WO2024126457A1 - Polythérapie impliquant des agents de liaison bispécifiques se liant à cldn18.2 et cd3 et des inhibiteurs de point de contrôle immunitaire - Google Patents

Polythérapie impliquant des agents de liaison bispécifiques se liant à cldn18.2 et cd3 et des inhibiteurs de point de contrôle immunitaire Download PDF

Info

Publication number
WO2024126457A1
WO2024126457A1 PCT/EP2023/085290 EP2023085290W WO2024126457A1 WO 2024126457 A1 WO2024126457 A1 WO 2024126457A1 EP 2023085290 W EP2023085290 W EP 2023085290W WO 2024126457 A1 WO2024126457 A1 WO 2024126457A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
acid sequence
seq
polypeptide chain
composition
Prior art date
Application number
PCT/EP2023/085290
Other languages
English (en)
Inventor
Aya Kikuchi
Taisuke NAKAZAWA
Matthew Bernett
Alex Nisthal
Gregory Moore
Original Assignee
Astellas Pharma Europe Bv
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 Astellas Pharma Europe Bv filed Critical Astellas Pharma Europe Bv
Publication of WO2024126457A1 publication Critical patent/WO2024126457A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • 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
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [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
    • C07K16/2818Immunoglobulins [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 CD28 or CD152
    • 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
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • Cancer is the second leading cause of death globally and is expected to be responsible for an estimated 9.6 million deaths in 2018 (Bray, F. et al., CA: A Cancer Journal for Clinicians, 68: 394- 424, 2018).
  • Bray, F. et al., CA: A Cancer Journal for Clinicians, 68: 394- 424, 2018 In general, once a solid tumor has metastasized, with a few exceptions such as germ cell and some carcinoid tumors, 5-year survival rarely exceeds 25%. The poor prognosis of certain cancers highlights the need for additional treatment approaches.
  • CLDN18 The tight junction molecule claudin 18 (CLDN18) is an integral transmembrane protein (tetraspanin) having four membrane spanning hydrophobic regions and two extracellular loops (loopl embraced by hydrophobic region 1 and hydrophobic region 2; loop2 embraced by hydrophobic regions 3 and 4). CLDN 18 exists in two different splice variants, which are described in mouse and in human (Niimi, Mol. Cell. Biol. 21:7380-90, 2001).
  • the splice variants (Genbank accession number: splice variant 1 (CLDN18.1): NP 057453, NM 016369, and splice variant 2 (CLDN18.2): NM_001002026, NP_001002026) have a molecular weight of approximately 27.9 / 27.72 kD.
  • the splice variants CLDN18.1 and CLDN18.2 differ in the N-terminal portion which comprises the first transmembrane (TM) region and loopl, whereas the primary protein sequence of the C-terminus is identical.
  • CLDN 18.2 In normal tissues, there is no detectable expression of CLDN 18.2 with exception of stomach where CLDN18.2 is expressed exclusively on short-lived differentiated gastric epithelial cells. CLDN18.2 is maintained in the course of malignant transformation and thus frequently displayed on the surface of human gastric cancer cells. Moreover, this pan-tumoral antigen is ectopically activated at significant levels in esophageal, pancreatic and lung adenocarcinomas. The CLDN 18.2 protein is also localized in lymph node metastases of gastric cancer adenocarcinomas and in distant metastases especially into the ovary (so-called Krukenberg tumors).
  • claudins such as CLDN 18.2 between cancer and normal cells, their membrane localization and their absence from the vast majority of toxicity relevant normal tissues makes these molecules attractive targets for cancer immunotherapy and the use of antibody-based therapeutics for targeting CLDN 18.2 in cancer therapy promises a high level of therapeutic specificity.
  • the chimeric IgGl antibody IMAB362 (Zolbetuximab (previously named Claudiximab)) which is directed against CLDN18.2 has been developed by Ganymed Pharmaceuticals AG.
  • IMAB362 recognizes the first extracellular domain (ECD1) of CLDN18.2 with high affinity and specificity.
  • IMAB362 does not bind to any other claudin family member including the closely related splice variant 1 of Claudin 18 (CLDN18.1).
  • IMAB362 shows precise tumor cell specificity and bundles two independent highly potent mechanisms of action.
  • IMAB362 Upon target binding IMAB362 mediates cell killing mainly by ADCC and CDC.
  • IMAB362 lyses efficiently CLDN18.2-positive cells, including human gastric cancer cell lines in vitro and in vivo.
  • Anti -tumor efficacy of IMAB362 was demonstrated in mice carrying xenografted tumors inoculated with CLDN18.2- positive cancer cell lines.
  • IgGl antibodies typically engage the cellular immune system via interaction of the Fc domain with Fey receptors (FcyRs) expressed on various immune cells including natural killer cells which are the main agents of ADCC.
  • FcyRs Fey receptors
  • IgGl monoclonal antibodies (mAbs) triggering ADCC face several limitations including broad distribution of low affinity Fc receptor variants in the population (up to 80%) as well as in vivo IgGl modifications reducing the mAb efficacy (Chames et al., (2009) Br J Pharmacol, 157(2):220-233).
  • Therapeutic antibodies also have to compete with the patients IgGs resulting in high doses of mAbs necessary in vivo.
  • therapeutic antibodies may interact with FcyRIIb (an inhibitory FcyR expressed by B cells, macrophages, dendritic cells and neutrophils) resulting in negative signaling that decreases their efficacy.
  • FcyRIIb an inhibitory FcyR expressed by B
  • the present invention generally embraces the treatment of a subject comprising the administration of a bispecific binding agent described herein that comprises two CLDN18.2 binding domains in the Fab format that are specific for cancer cells, and a CD3 binding domain in the scFv format that is specific for the T cell-specific antigen CD3 allowing to bind to T cells and to pull the T cells into the complex, thus making it possible to target the cytotoxic effect of the T cells to the cancer cells. Formation of this complex can induce signalling in cytotoxic T cells, either on its own or in combination with accessory cells, which leads to the release of cytotoxic mediators.
  • the treatment using a bispecific binding agent described herein is combined with an additional treatment comprising administration of one or more immune checkpoint inhibitors. It is demonstrated herein that a treatment involving a bispecific binding agent described herein and an immune checkpoint inhibitor can induce a potent antitumor effect.
  • the invention generally provides a combination therapy involving a bispecific binding agent that binds to CLDN18.2 and CD3 and an immune checkpoint inhibitor.
  • the bispecific binding agent is a bispecific tetrameric binding agent.
  • the immune checkpoint inhibitor is a PD-1 antibody.
  • composition or medical preparation comprising:
  • a bispecific binding agent comprising a first binding domain that binds to human CLDN18.2, a second binding domain that binds to human CLDN18.2, and a third binding domain that binds to human CD3, wherein the binding agent comprises four polypeptide chains, wherein
  • the first polypeptide chain comprises the amino acid sequence of SEQ ID NO: 27 or an amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 27;
  • the second polypeptide chain comprises the amino acid sequence of SEQ ID NO: 28 or an amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 28;
  • the third polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29 or an amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 29;
  • the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29 or an amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 29;
  • the first polypeptide chain consists of the amino acid sequence of SEQ ID NO: 27 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 27.
  • the second polypeptide chain consists of the amino acid sequence of SEQ ID NO: 28 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 28.
  • the third polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 29.
  • the fourth polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 29.
  • the first polypeptide chain consists of the amino acid sequence of SEQ ID NO: 27 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 27;
  • the second polypeptide chain consists of the amino acid sequence of SEQ ID NO: 28 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 28;
  • the third polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 29;
  • the fourth polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29 or the amino acid sequence which has at least 80% sequence identity to the amino acid sequence of SEQ ID NO: 29.
  • composition or medical preparation comprising:
  • a bispecific binding agent comprising a first binding domain that binds to human CLDN18.2, a second binding domain that binds to human CLDN18.2, and a third binding domain that binds to human CD3, wherein the binding agent comprises four polypeptide chains, wherein
  • the first polypeptide chain comprises the amino acid sequence of SEQ ID NO: 27;
  • the second polypeptide chain comprises the amino acid sequence of SEQ ID NO: 28;
  • the third polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29;
  • the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29;
  • the first polypeptide chain consists of the amino acid sequence of SEQ ID NO: 27.
  • the second polypeptide chain consists of the amino acid sequence of SEQ ID NO: 28.
  • the third polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29.
  • the fourth polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29.
  • the first polypeptide chain consists of the amino acid sequence of SEQ ID NO: 27;
  • the second polypeptide chain consists of the amino acid sequence of SEQ ID NO: 28;
  • the third polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29;
  • the fourth polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29.
  • composition or medical preparation comprising:
  • a bispecific binding agent comprising a first binding domain that binds to human CLDN18.2, a second binding domain that binds to human CLDN18.2, and a third binding domain that binds to human CD3, wherein the binding agent is encoded by one or more nucleic acid molecules comprising:
  • the one or more nucleic acid molecules is a set of nucleic acids.
  • the bispecific binding agent comprises four polypeptide chains, wherein (i) the first polypeptide chain is encoded by the first nucleic acid sequence;
  • the second polypeptide chain is encoded by the second nucleic acid sequence
  • the third polypeptide chain is encoded by the third nucleic acid sequence
  • the fourth polypeptide chain is encoded by the third nucleic acid sequence.
  • the first polypeptide chain comprises the amino acid sequence of SEQ ID NO: 27 or a C-terminal truncation variant thereof, wherein the C-terminal truncation variant of SEQ ID NO: 27 comprises a deletion of lysine at position 447 of SEQ ID NO: 27.
  • the second polypeptide chain comprises the amino acid sequence of SEQ ID NO: 28 or a C-terminal truncation variant thereof, wherein the C-terminal truncation variant of SEQ ID NO: 28 comprises a deletion of lysine at position 720 of SEQ ID NO: 28.
  • the third polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29.
  • the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29.
  • the first polypeptide chain comprises the amino acid sequence of SEQ ID NO: 27 or a C- terminal truncation variant thereof, wherein the C-terminal truncation variant of SEQ ID NO: 27 comprises a deletion of lysine at position 447 of SEQ ID NO: 27;
  • the second polypeptide chain comprises the amino acid sequence of SEQ ID NO: 28 or a C- terminal truncation variant thereof, wherein the C-terminal truncation variant of SEQ ID NO: 28 comprises a deletion of lysine at position 720 of SEQ ID NO: 28;
  • the third polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29;
  • the fourth polypeptide chain comprises the amino acid sequence of SEQ ID NO: 29.
  • the first polypeptide chain consists of the amino acid sequence of SEQ ID NO: 27 or a C- terminal truncation variant thereof, wherein the C-terminal truncation variant of SEQ ID NO: 27 comprises a deletion of lysine at position 447 of SEQ ID NO: 27;
  • the second polypeptide chain consists of the amino acid sequence of SEQ ID NO: 28 or a C- terminal truncation variant thereof, wherein the C-terminal truncation variant of SEQ ID NO: 28 comprises a deletion of lysine at position 720 of SEQ ID NO: 28;
  • the third polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29;
  • the fourth polypeptide chain consists of the amino acid sequence of SEQ ID NO: 29.
  • the first polypeptide chain interacts with the second polypeptide chain and with the third polypeptide chain.
  • the second polypeptide chain interacts with the fourth polypeptide chain.
  • the first polypeptide chain interacts with the second polypeptide chain and with the third polypeptide chain and the second polypeptide chain interacts with the fourth polypeptide chain.
  • the first polypeptide chain comprises, from N-terminus to C-terminus,
  • VH variable region of a heavy chain derived from an immunoglobulin that binds to human CLDN18.2 (VH(CLDN18.2)),
  • the second polypeptide chain comprises, from N-terminus to C-terminus,
  • VH variable region of a heavy chain derived from an immunoglobulin that binds to human CLDN18.2 (VH(CLDN18.2)),
  • VL variable region of a light chain (VL) derived from an immunoglobulin that binds to human CD3 (VL(CD3)),
  • VH variable region of a heavy chain derived from an immunoglobulin that binds to human CD3 (VH(CD3)),
  • VH(CD3) variable region 2 of a heavy chain (CH2) derived from an immunoglobulin or a functional variant thereof, and
  • the third polypeptide chain comprises, from N-terminus to C-terminus,
  • VL variable region of a light chain (VL) derived from an immunoglobulin that binds to human CLDN18.2 (VL(CLDN18.2)), and
  • the fourth polypeptide chain comprises, from N-terminus to C-terminus,
  • VL variable region of a light chain (VL) derived from an immunoglobulin that binds to human CLDN18.2 (VL(CLDN18.2)), and
  • the VH(CLDN18.2) on the first polypeptide chain and the VL(CLDN18.2) on the third polypeptide chain interact to form a binding domain that binds to human CLDN18.2.
  • the VH(CLDN18.2) on the second polypeptide chain and the VL(CLDN18.2) on the fourth polypeptide chain interact to form a binding domain that binds to human CLDN18.2.
  • the VH(CD3) and the VL(CD3) interact to form a binding domain that binds to human CD3.
  • the VH(CLDN18.2) comprises a CDR1 comprising the amino acid sequence SYWIN (SEQ ID NO: 10), a CDR2 comprising the amino acid sequence NIYPSDSYTNYNQKFQG (SEQ ID NO: 11), and a CDR3 comprising the amino acid sequence SWRGNSFDY (SEQ ID NO: 12).
  • the VL(CLDN18.2) comprises a CDR1 comprising the amino acid sequence KSSQSLLNSGNQKNYLT (SEQ ID NO: 13), a CDR2 comprising the amino acid sequence WASTRES (SEQ ID NO: 14), and a CDR3 comprising the amino acid sequence QNDYSYPFT (SEQ ID NO: 15).
  • the VH(CD3) comprises a CDR1 comprising the amino acid sequence TYAMN (SEQ ID NO: 18), a CDR2 comprising the amino acid sequence RIRSKANNYATYYADSVKG (SEQ ID NO: 23), and a CDR3 comprising the amino acid sequence HGNFGDSYVSWFAY (SEQ ID NO: 19).
  • the VL(CD3) comprises a CDR1 comprising the amino acid sequence GSSTGAVTTSNYAN (SEQ ID NO: 20), a CDR2 comprising the amino acid sequence GTNKRAP (SEQ ID NO: 21), and a CDR3 comprising the amino acid sequence ALWYSNHWV (SEQ ID NO: 22).
  • the CH2 on the first polypeptide chain interacts with the CH2 on the second polypeptide chain and/or the CH3 on the first polypeptide chain interacts with the CH3 on the second polypeptide chain.
  • the CHI on the first polypeptide chain interacts with the CL on the third polypeptide chain.
  • the CHI on the second polypeptide chain interacts with the CL on the fourth polypeptide chain.
  • the immunoglobulin is IgGl. In some embodiments, the IgGl is human IgGl.
  • the VH(CLDN18.2) comprises or consists of the amino acid sequence represented by SEQ ID NO: 16.
  • the VL(CLDN18.2) comprises or consists of the amino acid sequence represented by SEQ ID NO: 17.
  • the VL(CD3) comprises or consists of the amino acid sequence represented by SEQ ID NO: 24. In some embodiments, the VH(CD3) comprises or consists of the amino acid sequence represented by SEQ ID NO: 25.
  • the VH(CLDN 18.2) comprises or consists of the amino acid sequence represented by SEQ ID NO:
  • VL(CLDN 18.2) comprises or consists of the amino acid sequence represented by SEQ ID NO:
  • the VH(CD3) comprises or consists of the amino acid sequence represented by SEQ ID NO: 25
  • the VL(CD3) comprises or consists of the amino acid sequence represented by SEQ ID NO: 24.
  • the VH(CLDN18.2), VL(CLDN18.2), VH(CD3) and/or VL(CD3) are humanized.
  • the CHI is connected to the CH2 by a peptide linker LI.
  • the peptide linker LI comprises the amino acid sequence EPKSCDKTHTCPPCP (SEQ ID NO: 6) or a functional variant thereof.
  • the VL(CD3) is connected to the CHI by a peptide linker L2.
  • the peptide linker L2 comprises the amino acid sequence (G4S) X or a functional variant thereof, wherein x is 2, 3, 4, 5 or 6.
  • the peptide linker L2 comprises the amino acid sequence (648)2 (SEQ ID NO: 5) or a functional variant thereof.
  • the VL(CD3) and the VH(CD3) are connected to one another by a peptide linker L3.
  • the peptide linker L3 comprises the amino acid sequence (GKPGS)x or a functional variant thereof, wherein x is 2, 3, 4, 5 or 6.
  • the peptide linker L3 comprises the amino acid sequence (GKPGS)4 (SEQ ID NO: 2) or a functional variant thereof.
  • the VH(CD3) is connected to the CH2 by a peptide linker L4.
  • the peptide linker L4 comprises the amino acid sequence (G4SX or a functional variant thereof, wherein x is 2, 3, 4, 5 or 6.
  • the peptide linker L4 comprises the amino acid sequence (648)2 (SEQ ID NO: 5) or a functional variant thereof.
  • the CHI, CH2 and/or CH3 domains of the bispecific binding agent described herein comprise one or more amino acid modifications, in particular substitutions and/or deletions, in positions corresponding to positions of human IgGl according to EH numbering.
  • the CHI on the first and/or the second polypeptide chain comprises an amino acid sequence comprising an aspartic acid residue at position 208 according to EU numbering.
  • the CHI on the first polypeptide chain comprises an amino acid sequence comprising an aspartic acid residue at position 208 according to EU numbering and the CHI on the second polypeptide chain comprises an amino acid sequence comprising an asparagine residue at position 208 according to EU numbering.
  • the binding agent described herein does not substantially, e.g., not detectably, bind to human FcyRI, Ila, lib, and/or Illa.
  • the CH2 on the first and/or the second polypeptide chain comprises an amino acid sequence comprising one or more of the following: a proline residue at position 233, a valine residue at position 234, an alanine residue at position 235, a deletion at position 236, a lysine residue at position 267 and a glutamic acid residue at position 295 according to EU numbering.
  • the CH2s on the first and the second polypeptide chain comprise an amino acid sequence comprising a proline residue at position 233, a valine residue at position 234, an alanine residue at position 235, a deletion at position 236 and a lysine residue at position 267 according to EU numbering and wherein the CH2 on the first polypeptide chain further comprises a glutamic acid residue at position 295 according to EU numbering and the CH2 on the second polypeptide chain further comprises a glutamine residue at position 295 according to EU numbering.
  • the CH3 on the first and/or the second polypeptide chain comprises an amino acid sequence comprising one or more of the following: a glutamine residue at position 357, a lysine residue at position 364, an aspartic acid residue at position 368, a serine residue at position 370, an aspartic acid residue at position 384, a glutamic acid residue at position 418 and an aspartic acid residue at position 421 according to EU numbering.
  • the CH3 on the first polypeptide chain comprises an amino acid sequence comprising a glutamic acid residue at position 357, a serine residue at position 364, an aspartic acid residue at position 368, a serine residue at position 370, an aspartic acid residue at position 384, a glutamic acid residue at position 418 and an aspartic acid residue at position 421 according to EU numbering and the CH3 on the second polypeptide chain comprises an amino acid sequence comprising a glutamine residue at position 357, a lysine residue at position 364, a leucine residue at position 368, a lysine residue at position 370, an asparagine residue at position 384, a glutamine residue at position 418 and an asparagine residue at position 421 according to EU numbering.
  • the first polypeptide chain comprises the amino acid sequence represented by SEQ ID NO: 7.
  • the second polypeptide chain comprises the amino acid sequence represented by SEQ ID NO: 8.
  • the third and/or fourth polypeptide chain comprises the amino acid sequence represented by SEQ ID NO: 9.
  • CD3 is expressed on the surface of T cells.
  • the bispecific binding agent described herein binds to the epsilon chain of CD3.
  • binding of the binding agent to CD3 on T cells results in proliferation and/or activation of the T cells.
  • proliferation and/or activation of T cells includes proliferation and/or activation of CD4 and/or CD8 T cells, preferably CD107a+ T cells.
  • said proliferated and/or activated T cells are capable of degranulation.
  • said activated T cells release cytotoxic factors, e.g., perforins and granzymes, and initiate cytolysis and/or apoptosis of cancer cells.
  • CLDN18.2 is expressed in cancer cells. In some embodiments, CLDN18.2 is expressed on the surface of cancer cells. In some embodiments, the bispecific binding agent binds to an extracellular portion of CLDN18.2. In some embodiments, the binding agent induces T cell-mediated cytotoxicity against cancer cells expressing CLDN18.2.
  • the cancer cells are from a cancer selected from the group consisting of gastric cancer, particularly gastric adenocarcinoma, esophageal cancer, cancer of the gastroesophageal junction (GEJ), particularly GEJ adenocarcinoma, pancreatic cancer, particularly pancreatic adenocarcinoma, lung cancer such as non-small cell lung cancer (NSCLC), breast cancer, ovarian cancer, colon cancer, rectal cancer, colorectal cancer, hepatic cancer, head- neck cancer, bile duct cancer, cancer of the gallbladder and the metastasis thereof, a Krukenberg tumor, peritoneal metastasis and/or lymph node metastasis.
  • gastric cancer particularly gastric adenocarcinoma, esophageal cancer, cancer of the gastroesophageal junction (GEJ), particularly GEJ adenocarcinoma
  • pancreatic cancer particularly pancreatic adenocarcinoma
  • lung cancer such as
  • the bispecific binding agent comprises one or more posttranslational modifications.
  • the bispecific binding agent is derived from one or more posttranslational modifications of a binding agent described herein.
  • the one or more posttranslational modifications are selected from pyroglutamylation at the N-terminus of one or more VH(CLDN18.2), deletion of lysine at the C-terminus of the first polypeptide chain and deletion of lysine at the C-terminus of the second polypeptide chain.
  • a bispecific binding agent is provided as nucleic acid encoding the binding agent. In some embodiments, a bispecific binding agent is provided as a set of nucleic acids together encoding the binding agent. In some embodiments, the nucleic acid or the set of nucleic acids is capable of expressing the bispecific binding agent.
  • a vector comprises the nucleic acid or the set of nucleic acids. In some embodiments, a set of vectors comprises the set of nucleic acids. In some embodiments, each nucleic acid of the set of nucleic acids is contained in a vector of the set of vectors. In some embodiments, the vector or the set of vectors is capable of expressing the bispecific binding agent.
  • nucleic acid or set of nucleic acids may provide the bispecific binding agent.
  • a nucleic acid is operably linked to any number of regulatory elements (promoters, origin of replication, selectable markers, ribosomal binding sites, inducers, etc.).
  • the vector can be an expression vector and can be extra-chromosomal or integrating vector.
  • nucleic acids encoding a polypeptide chain of the binding agent described herein are each contained within a single expression vector.
  • the nucleic acids can be under control of different or the same promoters. In such embodiments, different vector ratios can be used to drive formation of the binding agent described herein.
  • the immune checkpoint inhibitor comprises one or more immune checkpoint inhibitors.
  • the immune checkpoint inhibitor comprises an antibody selected from an anti-PD-1 antibody, an anti-PD-Ll antibody and a combination thereof.
  • the immune checkpoint inhibitor comprises an anti-PD-1 antibody.
  • the immune checkpoint inhibitor comprises cemiplimab (LIBTAYO, REGN2810), nivolumab (OPDIVO; BMS-936558), pembrolizumab (KEYTRUDA; MK-3475), pidilizumab (CT-011), spartalizumab (PDR001), MEDI0680 (AMP-514), dostarlimab (TSR-042), cetrelimab (JNJ 63723283), toripalimab (JSOO1), AMP-224 (GSK-2661380), PF-06801591, tislelizumab (BGB-A317), ABBV-181, BI 754091, sintilimab (IBI308) or camrelizumab (SHR- 1210).
  • cemiplimab LIBTAYO, REGN2810
  • OPDIVO pembrolizumab
  • KEYTRUDA pembrolizumab
  • the immune checkpoint inhibitor comprises an anti-PD-Ll antibody.
  • the immune checkpoint inhibitor comprises atezolizumab (TECENTRIQ; RG7446; MPDL3280A; R05541267), durvalumab (MEDI4736), BMS-936559, avelumab (bavencio), lodapolimab (LY3300054), CX-072 (Proclaim-CX-072), FAZ053, KN035, sugemalimab (CS1001) or MDX-1105.
  • atezolizumab TECENTRIQ; RG7446; MPDL3280A; R05541267
  • durvalumab MEDI4736
  • BMS-936559 avelumab (bavencio)
  • lodapolimab LY3300054
  • CX-072 Proclaim-CX-072
  • FAZ053, KN035, sugemalimab (CS1001) or MDX-1105 sugemalimab
  • the composition or medical preparation is a pharmaceutical composition.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers, diluents and/or excipients.
  • composition or medical preparation is a kit.
  • the bispecific binding agent and the immune checkpoint inhibitor are in separate vials.
  • composition or medical preparation further comprises instructions for use of the bispecific binding agent and the immune checkpoint inhibitor for treating or preventing cancer.
  • the invention further provides the composition or medical preparation described herein for pharmaceutical use.
  • the pharmaceutical use comprises a therapeutic or prophylactic treatment of a disease or disorder.
  • the therapeutic or prophylactic treatment of a disease or disorder comprises treating or preventing cancer.
  • the cancer involves cancer cells expressing CLDN18.2.
  • the cancer is selected from the group consisting of gastric cancer, particularly gastric adenocarcinoma, esophageal cancer, cancer of the gastroesophageal junction (GEJ), particularly GEJ adenocarcinoma, pancreatic cancer, particularly pancreatic adenocarcinoma lung cancer such as non small cell lung cancer (NSCLC), breast cancer, ovarian cancer, colon cancer, rectal cancer, colorectal cancer, hepatic cancer, head-neck cancer, bile duct cancer, cancer of the gallbladder and the metastasis thereof, a Krukenberg tumor, peritoneal metastasis and/or lymph node metastasis.
  • gastric cancer particularly gastric adenocarcinoma, esophageal cancer, cancer of the gastroesophageal junction (GEJ), particularly GEJ adenocarcinoma
  • pancreatic cancer particularly pancreatic adenocarcinoma lung cancer such as non small cell lung cancer (NSCLC
  • composition or medical preparation described herein is for administration to a human.
  • the invention further provides a method of treating or preventing cancer in a subject comprising administering to the subject:
  • the bispecific binding agent is a bispecific binding agent as described in the context of the composition or medical preparation described herein.
  • the immune checkpoint inhibitor is an immune checkpoint inhibitor as described herein, e.g., as described in the context of the composition or medical preparation described herein.
  • the subject is a human.
  • CLDN18.2 preferably has the amino acid sequence according to SEQ ID NO: 1.
  • the first polypeptide chain does not comprise a VL(CLDN18.2). In some embodiments, the second polypeptide chain does not comprise a VL(CLDN18.2). In some embodiments, the third polypeptide chain does not comprise a VH(CLDN18.2). In some embodiments, the fourth polypeptide chain does not comprise a VH(CLDN18.2).
  • the CLDN18.2 binding domains of the bispecific binding agent are in the format of a Fab fragment.
  • the CD3 binding domain of the bispecific binding agent is in the format of an scFv moiety.
  • the bispecific binding agent does not bind to CLDN18.1. In some embodiments, the bispecific binding agent does not bind to CLDN18.1 of human, mouse or cynomolgus. In some embodiments, the bispecific binding agent does not bind to CLDN9, such as human CLDN9.
  • the bispecific binding agent binds to CLDN18.2 of more than one species such as CLDN18.2 of human, mouse and cynomolgus.
  • treating a subject such as a patient with the bispecific binding agent and an immune checkpoint inhibitor as described herein results in prolonged survival of said subject.
  • treating a patient with the bispecific binding agent and an immune checkpoint inhibitor as described herein reduces, preferably significantly reduces, growth and/or volume of a tumor of a subject such as a patient.
  • the bispecific binding agent described herein is able to redirect T cells to attack cancer cells and, thus, acts through redirected T cell cytotoxicity (RTCC).
  • RTCC redirected T cell cytotoxicity
  • the bispecific binding agent is not capable, or not substantially capable, of inducing ADCC.
  • the binding agent is not capable, or not substantially capable, of inducing CDC.
  • the bispecific binding agent is produced by a method comprising transfecting host cells with a nucleic acid, a set of nucleic acids, a vector or a set of vectors encoding the polypeptide chains of the bispecific binding agent.
  • the host cells express the nucleic acid, the set of nucleic acids, the vector or the set of vectors.
  • a host cell co-expresses a nucleic acid encoding the first polypeptide chain of the bispecific binding agent, a nucleic acid encoding the second polypeptide chain of the bispecific binding agent, a nucleic acid encoding the third polypeptide chain of the bispecific binding agent and a nucleic acid encoding the fourth polypeptide chain of the bispecific binding agent.
  • said nucleic acids are contained in a vector or in a set of vectors.
  • a host cell expresses all polypeptide chains of the bispecific binding agent.
  • the host cell after transfection produces the bispecific binding agent, preferably when grown under appropriate conditions for binding agent production such as those described herein or known in the art.
  • the bispecific binding agent can be obtained from the host cell.
  • a method of producing the bispecific binding agent comprises the steps of transfecting a host cell with a nucleic acid encoding the first polypeptide chain of the bispecific binding agent, a nucleic acid encoding the second polypeptide chain of the bispecific binding agent, a nucleic acid encoding the third polypeptide chain of the bispecific binding agent, and a nucleic acid encoding the fourth polypeptide chain of the bispecific binding agent, expressing said nucleic acids in the host cell and obtaining the bispecific binding agent.
  • the host cell is a mammalian cell, preferably selected from the group consisting of CHO cells, BHK cells, HeLa cells, COS cells, HEK293 cells, HEK293 T cells and the like.
  • the host cell is a bacterial cell, a yeast cell, a fungal cell, a plant cell or an insect cell.
  • the binding agent is produced in vitro.
  • the binding agent is produced in vivo, e.g., in a subject to be treated such as a subject having a disease, in particular a disease associated with cells expressing CLDN18.2, e.g. cancer.
  • the different polypeptide chains of the bispecific binding agent are produced in two or more different host cells. In some embodiments, all polypeptide chains of the bispecific binding agent are produced in the same host cell.
  • the polypeptide chains of the bi specific binding agent are linked to one another, e.g., covalently linked.
  • the polypeptide chains of the bispecific binding agent are produced as one polypeptide comprising all polypeptide chains of the bispecific binding agent.
  • at least two polypeptide chains of the bispecific binding agent are linked together and are produced as one polypeptide.
  • the polypeptide chains of the bispecific binding agent described herein are produced separately, i.e., as separate polypeptides, e.g., in the same or in different cells and interact upon or after production to form the bispecific binding agent, e.g., within a cell or outside of a cell.
  • the polypeptide chains are produced as separate polypeptides, i.e., the first polypeptide chain is produced as one polypeptide, the second polypeptide chain is produced as one polypeptide, the third polypeptide chain is produced as one polypeptide, and the fourth polypeptide chain is produced as one polypeptide, and the polypeptide chains interact to form the bispecific binding agent.
  • Figure 1 depicts the “Fab2-scFv” format of the bispecific binding agent described herein which comprises a VH recombinantly fused to one side of a heterodimeric Fc (the first polypeptide chain described herein), a VH recombinantly fused to an scFv fused to the other side of the heterodimeric Fc (the second polypeptide chain described herein), and a LC (third and fourth polypeptide chains described herein) forming Fab domains with the VH of the first polypeptide chain and the additional VH of the second polypeptide chain.
  • Figure 2 Anti-tumor Effect of ASP2138 Monotherapy in hCD3a KI Mouse Model Bearing the hCLDN18.2 - expressing MC38 Tumor Cells
  • MC38_hCLDN18.2 cells were subcutaneously inoculated into the flank of mice at 2.0 * 10 5 cells on day -5.
  • hCD3s KI mice received intraperitoneal administration of PBS or ASP2138 on days 0 and 7.
  • Upper: the tumor volumes in each group were plotted at each time point as the mean ⁇ SEM (n 8).
  • Lower: scatter plots indicate the individual tumor volume on day 13 and short horizontal lines and error bars represent the mean ⁇ SEM (n 8).
  • Statistical analysis was performed on the values on day 13. **: P ⁇ 0.01 compared with the value of the PBS control group (Dunnett’s multiple comparison test).
  • CLDN18.2 claudin 18.2;
  • hCD3s KI human CD3e knock-in;
  • MC38_hCLDN18.2 human CLDN 18.2 -expressing MC38
  • Figure 3 Combination Effect of ASP2138 and Anti-mPD-1 Antibody in hCD3e KI Mouse Model Bearing the hCLDN18.2 - expressing MC38 Tumor Cells
  • MC38_hCLDN18.2 cells were subcutaneously inoculated into the flank of mice at 2.0 x 10 5 cells on day -6.
  • hCD3e KI mice received intraperitoneal administration of PBS or ASP2138 (0.1 mg/kg) on days 0 and 7, and anti-mPD-1 antibody (anti-mPD-1 Ab and combination groups) or isotype control antibody (control and ASP2138 groups) at a dose of 100 pg/head on days 0, 4, 7 and 11.
  • Upper: the tumor volumes in each group were plotted at each time point as the mean ⁇ SEM (n 15).
  • Lower: scatter plots indicate the individual tumor volume on day 14 and short horizontal lines and error bars represent the mean ⁇ SEM (n 15).
  • Statistical analysis was performed on the values on day 14.
  • CLDN18.2 claudin 18.2; hCD3e KI: human CD3s knock-in; MC38_hCLDN18.2: human CLDN18.2-expressing MC38; anti-mPD-1 antibody: anti-mouse programmed death- 1 antibody.
  • Bispecific binding agent The first target molecule of the bispecific binding agents described herein is CLDN18.2.
  • Cell surface proteins of the claudin family are expressed in tumors of various origins, and are particularly suited as target structures in connection with antibody-mediated cancer immunotherapy due to their selective expression (no expression in a toxicity relevant normal tissue) and localization to the plasma membrane.
  • CLDN18.2 has been identified as differentially expressed in tumor tissues, with the only normal tissues expressing CLDN 18.2 being stomach. CLDN18.2 is selectively expressed in normal tissues in differentiated epithelial cells of the gastric mucosa. CLDN 18.2 is expressed in cancers of various origins such as pancreatic carcinoma, esophageal carcinoma, gastric carcinoma, bronchial carcinoma, breast carcinoma, and ENT tumors.
  • CLDN18.2 is a valuable target for the prevention and/or treatment of primary tumors, such as gastric cancer, particularly gastric adenocarcinoma, esophageal cancer, cancer of the gastroesophageal junction (GEJ), particularly GEJ adenocarcinoma, pancreatic cancer, particularly pancreatic adenocarcinoma, lung cancer such as non-small cell lung cancer (NSCLC), ovarian cancer, colon cancer, rectal cancer, colorectal cancer, hepatic cancer, head-neck cancer, bile duct cancer, cancers of the gallbladder, and metastases thereof, in particular gastric cancer metastasis such as Krukenberg tumors, peritoneal metastasis, and lymph node metastasis.
  • gastric cancer metastasis such as Krukenberg tumors, peritoneal metastasis, and lymph node metastasis.
  • claudin 18 or "CLDN18” relates to claudin 18 and includes any variants, including claudin 18 splice variant 1 (claudin 18.1 (CLDN 18.1)) and claudin 18 splice variant 2 (claudin 18.2 (CLDN 18.2)).
  • CLDN 18.2 preferably relates to human CLDN 18.2, and, in particular, to a protein comprising, preferably consisting of the amino acid sequence according to SEQ ID NO: 1 of the sequence listing or a variant of said amino acid sequence.
  • the first extracellular loop of CLDN18.2 preferably comprises amino acids 27 to 81, more preferably amino acids 29 to 78 of the amino acid sequence shown in SEQ ID NO: 1 .
  • the second extracellular loop of CLDN 18.2 preferably comprises amino acids 140 to 180 or 144 to 167 of the amino acid sequence shown in SEQ ID NO: 1. Said first and second extracellular loops preferably form the extracellular portion ofCLDN18.2.
  • the second target molecule of the bispecific binding agents described herein is CD3 (cluster of differentiation 3).
  • the CD3 complex denotes an antigen that is expressed on mature human T- cells, thymocytes and a subset of natural killer cells as part of the multimolecular T-cell receptor (TCR) complex.
  • the T-cell co-receptor is a protein complex and is composed of four distinct chains. In mammals, the complex contains a CD3y chain, a CD38 chain, and two CD3s chains. These chains associate with a molecule known as the T-cell receptor (TCR) and the (-chain to generate an activation signal in T lymphocytes.
  • TCR T-cell receptor
  • the TCR, ((-chain, and CD3 molecules together comprise the TCR complex.
  • the human CD3 a is indicated in GenBank Accession No. NM_OOO733.
  • the human CD3 y is indicated in GenBank Accession No. NM_000073.
  • the human CD3 5 is indicated in GenBank Accession No. NM 000732.
  • CD3 is responsible for the signal transduction of the TCR. As described by Lin and Weiss, Journal of Cell Science 114, 243-244 (2001), activation of the TCR complex by binding of MHC-presented specific antigen epitopes results in the phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) by Src family kinases, triggering recruitment of further kinases which results in T-cell activation including Ca 2+ release.
  • ITAMs immunoreceptor tyrosine-based activation motifs
  • CD3 includes human CD3 and denotes an antigen that is expressed on human T cells as part of the multimolecular T-cell receptor complex.
  • the binding agent described herein preferably recognizes the epsilon-chain of CD3. In some embodiments, it recognizes an epitope that corresponds to the first 27 N-terminal amino acids of CD3 epsilon or functional fragments of this 27 amino acid stretch.
  • CLDN18.2 positive cancer or similar terms mean a cancer involving cancer cells expressing CLDN18.2, preferably on the surface of said cancer cells.
  • Cell surface is used in accordance with its normal meaning in the art, and thus includes the outside of the cell which is accessible to binding by proteins and other molecules.
  • CLDN18.2 is expressed on the surface of cells if it is located at the surface of said cells and is accessible to binding by CLDN18.2-specific antibodies added to the cells.
  • CD3 is expressed on the surface of cells if it is located at the surface of said cells and is accessible to binding by CD3-specific antibodies added to the cells.
  • extracellular portion in the context of the present invention refers to a part of a molecule such as a protein that is facing the extracellular space of a cell and preferably is accessible from the outside of said cell, e.g., by antigen-binding molecules such as antibodies located outside the cell.
  • the term refers to one or more extracellular loops or domains or a fragment thereof.
  • part or “fragment” are used interchangeably herein and refer to a continuous element.
  • a part of a structure such as an amino acid sequence or protein refers to a continuous element of said structure.
  • a portion, a part or a fragment of a structure preferably comprises one or more functional properties of said structure.
  • a part or fragment of an amino acid sequence preferably comprises a sequence of at least 4, in particular at least 6, at least 8, at least 12, at least 15, at least 20, at least 30, at least 50, or at least 100 consecutive amino acids of the protein sequence.
  • “Fragment” with reference to an amino acid sequence (peptide or protein), relates to a part of an amino acid sequence, i.e. a sequence which represents the amino acid sequence shortened at the N-terminus and/or C-terminus.
  • a fragment shortened at the C-terminus is obtainable e.g. by translation of a truncated open reading frame that lacks the 3'-end of the open reading frame.
  • a fragment shortened at the N-terminus (C-terminal fragment) is obtainable e.g. by translation of a truncated open reading frame that lacks the 5 '-end of the open reading frame, as long as the truncated open reading frame comprises a start codon that serves to initiate translation.
