true US20210292418A1 (en) 2021-09-23

Family

ID=69846241

Family Applications (3)

Application Number Title Priority Date Filing Date
US17/204,604 Pending US20210292418A1 (en) 2020-03-18 2021-03-17 Antibodies
US17/494,545 Active US11261254B1 (en) 2020-03-18 2021-10-05 Antibodies
US17/695,550 Pending US20220324980A1 (en) 2020-03-18 2022-03-15 Antibodies

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/494,545 Active US11261254B1 (en) 2020-03-18 2021-10-05 Antibodies
US17/695,550 Pending US20220324980A1 (en) 2020-03-18 2022-03-15 Antibodies

Country Status (19)

Country Link
US (3) US20210292418A1 (de)
EP (1) EP4121456A1 (de)
JP (1) JP2023517753A (de)
KR (1) KR20220154757A (de)
CN (1) CN115298221A (de)
AR (1) AR121599A1 (de)
AU (1) AU2021236906A1 (de)
BR (1) BR112022015572A2 (de)
CA (1) CA3168613A1 (de)
CL (1) CL2022002494A1 (de)
CO (1) CO2022014603A2 (de)
CR (1) CR20220521A (de)
DO (1) DOP2022000181A (de)
EC (1) ECSP22080714A (de)
IL (1) IL296362A (de)
MX (1) MX2022009936A (de)
PE (1) PE20230444A1 (de)
TW (1) TW202200624A (de)
WO (1) WO2021185934A1 (de)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11261254B1 (en) 2020-03-18 2022-03-01 Genmab A/S Antibodies
WO2023156790A1 (en) * 2022-02-17 2023-08-24 Bivictrix Limited Novel methods of therapy
US11964024B2 (en) 2021-01-04 2024-04-23 Mersana Therapeutics, Inc. B7H4-targeted antibody-drug conjugates and methods of use thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20230162013A (ko) 2021-03-26 2023-11-28 이나뜨 파르마 에스.에이. Nk 세포 관여를 위해 사이토카인에 융합된 nkp46-결합 부위, 암 항원 결합 부위를 포함하는 다중특이적 단백질
AR125815A1 (es) * 2021-05-07 2023-08-16 Genmab As Composiciones farmacéuticas que comprenden anticuerpos biespecíficos que se unen a b7h4 y cd3
WO2022258678A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp30, a cytokine receptor, a tumour antigen and cd16a
CA3218793A1 (en) 2021-06-09 2022-12-15 Laurent Gauthier Multispecific proteins binding to nkp46, a cytokine receptor, a tumour antigen and cd16a
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, a cytokine receptor, a tumour antigen and cd16a
CN116867805A (zh) * 2021-09-27 2023-10-10 盛禾(中国)生物制药有限公司 一种异源二聚体蛋白质及其应用