  • a fragment of an amino acid sequence comprises e.g. at least 50%, at least 60%, at least 70%, at least 80%, at least 90% of the amino acid residues from an amino acid sequence.
  • variant of an amino acid sequence or similar expressions herein is meant an amino acid sequence that differs from a parent amino acid sequence by virtue of at least one amino acid modification.
  • the parent amino acid sequence may be a naturally occurring or wild type (WT) amino acid sequence, or may be a modified version of a wild type amino acid sequence.
  • WT wild type
  • the variant amino acid sequence has at least one amino acid modification compared to the parent amino acid sequence, e.g., from 1 to about 20 amino acid modifications, and preferably from 1 to about 10 or from 1 to about 5 amino acid modifications compared to the parent.
  • wild type or WT wild type or “WT” or “native” used with respect to an amino acid sequence herein is meant an amino acid sequence that is found in nature, including allelic variations.
  • a wild type amino acid sequence, peptide or protein has an amino acid sequence that has not been intentionally modified.
  • variants of an amino acid sequence comprise amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants.
  • variant includes all mutants, splice variants, posttranslationally modified variants, conformations, isoforms, allelic variants, species variants, and species homologs, in particular those which are naturally occurring.
  • variant includes, in particular, fragments of an amino acid sequence.
  • Amino acid insertion variants comprise insertions of single or two or more amino acids in a particular amino acid sequence.
  • amino acid sequence variants having an insertion one or more amino acid residues are inserted into a particular site in an amino acid sequence, although random insertion with appropriate screening of the resulting product is also possible.
  • Amino acid addition variants comprise amino- and/or carboxy-terminal fusions of one or more amino acids, such as 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids.
  • Amino acid deletion variants are characterized by the removal of one or more amino acids from the sequence, such as by removal of 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids.
  • the deletions may be in any position of the protein.
  • Amino acid deletion variants that comprise the deletion at the N-terminal and/or C-terminal end of the protein are also called N-terminal and/or C-terminal truncation variants.
  • amino acid substitution variants are characterized by at least one residue in the sequence being removed and another residue being inserted in its place. Preference is given to the modifications being in positions in the amino acid sequence which are not conserved between homologous proteins or peptides and/or to replacing amino acids with other ones having similar properties.
  • amino acid changes in peptide and protein variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids.
  • a conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains.
  • conservative substitutions may be defined by substitutions within the classes of amino acids reflected in the following table:
  • the degree of similarity, preferably identity between a given amino acid sequence and an amino acid sequence which is a variant of said given amino acid sequence will be at least about 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • the degree of similarity or identity is given preferably for an amino acid region which is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or about 100% of the entire length of the reference amino acid sequence.
  • the degree of similarity or identity is given preferably for at least about 20, at least about 40, at least about 60, at least about 80, at least about 100, at least about 120, at least about 140, at least about 160, at least about 180, or about 200 amino acids, in some embodiments, continuous amino acids.
  • the degree of similarity or identity is given for the entire length of the reference amino acid sequence.
  • the alignment for determining sequence similarity, preferably sequence identity can be done with art known tools, preferably using the best sequence alignment, for example, using Align, using standard settings, preferably EMBOSS ::needle, Matrix: Blosum62, Gap Open 10.0, Gap Extend 0.5.
  • Sequence similarity indicates the percentage of amino acids that either are identical or that represent conservative amino acid substitutions.
  • Sequence identity between two amino acid sequences indicates the percentage of amino acids that are identical between the sequences.
  • Sequnce identity between two nucleic acid sequences indicates the percentage of nucleotides that are identical between the sequences.
  • % identical refers, in particular, to the percentage of nucleotides or amino acids which are identical in an optimal alignment between the sequences to be compared. Said percentage is purely statistical, and the differences between the two sequences may be but are not necessarily randomly distributed over the entire length of the sequences to be compared. Comparisons of two sequences are usually carried out by comparing the sequences, after optimal alignment, with respect to a segment or "window of comparison", in order to identify local regions of corresponding sequences. The optimal alignment for a comparison may be carried out manually or with the aid of the local homology algorithm by Smith and Waterman, 1981, Ads App. Math. 2, 482, with the aid of the local homology algorithm by Neddleman and Wunsch, 1970, J.
  • NCBI National Center for Biotechnology Information
  • the algorithm parameters used for BLASTN algorithm on the NCBI website include: (i) Expect Threshold set to 10; (ii) Word Size set to 28; (iii) Max matches in a query range set to 0; (iv) Match/Mismatch Scores set to 1, -2; (v) Gap Costs set to Linear; and (vi) the filter for low complexity regions being used.
  • the algorithm parameters used for BLASTP algorithm on the NCBI website include: (i) Expect Threshold set to 10; (ii) Word Size set to 3; (iii) Max matches in a query range set to 0; (iv) Matrix set to BLOSUM62; (v) Gap Costs set to Existence: 11 Extension: 1; and (vi) conditional compositional score matrix adjustment.
  • Percentage identity is obtained by determining the number of identical positions at which the sequences to be compared correspond, dividing this number by the number of positions compared (e.g., the number of positions in the reference sequence) and multiplying this result by 100.
  • the degree of similarity or identity is given for a region which is at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100% of the entire length of the reference sequence.
  • the degree of identity is given for at least about 100, at least about 110, at least about 120, at least about 130, at least about 140, at least about 150, at least about 160, at least about 170, at least about 180, at least about 190, or about 200 nucleotides, in some embodiments, continuous nucleotides. In some embodiments, the degree of similarity or identity is given for the entire length of the reference sequence.
  • Homologous amino acid sequences exhibit according to the disclosure at least 40%, in particular at least 50%, at least 60%, at least 70%, at least 80%, at least 90% and preferably at least 95%, at least 98 or at least 99% identity of the amino acid residues.
  • amino acid sequence variants described herein may readily be prepared by the skilled person, for example, by recombinant DNA manipulation.
  • the manipulation of DNA sequences for preparing peptides or proteins having substitutions, additions, insertions or deletions, is described in detail in Sambrook et al. (1989), for example.
  • the peptides and amino acid variants described herein may be readily prepared with the aid of known peptide synthesis techniques such as, for example, by solid phase synthesis and similar methods.
  • a binding domain comprising functional VH and VL variants or functional CDR variant sequences has the same or similar binding characteristics compared to the parent molecule.
  • the modifications in the amino acid sequence of the parent molecule or sequence do not significantly affect or alter the characteristics of the molecule or sequence.
  • characteristics of a molecule comprising the functional fragment or functional variant may be reduced but still significantly present, e.g., binding characteristics such as binding strength of the binding domain comprising the functional variant may be at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the parent molecule or sequence.
  • a functional variant may comprise 1 , 2, 3, 4, 5, or more amino acid insertions, amino acid additions, amino acid substitutions and/or amino acid deletions compared to the parent molecule.
  • a "functional variant” is a "functional fragment”, e.g., an amino acid sequence that is shortened at the N-terminal and/or C-terminal end compared to the parent molecule, but retains or retains essentially one or more, or all, of the functions of the parent molecule, as described above, and in particular is functional equivalent to the parent molecule.
  • amino acid sequence "derived from" a designated amino acid sequence (peptide, protein or polypeptide, e.g., VH, VL, CHI, CH2 or CH3) refers to the origin of the first amino acid sequence.
  • amino acid sequence which is derived from a particular amino acid sequence has an amino acid sequence that is identical, essentially identical or homologous to that particular sequence or a fragment thereof.
  • Amino acid sequences derived from a particular amino acid sequence may be variants of that particular sequence or a fragment thereof, preferably functional variants thereof as described herein, including functional fragments.
  • amino acid sequences suitable for use herein may be altered such that they vary in sequence, including amino acid insertions, amino acid deletions, amino acid additions and/or amino acid substitutions, from the naturally occurring or native sequences from which they were derived, while retaining or essentially retaining the desirable activity of the native sequences.
  • amino acid sequences of the VH, VL, CHI , CH2 and/or CH3 domains on the peptide chains of the binding agent described herein are derived from amino acid sequences of VH, VL, CHI, CH2 and/or CH3 domains of immunoglobulins but maybe altered compared to the domains from which they are derived.
  • a VH or VL derived from an immunoglobulin comprises an amino acid sequence that can be identical to the amino acid sequence of the respective VH or VL it is derived from, or it can differ in one or more amino acid positions compared to the sequence of the respective parent VH or VL.
  • a VH domain of a binding agent described herein may comprise an amino acid sequence comprising one or more amino acid insertions, amino acid additions, amino acid deletions and/or amino acid substitutions compared to the amino acid sequence of the VH domain it is derived from.
  • a VL domain of a binding agent described herein may comprise an amino acid sequence comprising one or more amino acid insertions, amino acid additions, amino acid deletions and/or amino acid substitutions compared to the amino acid sequence of the VL domain it is derived from.
  • a VH or VL having an amino acid sequence that is a functional variant of the amino acid sequence of the parent VH or VL provides the same or essentially the same functions as the amino acid sequence of the parent VH or VL, e.g., in terms of binding specificity, binding strength etc.
  • an amino acid sequence e.g., of a VH or VL, which has altered characteristics compared to the amino acid sequence of the parent molecule.
  • the same considerations apply to amino acid sequences of, e.g., CDRs, and to other amino acid sequences, e.g., those of CHI, CH2, CH3 and/or CL domains.
  • bispecific binding agent When a bispecific binding agent is described to comprise a VH “derived from” an immunoglobulin and a VL “derived from” the same or a different immunoglobulin, the term “derived from” indicates that the bispecific binding agent was generated by recombining, by any known method, said VH and VL from said immunoglobulin(s) into the resulting bispecific binding agent.
  • recombining is not intended to be limited by any particular method of recombining and thus includes all of the methods for producing bispecific binding agents described herein or known in the art, including for example recombining at nucleic acid level and/or through co-expression of different molecules in the same cells.
  • binding agent refers to an agent having two different antigen-binding domains defined by different amino acid sequences.
  • said different antigen-binding domains bind different epitopes on the same antigen.
  • said different antigen-binding domains bind different target antigens.
  • a binding agent can bind each of the different antigens or different epitopes with one, two, or more binding domains, i.e., bind each of the different antigens or different epitopes monovalently, divalently (or bivalently), trivalently, tetravalently and even with valency of higher order.
  • CLDN18.2 is not substantially expressed in a cell if the level of expression is lower compared to expression in stomach cells or stomach tissue.
  • the level of expression is less than 10%, preferably less than 5%, 3%, 2%, 1%, 0.5%, 0.1% or 0.05% of the expression in stomach cells or stomach tissue or even lower.
  • CLDN18.2 is not substantially expressed in a cell if the level of expression exceeds the level of expression in non-cancerous tissue other than stomach by no more than 2-fold, preferably 1.5-fold, and preferably does not exceed the level of expression in said non-cancerous tissue.
  • CLDN 18.2 is not substantially expressed in a cell if the level of expression is below the detection limit and/or if the level of expression is too low to allow binding by CLDN18.2-specific antibodies added to the cell.
  • CLDN18.2 is expressed in a cell if the level of expression exceeds the level of expression in non- cancerous tissue other than stomach preferably by more than 2-fold, preferably 10-fold, 100-fold, 1000-fold, or 10000-fold.
  • CLDN18.2 is expressed in a cell if the level of expression is above the detection limit and/or if the level of expression is high enough to allow binding by CLDN18.2-specific antibodies added to the cell.
  • CLDN18.2 expressed in a cell is expressed or exposed on the surface of said cell.
  • the term "disease” refers to any pathological state, including cancer, in particular those forms of cancer described herein. Any reference herein to cancer or particular forms of cancer also includes cancer metastasis thereof.
  • a disease to be treated according to the present teaching involves cells expressing CLDN18.2.
  • expression of CLDN18.2 in cells of a diseased tissue or organ is increased compared to the state in a healthy tissue or organ.
  • An increase refers to an increase by at least 10%, in particular at least 20%, at least 50%, at least 100%, at least 200%, at least 500%, at least 1000%, at least 10000% or even more.
  • expression is only found in a diseased tissue, while expression in a healthy tissue is repressed.
  • diseases associated with cells expressing CLDN18.2 include cancer diseases.
  • cancer diseases preferably are those wherein the cancer cells express CLDN18.2.
  • a "cancer disease” or “cancer” includes a disease characterized by aberrantly regulated cellular growth, proliferation, differentiation, adhesion, and/or migration.
  • cancer cell is meant an abnormal cell that grows by a rapid, uncontrolled cellular proliferation and continues to grow after the stimuli that initiated the new growth cease.
  • a “cancer disease” is characterized by cells expressing CLDN18.2 and a cancer cell expresses CLDN18.2.
  • a cell expressing CLDN18.2 preferably is a cancer cell, preferably of the cancers described herein.
  • cancer comprises leukemias, seminomas, melanomas, teratomas, lymphomas, neuroblastomas, gliomas, rectal cancer, endometrial cancer, kidney cancer, adrenal cancer, thyroid cancer, blood cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, liver cancer, colon cancer, rectal cancer, colorectal cancer, stomach cancer, intestine cancer, head and neck cancer, bile duct cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, cancer of the gastroesophageal junction (GEJ), colorectal cancer, pancreas cancer, ear, nose and throat (ENT) cancer, breast cancer, prostate cancer, cancer of the uterus, ovarian cancer and lung cancer and the metastases thereof. Examples thereof are lung carcinomas, mamma carcinomas, prostate carcinomas, colon carcinomas, renal cell carcinomas, cervical carcinomas, or metastases of the cancer types or tumors described above.
  • cancer according to the invention also comprises cancer metastases thereof.
  • a "carcinoma” is a malignant tumor derived from epithelial cells. This group represents the most common cancers, including the common forms of breast, prostate, lung and colon cancer.
  • Adenocarcinoma is a cancer that originates in glandular tissue. This tissue is also part of a larger tissue category known as epithelial tissue. Epithelial tissue includes skin, glands and a variety of other tissue that lines the cavities and organs of the body. Epithelium is derived embryologically from ectoderm, endoderm and mesoderm. To be classified as adenocarcinoma, the cells do not necessarily need to be part of a gland, as long as they have secretory properties. This form of carcinoma can occur in some higher mammals, including humans. Well differentiated adenocarcinomas tend to resemble the glandular tissue that they are derived from, while poorly differentiated may not.
  • Adenocarcinomas can arise in many tissues of the body due to the ubiquitous nature of glands within the body. While each gland may not be secreting the same substance, as long as there is an exocrine function to the cell, it is considered glandular and its malignant form is therefore named adenocarcinoma. Malignant adenocarcinomas invade other tissues and often metastasize given enough time to do so. Ovarian adenocarcinoma is the most common type of ovarian carcinoma. It includes the serous and mucinous adenocarcinomas, the clear cell adenocarcinoma and the endometrioid adenocarcinoma.
  • metastasis is meant the spread of cancer cells from its original site to another part of the body.
  • the formation of metastasis is a very complex process and depends on detachment of malignant cells from the primary tumor, invasion of the extracellular matrix, penetration of the endothelial basement membranes to enter the body cavity and vessels, and then, after being transported by the blood, infiltration of target organs. Finally, the growth of a new tumor at the target site depends on angiogenesis. Tumor metastasis often occurs even after the removal of the primary tumor because tumor cells or components may remain and develop metastatic potential.
  • the term "metastasis” according to the invention relates to "distant metastasis” which relates to a metastasis which is remote from the primary tumor and the regional lymph node system.
  • the term "metastasis” according to the invention relates to lymph node metastasis.
  • One particular form of metastasis which is treatable using the therapy or methods of treatment of the invention is metastasis originating from gastric cancer as primary site.
  • gastric cancer metastasis is Krukenberg tumors, peritoneal metastasis and/or lymph node metastasis.
  • Krukenberg tumor is an uncommon metastatic tumor of the ovary accounting for 1% to 2% of all ovarian tumors. Prognosis of Krukenberg tumor is still very poor and there is no established treatment for Krukenberg tumors.
  • Krukenberg tumor is a metastatic signet ring cell adenocarcinoma of the ovary. Stomach is the primary site in most Krukenberg tumor cases (70%). Carcinomas of colon, appendix, and breast (mainly invasive lobular carcinoma) are the next most common primary sites. Rare cases of Krukenberg tumor originating from carcinomas of the gallbladder, biliary tract, pancreas, small intestine, ampulla of Vater, cervix, and urinary bladder/urachus have been reported.
  • treat is meant to administer a compound or composition or a combination of compounds or compositions to a subject in order to prevent, ameliorate or eliminate a disease, including reducing the size of a tumor or the number of tumors in a subject; arrest or slow a disease in a subject; inhibit or slow the development of a new disease in a subject; decrease the frequency or severity of symptoms and/or recurrences in a subject who currently has or who previously has had a disease; and/or prolong, i.e. increase the lifespan of the subject.
  • treatment of a disease includes curing, shortening the duration, ameliorating, preventing, slowing down or inhibiting progression or worsening, or preventing or delaying the onset of a disease or the symptoms thereof.
  • terms such as “protect”, “prevent”, “prophylactic”, “preventive”, or “protective” relate to the prevention or treatment or both of the occurrence and/or the propagation of a disease in a subject and, in particular, to minimizing the chance that a subject will develop a disease or to delaying the development of a disease.
  • a person at risk for cancer would be a candidate for therapy to prevent cancer.
  • being at risk is meant a subject that is identified as having a higher than normal chance of developing a disease, in particular cancer, compared to the general population.
  • a subject who has had, or who currently has, a disease, in particular cancer is a subject who has an increased risk for developing a disease, as such a subject may continue to develop a disease.
  • Subjects who currently have, or who have had, a cancer also have an increased risk for cancer metastases.
  • the terms “individual” and “subject” are used herein interchangeably. They refer to a human or another mammal (e.g. mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate) that can be afflicted with or is susceptible to a disease or disorder (e.g., cancer) but may or may not have the disease or disorder.
  • the individual is a human being.
  • the terms “individual” and “subject” do not denote a particular age, and thus encompass adults, elderlies, children, and newborns.
  • the "individual” or “subject” is a "patient”.
  • patient means according to the invention a subject for treatment, in particular a diseased subject, including human beings, nonhuman primates or another animals, in particular mammals such as cows, horses, pigs, sheeps, goats, dogs, cats or rodents such as mice and rats.
  • a patient is a human being.
  • “Target cell” shall mean any undesirable cell such as a cancer cell.
  • the target cell expresses CLDN18.2.
  • Activation refers to the state of an immune effector cell such as T cell that has been sufficiently stimulated to induce detectable cellular proliferation. Activation can also be associated with initiation of signaling pathways, induced cytokine production, and detectable effector functions.
  • activated immune effector cells refers to, among other things, immune effector cells that are undergoing cell division.
  • the term "priming" refers to a process wherein an immune effector cell such as a T cell has its first contact with its specific antigen and causes differentiation into effector cells such as effector T cells.
  • clonal expansion refers to a process wherein a specific entity is multiplied.
  • the term is preferably used in the context of an immunological response in which immune effector cells are stimulated by an antigen, proliferate, and the specific immune effector cell recognizing said antigen is amplified.
  • clonal expansion leads to differentiation of the immune effector cells.
  • the term "interact" means that two molecular species, e.g., two polypeptide chains or portions thereof, physically associate with each other.
  • the association that is characterized as an interaction can involve non-covalent and/or covalent, preferably non-covalent interactions, e.g., charge-charge interactions, charge-dipole interactions, dipole-dipole interactions, van der Waals forces, hydrogen bonding and/or hydrophobic forces.
  • bind or “binding” relates to the non-covalent interaction with a target.
  • the term “bind” or “binding” relates to a specific binding.
  • specific binding or “specifically binds”, as used herein, is meant a molecule such as an antibody which recognizes a specific target molecule, but does not substantially recognize or bind other molecules in a sample or in a subject.
  • an antibody that specifically binds to an antigen from one species may also bind to that antigen from one or more other species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific.
  • an antibody that specifically binds to an antigen may also bind to different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific.
  • the term "specific binding” or “specifically binds”, can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A” and the antibody, will reduce the amount of labeled A bound to the antibody.
  • a particular structure e.g., an antigenic determinant or epitope
  • binding moieties and binding agents may form complexes with CLDN 18.2 and/or CD3.
  • binding agent refers to any agent capable of binding to desired antigens.
  • a binding agent may also comprise synthetic, modified or non-naturally occurring moieties. Such moieties may, for example, link desired antigen-binding functionalities or regions such as antibodies or antibody fragments.
  • a binding agent is a synthetic construct comprising antigen-binding CDRs or variable regions.
  • binding agents disclosed herein comprise bispecific or multispecific binding agents such as bispecific antibody- derived binding agents comprising a first and a second binding domain, wherein the first binding domain is capable of binding to CLDN18.2 and the second binding domain is capable of binding to CD3.
  • binding domain refers to any region, moiety, group or domain which interacts with an antigen.
  • binding domain refers to the site of a binding agent described herein, that binds to an antigen and includes the antigen-binding portion of a binding agent.
  • the binding domain is or comprises an antibody, antibody fragment, or any other binding protein, or any combination thereof.
  • the binding domain may be comprised of heavy chain and light chain variable domains (VH and VL), each of which includes four conserved framework regions (FR) and three CDRs. The CDRs vary in sequence and determine the specificity to a particular antigen.
  • VH and VL domains together may form the site that binds, e.g., specifically binds, a particular antigen.
  • a "binding domain with specificity" for an antigen designates a binding domain, e.g., of a binding agent described herein, which binds to said antigen and preferably specifically binds to said antigen.
  • an agent such as an antibody is capable of binding to a predetermined target if it has a significant affinity for said predetermined target and binds to said predetermined target in standard assays.
  • "Affinity” or “binding affinity” is often measured by equilibrium dissociation constant (KD).
  • KD equilibrium dissociation constant
  • the term "significant affinity” refers to the binding to a predetermined target with a dissociation constant (KD) of 1 O’ 5 M or lower, 1 O' 6 M or lower, 10 ⁇ 7 M or lower, 10' 8 M or lower, 10' 9 M or lower, IO’ 10 M or lower, IO" 11 M or lower, or 10’ 12 M or lower.
  • An agent is not (substantially) capable of binding to a target if it has no significant affinity for said target and does not bind significantly, in particular does not bind detectably, to said target in standard assays.
  • the agent does not detectably bind to said target if present in a concentration of up to 2, preferably 10, more preferably 20, in particular 50 or 100 pg/ml or higher.
  • an agent has no significant affinity for a target if it binds to said target with a KD that is at least 10-fold, 100-fold, 10 3 -fold, 10 4 -fold, 10 5 -fold, or 10 6 -fold higher than the KD for binding to the predetermined target to which the agent is capable of binding.
  • the KD for binding of an agent to the target to which the agent is capable of binding is 10' 7 M
  • the KD for binding to a target for which the agent has no significant affinity would be at least 10’ 6 M, 10' 5 M, IO -4 M, 10’ 3 M, W 2 M, or 10’ 1 M.
  • a binding agent such as an antibody is specific for a predetermined target if it is capable of binding to said predetermined target while it is not capable of binding to other targets, i.e. has no significant affinity for other targets and does not significantly bind to other targets in standard assays.
  • a binding agent is specific for CLDN 18.2 if it is capable of binding to CLDN 18.2 but is not (substantially) capable of binding to other targets.
  • a binding agent is specific for CLDN18.2 if the affinity for and the binding to such other targets does not significantly exceed the affinity for or binding to CLDN18.2-unrelated proteins such as bovine serum albumin (BSA), casein, human serum albumin (HSA) or non-claudin transmembrane proteins such as MHC molecules or transferrin receptor or any other specified polypeptide.
  • BSA bovine serum albumin
  • HSA human serum albumin
  • a binding agent is specific for a predetermined target if it binds to said target with a KD that is at least 10-fold, 100-fold, 10 3 -fold, 10 4 -fold, 10 5 -fold, or 10 6 -fold lower than the KD for binding to a target for which it is not specific.
  • the KD for binding of a binding agent to the target for which it is specific is 10' 7 M
  • the KD for binding to a target for which it is not specific would be at least 10' 6 M, 1(F 5 M, 10' 4 M, 10’ 3 M, 10' 2 M, or 10' 1 M.
  • kd (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 kotr value.
  • KD (M), as used herein, refers to the dissociation equilibrium constant of a particular antibody-antigen interaction.
  • Binding of a binding agent to a target can be determined experimentally using any suitable method; see, for example, Berzofsky et al., "Antibody- Antigen Interactions" In Fundamental Immunology, Paul, W. E., Ed., Raven Press New York, N Y (1984), Kuby, Janis Immunology, W. H. Freeman and Company New York, N Y (1992), and methods described herein. Affinities may be readily determined using conventional techniques, such as by equilibrium dialysis; by using the BIAcore 2000 instrument, using general procedures outlined by the manufacturer; by radioimmunoassay using radiolabeled target antigen; or by another method known to the skilled artisan.
  • the affinity data maybe analyzed, for example, by the method of Scatchard et al., Ann N.Y. Acad. ScL, 51 :660 (1949).
  • the measured affinity of a particular interaction between binding agent and antigen can vary if measured under different conditions, e.g., salt concentration, pH.
  • affinity and other antigen-binding parameters e.g., KD, IC50, are preferably made with standardized solutions of binding agent and antigen, and a standardized buffer.
  • Compets refers to the competition between two binding agents such as antibodies for binding to a target antigen. If two binding agents do not block each other for binding to a target antigen, such binding agents are non-competing and this is an indication that said binding agents do not bind to the same part, i.e. epitope of the target antigen. It is well known to a person skilled in the art how to test for competition of binding agents for binding to a target antigen. An example of such a method is a so-called cross-competition assay, which may e.g. be performed as an ELISA or by flow- cytometry.
  • binding agents such as antibodies have the "same specificity" if they bind to the same antigen and to the same epitope. Such binding agents would compete for binding in a competition binding assay. In some embodiments, binding agents binding to the same epitope are considered to bind to the same amino acids on the target molecule. That antibodies bind to the same epitope on a target antigen may be determined by standard alanine scanning experiments or antibody-antigen crystallization experiments known to a person skilled in the art.
  • binding agents to compete for binding to an antigen indicates that these binding agents may bind to the same epitope region of the antigen or when binding to another epitope sterically hinder the binding of binding agents to that particular epitope region.
  • Competing binding agents can be readily identified based on their ability to compete with one or more binding agents in standard binding assays such as Surface Plasmon Resonance analysis, ELISA assays or flow cytometry (see WO 2013/173223).
  • the competition between binding agents can be detected by a cross-blocking assay.
  • a competitive ELISA assay may be performed by coating target antigen on the wells of a microtiter plate and adding antigen-binding agent and candidate competing test binding agent.
  • the amount of the antigen-binding agent bound to the antigen in the well indirectly correlates with the binding ability of the candidate competing test binding agent that competes therewith for binding, e.g., to the same epitope. Specifically, the larger the affinity of the candidate competing test binding agent is for the same epitope, the smaller the amount of the antigen-binding agent bound to the antigen-coated well.
  • the amount of the antigenbinding agent bound to the well can be measured by labelling the binding agent with detectable or measurable labelling substances. As described in WO 2013/173223 and as known in the art, Surface Plasmon Resonance analysis, e.g., using a Biacore instrument, can be used to identify overlapping versus different epitope regions recognized by binding agents. Alternatively, competition may be determined using biolayer interferometry.
  • a binding agent competing for binding to an antigen with another binding agent e.g., a binding agent comprising heavy and light chain variable regions as described herein, or a binding agent having the specificity for an antigen of another binding agent, e.g., a binding agent comprising heavy and light chain variable regions as described herein, such as an antibody, may be a binding agent comprising variants of said heavy and/or light chain variable regions as described herein, e.g. modifications in the CDRs and/or a certain degree of identity as described herein.
  • antigen relates to a molecule such as a protein or peptide comprising an epitope against which an agent is directed and/or is to be directed, preferably to induce an immune response.
  • An antigen or a procession product thereof such as a T-cell epitope is in some embodiments, bound by a T- or B-cell receptor, or by an immunoglobulin molecule such as an antibody. Accordingly, an antigen or a procession product thereof may react specifically with antibodies or T lymphocytes (T cells).
  • an antigen is a tumor-associated antigen, such as CLDN18.2, i.e., a constituent of cancer cells which may be derived from the cytoplasm, the cell surface and the cell nucleus, in particular those antigens which are produced, preferably in large quantity, intracellular or as surface antigens on cancer cells.
  • a tumor-associated antigen such as CLDN18.2
  • a constituent of cancer cells which may be derived from the cytoplasm, the cell surface and the cell nucleus, in particular those antigens which are produced, preferably in large quantity, intracellular or as surface antigens on cancer cells.
  • tumor-associated antigen or "cancer-associated antigen” preferably relates to proteins that are under normal conditions specifically expressed in a limited number of tissues and/or organs or in specific developmental stages and are expressed or aberrantly expressed in one or more tumor or cancer tissues.
  • the tumor- associated antigen is preferably associated with the cell surface of a cancer cell and is preferably not or only rarely expressed in normal tissues.
  • epitope refers to an antigenic determinant in a molecule, e.g., to the part in a molecule that is recognized by the immune system, for example, that is recognized by an antibody.
  • epitopes are the discrete, three-dimensional sites on an antigen, which are recognized by the immune system.
  • Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains 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.
  • An epitope of a protein preferably comprises a continuous or discontinuous portion of said protein and is preferably between 5 and 100, preferably between 5 and 50, more preferably between 8 and 30, most preferably between 10 and 25 amino acids in length, for example, the epitope may be preferably 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acids in length.
  • immunoglobulin relates to proteins of the immunoglobulin superfamily, preferably to antigen receptors such as antibodies or the B cell receptor (BCR).
  • the immunoglobulins are characterized by a structural domain, i.e., the immunoglobulin domain, having a characteristic immunoglobulin (Ig) fold.
  • the term encompasses membrane bound immunoglobulins as well as soluble immunoglobulins. Soluble immunoglobulins are generally termed antibodies.
  • Immunoglobulins generally comprise several chains, typically two identical heavy chains and two identical light chains which are linked via disulfide bonds.
  • immunoglobulin domains such as the VL (variable light chain) domain, CL (constant light chain) domain, the VH (variable heavy chain) and the CH (constant heavy chain) domains CHI, CH2, CH3, and CH4.
  • immunoglobulin heavy chains There are five types of mammalian immunoglobulin heavy chains, i.e., a, 5, e, y, and p which account for the different classes of immunoglobulins, i.e., IgA, IgD, IgE, IgG, and IgM.
  • Immunoglobulin classes are also referred to as "isotypes" (for instance IgGl, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM) referring to the immunoglobulin class that is encoded by heavy chain constant region genes.
  • isotypes for instance IgGl, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM
  • isotypes for instance IgGl, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM
  • isotypes for instance IgGl, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM
  • the term is not limited to a specific isotype sequence, e.g. a particular IgGl sequence, but is used to indicate that the antibody is closer in sequence to that isotype, e.g. IgG
  • the heavy chains of membrane or surface immunoglobulins comprise a transmembrane domain and a short cytoplasmic domain at their carboxy-terminus.
  • the immunoglobulin chains comprise a variable region and a constant region. The constant region is essentially conserved within the different isotypes of the immunoglobulins, wherein the variable part is highly divers and accounts for antigen recognition.
  • antibody refers to an immunoglobulin comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • antibody includes monoclonal antibodies, recombinant antibodies, human antibodies, humanized antibodies and chimeric antibodies.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region (amino acid residues 118-447 of human IgGl according to EU numbering) comprising CHI, CH2 and CH3 domains, wherein CHI is typically connected to CH2-CH3 by a peptide linker (also called “hinge”).
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region (abbreviated herein as CL).
  • VL light chain variable region
  • CL light chain constant region
  • region and the term “domain” are used interchangeably herein.
  • the VH and VL domains can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is 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 (see also Chothia and Lesk, J. Mol. Biol.
  • CDR sequences herein are identified according to the Kabat numbering system and reference to amino acid positions in the constant regions in the present invention is according to the EU-numbering (Edelman et al., (1969) Proc. Natl. Acad. Sci. USA 63(l):78-85; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Edit. 1991 NIH Publication No. 91-3242).
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • amino acid corresponding to position refers to an amino acid position number in a human IgGl heavy chain. Corresponding amino acid positions in other immunoglobulins may be found by alignment with human IgGl .
  • 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 IgGl 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.
  • IgG Fc ligand refers to a molecule, preferably a polypeptide, that binds to the Fc region of an IgG immunoglobulin to form an Fc/Fc ligand complex.
  • Fc ligands include but are not limited to I cyRIs, FcyRIIs, FcyRIIIs, FcRn, Clq, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcyR.
  • Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcyRs (Davis et al., (2002) Immunol. Rev. 190:123-136). Particular IgG Fc ligands are FcRn and Fc gamma receptors.
  • Fc ligand as used herein can be from any organism such as mouse, human and cynomolgus.
  • Fc gamma receptor refers to any member of the family of proteins that bind the IgG Fc region and is encoded by an FcyR gene. In humans this family includes but is not limited to FcyRI (CD64), including isoforms FcyRIa, FcyRIb, and FcyRIc; FcyRII (CD32), including isoforms FcyRIIa (including allotypes H131 and R131), FcyRIIb (including FcyRIIb-1 and FcyRIIb-2), and FcyRIIc; and FcyRIII (CD 16), including isoforms FcyRIIIa (including allotypes VI 58 and Fl 58) and FcyRIIIb (including allotypes FcyRIIb-NAl and FcyRIIb-NA2) (Jefferis et al., (2002) Immunol.
  • An FcyR may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Mouse FcyRs include but are not limited to FcyRI (CD64), FcyRII (CD32), FcyRIII (CD16), and FcyRIII-2 (CD16-2).
  • FcRn or "neonatal Fc Receptor” as used herein relates to a protein that binds the IgG Fc region and is encoded at least in part by an FcRn gene.
  • the FcRn may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • the functional FcRn protein comprises two polypeptides, often referred to as the heavy chain (encoded by the FcRn gene) and light chain (beta-2-microglobulin).
  • FcRn or “FcRn protein” refers to the complex of FcRn heavy chain with beta-2-microglobulin.
  • An "FcRn variant” as used herein is one that increases binding to the FcRn receptor and may also increase serum half-life.
  • Fc or Fc region or “Fc domain” as used herein refers to a polypeptide comprising the CH2 and CH3 domains of an IgG molecule and optionally a peptide linker such as the hinge.
  • the CH2-CH3 domains of human IgGl comprise amino acid positions 231-447, and the hinge comprises 216- 230, according to EU numbering.
  • the term “Fc domain” as used herein includes amino acid positions 231-447 (CH2-CH3) and 216-447 (hinge-CH2-CH3) according to EU numbering, and functional variants thereof including functional fragments thereof.
  • an "Fc fragment” may contain fewer amino acids, e.g., an N-terminal or C-terminal truncation variant, but still retains the ability to form a dimer with another Fc domain or Fc fragment, as can be detected using standard methods, e.g., based on size (e.g. non-denaturing chromatography, size exclusion chromatography, etc.), and thus is a functional variant.
  • an IgG Fc domain preferably is a human IgG Fc domain including the Fc domain from human IgGl, IgG2 or IgG4.
  • hinge refers to the peptide linker comprising the amino acids between CHI and CH2 of an immunoglobulin, e.g., IgG. Structurally, in naturally occurring IgG, e.g., IgGl, molecules, the CHI ends at amino acid position 215 according to EU numbering, and the CH2 begins at amino acid position 231 according to EU numbering.
  • the hinge includes amino acid positions 216 to 230 according to EU numbering.
  • a “variant Fc domain” contains amino acid modifications as compared to a parental Fc domain.
  • a “variant IgGl Fc domain” e.g., variant human IgGl Fc domain, contains amino acid modifications (e.g., amino acid substitutions and/or deletions) in positions corresponding to positions of an IgGl Fc domain, e.g., human IgGl Fc domain, and preferably is a functional variant of the parental Fc domain.
  • Such variant IgG Fc domains retain at least about 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to the corresponding parental human IgG Fc domain.
  • the variant Fc domains can have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11,12, 13, 14, 15, 16, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid modifications as compared to the parental Fc domain.
  • the variant Fc domains can have up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11,12, 13, 14, 15, 16, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid modifications as compared to the parental Fc domain.
  • a variant Fc domain retains the ability to form a dimer with another Fc domain as measured using techniques as described herein or known in the art, such as nondenaturing gel electrophoresis.
  • the binding agents described herein comprise three different polypeptide chains, wherein two polypeptide chains, e.g., the first polypeptide chain and the second polypeptide chain, comprise CH2-CH3 regions, preferably derived from IgG such as IgGl, in particular human IgGl.
  • said first and second polypeptide chains comprising CH2-CH3 regions are able to interact, e.g., dimerize thereby forming a heterodimer comprising said first and second polypeptide chain of a binding agent described herein.
  • the CH2-CH3 regions of the binding agents are derived from IgGl, more preferably human IgGl, even though CH2 and CH3 derived from other serotypes can be used as described herein.
  • the binding agents will self-assemble, e.g., within a producing host cell.
  • the CH2 on the first polypeptide chain interacts with the CH2 on the second polypeptide chain and/or the CH3 on the first polypeptide chain interacts with the CH3 on the second polypeptide chain, thereby forming an Fc fragment.
  • Fc fragments may comprise one or more amino acid modifications or "Fc modifications" as discussed herein with respect to human IgGl to promote interaction between the CH2 on the first and second polypeptide chains and/or CH3 on the first and second polypeptide chains, and/or allow for ease of purification of heteromultimers, such as heterodimers, containing first and second polypeptide chains interacting with one another over homomultimers only containing one type of polypeptide chain, and/or confer further beneficial functionalities as discussed herein.
  • Amino acid modifications discussed herein may also be contained in CHI domains, e.g., within the first and/or second polypeptide chains of a binding agent described herein.
  • peptide linkers such as peptide linkers within scFv moieties or peptide linkers connecting further domains of a binding agent, e.g., CHI and scFv, or CH2 and scFv, or CH 1 and CH2, may comprise one or more amino acid modifications as described herein.
  • a peptide linker connecting a CH2-CH3 region to another region or domain, such as VH(CD3), or CHI can have a serine at the amino acid position which corresponds to position 220 according to EU numbering in a naturally occurring IgGl (where normally a cysteine can be found).
  • a peptide linker comprising a serine at said position corresponding to position 220 of human IgGl reduces disulfide formation between the two chains comprising CH2-CH3 regions.
  • binding agents described herein is based on the use of different monomers (e.g., first and second polypeptide chains described herein) comprising CHI, CH2 and/or CH3 containing amino acid substitutions such as those that "skew” formation of heterodimers formed by said monomers over homodimers described herein, preferably coupled with "pl modifications” that allow for simple purification of the heterodimers away from the homodimers, and optionally in combination with “ablation modifications” and additional Fc modifications discussed herein.
  • monomers e.g., first and second polypeptide chains described herein
  • any of the amino acid modifications discussed herein with respect to CHI, CH2 and/or CH3 domains and peptide linkers, e.g., hinge variants, can be combined with further amino acid modifications discussed herein or known in the art.
  • any of the skew and pl modifications can independently be combined with ablation, and other Fc modifications.
  • An Fc modification according to the present invention can be an amino acid insertion, addition, deletion, or substitution.
  • N434S is an Fc modification with a substitution of serine at position 434 for asparagine relative to parental human IgGl Fc polypeptide and according to EU numbering.
  • the identity of the parental amino acid may be unspecified, in which case the aforementioned variant is referred to as 434S, i.e., the CH2-CH3 region comprising said Fc modification comprises a serine in an amino acid position that corresponds to position 434 in human IgGl according to EU numbering.
  • pl modifications pl modifications increase the isoelectric point (pl) difference between monomers what allows the isoelectric purification of homo- and heteromultimeric proteins.
  • a pl modification either increases the pl of the polypeptide chain (basic change) or decreases the pl of the polypeptide chain (acidic change).
  • a pl difference of at least 0.1, e.g., 0.2, 0.3, 0.4 or 0.5, between two polypeptide chains can allow separation by ion exchange chromatography or isoelectric focusing, or other methods sensitive to isoelectric point known in the art. Therefore, inclusion of pl modifications that alter the pl of each of the polypeptide chains interacting with one another, e.g., first and second polypeptide chains described herein, such that each of said polypeptide chains has a different pl and the heteromultimer formed by said polypeptide chains also has a distinct pl facilitates isoelectric purification of binding agents comprising said heteromultimer. These substitutions also aid in the determination and monitoring of any contaminating unwanted homo- or heteromultimer formation.
  • pl modifications can be contained within one or both chains of a heteromultimer comprising heavy chain constant regions, e.g., first and second polypeptide chains of a binding agent described herein, and within CHI, CH2 and/or CH3 and/or within peptide linkers such as linkers connecting the VH and VL domains of an scFv moiety.
  • pl variants can be contained in the first polypeptide chain and/or in the second polypeptide chain of a binding agent described herein to decrease or prevent homomultimer formation.
  • pl variants will be used such that the pl of one polypeptide chain is increased while the pl of the second polypeptide chain is decreased.
  • a sufficient change in pl in at least one of the polypeptide chains of a binding agent described herein is provided such that heteromultimers, e.g., of the first and second polypeptide chains, can be purified away from homomultimers.
  • a pl difference of as little as 0.1, 0.2, 0.3, 0.4 or 0.5 or greater in pH units is used in the present invention.
  • the amount of pl modifications to be included on one or more polypeptide chains of a binding agent described herein to get good separation will depend in part on the starting pl of the polypeptide chains, the pl of the CH regions, Fv scaffold regions etc. as will be understood by a person skilled in the art.
  • the change in pl can be calculated by any method known in the art, e.g., on the basis of the CH regions, e.g., using the method described by Sillero and Maldonado (Sillero, Maldonado, (2006) Comput. Biol. Med. 36(2):157-I66).
  • the pl of each polypeptide chain can be compared.
  • heteromultimers can be separated according to their size.
  • pl modifications, skew modifications, additional Fc modifications or ablation modifications etc. are not included in the variable regions of a binding agent described herein.
  • pl modifications are derived from different IgG isotypes such that the pl of the respective polypeptide chain is changed without introducing immunogenicity (cf., U.S. 2014/0370013).
  • pl modifications are derived from human IgG isotypes so as to decrease the risk of introducing immunogenicity.
  • the modifications discussed herein are described in relation to human IgGl, but all IgG isotypes can be altered this way, as well as isotype hybrids. In the case where the heavy chain constant domain is from IgG2-4, R133E and R133Q can also be used.
  • IgGl is a common isotype for therapeutic antibodies for a variety of reasons, including high effector function.
  • the CH region of IgGl has a higher pl than that of IgG2.
  • the pl of the resulting monomer is lowered or increased and, additionally, serum half-life maybe increased.
  • human IgGl has a glycine (pl approximately 5.97) at position 137 according to EU numbering
  • human IgG2 has a glutamic acid (pl approximately 3.22) at the corresponding position; substituting the glutamic acid residue for a glycine residue will affect the pl of the resulting polypeptide.
  • one polypeptide chain (e.g., the first polypeptide chain of a binding agent described herein, e.g. comprising VH(CLDN18.2), CHI , CH2 and CH3) comprises an aspartic acid residue at position 208, a glutamic acid residue at position 295, an aspartic acid residue at position 384, a glutamic acid residue at position 418 and an aspartic acid residue at position 421 (i.e., N208D/Q295E/N384D/Q418E/N421D with respect to human IgGl) according to EU numbering and another polypeptide chain (e.g., the second polypeptide chain of said binding agent, e.g.
  • VH(CLDN18.2), CHI, VL(CD3), VH(CD3), CH2 and CH3) comprises a positively charged peptide linker connecting VH(CD3) and VL(CD3) ("scFv linker"), such as a polypeptide linker comprising or consisting of the amino acid sequence (GKPGS)4 or a functional variant thereof.
  • polypeptide chains that do not comprise a CHI and, thus, do not include an amino acid position that corresponds to position 208 according to EU numbering the following negative pl modifications can be used: a glutamic acid residue at position 295, an aspartic acid residue at position 384, a glutamic acid residue at position 418 and an aspartic acid residue at position 421 (human IgGl: Q295E/N384D/Q418E/N421D) according to EU numbering.
  • one polypeptide chain e.g., the first polypeptide chain
  • a charged scFv linker such as a positively charged scFv linker of SEQ ID NO: 2 or a functional variant thereof, or a negatively charged scFv linker.
  • “Skew modifications” are steric modifications that facilitate interaction of the polypeptide chains comprising such modifications.
  • One strategy making use of steric modifications is referred to in the art as “knobs and holes”, referring to amino acid engineering, wherein a protuberance is introduced on a first heavy-chain polypeptide, typically in the Fc region (CH2-CH3), and a corresponding cavity in a second heavy-chain polypeptide, typically in the Fc region (CH2-CH3), such that the protuberance can be positioned in the cavity at the interface of these two heavy chains to promote heterodimer formation and hinder homodimer formation (see USSN 61/596,846, Ridgway et al., (1996) Protein Engineering 9(7):617; Atwell et al., (1997) J.
  • Protuberances are constructed by replacing small amino-acid sidechains from the interface of the first heavy-chain polypeptide with larger side chains. Compensatory "cavities” of identical or similar size to the protuberances are created in the interface of the second heavy-chain polypeptide by replacing large amino-acid side-chains with smaller ones (US patent 5,731,168). "Knobs and holes” modifications can be combined with disulfide bonds to skew fonnation to heteromultimerisation, e.g., heterodimerization of said first and second heavy-chain polypeptides (see Merchant et al., (1998) Nature Biotech. 16:677).
  • Useful skew modifications include without limitation the following pairs of double modifications, wherein one part of each pair of double modifications will be present in one polypeptide chain (e.g., the first polypeptide chain described herein) of the binding agent described herein and the second part will be present in another polypeptide chain (e.g., the second polypeptide chain described herein) of the binding agent described herein: S364K/E357Q : L368D/K37OS; L368D/K370S : S364K; L368D/K370S : S364K/E357Q; L368E/K37OS : S364K; T411E/K360E/Q362E : D401K; L368D/K370S : S364K/E357L; K370S : S364K/E357Q; T366S/L368A/Y407V: T366W and T366S/L368A/Y407V/
  • Skew modifications described herein may also have an effect on pl (cf., Gunasekaran et al., (2010) J. Biol. Chem. 285(25): 19637), and thus on purification, and, therefore, could also be considered pl variants.
  • ablation herein is meant a decrease or removal of activity.
  • “Ablating FcyR binding” means that an Fc region comprising one or more ablation modifications has more than 50% loss in FcyR binding activity as compared to an Fc region not containing the specific modifications.
  • the Fc region comprising the one or more ablation modifications has more than 70%, 80%, 90%, 95%, 98% or even more loss of FcyR binding activity.
  • the FcyR binding activity of an Fc region comprising the one or more ablation modifications as compared to an Fc region not containing the specific modifications is below the level of detectable binding in a Biacore, SPR or BLI assay.
  • the Fc domain of human IgGl has the highest binding to Fey receptors, and thus ablation modifications can be used when the constant domains of a binding agent are derived from IgGl.
  • ablation modifications mutations at the glycosylation position 297 (generally to A or S) can significantly ablate binding to FcyRIIIa, for example.
  • Human IgG2 and IgG4 have naturally lower binding to Fey receptors (Parren et al., 1992, J. Clin Invest.
  • CHI, CH2 and CH3 domains derived from IgG2 or IgG4 can be used with or without ablation modifications in binding agents described herein.
  • Amino acid modifications that ablate FcyR binding have, e.g., been described in Dall'Acqua WF et al., J Immunol. 177(2):1129-1138 (2006) and Hezareh M, J Virol.; 75(24): 12161-12168 (2001).
  • the Fc portion of a binding agent described herein may comprise one or more "FcyR ablation modifications” or “Fc knock out (FcKO or KO) modifications".
  • FcyR ablation modifications or "Fc knock out (FcKO or KO) modifications.
  • one or more of the polypeptide chains, e.g., the first polypeptide chain and the second polypeptide chain, of a binding agent described herein comprise one or more FcyR ablation variants.
  • the one or more ablation variants are selected from the group consisting of G236R, S239G, S239K, S239Q, S239R, V266D, S267K, S267R, H268K, E269R, 299R, 299K, K322A, A327G, A327L, A327N, A327Q, L328E, L328R, P329A, P329H, P329K, A330L, A330S/P331S, I332K, I332R, V266D/A327Q, V266D/P329K, S267R/A327Q, S267R/P329K, G236R
  • E233P/L234V/L235A/G236del/S239K/A327G E233P/L234V/L235A/G236del/S239K/A327G
  • E233P/L234V/L235A/G236del/S239K E233P/L234V/L235A/G236del/S239K/A327G
  • E233P/L234V/L235A/G236del/S267K/A327G E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V/L235A/G236del according to EU numbering and with respect to human IgGl, wherein "del" represents an amino acid deletion at the indicated position.
  • the modifications E233P/L234V/L235A/G236del/S267K according to EU numbering and with respect to human IgGl are used in both the first and the second polypeptide chain of the binding agent described herein.
  • the ablation modifications disclosed herein ablate FcyR binding but generally not FcRn binding.
  • techniques for reducing or increasing the binding to FcRn in order to reduce or increase serum half-life of the binding agent are known and can be used (see, e.g., Dall’Acqua et al. 2006, J. Biol. Chem., 281 :23514-24; Hinton et al. 2006, J. Immunol., 176:346-56; and Zalevsky et al. 2010 Nat. Biotechnol., 28:157-9).
  • the first polypeptide chain comprises an amino acid sequence according to SEQ ID NO: 7 and the second polypeptide chain comprises an amino acid sequence according to SEQ ID NO: 8, wherein the first and second polypeptide chains comprise the L368D/K370S : S364K/E357Q set of skew modifications, the first polypeptide chain further comprises the N208D/Q295E/N384D/Q418E/N421D set of pl modifications, and both the first and second polypeptide chain further comprises the E233P/L234V/L235A/G236del/S267K set of ablation modifications, e.g.
  • first polypeptide chain comprises VH(CLDN18.2) and CHI and the second polypeptide chain comprises scFv(CD3), VH(CLDN18.2) and CHI.
  • a binding agent may comprise additional amino acid modifications such as substitutions in addition to the modifications discussed above. Additional Fc modifications
  • Particular useful amino acid substitutions that can be incorporated into the binding agents described herein include but are not limited to 236A, 239D, 239E, 332E, 332D, 239D/332E, 267D, 267E, 328F, 267E/328F, 236A/332E, 239D/332E/330Y, 239D/332E/330L, 243A, 243L, 264A, 264V and 299T.
  • a CH3 on one or both polypeptide chains, preferably on both polypeptide chains, forming an Fc heterodimer may contain the modifications M428L/N434S resulting in longer serum half-life.
  • one polypeptide chain of a binding agent described herein comprises N208D, Q295E, N384D, Q418E and N481D according to EU numbering
  • another polypeptide chain of the binding agent e.g., the second polypeptide chain
  • the first polypeptide chain of the binding agent further comprises K370S and L368D
  • the second polypeptide chain of the binding agent further comprises E357Q and S364K, according to EU numbering.
  • both the first and second polypeptide chains further comprise E233P, L234V, L235A, G236del and S267K according to EU numbering.
  • the first polypeptide chain of a binding agent described herein comprises N208D, E233P, L234V, L235A, G236del, S267K, Q295E, L368D, K370S, N384D, Q418E and N481D
  • the second polypeptide chain of the binding agent comprises E233P, L234V, L235A, G236del, S267K, E357Q and S364K and, optionally, C220S to remove the cysteine that typically pairs with a light chain.
  • monoclonal binding agent as used herein includes a “monoclonal antibody” and refers to a preparation of binding agent molecules of single molecular composition.
  • a “monoclonal antibody” displays a single binding specificity and affinity for a particular epitope.
  • recombinant binding agent includes a “recombinant antibody” and includes all binding agents that are prepared, expressed, created or isolated by recombinant means.
  • human binding agent as used herein includes a "human antibody” and is intended to include binding agents having variable and constant regions derived from human germline immunoglobulin sequences. Human binding agents may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or sitespecific mutagenesis in vitro or by somatic mutation in vivo).
  • humanized binding agent includes a "humanized antibody” and refers to a molecule having an antigen binding site that is substantially derived from an immunoglobulin from a non-human species, wherein the remaining immunoglobulin structure of the molecule is based upon the structure and/or sequence of a human immunoglobulin. This can be achieved, e.g., by grafting of the six non-human antibody complementarity-determining regions (CDRs), which together form the antigen binding site, onto a homologous human acceptor framework region (FR) (see WO92/22653 and EP0629240).
  • CDRs non-human antibody complementarity-determining regions
  • the antigen binding site may either comprise complete variable domains fused onto constant domains or only the complementarity determining regions (CDR) grafted onto appropriate framework regions in the variable domains.
  • CDR complementarity determining regions
  • the substitution of framework residues from the parental binding agent i.e. the non-human binding agent, e.g., a murine antibody
  • Structural homology modeling may help to identify the amino acid residues in the framework regions that are important for the binding properties of the binding agent.
  • Antigen binding sites may be wild- type or modified by one or more amino acid substitutions, e.g., modified to resemble human immunoglobulins more closely.
  • humanized binding agents preserve all CDR sequences (for example a humanized mouse antibody which contains all six CDRs from the mouse antibody). Other forms have one or more CDRs which are altered with respect to the original binding agent, e.g., antibody.
  • a humanized binding agent 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.
  • chimeric binding agent includes a "chimeric antibody” and refers to those binding agents wherein one portion of each of the amino acid sequences of heavy and light chains is homologous to corresponding sequences in binding agents, e.g., antibodies, derived from a particular species or belonging to a particular class, while the remaining segment of the chain is homologous to corresponding sequences in another.
  • binding agents e.g., antibodies, derived from a particular species or belonging to a particular class
  • the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals, while the constant portions are homologous to sequences of antibodies derived from another.
  • variable region can conveniently be derived from presently known sources using readily available B cells or hybridomas from non-human host organisms in combination with constant regions derived from, for example, human cell preparations. While the variable region has the advantage of ease of preparation and the specificity is not affected by the source, the constant region being human, is less likely to elicit an immune response from a human subject when the binding agents are injected than would the constant region from a non-human source.
  • the definition is not limited to this particular example.
  • Binding agents or fragments thereof may be derived from different species, including but not limited to mouse, rat, rabbit, guinea pig and human.
  • Immunoglobulins described herein include IgA such as IgAl or IgA2, IgGl (including allotypes with polymorphisms at amino acid position 356 (D or E) and 358 (L or M) according to EU numbering), IgG2, IgG3, IgG4, IgE, IgM, and IgD antibodies.
  • the immunoglobulin is an IgGl antibody, more particularly an IgGl, kappa or IgGl, lambda isotype (i.e. IgGl, K, X), an IgG2a antibody (e.g. IgG2a, K, X), an IgG2b antibody (e.g.
  • IgG2b, K, X an IgG3 antibody (e.g. IgG3, K, X) or an IgG4 antibody (e.g. IgG4, K, X).
  • Amino acid sequences described herein with respect to IgGl allotype 356D/358M also include allotype 356E/358L.
  • the term "IgG subclass modification" or "isotype modification” means an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype. For example, because IgGl comprises a tyrosine and IgG2 a phenylalanine at amino acid position 296 according to EU numbering, a F296Y substitution in IgG2 is considered an IgG subclass modification.
  • a “heterologous binding agent” includes a “heterologous antibody” and is defined in relation to a transgenic organism producing such a binding agent. This term refers to a binding agent having an amino acid sequence or an encoding nucleic acid sequence corresponding to that found in an organism not consisting of the transgenic organism, and being generally derived from a species other than the transgenic organism.
  • a heterohybrid binding agent includes a “heterohybrid antibody” and refers to a binding agent having light and heavy chains of different organismal origins.
  • an antibody having a human heavy chain associated with a murine light chain is a heterohybrid antibody.
  • the binding agents including antibodies described herein are preferably isolated. "Isolated” as used herein, is intended to refer to a binding agent which is substantially free of other agents having different antigenic specificities (e.g., an isolated binding agent that specifically binds to CLDN18.2 and CD 3 is substantially free of binding agents that specifically bind antigens other than CLDN18.2 and CD3).
  • An isolated binding agent that specifically binds to an epitope, isofonn or variant of human CLDN18.2 may, however, have cross-reactivity to other related antigens, e.g., from other species (e.g., CLDN18.2 species homologs).
  • an isolated binding agent may be substantially free of other cellular material and/or chemicals.
  • antigen-binding portion of a binding agent such as an antibody (or simply “binding portion") or "antigen-binding fragment” of a binding agent such as an antibody (or simply “binding fragment”) or similar terms refer to one or more fragments of a binding agent that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding portion" of a binding agent such as an antibody include (i) Fab fragments, monovalent fragments consisting of the VL, VH, CL and CH domains; (ii) F(ab')2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fab' fragments, derived from a F(ab')2 fragment and containing a free sulfhydryl group that may be alkylated or utilized in conjugation with an enzyme, toxin or other protein of interest, wherein the Fab' may contain a small portion of Fc; (iv) Fd fragments consisting of the VH and CH domains; (v) Fv fragments consisting of the VL and VH domains of a single arm of an antibody, (vi) dAb fragments (Ward et al., (1989) Nature 341 : 544-546), which consist of a
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can 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 domains pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242: 423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of a binding agent such as an antibody.
  • a further example is binding-domain immunoglobulin fusion proteins comprising (i) a binding domain polypeptide that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region.
  • the binding domain polypeptide can be a heavy chain variable region or a light chain variable region.
  • the binding-domain immunoglobulin fusion proteins are further disclosed in US 2003/0118592 and US 2003/0133939. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • a single-chain variable fragment is a fusion protein of the variable regions of the heavy (VH) and light chains (VL) of immunoglobulins, connected with a short linker peptide of usually ten to about 30 amino acids such as an scFv linker, e.g., as represented by SEQ ID NO: 2.
  • the linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility, and can either connect the N-terminus of the VH with the C-terminus of the VL, or vice versa.
  • Divalent (or bivalent) single-chain variable fragments (di-scFvs, bi-scFvs) can be engineered by linking two scFvs.
  • the invention also includes multispecific molecules comprising more than one scFvs binding domain.
  • One common flexible linking peptide is (G4S) X , wherein x may be 2, 3, 4, 5 or 6.
  • a linker connecting the VH and VL domain of an scFv comprises, preferably consists of, the amino acid sequence (GKPGS) X or a functional variant thereof, wherein x may be 2, 3, 4, 5, or 6.
  • the association of the VH and VL can be stabilized by one or more intermolecular disulfide bonds.
  • Fab (fragment antigen binding) antibody fragments are immunoreactive polypeptides comprising monovalent antigen-binding domains of an antibody composed of a polypeptide consisting of a heavy chain variable region (VH) and heavy chain constant region 1 (CHI) portion and a polypeptide consisting of a light chain variable region (VL) and a light chain constant region (in which the CL and CHI portions are bound together, for example by a disulfide bond between Cys residues).
  • VH heavy chain variable region
  • CHI heavy chain constant region 1
  • VL light chain variable region
  • the CL and CHI portions are bound together, for example by a disulfide bond between Cys residues.
  • the CHI and the CL are of human origin.
  • the CL is a kappa-type CL.
  • the CHI is derived from IgGl, preferably from human IgGl.
  • antibody derivatives refers to any modified form of an antibody, e.g., a conjugate of the antibody and another agent or antibody, or an antibody fragment. Furthermore, the antibodies and derivatives of antibodies as described herein are useful for producing binding agents described herein.
  • Naturally occurring antibodies are generally monospecific, i.e. they bind to a single antigen.
  • the bispecific binding agents described herein bind to a cytotoxic cell such as a T cell (by engaging the CD3 receptor) and a target cell such as a cancer cell (by engaging CLDN18.2).
  • the binding agents described herein bind to at least two different types of antigen and are at least bispecific or multispecific such as trispecific, tetraspecific and so on.
  • a binding agent described herein is at least trivalent.
  • “valent”, “valence”, “valencies”, or other grammatical variations thereof, mean the number of antigen binding sites or binding domains in a binding agent. Antigen binding sites binding to the same antigen may recognize different epitopes or preferably the same epitope. A binding agent described herein may also have valencies of 4 or higher.
  • a binding agent described herein is preferably an artificial protein (including protein complexes) that may be composed of fragments of at least two different antibodies (said fragments of at least two different antibodies forming at least two different binding domains) and consequently binds to at least two different types of antigen.
  • a binding agent described herein is engineered to simultaneously bind to an immune cell, such as an immune effector cell, in particular a T cell such as a cytotoxic cell (e.g. by binding to CD3) and a target cell like a cancer cell (by binding to the tumor-associated antigen CLDN18.2) to be destroyed.
  • the binding agent described herein is in the format of a Fab2-scFv construct, i.e. a construct comprising two Fab fragments (each comprising VH and CHI domains on one polypeptide chain and corresponding VL und CL domains on another polypeptide chain, wherein an antigen binding domain is formed by interaction of each of said set of polypeptide chains) and an scFv moiety (comprising VH and VL domains connected to one another by a polypeptide linker on the same polypeptide chain, wherein the VH and VL interact to form an antigen binding domain).
  • a Fab2-scFv construct i.e. a construct comprising two Fab fragments (each comprising VH and CHI domains on one polypeptide chain and corresponding VL und CL domains on another polypeptide chain, wherein an antigen binding domain is formed by interaction of each of said set of polypeptide chains) and an scFv moiety (comprising VH and VL domain
  • the binding agent described herein is a tetramer composed of four polypeptide chains, wherein the first polypeptide chain comprises a VH derived from an immunoglobulin, e.g., an immunoglobulin with a first specificity, the second polypeptide comprises a VH derived from an immunoglobulin, e.g., an immunoglobulin with a first specificity, and an scFv moiety comprising a VH derived from an immunoglobulin, e.g., an immunoglobulin with a second specificity, and a VL derived from an immunoglobulin, e.g., an immunoglobulin with a second specificity, the third polypeptide chain comprises a VL derived from an immunoglobulin, e.g., an immunoglobulin with a first specificity, and the fourth polypeptide chain is identical to the third polypeptide chain.
  • the first polypeptide chain comprises a VH derived from an immunoglobulin, e.g
  • the first and second polypeptide chains further comprise a CHI derived from an immunoglobulin, e.g., C-terminal to the VH derived from an immunoglobulin with a first specificity
  • the third and fourth polypeptide chains further comprise a CL derived from an immunoglobulin.
  • the first and second polypeptide chains further comprise CH2 and CH3 (CH2-CH3) domains derived from an immunoglobulin, e.g., C-terminal to the Fab fragment and scFv moiety, respectively.
  • the binding agent described herein comprises a first polypeptide chain comprising VH-CH1 connected to CH2-CH3, a second polypeptide chain comprising VH-CH1 connected to an scFv moiety connected to CH2-CH3, and third and fourth polypeptide chains each comprising VL-CL.
  • the first polypeptide chain interacts with the second polypeptide chain.
  • the first polypeptide chain interacts with the third polypeptide chain.
  • the second polypeptide chain interacts with the fourth polypeptide chain.
  • the first and second polypeptide chains interact, the first polypeptide chain further interacts with the third polypeptide chain, and the second polypeptide chain further interacts with the fourth polypeptide chain.
  • the CH2 on the first polypeptide chain interacts with the CH2 on the second polypeptide chain and/or the CH3 on the first polypeptide chain interacts with the CH3 on the second polypeptide chain.
  • the VH on the first polypeptide chain interacts with the VL on the third polypeptide chain to form a binding domain and/or the CHI on the first polypeptide chain interacts with the CL on the third polypeptide chain.
  • the VH on the second polypeptide chain (which is not part of the scFv moiety) interacts with the VL on the fourth polypeptide chain to form a binding domain and/or the CHI on the second polypeptide chain interacts with the CL on the fourth polypeptide chain.
  • a disulfide bridge is formed between a cysteine residue in the CL and a cysteine residue in the CHI.
  • One or both polypeptide chains comprising CH2-CH3 may comprise one or more amino acid modifications, e.g., Fc modifications, described herein, such as pl, skew, additional Fc and ablation modifications, e.g., to promote polypeptide chain interaction.
  • the VH and VL of the scFv moiety are preferably connected by a peptide linker ("scFv linker").
  • scFv linker a peptide linker
  • the CHI on the first and/or second polypeptide chain is connected to the CH2 on the same polypeptide chain by a peptide linker.
  • the scFv is connected to the CH2 by a peptide linker.
  • the VH on the first polypeptide chain and the VL on the third polypeptide chain interact to form a binding domain with specificity for CLDN18.2
  • the additional VH on the second polypeptide chain that is not part of the scFv and the VL on the fourth polypeptide chain interact to form a binding domain with specificity for CLDN 18.2
  • the VH and VL of the scFv on the second polypeptide chain interact to form a binding domain with specificity for CDS.
  • linker refers to any means that serves to join two distinct functional units (e.g. domains or regions on a polypeptide chain).
  • Types of linkers include, but are not limited to, chemical linkers, peptide and polypeptide linkers.
  • the sequences of the peptide and polypeptide linkers are not limited.
  • Peptide linkers are preferably non-immunogenic and flexible, such as those comprising serine and glycine sequences. Depending on the particular construct, the linkers may be long or short.
  • the scFv linker i.e., the linker connecting VH and VL that form the scFv moiety, preferably comprises, and preferably consists of, a flexible peptide linker as described herein, preferably the amino acid sequence (GKPGS) X or a functional variant thereof, wherein x is 2, 3, 4, 5 or 6.
  • the scFv linker comprises, and preferably consists of, the amino acid sequence (GKPGS)4 (SEQ ID NO: 2) or a functional variant thereof.
  • the scFv moiety is connected to the CH2 on the second polypeptide chain by a peptide linker comprising the amino acid sequence (G4S)2KTHTCPPC (SEQ ID NO: 4) or a functional variant thereof.
  • a linker connecting an scFv and a CHI preferably at the C-terminus of the CHI, preferably comprises, preferably consists of, the amino acid sequence (G4S) X or a functional variant thereof, wherein x is 2, 3, 4, 5 or 6, preferably (G4S)2 (SEQ ID NO: 5) or a functional variant thereof.
  • a linker connecting CH2 and scFv, preferably at the N-terminus of CH2, preferably comprises, and preferably consists of, the amino acid sequence (G4S)2KTHTCPPC (SEQ ID NO: 4) or a variant thereof.
  • a linker connecting CH 1 and CH2 preferably comprises, and preferably consists of, the amino acid sequence EPKSCDKTHTCPPCP (SEQ ID NO: 6) or a functional variant thereof.
  • CHI and scFv, e.g., on the second polypeptide chain are connected by a peptide linker preferably comprising, preferably consisting of, the amino acid sequence (G4S)2 (SEQ ID NO: 5) or a functional variant thereof.
  • G4S amino acid sequence
  • other linkers can be used as known in the art.
  • the binding agent described herein comprises a first, a second, a third and a fourth polypeptide chain, wherein i) the first polypeptide chain comprises from N-terminus to C-terminus the following domains: VH(CLDN 18.2)-CH 1 -CH2-CH3 , ii) the second polypeptide chain comprises from N-terminus to C-terminus the following domains: VH(CLDN18.2)-CH1-VL(CD3)-VH(CD3)-CH2-CH3, iii) the third polypeptide chain comprises from N-terminus to C-terminus the following domains: VL(CLDN18.2)-CL, and iv) the fourth polypeptide chain is identical to the third polypeptide chain, preferably wherein VH(CLDN 18.2) on the first polypeptide chain and VL(CLDN 18.2) on the third polypeptide chain interact to form a binding domain with specificity for CLDN18.2, VH(CLDN18.2) on the second polypeptide chain and
  • the binding agent described herein comprises: a) a first polypeptide chain comprising a VH(CLDN18.2), and CHI, CH2 and CH3 comprising an aspartic acid residue at position 208, a proline residue at position 233, a valine residue at position 234, an alanine residue at position 235, a deletion at position 236, a lysine residue at position 267, a glutamic acid residue at position 295, an aspartic acid residue at position 368, a serine residue at position 370, an aspartic acid residue at position 384, a glutamic acid residue at position 418 and an aspartic acid residue at position 421 according to EU numbering; b) a second polypeptide chain comprising a VH(CLDN18.2) and a CHI as described herein, VH(CD3) and VL(CD3) connected to one another through a charged scFv linker with the amino acid sequence (GKPGS)4 (SEQ ID NO:
  • the binding agent described herein comprises a first, a second, a third and a fourth polypeptide chain, wherein i) the first polypeptide chain comprises from N-terminus to C-terminus: VH(CLDN 18.2)-CH 1 -linker-CH2-CH3 , ii) the second polypeptide chain comprises from N-terminus to C-terminus: VH(CLDN18.2)-CHl-linker-VL(CD3)-linker-VH(CD3)-linker-CH2-CH3, iii) the third polypeptide chain comprises from N-terminus to C-terminus the following domains: VL(CLDN18.2)-CL, and iv) the fourth polypeptide chain is identical to the third polypeptide chain, preferably wherein VH(CLDN 18.2) on the first polypeptide chain and VL(CLDN 18.2) on the third polypeptide chain interact to form a binding domain with specificity for CLDN18.2, VH(CLDN18.2) on the second polypeptide chain
  • the binding agent described herein comprises a first, a second, a third and a fourth polypeptide chain, wherein i) the first polypeptide chain comprises from N-terminus to C-terminus: VH(CLDN 18 ,2)-CH 1 -linker 1 -CH2-CH3 , ii) the second polypeptide chain comprises from N-terminus to C-terminus: VH(CLDN18.2)-CHl -Iinker2-VL(CD3)-Iinker3-VH(CD3)-Iinker4-CH2-CH3, iii) the third polypeptide chain comprises from N-terminus to C-terminus: VL(CLDN18.2)-CL, and iv) the fourth polypeptide chain is identical to the third polypeptide chain, wherein linkerl comprises the amino acid sequence EPKSCDKTHTCPPCP or a functional variant thereof, linker2 comprises the amino acid sequence (G4S) X or a functional variant thereof, wherein x is 2, 3, 4,
  • Binding agents described herein, and/or first, second, third, and fourth polypeptide chains of said binding agents described herein, may also comprise an amino acid sequence for facilitating secretion of the binding agent or polypeptide chain, such as a N -terminal secretion signal, and/or one or more epitope tags facilitating binding, purification or detection of the molecule.
  • the secretion signal is a signal sequence that allows a sufficient passage through the secretory pathway and/or secretion of the binding agent or the polypeptide chains thereof into the extracellular environment.
  • the secretion signal sequence is cleavable and is removed from the mature binding agent or polypeptide chain.
  • the secretion signal sequence preferably is chosen with respect to the cell or organism wherein the binding agent or polypeptide chain is produced in.
  • the amino acid sequence of an epitope tag may be introduced to any position within the amino acid sequence of the binding agent or polypeptide chain, and may take the shape of a loop within the encoded protein structure, or it may be N-terminally or C-terminally fused to the binding agent or polypeptide chain.
  • the epitope tag is C-terminally fused to the binding agent or polypeptide chain.
  • the epitope tag may contain a cleavage site that allows a removal of the tag from the binding agent or polypeptide chain.
  • Said epitope tag can be any kind of epitope tag that is functional under native and/or denaturing conditions, preferable a histidine tag, most preferable a tag comprising six histidines.
  • the binding agent described herein may contain, in addition to said first, second and third binding domains one or more further binding domains which serve e.g. to enhance selectivity for tumor cells. This can be achieved e.g. by providing binding domains that bind to other antigens expressed on tumor cells.
  • posttranslational modification refers to modifications of a protein, such as covalent and enzymatic modifications, that occur following protein biosynthesis.
  • binding agents such as antibodies that are expressed in cells are often modified after translation.
  • posttranslational modifications of binding agents can occur on the amino acid side chains or at the N- or C-termini of heavy or light chains, e.g., of the first and/or the second polypeptide chain described herein.
  • posttranslational modifications that may occur in binding agents described herein include, without limitation, cleavage of lysine at the C-terminus of a heavy chain, e.g., of the first and/or second polypeptide chain, e.g., by a carboxypeptidase; modification of glutamine or glutamic acid at the N-terminus of a heavy chain, e.g., of the first and/or second polypeptide chain, to pyroglutamic acid bypyroglutamylation; modification of glutamine or glutamic acid at the N-terminus of a light chain, e.g., of the third and/or fourth polypeptide chain, to pyroglutamic acid by pyroglutamylation; glycosylation; oxidation; deamidation; and glycation.
  • a binding agent described herein may comprise one or more posttranslational modifications.
  • the one or more posttranslational modifications comprise pyroglutamylation at the N-terminus of one or more polypeptide chains of the binding agent.
  • the one or more posttranslational modifications comprise pyroglutamylation at the N-terminus of one or more VH(CLDN18.2).
  • the one or more posttranslational modifications comprise deletion of lysine at the C-terminus of the first polypeptide chain.
  • the one or more posttranslational modifications comprise deletion of lysine at the C-terminus of the second polypeptide chain.
  • the binding agents described herein are preferably capable of eliciting one or more immune effector functions as described herein.
  • said immune effector functions are directed against cells carrying the cancer- associated antigen CLDN18.2 on their surface.
  • immune effector functions in the context of the present invention includes any functions mediated by components of the immune system that result e.g. in the inhibition of cancer growth and/or inhibition of cancer development, including inhibition of cancer dissemination and metastasis.
  • immune effector functions result in killing of cancer cells.
  • Immune effector functions comprise complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), induction of apoptosis in the cells carrying the cancer-associated antigen, cytolysis of the cells carrying the cancer-associated antigen, and/or inhibition of proliferation of the cells carrying the cancer- associated antigen.
  • the binding agents described herein preferably are able to recruit and redirect T cells such as CD4 and/or CD8 T cells, in particular CD107a+ T cells, to disease-associated cells such as cancer cells and, thus, act through redirected T cell cytotoxicity (RTCC), i.e., the T cells upon redirection preferably kill the disease-associated cells, e.g., cancer cells.
  • RTCC redirected T cell cytotoxicity
  • CD107a expression is known to be associated with cytolytic potential of CD4 and CD8 T cells.
  • said CD107a+ T cells are capable to degranulate, i.e., they are capable to release cytotoxic molecules such as perforines, granzymes, etc., and may also release cytokines such as one or more of Tumor Necrosis Factor a (TNFa), interleukine-2 (IL2), Interferon y (IFNy) etc., thereby causing death of target cells, e.g., cancer cells the T cells are redirected to by the binding agents described herein.
  • Binding agents may also exert an effect simply by binding to cancer-associated antigens on the surface of a cancer cell. For example, binding agents may block the function of the cancer- associated antigen or induce apoptosis just by binding to the cancer-associated antigen on the surface of a cancer cell.
  • immune effector cell or “effector cell” in the context of the present invention relates to a cell which exerts effector functions during an immune reaction.
  • immune effector cells comprise T cells (cytotoxic T cells, helper T cells, tumor infiltrating T cells), B cells, natural killer cells, neutrophils, macrophages, and dendritic cells.
  • T cell and “T lymphocyte” are used interchangeably herein and include T helper cells (CD4+ T cells) and cytotoxic T cells (CTLs, CD8+ T cells) which comprise cytolytic T cells.
  • MHC-dependent T cell or similar terms relate to a T cell which recognizes an antigen when presented in the context of MHC and preferably exerts effector functions of T cells, e.g., killing of target cells expressing an antigen.
  • T cells belong to a group of white blood cells known as lymphocytes, and play a central role in cell-mediated immunity. They can be distinguished from other lymphocyte types, such as B cells and natural killer cells by the presence of a special receptor on their cell surface called T cell receptor (TCR).
  • TCR T cell receptor
  • the thymus is the principal organ responsible for the maturation of T cells. Several different subsets of T cells have been discovered, each with a distinct function.
  • T helper cells assist other white blood cells in immunologic processes, including maturation of B cells into plasma cells and activation of cytotoxic T cells and macrophages, among other functions. These cells are also known as CD4+ T cells because they express the CD4 glycoprotein on their surface. Helper T cells usually become activated when they are presented with peptide antigens by MHC class II molecules that are expressed on the surface of antigen presenting cells (APCs). Once activated, they divide rapidly and secrete small proteins called cytokines that regulate or assist in the active immune response.
  • APCs antigen presenting cells
  • Cytotoxic T cells destroy virally infected cells and cancer cells, and are also implicated in transplant rejection. These cells are also known as CD8+ T cells since they express the CD 8 glycoprotein on their surface. These cells usually recognize their targets by binding to antigen associated with MHC class I, which is present on the surface of nearly every cell of the body.
  • T cells have a T cell receptor (TCR) existing as a complex of several proteins.
  • TCR T cell receptor
  • the TCR of a T cell is able to interact with immunogenic peptides (epitopes) bound to major histocompatibility complex (MHC) molecules and presented on the surface of target cells. Specific binding of the TCR triggers a signal cascade inside the T cell leading to proliferation and differentiation into a maturated effector T cell.
  • MHC major histocompatibility complex
  • TCRa and TCR0 major histocompatibility complex
  • TCRa and TCR0 T cell receptor alpha and beta
  • P-TCR chains A much less common (2% of total T cells) group of T cells, the y8 T cells (gamma delta T cells) possess a distinct T cell receptor (TCR) on their surface, which is made up of one y-chain and one 8-chain.
  • T cells originate from hematopoietic stem cells in the bone marrow.
  • Hematopoietic progenitors derived from hematopoietic stem cells populate the thymus and expand by cell division to generate a large population of immature thymocytes.
  • the earliest thymocytes express neither CD4 nor CD8, and are therefore classed as double-negative (CD4-CD8-) cells.
  • CD4+CD8+ double-positive thymocytes
  • CD4+CD8+ single- positive thymocytes that are then released from the thymus to peripheral tissues.
  • NK cell or “Natural Killer cell” refers to a subset of peripheral blood lymphocytes defined by the expression of CD56 or CD 16 and the absence of the T cell receptor.
  • MHC molecules in humans are normally referred to as HLA (human leukocyte antigen) molecules.
  • HLA human leukocyte antigen
  • MHC class I antigens are found on nearly all nucleated cells of the body.
  • the primary function of this class of MHC molecules is to display (or present) peptide fragments of intracellular proteins to CTLs. Based on this display, CTLs will attack those displaying MHC-bound peptides, including disease-associated peptides (antigens) such as cancer antigens.
  • cytotoxic T cells CTL
  • MHC class I molecules MHC class I molecules
  • binding agents described herein may be conjugated to a therapeutic moiety or agent, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radioisotope.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to and, in particular, kills cells.
  • Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1 -dehydrotesto sterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Suitable therapeutic agents for forming conjugates include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, fludarabin, 5 -fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (A
  • the therapeutic agent is a cytotoxic agent or a radiotoxic agent.
  • the therapeutic agent is an immunosuppressant.
  • the therapeutic agent is GM-CSF.
  • the therapeutic agent is doxorubicin, cisplatin, bleomycin, sulfate, carmustine, chlorambucil, cyclophosphamide or ricin A.
  • Binding agents also can be conjugated to a radioisotope, e.g., iodine-131, yttrium-90 or indium- 111 , to generate cytotoxic radiopharmaceuticals.
  • a radioisotope e.g., iodine-131, yttrium-90 or indium- 111 .
  • isotype refers to the antibody class (e.g., IgM or IgGl) that is encoded by heavy chain constant region genes.
  • isotype switching refers to the phenomenon by which the class, or isotype, of an antibody changes from one Ig class to one of the other Ig classes.
  • naturally occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.
  • rearranged refers to a configuration of a heavy chain or light chain immunoglobulin locus wherein a V segment is positioned immediately adjacent to a D-J or J segment in a conformation encoding essentially a complete VH or VL domain, respectively.
  • a rearranged immunoglobulin (antibody) gene locus can be identified by comparison to germline DNA; a rearranged locus will have at least one recombined heptamer/nonamer homology element.
  • unrearranged or “germline configuration” as used herein in reference to a V segment refers to the configuration wherein the V segment is not recombined so as to be immediately adjacent to a D or J segment.
  • a binding agent described herein has the ability of binding to CLDN18.2, i.e. the ability of binding to, and preferably binds to, an epitope present in CLDN18.2, preferably an epitope located within the extracellular domains of CLDN18.2, in particular the first extracellular loop, preferably amino acid positions 29 to 78 of CLDN18.2.
  • an agent having the ability of binding to CLDN 18.2 binds to an epitope on CLDN 18.2 which is not present on CLDN18.1.
  • An agent having the ability of binding to CLDN 18.2 preferably binds to CLDN 18.2, preferably of human, mouse and/or cynomolgus, but not to CLDN18.1, preferably of human, mouse and/or cynomolgus.
  • an agent binding to CLDN18.2 does not bind to CLDN9, preferably of human, mouse and/or cynomolgus.
  • an agent having the ability of binding to CLDN 18.2 is specific for CLDN18.2.
  • an agent having the ability of binding to CLDN18.2 binds to CLDN18.2 expressed on the cell surface.
  • an agent having the ability of binding to CLDN18.2 binds to native epitopes of CLDN18.2 present on the surface of living cells.
  • a binding domain comprises an antibody fragment.
  • fragment refers, in particular, to one or more of the complementarity-determining regions (CDRs), preferably at least the CDR3 variable region, of the heavy chain variable region (VH) and/or of the light chain variable region (VL).
  • said one or more of the complementarity-determining regions (CDRs) are selected from a set of complementaritydetermining regions CDR1, CDR2 and CDR3.
  • fragment refers to the complementarity-determining regions CDR1, CDR2 and CDR3 of the heavy chain variable region (VH) and/or of the light chain variable region (VL).
  • a binding domain comprising one or more CDRs, a set of CDRs or a combination of sets of CDRs as described herein comprises said CDRs together with their intervening framework regions.
  • the portion will also include at least about 50% of either or both of the first and fourth framework regions, the 50% being the C-terminal 50% of the first framework region and the N-terminal 50% of the fourth framework region. Construction of binding agents made by recombinant DNA techniques may result in the introduction of residues N- or C-terminal to the variable regions encoded by linkers introduced to facilitate cloning or other manipulation steps, including the introduction of linkers to join variable regions to further protein sequences including immunoglobulin heavy chains, other variable regions or protein labels.
  • a binding domain comprising one or more CDRs, a set of CDRs or a combination of sets of CDRs as described herein comprises said CDRs in a human antibody framework.
  • variable regions generally encompass amino acid residues 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3) in the VL and around about 31-35 (CDR1), 50-65 (CDR2), and 95-102 (CDR3) in the VH; variable regions may also encompass those residues forming a hypervariable loop (e.g.
  • the CDR sequences disclosed herein also comprise variants thereof derived according to different numbering systems. Accordingly, the disclosure of each VH is a disclosure of the CDRs (e.g., CDR1, CDR2 and CDR3) derivable therefrom and the disclosure of each VL is a disclosure of the CDRs (e.g., CDR1 , CDR2 and CDR3) derivable therefrom.
  • the Kabat numbering system is used when referring to residues in variable regions (approximately, residues 1-107 of the light chain and residues 1-113 of the heavy chain) of binding domains with specificity for CD3 or CLDN18.2, and the EU numbering system for CHI and CH2-CH3 (optionally including the hinge) regions, as described herein.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises a VH comprising complementarity determining regions CDR1, CDR2 and/or CDR3 identified within the amino acid sequence of SEQ ID NO: 16.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises a VL comprising complementarity determining regions CDR1, CDR2 and/or CDR3 identified within the amino acid sequence represented by SEQ ID NO: 17.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises the following set of CDRs: the VH comprises a CDR3 comprising the sequence set forth in SEQ ID NO: 12 or a functional variant thereof, and the VL comprises a CDR3 comprising the sequence set forth in SEQ ID NO: 15 or a functional variant thereof.
  • the VH further comprises a CDR1 comprising the sequence set forth in SEQ ID NO: 10 or a functional variant thereof and/or a CDR2 comprising the sequence set forth in SEQ ID NO: 11 or a functional variant thereof
  • the VL further comprises a CDR1 comprising the sequence set forth in SEQ ID NO: 13 or a functional variant thereof, and/or a CDR2 comprising the sequence set forth in SEQ ID NO: 14 or a functional variant thereof.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises the following set of CDRs: the VH comprises a CDR1 comprising the sequence set forth in SEQ ID NO: 10 or a functional variant thereof, a CDR2 comprising the sequence set forth in SEQ ID NO: 11 or a functional variant thereof and a CDR3 comprising the sequence set forth in SEQ ID NO: 12 or a functional variant thereof and the VL comprises a CDR1 comprising the sequence set forth in SEQ ID NO: 13 or a functional variant thereof, a CDR2 comprising the sequence set forth in SEQ ID NO: 14 or a functional variant thereof and a CDR3 comprising the sequence set forth in SEQ ID NO: 15 or a functional variant thereof.
  • the VH comprises CDR1, 2 and 3 of SEQ ID NOs: 10, 11 and 12
  • the VL comprises CDR1, 2 and 3 of SEQ ID NOs: 13, 14 and 15.
  • said heavy and light chain variable regions comprise said complementarity determining regions interspersed within framework regions.
  • each variable region comprises three complementarity determining regions (CDR1, 2, and 3) and four framework regions (FR1, 2, 3, and 4).
  • said complementarity determining regions and said framework regions are arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises a VH(CLDN18.2) comprising the amino acid sequences represented by SEQ ID NO: 16 or a functional variant thereof.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises VL(CLDN18.2) comprising the amino acid sequences represented by SEQ ID NO: 17 or a functional variant thereof.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises the following combination of VH(CLDN18.2) and VL(CLDN18.2): the VH(CLDN18.2) comprises an amino acid sequence represented by SEQ ID NO: 16 or a functional variant thereof and the VL(CLDN18.2) comprises an amino acid sequence represented by SEQ ID NO: 17 or a functional variant thereof.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein comprises heavy and light chain variable regions of an antibody which (i) competes for CLDN18.2 binding with an antibody comprising heavy and light chain variable regions as described above and/or (ii) has the specificity for CLDN18.2 of an antibody comprising heavy and light chain variable regions as described above.
  • a binding domain with specificity for CLDN18.2 of a binding agent described herein has the format of a Fab molecule as described herein.
  • a VH(CLDN18.2) is part of the first and the second polypeptide chains as described herein and a VL(CLDN 18.2) is part of the third polypeptide chain and the fourth polypeptide chain identical to the third polypeptide chain of a binding agent described herein.
  • binding domains binding to CLDN18.2 of a binding agent described herein in the Fab2-scFv format may be identical or essentially identical and thus may bind to identical or essentially identical epitopes of CLDN18.2.
  • both binding domains binding to CLDN18.2 of a binding agent described herein in the Fab2-scFv format may correspond or correspond essentially to one of the binding domains binding to CLDN18.2 described herein.
  • the binding domain with specificity for CD3 is capable of specifically recognizing human CD3 in the context of other TCR subunits as present on activated primary human T cells expressing the TCR in its native configuration.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises a VH comprising CDR1, CDR2 and/or CDR3 identified within the amino acid sequence of SEQ ID NO: 25.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises a VH comprising the following set of CDR1, CDR2 and CDR3:
  • CDR1 SEQ ID NO: 18 or a functional variant thereof
  • CDR2 SEQ ID NO: 23 or a functional variant thereof
  • CDR3 SEQ ID NO: 19 or a functional variant thereof.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises a VL comprising CDR1, CDR2 and/or CDR3 identified within the amino acid sequence according to SEQ ID NO: 24.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises a VL comprising the following set of CDR1 , CDR2 and CDR3 :
  • CDR1 SEQ ID NO: 20 or a functional variant thereof
  • CDR2 SEQ ID NO: 21 or a functional variant thereof
  • CDR3 SEQ ID NO: 22 or a functional variant thereof.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises the following combination of VH and VL each comprising a set of CDR1 , CDR2 and CDR3:
  • VH CDR1 : SEQ ID NO: 18 or a functional variant thereof
  • CDR2 SEQ ID NO: 23 or a functional variant thereof
  • CDR3 SEQ ID NO: 19 or a functional variant thereof
  • VL CDR1: SEQ ID NO: 20 or a functional variant thereof
  • CDR2 SEQ ID NO: 21 or a functional variant thereof
  • CDR3 SEQ ID NO: 22 or a functional variant thereof.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO: 25 or a functional variant thereof.
  • VH heavy chain variable region
  • a binding domain with specificity for CD3 of a binding agent described herein comprises a VL comprising the amino acid sequence set forth in SEQ ID NO: 24 or a functional variant thereof.
  • a binding domain with specificity for CD3 of a binding agent described herein comprises the following VH and a VL: the VH comprises or consists of an amino acid sequence according to SEQ ID NO: 25 or a functional variant thereof and the VL comprises or consists of an amino acid sequence according to SEQ ID NO: 24 or a functional variant thereof.
  • the binding domain with specificity for CD3 comprises or consists of the amino acid sequence of SEQ ID NO: 26 or a functional variant thereof.
  • the CHI of the first and/or second polypeptide chain of a binding agent described herein is derived from IgG, preferably IgGl, more preferably human IgGl.
  • the CH2 and CH3 domains of the first and/or second polypeptide chain of a binding agent described herein are derived from IgG, preferably IgGl , more preferably human IgGl .
  • the CL of the third and/or fourth polypeptide chain of a binding agent described herein is derived from IgK or IgA, preferably IgK, more preferably human IgK.
  • the first polypeptide chain of a binding agent described herein comprises an amino acid sequence represented by SEQ ID NO: 7 or a functional variant thereof.
  • the second polypeptide chain of a binding agent described herein comprises an amino acid sequence represented by SEQ ID NO: 8 or a functional variant thereof.
  • the third and/or fourth polypeptide chain of a binding agent described herein comprises an amino acid sequence represented by SEQ ID NO: 9 or a functional variant thereof.
  • a binding agent described herein comprises first, second, third, and fourth polypeptide chains comprising the following amino acid sequences: the first polypeptide chain comprises SEQ ID NO: 7 or a functional variant thereof, the second polypeptide chain comprises SEQ ID NO: 8 or a functional variant thereof, and the third polypeptide chain comprises SEQ ID NO: 9 or a functional variant thereof, wherein the fourth polypeptide chain is identical to the third polypeptide chain.
  • a binding agent described herein comprises at least two binding domains with specificity for CLN18.2 and at least one binding domain with specificity for CD3.
  • the first polypeptide chain of a binding agent described herein comprises or consists of the amino acid sequence set forth in SEQ ID NO: 27 or a functional variant thereof.
  • the second polypeptide chain of a binding agent described herein comprises or consists of the amino acid sequence set forth in SEQ ID NO: 28 or a functional variant thereof.
  • the third polypeptide chain of a binding agent described herein comprises or consists of the amino acid sequence set forth in SEQ ID NO: 29 or a functional variant thereof.
  • the fourth polypeptide chain of a binding agent described herein comprises or consists of the amino acid sequence set forth in SEQ ID NO: 29 or a functional variant thereof.
  • a binding agent described herein comprising at least two binding domains binding to CLDN18.2 and at least one binding domain binding to CD3 comprises a set of first, second, third and fourth polypeptide chains according to SEQ ID NOs: 27, 28, 29, and 29. It is to be understood that the binding agents described herein may be delivered to a patient by administering a nucleic acid such as RNA encoding the agent and/or by administering a host cell comprising a nucleic acid such as RNA encoding the agent.
  • the different polypeptide chains may be encoded on the same nucleic acid or on different nucleic acids, e.g., a set of nucleic acids.
  • a nucleic acid to be administered may be a mixture of different nucleic acid molecules such as a set of nucleic acids.
  • a nucleic acid or set of nucleic acids encoding a binding agent, e.g., when administered to a subject such as a patient, may be present in naked form or in a suitable delivery vehicle such as in the form of liposomes or nanoparticles or viral particles, or within a host cell.
  • nucleic acid or set of nucleic acids provided can produce the agent over extended time periods in a sustained manner mitigating the instability at least partially observed for therapeutic antibodies.
  • Nucleic acids or sets of nucleic acids to be delivered to a patient can be produced by recombinant means. If a nucleic acid or set of nucleic acids is administered to a patient without being present within a host cell, it is preferably taken up by cells of the patient for expression of the binding agent encoded by the nucleic acid(s). If a nucleic acid or set of nucleic acids is administered to a patient while being present within a host cell, it is preferably expressed by the host cell within the patient so as to produce the binding agent encoded by the nucleic acid(s).
  • recombinant in the context of the present invention means "made through genetic engineering”.
  • a “recombinant object” such as a recombinant nucleic acid in the context of the present invention is not occurring naturally.
  • naturally occurring refers to the fact that an object can be found in nature.
  • a peptide or nucleic acid that is present in an organism (including viruses) and can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.
  • nucleic acid is intended to include DNA and RNA such as genomic DNA, cDNA, mRNA, recombinantly produced and chemically synthesized molecules.
  • a nucleic acid may be single-stranded or double-stranded.
  • RNA includes in vitro transcribed RNA (IVT RNA) or synthetic RNA.
  • Nucleic acids or sets of nucleic acids may be comprised in a vector.
  • Sets of nucleic acids may also be comprised in sets of vectors such that each nucleic acid of the set of nucleic acids is comprised in a vector.
  • the term "vector" as used herein includes any vectors known to the skilled person including plasmid vectors, cosmid vectors, phage vectors such as lambda phage, viral vectors such as adenoviral or baculoviral vectors, or artificial chromosome vectors such as bacterial artificial chromosomes (BAC), yeast artificial chromosomes (YAC), or Pl artificial chromosomes (PAC). Said vectors include expression as well as cloning vectors.
  • Expression vectors comprise plasmids as well as viral vectors and generally contain a desired coding sequence and appropriate DNA sequences necessary for the expression of the operably linked coding sequence in a particular host organism (e.g., bacteria, yeast, plant, insect, or mammal) or in in vitro expression systems.
  • Cloning vectors are generally used to engineer and amplify a certain desired DNA fragment and may lack functional sequences needed for expression of the desired DNA fragments.
  • RNA relates to a molecule which comprises ribonucleotide residues and preferably is entirely or substantially composed of ribonucleotide residues.
  • “Ribonucleotide” relates to a nucleotide with a hydroxyl group at the 2'-position of a p- D-ribofuranosyl group.
  • the term includes double stranded RNA, single stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as modified RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides.
  • Such alterations can include addition of non-nucleotide material, such as to the end(s) of a RNA or internally, for example at one or more nucleotides of the RNA.
  • Nucleotides in RNA molecules can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • RNA includes and preferably relates to "mRNA” which means “messenger RNA” and relates to a “transcript” which may be produced using DNA as template and encodes a peptide or protein.
  • mRNA typically comprises a 5' non translated region (5'-UTR), a protein or peptide coding region and a 3' non translated region (3'-UTR).
  • mRNA has a limited halftime in cells and in vitro.
  • mRNA is produced by in vitro transcription using a DNA template.
  • the RNA is obtained by in vitro transcription or chemical synthesis.
  • the in vitro transcription methodology is known to the skilled person. For example, there is a variety of in vitro transcription kits commercially available.
  • RNA is self-replicating RNA, such as single stranded self-replicating RNA.
  • the self-replicating RNA is single stranded RNA of positive sense.
  • the self-replicating RNA is viral RNA or RNA derived from viral RNA.
  • the self-replicating RNA is alphaviral genomic RNA or is derived from alphaviral genomic RNA.
  • Alphaviral RNA may act as mRNA, as is known in the art.
  • the self-replicating RNA is a viral gene expression vector.
  • the virus is Semliki forest virus.
  • the self-replicating RNA contains one or more transgenes at least one of said transgenes encoding the binding agent described herein.
  • the transgenes may partially or completely replace viral sequences such as viral sequences encoding structural proteins.
  • the self-replicating RNA is in vitro transcribed RNA.
  • RNA used according to the present invention may be modified, preferably without altering the sequence of the expressed peptide or protein.
  • modification in the context of RNA as used according to the present invention includes any modification of RNA which is not naturally present in said RNA.
  • the RNA used according to the invention does not have uncapped 5 '-triphosphates. Removal of such uncapped 5'-triphosphates can be achieved by treating RNA with a phosphatase.
  • RNA according to the invention may have modified naturally occurring or synthetic ribonucleotides in order to increase its stability and/or decrease cytotoxicity and/or immunogenicity.
  • 5-methylcytidine is substituted partially or completely, preferably completely, for cytidine.
  • pseudouridine is substituted partially or completely, preferably completely, for uridine.
  • the term "modification” relates to providing an RNA with a 5'-cap or 5'- cap analog.
  • the term “5'-cap” refers to a cap structure found on the 5'-end of an mRNA molecule and generally consists of a guanosine nucleotide connected to the mRNA via an unusual 5' to 5'- triphosphate linkage. In some embodiments, this guanosine is methylated at the 7-position.
  • the term “conventional 5'-cap” refers to a naturally occurring RNA 5'-cap, preferably to the 7- methylguanosine cap (m7G).
  • 5'-cap includes a 5'-cap analog that resembles the RNA cap structure and is modified to possess the ability to stabilize RNA if attached thereto, preferably in vivo and/or in a cell.
  • RNA with a 5'-cap or 5'-cap analog may be achieved by in vitro transcription of a DNA template in the presence of said 5'-cap or 5'-cap analog, wherein said 5'-cap is co- transcriptionally incorporated into the generated RNA strand, or the RNA may be generated, for example, by in vitro transcription, and the 5'-cap maybe attached to the RNA post-transcriptionally using capping enzymes, for example, capping enzymes of vaccinia virus.
  • capping enzymes for example, capping enzymes of vaccinia virus.
  • RNA may comprise further modifications.
  • a further modification of the RNA used in the present invention may be an extension or truncation of the naturally occurring poly(A) tail or an alteration of the 5'- or 3 '-untranslated regions (UTR) such as introduction of a UTR which is not related to the coding region of said RNA, for example, the insertion of one or more, preferably two copies of a 3 '-UTR derived from a globin gene, such as alpha2-globin, alphal- globin, beta-globin, preferably beta-globin, more preferably human beta-globin.
  • UTR 5'- or 3 '-untranslated regions
  • the RNA used according to the present invention may be modified so as to be present in conjunction with a poly- A sequence, preferably having a length of 10 to 500, more preferably 30 to 300, even more preferably 65 to 200 and especially 100 to 150 adenosine residues.
  • the poly- A sequence has a length of approximately 120 adenosine residues.
  • incorporation of two or more 3'-non translated regions (UTR) into the 3'-non translated region of an RNA molecule can result in an enhancement in translation efficiency.
  • the 3 '-UTR is derived from the human P-globin gene.
  • RNA if delivered to, i.e. transfected into, a cell, in particular a cell present in vivo, expresses the protein, peptide or antigen it encodes.
  • transfection relates to the introduction of nucleic acids, in particular RNA, into a cell.
  • the term “transfection” also includes the introduction of a nucleic acid into a cell or the uptake of a nucleic acid by such cell, wherein the cell may be present in a subject, e.g., a patient.
  • a cell for transfection of a nucleic acid described herein can be present in vitro or in vivo, e.g. the cell can form part of an organ, a tissue and/or an organism of a patient.
  • transfection can be transient or stable.
  • transfected genetic material is only transiently expressed. Since the nucleic acid introduced in the transfection process is usually not integrated into the nuclear genome, the foreign nucleic acid will be diluted through mitosis or degraded. Cells allowing episomal amplification of nucleic acids greatly reduce the rate of dilution. If it is desired that the transfected nucleic acid actually remains in the genome of the cell and its daughter cells, a stable transfection must occur. RNA can be transfected into cells to transiently express its coded protein.
  • RNA relates to the "half-life" of RNA.
  • "Half-life” relates to the period of time which is needed to eliminate half of the activity, amount, or number of molecules.
  • the half-life of an RNA is indicative for the stability of said RNA.
  • the half-life of RNA may influence the "duration of expression" of the RNA. It can be expected that RNA having a long half-life will be expressed for an extended time period.
  • the term “transcription” relates to a process, wherein the genetic code in a DNA sequence is transcribed into RNA. Subsequently, the RNA may be translated into protein.
  • the term “transcription” comprises "in vitro transcription", wherein the term “in vitro transcription” relates to a process wherein RNA, in particular mRNA, is in vitro synthesized in a cell-free system, preferably using appropriate cell extracts.
  • cloning vectors are applied for the generation of transcripts. These cloning vectors are generally designated as transcription vectors and are according to the present invention encompassed by the term "vector".
  • translation relates to the process in the ribosomes of a cell by which a strand of messenger RNA directs the assembly of a sequence of amino acids to make a peptide or protein.
  • expression is used according to the invention in its most general meaning and comprises the production of RNA and/or peptides or proteins, e.g. by transcription and/or translation.
  • expression or “translation” relates in particular to the production of peptides or proteins. It also comprises partial expression of nucleic acids. Moreover, expression can be transient or stable.
  • expression also includes an "aberrant expression” or "abnormal expression”. "Aberrant expression” or "abnormal expression” means according to the invention that expression is altered, preferably increased, compared to a reference, e.g.
  • an increase in expression refers to an increase by at least 10%, in particular at least 20%, at least 50% or at least 100%, or more. In some embodiments, expression is only found in a diseased tissue, while expression in a healthy tissue is repressed.
  • a tumor antigen specifically expressed in gastric mucosa means that said protein is primarily expressed in gastric mucosa and is not expressed in other tissues or is not expressed to a significant extent in other tissue or organ types.
  • a protein that is exclusively expressed in cells of the gastric mucosa and to a significantly lesser extent in any other tissue, such as testis is specifically expressed in cells of the gastric mucosa.
  • a tumor antigen may also be specifically expressed under normal conditions in more than one tissue type or organ, such as in 2 or 3 tissue types or organs, but preferably in not more than 3 different tissue or organ types. In this case, the tumor antigen is then specifically expressed in these organs. For example, if a tumor antigen is expressed under normal conditions preferably to an approximately equal extent in lung and stomach, said tumor antigen is specifically expressed in lung and stomach.
  • RNA encoding means that RNA, if present in the appropriate environment, preferably within a cell, can be expressed to produce a protein or peptide it encodes.
  • Some aspects of the invention rely on the adoptive transfer of host cells which are transfected in vitro with a nucleic acid such as RNA encoding a binding agent described herein and transferred to recipients such as patients, preferably after ex vivo expansion from low precursor frequencies to clinically relevant cell numbers.
  • the host cells used for treatment according to the invention may be autologous, allogeneic, or syngeneic to a treated recipient.
  • autologous is used to describe anything that is derived from the same subject.
  • autologous transplant refers to a transplant of tissue or organs derived from the same subject. Such procedures are advantageous because they overcome the immunological barrier which otherwise results in rejection.
  • allogeneic is used to describe anything that is derived from different individuals of the same species. Two or more individuals are said to be allogeneic to one another when the genes at one or more loci are not identical.
  • genotypeic is used to describe anything that is derived from individuals or tissues having identical genotypes, i.e., identical twins or animals of the same inbred strain, or their tissues.
  • heterologous is used to describe something consisting of multiple different elements. As an example, the transfer of one individual's bone marrow into a different individual constitutes a heterologous transplant.
  • a heterologous gene is a gene derived from a source other than the subject.
  • peptide comprises oligo- and polypeptides and refers to substances comprising two or more, preferably 3 or more, preferably 4 or more, preferably 6 or more, preferably 8 or more, preferably 9 or more, preferably 10 or more, preferably 13 or more, preferably 16 more, preferably 21 or more and up to preferably 8, 10, 20, 30, 40 or 50, in particular 100 amino acids joined covalently by peptide bonds.
  • protein refers to large peptides, preferably to peptides with more than 100 amino acid residues, but in general the terms “peptides” and “proteins” are synonyms and are used interchangeably herein.
  • a sequence which is a variant with respect to a specific sequence when it replaces the specific sequence in a binding agent retains binding of said binding agent to CLDN18.2 and/or CD3 and preferably functions of said binding agent as described herein, e.g. cytotoxic T-cell mediated lysis.
  • sequences shown in the sequence listing can be modified so as to remove one or more, preferably all free cysteine residues, in particular by replacing the cysteine residues by amino acids other than cysteine, preferably serine, alanine, threonine, glycine, tyrosine, tryptophan, leucine or methionine.
  • CDR regions will be either identical or highly homologous to the regions specified herein.
  • highly homologous it is contemplated that from 1 to 5, preferably from 1 to 4, such as 1 to 3 or 1 or 2 substitutions may be made in the CDRs.
  • the hypervariable and variable regions may be modified so that they show substantial homology with the regions specifically disclosed herein.
  • the variable region sequences only deviate in the framework sequences from the variable region sequences specifically disclosed herein.
  • the binding agents described herein can be produced either intracellularly (e.g. in the cytosol, in the periplasma or in inclusion bodies) and then isolated from the host cells and optionally further purified; or they can be produced extracellularly (e.g. in the medium in which the host cells are cultured) and then isolated from the culture medium and optionally further purified.
  • Methods and reagents used for the recombinant production of polypeptides such as specific suitable expression vectors, transformation or transfection methods, selection markers, methods of induction of protein expression, culture conditions, and the like, are known in the art.
  • protein isolation and purification techniques are well known to the skilled person.
  • cell or "host cell” preferably relates to an intact cell, i.e. a cell with an intact membrane that has not released its normal intracellular components such as enzymes, organelles, or genetic material.
  • An intact cell preferably is a viable cell, i.e. a living cell capable of carrying out its normal metabolic functions.
  • said term relates according to the invention to any cell which can be transfected with an exogenous nucleic acid.
  • the cell when transfected with an exogenous nucleic acid and transferred to a recipient can express the nucleic acid in the recipient.
  • the term "cell” includes bacterial cells; other useful cells are yeast cells, fungal cells or mammalian cells.
  • Suitable bacterial cells include cells from gram-negative bacterial strains such as strains of Escherichia coli, Proteus, and Pseudomonas, and gram-positive bacterial strains such as strains of Bacillus, Streptomyces, Staphylococcus, and Lactococcus.
  • Suitable fungal cells include cells from species of Trichoderma, Neurospora, and Aspergillus.
  • Suitable yeast cells include cells from species of Saccharomyces (for example Saccharomyces cerevisiae), Schizosaccharomyces (for example Schizosaccharomyces pombe), Pichia (for example Pichia pastoris and Pichia methanolica), and Hansenula.
  • Suitable mammalian cells include for example CHO cells, BHK cells, HeLa cells, COS cells, 293 HEK and the like. However, amphibian cells, insect cells, plant cells, and any other cells used in the art for the expression of heterologous proteins can be used as well. Mammalian cells are particularly preferred for adoptive transfer, such as cells from humans, mice, hamsters, pigs, goats, and primates.
  • the cells may be derived from a large number of tissue types and include primary cells and cell lines such as cells of the immune system, in particular antigen-presenting cells such as dendritic cells and T cells, stem cells such as hematopoietic stem cells and mesenchymal stem cells and other cell types.
  • An antigen-presenting cell is a cell that displays antigen in the context of major histocompatibility complex on its surface. T cells may recognize this complex using their T-cell receptor (TCR).
  • TCR T-cell receptor
  • the ability of antibodies and other binding agents to bind an antigen can be determined using standard binding assays (e.g., ELISA, Western Blot, immunofluorescence and flow cytometric analysis).
  • standard binding assays e.g., ELISA, Western Blot, immunofluorescence and flow cytometric analysis.
  • Binding agents described herein also can be tested in an in vivo model (e.g. in immunodeficient mice carrying xenografted tumors inoculated with cell lines expressing CLDN18.2) to determine their efficacy in controlling growth of CLDN18.2-expressing tumor cells.
  • Reduce means an overall decrease or the ability to cause an overall decrease, preferably of 5% or greater, 10% or greater, 20% or greater, more preferably of 50% or greater, and most preferably of 75% or greater, in the level, e.g. in the level of expression or in the level of proliferation of cells.
  • Terms such as "increase” or “enhance” preferably relate to an increase or enhancement by about at least 10%, preferably at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 80%, and most preferably at least 100%, at least 200%, at least 500%, at least 1000%, at least 10000% or even more.
  • ADCC describes the cell-killing ability of effector cells as described herein, in particular lymphocytes, which preferably requires the target cell being marked by an antibody.
  • ADCC preferably occurs when antibodies bind to antigens on tumor cells and the antibody Fc domains engage Fc receptors (FcR) on the surface of immune effector cells.
  • FcR Fc receptors
  • Several families of Fc receptors have been identified, and specific cell populations characteristically express defined Fc receptors.
  • ADCC can be viewed as a mechanism to directly induce a variable degree of immediate tumor destruction that leads to antigen presentation and the induction of tumor-directed T-cell responses.
  • in vivo induction of ADCC will lead to tumor-directed T-cell responses and host-derived antibody responses.
  • ADCP is one mechanism of action of many antibody therapies. It is defined as a highly regulated process by which antibodies eliminate bound targets via connecting their Fc domain to specific receptors on phagocytic cells, and eliciting phagocytosis. ADCP can be mediated by monocytes, macrophages, neutrophils, and dendritic cells, through FcyRIIa, FcyRI, and FcyRlIIa, of which FcyRIIa (CD32a) on macrophages represent the predominant pathway.
  • ADCP preferably occurs when non-specific phagocytic cells that express FcyRs recognize antibody that is bond to target cells such as diseased cells including tumor cells and subsequently cause phagocytosis of the target cells such as the diseased cells including tumor cells.
  • ADCP also provides for stimulation of downstream adaptive immune responses by facilitating antigen presentation or by stimulating the secretion of inflammatory mediators.
  • ADCP may be improved in vivo by simultaneous treatment with immunomodulatory agents.
  • the Fc receptor-dependent function of ADCP provides mechanisms for clearance of virus and virus-infected cells, as well as for stimulation of downstream adaptive immune responses by facilitating antigen presentation, or by stimulating the secretion of inflammatory mediators.
  • CDC is yet another cell-killing method that can be directed by antibodies.
  • IgM is the most effective isotype for complement activation.
  • IgGl and IgG3 are also both very effective at directing CDC via the classical complement-activation pathway.
  • the formation of antigen-antibody complexes results in the uncloaking of multiple Clq binding sites in close proximity on the CH2 domains of participating antibody molecules such as IgG molecules (Clq is one of three subcomponents of complement Cl).
  • these uncloaked Clq binding sites convert the previously low-affinity Clq-IgG interaction to one of high avidity, which triggers a cascade of events involving a series of other complement proteins and leads to the proteolytic release of the effector-cell chemotactic/activating agents C3a and C5a.
  • the complement cascade ends in the formation of a membrane attack complex, which creates pores in the cell membrane that facilitate free passage of water and solutes into and out of the cell.
  • Non-labeled murine antibodies are highly immunogenic in man when repetitively applied leading to reduction of the therapeutic effect.
  • the main immunogenicity is mediated by the heavy chain constant regions.
  • the immunogenicity of binding agents derived from murine antibodies in man can be reduced or completely avoided if respective binding agents are chimerized or humanized.
  • Chimeric binding agents are binding agents, the different portions of which are derived from different animal species, such as those having a variable region derived from a murine antibody and a human immunoglobulin constant region. Chimerization of binding agents is achieved by joining of the variable regions of the murine antibody heavy and light chain with the constant region of human heavy and light chain (e.g.
  • chimeric binding agents are generated by joining human kappa-light chain constant region to murine light chain variable region.
  • chimeric binding agents can be generated by joining human lambda-light chain constant region to murine light chain variable region.
  • the preferred heavy chain constant regions for generation of chimeric binding agents are IgGl, IgG3 and IgG4.
  • Other preferred heavy chain constant regions for generation of chimeric binding agents are IgG2, IgA, IgD and IgM.
  • Binding agents such as antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody- antigen interactions, it is possible to express recombinant binding agents, e.g., antibodies, that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann, L. et al. (1998) Nature 332: 323-327; Jones, P. et al.
  • Such framework sequences can be obtained from public DNA databases that include germline antibody gene sequences. These germline sequences will differ from mature antibody gene sequences because they will not include completely assembled variable genes, which are formed by V (D) J joining during B cell maturation. Germline gene sequences will also differ from the sequences of a high affinity secondary repertoire antibody at individual evenly across the variable region.
  • immune checkpoint refers to regulators of the immune system, and, in particular, co-stimulatory and inhibitory signals that regulate the amplitude and quality of T cell receptor recognition of an antigen.
  • the immune checkpoint is an inhibitory signal.
  • the inhibitory signal is the interaction between PD-1 and PD-L1 and/or PD-L2.
  • the inhibitory signal is the interaction between CTLA-4 and CD80 or CD86 to displace CD28 binding.
  • the inhibitory signal is the interaction between LAG-3 and MHC class II molecules.
  • the inhibitory signal is the interaction between TIM-3 and one or more of its ligands, such as galectin 9, PtdSer, HMGB 1 and CEACAM1 .
  • the inhibitory signal is the interaction between one or several KIRs and their ligands.
  • the inhibitory signal is the interaction between TIGIT and one or more of its ligands, PVR, PVRL2 and PVRL3.
  • the inhibitory signal is the interaction between CD94/NKG2A and HLA-E.
  • the inhibitory signal is the interaction between VISTA and its binding partner(s).
  • the inhibitory signal is the interaction between one or more Siglecs and their ligands.
  • the inhibitory signal is the interaction between GARP and one or more of it ligands. In certain embodiments, the inhibitory signal is the interaction between CD47 and SIRPa. In certain embodiments, the inhibitory signal is the interaction between PVRIG and PVRL2. In certain embodiments, the inhibitory signal is the interaction between CSF 1 R and CSF 1. In certain embodiments, the inhibitory signal is the interaction between BTLA and HVEM. In certain embodiments, the inhibitory signal is part of the adenosinergic pathway, e.g., the interaction between A2AR and/or A2BR and adenosine, produced by CD39 and CD73. In certain embodiments, the inhibitory signal is the interaction between B7-H3 and its receptor and/or B7- H4 and its receptor. In certain embodiments, the inhibitory signal is mediated by IDO, CD20, NOX or TDO.
  • the "Programmed Death- 1 (PD-1)" receptor refers to an immuno-inhibitory receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo, and binds to two ligands, PD-L1 (also known as B7-H1 or CD274) and PD-L2 (also known as B7-DC or CD273).
  • PD-1 as used herein includes human PD-1 (hPD-1), variants, isoforms, and species homologs of hPD-1, and analogs having at least one common epitope with hPD-1.
  • P-L1 Programmed Death Ligand-1
  • PD-L1 is one of two cell surface glycoprotein ligands for PD-1 (the other being PD-L2) that downregulates T cell activation and cytokine secretion upon binding to PD-1.
  • the term "PD-L1” as used herein includes human PD-L1 (hPD-Ll), variants, isoforms, and species homologs of hPD-Ll, and analogs having at least one common epitope with hPD-Ll .
  • PD-L2 includes human PD-L2 (hPD-L2), variants, isoforms, and species homologs of hPD-L2, and analogs having at least one common epitope with hPD-L2.
  • the ligands of PD-1 (PD-L1 and PD-L2) are expressed on the surface of antigen-presenting cells, such as dendritic cells or macrophages, and other immune cells. Binding of PD-1 to PD-L1 or PD-L2 results in downregulation of T cell activation. Cancer cells expressing PD-L1 and/or PD-L2 are able to switch off T cells expressing PD-1 what results in suppression of the anticancer immune response.
  • Cytotoxic T Lymphocyte Associated Antigen-4 (CTLA-4) is a T cell surface molecule and is a member of the immunoglobulin superfamily. This protein downregulates the immune system by binding to CD80 (B7-1) and CD86 (B7-2).
  • CTLA-4" as used herein includes human CTLA-4 (hCTLA-4), variants, isoforms, and species homologs of hCTLA- 4, and analogs having at least one common epitope with hCTLA-4.
  • CTLA-4 is a homolog of the stimulatory checkpoint protein CD28 with much higher binding affinity for CD80 and CD86.
  • CTLA4 is expressed on the surface of activated T cells and its ligands are expressed on the surface of professional antigen-presenting cells. Binding of CTLA-4 to its ligands prevents the costimulatory signal of CD28 and produces an inhibitory signal. Thus, CTLA-4 downregulates T cell activation.
  • T cell Immunoreceptor with Ig and ITIM domains (TIGIT, also known as WUCAM or Vstm3) is an immune receptor on T cells and Natural Killer (NK) cells and binds to PVR (CD155) on DCs, macrophages etc., and PVRL2 (CD112; nectin-2) and PVRL3 (CD113; nectin-3) and regulates T cell-mediated immunity.
  • TIGIT includes human TIGIT (hTIGIT), variants, isoforms, and species homologs of hTIGIT, and analogs having at least one common epitope with hTIGIT.
  • PVR includes human PVR (hPVR), variants, isoforms, and species homologs of hPVR, and analogs having at least one common epitope with hPVR.
  • PVRL2 includes human PVRL2 (hPVRL2), variants, isoforms, and species homologs of hPVRL2, and analogs having at least one common epitope with hPVRL2.
  • PVRL3 includes human PVRL3 (hPVRL3), variants, isofonns, and species homologs of hPVRL3, and analogs having at least one common epitope with hPVRL3.
  • B7 family refers to inhibitory ligands with undefined receptors.
  • the B7 family encompasses B7-H3 and B7-H4, both upregulated on tumor cells and tumor infiltrating cells.
  • B7- H3 and B7-H4 as used herein include human B7-H3 (hB7-H3) and human B7-H4 (hB7-H4), variants, isofonns, and species homologs thereof, and analogs having at least one common epitope with B7-H3 and B7-H4, respectively.
  • B and T Lymphocyte Attenuator (BTLA, also known as CD272) is a TNFR family member expressed in Thl but not Th2 cells. BTLA expression is induced during activation of T cells and is in particular expressed on surfaces of CD8+ T cells.
  • BTLA as used herein includes human BTLA (hBTLA), variants, isoforms, and species homologs of hBTLA, and analogs having at least one common epitope with hBTLA.
  • BTLA expression is gradually downregulated during differentiation of human CD8+ T cells to effector cell phenotype. Tumor-specific human CD8+ T cells express high levels of BTLA.
  • HVEM Herpesvirus entry mediator
  • TNFRSF14 TNFRSF14 or CD270
  • HVEM human HVEM
  • variants variants
  • isoforms and species homologs of hHVEM
  • analogs having at least one common epitope with hHVEM.
  • BTLA-HVEM complexes negatively regulate T cell immune responses.
  • KIRs KIRs are receptors for MHC Class I molecules on NK T cells and NK cells that are involved in differentiation between healthy and diseased cells. KIRs bind to human leukocyte antigen (HLA) A, B and C, what suppresses normal immune cell activation.
  • HLA human leukocyte antigen
  • KIRs as used herein includes human KIRs (hKIRs), variants, isoforms, and species homologs of hKIRs, and analogs having at least one common epitope with a hKIR.
  • HLA as used herein includes variants, isoforms, and species homologs of HLA, and analogs having at least one common epitope with a HLA.
  • KIR as used herein in particular refers to KIR2DL1, KIR2DL2, and/or KIR2DL3.
  • LAG-3 Lymphocyte Activation Gene-3 (LAG-3) (also known as CD223) is an inhibitory receptor associated with inhibition of lymphocyte activity by binding to MHC class II molecules. This receptor enhances the function of Treg cells and inhibits CD8+ effector T cell function leading to immune response suppression. LAG-3 is expressed on activated T cells, NK cells, B cells and DCs.
  • LAG-3 as used herein includes human LAG-3 (hLAG-3), variants, isoforms, and species homologs of hLAG-3, and analogs having at least one common epitope.
  • T Cell Membrane Protein-3 (TIM-3) (also known as HAVcr-2) is an inhibitory receptor involved in the inhibition of lymphocyte activity by inhibition of Thl cell responses. Its ligand is galectin 9 (GAL9), which is upregulated in various types of cancers. Other TIM-3 ligands include phosphatidyl serine (PtdSer), High Mobility Group Protein 1 (HMGB1) and Carcinoembryonic Antigen Related Cell Adhesion Molecule 1 (CEACAM1).
  • PtdSer phosphatidyl serine
  • HMGB1 High Mobility Group Protein 1
  • CEACAM1 Carcinoembryonic Antigen Related Cell Adhesion Molecule 1
  • TIM-3 as used herein includes human TIM3 (hTIM-3), variants, isoforms, and species homologs ofhTIM-3, and analogs having at least one common epitope.
  • GAL9 as used herein includes human GAL9 (hGAL9), variants, isoforms, and species homologs of hGAL9, and analogs having at least one common epitope.
  • PdtSer as used herein includes variants and analogs having at least one common epitope.
  • HMGB1 as used herein includes human HMGB1 (hHMGBl), variants, isoforms, and species homologs of hHMGBl, and analogs having at least one common epitope.
  • CEACAM1 as used herein includes human CEACAM1 (hCEACAMl), variants, isoforms, and species homologs of hCEACAMl, and analogs having at least one common epitope.
  • CD94/NKG2A is an inhibitory receptor predominantly expressed on the surface of natural killer cells and of CD8+ T cells.
  • the term "CD94/NKG2A” as used herein includes human CD94/NKG2A (hCD94/NKG2A), variants, isoforms, and species homologs of hCD94/NKG2A, and analogs having at least one common epitope.
  • the CD94/NKG2A receptor is a heterodimer comprising CD94 and NKG2A. It suppresses NK cell activation and CD8+ T cell function, probably by binding to ligands such as HLA-E.
  • CD94/NKG2A restricts cytokine release and cytotoxic response of natural killer cells (NK cells), Natural Killer T cells (NK-T cells) and T cells (a/p and y/8). NKG2A is frequently expressed in tumor infiltrating cells and HLA-E is overexpressed in several cancers.
  • IDO Indoleamine 2,3-dioxygenase
  • IDO is a tryptophan catabolic enzyme with immune-inhibitory properties.
  • the term "IDO” as used herein includes human IDO (hIDO), variants, isoforms, and species homologs of hIDO, and analogs having at least one common epitope.
  • IDO is the rate limiting enzyme in tryptophan degradation catalyzing its conversion to kynurenine. Therefore, IDO is involved in depletion of essential amino acids. It is known to be involved in suppression of T and NK cells, generation and activation of Tregs and myeloid-derived suppressor cells, and promotion of tumor angiogenesis. IDO is overexpressed in many cancers and was shown to promote immune system escape of tumor cells and to facilitate chronic tumor progression when induced by local inflammation.
  • ATP is converted to adenosine by the ectonucleotidases CD39 and CD73 resulting in inhibitory signaling through adenosine binding by one or more of the inhibitory adenosine receptors "Adenosine A2A Receptor" (A2AR, also known as ADORA2A) and “Adenosine A2B Receptor” (A2BR, also known as ADORA2B).
  • Adenosine is a nucleoside with immunosuppressive properties and is present in high concentrations in the tumor microenvironment restricting immune cell infiltration, cytotoxicity and cytokine production.
  • adenosine signaling is a strategy of cancer cells to avoid host immune system clearance.
  • Adenosine signaling through A2AR and A2BR is an important checkpoint in cancer therapy that is activated by high adenosine concentrations typically present in the tumor microenvironment.
  • CD39, CD73, A2AR and A2BR are expressed by most immune cells, including T cells, invariant natural killer cells, B cells, platelets, mast cells and eosinophils.
  • Adenosine signaling through A2AR and A2BR counteracts T cell receptor mediated activation of immune cells and results in increased numbers of Tregs and decreased activation of DCs and effector T cells.
  • CD39 as used herein includes human CD39 (11CD39), variants, isoforms, and species homologs of hCD39, and analogs having at least one common epitope.
  • CD73 as used herein includes human CD73 (hCD73), variants, isoforms, and species homologs of hCD73, and analogs having at least one common epitope.
  • A2AR as used herein includes human A2AR (hA2AR), variants, isoforms, and species homologs of hA2AR, and analogs having at least one common epitope.
  • A2BR as used herein includes human A2BR (hA2BR), variants, isoforms, and species homologs of hA2BR, and analogs having at least one common epitope.
  • V-domain Ig suppressor of T cell activation (VISTA, also known as C10orf54) bears homology to PD-L1 but displays a unique expression pattern restricted to the hematopoietic compartment.
  • VISTA includes human VISTA (h VISTA), variants, isoforms, and species homologs of hVISTA, and analogs having at least one common epitope. VISTA induces T cell suppression and is expressed by leukocytes within tumors.
  • the "Sialic acid binding immunoglobulin type lectin” (Siglec) family members recognize sialic acids and are involved in distinction between “self and “non-self” .
  • the term “Siglecs” as used herein includes human Siglecs (hSiglecs), variants, isoforms, and species homologs of hSiglecs, and analogs having at least one common epitope with one or more hSiglecs.
  • the human genome contains 14 Siglecs of which several are involved in immunosuppression, including, without limitation, Siglec-2, Siglec-3, Siglec-7 and Siglec-9.
  • Siglec receptors bind glycans containing sialic acid, but differ in their recognition of the linkage regiochemistry and spatial distribution of sialic residues. The members of the family also have distinct expression patterns. A broad range of malignancies overexpress one or more Siglecs.
  • CD20 is an antigen expressed on the surface of B and T cells. High expression of CD20 can be found in cancers, such as B cell lymphomas, hairy cell leukemia, B cell chronic lymphocytic leukemia, and melanoma cancer stem cells.
  • the term "CD20” as used herein includes human CD20 (hCD20), variants, isoforms, and species homologs of hCD20, and analogs having at least one common epitope.
  • GARP Glycoprotein A repetitions predominant
  • hGARP human GARP
  • variants isoforms
  • species homologs of hGARP and analogs having at least one common epitope.
  • GARP is expressed on lymphocytes including Treg cells in peripheral blood and tumor infiltrating T cells at tumor sites. It probably binds to latent "transforming growth factor P” (TGF-p). Disruption of GARP signaling in Tregs results in decreased tolerance and inhibits migration of Tregs to the gut and increased proliferation of cytotoxic T cells.
  • CD47 is a transmembrane protein that binds to the ligand “signal-regulatory protein alpha” (SIRPa).
  • SIRPa signal-regulatory protein alpha
  • CD47 signaling is involved in a range of cellular processes including apoptosis, proliferation, adhesion and migration.
  • CD47 is overexpressed in many cancers and functions as "don’t eat me” signal to macrophages. Blocking CD47 signaling through inhibitory anti-CD47 or anti-SIRPa antibodies enables macrophage phagocytosis of cancer cells and fosters the activation of cancer-specific T lymphocytes.
  • PVRIG Polyovirus receptor related immunoglobulin domain containing
  • CD112R Polypeptide-binds to "Poliovirus receptor-related 2"
  • PVRIG and PVRL2 are overexpressed in a number of cancers. PVRIG expression also induces TIGIT and PD- 1 expression and PVRL2 and PVR (a TIGIT ligand) are co-overexpressed in several cancers. Blockade of the PVRIG signaling pathway results in increased T cell function and CD8+ T cell responses and, therefore, reduced immune suppression and elevated interferon responses.
  • PVRIG includes human PVRIG (hPVRIG), variants, isoforms, and species homologs of hPVRIG, and analogs having at least one common epitope with hPVRIG.
  • PVRL2 as used herein includes hPVRL2, as defined above.
  • CSF1R is a myeloid growth factor receptor that binds CSF1. Blockade of the CSF1R signaling can functionally reprogram macrophage responses, thereby enhancing antigen presentation and anti-tumor T cell responses.
  • CSF1R as used herein includes human CSF1R (hCSFIR), variants, isoforms, and species homologs of hCSFIR, and analogs having at least one common epitope with hCSFIR.
  • CSF1 as used herein includes human CSF1 (hCSFl), variants, isoforms, and species homologs of hCSFl, and analogs having at least one common epitope with hCSFl .
  • NADPH oxidase refers to an enzyme of the NOX family of enzymes of myeloid cells that generate immunosuppressive reactive oxygen species (ROS).
  • NOX reactive oxygen species
  • NOX as used herein includes human NOX (hNOX), variants, isoforms, and species homologs of hNOX, and analogs having at least one common epitope with hNOX.
  • TDO tumor-derived antigenase
  • TDO represents an alternative route to IDO in tryptophan degradation and is involved in immune suppression. Since tumor cells may catabolize tryptophan via TDO instead of IDO, TDO may represent an additional target for checkpoint blockade. Indeed, several cancer cell lines have been found to upregulate TDO and TDO may complement IDO inhibition.
  • TDO includes human TDO (hTDO), variants, isoforms, and species homologs of hTDO, and analogs having at least one common epitope with hTDO.
  • immune checkpoint proteins mediate immune checkpoint signaling.
  • checkpoint proteins directly or indirectly regulate T cell activation, T cell proliferation and/or T cell function. Cancer cells often exploit these checkpoint pathways to protect themselves from being attacked by the immune system.
  • the function of checkpoint proteins, which is modulated according to the present disclosure is typically the regulation of T cell activation, T cell proliferation and/or T cell function. Immune checkpoint proteins thus regulate and maintain self-tolerance and the duration and amplitude of physiological immune responses.
  • immune checkpoint proteins belong to the B7:CD28 family or to the tumor necrosis factor receptor (TNFR) super family and, by binding to specific ligands, activate signaling molecules that are recruited to the cytoplasmic domain (Suzuki et al., 2016, Jap J Clin One, 46:191-203).
  • the term "immune checkpoint modulator” or “checkpoint modulator” refers to a molecule or to a compound that modulates the function of one or more checkpoint proteins. Immune checkpoint modulators are typically able to modulate self-tolerance and/or the amplitude and/or the duration of the immune response. Preferably, the immune checkpoint modulator used according to the present disclosure modulates the function of one or more human checkpoint proteins and is, thus, a "human checkpoint modulator”. In a preferred embodiment, the human checkpoint modulator as used herein is an immune checkpoint inhibitor.
  • immune checkpoint inhibitor refers to a molecule that totally or partially reduces, inhibits, interferes with or negatively modulates one or more checkpoint proteins or that totally or partially reduces, inhibits, interferes with or negatively modulates expression of one or more checkpoint proteins.
  • the immune checkpoint inhibitor binds to one or more checkpoint proteins.
  • the immune checkpoint inhibitor binds to one or more molecules regulating checkpoint proteins.
  • the immune checkpoint inhibitor binds to precursors of one or more checkpoint proteins e.g., on DNA- or RNA-level. Any agent that functions as a checkpoint inhibitor according to the present disclosure can be used.
  • the term “partially” as used herein means at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% in the level, e.g., in the level of inhibition of a checkpoint protein.
  • the immune checkpoint inhibitor suitable for use herein is an antagonist of inhibitory signals, e.g., an antibody which targets, for example, PD-1, PD-L1, CTLA-4, LAG- 3, B7-H3, B7-H4, or TIM-3.
  • inhibitory signals e.g., an antibody which targets, for example, PD-1, PD-L1, CTLA-4, LAG- 3, B7-H3, B7-H4, or TIM-3.
  • the immune checkpoint inhibitor prevents inhibitory signals associated with the immune checkpoint.
  • the immune checkpoint inhibitor is an antibody, or fragment thereof that disrupts inhibitory signaling associated with the immune checkpoint.
  • the immune checkpoint inhibitor is a small molecule inhibitor that disrupts inhibitory signaling.
  • the immune checkpoint inhibitor is a peptide-based inhibitor that disrupts inhibitory signaling.
  • the immune checkpoint inhibitor is an inhibitory nucleic acid molecule that disrupts inhibitory signaling.
  • the immune checkpoint inhibitor is an antibody, fragment thereof, or antibody mimic, that prevents the interaction between checkpoint blocker proteins, e.g., an antibody, or fragment thereof that prevents the interaction between PD-1 and PD-L1 or PD-L2.
  • the immune checkpoint inhibitor is an antibody, fragment thereof, or antibody mimic, that prevents the interaction between CTLA-4 and CD80 or CD86.
  • the immune checkpoint inhibitor is an antibody, fragment thereof, or antibody mimic, that prevents the interaction between LAG-3 and its ligands, or TIM-3 and its ligands.
  • the immune checkpoint inhibitor prevents inhibitory signaling through CD39 and/or CD73 and/or the interaction of A2AR and/or A2BR with adenosine. In certain embodiments, the immune checkpoint inhibitor prevents interaction of B7-H3 with its receptor and/or of B7-H4 with its receptor. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of BTLA with its ligand HVEM. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of one or more KIRs with their respective ligands. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of LAG-3 with one or more of its ligands.
  • the immune checkpoint inhibitor prevents the interaction of TIM-3 with one or more of its ligands Galectin-9, PtdSer, HMGB1 and CEACAM1. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of TIGIT with one or more of its ligands PVR, PVRL2 and PVRL3. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of CD94/NKG2A with HLA-E. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of VISTA with one or more of its binding partners. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of one or more Siglecs and their respective ligands. In certain embodiments, the immune checkpoint inhibitor prevents CD20 signaling.
  • the immune checkpoint inhibitor prevents the interaction of GARP with one or more of its ligands. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of CD47 with SIRPa. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of PVRIG with PVRL2. In certain embodiments, the immune checkpoint inhibitor prevents the interaction of CSF1R with CSF1. In certain embodiments, the immune checkpoint inhibitor prevents NOX signaling. In certain embodiments, the immune checkpoint inhibitor prevents IDO and/or TDO signaling.
  • Inhibiting or blocking of inhibitory immune checkpoint signaling results in preventing or reversing immune- suppression and establishment or enhancement of T cell immunity against cancer cells.
  • inhibition of immune checkpoint signaling reduces or inhibits dysfunction of the immune system.
  • inhibition of immune checkpoint signaling renders dysfunctional immune cells less dysfunctional.
  • inhibition of immune checkpoint signaling renders a dysfunctional T cell less dysfunctional.
  • Dysfunction refers to a state of reduced immune responsiveness to antigenic stimulation.
  • the term includes the common elements of both exhaustion and/or anergy in which antigen recognition may occur, but the ensuing immune response is ineffective to control infection or tumor growth.
  • Dysfunction also includes a state in which antigen recognition is retarded due to dysfunctional immune cells.
  • Dysfunctional refers to an immune cell that is in a state of reduced immune responsiveness to antigen stimulation. Dysfunctional includes unresponsive to antigen recognition and impaired capacity to translate antigen recognition into downstream T cell effector functions, such as proliferation, cytokine production (e.g., IL-2) and/or target cell killing.
  • T cell effector functions such as proliferation, cytokine production (e.g., IL-2) and/or target cell killing.
  • T cell anergy refers to the state of unresponsiveness to antigen stimulation resulting from incomplete or insufficient signals delivered through the T cell receptor (TCR). T cell anergy can also result upon stimulation with antigen in the absence of co-stimulation, resulting in the cell becoming refractory to subsequent activation by the antigen even in the context of costimulation. The unresponsive state can often be overridden by the presence of IL-2. Anergic T cells do not undergo clonal expansion and/or acquire effector functions.
  • exhaust refers to immune cell exhaustion, such as T cell exhaustion as a state of T cell dysfunction that arises from sustained TCR signaling that occurs during many chronic infections and cancer. It is distinguished from anergy in that it arises not through incomplete or deficient signaling, but from sustained signaling. Exhaustion is defined by poor effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T cells. Exhaustion prevents optimal control of diseases (e.g., infection and tumors). Exhaustion can result from both extrinsic negative regulatory pathways (e.g., immunoregulatory cytokines) as well as cell intrinsic negative regulatory pathways (inhibitory immune checkpoint pathways, such as described herein).
  • extrinsic negative regulatory pathways e.g., immunoregulatory cytokines
  • cell intrinsic negative regulatory pathways inhibitory immune checkpoint pathways, such as described herein.
  • Enhancing T cell function means to induce, cause or stimulate a T cell to have a sustained or amplified biological function, or renew or reactivate exhausted or inactive T cells.
  • enhancing T cell function include increased secretion of y-interferon from CD8+ T cells, increased proliferation, increased antigen responsiveness (e.g., tumor clearance) relative to such levels before the intervention.
  • the level of enhancement is as least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 110%, 120%, 130%, 140%, 150%, 200%, or more. Manners of measuring this enhancement are known to one of ordinary skill in the art.
  • the immune checkpoint inhibitor may be an inhibitory nucleic acid molecule.
  • inhibitory nucleic acid or “inhibitory nucleic acid molecule” as used herein refers to a nucleic acid molecule, e.g., DNA or RNA, that totally or partially reduces, inhibits, interferes with or negatively modulates one or more checkpoint proteins.
  • Inhibitory nucleic acid molecules include, without limitation, oligonucleotides, siRNA, shRNA, antisense DNA or RNA molecules, and aptamers (e.g., DNA or RNA aptamers).
  • oligonucleotide refers to a nucleic acid molecule that is able to decrease protein expression, in particular expression of a checkpoint protein, such as the checkpoint proteins described herein.
  • Oligonucleotides are short DNA or RNA molecules, typically comprising from 2 to 50 nucleotides. Oligonucleotides maybe single-stranded or double-stranded.
  • a checkpoint inhibitor oligonucleotide may be an antisense-oligonucleotide.
  • Antisense-oligonucleotides are single-stranded DNA or RNA molecules that are complementary to a given sequence, in particular to a sequence of the nucleic acid sequence (or a fragment thereof) of a checkpoint protein.
  • Antisense RNA is typically used to prevent protein translation of mRNA, e.g., of mRNA encoding a checkpoint protein, by binding to said mRNA.
  • Antisense DNA is typically used to target a specific, complementary (coding or non-coding) RNA. If binding takes place, such a DNA/RNA hybrid can be degraded by the enzyme RNase H.
  • morpholino antisense oligonucleotides can be used for gene knockdowns in vertebrates.
  • Kryczek et al., 2006 (J Exp Med, 203:871-81) designed B7-H4-specific morpholines that specifically blocked B7-H4 expression in macrophages, resulting in increased T cell proliferation and reduced tumor volumes in mice with tumor associated antigen (TAA)-specific T cells.
  • TAA tumor associated antigen
  • siRNA or "small interfering RNA” or “small inhibitory RNA” are used interchangeably herein and refer to a double-stranded RNA molecule with a typical length of 20- 25 base pairs that interferes with expression of a specific gene, such as a gene coding for a checkpoint protein, with a complementary nucleotide sequence.
  • siRNA interferes with mRNA therefore blocking translation, e.g., translation of an immune checkpoint protein.
  • Transfection of exogenous siRNA may be used for gene knockdown, however, the effect maybe only transient, especially in rapidly dividing cells. Stable transfection may be achieved, e.g., by RNA modification or by using an expression vector.
  • siRNA sequences may also be modified to introduce a short loop between the two strands resulting in a "small hairpin RNA” or “shRNA".
  • shRNA can be processed into a functional siRNA by Dicer.
  • shRNA has a relatively low rate of degradation and turnover. Accordingly, the immune checkpoint inhibitor may be a shRNA.
  • aptamer refers to a single-stranded nucleic acid molecule, such as DNA or RNA, typically in a length of 25-70 nucleotides that is capable of binding to a target molecule, such as a polypeptide.
  • the aptamer binds to an immune checkpoint protein such as the immune checkpoint proteins described herein.
  • an aptamer according to the disclosure can specifically bind to an immune checkpoint protein or polypeptide, or to a molecule in a signaling pathway that modulates the expression of an immune checkpoint protein or polypeptide.
  • the generation and therapeutic use of aptamers is well known in the art (see, e.g., US 5,475,096).
  • small molecule inhibitor or “small molecule” are used interchangeably herein and refer to a low molecular weight organic compound, usually up to 1000 daltons, that totally or partially reduces, inhibits, interferes with, or negatively modulates one or more checkpoint proteins as described above.
  • small molecular inhibitors are usually synthesized by organic chemistry, but may also be isolated from natural sources, such as plants, fungi, and microbes.
  • the small molecular weight allows a small molecule inhibitor to rapidly diffuse across cell membranes.
  • various A2AR antagonists known in the art are organic compounds having a molecular weight below 500 daltons.
  • the immune checkpoint inhibitor may be an antibody, an antigen-binding fragment thereof, an antibody mimic or a fusion protein comprising an antibody portion with an antigen-binding fragment of the required specificity.
  • Antibodies or antigen-binding fragments thereof are as described herein.
  • Antibodies or antigen-binding fragments thereof that are immune checkpoint inhibitors include in particular antibodies or antigen-binding fragments thereof that bind to immune checkpoint proteins, such as immune checkpoint receptors or immune checkpoint receptor ligands.
  • Antibodies or antigen-binding fragments may also be conjugated to further moieties, as described herein.
  • antibodies or antigen-binding fragments thereof are chimerized, humanized or human antibodies.
  • immune checkpoint inhibitor antibodies or antigenbinding fragments thereof are antagonists of immune checkpoint receptors or of immune checkpoint receptor ligands.
  • an antibody that is an immune checkpoint inhibitor is an isolated antibody.
  • the antibody that is an immune checkpoint inhibitor or the antigen-binding fragment thereof according to the present disclosure may also be an antibody that cross-competes for antigen binding with any known immune checkpoint inhibitor antibody.
  • an immune checkpoint inhibitor antibody cross-competes with one or more of the immune checkpoint inhibitor antibodies described herein. The ability of antibodies to cross-compete for binding to an antigen indicates that these antibodies may bind to the same epitope region of the antigen or when binding to another epitope sterically hinder the binding of known immune checkpoint inhibitor antibodies to that particular epitope region.
  • cross-competing antibodies may have functional properties very similar to those they are cross-competing with as they are expected to block binding of the immune checkpoint to its ligand either by binding to the same epitope or by sterically hindering the binding of the ligand.
  • Cross-competing antibodies can be readily identified based on their ability to cross-compete with one or more of known antibodies in standard binding assays such as Surface Plasmon Resoncance analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • antibodies or antigen binding fragments thereof that cross-compete for binding to a given antigen with, or bind to the same epitope region of a given antigen as, one or more known antibodies are monoclonal antibodies.
  • these cross-competing antibodies can be chimeric antibodies, or humanized or human antibodies.
  • Such chimeric, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
  • the checkpoint inhibitor may also be in the form of the soluble form of the molecules (or variants thereof) themselves, e.g., a soluble PD-L1 or PD-L1 fusion.
  • more than one checkpoint inhibitor can be used, wherein the more than one checkpoint inhibitors are targeting distinct checkpoint pathways or the same checkpoint pathway.
  • the more than one checkpoint inhibitors are distinct checkpoint inhibitors.
  • more than one distinct checkpoint inhibitor in particular at least 2, 3, 4, 5, 6, 7, 8, 9 or 10 distinct checkpoint inhibitors are used, preferably 2, 3, 4 or 5 distinct checkpoint inhibitors are used, more preferably 2, 3 or 4 distinct checkpoint inhibitors are used, even more preferably 2 or 3 distinct checkpoint inhibitors are used and most preferably 2 distinct checkpoint inhibitors are used.
  • Preferred examples of combinations of distinct checkpoint inhibitors include combination of an inhibitor of PD-1 signaling and an inhibitor of CTLA-4 signaling, an inhibitor of PD-1 signaling and an inhibitor of TIGIT signaling, an inhibitor of PD-1 signaling and an inhibitor of B7-H3 and/or B7-H4 signaling, an inhibitor of PD-1 signaling and an inhibitor of BTLA signaling, an inhibitor of PD-1 signaling and an inhibitor of KIR signaling, an inhibitor of PD-1 signaling and an inhibitor of LAG-3 signaling, an inhibitor of PD-1 signaling and an inhibitor of TIM-3 signaling, an inhibitor of PD-1 signaling and an inhibitor of CD94/NKG2A signaling, an inhibitor of PD-1 signaling and an inhibitor of IDO signaling, an inhibitor of PD-1 signaling and an inhibitor of adenosine signaling, an inhibitor of PD-1 signaling and an inhibitor of VISTA signaling, an inhibitor of PD-1 signaling and an inhibitor of Siglec signaling, an inhibitor of PD-1 signaling and
  • the inhibitory immunoregulator is a component of the PD-1/PD-L1 or PD-1/PD-L2 signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the PD-1 signaling pathway.
  • the checkpoint inhibitor of the PD-1 signaling pathway is a PD-1 inhibitor.
  • the checkpoint inhibitor of the PD-1 signaling pathway is a PD-1 ligand inhibitor, such as a PD-L1 inhibitor or a PD-L2 inhibitor.
  • the checkpoint inhibitor of the PD-1 signaling pathway is an antibody or an antigen-binding portion thereof that disrupts the interaction between the PD-1 receptor and one or more of its ligands, PD-L1 and/or PD-L2.
  • Antibodies which bind to PD-1 and disrupt the interaction between PD-1 and one or more of its ligands are known in the art.
  • the antibody or antigen-binding portion thereof binds specifically to PD- 1.
  • the antibody or antigen-binding portion thereof binds specifically to PD-L1 and inhibits its interaction with PD-1 , thereby increasing immune activity.
  • the antibody or antigen-binding portion thereof binds specifically to PD-L2 and inhibits its interaction with PD- 1 , thereby increasing immune activity.
  • the inhibitory immunoregulator is a component of the CTLA-4 signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the CTLA-4 signaling pathway. In certain embodiments, the checkpoint inhibitor of the CTLA-4 signaling pathway is a CTLA-4 inhibitor. In certain embodiments, the checkpoint inhibitor of the CTLA-4 signaling pathway is a CTLA-4 ligand inhibitor.
  • the inhibitory immunoregulator is a component of the TIGIT signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the TIGIT signaling pathway.
  • the checkpoint inhibitor of the TIGIT signaling pathway is a TIGIT inhibitor. In certain embodiments, the checkpoint inhibitor of the TIGIT signaling pathway is a TIGIT ligand inhibitor.
  • the inhibitory immunoregulator is a component of the B7 family signaling pathway.
  • the B7 family members are B7-H3 and B7-H4.
  • Certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of B7- H3 and/or B7-4. Accordingly, certain embodiments of the disclosure provide for administering to a subject an antibody or an antigen-binding portion thereof that targets B7-H3 or B7-H4.
  • the B7 family does not have any defined receptors but these ligands are upregulated on tumor cells or tumor-infiltrating cells. Preclinical mouse models have shown that blockade of these ligands can enhance anti-tumor immunity.
  • the inhibitory immunoregulator is a component of the BTLA signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the BTLA signaling pathway.
  • the checkpoint inhibitor of the BTLA signaling pathway is a BTLA inhibitor. In certain embodiments, the checkpoint inhibitor of the BTLA signaling pathway is a HVEM inhibitor.
  • the inhibitory immunoregulator is a component of one or more KIR signaling pathways. Accordingly, certain embodiments of the disclosure provide for administering to a subj ect a checkpoint inhibitor of one or more KIR signaling pathways.
  • the checkpoint inhibitor of one or more KIR signaling pathways is a KIR inhibitor.
  • the checkpoint inhibitor one or more KIR signaling pathways is a KIR ligand inhibitor.
  • the KIR inhibitor according to the present disclosure may be an anti-KIR antibody that binds to KIR2DL1, KIR2DL2, and/or KIR2DL3.
  • the inhibitory immunoregulator is a component of the LAG-3 signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of LAG-3 signaling.
  • the checkpoint inhibitor of the LAG-3 signaling pathway is a LAG-3 inhibitor. In certain embodiments, the checkpoint inhibitor of the LAG-3 signaling pathway is a LAG-3 ligand inhibitor.
  • the inhibitory immunoregulator is a component of the TIM-3 signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the TIM-3 signaling pathway. In certain embodiments, the checkpoint inhibitor of the TIM-3 signaling pathway is a TIM-3 inhibitor. In certain embodiments, the checkpoint inhibitor of the TIM-3 signaling pathway is a TIM-3 ligand inhibitor.
  • the inhibitory immunoregulator is a component of the CD94/NKG2A signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the CD94/NKG2A signaling pathway. In certain embodiments, the checkpoint inhibitor of the CD94/NKG2A signaling pathway is a CD94/NKG2A inhibitor. In certain embodiments, the checkpoint inhibitor of the CD94/NKG2A signaling pathway is a CD94/NKG2A ligand inhibitor.
  • the inhibitory immunoregulator is a component of the IDO signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the IDO signaling pathway, e.g., an IDO inhibitor.
  • the inhibitory immunoregulator is a component of the adenosine signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the adenosine signaling pathway.
  • the checkpoint inhibitor of the adenosine signaling pathway is a CD39 inhibitor.
  • the checkpoint inhibitor of the adenosine signaling pathway is a CD73 inhibitor.
  • the checkpoint inhibitor of the adenosine signaling pathway is an A2AR inhibitor.
  • the checkpoint inhibitor of the adenosine signaling pathway is an A2BR inhibitor.
  • the inhibitory immunoregulator is a component of the VISTA signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the VISTA signaling pathway. In certain embodiments, the checkpoint inhibitor of the VISTA signaling pathway is a VISTA inhibitor.
  • the inhibitory immunoregulator is a component of one or more Siglec signaling pathways. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of one or more Siglec signaling pathways. In certain embodiments, the checkpoint inhibitor of one or more Siglec signaling pathways is a Siglec inhibitor. In certain embodiments, the checkpoint inhibitor of one or more Siglec signaling pathways is a Siglec ligand inhibitor.
  • the inhibitory immunoregulator is a component of the CD20 signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a checkpoint inhibitor of the CD20 signaling pathway is a CD20 inhibitor.
  • the inhibitory immunoregulator is a component of the GARP signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the GARP signaling pathway. In certain embodiments, the checkpoint inhibitor of the GARP signaling pathway is a GARP inhibitor.
  • the inhibitory immunoregulator is a component of the CD47 signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the CD47 signaling pathway.
  • the checkpoint inhibitor of the CD47 signaling pathway is a CD47 inhibitor. In certain embodiments, the checkpoint inhibitor of the CD47 signaling pathway is a SIRPa inhibitor.
  • the inhibitory immunoregulator is a component of the PVRIG signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the PVRIG signaling pathway. In certain embodiments, the checkpoint inhibitor of the PVRIG signaling pathway is a PVRIG inhibitor. In certain embodiments, the checkpoint inhibitor of the PVRIG signaling pathway is a PVRIG ligand inhibitor. In certain embodiments, the inhibitory immunoregulator is a component of the CSF1R signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the CSF1R signaling pathway. In certain embodiments, the checkpoint inhibitor of the CSF1R signaling pathway is a CSF1R inhibitor. In certain embodiments, the checkpoint inhibitor of the CSF1R signaling pathway is a CSF1 inhibitor.
  • the inhibitory immunoregulator is a component of the NOX signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the NOX signaling pathway, e.g., a NOX inhibitor.
  • the inhibitory immunoregulator is a component of the TDO signaling pathway. Accordingly, certain embodiments of the disclosure provide for administering to a subject a checkpoint inhibitor of the TDO signaling pathway, e.g., a TDO inhibitor.
  • Exemplary PD-1 inhibitors include, without limitation, anti-PD-1 antibodies such as BGB-A317 (BeiGene; see US 8,735,553, WO 2015/35606 and US 2015/0079109), cemiplimab (Regeneron; see WO 2015/112800) and lambrolizumab (e.g., disclosed as hPD109A and its humanized derivatives h409Al, h409A16 and h409A17 in WO2008/156712), AB137132 (Abeam), EH12.2H7 and RMP1-14 (#BE0146; Bioxcell Lifesciences Pvt.
  • anti-PD-1 antibodies such as BGB-A317 (BeiGene; see US 8,735,553, WO 2015/35606 and US 2015/0079109), cemiplimab (Regeneron; see WO 2015/112800) and lambrolizumab (e.g., disclosed as hPD109A and its humanized derivatives h
  • JS001 TAIZHOU JUNSHI PHARMA; see Si- Yang Liu et al., 2007, J. Hematol. Oncol. 70: 136
  • AMP- 224 GSK-2661380; cf.
  • STI-1110 Suddeno Therapeutics; see WO 2014/194302), AGEN2034 (Agenus; see WO 2017/040790), MGA012 (Macrogenics; see WO 2017/19846), IBI308 (Innovent; see WO 2017/024465, WO 2017/025016, WO 2017/132825, and WO 2017/133540), anti-PD-1 antibodies as described, e.g., in US 7,488,802, US 8,008,449, US 8,168,757, WO 03/042402, WO 2010/089411 (further disclosing anti-PD-Ll antibodies), WO 2010/036959, WO 2011/159877 (further disclosing antibodies against TIM-3), WO 2011/082400, WO 2011/161699, WO 2009/014708, WO 03/099196, WO 2009/114335, WO 2012/145493 (further disclosing antibodies against PD-
  • the PD-1 inhibitor is nivolumab (OPDIVO; BMS-936558), pembrolizumab (KEYTRUDA; MK-3475), pidilizumab (CT-011), PDR001, MEDI0680 (AMP- 514), TSR-042, REGN2810, JS001, AMP-224 (GSK-2661380), PF-06801591, BGB-A317, Bl 754091, sintilimab (IBI308), or camrelizumab (SHR-1210).
  • Exemplary PD-1 ligand inhibitors are PD-L1 inhibitors and PD-L2 inhibitors and include, without limitation, anti-PD-Ll antibodies such as MEDI4736 (durvalumab; AstraZeneca; see WO 2011/066389), MSB-0010718C (see US 2014/0341917), YW243.55.S70 (see SEQ ID NO: 20 of WO 2010/077634 and US 8,217,149), MIH1 (Asymetrix eBioscience; cf.
  • anti-PD-Ll antibodies such as MEDI4736 (durvalumab; AstraZeneca; see WO 2011/066389), MSB-0010718C (see US 2014/0341917), YW243.55.S70 (see SEQ ID NO: 20 of WO 2010/077634 and US 8,217,149), MIH1 (Asymetrix eBioscience; cf.
  • CTLA-4 inhibitors include, without limitation, the monoclonal antibodies ipilimumab (Yervoy; Bristol Myers Squibb) and tremelimumab (Pfizer/Medlmmune), trevilizumab, AGEN- 1884 (Agenus) and ATOR-1015, the anti-CTLA4 antibodies disclosed in WO 2001/014424, US 2005/0201994, EP 1212422, US 5,811,097, US 5,855,887, US 6,051,227, US 6,682,736, US 6,984,720, WO 01/14424, WO 00/37504, US 2002/0039581, US 2002/086014, WO 98/42752, US 6,207,156, US 5,977,318, US 7,109,003, and US 7,132,281, the dominant negative proteins abatacept (Orencia; see EP 2 855 533 ), which comprises the Fe region of IgG 1 fused to the CTLA-4 ECD, and belatacept (N
  • Exemplary checkpoint inhibitors of the TIGIT signaling pathway include, without limitation, anti- TIGIT antibodies, such as BMS-986207, COM902 (CGEN-15137; Compugen), AB154 (Arcus Biosciences) or etigilimab (OMP-313M32; OncoMed Pharmaceuticals), or the antibodies disclosed in WO2017/059095, in particular "MAB10", US 2018/0185482, WO 2015/009856, and US 2019/0077864.
  • Exemplary checkpoint inhibitors of B7-H3 include, without limitation, the Fc-optimized monoclonal antibody enoblituzumab (MGA271; Macrogenics; see US 2012/0294796) and the anti-B7-H3 antibodies MGD009 (Macrogenics) and pidilizumab (see US 7,332,582).
  • Exemplary B7-H4 inhibitors include, without limitation, antibodies as described in Dangaj et al., 2013 (Cancer Research 73:4820-9) and in Smith et al., 2014 (Gynecol Oncol, 134:181-189), WO 2013/025779 (e.g., 2D1 encoded by SEQ ID NOs: 3 and 4, 2H9 encoded by SEQ ID NO: 37 and 39, and 2E11 encoded by SEQ ID NOs: 41 and 43) and in WO 2013/067492 (e.g., an antibody with an amino acid sequence selected from SEQ ID NOs: 1-8), morpholino antisense oligonucleotides, e.g., as described by Kryczek et al., 2006 (J Exp Med, 203:871-81), or soluble recombinant forms of B7-H4, such as disclosed in US 2012/0177645.
  • WO 2013/025779 e.g., 2
  • Exemplary BTLA inhibitors include, without limitation, the anti-BTLA antibodies described in Crawford and Wherry, 2009 (J Leukocyte Biol 86:5-8), WO 2011/014438 (e.g., 4C7 or an antibody comprising heavy and light chains according to SEQ ID NOs: 8 and 15 and/or SEQ ID NOs: 11 and 18), WO 2014/183885 (e.g., the antibody deposited under the number CNCM 1-4752) and US 2018/155428.
  • WO 2011/014438 e.g., 4C7 or an antibody comprising heavy and light chains according to SEQ ID NOs: 8 and 15 and/or SEQ ID NOs: 11 and 18
  • WO 2014/183885 e.g., the antibody deposited under the number CNCM 1-4752
  • US 2018/155428 e.g., the antibody deposited under the number CNCM 1-4752
  • Checkpoint inhibitors of KIR signaling include, without limitation, the monoclonal antibodies lirilumab (1-7F9; IPH2102; see see US 8,709,411), IPH4102 (Innate Pharma; see Marie-Cardine et al., 2014, Cancer 74(21): 6060-70), anti-KIR antibodies as disclosed, e.g., in US 2018/208652, US 2018/117147, US 2015/344576, WO 2005/003168, WO 2005/009465, WO 2006/072625, WO 2006/072626, WO 2007/042573, WO 2008/084106 (e.g., an antibody comprising heavy and light chains according to SEQ ID NOs: 2 and 3), WO 2010/065939, WO 2012/071411, WO 2012/160448 and WO 2014/055648.
  • WO 2010/065939 WO 2012/071411, WO 2012/160448 and WO 2014/055648.
  • LAG-3 inhibitors include, without limitation, the anti-LAG-3 antibodies BMS-986016 (Bristol-Myers Squibb; see WO 2014/008218 and WO 2015/116539), 25F7 (see US2011/0150892), IMP731 (see WO 2008/132601), H5L7BW (cf.
  • W02014140180 MK-4280 (28G-10; Merck; see WO 2016/028672), REGN3767 (Regneron/Sanofi), BAP050 (see WO 2017/019894), IMP-701 (LAG-525; Novartis) Sym022 (Symphogen), TSR-033 (Tesaro), MGD013 (a bispecific DART antibody targeting LAG-3 and PD-1 developed by MacroGenics), BI754111 (Boehringer Ingelheim), FS118 (a bispecific antibody targeting LAG-3 and PD-1 developed by F-star), GSK2831781 (GSK) and antibodies as disclosed in WO 2009/044273, WO 2008/132601, WO 2015/042246, EP 2 320 940, US 2019/169294, US 2019/169292, WO 2016/028672, WO 2016/126858, WO 2016/200782, WO 2015/200119, WO 2017/220569, WO 2017/087589, WO 2017
  • TIM-3 inhibitors include, without limitation, antibodies targeting TIM-3 such as F38-2E2 (BioLegend), cobolimab (TSR-022; Tesaro), LY3321367 (Eli Lilly), MBG453 (Novartis) and antibodies as disclosed in, e.g., WO 2013/006490, WO 2018/085469 (e.g., antibodies comprising heavy and light chain sequences encoded by nucleic acid sequences according to SEQ ID NOs: 3 and 4), WO 2018/106588, WO 2018/106529 (e.g., an antibody comprising heavy and light chain sequences according to SEQ ID NOs: 8-11).
  • antibodies targeting TIM-3 such as F38-2E2 (BioLegend), cobolimab (TSR-022; Tesaro), LY3321367 (Eli Lilly), MBG453 (Novartis) and antibodies as disclosed in, e.g., WO 2013/006490, WO 2018/08
  • TIM-3 ligand inhibitors include, without limitation, CEACAM1 inhibitors such as the anti- CEACAM1 antibody CM10 (cCAM Biotherapeutics; see WO 2013/054331), antibodies disclosed in WO 2015/075725 (e.g., CM-24, 26H7, 5F4, TEC-11, 12-140-4, 4/3/17, COL-4, F36-54, 34B1, YG-C28F2, D14HD11, M8.7.7, DI 1-AD11 , HEA81, B 1.
  • CEACAM1 inhibitors such as the anti- CEACAM1 antibody CM10 (cCAM Biotherapeutics; see WO 2013/054331), antibodies disclosed in WO 2015/075725 (e.g., CM-24, 26H7, 5F4, TEC-11, 12-140-4, 4/3/17, COL-4, F36-54, 34B1, YG-C28F2, D14HD11, M8.7.7, DI 1-AD11 , HEA81
  • CD94/NKG2A inhibitors include, without limitation, monalizumab (IPH2201; Innate Pharma) and the antibodies and method for their production as disclosed in US 9,422,368 (e.g., humanized Z199; see EP 2 628 753), EP 3 193 929 and WO2016/032334 (e.g., humanized Z270; see EP 2 628 753).
  • IDO inhibitors include, without limitation, exiguamine A, epacadostat (INCB024360; InCyte; see US 9,624,185), indoximod (Newlink Genetics; CAS#: 110117-83-4), NLG919 (Newlink Genetics/Genentech; CAS#: 1402836-58-1), GDC-0919 (Newlink Genetics/Genentech; CAS#: 1402836-58-1), F001287 (Flexus Biosciences/BMS; CAS#: 2221034-29-1), KHK2455 (Cheong et al., 2018, Expert Opin Ther Pat.
  • CD39 inhibitors include, without limitation, A001485 (Arcus Biosciences), PSB 069 (CAS#: 78510-31-3) and the anti-CD39 monoclonal antibody IPH5201 (Innate Pharma; see Perrot et al., 2019, Cell Reports 8:2411-2425.E9).
  • CD73 inhibitors include, without limitation, anti-CD73 antibodies such as CPI-006 (Corvus Pharmaceuticals), MEDI9447 (Medlmmune; see W02016075099), IPH5301 (Innate Pharma; see Perrot et al., 2019, Cell Reports 8:2411-2425.E9), the anti-CD73 antibodies described in WO2018/110555, the small molecule inhibitors PBS 12379 (Tocris Bioscience; CAS#: 1802226- 78-3), A000830, A001190 and A001421 (Arcus Biosciences; see Becker et al., 2018, Cancer Research 78(13 Supplement):3691-3691, doi: 10.1158/1538-7445.AM2018-3691), CB-708 (Calithera Biosciences) and purine cytotoxic nucleoside analogue-based diphosphonates as described by Allard et ah, 2018 (Immunol Rev., 276(1): 121-144).
  • A2AR inhibitors include, without limitation, small molecule inhibitors such as istradefylline (KW- 6002; CAS#: 155270-99-8), PBF-509 (Palobiopharma), ciforadenant (CPI-444: Corvus Pharma/Genentech; CAS#: 1202402-40-1), ST1535 ([2butyl-9-methyl-8-(2H-l,2,3-triazol 2-yl)- 9H-purin-6-xylamine]; CAS#: 496955-42-1), ST4206 (see Stasi et al., 2015, Europ J Pharm 761:353-361; CAS#: 1246018-36-9), tozadenant (SYN115; CAS#: 870070-55-6), V81444 (see WO 2002/055082), preladenant (SCH420814; Merck; CAS#: 377727-87-2), vipadenant (BIIB014; CAS#: 442908
  • A2BR inhibitors include, without limitation, AB928 (a dual A2AR/A2BR small molecule inhibitor; Arcus Biosciences), MRS 1706 (CAS#: 264622-53-9), GS6201 (CAS#: 752222-83-6) and PBS 1115 (CAS#: 152529-79-8).
  • VISTA inhibitors include, without limitation, anti-VISTA antibodies such as JNJ-61610588 (onvatilimab; Janssen Biotech) and the small molecule inhibitor CA-170 (anti-PD-Ll/L2 and anti- VISTA small molecule; CAS#: 1673534-76-3).
  • Siglec inhibitors include, without limitation, the anti-Sigle-7 antibodies disclosed in US 2019/023786 and WO 2018/027203 (e.g., an antibody comprising a variable heavy chain region according to SEQ ID NO: 1 and a variable light chain region according to SEQ ID NO: 15), the anti-Siglec-2 antibody inotuzumab ozogamicin (Besponsa; see US 8,153,768 and US 9,642,918), the anti-Siglec-3 antibody gemtuzumab ozogamicin (Mylotarg; see US 9,359,442) or the anti- Siglec-9 antibodies disclosed in US 2019/062427, US 2019/023786, WO 2019/011855, WO 2019/011852 (e.g., an antibody comprising the CDRs according to SEQ ID NOs: 171-176, or 3 and 4, or 5 and 6, or 7 and 8, or 9 and 10, or 11 and 12, or 13 and 14, or 15 and 16, or 17 and 18, or 19 and 20, or 21 and 22, or
  • CD20 inhibitors include, without limitation, anti-CD20 antibodies such as rituximab (RITUXAN; IDEC-102; IDEC-C2B8; see US 5,843,439), ABP 798 (rituximab biosimilar), ofatumumab (2F2; see W02004/035607), obinutuzumab, ocrelizumab (2h7; see WO 2004/056312), ibritumomab tiuxetan (Zevalin), tositumomab, ublituximab (LFB-R603; LFB Biotechnologies) and the antibodies disclosed in US 2018/0036306 (e.g., an antibody comprising light and heavy chains according to SEQ ID NOs: 1-3 and 4-6, or 7 and 8, or 9 and 10).
  • anti-CD20 antibodies such as rituximab (RITUXAN; IDEC-102; IDEC-C2B8; see US 5,843,43
  • GARP inhibitors include, without limitation, anti-GARP antibodies such as ARGX-115 (arGEN- X) and the antibodies and methods for their production as disclosed in US 2019/127483, US 2019/016811, US 2018/327511, US 2016/251438, EP 3 253 796.
  • anti-GARP antibodies such as ARGX-115 (arGEN- X) and the antibodies and methods for their production as disclosed in US 2019/127483, US 2019/016811, US 2018/327511, US 2016/251438, EP 3 253 796.
  • CD47 inhibitors include, without limitation, anti-CD47 antibodies such as HuF9-G4 (Stanford University/Forty Seven), CC-90002/INBRX-103 (Celgene/Inhibrx), SRF231 (Surface Oncology), IBI188 (Innovent Biologies), AO-176 (Arch Oncology), bispecific antibodies targeting CD47 including TG-1801 (NI-1701; bispecific monoclonal antibody targeting CD47 and CD19; Novimmune/TG Therapeutics) and NI- 1801 (bispecific monoclonal antibody targeting CD47 and mesothelin; Novimmune), and CD47 fusion proteins such as ALX148 (ALX Oncology; see Kauder et al., 2019, PLoS One, doi: 10.1371/joumal.pone.0201832).
  • anti-CD47 antibodies such as HuF9-G4 (Stanford University/Forty Seven), CC-90002/INBRX-103 (Celgene/
  • SIRPa inhibitors include, without limitation, anti-SIRPa antibodies such as OSE-172 (Boehringer Ingelheim/OSE), FSI-189 (Forty Seven), anti-SIRPa fusion proteins such as TTI-621 and TTI-662 (Trillium Therapeutics; see WO 2014/094122).
  • anti-SIRPa antibodies such as OSE-172 (Boehringer Ingelheim/OSE), FSI-189 (Forty Seven), anti-SIRPa fusion proteins such as TTI-621 and TTI-662 (Trillium Therapeutics; see WO 2014/094122).
  • PVRIG inhibitors include, without limitation, anti-PVRIG antibodies such as COM701 (CGEN- 15029) and antibodies and method for their manufacture as disclosed in, e.g., WO 2018/033798 (e.g., CHA.7.518.1H4(S241P), CHA.7.538.1.2.H4(S241P), CPA.9.086H4(S241P),
  • anti-PVRIG antibodies such as COM701 (CGEN- 15029) and antibodies and method for their manufacture as disclosed in, e.g., WO 2018/033798 (e.g., CHA.7.518.1H4(S241P), CHA.7.538.1.2.H4(S241P), CPA.9.086H4(S241P),
  • CSF1R inhibitors include, without limitation, anti-CSFIR antibodies cabiralizumab (FPA008; FivePrime; see WO 2011/140249, WO 2013/169264 and WO 2014/036357), IMC-CS4 (EiiLilly), emactuzumab (R05509554; Roche), RG7155 (WO 2011/70024, WO 2011/107553, WO 2011/131407, WO 2013/87699, WO 2013/119716, WO 2013/132044) and the small molecule inhibitors BLZ945 (CAS#: 953769-46-5) and pexidartinib (PLX3397; Selleckchem; CAS#: 1029044-16-3).
  • anti-CSFIR antibodies cabiralizumab (FPA008; FivePrime; see WO 2011/140249, WO 2013/169264 and WO 2014/036357
  • IMC-CS4 EdiiLilly
  • emactuzumab
  • CSF1 inhibitors include, without limitation, anti-CSFl antibodies disclosed in EP 1 223 980 and Weir et al., 1996 (J Bone Mineral Res 11 : 1474-1481), WO 2014/132072, and antisense DNA and RNA as disclosed in WO 2001/030381.
  • NOX inhibitors include, without limitation, NOXI inhibitors such as the small molecule ML171 (Gianni et al., 2010, ACS Chem Biol 5(10):981 -93, NOS31 (Yamamoto et al., 2018, Biol Pharm Bull.
  • NOX2 inhibitors such as the small molecules ceplene (histamine dihydrochloride; CAS#: 56-92-8), BJ-1301 (Gautam et al., 2017, Mol Cancer Ther 16(10):2144-2156; CAS#: 1287234-48-3) and inhibitors described by Lu et al., 2017, Biochem Pharmacol 143:25-38, NOX4 inhibitors such as the small molecule inhibitors VAS2870 (Altenhofer et al., 2012, Cell Mol Life Sciences 69(14):2327-2343), diphenylene iodonium (CAS#: 244-54-2) and GKT137831 (CAS#: 1218942-37-0; see Tang et al., 2018, 19(10):578- 585).
  • NOX2 inhibitors such as the small molecules ceplene (histamine dihydrochloride; CAS#: 56-92-8), BJ-1301 (Gautam et al., 2017, Mol Cancer Ther
  • TDO inhibitors include, without limitation, 4-(indol-3-yl)-pyrazole derivatives (see US 9,126,984 and US 2016/0263087), 3-indol substituted derivatives (see WO 2015/140717, WO 2017/025868, WO 2016/147144), 3-(indol-3-yl)-pyridine derivatives (see US 2015/0225367 and WO 2015/121812), dual IDO/TDO antagonist, such as small molecule dual IDO/TDO inhibitors disclosed in WO 2015/150097, WO 2015/082499, WO 2016/026772, WO 2016/071283, WO 2016/071293, WO 2017/007700, and the small molecule inhibitor CB548 (Kim, C, et al., 2018, Annals Oncol 29 (suppl_8): viii400-viii441).
  • the immune checkpoint inhibitor is an inhibitor of an inhibitory checkpoint protein but preferably not an inhibitor of a stimulatory checkpoint protein.
  • a number of CTLA-4, PD-1, TIGIT, B7-H3, B7-H4, BTLA, KIR, LAG-3, TIM-3, CD94/NKG2A, IDO, A2AR, A2BR, VISTA, Siglec, CD20, CD39, CD73, GARP, CD47, PVRIG, CSF1R, NOX and TDO inhibitors and inhibitors of respective ligands are known and several of them are already in clinical trials or even approved.
  • alternative immune checkpoint inhibitors may be developed.
  • known inhibitors of the preferred immune checkpoint proteins may be used as such or analogues thereof may be used, in particular chimerized, humanized or human forms of antibodies and antibodies cross-competing with any of the antibodies described herein.
  • immune checkpoint targets can also be targeted by antagonists or antibodies, provided that the targeting results in the stimulation of an immune response such as an anti-tumor immune response as reflected in an increase in T cell proliferation, enhanced T cell activation, and/or increased cytokine production (e.g., IFN-y, IL2).
  • an immune response such as an anti-tumor immune response as reflected in an increase in T cell proliferation, enhanced T cell activation, and/or increased cytokine production (e.g., IFN-y, IL2).
  • Checkpoint inhibitors may be administered in any manner and by any route known in the art. The mode and route of administration will depend on the type of checkpoint inhibitor to be used.
  • Checkpoint inhibitors maybe administered in the form of any suitable pharmaceutical composition as described herein.
  • Checkpoint inhibitors may be administered in the form of nucleic acid, such DNA or RNA molecules, encoding an immune checkpoint inhibitor, e.g., an inhibitory nucleic acid molecule or an antibody or fragment thereof.
  • an immune checkpoint inhibitor e.g., an inhibitory nucleic acid molecule or an antibody or fragment thereof.
  • antibodies can be delivered encoded in expression vectors, as described herein.
  • Nucleic acid molecules can be delivered as such, e.g., in the form of a plasmid or mRNA molecule, or complexed with a delivery vehicle, e.g., a liposome, lipoplex or nucleic-acid lipid particles.
  • Checkpoint inhibitors may also be administered via an oncolytic virus comprising an expression cassette encoding the checkpoint inhibitor.
  • Checkpoint inhibitors may also be administered by administration of endogeneic or allogeneic cells able to express a checkpoint inhibitor, e.g., in the form of a cell based therapy.
  • cell based therapy refers to the transplantation of cells (e.g., T lymphocytes, dendritic cells, or stem cells) expressing an immune checkpoint inhibitor into a subject for the purpose of treating a disease or disorder (e.g., a cancer disease).
  • the cell based therapy comprises genetically engineered cells.
  • the genetically engineered cells express an immune checkpoint inhibitor, such as described herein.
  • the genetically engineered cells express an immune checkpoint inhibitor that is an inhibitory nucleic acid molecule, such as a siRNA, shRNA, an oligonucleotide, antisense DNA or RNA, an aptamer, an antibody or a fragment thereof or a soluble immune checkpoint protein or fusion.
  • Genetically engineered cells may also express further agents that enhance T cell function. Such agents are known in the art.
  • Cell based therapies for the use in inhibition of immune checkpoint signaling are disclosed, e.g., in WO 2018/222711, herein incorporated by reference in its entirety.
  • oncolytic virus refers to a virus capable of selectively replicating in and slowing the growth or inducing the death of a cancerous or hyperproliferative cell, either in vitro or in vivo, while having no or minimal effect on normal cells.
  • An oncolytic virus for the delivery' of an immune checkpoint inhibitor comprises an expression cassette that may encode an immune checkpoint inhibitor that is an inhibitory nucleic acid molecule, such as a siRNA, shRNA, an oligonucleotide, antisense DNA or RNA, an aptamer, an antibody or a fragment thereof or a soluble immune checkpoint protein or fusion.
  • the oncolytic virus preferably is replication competent and the expression cassette is under the control of a viral promoter, e.g., synthetic early/late poxvirus promoter.
  • exemplary oncolytic viruses include vesicular stomatitis virus (VSV), rhabdoviruses (e.g., picomaviruses such as Seneca Valley virus; SVV-001), coxsackievirus, parvovirus, Newcastle disease virus (NDV), herpes simplex virus (HSV; OncoVEX GMCSF), retroviruses (e.g., influenza viruses), measles virus, reovirus, Sinbis virus, vaccinia virus, as exemplarily described in WO 2017/209053 (including Copenhagen, Western Reserve, Wyeth strains), and adenovirus (e.g., Delta-24, Delta-24-RGD, ICOVIR-5, ICOVIR-7, Onyx-015, ColoAdl, H101, AD5/3-D24-GMCSF).
  • oncolytic viruses comprising a soluble form of an immune checkpoint inhibitor and methods for their use are disclosed in WO 2018/022831, herein incorporated by reference in its entirety.
  • Oncolytic viruses can be used as attenuated viruses.
  • the immune checkpoint inhibitor comprises an antibody selected from an anti-PD-1 antibody, an anti-PD-Ll antibody and a combination thereof.
  • the immune checkpoint inhibitor comprises an anti-PD-1 antibody.
  • the anti-PD-1 antibody comprises cemiplimab (LIBTAYO, REGN2810), nivolumab (OPDIVO; BMS-936558), pembrolizumab (KEYTRUDA; MK-3475), pidilizumab (CT-011), spartalizumab (PDR001), MED 10680 (AMP-514), dostarlimab (TSR-042), cetrelimab (JNJ 63723283), toripalimab (JS001), AMP-224 (GSK-2661380), PF-06801591, tislelizumab (BGB-A317), ABBV-181, BI 754091, sintilimab (IBI308), or camrelizumab (SHR-1210).
  • cemiplimab LIBTAYO, REGN2810
  • OPDIVO pembrolizumab
  • KEYTRUDA pembrolizumab
  • MK-3475
  • the immune checkpoint inhibitor comprises an anti-PD-Ll antibody.
  • the anti-PD-Ll antibody comprises atezolizumab (TECENTRIQ; RG7446; MPDL3280A; R05541267), durvalumab (MEDI4736), BMS-936559, avelumab (bavencio), lodapolimab (LY3300054), CX-072 (Proclaim-CX-072), FAZ053, KN035, sugemalimab (CS1001), or MDX-1105.
  • atezolizumab TECENTRIQ; RG7446; MPDL3280A; R05541267
  • durvalumab MEDI4736
  • BMS-936559 avelumab (bavencio)
  • lodapolimab LY3300054
  • CX-072 Proclaim-CX-072
  • FAZ053, KN035, sugemalimab (CS1001), or MDX-1105 or MDX-1105.
  • a bispecific binding agent is administered together, i.e., co-administered, with a checkpoint inhibitor to a subject, e.g., a patient.
  • the checkpoint inhibitor and the bispecific binding agent are administered as a single composition to the subject.
  • the checkpoint inhibitor and the bispecific binding agent are administered concurrently (as separate compositions at the same time) to the subject.
  • the checkpoint inhibitor and the bispecific binding agent are administered separately to the subj ect.
  • the checkpoint inhibitor is administered before the bispecific binding agent to the subject.
  • the checkpoint inhibitor is administered after the bispecific binding agent to the subject.
  • the checkpoint inhibitor and the bispecific binding agent are administered to the subject on the same day.
  • the checkpoint inhibitor and the bispecific binding agent are administered to the subject on different days.
  • medical preparation refers to any product that is intended for a medical use.
  • the term includes pharmaceutical compositions containing one or more active ingredients, as well as arrangements, e.g. kits, of one or more active ingredients, which may be present together or separately, e.g., in separate vials, optionally together with information material concerning, for example, their administration, effect, etc.
  • the compounds and agents described herein may be administered in the form of any suitable pharmaceutical composition.
  • compositions described herein are preferably sterile and contain an effective amount of the compounds and agents described herein and optionally of further agents as discussed herein to generate the desired reaction or the desired effect.
  • compositions are usually provided in a uniform dosage form and may be prepared in a manner known per se.
  • a pharmaceutical composition may e.g. be in the form of a solution or suspension.
  • a pharmaceutical composition may comprise salts, buffer substances, preservatives, carriers, diluents and/or excipients all of which are preferably pharmaceutically acceptable.
  • pharmaceutically acceptable refers to the non-toxicity of a material which does not interact with the action of the active component of the pharmaceutical composition.
  • Salts which are not pharmaceutically acceptable may be used for preparing pharmaceutically acceptable salts and are included in the invention.
  • Pharmaceutically acceptable salts of this kind comprise in a non-limiting way those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic acids, and the like.
  • Pharmaceutically acceptable salts may also be prepared as alkali metal salts or alkaline earth metal salts, such as sodium salts, potassium salts or calcium salts.
  • Suitable buffer substances for use in a pharmaceutical composition include acetic acid in a salt, citric acid in a salt, boric acid in a salt and phosphoric acid in a salt.
  • Suitable preservatives for use in a pharmaceutical composition include benzalkonium chloride, chlorobutanol, paraben and thimerosal.
  • An injectable formulation may comprise a pharmaceutically acceptable excipient such as Ringer Lactate.
  • carrier refers to an organic or inorganic component, of a natural or synthetic nature, in which the active component is combined in order to facilitate, enhance or enable application. According to the invention, the term “carrier” also includes one or more compatible solid or liquid fillers, diluents or encapsulating substances, which are suitable for administration to a patient.
  • Possible carrier substances for parenteral administration are e.g. sterile water, Ringer, Ringer lactate, sterile sodium chloride solution, polyalkylene glycols, hydrogenated naphthalenes and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy- propylene copolymers.
  • excipient when used herein is intended to indicate all substances which may be present in a pharmaceutical composition and which are not active ingredients such as, e.g., carriers, binders, lubricants, thickeners, surface active agents, preservatives, emulsifiers, buffers, flavoring agents, or colorants.
  • agents and compositions described herein may be administered via any conventional route, such as by parenteral administration including by injection or infusion. Administration is intramuscularly.
  • compositions suitable for parenteral administration usually comprise a sterile aqueous or nonaqueous preparation of the active compound, which is preferably isotonic to the blood of the recipient.
  • suitable carriers and solvents are Ringer solution and isotonic sodium chloride solution.
  • sterile, fixed oils are used as solution or suspension medium.
  • an "effective amount” refers to the amount which achieves a desired reaction or a desired effect alone or together with further doses.
  • the desired reaction preferably relates to inhibition of the course of the disease. This comprises slowing down the progress of the disease and, in particular, interrupting or reversing the progress of the disease.
  • the desired reaction in a treatment of a disease or of a condition may also be delay of the onset or a prevention of the onset of said disease or said condition.
  • an effective amount of an agent or composition described herein will depend on the condition to be treated, the severeness of the disease, the individual parameters of the patient, including age, physiological condition, size and weight, the duration of treatment, the type of an accompanying therapy (if present), the specific route of administration and similar factors. Accordingly, the doses administered of the agents described herein may depend on various of such parameters. In the case that a reaction in a patient is insufficient with an initial dose, higher doses (or effectively higher doses achieved by a different, more localized route of administration) may be used.
  • the agents and compositions described herein can be administered to patients, e.g., in vivo, to treat or prevent a variety of disorders such as those described herein.
  • Preferred patients include human patients having disorders that can be corrected or ameliorated by administering the agents and compositions described herein. This includes disorders involving cells characterized by an altered expression pattern of CLDN18.2.
  • agents described herein can be used to treat a patient with a cancer disease, e.g., a cancer disease such as described herein characterized by the presence of cancer cells expressing CLDN18.2.
  • a cancer disease e.g., a cancer disease such as described herein characterized by the presence of cancer cells expressing CLDN18.2.
  • compositions described herein may be administered together with supplementing immunity- enhancing substances such as one or more adjuvants and may comprise one or more immunity-enhancing substances to further increase its effectiveness, preferably to achieve an additive effect of immunostimulation.
  • adjuvant relates to compounds which prolong or enhance or accelerate an immune response. Various mechanisms are possible in this respect, depending on the various types of adjuvants. For example, compounds which allow the maturation of the DC, e.g. lipopolysaccharides or CD40 ligand, form a first class of suitable adjuvants.
  • any agent which influences the immune system of the type of a "danger signal" (LPS, GP96, dsRNA etc.) or cytokines, such as GM-CSF, can be used as an adjuvant which enables an immune response to be intensified and/or influenced in a controlled manner.
  • CpG oligodeoxynucleotides can optionally also be used in this context, although their side effects which occur under certain circumstances, as explained above, are to be considered.
  • Particularly preferred adjuvants are cytokines, such as monokines, lymphokines, interleukins or chemokines, e.g.
  • Further known adjuvants are aluminium hydroxide, Freund's adjuvant or oil such as Montanide®, most preferred Montanide® ISA51.
  • Lipopeptides, such as Pam3Cys are also suitable for use as adjuvants in the pharmaceutical composition of the present invention.
  • agents and compositions provided herein may be used alone or in combination with conventional therapeutic regimens such as surgery, irradiation, chemotherapy and/or bone marrow transplantation (autologous, syngeneic, allogeneic or unrelated).
  • a cancer treatment which utilizes immune- or vaccination-based mechanisms such as the methods and compositions of the present invention may be effectively combined with various other drugs and/or methods targeting similar or other specific mechanisms.
  • those are e.g. combinations with conventional tumor therapies, multi-epitope strategies, additional immunotherapy, and treatment approaches targeting angiogenesis or apoptosis (for review see, e.g., Andersen et al. 2008: Cancer treatment: the combination of vaccination with other therapies.
  • Chemotherapy is the standard of care for multiple types of cancer.
  • the most common chemotherapy agents act by killing cells that divide rapidly, one of the main properties of cancer cells.
  • conventional chemotherapeutic drugs such as e.g. alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents which either affect cell division or DNA synthesis may significantly improve the therapeutic effects of the present invention by clearing suppressor cells, reboot of the immune system, by rendering tumor cells more susceptible to immune mediated killing, or by additional activation of cells of the immune system.
  • chemotherapeutic drugs available which are basically suitable for combination therapies.
  • Some (non-limiting) examples of chemotherapeutic drugs which can be combined with the present invention are carboplatin (Paraplatin), cisplatin (Platinol, Platinol-AQ), cyclophosphamide (Cytoxan, Neosar), docetaxel (Taxotere), doxorubicin (Adriamycin), erlotinib (Tarceva), etoposide (VePesid), gemcitabine (Gemzar), imatinib mesylate (Gleevec), irinotecan (Camptosar), methotrexate (Folex, Mexate, Amethopterin), paclitaxel (Taxol, Abraxane), sorafinib (Nexavar), sunitinib (Sutent), topotecan (
  • Radiation therapy remains an important component of cancer treatment with approximately 50% of all cancer patients receiving radiation therapy during their course of illness.
  • the main goal of radiation therapy is to deprive cancer cells of their multiplication (cell division) potential.
  • the types of radiation used to treat cancer are photons radiation (x-rays and gamma rays) and particle radiations (electron, proton and neutron beams.)
  • Photons radiation x-rays and gamma rays
  • particle radiations electron, proton and neutron beams.
  • External beam radiation is delivered from outside the body by aiming high- energy rays (photons, protons or particle radiation) to the location of the tumor.
  • Internal radiation or brachytherapy is delivered from inside the body by radioactive sources, sealed in catheters or seeds directly into the tumor site.
  • Radiation therapy techniques which are applicable in combination with the present invention are e.g. fractionation (radiation therapy delivered in a fractionated regime, e.g. daily fractions of 1.5 to 3 Gy given over several weeks), 3D conformal radiotherapy (3DCRT; delivering radiation to the gross tumor volume), intensity modulated radiation therapy (IMRT; computer-controlled intensity modulation of multiple radiation beams), image guided radiotherapy (IGRT; a technique comprising pre-radiotherapy imaging which allows for correction), and stereotactic body radiation therapy (SRBT, delivers very high individual doses of radiation over only a few treatment fractions).
  • fractionation radiation therapy delivered in a fractionated regime, e.g. daily fractions of 1.5 to 3 Gy given over several weeks
  • 3DCRT 3D conformal radiotherapy
  • IMRT intensity modulated radiation therapy
  • IGRT image guided radiotherapy
  • SRBT stereotactic body radiation therapy
  • Example 1 Anti-tumor Effect of ASP2138 Monotherapy and Combination Therapy with Anti-mouse PD-1 Antibody in a Human CLDN18.2-Expressing MC38 Tumor-Bearing Human CD3e Knock-in Mouse Model
  • ASP2138 is a bispecific antibody binding to claudin 18.2 (CLDN18.2) and cluster of differentiation 3 (CD3), comprising a set of first, second, third and fourth polypeptide chains according to SEQ ID NOs: 27, 28, 29, and 29, respectively.
  • ASP2138 (Lot No. DSP20804; Astellas Pharma Inc., Tokyo, Japan) was dissolved in phosphate buffered saline (PBS).
  • Anti-mPD-1 antibody (clone RMP1-14) and rat IgG2a isotype antibody (clone 2A3) were purchased from Bio X cell (Lebanon, NH, USA). Both antibodies were diluted in PBS prior to administration.
  • MC38 a mouse colon adenocarcinoma cell line, was obtained from National Cancer Institute (USA).
  • MC38 hCLDN18.2 was established at Astellas Pharma Inc. from MC38 by transduction of human CLDN18.2 and cultured in Dulbecco’s modified eagle’s medium supplemented with 10% heat inactivated fetal bovine serum and 6 pg/mL of blasticidin at 37°C in 5% CO2.
  • mice Five or six-week-old female hCD3s KI mice were purchased from Biocytogen Jiangsu Co., Ltd (Jiangsu, China). This study was approved by the Institutional Animal Care and Use Committee of Astellas Pharma Inc., Tsukuba Research Center, which is accredited by AAALAC International.
  • MC38_hCLDN18.2 cells were suspended in PBS at 4.0 * IO 6 cells/mL and subcutaneously inoculated into the flank of 6 or 7-week-old mice at 2.0 x 10 5 cells/50 pL/mouse and allowed to grow.
  • Tumor volume [mm 3 ] was calculated with the formula: (Length of tumor long axis [mm]) x (Length of tumor short axis [mm]) 2 x 0.5.
  • Percent inhibition of tumor growth was calculated using the following formula:
  • mice received intraperitoneal administration of PBS on days 0 and 7, and intraperitoneal administration of isotype control antibody at 100 pg/head on days 0, 4, 7 and 11.
  • mice received intraperitoneal administration of ASP2138 (0.1 mg/kg) on days 0 and 7, and intraperitoneal administration of isotype control antibody at 100 pg/head on days 0, 4, 7 and 11.
  • Group 3 anti-mPD-1 antibody
  • mice received intraperitoneal administration of PBS on days 0 and 7, and intraperitoneal administration of anti-mPD-1 antibody at 100 pg/head on days 0, 4, 7 and 11.
  • mice received intraperitoneal administration of ASP2138 (0.1 mg/kg) on days 0 and 7, and intraperitoneal administration of anti-mPD-1 antibody at 100 pg/head on days 0, 4, 7 and 11.
  • Tumor diameters were measured with a caliper on days 0, 4, 7, 11 and 14.
  • Tumor volume [mm 3 ] was calculated with the formula: (Length of tumor long axis [mm]) x (Length of tumor short axis [mm]) 2 x 0.5.
  • Mean tumor volumes on day 14 of Group 2 and Group 3 were compared to Group 4 separately using unpaired Student’s t test. P ⁇ 0.05 was considered statistically significant.
  • GraphPad Prism (ver 8.0.2, GraphPad Software, San Diego, CA, USA) was used for data processing.
  • ASP2138 induced significant growth inhibition of MC38_hCLDN18.2 tumors at 0.1, 0.3, 1 and 3 mg/kg by 69%, 77%, 76% and 82% on day!3, respectively, in the hCD3s KI mouse model (Table 2 and Figure 2).
  • administration of ASP2138 or anti-mPD-1 antibody alone inhibited the tumor growth by 47% or 61%, respectively, on day 14.
  • the combination of ASP2138 and anti-mPD-1 antibody inhibited the tumor growth by 83% on day 14.
  • Tumor volume in the combination group was significantly smaller than that in either the ASP2138 group or the anti-mPD-1 antibody group.
  • 3 out of 15 mice in the combination group achieved complete tumor regression (Table 3 and Figure 3).
  • mice were treated with ASP2138 on days 0 and 7 and anti-mPD-1 antibody on days 0, 4, 7 and 11.
  • Anti-mPD-1 antibody anti -mouse PD-1 antibody
  • KPGSGKPGSE VQLVESGGGL VQPGGSLRLS CAASGFTFST YAMNWVRQAP GKGLEWVGRI 180
  • DIVMTQSPDS LAVSLGERAT INCKSSQSLL NSGNQKNYLT WYQQKPGQPP KLLIYWASTR 60 ESGVPDRFTG SGSGTDFTLT ISSLQAEDVA VYYCQNDYSY PFTFGSGTKL EIKRTVAAPS 120 VFIFPPSDEQ LKSGTASWC LLNNFYPREA KVQWKVDNAL QSGNSQESVT EQDSKDSTYS 180 LSSTLTLSKA DYEKHKVYAC EVTHQGLSSP VTKSFNRGEC 220

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne une polythérapie impliquant des agents de liaison bispécifiques comprenant deux domaines de liaison se liant à CLDN18.2 et un domaine de liaison se liant à CD3 ainsi que des inhibiteurs de point de contrôle immunitaire. La polythérapie est efficace dans le traitement du cancer impliquant des cellules cancéreuses exprimant CLDN18.2.
PCT/EP2023/085290 2022-12-14 2023-12-12 Polythérapie impliquant des agents de liaison bispécifiques se liant à cldn18.2 et cd3 et des inhibiteurs de point de contrôle immunitaire WO2024126457A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263387403P 2022-12-14 2022-12-14
US63/387,403 2022-12-14
EP2023061047 2023-04-26
EPPCT/EP2023/061047 2023-04-26

Publications (1)

Publication Number Publication Date
WO2024126457A1 true WO2024126457A1 (fr) 2024-06-20

Family

ID=89222231

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/085290 WO2024126457A1 (fr) 2022-12-14 2023-12-12 Polythérapie impliquant des agents de liaison bispécifiques se liant à cldn18.2 et cd3 et des inhibiteurs de point de contrôle immunitaire

Country Status (1)

Country Link
WO (1) WO2024126457A1 (fr)

Citations (205)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992022653A1 (fr) 1991-06-14 1992-12-23 Genentech, Inc. Procede de production d'anticorps humanises
EP0629240A1 (fr) 1992-02-19 1994-12-21 Scotgen Limited Anticorps modifies, produits et procedes s'y rapportant
US5475096A (en) 1990-06-11 1995-12-12 University Research Corporation Nucleic acid ligands
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO1998042752A1 (fr) 1997-03-21 1998-10-01 Brigham And Women's Hospital Inc. Peptides immunotherapeutiques se liant a ctla-4
US5843439A (en) 1992-11-13 1998-12-01 Anderson; Darrell R. Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5855887A (en) 1995-07-25 1999-01-05 The Regents Of The University Of California Blockade of lymphocyte down-regulation associated with CTLA-4 signaling
US5977318A (en) 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO2000037504A2 (fr) 1998-12-23 2000-06-29 Pfizer Inc. Anticorps monoclonaux humains diriges contre l'antigene ctla-4
WO2001014424A2 (fr) 1999-08-24 2001-03-01 Medarex, Inc. Anticorps contre l'antigene ctla-4 humain et utilisation
WO2001030381A2 (fr) 1999-10-28 2001-05-03 Hofbauer, Reinhold Utilisation d'inhibiteurs csf-1
US20020039581A1 (en) 2000-01-27 2002-04-04 Carreno Beatriz M. Antibodies against CTLA4 and uses therefor
US20020086014A1 (en) 1999-08-24 2002-07-04 Korman Alan J. Human CTLA-4 antibodies and their uses
WO2002055082A1 (fr) 2001-01-10 2002-07-18 Vernalis Research Limited Dérivés pyrazolo[3,4-d]pyrimidiniques et leur utilisation comme antagonistes des récepteurs purinergiques
WO2003042402A2 (fr) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents modulant l'activite de cellules immunes et procedes d'utilisation associes
US20030118592A1 (en) 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
WO2003099196A2 (fr) 2002-05-23 2003-12-04 Cure Tech Ltd. Anticorps monoclonaux humanises immunomodulateurs servant a traiter une maladie neoplasique ou une immunodeficience
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
WO2004035607A2 (fr) 2002-10-17 2004-04-29 Genmab A/S Anticorps monoclonaux humains anti-cd20
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
WO2004056312A2 (fr) 2002-12-16 2004-07-08 Genentech, Inc. Variants d'immunoglobuline et utilisations
US6808710B1 (en) 1999-08-23 2004-10-26 Genetics Institute, Inc. Downmodulating an immune response with multivalent antibodies to PD-1
WO2005003168A2 (fr) 2003-07-02 2005-01-13 Novo Nordisk A/S Compositions et procedes pour la regulation d'activite de cellules nk
WO2005009465A1 (fr) 2003-07-24 2005-02-03 Innate Pharma Methodes et compositions pour augmenter l'efficacite d'anticorps therapeutiques au moyen de composes de potentialisation de cellules nk
WO2006072626A1 (fr) 2005-01-06 2006-07-13 Novo Nordisk A/S Agents de liaison kir et leurs procedes d'utilisation
WO2006072625A2 (fr) 2005-01-06 2006-07-13 Novo Nordisk A/S Procedes et traitements combines anti-kir
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007042573A2 (fr) 2005-10-14 2007-04-19 Innate Pharma Compositions et procedes pour traiter des troubles de proliferation
WO2008084106A1 (fr) 2007-01-11 2008-07-17 Novo Nordisk A/S Anticorps anti-kir, formulations et utilisations de celles-ci
WO2008132601A1 (fr) 2007-04-30 2008-11-06 Immutep Anticorps monoclonal anti-lag-3 cytotoxique et son utilisation dans le traitement ou la prévention d'un rejet du greffon d'organe et de maladies auto-immunes
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
WO2009014708A2 (fr) 2007-07-23 2009-01-29 Cell Genesys, Inc. Anticorps pd-1 en combinaison avec une cellule sécrétant de la cytokine et leurs procédés d'utilisation
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2009044273A2 (fr) 2007-10-05 2009-04-09 Immutep Utilisation d'une protéine lag-3 recombinée ou de dérivés de celle-ci pour produire une réponse immunitaire des monocytes
WO2009101611A1 (fr) 2008-02-11 2009-08-20 Curetech Ltd. Anticorps monoclonaux pour le traitement de tumeurs
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
US7635757B2 (en) 1999-08-23 2009-12-22 Dana-Farber Cancer Institute, Inc. B7-4 Antibodies and uses therefor
US20090317368A1 (en) 1999-11-30 2009-12-24 Lieping Chen B7-h1, a novel immunoregulatory molecule
WO2010012557A1 (fr) 2008-08-01 2010-02-04 Emitec Gesellschaft Für Emissionstechnologie Mbh Procédé de conduite d'une installation de traitement des gaz d'échappement à régulation lambda
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
WO2010036959A2 (fr) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Anticorps anti-pd-1, pd-l1, et pd-l2 humains et leurs utilisations
WO2010065939A1 (fr) 2008-12-05 2010-06-10 Indiana University Research & Technology Corporation Traitement combiné pour améliorer la cytotoxicité induite par les cellules nk
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2011014438A1 (fr) 2009-07-31 2011-02-03 N.V. Organon Anticorps totalement humains dirigés contre le btla
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
EP2320940A2 (fr) 2008-08-11 2011-05-18 Medarex, Inc. Anticorps humains qui se lient au gène 3 d'activation des lymphocytes (lag-3), et leurs utilisations
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011070024A1 (fr) 2009-12-10 2011-06-16 F. Hoffmann-La Roche Ag Anticorps se liant de façon préférentielle au domaine extracellulaire 4 de csf1r humain et leur utilisation
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011107553A1 (fr) 2010-03-05 2011-09-09 F. Hoffmann-La Roche Ag Anticorps dirigés contre le csf-1r humain et utilisations associées
WO2011131407A1 (fr) 2010-03-05 2011-10-27 F. Hoffmann-La Roche Ag Anticorps contre le csf-1r humain et leurs utilisations
WO2011140249A2 (fr) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Anticorps liant csf1r
WO2011159877A2 (fr) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Anticorps di-spécifiques anti-tim-3 et pd-1 pour immunothérapie dans des états pathologiques immuns chroniques
WO2011161699A2 (fr) 2010-06-25 2011-12-29 Aurigene Discovery Technologies Limited Composés modulateurs de l'immunosuppression
US8153768B2 (en) 2002-05-02 2012-04-10 Wyeth Holdings Corporation Calicheamicin derivative-carrier conjugates
WO2012071411A2 (fr) 2010-11-22 2012-05-31 Innate Pharma Sa Traitements modulant les cellules tueuses naturelles et méthodes de traitement d'hémopathies malignes
US20120177645A1 (en) 2009-08-31 2012-07-12 Solomon Langermann Methods and compositions for the inhibition of transplant rejection
WO2012145493A1 (fr) 2011-04-20 2012-10-26 Amplimmune, Inc. Anticorps et autres molécules qui se lient à b7-h1 et à pd-1
US20120294796A1 (en) 2010-03-04 2012-11-22 Macrogenics, Inc. Antibodies Reactive with B7-H3 and Uses Thereof
WO2012160448A2 (fr) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anticorps anti-kir destinés au traitement de troubles inflammatoires
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013019906A1 (fr) 2011-08-01 2013-02-07 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2013025779A1 (fr) 2011-08-15 2013-02-21 Amplimmune, Inc. Anticorps anti-b7-h4 et leurs utilisations
WO2013054331A1 (fr) 2011-10-11 2013-04-18 Tel Hashomer Medical Research Infrastructure And Services Ltd. Anticorps dirigés contre la molécule d'adhésion cellulaire associée à l'antigène carcinoembryonnaire (ceacam)
WO2013067492A1 (fr) 2011-11-03 2013-05-10 The Trustees Of The University Of Pennsylvania Compositions spécifiques de b7-h4 isolé et procédés d'utilisation associés
WO2013087699A1 (fr) 2011-12-15 2013-06-20 F. Hoffmann-La Roche Ag Anticorps contre le csf-1r humain et leurs utilisations
WO2013119716A1 (fr) 2012-02-06 2013-08-15 Genentech, Inc. Compositions et procédés d'utilisation d'inhibiteurs de csf1r
EP2628753A1 (fr) 2008-01-24 2013-08-21 Novo Nordisk A/S Anticorps monoclonal NKG2A antihumain humanisé
WO2013132044A1 (fr) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Thérapie combinée d'anticorps contre le csf -1r humain et ses utilisations
WO2013169264A1 (fr) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Méthodes destinées à traiter des affections avec des anticorps qui se lient au récepteur du facteur 1 de stimulation des colonies (csf1r)
WO2013173223A1 (fr) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Immunothérapie anticancéreuse par rupture de la signalisation pd-1/pd-l1
WO2013181634A2 (fr) 2012-05-31 2013-12-05 Sorrento Therapeutics Inc. Protéines liant un antigène qui lient pd-l1
WO2014008218A1 (fr) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimisation d'anticorps se liant à la protéine lag-3 exprimée par le gène 3 d'activation des lymphocytes, et leurs utilisations
WO2014036357A1 (fr) 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Méthodes de traitement de pathologies par des anticorps qui se lient au récepteur du facteur stimulant les colonies 1 (csf1r)
WO2014055648A1 (fr) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2014094122A1 (fr) 2012-12-17 2014-06-26 Trillium Therapeutics Inc. Traitement de cellules tumorales à cd47+ avec des fusions sirp alpha/fc
WO2014100079A1 (fr) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Anticorps qui se lient au ligand 1 de la mort programmée humaine (pd-l1)
WO2014132072A1 (fr) 2013-02-28 2014-09-04 University Court Of The University Of Edinburgh Thérapie à base de csf1
WO2014140180A1 (fr) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Protéines de liaison anti-lag-3
WO2014159562A1 (fr) 2013-03-14 2014-10-02 Bristol-Myers Squibb Company Combinaison d'agoniste de dr5 et d'antagoniste anti-pd-1 et méthodes d'utilisation associées
WO2014165082A2 (fr) 2013-03-13 2014-10-09 Medimmune, Llc Anticorps et procédés de détection
WO2014179664A2 (fr) 2013-05-02 2014-11-06 Anaptysbio, Inc. Anticorps dirigés contre la protéine de mort programmée 1 (pd-1)
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2014183885A1 (fr) 2013-05-17 2014-11-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antagoniste de l'interaction btla/hvem pour une utilisation en thérapie
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
US20140370013A1 (en) 2013-01-14 2014-12-18 Xencor, Inc. Novel heterodimeric proteins
WO2014207748A1 (fr) 2013-06-27 2014-12-31 Alexander Biro Molécules ctla-4 solubles et leurs dérivés pour le traitement d'une néphrose lipoïdique
WO2015009856A2 (fr) 2013-07-16 2015-01-22 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2015042246A1 (fr) 2013-09-20 2015-03-26 Bristol-Myers Squibb Company Combinaison d'anticorps anti-lag-3 et d'anticorps anti-pd-1 pour traiter des tumeurs
EP2855533A1 (fr) 2013-03-15 2015-04-08 Momenta Pharmaceuticals, Inc. Procédés se rapportant à des protéines de fusion fc-ctla4
WO2015061668A1 (fr) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux anti-pd-l1 et fragments de ceux-ci
WO2015075725A1 (fr) 2013-11-25 2015-05-28 Ccam Biotherapeutics Ltd. Compositions comprenant des anticorps anti-ceacam1 et anti-pd pour la cancérothérapie
WO2015082499A2 (fr) 2013-12-03 2015-06-11 Iomet Pharma Ltd Composé pharmaceutique
WO2015085847A1 (fr) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Anticorps anti-pd-1, son fragment de liaison à l'antigène, et son application médicale
US20150203579A1 (en) 2014-01-23 2015-07-23 Regeneron Pharmaceuticals, Inc. Human Antibodies to PD-1
US20150203848A1 (en) 2014-01-21 2015-07-23 City Of Hope Ctla-4 aptamer sirna species
WO2015109124A2 (fr) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Agents immunomodulateurs
WO2015112900A1 (fr) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Molécules d'anticorps anti-pd-1 et leurs utilisations
WO2015112805A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains dirigés contre pd-l1
WO2015116539A1 (fr) 2014-01-28 2015-08-06 Bristol-Myers Squibb Company Anticorps anti-lag-3 pour traiter des hémopathies malignes
US20150225367A1 (en) 2014-02-12 2015-08-13 Iteos Therapeutics Novel 3-(indol-3-yl)-pyridine derivatives, pharmaceutical compositions and methods for use
US9126984B2 (en) 2013-11-08 2015-09-08 Iteos Therapeutics 4-(indol-3-yl)-pyrazole derivatives, pharmaceutical compositions and methods for use
WO2015140717A1 (fr) 2014-03-18 2015-09-24 Iteos Therapeutics Dérivés substitués par 3-indole, compositions pharmaceutiques et procédés d'utilisation
WO2015150097A1 (fr) 2014-04-04 2015-10-08 Iomet Pharma Ltd Dérivés indolés destinés à être utilisés dans le domaine de la médecine
US20150320859A1 (en) 2012-05-31 2015-11-12 Genentech, Inc. Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
WO2015173267A1 (fr) 2014-05-13 2015-11-19 Medimmune Limited Anticorps anti-b7-h1 et anti-ctla -4 pour le traitement du cancer du poumon non à petites cellules
WO2015173764A1 (fr) 2014-05-15 2015-11-19 Iteos Therapeutics Dérivés de pyrrolidine -2,5-dione, compositions pharmaceutiques et procédés pour une utilisation en tant qu'inhibiteursde de ido1
WO2015179654A1 (fr) 2014-05-22 2015-11-26 Mayo Foundation For Medical Education And Research Distinction d'anticorps anti-b7-h1 agonistes et antagonistes
WO2015181342A1 (fr) 2014-05-29 2015-12-03 Spring Bioscience Corporation Anticorps dirigés contre pd-l1 et leurs utilisations
US20150344576A1 (en) 2004-07-01 2015-12-03 Novo Nordisk A/S - Novo Alle Human anti-kir antibodies
WO2015200119A1 (fr) 2014-06-26 2015-12-30 Macrogenics, Inc. Dianticorps liés par covalence, présentant une immunoréactivité avec pd-1 et lag-3 et leurs procédés d'utilisation
WO2016000619A1 (fr) 2014-07-03 2016-01-07 Beigene, Ltd. Anticorps anti-pd-l1 et leur utilisation comme agents thérapeutiques et diagnostiques
WO2016007235A1 (fr) 2014-07-11 2016-01-14 Genentech, Inc. Anticorps anti-pd-l1 et leurs utilisations
WO2016022630A1 (fr) 2014-08-05 2016-02-11 Jiping Zha Anticorps anti-pd-l1
WO2016028672A1 (fr) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anticorps et fragments de fixation à l'antigène anti-lag3
WO2016026772A1 (fr) 2014-08-19 2016-02-25 Iomet Pharma Ltd Composé pharmaceutique
WO2016032334A1 (fr) 2014-08-28 2016-03-03 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Combinaisons d'anticorps cd94/nkg2a et cd94/nkg2b et de vaccin
WO2016061142A1 (fr) 2014-10-14 2016-04-21 Novartis Ag Molécules d'anticorps de pd-l1 et leurs utilisations
WO2016071293A2 (fr) 2014-11-03 2016-05-12 Iomet Pharma Ltd Composé pharmaceutique
WO2016071283A1 (fr) 2014-11-03 2016-05-12 Iomet Pharma Ltd Inhibiteurs de la tryptophane-2,3-dioxygénase ou de l'indoleamine-2,3-dioxygénase
WO2016075099A1 (fr) 2014-11-10 2016-05-19 Medimmune Limited Molécules de liaison spécifiques du cd73 et leur utilisation
US9359442B2 (en) 2002-11-07 2016-06-07 Immunogen Inc. Anti-CD33 antibodies and methods for treatment of acute myeloid leukemia using the same
WO2016111645A1 (fr) 2015-01-09 2016-07-14 Agency For Science, Technology And Research Anticorps anti-pd-l1
WO2016126858A2 (fr) 2015-02-03 2016-08-11 Anaptysbio, Inc. Anticorps dirigés contre le gène d'activation 3 des lymphocytes (lag-3)
WO2016134335A2 (fr) 2015-02-19 2016-08-25 Compugen Ltd. Polypeptides pvrig et méthodes de traitement
WO2016134333A1 (fr) 2015-02-19 2016-08-25 Compugen Ltd. Anticorps anti-pvrig et méthodes d'utilisation
US20160251438A1 (en) 2013-08-01 2016-09-01 Argen-X N.V. Anti-garp protein and uses thereof
US20160263087A1 (en) 2013-11-08 2016-09-15 Iteos Therapeutics Novel 4-(indol-3-yl)-pyrazole derivatives, pharmaceutical compositions and methods for use
WO2016149201A2 (fr) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anticorps anti-pdl1, anticorps anti-pld1 activables, et leurs procédés d'utilisation
US20160272708A1 (en) 2013-06-26 2016-09-22 Shanghai Junshi Biosciences Inc. Anti-pd-1 antibody and use thereof
WO2016147144A1 (fr) 2015-03-17 2016-09-22 Pfizer Inc. Nouveaux dérivés substitués par un 2-indole, compositions pharmaceutiques et leurs procédés d'utilisation
WO2016160792A1 (fr) 2015-03-30 2016-10-06 Stcube & Co., Inc. Anticorps spécifiques de la protéine pd-l1 glycosylée et leurs procédés d'utilisation
WO2016181348A1 (fr) 2015-05-14 2016-11-17 Pfizer Inc. Combinaisons comprenant un inhibiteur d'ido1 de type pyrrolidine-2,5-dione et un anticorps
WO2016197367A1 (fr) 2015-06-11 2016-12-15 Wuxi Biologics (Shanghai) Co. Ltd. Nouveaux anticorps anti-pd-l1
WO2016200782A1 (fr) 2015-06-08 2016-12-15 Macrogenics, Inc. Molécules se liant à lag-3 et méthodes d'utilisation de ces dernières
WO2017007700A1 (fr) 2015-07-06 2017-01-12 Iomet Pharma Ltd. Composé pharmaceutique
WO2017015560A2 (fr) 2015-07-22 2017-01-26 Sorrento Therapeutics, Inc. Anticorps thérapeutiques qui se lient à lag3
WO2017019894A1 (fr) 2015-07-29 2017-02-02 Novartis Ag Polythérapies comprenant des molécules d'anticorps dirigées contre lag-3
WO2017019846A1 (fr) 2015-07-30 2017-02-02 Macrogenics, Inc. Molécules se liant à pd-1 et méthodes d'utilisation correspondantes
WO2017020802A1 (fr) 2015-07-31 2017-02-09 苏州康宁杰瑞生物科技有限公司 Anticorps à domaine unique pour le ligand du récepteur de mort cellulaire programmée (pd-l1) et protéine dérivée de celui-ci
WO2017020858A1 (fr) 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Nouveaux anticorps anti-pd-l1
WO2017025868A1 (fr) 2015-08-10 2017-02-16 Pfizer Inc. Dérivés substitués par un 3-indole, compositions pharmaceutiques et leurs procédés d'utilisation
WO2017025498A1 (fr) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Nouveau polypeptide de fusion spécifique de lag-3 et pd-1
WO2017025016A1 (fr) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017034916A1 (fr) 2015-08-24 2017-03-02 Eli Lilly And Company Anticorps anti-pd-l1 (« ligand de mort programmée 1 »)
WO2017040790A1 (fr) 2015-09-01 2017-03-09 Agenus Inc. Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
EP3146979A1 (fr) 2005-10-28 2017-03-29 University Court of The University of Dundee Agents de liaison siglec-9
WO2017059095A1 (fr) 2015-10-01 2017-04-06 Potenza Therapeutics, Inc. Protéines de liaison à un antigène anti-tigit et leurs méthodes d'utilisation
WO2017062888A1 (fr) 2015-10-09 2017-04-13 Regeneron Pharmaceuticals, Inc. Anticorps anti-lag3 et leurs utilisations
US9624185B1 (en) 2016-01-20 2017-04-18 Yong Xu Method for preparing IDO inhibitor epacadostat
US9642918B2 (en) 2011-12-16 2017-05-09 Pfizer Inc. Combination of inotuzumab ozogamicin and torisel for the treatment of cancer
WO2017087901A2 (fr) 2015-11-19 2017-05-26 Sutro Biopharma, Inc. Anticorps anti-lag3, compositions comprenant des anticorps anti-lag3 et méthodes de production et d'utilisation d'anticorps anti-lag3
WO2017087589A2 (fr) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Liants pd1 et/ou lag3
WO2017106129A1 (fr) 2015-12-16 2017-06-22 Merck Sharp & Dohme Corp. Anticorps anti-lag3 et fragments de fixation à l'antigène
EP3193929A1 (fr) 2014-09-16 2017-07-26 Innate Pharma Régimes de traitement utilisant des anticorps anti-nkg2a
WO2017132825A1 (fr) 2016-02-02 2017-08-10 华为技术有限公司 Procédé de vérification de puissance d'émission, équipement utilisateur et station de base
WO2017133540A1 (fr) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017149143A1 (fr) 2016-03-04 2017-09-08 Agency For Science, Technology And Research Anticorps anti-lag-3
US20170260271A1 (en) 2014-05-13 2017-09-14 Chugai Seiyaku Kabushiki Kaisha T Cell-Redirected Antigen-Binding Molecule For Cells Having Immunosuppression Function
EP3230319A1 (fr) 2014-12-09 2017-10-18 Rinat Neuroscience Corporation Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
US20170306014A1 (en) 2014-09-10 2017-10-26 Innate Pharma Cross reactive siglec antibodies
WO2017198741A1 (fr) 2016-05-18 2017-11-23 Boehringer Ingelheim International Gmbh Anticorps anti-pd-1 et anti-lag3 pour le traitement du cancer
WO2017209053A1 (fr) 2016-05-30 2017-12-07 アステラス製薬株式会社 Nouveau virus de la vaccine génétiquement modifié
EP3253796A1 (fr) 2015-02-03 2017-12-13 Université Catholique de Louvain Protéine anti-garp et ses utilisations
WO2017220569A1 (fr) 2016-06-20 2017-12-28 F-Star Delta Limited Molécules de liaison liant pd-l1 et lag -3
WO2017220555A1 (fr) 2016-06-20 2017-12-28 F-Star Beta Limited Éléments de liaison lag-3
WO2017219995A1 (fr) 2016-06-23 2017-12-28 江苏恒瑞医药股份有限公司 Anticorps anti-lag-3, fragment de celui-ci se liant à l'antigène, et son application pharmaceutique
WO2018017864A2 (fr) 2016-07-20 2018-01-25 Oncomed Pharmaceuticals, Inc. Agents de liaison à pvrig et leurs utilisations
WO2018022831A1 (fr) 2016-07-28 2018-02-01 Musc Foundation For Research Development Méthodes et compositions permettant le traitement du cancer associant un anticorps anti-smic et des inhibiteurs de point de contrôle immunitaire
US20180036306A1 (en) 2016-08-08 2018-02-08 Acetylon Pharmaceuticals Inc. Methods of use and pharmaceutical combinations of histone deacetylase inhibitors and cd20 inhibitory antibodies
WO2018027203A1 (fr) 2016-08-05 2018-02-08 Allakos, Inc. Anticorps anti-siglec-7 pour le traitement du cancer
WO2018034227A1 (fr) 2016-08-15 2018-02-22 国立大学法人北海道大学 Anticorps anti-lag-3
WO2018033798A1 (fr) 2016-08-17 2018-02-22 Compugen Ltd. Anticorps anti-tigit, anticorps anti-pvrig et combinaisons associées
WO2018054484A1 (fr) * 2016-09-23 2018-03-29 Biontech Ag Anticorps trivalents bispécifiques se liant à claudine 6 ou claudin18.2 et cd3 pour le traitement de maladies cancéreuses exprimant la claudine
WO2018069500A2 (fr) 2016-10-13 2018-04-19 Symphogen A/S Anticorps anti-lag-3 et compositions
WO2018071500A1 (fr) 2016-10-11 2018-04-19 Agenus Inc. Anticorps anti-lag-3 et leurs procédés d'utilisation
US20180117147A1 (en) 2012-10-23 2018-05-03 Bristol-Myers Squibb Company Combination of anti-kir and anti-ctla-4 antibodies to treat cancer
WO2018085469A2 (fr) 2016-11-01 2018-05-11 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de lymphocyte t et la protéine 3 de mucine (tim-3)
WO2018083087A2 (fr) 2016-11-02 2018-05-11 Glaxosmithkline Intellectual Property (No.2) Limited Protéines de liaison
US20180155428A1 (en) 2009-03-17 2018-06-07 Université D'aix Marseille BTLA Antibodies and Uses Thereof
WO2018106529A1 (fr) 2016-12-08 2018-06-14 Eli Lilly And Company Anticorps anti-tim-3 pour combinaison avec des anticorps anti-pd-l1
WO2018103501A1 (fr) 2016-12-08 2018-06-14 上海优卡迪生物医药科技有限公司 Arnsi d'inactivation de pd-1 humain, vecteur de car-t d'expression recombinée, procédé de préparation et application associés
WO2018106588A1 (fr) 2016-12-08 2018-06-14 Eli Lilly And Company Anticorps anti-tim-3 pour une combinaison avec des anticorps anti-pd-1
WO2018110555A1 (fr) 2016-12-13 2018-06-21 アステラス製薬株式会社 Anticorps anti-cd73 humain
US20180185482A1 (en) 2017-01-05 2018-07-05 Gensun Biopharma Inc. Checkpoint regulator antagonists
US20180327511A1 (en) 2015-09-24 2018-11-15 Daiichi Sankyo Company, Limited Anti-garp antibody
WO2018222711A2 (fr) 2017-05-30 2018-12-06 Bristol-Myers Squibb Company Compositions comprenant une combinaison d'un anticorps anti-lag-3, d'un inhibiteur de voie pd-1 et d'un agent immunothérapeutique
WO2019000146A1 (fr) 2017-06-26 2019-01-03 深圳市博奥康生物科技有限公司 Siarn de récepteur de mort cellulaire programmée humaine 1 et son utilisation
WO2019011852A1 (fr) 2017-07-10 2019-01-17 Innate Pharma Polythérapie utilisant un anticorps dirigé contre siglec-9 humain et anticorps dirigé contre nkg2a humain pour le traitement du cancer
WO2019011855A1 (fr) 2017-07-10 2019-01-17 Innate Pharma Anticorps neutralisant la siglec-9
US20190023786A1 (en) 2016-01-12 2019-01-24 Palleon Pharmaceuticals Inc. Use of siglec-7 or siglec-9 antibodies for the treatment of cancer
US20190062427A1 (en) 2015-10-29 2019-02-28 Alector Llc Anti-siglec-9 antibodies and methods of use thereof
US20190127483A1 (en) 2016-03-30 2019-05-02 Musc Foundation For Research Development Methods for treatment and diagnosis of cancer by targeting glycoprotein a repetitions predominant (garp) and for providing effective immunotherapy alone or in combination
WO2020025792A1 (fr) * 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Constructions d'anticorps pour cldn18.2 et cd3
WO2021011885A1 (fr) * 2019-07-17 2021-01-21 The Regents Of The University Of California Anticorps dirigés contre la claudine 18 et méthodes de traitement du cancer
WO2021025177A1 (fr) * 2019-08-06 2021-02-11 Astellas Pharma Inc. Polythérapie impliquant des anticorps dirigés contre la claudine 18.2 et inhibiteurs de point de contrôle immunitaire pour le traitement du cancer
WO2022203090A1 (fr) * 2021-03-25 2022-09-29 Astellas Pharma Inc. Polythérapie impliquant des anticorps dirigés contre la claudine 18.2 pour le traitement du cancer
WO2022263508A1 (fr) * 2021-06-15 2022-12-22 Astellas Pharma Europe Bv Agents de liaison bispécifiques se liant à la cldn18.2 et au cd3

Patent Citations (234)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5475096A (en) 1990-06-11 1995-12-12 University Research Corporation Nucleic acid ligands
WO1992022653A1 (fr) 1991-06-14 1992-12-23 Genentech, Inc. Procede de production d'anticorps humanises
US5977318A (en) 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
EP0629240A1 (fr) 1992-02-19 1994-12-21 Scotgen Limited Anticorps modifies, produits et procedes s'y rapportant
US5843439A (en) 1992-11-13 1998-12-01 Anderson; Darrell R. Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US8216805B2 (en) 1995-03-01 2012-07-10 Genentech, Inc. Knobs and holes heteromeric polypeptides
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
US5855887A (en) 1995-07-25 1999-01-05 The Regents Of The University Of California Blockade of lymphocyte down-regulation associated with CTLA-4 signaling
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
WO1998042752A1 (fr) 1997-03-21 1998-10-01 Brigham And Women's Hospital Inc. Peptides immunotherapeutiques se liant a ctla-4
US6207156B1 (en) 1997-03-21 2001-03-27 Brigham And Women's Hospital, Inc. Specific antibodies and antibody fragments
WO2000037504A2 (fr) 1998-12-23 2000-06-29 Pfizer Inc. Anticorps monoclonaux humains diriges contre l'antigene ctla-4
US7132281B2 (en) 1998-12-23 2006-11-07 Amgen Fremont Inc. Methods and host cells for producing human monoclonal antibodies to CTLA-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
US7635757B2 (en) 1999-08-23 2009-12-22 Dana-Farber Cancer Institute, Inc. B7-4 Antibodies and uses therefor
US6808710B1 (en) 1999-08-23 2004-10-26 Genetics Institute, Inc. Downmodulating an immune response with multivalent antibodies to PD-1
WO2001014424A2 (fr) 1999-08-24 2001-03-01 Medarex, Inc. Anticorps contre l'antigene ctla-4 humain et utilisation
US20050201994A1 (en) 1999-08-24 2005-09-15 Medarex, Inc. Human CTLA-4 antibodies and their uses
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
US20020086014A1 (en) 1999-08-24 2002-07-04 Korman Alan J. Human CTLA-4 antibodies and their uses
EP1212422A2 (fr) 1999-08-24 2002-06-12 Medarex, Inc. Anticorps contre l'antigene ctla-4 humain et utilisation
EP1223980A2 (fr) 1999-10-28 2002-07-24 Hofbauer, Reinhold Utilisation d'inhibiteurs csf-1
WO2001030381A2 (fr) 1999-10-28 2001-05-03 Hofbauer, Reinhold Utilisation d'inhibiteurs csf-1
US20090317368A1 (en) 1999-11-30 2009-12-24 Lieping Chen B7-h1, a novel immunoregulatory molecule
US20020039581A1 (en) 2000-01-27 2002-04-04 Carreno Beatriz M. Antibodies against CTLA4 and uses therefor
WO2002055082A1 (fr) 2001-01-10 2002-07-18 Vernalis Research Limited Dérivés pyrazolo[3,4-d]pyrimidiniques et leur utilisation comme antagonistes des récepteurs purinergiques
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030118592A1 (en) 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
WO2003042402A2 (fr) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents modulant l'activite de cellules immunes et procedes d'utilisation associes
US8153768B2 (en) 2002-05-02 2012-04-10 Wyeth Holdings Corporation Calicheamicin derivative-carrier conjugates
WO2003099196A2 (fr) 2002-05-23 2003-12-04 Cure Tech Ltd. Anticorps monoclonaux humanises immunomodulateurs servant a traiter une maladie neoplasique ou une immunodeficience
US7332582B2 (en) 2002-05-23 2008-02-19 Curetech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
WO2004035607A2 (fr) 2002-10-17 2004-04-29 Genmab A/S Anticorps monoclonaux humains anti-cd20
US9359442B2 (en) 2002-11-07 2016-06-07 Immunogen Inc. Anti-CD33 antibodies and methods for treatment of acute myeloid leukemia using the same
WO2004056312A2 (fr) 2002-12-16 2004-07-08 Genentech, Inc. Variants d'immunoglobuline et utilisations
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2005003168A2 (fr) 2003-07-02 2005-01-13 Novo Nordisk A/S Compositions et procedes pour la regulation d'activite de cellules nk
WO2005009465A1 (fr) 2003-07-24 2005-02-03 Innate Pharma Methodes et compositions pour augmenter l'efficacite d'anticorps therapeutiques au moyen de composes de potentialisation de cellules nk
US20150344576A1 (en) 2004-07-01 2015-12-03 Novo Nordisk A/S - Novo Alle Human anti-kir antibodies
WO2006072625A2 (fr) 2005-01-06 2006-07-13 Novo Nordisk A/S Procedes et traitements combines anti-kir
WO2006072626A1 (fr) 2005-01-06 2006-07-13 Novo Nordisk A/S Agents de liaison kir et leurs procedes d'utilisation
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
US8779105B2 (en) 2005-05-09 2014-07-15 Medarex, L.L.C. Monoclonal antibodies to programmed death 1 (PD-1)
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007042573A2 (fr) 2005-10-14 2007-04-19 Innate Pharma Compositions et procedes pour traiter des troubles de proliferation
EP3146979A1 (fr) 2005-10-28 2017-03-29 University Court of The University of Dundee Agents de liaison siglec-9
WO2008084106A1 (fr) 2007-01-11 2008-07-17 Novo Nordisk A/S Anticorps anti-kir, formulations et utilisations de celles-ci
US20180208652A1 (en) 2007-01-11 2018-07-26 Novo Nordisk A/S Anti-kir antibodies, formulations, and uses thereof
WO2008132601A1 (fr) 2007-04-30 2008-11-06 Immutep Anticorps monoclonal anti-lag-3 cytotoxique et son utilisation dans le traitement ou la prévention d'un rejet du greffon d'organe et de maladies auto-immunes
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
WO2009014708A2 (fr) 2007-07-23 2009-01-29 Cell Genesys, Inc. Anticorps pd-1 en combinaison avec une cellule sécrétant de la cytokine et leurs procédés d'utilisation
WO2009044273A2 (fr) 2007-10-05 2009-04-09 Immutep Utilisation d'une protéine lag-3 recombinée ou de dérivés de celle-ci pour produire une réponse immunitaire des monocytes
EP2205257A2 (fr) 2007-10-05 2010-07-14 Immutep Utilisation de lag-3 recombinant ou ses dérivatifs pour déclencher la réponse immune des monocytes
EP2628753A1 (fr) 2008-01-24 2013-08-21 Novo Nordisk A/S Anticorps monoclonal NKG2A antihumain humanisé
US9422368B2 (en) 2008-01-24 2016-08-23 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
WO2009101611A1 (fr) 2008-02-11 2009-08-20 Curetech Ltd. Anticorps monoclonaux pour le traitement de tumeurs
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
WO2010012557A1 (fr) 2008-08-01 2010-02-04 Emitec Gesellschaft Für Emissionstechnologie Mbh Procédé de conduite d'une installation de traitement des gaz d'échappement à régulation lambda
EP2320940A2 (fr) 2008-08-11 2011-05-18 Medarex, Inc. Anticorps humains qui se lient au gène 3 d'activation des lymphocytes (lag-3), et leurs utilisations
US20110150892A1 (en) 2008-08-11 2011-06-23 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
WO2010036959A2 (fr) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Anticorps anti-pd-1, pd-l1, et pd-l2 humains et leurs utilisations
US8709411B2 (en) 2008-12-05 2014-04-29 Novo Nordisk A/S Combination therapy to enhance NK cell mediated cytotoxicity
WO2010065939A1 (fr) 2008-12-05 2010-06-10 Indiana University Research & Technology Corporation Traitement combiné pour améliorer la cytotoxicité induite par les cellules nk
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
US20180155428A1 (en) 2009-03-17 2018-06-07 Université D'aix Marseille BTLA Antibodies and Uses Thereof
WO2011014438A1 (fr) 2009-07-31 2011-02-03 N.V. Organon Anticorps totalement humains dirigés contre le btla
US20120177645A1 (en) 2009-08-31 2012-07-12 Solomon Langermann Methods and compositions for the inhibition of transplant rejection
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011070024A1 (fr) 2009-12-10 2011-06-16 F. Hoffmann-La Roche Ag Anticorps se liant de façon préférentielle au domaine extracellulaire 4 de csf1r humain et leur utilisation
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
US20120294796A1 (en) 2010-03-04 2012-11-22 Macrogenics, Inc. Antibodies Reactive with B7-H3 and Uses Thereof
WO2011107553A1 (fr) 2010-03-05 2011-09-09 F. Hoffmann-La Roche Ag Anticorps dirigés contre le csf-1r humain et utilisations associées
WO2011131407A1 (fr) 2010-03-05 2011-10-27 F. Hoffmann-La Roche Ag Anticorps contre le csf-1r humain et leurs utilisations
WO2011140249A2 (fr) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Anticorps liant csf1r
WO2011159877A2 (fr) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Anticorps di-spécifiques anti-tim-3 et pd-1 pour immunothérapie dans des états pathologiques immuns chroniques
WO2011161699A2 (fr) 2010-06-25 2011-12-29 Aurigene Discovery Technologies Limited Composés modulateurs de l'immunosuppression
WO2012071411A2 (fr) 2010-11-22 2012-05-31 Innate Pharma Sa Traitements modulant les cellules tueuses naturelles et méthodes de traitement d'hémopathies malignes
WO2012145493A1 (fr) 2011-04-20 2012-10-26 Amplimmune, Inc. Anticorps et autres molécules qui se lient à b7-h1 et à pd-1
WO2012160448A2 (fr) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anticorps anti-kir destinés au traitement de troubles inflammatoires
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
US9724413B2 (en) 2011-08-01 2017-08-08 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
WO2013019906A1 (fr) 2011-08-01 2013-02-07 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2013025779A1 (fr) 2011-08-15 2013-02-21 Amplimmune, Inc. Anticorps anti-b7-h4 et leurs utilisations
WO2013054331A1 (fr) 2011-10-11 2013-04-18 Tel Hashomer Medical Research Infrastructure And Services Ltd. Anticorps dirigés contre la molécule d'adhésion cellulaire associée à l'antigène carcinoembryonnaire (ceacam)
WO2013067492A1 (fr) 2011-11-03 2013-05-10 The Trustees Of The University Of Pennsylvania Compositions spécifiques de b7-h4 isolé et procédés d'utilisation associés
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013087699A1 (fr) 2011-12-15 2013-06-20 F. Hoffmann-La Roche Ag Anticorps contre le csf-1r humain et leurs utilisations
US9642918B2 (en) 2011-12-16 2017-05-09 Pfizer Inc. Combination of inotuzumab ozogamicin and torisel for the treatment of cancer
WO2013119716A1 (fr) 2012-02-06 2013-08-15 Genentech, Inc. Compositions et procédés d'utilisation d'inhibiteurs de csf1r
WO2013132044A1 (fr) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Thérapie combinée d'anticorps contre le csf -1r humain et ses utilisations
WO2013169264A1 (fr) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Méthodes destinées à traiter des affections avec des anticorps qui se lient au récepteur du facteur 1 de stimulation des colonies (csf1r)
WO2013173223A1 (fr) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Immunothérapie anticancéreuse par rupture de la signalisation pd-1/pd-l1
US20150320859A1 (en) 2012-05-31 2015-11-12 Genentech, Inc. Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
WO2013181634A2 (fr) 2012-05-31 2013-12-05 Sorrento Therapeutics Inc. Protéines liant un antigène qui lient pd-l1
WO2014008218A1 (fr) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimisation d'anticorps se liant à la protéine lag-3 exprimée par le gène 3 d'activation des lymphocytes, et leurs utilisations
WO2014036357A1 (fr) 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Méthodes de traitement de pathologies par des anticorps qui se lient au récepteur du facteur stimulant les colonies 1 (csf1r)
WO2014055648A1 (fr) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
US20180117147A1 (en) 2012-10-23 2018-05-03 Bristol-Myers Squibb Company Combination of anti-kir and anti-ctla-4 antibodies to treat cancer
WO2014094122A1 (fr) 2012-12-17 2014-06-26 Trillium Therapeutics Inc. Traitement de cellules tumorales à cd47+ avec des fusions sirp alpha/fc
WO2014100079A1 (fr) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Anticorps qui se lient au ligand 1 de la mort programmée humaine (pd-l1)
US20140370013A1 (en) 2013-01-14 2014-12-18 Xencor, Inc. Novel heterodimeric proteins
WO2014132072A1 (fr) 2013-02-28 2014-09-04 University Court Of The University Of Edinburgh Thérapie à base de csf1
WO2014165082A2 (fr) 2013-03-13 2014-10-09 Medimmune, Llc Anticorps et procédés de détection
WO2014159562A1 (fr) 2013-03-14 2014-10-02 Bristol-Myers Squibb Company Combinaison d'agoniste de dr5 et d'antagoniste anti-pd-1 et méthodes d'utilisation associées
EP2855533A1 (fr) 2013-03-15 2015-04-08 Momenta Pharmaceuticals, Inc. Procédés se rapportant à des protéines de fusion fc-ctla4
WO2014140180A1 (fr) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Protéines de liaison anti-lag-3
WO2014179664A2 (fr) 2013-05-02 2014-11-06 Anaptysbio, Inc. Anticorps dirigés contre la protéine de mort programmée 1 (pd-1)
WO2014183885A1 (fr) 2013-05-17 2014-11-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antagoniste de l'interaction btla/hvem pour une utilisation en thérapie
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
US20160272708A1 (en) 2013-06-26 2016-09-22 Shanghai Junshi Biosciences Inc. Anti-pd-1 antibody and use thereof
WO2014207748A1 (fr) 2013-06-27 2014-12-31 Alexander Biro Molécules ctla-4 solubles et leurs dérivés pour le traitement d'une néphrose lipoïdique
WO2015009856A2 (fr) 2013-07-16 2015-01-22 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
US20190016811A1 (en) 2013-08-01 2019-01-17 Ludwig Institute For Cancer Research Ltd Anti-garp protein and uses thereof
US20160251438A1 (en) 2013-08-01 2016-09-01 Argen-X N.V. Anti-garp protein and uses thereof
WO2015035606A1 (fr) 2013-09-13 2015-03-19 Beigene, Ltd. Anticorps anti-pd1 et leur utilisation comme produits thérapeutiques et produits de diagnostic
US20150079109A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2015042246A1 (fr) 2013-09-20 2015-03-26 Bristol-Myers Squibb Company Combinaison d'anticorps anti-lag-3 et d'anticorps anti-pd-1 pour traiter des tumeurs
WO2015061668A1 (fr) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux anti-pd-l1 et fragments de ceux-ci
US20160263087A1 (en) 2013-11-08 2016-09-15 Iteos Therapeutics Novel 4-(indol-3-yl)-pyrazole derivatives, pharmaceutical compositions and methods for use
US9126984B2 (en) 2013-11-08 2015-09-08 Iteos Therapeutics 4-(indol-3-yl)-pyrazole derivatives, pharmaceutical compositions and methods for use
WO2015075725A1 (fr) 2013-11-25 2015-05-28 Ccam Biotherapeutics Ltd. Compositions comprenant des anticorps anti-ceacam1 et anti-pd pour la cancérothérapie
WO2015082499A2 (fr) 2013-12-03 2015-06-11 Iomet Pharma Ltd Composé pharmaceutique
WO2015085847A1 (fr) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Anticorps anti-pd-1, son fragment de liaison à l'antigène, et son application médicale
WO2015109124A2 (fr) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Agents immunomodulateurs
US20150203848A1 (en) 2014-01-21 2015-07-23 City Of Hope Ctla-4 aptamer sirna species
WO2015112800A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains se liant à pd-1
US20150203579A1 (en) 2014-01-23 2015-07-23 Regeneron Pharmaceuticals, Inc. Human Antibodies to PD-1
WO2015112805A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains dirigés contre pd-l1
WO2015112900A1 (fr) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Molécules d'anticorps anti-pd-1 et leurs utilisations
WO2015116539A1 (fr) 2014-01-28 2015-08-06 Bristol-Myers Squibb Company Anticorps anti-lag-3 pour traiter des hémopathies malignes
WO2015121812A1 (fr) 2014-02-12 2015-08-20 Iteos Therapeutics Dérivés de 3-(indol-3-yl)piridine, compositions pharmaceutiques et procédés d'utilisation
US20150225367A1 (en) 2014-02-12 2015-08-13 Iteos Therapeutics Novel 3-(indol-3-yl)-pyridine derivatives, pharmaceutical compositions and methods for use
WO2015140717A1 (fr) 2014-03-18 2015-09-24 Iteos Therapeutics Dérivés substitués par 3-indole, compositions pharmaceutiques et procédés d'utilisation
WO2015150097A1 (fr) 2014-04-04 2015-10-08 Iomet Pharma Ltd Dérivés indolés destinés à être utilisés dans le domaine de la médecine
WO2015173267A1 (fr) 2014-05-13 2015-11-19 Medimmune Limited Anticorps anti-b7-h1 et anti-ctla -4 pour le traitement du cancer du poumon non à petites cellules
US20170260271A1 (en) 2014-05-13 2017-09-14 Chugai Seiyaku Kabushiki Kaisha T Cell-Redirected Antigen-Binding Molecule For Cells Having Immunosuppression Function
WO2015173764A1 (fr) 2014-05-15 2015-11-19 Iteos Therapeutics Dérivés de pyrrolidine -2,5-dione, compositions pharmaceutiques et procédés pour une utilisation en tant qu'inhibiteursde de ido1
WO2015179654A1 (fr) 2014-05-22 2015-11-26 Mayo Foundation For Medical Education And Research Distinction d'anticorps anti-b7-h1 agonistes et antagonistes
WO2015181342A1 (fr) 2014-05-29 2015-12-03 Spring Bioscience Corporation Anticorps dirigés contre pd-l1 et leurs utilisations
WO2015200119A1 (fr) 2014-06-26 2015-12-30 Macrogenics, Inc. Dianticorps liés par covalence, présentant une immunoréactivité avec pd-1 et lag-3 et leurs procédés d'utilisation
US20190169292A1 (en) 2014-06-26 2019-06-06 Macrogenics, Inc. Covalently Bonded Diabodies Having Immunoreactivity with PD-1 and LAG-3, and Methods of Use Thereof
WO2016000619A1 (fr) 2014-07-03 2016-01-07 Beigene, Ltd. Anticorps anti-pd-l1 et leur utilisation comme agents thérapeutiques et diagnostiques
WO2016007235A1 (fr) 2014-07-11 2016-01-14 Genentech, Inc. Anticorps anti-pd-l1 et leurs utilisations
WO2016022630A1 (fr) 2014-08-05 2016-02-11 Jiping Zha Anticorps anti-pd-l1
WO2016028672A1 (fr) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anticorps et fragments de fixation à l'antigène anti-lag3
WO2016026772A1 (fr) 2014-08-19 2016-02-25 Iomet Pharma Ltd Composé pharmaceutique
WO2016032334A1 (fr) 2014-08-28 2016-03-03 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Combinaisons d'anticorps cd94/nkg2a et cd94/nkg2b et de vaccin
US20170306014A1 (en) 2014-09-10 2017-10-26 Innate Pharma Cross reactive siglec antibodies
EP3193929A1 (fr) 2014-09-16 2017-07-26 Innate Pharma Régimes de traitement utilisant des anticorps anti-nkg2a
WO2016061142A1 (fr) 2014-10-14 2016-04-21 Novartis Ag Molécules d'anticorps de pd-l1 et leurs utilisations
WO2016071283A1 (fr) 2014-11-03 2016-05-12 Iomet Pharma Ltd Inhibiteurs de la tryptophane-2,3-dioxygénase ou de l'indoleamine-2,3-dioxygénase
WO2016071293A2 (fr) 2014-11-03 2016-05-12 Iomet Pharma Ltd Composé pharmaceutique
WO2016075099A1 (fr) 2014-11-10 2016-05-19 Medimmune Limited Molécules de liaison spécifiques du cd73 et leur utilisation
EP3230319A1 (fr) 2014-12-09 2017-10-18 Rinat Neuroscience Corporation Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
WO2016111645A1 (fr) 2015-01-09 2016-07-14 Agency For Science, Technology And Research Anticorps anti-pd-l1
WO2016126858A2 (fr) 2015-02-03 2016-08-11 Anaptysbio, Inc. Anticorps dirigés contre le gène d'activation 3 des lymphocytes (lag-3)
EP3253796A1 (fr) 2015-02-03 2017-12-13 Université Catholique de Louvain Protéine anti-garp et ses utilisations
WO2016134335A2 (fr) 2015-02-19 2016-08-25 Compugen Ltd. Polypeptides pvrig et méthodes de traitement
WO2016134333A1 (fr) 2015-02-19 2016-08-25 Compugen Ltd. Anticorps anti-pvrig et méthodes d'utilisation
WO2016149201A2 (fr) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anticorps anti-pdl1, anticorps anti-pld1 activables, et leurs procédés d'utilisation
WO2016147144A1 (fr) 2015-03-17 2016-09-22 Pfizer Inc. Nouveaux dérivés substitués par un 2-indole, compositions pharmaceutiques et leurs procédés d'utilisation
WO2016160792A1 (fr) 2015-03-30 2016-10-06 Stcube & Co., Inc. Anticorps spécifiques de la protéine pd-l1 glycosylée et leurs procédés d'utilisation
WO2016181348A1 (fr) 2015-05-14 2016-11-17 Pfizer Inc. Combinaisons comprenant un inhibiteur d'ido1 de type pyrrolidine-2,5-dione et un anticorps
WO2016200782A1 (fr) 2015-06-08 2016-12-15 Macrogenics, Inc. Molécules se liant à lag-3 et méthodes d'utilisation de ces dernières
WO2016197367A1 (fr) 2015-06-11 2016-12-15 Wuxi Biologics (Shanghai) Co. Ltd. Nouveaux anticorps anti-pd-l1
WO2017007700A1 (fr) 2015-07-06 2017-01-12 Iomet Pharma Ltd. Composé pharmaceutique
WO2017015560A2 (fr) 2015-07-22 2017-01-26 Sorrento Therapeutics, Inc. Anticorps thérapeutiques qui se lient à lag3
WO2017019894A1 (fr) 2015-07-29 2017-02-02 Novartis Ag Polythérapies comprenant des molécules d'anticorps dirigées contre lag-3
WO2017019846A1 (fr) 2015-07-30 2017-02-02 Macrogenics, Inc. Molécules se liant à pd-1 et méthodes d'utilisation correspondantes
WO2017020802A1 (fr) 2015-07-31 2017-02-09 苏州康宁杰瑞生物科技有限公司 Anticorps à domaine unique pour le ligand du récepteur de mort cellulaire programmée (pd-l1) et protéine dérivée de celui-ci
WO2017020801A1 (fr) 2015-07-31 2017-02-09 苏州康宁杰瑞生物科技有限公司 Anticorps à domaine unique pour le ligand du récepteur de mort cellulaire programmée (pd-l1) et protéine dérivée de celui-ci
WO2017020291A1 (fr) 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Nouveaux anticorps anti-pd-l1
WO2017020858A1 (fr) 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Nouveaux anticorps anti-pd-l1
WO2017025498A1 (fr) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Nouveau polypeptide de fusion spécifique de lag-3 et pd-1
WO2017024465A1 (fr) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017025868A1 (fr) 2015-08-10 2017-02-16 Pfizer Inc. Dérivés substitués par un 3-indole, compositions pharmaceutiques et leurs procédés d'utilisation
WO2017025016A1 (fr) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017034916A1 (fr) 2015-08-24 2017-03-02 Eli Lilly And Company Anticorps anti-pd-l1 (« ligand de mort programmée 1 »)
WO2017040790A1 (fr) 2015-09-01 2017-03-09 Agenus Inc. Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
US20180327511A1 (en) 2015-09-24 2018-11-15 Daiichi Sankyo Company, Limited Anti-garp antibody
WO2017059095A1 (fr) 2015-10-01 2017-04-06 Potenza Therapeutics, Inc. Protéines de liaison à un antigène anti-tigit et leurs méthodes d'utilisation
WO2017062888A1 (fr) 2015-10-09 2017-04-13 Regeneron Pharmaceuticals, Inc. Anticorps anti-lag3 et leurs utilisations
US20190062427A1 (en) 2015-10-29 2019-02-28 Alector Llc Anti-siglec-9 antibodies and methods of use thereof
WO2017087589A2 (fr) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Liants pd1 et/ou lag3
WO2017087901A2 (fr) 2015-11-19 2017-05-26 Sutro Biopharma, Inc. Anticorps anti-lag3, compositions comprenant des anticorps anti-lag3 et méthodes de production et d'utilisation d'anticorps anti-lag3
WO2017106129A1 (fr) 2015-12-16 2017-06-22 Merck Sharp & Dohme Corp. Anticorps anti-lag3 et fragments de fixation à l'antigène
US20190023786A1 (en) 2016-01-12 2019-01-24 Palleon Pharmaceuticals Inc. Use of siglec-7 or siglec-9 antibodies for the treatment of cancer
US9624185B1 (en) 2016-01-20 2017-04-18 Yong Xu Method for preparing IDO inhibitor epacadostat
WO2017133540A1 (fr) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017132825A1 (fr) 2016-02-02 2017-08-10 华为技术有限公司 Procédé de vérification de puissance d'émission, équipement utilisateur et station de base
WO2017149143A1 (fr) 2016-03-04 2017-09-08 Agency For Science, Technology And Research Anticorps anti-lag-3
US20190127483A1 (en) 2016-03-30 2019-05-02 Musc Foundation For Research Development Methods for treatment and diagnosis of cancer by targeting glycoprotein a repetitions predominant (garp) and for providing effective immunotherapy alone or in combination
WO2017198741A1 (fr) 2016-05-18 2017-11-23 Boehringer Ingelheim International Gmbh Anticorps anti-pd-1 et anti-lag3 pour le traitement du cancer
WO2017209053A1 (fr) 2016-05-30 2017-12-07 アステラス製薬株式会社 Nouveau virus de la vaccine génétiquement modifié
WO2017220555A1 (fr) 2016-06-20 2017-12-28 F-Star Beta Limited Éléments de liaison lag-3
WO2017220569A1 (fr) 2016-06-20 2017-12-28 F-Star Delta Limited Molécules de liaison liant pd-l1 et lag -3
WO2017219995A1 (fr) 2016-06-23 2017-12-28 江苏恒瑞医药股份有限公司 Anticorps anti-lag-3, fragment de celui-ci se liant à l'antigène, et son application pharmaceutique
WO2018017864A2 (fr) 2016-07-20 2018-01-25 Oncomed Pharmaceuticals, Inc. Agents de liaison à pvrig et leurs utilisations
WO2018022831A1 (fr) 2016-07-28 2018-02-01 Musc Foundation For Research Development Méthodes et compositions permettant le traitement du cancer associant un anticorps anti-smic et des inhibiteurs de point de contrôle immunitaire
WO2018027203A1 (fr) 2016-08-05 2018-02-08 Allakos, Inc. Anticorps anti-siglec-7 pour le traitement du cancer
US20180036306A1 (en) 2016-08-08 2018-02-08 Acetylon Pharmaceuticals Inc. Methods of use and pharmaceutical combinations of histone deacetylase inhibitors and cd20 inhibitory antibodies
US20190169294A1 (en) 2016-08-15 2019-06-06 National University Corporation Hokkaido University Anti-lag-3 antibody
WO2018034227A1 (fr) 2016-08-15 2018-02-22 国立大学法人北海道大学 Anticorps anti-lag-3
WO2018033798A1 (fr) 2016-08-17 2018-02-22 Compugen Ltd. Anticorps anti-tigit, anticorps anti-pvrig et combinaisons associées
WO2018054484A1 (fr) * 2016-09-23 2018-03-29 Biontech Ag Anticorps trivalents bispécifiques se liant à claudine 6 ou claudin18.2 et cd3 pour le traitement de maladies cancéreuses exprimant la claudine
WO2018071500A1 (fr) 2016-10-11 2018-04-19 Agenus Inc. Anticorps anti-lag-3 et leurs procédés d'utilisation
WO2018069500A2 (fr) 2016-10-13 2018-04-19 Symphogen A/S Anticorps anti-lag-3 et compositions
WO2018085469A2 (fr) 2016-11-01 2018-05-11 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de lymphocyte t et la protéine 3 de mucine (tim-3)
WO2018083087A2 (fr) 2016-11-02 2018-05-11 Glaxosmithkline Intellectual Property (No.2) Limited Protéines de liaison
WO2018106588A1 (fr) 2016-12-08 2018-06-14 Eli Lilly And Company Anticorps anti-tim-3 pour une combinaison avec des anticorps anti-pd-1
WO2018103501A1 (fr) 2016-12-08 2018-06-14 上海优卡迪生物医药科技有限公司 Arnsi d'inactivation de pd-1 humain, vecteur de car-t d'expression recombinée, procédé de préparation et application associés
WO2018106529A1 (fr) 2016-12-08 2018-06-14 Eli Lilly And Company Anticorps anti-tim-3 pour combinaison avec des anticorps anti-pd-l1
WO2018110555A1 (fr) 2016-12-13 2018-06-21 アステラス製薬株式会社 Anticorps anti-cd73 humain
US20180185482A1 (en) 2017-01-05 2018-07-05 Gensun Biopharma Inc. Checkpoint regulator antagonists
WO2018222711A2 (fr) 2017-05-30 2018-12-06 Bristol-Myers Squibb Company Compositions comprenant une combinaison d'un anticorps anti-lag-3, d'un inhibiteur de voie pd-1 et d'un agent immunothérapeutique
WO2019000146A1 (fr) 2017-06-26 2019-01-03 深圳市博奥康生物科技有限公司 Siarn de récepteur de mort cellulaire programmée humaine 1 et son utilisation
WO2019011855A1 (fr) 2017-07-10 2019-01-17 Innate Pharma Anticorps neutralisant la siglec-9
WO2019011852A1 (fr) 2017-07-10 2019-01-17 Innate Pharma Polythérapie utilisant un anticorps dirigé contre siglec-9 humain et anticorps dirigé contre nkg2a humain pour le traitement du cancer
WO2020025792A1 (fr) * 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Constructions d'anticorps pour cldn18.2 et cd3
WO2021011885A1 (fr) * 2019-07-17 2021-01-21 The Regents Of The University Of California Anticorps dirigés contre la claudine 18 et méthodes de traitement du cancer
WO2021025177A1 (fr) * 2019-08-06 2021-02-11 Astellas Pharma Inc. Polythérapie impliquant des anticorps dirigés contre la claudine 18.2 et inhibiteurs de point de contrôle immunitaire pour le traitement du cancer
WO2022203090A1 (fr) * 2021-03-25 2022-09-29 Astellas Pharma Inc. Polythérapie impliquant des anticorps dirigés contre la claudine 18.2 pour le traitement du cancer
WO2022263508A1 (fr) * 2021-06-15 2022-12-22 Astellas Pharma Europe Bv Agents de liaison bispécifiques se liant à la cldn18.2 et au cd3

Non-Patent Citations (83)

* Cited by examiner, † Cited by third party
Title
"A multilingual glossary of biotechnological terms: (IUPAC Recommendations", 1995, HELVETICA CHIMICA ACTA
"GenBank", Database accession no. NM_000732
"Genbank", Database accession no. NP_001002026
"Monoclonal Antibodies For Cancer Detection And Therapy", 1985, ACADEMIC PRESS, article "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", pages: 303 - 16
ALLARD ET AL., IMMUNOL REV., vol. 276, no. 1, 2018, pages 121 - 144
ALTENHOFER ET AL., CELL MOL LIFE SCIENCES, vol. 69, no. 14, 2012, pages 2327 - 2343
AMDERSEN: "Cancer treatament: the combination of vaccination with other therapies", CANCER IMMUNOLOGY IMMUNOTHERAPY, vol. 57, no. 11, 2008, pages 1735 - 1743, XP019624403
AMON ET AL.: "Monoclonal Antibodies And Cancer Therapy", 1985, ALAN R. LISS, INC., article "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", pages: 243 - 56
ANONYMOUS: "NCT03505320 - A Study to Assess the Antitumor Activity, Safety, Pharmacokinetics and Biomarkers of Zolbetuximab (IMAB362) in Participants With Claudin (CLDN) 18.2 Positive, Metastatic or Advanced Unresectable Gastric and Gastroesophageal Junction (GEJ) Adenocarcinoma", 9 December 2022 (2022-12-09), XP093069794, Retrieved from the Internet <URL:https://classic.clinicaltrials.gov/ct2/history/NCT03505320?A=45&B=45&C=merged#StudyPageTop> [retrieved on 20230802] *
ANONYMOUS: "NCT05365581 - A Study of ASP2138 in Adults With Stomach Cancer or Pancreatic Cancer", CLINICALTRIALS.GOV, 13 December 2022 (2022-12-13), XP093069389, Retrieved from the Internet <URL:https://classic.clinicaltrials.gov/ct2/history/NCT05365581?A=9&B=9&C=merged#StudyPageTop> [retrieved on 20230801] *
ATWELL ET AL., J. MOL. BIOL., vol. 270, 1997, pages 26
BASKAR ET AL.: "Cancer and radiation therapy: current advances and future directions", INT. J MED SCI., vol. 9, no. 3, 2012, pages 193 - 199, XP055668349, DOI: 10.7150/ijms.3635
BECKER ET AL., CANCER RESEARCH, vol. 78, no. 1802226-78-3, 2018, pages 3691 - 3691
BERZOFSKY ET AL.: "Fundamental Immunology", 1984, RAVEN PRESS, article "Antibody-Antigen Interactions"
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BRAY, F. ET AL., CA: A CANCER JOURNAL FOR CLINICIANS, vol. 68, 2018, pages 394 - 424
BRIGNONE ET AL., J. IMMUNOL., vol. 179, 2007, pages 4202 - 4211
BRUHNS ET AL., BLOOD, vol. 113, 2009, pages 3716 - 3725
CHAMES ET AL., BR J PHARMACOL, vol. 157, no. 2, 2009, pages 220 - 233
CHEONG ET AL., EXPERT OPIN THER PAT., vol. 28, no. 2221034-29-1, 2018, pages 317 - 330
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
DALL'ACQUA ET AL., J. BIOL. CHEM., vol. 281, pages 23514 - 24
DALL'ACQUA WF ET AL., J IMMUNOL., vol. 177, no. 2, 2006, pages 1129 - 1138
DANGAJ ET AL., CANCER RESEARCH, vol. 73, 2013, pages 4820 - 9
DAVIS ET AL., IMMUNOL. REV., vol. 190, 2002, pages 123 - 136
EDELMAN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 63, no. 1, 1969, pages 78 - 85
GAUTAM ET AL., MOL CANCER THER, vol. 16, no. 56-92-8, 2017, pages 2144 - 2156
GHETIEWARD, IMMUNOL TODAY., vol. 18, no. 12, 1997, pages 592 - 598
GIANNI ET AL., ACS CHEM BIOL, vol. 5, no. 10, 2010, pages 981 - 93
GUNASEKARAN ET AL., J. BIOL. CHEM., vol. 285, no. 25, 2010, pages 19637
HARDY ET AL., CANCER RES., vol. 54, no. 22, 1994, pages 5793 - 6
HELLSTROM ET AL.: "Controlled Drug Delivery", 1987, MARCEL DEKKER, INC., article "Antibodies For Drug Delivery", pages: 623 - 53
HEZAREH M, J VIROL., vol. 75, no. 24, 2001, pages 12161 - 12168
HINTON ET AL., J. IMMUNOL., vol. 176, 2006, pages 346 - 56
HIROOKA ET AL.: "A combination therapy of gemcitabine with immunotherapy for patients with inoperable locally advanced pancreatic cancer", PANCREAS, vol. 38, no. 3, 2009, pages 69 - 74
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
IGAWA ET AL., PEDS, vol. 23, no. 5, 2010, pages 385 - 392
JEFFERIS ET AL., IMMUNOL. LETT., vol. 82, 2002, pages 57 - 65
JONES, P. ET AL., NATURE, vol. 321, 1986, pages 522 - 525
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NIH PUBLICATION
KAUDER ET AL., PLOS ONE, no. 1673534-76-3, 2019
KIM, C ET AL., ANNALS ONCOL, vol. 29, no. 1218942-37-0, 2018, pages 400 - 441
KRAUS ET AL.: "Recombinant antibodies for cancer therapy", METHODS IN MOLECULAR BIOLOGY
KRYCZEK ET AL., J EXP MED, vol. 203, 2006, pages 871 - 81
LAFRANC ET AL., DEV. COMP. IMMUNOL., vol. 27, no. 1, 2003, pages 55 - 77
LI ET AL., INT J MOL SCI, vol. 17, no. 7, 2016, pages 1151
LINWEISS, JOURNAL OF CELL SCIENCE, vol. 114, 2001, pages 243 - 244
LISETH ET AL.: "Combination of intensive chemotherapy and anticancer vaccines in the treatment of human malignancies: the hematological experience", J BIOMED BIOTECHNOL., 2010, pages 6920979
LIU ET AL., J. PHARMACOL. SCI., vol. 97, no. 7, 2008, pages 2426 - 2447
LU ET AL., BIOCHEM PHARMACOL, vol. 143, no. 1287234-48-3, 2017, pages 25 - 38
LYUBARSKAYA ET AL., ANALYT. BIOCHEM., vol. 348, no. 1, 2006, pages 24 - 39
MARIE-CARDINE ET AL., CANCER, vol. 74, no. 21, 2014, pages 6060 - 70
MERCHANT ET AL., NATURE BIOTECH., vol. 16, 1998, pages 677
NEDDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
NIIMI, MOL. CELL. BIOL., vol. 21, 2001, pages 7380 - 90
PARDOLL, D., NATURE, vol. 12, 2012, pages 252 - 264
PARREN ET AL., J. CLIN INVEST., vol. 90, 1992, pages 1537 - 1546
PEARSONLIPMAN, PROC. NATL ACAD. SCI. USA, vol. 88, 1988, pages 2444
PERROT ET AL., CELL REPORTS, vol. 8, no. 1923833-60-6, 2019, pages 2411 - 2425
POPOLI ET AL., NEUROPSYCHOPHARM, vol. 22, no. 1246018-36-9, 2000, pages 522 - 529
QUEEN, C. ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 86, 1989, pages 10029 - 10033
QUOIX ET AL.: "Therapeutic vaccination with TG4010 and first-line chemotherapy in advanced non-small-cell lung cancer: a controlled phase 2B trial", LANCET ONCOL., vol. 12, no. 12, 2011, pages 1125 - 33, XP055079143, DOI: 10.1016/S1470-2045(11)70259-5
RIDGWAY ET AL., PROTEIN ENGINEERING, vol. 9, no. 7, 1996, pages 617
RIECHMANN, L. ET AL., NATURE, vol. 332, 1998, pages 323 - 327
SASIKUMARRAMACHANDRA, BIODRUGS, vol. 32, no. 5, 2018, pages 481 - 497
SCATCHARD ET AL.: "Ann N.Y.", vol. 51, 1949, ACAD. SCL, pages: 660
SHAABANI ET AL., EXPERT OP THER PAT., vol. 28, no. 9, 2018, pages 665 - 678
SHERIDAN, NAT BIOTECHNOL, vol. 33, 2015, pages 321 - 322
SILLEROMALDONADO, COMPUT. BIOL. MED., vol. 36, no. 2, 2006, pages 157 - 166
SI-YANG ET AL., J. HEMATOL. ONCOL., vol. 70, 2017, pages 136
SI-YANG LIU ET AL., J. HEMATOL. ONCOL., vol. 70, 2007, pages 136
SMITH ET AL., GYNECOL ONCOL, vol. 134, 2014, pages 181 - 189
STASI ET AL., EUROP J PHARM, vol. 761, no. 1202402-40-1, 2015, pages 353 - 361
SUZUKI ET AL., JAP J CLIN ONC, vol. 46, 2016, pages 191 - 203
THORPE ET AL.: "Monoclonal Antibodies '84: Biological And Clinical Applications", 1985, article "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", pages: 475 - 506
THORPE ET AL.: "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", IMMUNOL. REV., vol. 62, 1982, pages 119 - 58, XP001179872, DOI: 10.1111/j.1600-065X.1982.tb00392.x
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WATT ET AL., BLOOD, vol. 98, 2001, pages 1469 - 1479
WEIR ET AL., J BONE MINERAL RES, vol. 11, no. 1029044-16-3, 1996, pages 1474 - 1481
WHERRY, J LEUKOCYTE BIOL, vol. 86, 2009, pages 5 - 8
YAMAMOTO ET AL., BIOL PHARM BULL., vol. 41, no. 3, 2018, pages 419 - 426
ZALEVSKY ET AL., NAT. BIOTECHNOL., vol. 28, 2010, pages 157 - 9
ZHANG ET AL., CELL DISCOV., vol. 3, 2017, pages 17004

Similar Documents

Publication Publication Date Title
US20240158501A1 (en) Bispecific trivalent antibodies binding to claudin6 or claudin18.2 and cd3 for treatment of claudin expressing cancer diseases
AU2022201762A1 (en) IL-15 variants and uses thereof
AU2013347184B2 (en) Agents for treatment of claudin expressing cancer diseases
KR20190015520A (ko) 조합 치료법
US20240209061A1 (en) Fusion proteins and uses thereof
US20240018234A1 (en) Bispecific binding agents binding to cldn18.2 and cd3
EP3766903A2 (fr) Anticorps bispecifiques anti claudin xcd3 pour le traitement de maladies cancéreuses exprimant claudine
WO2022216723A9 (fr) Anticorps bispécifiques ciblant nkp46 et cd38 ainsi que leurs méthodes d&#39;utilisation
WO2024126457A1 (fr) Polythérapie impliquant des agents de liaison bispécifiques se liant à cldn18.2 et cd3 et des inhibiteurs de point de contrôle immunitaire
JP7513854B2 (ja) Cldn18.2及びcd3に結合する二重特異性結合剤
WO2023143478A1 (fr) Nouveaux anticorps bispécifiques anti-cd4 et anti-pd-l1
RU2798988C2 (ru) Биспецифические трехвалентные антитела, связывающиеся с клаудином 6 или клаудином 18.2, и cd3, для лечения онкологических заболеваний с экспрессией клаудина
WO2022216744A2 (fr) Anticorps bispécifiques ciblant nkp46 et gpc3 et leurs méthodes d&#39;utilisation