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
LU91067I2 (fr) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab et ses variantes et dérivés immuno chimiques y compris les immotoxines
GB9203459D0 (en) 1992-02-19 1992-04-08 Scotgen Ltd Antibodies with germ-line variable regions
US6077835A (en) 1994-03-23 2000-06-20 Case Western Reserve University Delivery of compacted nucleic acid to cells
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
KR970029803A (ko) 1995-11-03 1997-06-26 김광호 반도체 메모리장치의 프리차지 회로
DE69830901T2 (de) 1997-05-02 2006-05-24 Genentech Inc., San Francisco ein verfahren zur herstellung multispezifischer antikörper die heteromultimere und gemeinsame komponenten besitzen
CA2361421A1 (en) 1999-02-03 2000-08-10 Biosante Pharmaceuticals, Inc. Therapeutic calcium phosphate particles and methods of manufacture and use
WO2004009618A2 (en) 2002-07-18 2004-01-29 Crucell Holland B.V. Recombinant production of mixtures of antibodies
US6281005B1 (en) 1999-05-14 2001-08-28 Copernicus Therapeutics, Inc. Automated nucleic acid compaction device
US20100081792A1 (en) 2001-06-28 2010-04-01 Smithkline Beecham Corporation Ligand
US20040018557A1 (en) 2002-03-01 2004-01-29 Immunomedics, Inc. Bispecific antibody point mutations for enhancing rate of clearance
CA2531118C (en) 2003-07-01 2013-01-08 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
US7235641B2 (en) 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
US7741568B2 (en) 2005-01-13 2010-06-22 The Wiremold Company Downward facing receptacle assembly for cable raceway
ES2592271T3 (es) 2005-03-31 2016-11-29 Chugai Seiyaku Kabushiki Kaisha Métodos de producción de polipéptidos mediante la regulación de la asociación de los polipéptidos
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US10155816B2 (en) 2005-11-28 2018-12-18 Genmab A/S Recombinant monovalent antibodies and methods for production thereof
AU2007227292B2 (en) 2006-03-17 2012-04-12 Biogen Ma Inc. Stabilized polypeptide compositions
EP1999154B1 (de) 2006-03-24 2012-10-24 Merck Patent GmbH Gentechnisch hergestellte heterodimere proteindomänen
AT503902B1 (de) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw Verfahren zur manipulation von immunglobulinen
EP2139924B1 (de) 2007-03-29 2016-07-06 Genmab A/S Bispezifische antikörper und verfahren zu deren herstellung
CN107226864A (zh) 2007-06-21 2017-10-03 宏观基因有限公司 共价双抗体及其用途
US8629246B2 (en) 2007-09-26 2014-01-14 Ucb Pharma S.A. Dual specificity antibody fusions
EP2050764A1 (de) 2007-10-15 2009-04-22 sanofi-aventis Neues polyvalentes bispezifisches Antikörperformat und Verwendung
EP2224958A2 (de) 2007-11-30 2010-09-08 Bristol-Myers Squibb Company Anti-b7h4 monoklonales antikörper-arzneimittel-konjugat und anwendungsverfahren
US8227577B2 (en) 2007-12-21 2012-07-24 Hoffman-La Roche Inc. Bivalent, bispecific antibodies
JP6157046B2 (ja) 2008-01-07 2017-07-05 アムジェン インコーポレイテッド 静電的ステアリング(electrostaticsteering)効果を用いた抗体Fcヘテロ二量体分子を作製するための方法
WO2010015792A1 (en) 2008-08-06 2010-02-11 Argenta Discovery Limited Nitrogen containing heterocyclic compounds useful as bifunctional modulators of m3 receptors and beta-2 receptors
JP5397668B2 (ja) 2008-09-02 2014-01-22 ソニー株式会社 記憶素子および記憶装置
KR20110112301A (ko) 2008-11-18 2011-10-12 메리맥 파마슈티컬즈, 인크. 인간 혈청 알부민 링커 및 그 콘쥬게이트
EP2786762B1 (de) 2008-12-19 2019-01-30 MacroGenics, Inc. Kovalente diabodies und verwendungen davon
JP2012521768A (ja) 2009-03-27 2012-09-20 ジモジェネティクス・インコーポレイテッド 抗体−受容体の組み合わせを含む多特異的結合性タンパク質を用いるための組成物および方法
EP3505636A1 (de) 2009-04-27 2019-07-03 OncoMed Pharmaceuticals, Inc. Verfahren zur herstellung von heteromultimeren molekülen
KR101224468B1 (ko) 2009-05-20 2013-01-23 주식회사 파멥신 신규한 형태의 이중표적항체 및 그 용도
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
CN102770456B (zh) 2009-12-04 2018-04-06 弗·哈夫曼-拉罗切有限公司 多特异性抗体、抗体类似物、组合物和方法
PH12018501083A1 (en) 2010-03-04 2019-02-18 Macrogenics Inc Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
TWI426920B (zh) 2010-03-26 2014-02-21 Hoffmann La Roche 雙專一性、雙價抗-vegf/抗-ang-2抗體
US9150663B2 (en) 2010-04-20 2015-10-06 Genmab A/S Heterodimeric antibody Fc-containing proteins and methods for production thereof
EP2569337A1 (de) 2010-05-14 2013-03-20 Rinat Neuroscience Corp. Heterodimere proteine sowie verfahren zu ihrer herstellung und aufreinigung
CN103154035B (zh) 2010-05-27 2017-05-10 根马布股份公司 针对her2的单克隆抗体
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
BR112013002167A2 (pt) 2010-08-24 2016-05-31 Roche Glycart Ag anticorpo biespecífico, composição farmacêutica, uso, método de tratamento de um paciente que sofre de câncer e de um paciente que sofre de inflamação
JP5758004B2 (ja) 2010-08-24 2015-08-05 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ジスルフィドによって安定化されたFv断片を含む二重特異性抗体
CA2815266C (en) 2010-11-05 2023-09-05 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
CN102250246A (zh) 2011-06-10 2011-11-23 常州亚当生物技术有限公司 抗VEGF/PDGFRβ双特异性抗体及其应用
CN118290588A (zh) 2011-10-27 2024-07-05 健玛保 异二聚体蛋白的生成
MX360110B (es) 2012-04-20 2018-10-23 Merus Nv Metodos y medios para la produccion de moleculas de tipo ig.
DK2869845T3 (da) 2012-07-06 2019-12-09 Genmab Bv Dimert protein med tredobbelte mutationer
SG10201605703TA (en) 2012-07-06 2016-09-29 Genmab Bv Dimeric protein with triple mutations
WO2014081202A1 (ko) 2012-11-21 2014-05-30 주식회사 파멥신 Vegfr-2와 dll4를 표적으로 하는 이중표적항체 및 이를 포함하는 약학적 조성물
PE20151672A1 (es) 2013-03-14 2015-11-27 Genentech Inc Anticuerpos e inmunoconjugados anti-b7-h4
JP6546162B2 (ja) 2013-07-05 2019-07-17 ゲンマブ エー/エス ヒト化またはキメラcd3抗体
EA201791554A1 (ru) 2015-01-08 2017-12-29 Генмаб А/С Биспецифичные антитела против cd3 и cd20
MX2018000347A (es) 2015-07-15 2018-03-14 Genmab As Anticuerpos de grupo de diferenciacion 3 (cd3) humanizados o quimericos.
CN109563503B (zh) * 2016-07-26 2023-09-29 静冈县 抗b7-h4抗体
EP3672990A1 (de) 2017-08-25 2020-07-01 Five Prime Therapeutics, Inc. B7-h4-antikörper und verfahren zur verwendung davon
AR121599A1 (es) 2020-03-18 2022-06-22 Genmab As Anticuerpos

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11261254B1 (en) 2020-03-18 2022-03-01 Genmab A/S Antibodies
US11964024B2 (en) 2021-01-04 2024-04-23 Mersana Therapeutics, Inc. B7H4-targeted antibody-drug conjugates and methods of use thereof
WO2023156790A1 (en) * 2022-02-17 2023-08-24 Bivictrix Limited Novel methods of therapy

Also Published As

Publication number Publication date
BR112022015572A2 (pt) 2022-09-27
WO2021185934A1 (en) 2021-09-23
AU2021236906A1 (en) 2022-08-18
EP4121456A1 (de) 2023-01-25
TW202200624A (zh) 2022-01-01
AR121599A1 (es) 2022-06-22
CL2022002494A1 (es) 2023-03-03
KR20220154757A (ko) 2022-11-22
CR20220521A (es) 2023-01-23
DOP2022000181A (es) 2022-10-16
US20220048999A1 (en) 2022-02-17
US11261254B1 (en) 2022-03-01
CA3168613A1 (en) 2021-09-23
JP2023517753A (ja) 2023-04-26
ECSP22080714A (es) 2022-11-30
IL296362A (en) 2022-11-01
PE20230444A1 (es) 2023-03-08
CO2022014603A2 (es) 2023-02-16
US20220324980A1 (en) 2022-10-13
MX2022009936A (es) 2022-09-12
CN115298221A (zh) 2022-11-04

Similar Documents

Publication Publication Date Title
US11261254B1 (en) Antibodies
EP3592769B1 (de) Antikörper gegen pd-l1
US11130819B2 (en) Antibodies
US20240076397A1 (en) Bispecific anti-cd37 antibodies, monoclonal anti-cd37 antibodies and methods of use thereof
AU2022268652A9 (en) PHARMACEUTICAL COMPOSITIONS COMPRISING BISPECIFIC ANTIBODIES BINDING TO B7H4 and CD3
US20230399414A1 (en) Bispecific anti-cd37 antibodies, monoclonal anti-cd37 antibodies and methods of use thereof
US20230365714A1 (en) Antibodies capable of binding to ror2 and bispecific antibodies binding to ror2 and cd3

Legal Events

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

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: GENMAB A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KOOPMAN, LOUISE;ENGELBERTS, PATRICK;VERZIJL, DENNIS;AND OTHERS;SIGNING DATES FROM 20210422 TO 20210809;REEL/FRAME:058018/0009

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

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

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

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED