[go: up one dir, main page]

WO2025169945A1 - Cd3-targeting antigen-binding molecule with improved stability - Google Patents

Cd3-targeting antigen-binding molecule with improved stability

Info

Publication number
WO2025169945A1
WO2025169945A1 PCT/JP2025/003698 JP2025003698W WO2025169945A1 WO 2025169945 A1 WO2025169945 A1 WO 2025169945A1 JP 2025003698 W JP2025003698 W JP 2025003698W WO 2025169945 A1 WO2025169945 A1 WO 2025169945A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
acid sequence
seq
sequence shown
antigen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/JP2025/003698
Other languages
French (fr)
Japanese (ja)
Inventor
直人 谷舗
和英 白井
啓太 森
秀季 滝沢
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chugai Pharmaceutical Co Ltd
Original Assignee
Chugai Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chugai Pharmaceutical Co Ltd filed Critical Chugai Pharmaceutical Co Ltd
Publication of WO2025169945A1 publication Critical patent/WO2025169945A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the present disclosure relates to an antigen-binding molecule comprising a CD3-binding domain with improved stability, a pharmaceutical composition comprising the antigen-binding molecule, and uses thereof.
  • Antibodies are attracting attention as pharmaceuticals due to their high stability in plasma and minimal side effects (Non-Patent Documents 1 and 2). Antibodies not only bind to antigens and have agonistic and antagonistic effects, but are also known to exert antitumor effects against cancer cells by inducing cytotoxic activity (also known as effector function) by effector cells, such as ADCC (Antibody Dependent Cytotoxicity), ADCP (Antibody Dependent Cell phagocytosis), and CDC (Complement-Dependent Cytotoxicity) (Non-Patent Document 3).
  • ADCC is the cytotoxicity exerted by effector cells against antibody-bound target cancer cells when the Fc region of an antibody binds to Fc receptors present on effector cells such as NK cells and macrophages.
  • Natural immunoglobulins bind to antigens through their variable regions, and to receptors such as Fc ⁇ R, FcRn, Fc ⁇ R, and Fc ⁇ R, or to complements, through their constant regions.
  • Normal natural IgG antibodies recognize and bind to a single epitope through their variable region (Fab), and can therefore only bind to a single antigen.
  • Fab variable region
  • Bispecific antibodies which bind to two or more antigens with a single molecule, are being researched as molecules that inhibit multiple targets.
  • IgG antibodies By improving natural IgG antibodies, it is possible to confer binding activity to two different antigens (a first antigen and a second antigen) (Non-Patent Document 4). Therefore, it is possible to neutralize two or more antigens with a single molecule, or to enhance anti-tumor activity by cross-linking cytotoxic cells with cancer cells.
  • T cell-redirecting antibodies a type of bispecific antibody known since the 1980s, are antibodies whose antitumor effect is mediated by cytotoxicity, which recruits T cells as effector cells (Non-Patent Documents 5, 6, 7). Unlike antibodies whose antitumor effect is mediated by ADCC, which recruits NK cells and macrophages as effector cells, T cell-redirecting antibodies are bispecific antibodies that contain an antibody against one of the constituent subunits of the T cell receptor (TCR) complex on T cells, specifically an antibody that binds to the CD3 ⁇ chain, and an antibody that binds to an antigen on the target cancer cell.
  • TCR T cell receptor
  • the simultaneous binding of T cell-redirecting antibodies to the CD3 ⁇ chain and the cancer antigen allows T cells to approach the cancer cells. As a result, it is believed that the cytotoxic activity of T cells exerts an antitumor effect on cancer cells.
  • Catumaxomab a T cell-redirecting antibody, uses two Fab fragments to bind to a cancer antigen (EpCAM) and the CD3 ⁇ chain expressed on T cells.
  • Catumaxomab is a trifunctional antibody that simultaneously binds to both a cancer antigen and CD3 ⁇ , thereby inducing cytotoxic activity by T cells, and to both a cancer antigen and Fc ⁇ R, thereby inducing cytotoxic activity by antigen-presenting cells such as NK cells and macrophages.
  • BiTE bispecific T-cell engager
  • NK cells NK cells, macrophages, etc.
  • BiTE bispecific T-cell engager
  • NK cells NK cells, macrophages, etc.
  • the cancer antigen-independent cytokine induction observed when catumaxomab is administered does not occur.
  • BiTE is a low-molecular-weight modified antibody molecule lacking the Fc region, there is a problem in that the half-life of BiTE administered to patients in the blood is significantly shorter than that of IgG antibodies, which are commonly used as therapeutic antibodies.
  • Patent Document 1 new polypeptide complexes have been provided that use an Fc region with reduced Fc ⁇ R-binding activity to bring T cells into close proximity with target cancer cells, exhibiting excellent safety characteristics by not inducing cytokine storms or other conditions in a cancer antigen-independent manner, and have a long blood half-life. It has been reported that new T cell redirecting antibodies with such advantageous effects are useful not only for targeting cancer cells, but also for targeting regulatory T cells and exhausted T cells, which are cells present in the cancer microenvironment that function to suppress immune responses (Patent Documents 2 and 3).
  • the present invention was made in light of the above circumstances, and focuses particularly on the CD3-binding domain used in T cell redirecting antibodies. It aims to provide a novel CD3-binding domain that has greater stability than the CD3-binding domains used in conventional T cell redirecting antibodies, an antigen-binding molecule containing said CD3-binding domain, a method for producing said antigen-binding molecule, a pharmaceutical composition containing said antigen-binding molecule as an active ingredient, and uses thereof.
  • the present inventors introduced various modifications into the CDRs of the CD3-binding Fab contained in the bispecific antibody described in WO2015174439 and succeeded in obtaining modified antibodies that showed a smaller degree of decline in CD3-binding activity when stored at room temperature or above than the parent Fab.
  • the present inventors further combined these modified CD3-binding domains with other antigen-binding domains, such as a cancer-specific antigen-binding domain, to prepare bispecific antigen-binding molecules and found that the degree of decline in T cell-dependent cytotoxicity when stored at room temperature or above was also small. Based on these findings, the present inventors have demonstrated that the multispecific antigen-binding molecules of the present invention can damage tissues containing target cells expressing a target antigen, such as a cancer-specific antigen.
  • An antigen-binding molecule comprising a first antigen-binding domain and a second antigen-binding domain, wherein the first antigen-binding domain comprises an antibody H chain variable region and L chain variable region that have binding activity to CD3; the heavy chain variable region H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1); an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3), wherein X3 is A or P, and X4 is I, M, or L.
  • L chain CDR1 comprising the amino acid sequence RSX5X6X7VVHENRX8TYLH ( SEQ ID NO: 4 ), wherein X5 is S or T, X6 is Q or M, X7 is S or T, and X8 is Q or N; L chain CDR2 comprising the amino acid sequence KVSNRFS (SEQ ID NO: 5); and L chain CDR3 comprising the amino acid sequence GQGTQVPYT (SEQ ID NO: 6).
  • An antigen-binding molecule comprising: [2] The antigen-binding molecule of [1], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain CDR1, CDR2, and CDR3, and L chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8): (a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6; (a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid
  • [3] The antigen-binding molecule of [1] or [2], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain variable region and L chain variable region selected from the following (a1) to (a8): (a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26; (a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in S
  • [11] A cell comprising the nucleic acid of [9] or the vector of [10].
  • [12] A method for producing the antigen-binding molecule of any one of [1] to [8], comprising the step of culturing the cell of [11].
  • [14] A pharmaceutical composition comprising the antigen-binding molecule of any of [1] to [8] and a pharmaceutically acceptable carrier.
  • [15-1] The pharmaceutical composition according to [14], for inducing cell damage.
  • [15-2] The pharmaceutical composition according to [14], for inducing T cell-dependent cytotoxicity.
  • [15-3] The pharmaceutical composition according to [14], for use in the treatment or prevention of cancer.
  • [16-1] A method for inducing cytotoxicity, comprising the step of administering the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14].
  • [16-2] A method for inducing T cell-dependent cytotoxicity, comprising the step of administering the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14].
  • [16-3] A method for treating or preventing cancer, comprising the step of administering the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14].
  • [17-1] A kit for inducing cytotoxicity, comprising the antigen-binding molecule of any one of [1] to [8] or the pharmaceutical composition of [14], and instructions for use.
  • [17-2] A kit for inducing T cell-dependent cytotoxicity, comprising the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14], and instructions for use.
  • [17-3] A kit for treating or preventing cancer, comprising the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14], and instructions for use.
  • [18-1] The antigen-binding molecule of any one of [1] to [8], for use in inducing cytotoxicity.
  • [18-2] The antigen-binding molecule of any one of [1] to [8], for use in inducing T cell-dependent cytotoxicity.
  • a monovalent antigen-binding molecule comprising a single antigen-binding domain, wherein the single antigen-binding domain comprises an antibody H chain variable region and an L chain variable region that have binding activity to CD3; the heavy chain variable region H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1); an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3), wherein X3 is A or P, and X4 is I, M, or L.
  • An antigen-binding molecule comprising: [A2] The antigen-binding molecule of [A1], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain CDR1, CDR2, and CDR3, and L chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8): (a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6; (a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the
  • [A3] The antigen-binding molecule of [A1] or [A2], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain variable region and L chain variable region selected from the following (a1) to (a8): (a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26; (a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence
  • [A4-1] The antigen-binding molecule of any of [A1] to [A3], which further comprises an Fc region.
  • [A4-2] The antigen-binding molecule of any of [A1] to [A3], further comprising an Fc region with reduced binding activity to an Fc ⁇ receptor.
  • [A4-3] The antigen-binding molecule of [A4-1] or [A4-2], which comprises any one of the combinations of heavy chains and light chains selected from the following (a1) to (a8): (a1) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 30 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35; (a2) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35; (a3) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 36; (a4) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35; (a5) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO:
  • [A5] A nucleic acid encoding the antigen-binding molecule of any of [A1] to [A4].
  • [A6] A vector into which the nucleic acid described in [A5] has been introduced.
  • [A7] A cell comprising the nucleic acid according to [A5] or the vector according to [A6].
  • [A8] A method for producing the antigen-binding molecule of any of [A1] to [A4], comprising the step of culturing the cell of [A7].
  • [A9] An antigen-binding molecule produced by the method described in [A8].
  • antigen-binding molecule refers to a molecule capable of binding to its antigen with sufficient affinity. In one embodiment, the extent of binding of an antigen-binding molecule to proteins unrelated to the antigen is less than about 10% of the binding of the antigen-binding molecule to the antigen, as measured (e.g., by radioimmunoassay (RIA)).
  • RIA radioimmunoassay
  • the antigen-binding molecule has a dissociation constant (Kd) for the antigen of ⁇ 1 ⁇ M, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g., 10 ⁇ 8 M or less, e.g., 10 ⁇ 8 M to 10 ⁇ 13 M, e.g., 10 ⁇ 9 M to 10 ⁇ 13 M).
  • the antigen-binding molecule binds to an epitope of the antigen that is conserved among the antigens from different species.
  • the antigen-binding molecule is an antibody.
  • antibody refers to an immunoglobulin that is natural or partially or fully synthetically produced. Antibodies can be isolated from natural sources such as plasma or serum where they occur, or from the culture supernatant of antibody-producing hybridoma cells, or can be partially or fully synthesized using techniques such as genetic recombination. Suitable examples of antibodies include immunoglobulin isotypes and their isotype subclasses. Nine known classes (isotypes) of human immunoglobulins are IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgD, IgE, and IgM. Of these isotypes, antibodies of the present invention may include IgG1, IgG2, IgG3, and IgG4.
  • An antibody's "class” refers to the type of constant domain or constant region present in its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM. Some of these may be further divided into subclasses (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2.
  • the heavy-chain constant domains corresponding to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • Native antibodies refer to immunoglobulin molecules with a variety of naturally occurring structures.
  • native IgG antibodies are heterotetrameric glycoproteins of approximately 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From the N-terminus to the C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3). Similarly, from the N-terminus to the C-terminus, each light chain has a variable region (VL), also called a variable light domain or light chain variable domain, followed by a constant light (CL) domain.
  • VH variable region
  • VL variable region
  • CL constant light domain
  • the light chains of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of their constant domains.
  • variable region or “variable domain” refer to the domains of an antibody heavy or light chain that are involved in binding the antibody to antigen.
  • the heavy and light chain variable domains (VH and VL, respectively) of natural antibodies typically have a similar structure, with each domain containing four conserved framework regions (FR) and three hypervariable regions (HVR).
  • FR conserved framework regions
  • HVR hypervariable regions
  • antibodies that bind to a particular antigen may be isolated by screening a complementary library of VL or VH domains, respectively, with a VH or VL domain from an antibody that binds to that antigen. See, for example, Portolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
  • hypervariable region refers to each region of an antibody variable domain that is hypervariable in sequence (the “complementarity determining region” or “CDR") and/or forms structurally defined loops (the “hypervariable loops") and/or contains antigen-contacting residues (the “antigen contacts”).
  • CDR complementarity determining region
  • hypervariable loops the region of an antibody variable domain that is hypervariable in sequence
  • antigen contacts the region of antibodies
  • antibodies typically contain six HVRs: three in the VH (H1, H2, H3) and three in the VL (L1, L2, L3).
  • Exemplary HVRs herein include the following: (a) hypervariable loops occurring at amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3) (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987)); (b) CDRs occurring at amino acid residues 24-34 (L1), 50-56 (L2), 89-97 (L3), 31-35b (H1), 50-65 (H2), and 95-102 (H3) (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al., supra.
  • FR Framework or "FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain typically consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences typically appear in VH (or VL) in the following order: FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.
  • Fc region is used herein to define the C-terminal region of an immunoglobulin heavy chain comprising at least a portion of the constant region. This term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226 or from Pro230 to the carboxyl terminus of the heavy chain, except that the C-terminal lysine (Lys447) or glycine-lysine (Gly446-Lys447) residue at the Fc region may or may not be present.
  • the C-terminal lysine (Lys447) or glycine-lysine (Gly446-Lys447) residue at the Fc region may be removed by degradation.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system (also known as the EU index) as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD 1991.
  • a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • Native sequence human Fc regions include native sequence human IgG1 Fc regions (non-A and A allotypes); native sequence human IgG2 Fc regions; native sequence human IgG3 Fc regions; and native sequence human IgG4 Fc regions, as well as naturally occurring variants thereof.
  • a “variant Fc region” comprises an amino acid sequence that differs from that of a native-sequence Fc region by at least one amino acid modification, preferably one or more amino acid substitutions.
  • the variant Fc region has at least one amino acid substitution, e.g., about 1 to about 10 amino acid substitutions, preferably about 1 to about 5 amino acid substitutions, in the native-sequence Fc region or in the Fc region of the parent polypeptide compared to the native-sequence Fc region or the Fc region of the parent polypeptide.
  • the variant Fc region herein preferably has at least about 80% sequence identity with the native-sequence Fc region and/or the Fc region of the parent polypeptide, more preferably at least about 90% sequence identity thereto, and most preferably at least about 95% sequence identity thereto.
  • Fc receptor refers to a receptor that binds to the Fc region of an antibody.
  • the FcR is a native human FcR.
  • the FcR is one that binds IgG antibodies (gamma receptors) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA ("activating receptors") and Fc ⁇ RIIB ("inhibiting receptors”), which have similar amino acid sequences that differ primarily in their cytoplasmic domains.
  • Activating receptor Fc ⁇ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain.
  • ITAM immunoreceptor tyrosine-based activation motif
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • Fc receptor or “FcR” also includes the neonatal receptor FcRn, which is responsible for regulating maternal IgG transfer to the fetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)) and immunoglobulin homeostasis. Methods for measuring binding to FcRn are known (see, e.g., Ghetie and Ward., Immunol. Today 18(12):592-598 (1997); Ghetie et al., Nature Biotechnology, 15(7):637-640 (1997); Hinton et al., J. Biol. Chem. 279(8):6213-6216 (2004); WO2004/92219 (Hinton et al.)).
  • human FcRn high-affinity binding polypeptides can be measured, for example, in transgenic mice or transfected human cell lines expressing human FcRn, or in primates to which polypeptides with mutant Fc regions are administered. Instead of plasma half-life, blood half-life or serum half-life can be measured.
  • WO2000/42072 (Presta) describes antibody variants with increased or decreased binding to FcRs. See also, e.g., Shields et al. J. Biol. Chem. 9(2):6591-6604 (2001).
  • Fc region-containing antibody refers to an antibody that contains an Fc region.
  • the C-terminal lysine (residue 447 according to the EU numbering system) or the C-terminal glycine-lysine (residues 446-447) of the Fc region can be removed, for example, during antibody purification or by recombinant manipulation of the nucleic acid encoding the antibody.
  • composition containing an antibody with an Fc region can include an antibody with G446-K447, an antibody with G446 but no K447, an antibody from which G446-K447 have been completely removed (in the case of IgG, an antibody whose C-terminus is P445 or an antibody with an amide group (NH2) added to P445, resulting in proline amide (Pro-NH2)), or a mixture of the above three types of antibodies.
  • Antibody function refers to a biological activity attributable to the Fc region of an antibody, which varies depending on the antibody isotype.
  • Examples of antibody effector functions include: C1q binding and complement-dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down-regulation of cell surface receptors (e.g., B cell receptors); and B cell activation.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs Fc receptors
  • cytotoxic cells e.g., NK cells, neutrophils, and macrophages
  • NK cells the primary cells mediating ADCC
  • monocytes express Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev.
  • ADCC activity of a molecule of interest may be assessed in vitro in an animal model, such as that disclosed in Clynes et al. PNAS (USA) 95:652-656 (1998).
  • the term "monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies. That is, the individual antibodies comprising the population are identical and/or bind the same epitope (determinant), except for possible variants (e.g., variants containing naturally occurring mutations or variants that arise during the production of a monoclonal antibody preparation; such variants are typically present in small amounts).
  • each monoclonal antibody in a monoclonal antibody preparation is directed against a single epitope on an antigen.
  • monoclonal indicates the character of the antibody as being obtained from a population of substantially homogeneous antibodies, and should not be construed as requiring production of the antibody by any particular method.
  • monoclonal antibodies for use in accordance with the present invention may be produced by a variety of techniques, including, but not limited to, hybridoma technology, recombinant DNA technology, phage display technology, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci.
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • a “humanized” antibody refers to a chimeric antibody that contains amino acid residues from non-human HVRs and human FRs.
  • a humanized antibody contains substantially all of at least one, and typically two, variable domains, in which all or substantially all HVRs (e.g., CDRs) correspond to those of a non-human antibody and all or substantially all FRs correspond to those of a human antibody.
  • a humanized antibody may optionally contain at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of an antibody (e.g., a non-human antibody) refers to an antibody that has undergone humanization.
  • a “human antibody” is an antibody with an amino acid sequence that corresponds to that of an antibody produced by a human or human cell, or an antibody derived from the human antibody repertoire or other non-human source that uses human antibody coding sequences. This definition of a human antibody specifically excludes humanized antibodies, which contain non-human antigen-binding residues.
  • full length antibody “full length antibody,” “complete antibody,” and “whole antibody” are used interchangeably herein and refer to an antibody having a structure substantially similar to that of a native antibody or having a heavy chain that includes an Fc region as defined herein.
  • antibody fragment refers to a molecule other than a complete antibody that contains a portion of the complete antibody that binds to the antigen to which the complete antibody binds.
  • antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabodies; linear antibodies; single-chain antibody molecules (e.g., scFv); and multispecific antigen-binding molecules formed from antibody fragments.
  • Fv variable fragment
  • VL light chain variable region
  • VH heavy chain variable region
  • single-chain antibodies refers to an antibody fragment that contains, in a single polypeptide chain, the variable regions from both the heavy and light chains, but lacks a constant region.
  • single-chain antibodies further comprise a polypeptide linker between the VH and VL domains, which enables them to form the desired structure that may enable antigen binding.
  • Single-chain antibodies are discussed in detail by Plückthun in *The Pharmacology of Monoclonal Antibodies*, Vol. 113, Rosenburg and Moore (eds.), Springer-Verlag, New York, pp. 269-315 (1994). See also International Patent Application Publication No. WO 1988/001649 and U.S. Pat. Nos. 4,946,778 and 5,260,203.
  • single-chain antibodies may also be bispecific and/or humanized.
  • scFv is an antigen-binding domain in which the VH and VL constituting the Fv are linked by a peptide linker (Proc. Natl. Acad. Sci. U.S.A. (1988) 85 (16), 5879-5883).
  • the peptide linker allows the VH and VL to be held in close proximity.
  • sc(Fv) 2 is a single-chain antibody in which four variable regions, two VL and two VH, are linked by a linker such as a peptide linker to form a single chain (J Immunol. Methods (1999) 231 (1-2), 177-189).
  • the two VH and VL may be derived from different monoclonal antibodies. Suitable examples include bispecific sc(Fv) 2s that recognize two different epitopes present in the same antigen, as disclosed in Journal of Immunology (1994) 152 (11), 5368-5374.
  • sc(Fv) 2s can be produced by methods known to those skilled in the art. For example, they can be produced by linking scFvs with a linker such as a peptide linker.
  • the configuration of the antigen-binding domain constituting sc(Fv) 2 includes antigen-binding molecules in which two VHs and two VLs are arranged in the following order, starting from the N-terminus of the single-chain polypeptide: VH, VL, VH, VL ([VH] linker-[VL] linker-[VH] linker-[VL]).
  • the order of the two VHs and two VLs is not limited to the above configuration and may be arranged in any order.
  • sc(Fv) 2 The molecular structure of sc(Fv) 2 is also described in detail in WO2006/132352, and those skilled in the art can prepare the desired sc(Fv) 2 appropriately for producing the antigen-binding molecules disclosed herein based on these descriptions.
  • the antigen-binding molecules of the present invention may also be conjugated to carrier polymers such as PEG or organic compounds such as anticancer drugs. Furthermore, by inserting a glycosylation sequence into the amino acid sequence of the antigen-binding molecules of the present invention, a glycosylation sequence can be suitably added to the antigen-binding molecules in order to obtain the desired effects of the glycosylation.
  • the linker linking the antibody variable regions can be any peptide linker that can be introduced by genetic engineering or a synthetic compound linker (e.g., the linkers disclosed in Protein Engineering, 9 (3), 299-305, 1996). In the present invention, however, a peptide linker is preferred.
  • the length of the peptide linker is not particularly limited and can be selected appropriately by those skilled in the art depending on the purpose. A preferred length is 5 amino acids or more (the upper limit is not particularly limited, but typically 30 amino acids or less, preferably 20 amino acids or less), with 15 amino acids being particularly preferred.
  • sc(Fv) 2 contains three peptide linkers, all of the peptide linkers may be the same length, or peptide linkers of different lengths may be used.
  • a widely known example of a peptide linker is a flexible polypeptide linker composed of glycine and serine. The length and sequence of the peptide linker can be selected appropriately by those skilled in the art depending on the purpose.
  • Synthetic chemical linkers are crosslinkers commonly used for crosslinking peptides, such as N-hydroxysuccinimide (NHS), disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl)suberate (BS3), dithiobis(succinimidyl propionate) (DSP), dithiobis(sulfosuccinimidyl propionate) (DTSSP), ethylene glycol bis(succinimidyl succinate) (EGS), ethylene glycol bis(sulfosuccinimidyl succinate) (sulfo-EGS), disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST), bis[2-(succinimidooxycarbonyloxy)ethyl]sulfone (BSOCOES), and bis[2-(sulfosuccinimidooxycarbonyloxycarbonyloxy
  • linkers When linking four antibody variable regions, three linkers are usually required, but the same linker may be used for all three, or different linkers may be used.
  • Fab consists of one light chain and the CH1 and variable regions of one heavy chain.
  • the heavy chain of a Fab molecule cannot form disulfide bonds with another heavy chain molecule.
  • treating IgG with papain cleaves it upstream of the disulfide bond between the two heavy chains in the hinge region, producing two homologous antibody fragments in which an light chain consisting of a VL (light chain variable region) and a CL (light chain constant region) and an heavy chain fragment consisting of a VH (heavy chain variable region) and a CH ⁇ 1 (the ⁇ 1 region of the heavy chain constant region) are linked by a disulfide bond at their C-terminal regions.
  • These two homologous antibody fragments are each referred to as Fab'.
  • F(ab') 2 comprises two light chains and two heavy chains comprising constant regions, i.e., portions of the CH1 and CH2 domains, such that interchain disulfide bonds are formed between the two heavy chains.
  • the F(ab') 2 constituting the antigen-binding molecule disclosed herein can be suitably obtained by partially digesting a full-length monoclonal antibody or the like having the desired antigen-binding domain with a protease such as pepsin, followed by removal of the Fc fragment by adsorption onto a protein A column.
  • Such a protease is not particularly limited, as long as it can digest a full-length antibody so as to produce F(ab') 2 in a limited manner by appropriately setting the enzyme reaction conditions, such as pH, and examples thereof include pepsin and ficin.
  • Bind refers to the strength of the total non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and the molecule's binding partner (e.g., an antigen).
  • binding affinity refers to the intrinsic binding affinity that reflects a 1:1 interaction between members of a binding pair (e.g., an antibody and an antigen).
  • the affinity of a molecule X for its partner Y can generally be expressed by the dissociation constant (Kd). Affinity can be measured by conventional methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described below.
  • Specific binding refers to the state in which one of the specifically binding molecules does not exhibit any significant binding to any molecules other than the one or more molecules to which it binds.
  • the term is also used when a domain containing an antibody variable region is specific to a particular epitope among multiple epitopes contained in an antigen.
  • an antigen-binding molecule having the domain containing the antibody variable region can bind to various antigens containing that epitope.
  • epitope refers to a site on an antigen to which an antigen-binding molecule (e.g., an antibody), whether proteinaceous or non-proteinaceous, binds.
  • an epitope can be defined by its structure.
  • an epitope can be defined by the binding activity of an antigen-binding molecule (e.g., an antibody) that recognizes the epitope.
  • the antigen is a peptide or polypeptide
  • the epitope can also be identified by the amino acid residues that make up the epitope.
  • the epitope is a glycan
  • the epitope can also be identified by a specific glycan structure.
  • Linear epitopes are epitopes that contain a recognized primary amino acid structure. Linear epitopes typically contain at least three, and most usually at least five, e.g., about 8 to about 10, or 6 to 20 amino acids in a unique sequence.
  • conformational epitopes are epitopes in which the amino acids constituting the epitope are typically not contiguous as a primary structure but are composed of discontinuous amino acid residues (sometimes referred to as discontinuous epitopes). Conformational epitopes may encompass an increased number of amino acids compared to linear epitopes.
  • antigen-binding molecules e.g., antibodies
  • a protein molecule folds to form a three-dimensional structure
  • certain amino acids and/or polypeptide backbones that form a conformational epitope are juxtaposed, allowing antigen-binding molecules (e.g., antibodies) to recognize the epitope.
  • antigen-binding molecules e.g., antibodies
  • Methods for determining the conformational structure of an epitope include, but are not limited to, X-ray crystallography, two-dimensional nuclear magnetic resonance spectroscopy, and site-directed spin labeling and electromagnetic paramagnetic resonance spectroscopy. See, for example, Epitope Mapping Protocols in Methods in Molecular Biology (1996), Vol. 66, Morris (ed.).
  • an antigen-binding molecule e.g., an antibody
  • the structure of an antigen-binding molecule is called the "paratope,” “antigen-binding site,” “antigen-binding region,” or “antigen-binding domain.”
  • the epitope and paratope bind stably due to hydrogen bonds, electrostatic forces, van der Waals forces, hydrophobic bonds, and other forces that act between them.
  • affinity The binding strength between these epitopes and paratopes is called affinity.
  • avidity When multiple antigen-binding molecules (e.g., antibodies) bind to multiple antigens, the sum of their binding strengths is called avidity.
  • an antigen-binding molecule containing multiple antigen-binding domains i.e., a multivalent antigen-binding molecule
  • the affinities act synergistically, resulting in avidity being higher than affinity.
  • an "antigen-binding molecule that competes with a reference antigen-binding molecule” refers to an antigen-binding molecule that inhibits the binding of the reference antigen-binding molecule to its own antigen by 50% or more in a competition assay; conversely, a reference antigen-binding molecule inhibits the binding of the aforementioned antigen-binding molecule to its own antigen by 50% or more in a competition assay.
  • Competitive antigen-binding molecules include antigen-binding molecules that bind to the same or overlapping epitope as the reference antigen-binding molecule, and antigen-binding molecules that bind to an adjacent epitope that is sufficiently proximal to the epitope bound by the reference antigen-binding molecule and cause steric hindrance.
  • An exemplary competition assay is provided herein.
  • an “isolated” antibody is one that has been separated from components of its original environment.
  • the antibody is purified to greater than 95% or 99% purity, as measured, for example, by electrophoresis (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (e.g., ion exchange or reverse-phase HPLC).
  • electrophoresis e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatography e.g., ion exchange or reverse-phase HPLC.
  • Isolated nucleic acid refers to a nucleic acid molecule that has been separated from a component of its original environment. Isolated nucleic acid includes a nucleic acid molecule contained in a cell that normally contains that nucleic acid molecule, but where the nucleic acid molecule is present extrachromosomally or in a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an antigen-binding molecule refers to one or more nucleic acid molecules that encode an antigen-binding molecule, preferably comprising an antibody heavy chain variable region and a light chain variable region, and includes nucleic acid molecules carried on a single vector or separate vectors, and nucleic acid molecules present at one or more locations in a host cell.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived from that cell regardless of the number of passages.
  • the progeny may not be completely identical in nucleic acid content to the parent cell and may contain mutations. Mutant progeny that have the same function or biological activity as that for which the original transformed cell was screened or selected are also included herein.
  • pharmaceutical formulation and “pharmaceutical composition” refer to a preparation in a form that allows the biological activity of the active ingredient contained therein to be effective, and that does not contain additional components that are unacceptably toxic to the subject to whom the formulation is administered.
  • “Pharmaceutically acceptable carrier” refers to any ingredient, other than an active ingredient, in a pharmaceutical formulation or composition that is non-toxic to a subject.
  • Pharmaceutically acceptable carriers include, but are not limited to, buffers, excipients, stabilizers, or preservatives.
  • mammals include, but are not limited to, domestic animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domestic animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • an “effective amount” of an agent refers to the amount, at the dosage and for the period of time necessary, that is effective to achieve the desired therapeutic or prophylactic result.
  • package insert is used to refer to instructions typically included in commercial packaging for therapeutic products that contain information about the indications, usage, dosage, method of administration, concomitant therapy, contraindications, and/or warnings regarding the use of such therapeutic product.
  • treatment refers to a clinical intervention intended to alter the natural course of the individual being treated and may be performed prophylactically or during the course of a clinical condition. Desirable effects of treatment include, but are not limited to, prevention of disease onset or recurrence, alleviation of symptoms, attenuation of any direct or indirect pathological effects of the disease, prevention of metastasis, reduction in the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • the antibodies of the invention are used to delay the onset of disease or slow the progression of disease.
  • cell proliferative disorder and “proliferative disorder” refer to disorders associated with some degree of abnormal cell proliferation.
  • the cell proliferative disorder is cancer.
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation.
  • Examples of cancer include, but are not limited to, carcinoma, lymphoma (e.g., Hodgkin's and non-Hodgkin's lymphoma), blastoma, sarcoma, and leukemia.
  • cancers include squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, cancer of the peritoneum, hepatocellular carcinoma, gastrointestinal cancer, pancreatic cancer, glioma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, liver cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatocellular carcinoma, leukemia and other lymphoproliferative disorders, and various types of head and neck cancer.
  • tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • cancer refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • cancer refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • Inhibiting cell growth or proliferation means reducing cell growth or proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, including inducing cell death.
  • Percent (%) amino acid sequence identity to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical to amino acid residues in the reference polypeptide sequence, after aligning the sequences to achieve the maximum percent sequence identity and introducing gaps, if necessary, and after not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved by a variety of methods within the skill of the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN, Megalign (DNASTAR) software, or GENETYX® (Genetyx Corporation). Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms necessary to achieve maximum alignment over the entire length of the sequences being compared.
  • the ALIGN-2 sequence comparison computer program is the copyright of Genentech, Inc., and its source code, together with user documentation, has been filed with the U.S. Copyright Office, Washington, DC 20559, where it is registered under U.S. Copyright Registration No. TXU510087.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program is compiled for use on UNIX operating systems, including Digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows: 100 times the fraction X/Y, where X is the number of amino acid residues scored by the sequence alignment program ALIGN-2 as identical matches in that program's alignment of A and B, and Y is the total number of amino acid residues in B.
  • the invention of this disclosure generally relates to antigen-binding molecules comprising a CD3-binding domain with excellent stability, pharmaceutical compositions comprising them, and uses thereof.
  • the antigen-binding molecules of the present invention comprise a CD3-binding domain with improved stability compared to the CD3-binding domain contained in conventional T cell-redirecting antibodies (e.g., the bispecific antibodies described in WO2015174439).
  • the structure of the antigen-binding molecules of the present invention is not particularly limited, as long as they comprise the CD3-binding domain of the present invention with improved stability.
  • the antigen-binding molecules may be molecules having a natural antibody structure or polypeptides having other artificially designed structures.
  • the antigen-binding molecule of the present invention is an antibody. In some embodiments, the antigen-binding molecule of the present invention is a monoclonal antibody, including a chimeric, humanized, or human antibody. In one embodiment, the antigen-binding molecule of the present invention is an antibody fragment, such as an Fv, Fab, Fab', scFv, diabody, single-domain antibody, or F(ab') 2 fragment. In another embodiment, the antibody is a full-length antibody, such as a complete IgG1 antibody, a complete IgG2 antibody, a complete IgG3 antibody, or a complete IgG4 antibody.
  • Antigen-binding molecules comprising a CD3-binding domain with excellent stability .
  • the present invention provides antigen-binding molecules comprising a first antigen-binding domain (CD3-binding domain) having binding activity to CD3 and a second antigen-binding domain.
  • the present invention provides monovalent antigen-binding molecules comprising a single antigen-binding domain (CD3-binding domain) having binding activity to CD3.
  • the CD3-binding domain contained in the antigen-binding molecules of the present invention is characterized by excellent stability and a small decrease in CD3-binding activity after storage at room temperature or higher.
  • the CD3-binding activity of the CD3-binding domain contained in the antigen-binding molecules of the present invention after storage at room temperature or higher (e.g., 50°C) for a predetermined period of time (e.g., 3 days) maintains 65% or more, 70% or more, or 75% or more of the CD3-binding activity after storage at a low temperature (e.g., 4°C).
  • the CD3-binding domain contained in the antigen-binding molecules of the present invention has binding activity to the CD3 epsilon chain.
  • the CD3 is mammalian CD3.
  • the CD3 is human CD3.
  • the antigen-binding domain comprised in the antigen-binding molecule of the present invention is Fv, Fab, Fab', or scFv.
  • the CD3-binding domain contained in the antigen-binding molecule of the present invention comprises an antibody H chain variable region and L chain variable region that have binding activity to CD3,
  • the H chain variable region H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1); an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3 ), wherein X3 is A or P, and X4 is I, M, or L.
  • L chain variable region an L chain CDR1 comprising the amino acid sequence RSX5X6X7VVHENRX8TYLH ( SEQ ID NO: 4 ), wherein X5 is S or T, X6 is Q or M, X7 is S or T, and X8 is Q or N; L chain CDR2 comprising the amino acid sequence KVSNRFS (SEQ ID NO: 5); and L chain CDR3 comprising the amino acid sequence GQGTQVPYT (SEQ ID NO: 6).
  • H chain and heavy chain are used interchangeably, and the terms L chain and light chain are used interchangeably.
  • the antibody heavy chain variable region and light chain variable region having binding activity to CD3 comprise any combination of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8): (a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6; (a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ
  • the antibody heavy chain variable region and light chain variable region having binding activity to CD3 comprise any combination of heavy chain variable region and light chain variable region selected from the following (a1) to (a8): (a1) a combination of an H chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 20 and an L chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25; (a2) a combination of a heavy chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 21 and a light chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25; (a3) a combination of a heavy chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 21 and a light chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO:
  • the antibody H chain variable region and L chain variable region having binding activity to CD3 comprise any combination of an H chain variable region and an L chain variable region selected from the following (a1) to (a8): (a1) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 20, and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 25, and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6; (a2)
  • the antibody heavy chain variable region and light chain variable region having binding activity to CD3 comprises any one of the combinations of heavy chain variable region and light chain variable region selected from the following (a1) to (a8): (a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26; (a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25; (a5) a combination of an H-chain variable region
  • the antigen-binding molecule of the present invention comprises, as heavy and light chains having binding activity to CD3, any of the combinations of heavy and light chains selected from the following (a1) to (a8): (a1) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 30 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 35; (a2) a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 31 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 35; (a3) a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 31 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 36; (a4) a combination of a heavy chain having a heavy
  • the antigen-binding molecule of the present invention comprises, as a heavy chain and a light chain having binding activity to CD3, any combination of a heavy chain and a light chain selected from the following (a1) to (a8): (a1) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 30 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 7, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 10, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 35 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6; (a2) a combination of a heavy chain and
  • the antigen-binding molecule of the present invention comprises, as a heavy chain and a light chain having binding activity to CD3, any of the following combinations of heavy chains and light chains selected from (a1) to (a8): (a1) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 30 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35; (a2) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35; (a3) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 36; (a4) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35; (a5) a combination of a heavy chain comprising the amino acid sequence shown
  • the structure of the second antigen-binding domain is not particularly limited, as long as it has binding activity to a target antigen.
  • the second antigen-binding domain comprises an antibody heavy chain variable region and light chain variable region.
  • the antigen-binding molecules of the present invention are monospecific antigen-binding molecules, and both the first and second antigen-binding domains have binding activity to CD3 (e.g., human CD3).
  • the first and second antigen-binding domains have binding activity to the same epitope on CD3.
  • both the first and second antigen-binding domains have binding activity to the CD3 ⁇ chain (e.g., human CD3 ⁇ chain).
  • the first antigen-binding domain and the second antigen-binding domain comprise the same amino acid sequence.
  • the first antigen-binding domain and the second antigen-binding domain comprise different amino acid sequences from each other.
  • the antigen-binding molecules of the present invention are multispecific antigen-binding molecules having binding specificities at at least two different sites.
  • the multispecific antigen-binding molecules of the present invention are bispecific antigen-binding molecules.
  • the multispecific antigen-binding molecules of the present invention are antigen-binding molecules with three or more specificities, such as trispecific, tetraspecific, pentaspecific, or hexaspecific antigen-binding molecules.
  • the first antigen-binding domain has binding activity to CD3, and the second antigen-binding domain has binding activity to an antigen other than CD3.
  • the "antigen other than CD3" recognized and bound by the second antigen-binding domain is preferably a molecule expressed on the surface of cells that are targeted by the cytotoxic activity of T cells (T cell-dependent cytotoxicity), and examples include molecules specifically expressed on the surface of cancer cells (also referred to herein as cancer antigens or cancer-specific antigens) and molecules expressed on the surface of cells that have the function of suppressing immune responses (also referred to herein as immunosuppressive cell surface antigens).
  • the second antigen-binding domain has binding activity to a cancer antigen.
  • the multispecific antigen-binding molecule of the present invention binds to CD3 via the first antigen-binding domain and binds to a cancer antigen via the second antigen-binding domain, thereby bringing T cells into proximity with target cancer cells and enabling the treatment or prevention of cancer through the cytotoxic activity of T cells against tissues containing the target cancer cells.
  • the second antigen-binding domain has binding activity to an immunosuppressive cell surface antigen.
  • the multispecific antigen-binding molecule of the present invention binds to CD3 via the first antigen-binding domain and binds to the immunosuppressive cell surface antigen via the second antigen-binding domain, thereby bringing T cells into proximity with cells that have the function of suppressing immune responses (also referred to herein as immunosuppressive cells), and enabling the treatment or prevention of cancer through the cytotoxic activity of T cells against the immune response-suppressing cells.
  • known antigen-binding domains and other antigen-binding domains including those to be identified in the future, that have binding activity for cancer antigens or immunosuppressive cell surface antigens can be used as the second antigen-binding domain.
  • the multispecific antigen-binding molecule of the present invention has a cytotoxic activity that is equivalent to or greater than that of a control multispecific antigen-binding molecule that comprises, as a first antigen-binding domain having binding activity to CD3, an antibody heavy chain variable region (e.g., an heavy chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20) comprising an heavy chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an heavy chain CDR2 having the amino acid sequence shown in SEQ ID NO: 7, and an heavy chain CDR3 having the amino acid sequence shown in SEQ ID NO: 10, and an antibody light chain variable region (e.g., an light chain variable region comprising the amino acid sequence shown in SEQ ID NO: 24) comprising an light chain CDR1 having the amino acid sequence shown in SEQ ID NO: 14, an light chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an light chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6.
  • an antibody heavy chain variable region
  • multispecific antigen-binding molecules examples include multispecific antigen-binding molecules (e.g., bispecific antigen-binding molecules, e.g., bispecific antibodies) that comprise, as a first antigen-binding domain, the heavy and light chain CDRs, heavy and light chain variable regions, or heavy and light chains of antibody Nos. 2, 14, 25, 29, 30 to 32, and 34 listed in Table 5 below.
  • multispecific antigen-binding molecules e.g., bispecific antigen-binding molecules, e.g., bispecific antibodies
  • the terms "antibody variable region with binding activity to CD3” and “domain containing an antibody variable region with binding activity to CD3” are used interchangeably and refer to a portion of an anti-CD3 antibody comprising a region that specifically binds to and is complementary to part or all of CD3, an adaptor molecule that forms the T cell receptor complex together with the T cell receptor.
  • the domain contains the light chain variable region (VL) and heavy chain variable region (VH) of an anti-CD3 antibody.
  • Suitable examples of such domains include "scFv (single chain Fv),""single chain antibody,””Fv,”” scFv2 (single chain Fv2),””Fab,” and "F(ab') 2 .”
  • the CD3-binding domain of the present invention may bind to any epitope present in the gamma, delta, or epsilon chain sequence that constitutes CD3.
  • a domain containing the light chain variable region (VL) and heavy chain variable region (VH) of an anti-CD3 antibody that binds to an epitope present in the extracellular domain of the epsilon chain of the human CD3 complex is preferably used.
  • the light chain variable region (VL) and heavy chain variable region (VH) of an anti-CD3 antibody described in the Examples are preferably used as such a domain.
  • an appropriate humanized antibody or human antibody is used as the anti-CD3 antibody that is the source of the domain containing an antibody variable region having CD3-binding activity.
  • the polynucleotide sequences of the gamma, delta, and epsilon chains that make up CD3 are registered under RefSeq accession numbers NM_000073.2, NM_000732.4, and NM_000733.3, respectively, and their polypeptide sequences are registered under RefSeq accession numbers NP_000064.1, NP_000723.1, and NP_000724.1, respectively.
  • antibody variable region with binding activity to antigens other than CD3 and “domains comprising antibody variable regions with binding activity to antigens other than CD3” are used interchangeably and refer to portions of antibodies comprising a region that specifically binds to and is complementary to part or all of the antigen (e.g., a cancer antigen; also referred to herein as a cancer-specific antigen).
  • a domain comprising an antibody variable region can be provided by one or more antibody variable domains.
  • a domain comprising an antibody variable region comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).
  • Suitable examples of such domains comprising antibody variable regions include “scFv (single chain Fv),”"single chain antibody,””Fv,”” scFv2 (single chain Fv2),””Fab,” and “F(ab') 2 .”
  • Antigen is limited to including only the epitope to which the antigen-binding domain binds.
  • Suitable examples of antigens include, but are not limited to, peptides, polypeptides, and proteins derived from animals or humans.
  • Suitable examples of antigens used to treat diseases caused by target tissues include, but are not limited to, molecules expressed on the surface of target cells (e.g., cancer cells and inflammatory cells), molecules expressed on the surface of other cells in tissues containing target cells, molecules expressed on the surface of cells that play an immunological role in target cells and tissues containing target cells, and large molecules present in the interstitium of tissues containing target cells.
  • the antigen may be derived from any animal species (e.g., human; or non-human animal, e.g., mouse, rat, hamster, guinea pig, rabbit, monkey, cynomolgus monkey, rhesus monkey, hamadryas baboon, chimpanzee, goat, sheep, dog, horse, pig, cow, or camel), or any bird; preferably, the antigen is derived from human, rabbit, monkey, rat, or mouse.
  • animal species e.g., human
  • non-human animal e.g., mouse, rat, hamster, guinea pig, rabbit, monkey, cynomolgus monkey, rhesus monkey, hamadryas baboon, chimpanzee, goat, sheep, dog, horse, pig, cow, or camel
  • the antigen is derived from human, rabbit, monkey, rat, or mouse.
  • the "antigen” e.g., CD3) recognized and bound by the antigen-binding molecules of the present invention refers to any naturally occurring “antigen” (e.g., CD3) from any vertebrate source, including mammals such as primates (e.g., humans) and rodents (e.g., mice and rats).
  • This term encompasses "full-length” unprocessed “antigen” (e.g., CD3) as well as any form of "antigen” (e.g., CD3) that results from processing within a cell.
  • These terms also encompass naturally occurring variants of the "antigen” (e.g., CD3), such as splice variants and allelic variants.
  • antigens include 17-IA, 4-1BB, 4Dc, 6-keto-PGF1a, 8-iso-PGF2a, 8-oxo-dG, A1 adenosine receptor, A33, ACE, ACE-2, activin, activin A, activin AB, activin B, activin C, activin RIA, activin RIA ALK-2, activin RIB ALK-4, activin RIIA, activin RIIB, ADAM, ADAM10, ADAM12, ADAM15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, addressin, aFGF, ALCAM, ALK, ALK-1, ALK-7, alpha-1-antitrypsin, alpha-V/beta-1 antagonist, ANG, Ang, APAF-1, APE, APJ, APP, APRIL, AR, A RC, ART, Artemin, Anti-Id,
  • integrin alpha V integrin alpha 6 integrin beta 1, integrin beta 2, interferon gamma, IP-10, I-TAC, JE, kallikrein 2, kallikrein 5, kallikrein 6, kallikrein 11, kallikrein 12, kallikrein 14, kallikrein 15, kallikrein L1, kallikrein L2, kallikrein L3, kallikrein L4, KC, KDR, keratinocyte growth factor (KGF), laminin 5, LAMP, LAP, LAP (TGF-1), latent TGF-1, latent TGF-1 bp1, LBP, LDGF, LECT2, Lefty, Lewis-Y antigen, Lewis-Y related antigen,
  • Cancer-specific antigens as non-limiting examples of antigens on target cells refer to antigens expressed by cancer cells that enable the distinction between cancer cells and healthy cells. Examples include antigens expressed as cells become malignant, and abnormal sugar chains that appear on the cell surface or protein molecules when cells become cancerous.
  • ALK pleiotrophin
  • PPN pleiotrophin
  • EpCAM EpCAM
  • CA125 prostatic acid phosphatase
  • PAP prostatic acid phosphatase
  • PSA prostate-specific antigen
  • PSMA TYRP1, HMW-MAA
  • PSMA prostate-specific membrane antigen
  • CEA CEA
  • MUC1 MUC1
  • HMFG1 TAG-72
  • GICA GICA (CA19-9)
  • NY-ESO-1 LEA, CD15, CD17, CD19, CD20, CD22, CD30, CD33, CD38, CD77, CD79b, CD147, CD228, GD2, GD3, GM2, GM3, TSTA, and virus-induced tumors.
  • Antigens e.g., envelope antigens of DNA tumor viruses and RNA tumor viruses), alpha-fetoprotein (AFP), 5T4, differentiation antigens (e.g., L6 and L20 antigens), CD165, EGFR, ANKRD17, ErbB2, APO-1, SSEA-1, SCP-1, LeY, oligosaccharide antigens, SSEA-3, SSEA-4, CTAGE1, MART-1, sialyl Tn (STn), NY-CO-45, NY-LU-12, ART1, MA2, NOVA2, TSPAN8, MAGE-C1, MAGE-B1, MAGE-B2, MAGE-4A, Examples of polypeptides include MAGE-X2, YKL-40, EREG, CA15-3, CLEC12A, Nectin4, Trop2, BCMA, Tissue factor, FR ⁇ (FOLR1), ErbB3, Claudin18 (Claudin18.2), B7-H3 (CD276), MET, PSCA, PTK7,
  • Specific examples of molecules expressed on the surface of cells that have the function of suppressing these immune responses include CTLA4, PD1, TIM3, LAG-3, CD244 (2B4), CD160, GARP, OX40, CD137 (4-1BB), CD25, VISTA, VISATA, BTLA, TNFR25, CD57, KLRG1, CCR2, CCR5, CCR6, CD39, CD73, CD4, CD18, CD49b, CD1d, CD5, CD21, TIM1, CD19, CD20, CD23, CD24, CD38, CD93, IgM, B220 (CD45R), CD317, PD-L1, CD11b, Ly6G, ICAM-1, FAP, PDGFR, podoplanin, and TIGIT.
  • preferred target molecules for the binding domains of the present invention include, for example, CTLA4, TIM3, LAG3, CD137 (4-1BB), CD25, CCR5, CCR6, CD38, and TIGIT, which are cell surface molecules specifically expressed on cell fractions (CD4 + , CD25 high , CD45RA - ) that have been reported to have high immune response suppression function.
  • Preferred target molecules for the binding domains of the present invention include, in particular, CTLA4, LAG3, and OX40.
  • Antigen-binding activity Methods for confirming epitope binding by test antigen-binding molecules having domains containing antibody variable regions with binding activity to CD3 are exemplified below. Methods for confirming epitope binding by test antigen-binding molecules having domains containing antibody variable regions with binding activity to other antigens can also be appropriately performed in accordance with the examples below.
  • a test antigen-binding molecule containing a domain comprising an antibody variable region with binding activity to CD3 recognizes a linear epitope present in the CD3 molecule can be confirmed, for example, as follows.
  • a linear peptide consisting of the amino acid sequence constituting the extracellular domain of CD3 is synthesized.
  • This peptide can be chemically synthesized.
  • it can be obtained by genetic engineering techniques using a region in CD3 cDNA that encodes the amino acid sequence corresponding to the extracellular domain.
  • the binding activity of the linear peptide consisting of the amino acid sequence constituting the extracellular domain to a test antigen-binding molecule containing a domain comprising an antibody variable region with binding activity to CD3 is assessed.
  • the binding activity of the antigen-binding molecule to the peptide can be assessed by ELISA using immobilized linear peptide as the antigen.
  • the binding activity of the antigen-binding molecule to the linear peptide can be determined based on the level of inhibition by the linear peptide of binding of the antigen-binding molecule to CD3-expressing cells. These tests can determine the binding activity of the antigen-binding molecule to the linear peptide.
  • test antigen-binding molecule having a domain containing an antibody variable region that has binding activity to CD3 recognizes a conformational epitope
  • CD3-expressing cells are prepared. Examples of such cases include when a test antigen-binding molecule having a domain containing an antibody variable region that has binding activity to CD3 binds strongly to CD3-expressing cells upon contact with the cells, but does not substantially bind to a linear peptide consisting of the amino acid sequence that constitutes the extracellular domain of immobilized CD3.
  • “not substantially binding” refers to a binding activity that is 80% or less, typically 50% or less, preferably 30% or less, and particularly preferably 15% or less of the binding activity toward human CD3-expressing cells.
  • Methods for measuring the binding activity of a test antigen-binding molecule containing a CD3 antigen-binding domain to CD3-expressing cells include, for example, the method described in Antibodies: A Laboratory Manual (Ed Harlow, David Lane, Cold Spring Harbor Laboratory (1988) 359-420). Specifically, evaluation can be performed using ELISA or FACS (fluorescence activated cell sorting) principles, using CD3-expressing cells as antigens.
  • the binding activity of a test antigen-binding molecule containing a CD3 antigen-binding domain for CD3-expressing cells is quantitatively assessed by comparing the signal levels generated by the enzymatic reaction. That is, the test antigen-binding molecule is added to an ELISA plate on which CD3-expressing cells have been immobilized, and the test antigen-binding molecule bound to the cells is detected using an enzyme-labeled antibody that recognizes the test antigen-binding molecule.
  • test antigen-binding molecule a dilution series of the test antigen-binding molecule is prepared, and the antibody binding titer for CD3-expressing cells is determined, allowing the binding activity of the test antigen-binding molecule for CD3-expressing cells to be compared.
  • the binding of a test antigen-binding molecule to an antigen expressed on the surface of cells suspended in a buffer solution or the like can be detected using a flow cytometer.
  • Known flow cytometers include, for example, the following: FACSCanto TM II FACSAria TM FACSArray TM FACSVantage TM SE FACSCalibur TM (both are trade names of BD Biosciences) EPICS ALTRA HyperSort Cytomics FC 500 EPICS XL-MCL ADC EPICS XL ADC Cell Lab Quanta / Cell Lab Quanta SC (both are trade names of Beckman Coulter)
  • the following method is an example of a suitable method for measuring the antigen-binding activity of a test antigen-binding molecule containing a CD3 antigen-binding domain.
  • the test antigen-binding molecule is reacted with CD3-expressing cells and stained with an FITC-labeled secondary antibody that recognizes the test antigen-binding molecule.
  • the test antigen-binding molecule is diluted with an appropriate buffer solution to prepare the complex at the desired concentration. For example, it can be used at a concentration between 10 ⁇ g/ml and 10 ng/ml.
  • the fluorescence intensity and cell count are measured using a FACSCalibur (BD).
  • the amount of antibody binding to the cells is reflected in the fluorescence intensity, i.e., the Geometric Mean value, obtained by analysis using CELL QUEST Software (BD).
  • the Geometric Mean value obtained by analysis using CELL QUEST Software (BD).
  • test antigen-binding molecule containing a CD3 antigen-binding domain shares an epitope with another antigen-binding molecule can be confirmed by competition between the two for the same epitope. Competition between antigen-binding molecules can be detected by cross-blocking assays, for example.
  • a competitive ELISA assay is a preferred cross-blocking assay.
  • the amount of test antigen-binding molecules bound to the wells via CD3 protein can be easily measured by labeling the antigen-binding molecules in advance.
  • biotin-labeled antigen-binding molecules can be measured using an avidin-peroxidase conjugate and an appropriate substrate.
  • Cross-blocking assays using enzyme labels such as peroxidase are specifically referred to as competitive ELISA assays.
  • Antigen-binding molecules can also be labeled with other detectable or measurable labeling substances. Specific examples include radiolabels and fluorescent labels.
  • a competitor antigen-binding molecule can block the binding of a test antigen-binding molecule comprising an antigen-binding domain to CD3 by at least 20%, preferably at least 20-50%, and more preferably at least 50%, compared to the binding activity obtained in a control test performed in the absence of the candidate competitor antigen-binding molecule, the test antigen-binding molecule binds to substantially the same epitope as the competitor antigen-binding molecule, or is an antigen-binding molecule that competes for binding to the same epitope.
  • test antigen-binding molecule containing a CD3 antigen-binding domain binds whether the test antigen-binding molecule and a control antigen-binding molecule share the same epitope can be assessed by comparing the binding activity of both antigen-binding molecules to a peptide in which amino acid mutations have been introduced into the peptide constituting the epitope.
  • binding activity can be measured, for example, by comparing the binding activity of test and control antigen-binding molecules to a mutated linear peptide in the ELISA format described above.
  • the binding activity to the mutant peptide bound to a column can also be measured by passing the test and control antigen-binding molecules down the column and then quantifying the antigen-binding molecules eluted in the eluate.
  • Methods for adsorbing mutant peptides to a column, for example as fusion peptides with GST, are known.
  • the identified epitope is a conformational epitope
  • whether the test and control antigen-binding molecules share the same epitope can be assessed using the following method.
  • CD3-expressing cells and cells expressing CD3 with an epitope mutation introduced are prepared. These cells are suspended in an appropriate buffer solution such as PBS, and the test and control antigen-binding molecules are added to the cell suspension.
  • an FITC-labeled antibody that can recognize the test and control antigen-binding molecules is added to the cell suspension after washing with an appropriate buffer solution.
  • the fluorescence intensity and cell count of cells stained with the labeled antibody are measured using a FACSCalibur (BD).
  • the test and control antigen-binding molecules are diluted with an appropriate buffer solution to the desired concentration before use.
  • the amount of labeled antibody bound to the cells is reflected in the fluorescence intensity, i.e., the geometric mean value, obtained by analysis using CELLQUEST Software (BD).
  • BD CELLQUEST Software
  • substantially no binding to mutant CD3-expressing cells can be determined by the following method. First, test and control antigen-binding molecules bound to cells expressing mutant CD3 are stained with a labeled antibody. The fluorescence intensity of the cells is then detected. When a FACSCalibur is used for flow cytometry to detect fluorescence, the obtained fluorescence intensity can be analyzed using CELL QUEST Software. The percentage increase in fluorescence intensity due to antigen-binding molecule binding can be determined by calculating the comparative value ( ⁇ Geo-Mean) from the Geometric Mean values in the presence and absence of the antigen-binding molecule using the formula below.
  • Geo-Mean Geo-Mean (in the presence of antigen-binding molecules) / Geo-Mean (in the absence of antigen-binding molecules)
  • the Geometric Mean comparison value (mutant CD3 molecule ⁇ Geo-Mean value) obtained by analysis, which reflects the binding amount of the test antigen-binding molecule to mutant CD3-expressing cells, is compared with the ⁇ Geo-Mean comparison value, which reflects the binding amount of the test antigen-binding molecule to CD3-expressing cells.
  • concentrations of the test antigen-binding molecule used to determine the ⁇ Geo-Mean comparison values for mutant CD3-expressing cells and CD3-expressing cells are adjusted to be identical or substantially identical to each other.
  • An antigen-binding molecule that has been previously confirmed to recognize an epitope in CD3 is used as a control antigen-binding molecule.
  • the antigen-binding molecule is deemed to "not substantially bind to mutant CD3-expressing cells."
  • the formula for calculating the Geo-Mean value is described in the CELL QUEST Software User's Guide (BD biosciences). If the comparison values are substantially equivalent, the epitopes of the test antigen-binding molecule and the control antigen-binding molecule can be determined to be identical.
  • Antigen-binding molecules containing an FcRn-binding domain contained in the Fc region Preferred examples of the antigen-binding molecules of the present invention include antigen-binding molecules containing an FcRn-binding domain contained in the Fc region of an antibody.
  • a well-known method for extending the blood half-life of a protein administered to the body involves adding an antibody FcRn-binding domain to the protein of interest and utilizing the FcRn-mediated recycling function.
  • FcRn-binding domain is not particularly limited as long as it has binding activity to FcRn, and may be a domain that directly binds to FcRn or a domain that indirectly binds to FcRn.
  • domains that directly bind to FcRn include the variable region of an antibody whose antigen is FcRn, Fab, the Fc region of an antibody, fragments thereof, albumin, albumin domain 3, human serum albumin (HSA), and transferrin.
  • domains that indirectly bind to FcRn include domains that have binding activity to the above-mentioned domains that directly bind to FcRn.
  • an FcRn-binding domain is the Fc region of an antibody, or a fragment of the Fc region that contains the FcRn-binding region.
  • Fc region include IgA1, IgA2, IgD, IgE, IgG1, IgG2, IgG3, IgG4, and IgM Fc regions, and an Fc region derived from a native IgG can be used.
  • Native IgG refers to a polypeptide that includes the same amino acid sequence as IgG found in nature and belongs to the class of antibodies substantially encoded by immunoglobulin gamma genes.
  • native human IgG refers to native human IgG1, native human IgG2, native human IgG3, native human IgG4, etc.
  • Native IgG also includes naturally occurring variants thereof.
  • human IgG1, human IgG2, human IgG3, and human IgG4 antibodies multiple allotype sequences due to genetic polymorphisms are described in Sequences of proteins of immunological interest, NIH Publication No. 91-3242, and any of these may be used in the present invention.
  • the amino acid sequence at EU numbering positions 356 to 358 may be DEL or EEM.
  • the antigen-binding molecules of the present invention comprise an antibody Fc region.
  • the antigen-binding molecules of the present invention comprise a single antigen-binding domain
  • the antigen-binding domain may be linked to one of the two polypeptides constituting the dimeric Fc region.
  • the antigen-binding molecules of the present invention comprise a first antigen-binding domain and a second antigen-binding domain
  • the first antigen-binding domain may be linked to one of the two polypeptides constituting the dimeric Fc region
  • the second antigen-binding domain may be linked to the other, but this is not a limitation.
  • the antigen-binding molecules of the present invention comprise an antibody Fc region with reduced Fc ⁇ receptor-binding activity.
  • the multispecific antigen-binding molecules of the present invention comprise an antibody Fc region
  • Various amino acid mutations (modifications) in the Fc region of an antibody that result in reduced binding activity to Fc ⁇ receptors are known, and Fc regions bearing such known amino acid mutations (modifications) or other mutations (modifications) that will be identified in the future can be used in the antigen-binding molecules of the present invention.
  • the strength of the effector function acting through the binding of the antibody Fc region to the Fc ⁇ receptor varies depending on the IgG subclass, and in humans, it is known to be high in IgG1 and IgG3 and low in IgG2 and IgG4. Therefore, native Fc regions of human IgG2 or IgG4 can also be used as the Fc region of an antibody with reduced binding activity to Fc ⁇ receptors that is included in the antigen-binding molecules of the present invention.
  • Fc ⁇ Receptor refers to a receptor that can bind to the Fc region of an IgG1, IgG2, IgG3, or IgG4 monoclonal antibody, and refers to any member of a family of proteins substantially encoded by the Fc ⁇ receptor gene.
  • this family includes, but is not limited to, Fc ⁇ RI (CD64), which includes the isoforms Fc ⁇ RIa, Fc ⁇ RIb, and Fc ⁇ RIc; Fc ⁇ RII (CD32), which includes the isoforms Fc ⁇ RIIa (including allotypes H131 and R131), Fc ⁇ RIIb (including Fc ⁇ RIIb-1 and Fc ⁇ RIIb-2), and Fc ⁇ RIIc; and Fc ⁇ RIII (CD16), which includes the isoforms Fc ⁇ RIIIa (including allotypes V158 and F158) and Fc ⁇ RIIIb (including allotypes Fc ⁇ RIIIb-NA1 and Fc ⁇ RIIIb-NA2), as well as any unidentified human Fc ⁇ Rs or Fc ⁇ R isoforms or allotypes.
  • Fc ⁇ RI CD64
  • Fc ⁇ RII CD32
  • Fc ⁇ RIIa including allotypes H131 and R131
  • Fc ⁇ RIIb including Fc ⁇ RI
  • Fc ⁇ Rs may be derived from any organism, including, but not limited to, humans, mice, rats, rabbits, and monkeys.
  • Mouse Fc ⁇ Rs include, but are not limited to, Fc ⁇ RI (CD64), Fc ⁇ RII (CD32), Fc ⁇ RIII (CD16), and Fc ⁇ RIII-2 (CD16-2), as well as any unidentified mouse Fc ⁇ Rs or Fc ⁇ R isoforms or allotypes.
  • Preferred examples of such Fc ⁇ receptors include human Fc ⁇ RI (CD64), Fc ⁇ RIIA (CD32), Fc ⁇ RIIB (CD32), Fc ⁇ RIIIA (CD16), and/or Fc ⁇ RIIIB (CD16).
  • the polynucleotide and amino acid sequences of Fc ⁇ RI are registered under RefSeq accession numbers NM_000566.3 and NP_000557.1, respectively; the polynucleotide and amino acid sequences of Fc ⁇ RIIA are registered under RefSeq accession numbers BC020823.1 and AAH20823.1, respectively; the polynucleotide and amino acid sequences of Fc ⁇ RIIB are registered under RefSeq accession numbers BC146678.1 and AAI46679.1, respectively; the polynucleotide and amino acid sequences of Fc ⁇ RIIIA are registered under RefSeq accession numbers BC033678.1 and AAH33678.1, respectively; and the polynucleotide and amino acid sequences of Fc ⁇ RIIIB are registered under RefSeq accession numbers BC128562.1 and AAI28563.1, respectively.
  • an Fc ⁇ receptor has binding activity to the Fc region of an IgG1, IgG2, IgG3, or IgG4 monoclonal antibody can be confirmed by the FACS or ELISA formats described above, as well as by ALPHA screen (Amplified Luminescent Proximity Homogeneous Assay) and the BIACORE method using surface plasmon resonance (SPR) (Proc. Natl. Acad. Sci. USA (2006) 103 (11), 4005-4010).
  • Fc ⁇ Receptor-Binding Activity Reduced binding activity of an Fc region to any of the Fc ⁇ receptors Fc ⁇ I, Fc ⁇ IIA, Fc ⁇ IIB, Fc ⁇ IIIA, and/or Fc ⁇ IIIB can be confirmed by the FACS and ELISA formats described above, as well as ALPHA screen (Amplified Luminescent Proximity Homogeneous Assay) and the BIACORE method using surface plasmon resonance (SPR) (Proc. Natl. Acad. Sci. USA (2006) 103 (11), 4005-4010).
  • the ALPHA screen is performed using ALPHA technology, which uses two beads, donor and acceptor, based on the following principle: A luminescent signal is detected only when a molecule bound to the donor bead biologically interacts with a molecule bound to the acceptor bead and the two beads are in close proximity. A photosensitizer inside the donor bead, excited by a laser, converts surrounding oxygen into excited singlet oxygen. The singlet oxygen diffuses around the donor bead and, when it reaches a nearby acceptor bead, triggers a chemiluminescent reaction within the bead, ultimately resulting in the emission of light. If the molecules bound to the donor bead and the molecules bound to the acceptor bead do not interact, the singlet oxygen produced by the donor bead does not reach the acceptor bead, and no chemiluminescent reaction occurs.
  • biotin-labeled antigen-binding molecules are bound to donor beads, and glutathione S-transferase (GST)-tagged Fc ⁇ receptors are bound to acceptor beads.
  • GST glutathione S-transferase
  • antigen-binding molecules with wild-type Fc regions interact with Fc ⁇ receptors, generating a signal at 520-620 nm.
  • Antigen-binding molecules with untagged mutant Fc regions compete with the interaction between antigen-binding molecules with wild-type Fc regions and Fc ⁇ receptors. Relative binding affinity can be determined by quantifying the decrease in fluorescence that occurs as a result of competition.
  • Methods for tagging Fc ⁇ receptors with GST include expressing a fusion gene in which a polynucleotide encoding the Fc ⁇ receptor and a polynucleotide encoding GST are fused in-frame in cells harboring an expression vector, followed by purification using a glutathione column.
  • the resulting signals are suitably analyzed by fitting them to a one-site competition model using nonlinear regression analysis, for example, using software such as GRAPHPAD PRISM (GraphPad, San Diego).
  • One of the substances (ligand) whose interaction is to be observed is immobilized on the gold film of a sensor chip.
  • SPR signal reflection intensity
  • the other substance (analyte) whose interaction is to be observed is poured over the surface of the sensor chip and the ligand and analyte bind, the mass of the immobilized ligand molecule increases, changing the refractive index of the solvent on the sensor chip surface. This change in refractive index shifts the position of the SPR signal (conversely, when the bond dissociates, the signal position returns).
  • the Biacore system plots the amount of shift, i.e., the change in mass on the sensor chip surface, on the vertical axis, and displays the change in mass over time as measurement data (sensorgram).
  • the kinetics binding rate constant (ka) and dissociation rate constant (kd)
  • affinity KD
  • Inhibition measurement methods are also suitable for use with the BIACORE method. An example of an inhibition assay is described in Proc. Natl. Acad. Sci. USA (2006) 103 (11), 4005-4010.
  • Antigen-binding molecules having the Fc region of an IgG1, IgG2, IgG3, or IgG4 monoclonal antibody can be used as a control, as appropriate.
  • These Fc regions may have structures in which an A is added to the N-terminus of the amino acid sequences registered under RefSeq accession numbers AAC82527.1, AAB59393.1, CAA27268.1, and AAB59394.1, respectively.
  • an antigen-binding molecule having a mutant Fc region of an antibody of a certain isotype when used as a test substance, the effect of the mutation in the mutant on the binding activity to Fc ⁇ receptors can be verified by using an antigen-binding molecule having the Fc region of the antibody of that specific isotype as a control. As described above, antigen-binding molecules having Fc region mutants verified to have reduced binding activity to Fc ⁇ receptors can be appropriately prepared.
  • the Fc region of an antibody of a certain isotype is compared with the Fc region of an antibody of another certain isotype in terms of Fc ⁇ receptor-binding activity, and the Fc region with lower Fc ⁇ receptor-binding activity can be used in the antigen-binding molecules of the present invention.
  • amino acid mutations can be introduced into such an Fc region with low Fc ⁇ receptor-binding activity to obtain an Fc region with even lower Fc ⁇ receptor-binding activity.
  • Such mutations in the Fc region include deletion of amino acids 231A-238S, identified according to EU numbering (WO 2009/011941), C226S, C229S, P238S, (C220S) (J. Rheumatol (2007) 34, 11), C226S, C229S (Hum. Antibod. Hybridomas (1990) 1(1), 47-54), and C226S, C229S, E233P, L234V, L235A (Blood (2007) 109, 1185-1192).
  • antigen-binding molecules include those having an Fc region in which any of the following amino acids, as specified by EU numbering, have been substituted among the amino acids constituting the Fc region of an antibody of a specific isotype: 220, 226, 229, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 264, 265, 266, 267, 269, 270, 295, 296, 297, 298, 299, 300, 325, 327, 328, 329, 330, 331, or 332.
  • the antibody isotype from which the Fc region originates is not particularly limited, and Fc regions derived from IgG1, IgG2, IgG3, or IgG4 monoclonal antibodies can be used as appropriate.
  • any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG1 antibody (the numbers indicate the positions of the amino acid residues specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution): (a) L234F, L235E, P331S, (b) C226S, C229S, P238S, (c) C226S, C229S, (d) C226S, C229S, E233P, L234V, L235A (e) L234A, L235A or L235R, N297A (f) L235A or L235R, S239K, N297A Alternatively, antigen-binding molecules having an Fc region in which the amino acid sequence at positions 231 to 238 has been deleted may also be used as appropriate.
  • any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG3 antibody (the numbers indicate the amino acid residue positions specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution): (m) F241A (n) D265A (o) V264A Antigen-binding molecules having an Fc region modified with ⁇ -glucan may also be used appropriately.
  • any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG4 antibody (the numbers indicate the amino acid residue positions specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution): (p) L235A, G237A, E318A (q) L235E (r) F234A, L235A Antigen-binding molecules having an Fc region modified with ⁇ -glucan may also be used appropriately.
  • antigen-binding molecules having an Fc region in which any of the following amino acids, identified according to EU numbering, that constitute the Fc region of an IgG1 antibody are substituted with amino acids corresponding to the corresponding EU numbering in the corresponding IgG2 or IgG4: 233, 234, 235, 236, 237, 327, 330, and 331.
  • the type of amino acid present after substitution is not particularly limited, but antigen-binding molecules having an Fc region in which one or more of the amino acids at positions 234, 235, and 297 have been substituted with alanine are particularly preferred.
  • Another preferred example is an antigen-binding molecule having an Fc region in which the amino acid at position 265, as specified by EU numbering, among the amino acids constituting the Fc region of an IgG1 antibody, has been substituted with another amino acid.
  • the type of amino acid present after substitution is not particularly limited, but antigen-binding molecules having an Fc region in which the amino acid at position 265 has been substituted with alanine are particularly preferred.
  • the antigen-binding molecules of the present invention comprise an Fc region with reduced binding activity to Fc ⁇ receptors.
  • Fc ⁇ receptors include antibodies with reduced effector functions attributable to the Fc region, which are desirable when certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • antibodies with reduced effector functions include those with one or more substitutions at Fc region residues 238, 265, 269, 270, 297, 327, and 329 (U.S. Patent No. 6,737,056).
  • Such Fc variants include Fc variants with two or more substitutions at amino acid positions 265, 269, 270, 297, and 327, including the so-called "DANA" Fc variant with substitutions of residues 265 and 297 to alanine (U.S. Patent No. 7,332,581).
  • One preferred embodiment of the antigen-binding molecule of the present invention is an antibody comprising the variable region of the antibody of the present invention.
  • Monoclonal antibodies derived from mammals are preferably produced as antibodies of the present invention.
  • Mammalian-derived monoclonal antibodies include those produced by hybridomas and those produced by host cells transformed by genetic engineering techniques with an expression vector containing an antibody gene.
  • the mammal to be immunized to obtain antibodies is not limited to a specific animal, but it is preferable to select it taking into consideration its compatibility with the parent cells used in cell fusion to produce hybridomas.
  • rodents such as mice, rats, hamsters, rabbits, and monkeys are preferably used.
  • the above-mentioned animals are immunized with the sensitizing antigen according to known methods.
  • a common method is to administer the sensitizing antigen to the mammal by intraperitoneal or subcutaneous injection.
  • the sensitizing antigen is diluted at an appropriate dilution ratio with PBS (Phosphate-Buffered Saline) or physiological saline, and if desired, mixed with a conventional adjuvant, such as Freund's complete adjuvant, and emulsified.
  • the sensitizing antigen is then administered to the mammal several times every 4 to 21 days.
  • An appropriate carrier may also be used when immunizing with the sensitizing antigen.
  • sensitizing antigen when a partial peptide with a small molecular weight is used as the sensitizing antigen, it may be desirable to immunize with the sensitizing antigen peptide bound to a carrier protein such as albumin or keyhole limpet hemocyanin.
  • a carrier protein such as albumin or keyhole limpet hemocyanin.
  • Hybridomas producing the desired antibodies can also be prepared using DNA immunization as follows.
  • DNA immunization is an immunization method in which a vector DNA constructed in such a manner that a gene encoding an antigen protein can be expressed in the immunized animal is administered to the immunized animal, and a sensitizing antigen is expressed in the immunized animal's body, thereby conferring immune stimulation.
  • DNA immunization is expected to have the following advantages: - Immunostimulation can be achieved by maintaining the structure of membrane proteins - No need to purify the immunizing antigen
  • DNA that expresses the antigen protein is administered to the animal to be immunized.
  • DNA encoding the antigen protein can be synthesized by known methods such as PCR.
  • the obtained DNA is inserted into an appropriate expression vector and administered to the animal to be immunized.
  • expression vector commercially available expression vectors such as pcDNA3.1 can be suitably used. Commonly used methods can be used to administer the vector into the living body. For example, DNA immunization is performed by introducing gold particles to which the expression vector has been adsorbed into the cells of the animal to be immunized using a gene gun.
  • immune cells are collected from the mammal and subjected to cell fusion. Splenocytes, in particular, are preferred as immune cells.
  • Mammalian myeloma cells are used as the cells to be fused with the immune cells. It is preferable that the myeloma cells have an appropriate selection marker for screening.
  • a selection marker refers to a trait that allows (or prevents) survival under specific culture conditions.
  • Known selection markers include hypoxanthine-guanine-phosphoribosyltransferase deficiency (hereinafter abbreviated as HGPRT deficiency) and thymidine kinase deficiency (hereinafter abbreviated as TK deficiency).
  • HGPRT deficiency hypoxanthine-guanine-phosphoribosyltransferase deficiency
  • TK deficiency thymidine kinase deficiency
  • Cells that are HGPRT or TK deficient are hypoxanthine-aminopterin-thymidine sensitive (hereinafter abbreviated as HAT sensitive).
  • HGPRT-deficient or TK-deficient cells can be selected on media containing 6-thioguanine, 8-azaguanine (hereafter abbreviated as 8AG), or 5'-bromodeoxyuridine, respectively.
  • 8AG 8-azaguanine
  • 5'-bromodeoxyuridine normal cells that incorporate these pyrimidine analogs into their DNA die.
  • G418 resistance Another selection marker, called G418 resistance, confers resistance to 2-deoxystreptamine antibiotics (gentamicin analogs) via the neomycin resistance gene.
  • G418 resistance confers resistance to 2-deoxystreptamine antibiotics (gentamicin analogs) via the neomycin resistance gene.
  • gentamicin analogs gentamicin analogs
  • myeloma cell lines suitable for cell fusion are known.
  • Such myeloma cells include, for example, P3 (P3x63Ag8.653) (J. Immunol. (1979) 123 (4), 1548-1550), P3x63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (C. Eur. J. Immunol. (1976) 6 (7), 511-519), and MPC-11 (Cell ( 1976) 8 (3), 405-415), SP2/0 (Nature (1978) 276 (5685), 269-270), FO (J. Immunol. Methods (1980) 35 (1-2), 1-21), S194/5.XX0.BU.1 (J. Exp. Med. (1978) 148 (1), 313-323), R210 (Nature (1979) 277 (5692), 131-133), etc. can be suitably used.
  • the immune cells are fused with myeloma cells essentially according to known methods, such as the method of Köhler and Milstein et al. (Methods Enzymol. (1981) 73, 3-46).
  • the cell fusion can be carried out in a standard nutrient culture medium in the presence of a cell fusion promoter.
  • Fusion promoters such as polyethylene glycol (PEG) and Sendai virus (HVJ) can be used, and if desired, an adjuvant such as dimethyl sulfoxide can be added to further enhance fusion efficiency.
  • the ratio of immune cells to myeloma cells can be set as desired. For example, a ratio of immune cells to myeloma cells of 1 to 10 is preferred.
  • the culture medium used for the cell fusion may be, for example, RPMI1640 culture medium, MEM culture medium, or any other conventional culture medium used for this type of cell culture, which is suitable for growing the myeloma cell line.
  • serum supplements such as fetal calf serum (FCS) may be suitably added.
  • Cell fusion is performed by thoroughly mixing a predetermined amount of the immune cells and myeloma cells in the culture medium, and then adding a PEG solution (for example, an average molecular weight of approximately 1000 to 6000) that has been preheated to approximately 37°C, usually at a concentration of 30 to 60% (w/v). The mixture is gently mixed to form the desired fused cells (hybridomas). Next, the appropriate culture medium listed above is added sequentially, and the mixture is centrifuged and the supernatant is removed repeatedly, allowing cell fusion agents and other substances that are undesirable for hybridoma growth to be removed.
  • a PEG solution for example, an average molecular weight of approximately 1000 to 6000
  • the hybridomas obtained in this manner can be selected by culturing them in a conventional selective culture medium, such as HAT culture medium (a culture medium containing hypoxanthine, aminopterin, and thymidine). Culture can be continued using the above-mentioned HAT culture medium for a sufficient period of time (usually several days to several weeks) for cells other than the desired hybridoma (unfused cells) to die. Hybridomas producing the desired antibody are then screened and single-cloned using the conventional limiting dilution method.
  • HAT culture medium a culture medium containing hypoxanthine, aminopterin, and thymidine.
  • Hybridomas obtained in this manner can be selected using a selective culture medium that corresponds to the selection marker possessed by the myeloma used in cell fusion.
  • a selective culture medium that corresponds to the selection marker possessed by the myeloma used in cell fusion.
  • HAT culture medium a culture medium containing hypoxanthine, aminopterin, and thymidine. That is, when HAT-sensitive myeloma cells are used for cell fusion, cells that have successfully fused with normal cells can selectively grow in HAT culture medium.
  • Culture in the above HAT culture medium is continued for a period of time sufficient for cells other than the desired hybridoma (non-fused cells) to die.
  • the desired hybridoma can generally be selected by culturing for several days to several weeks. Hybridomas producing the desired antibody can then be screened and single-cloned using the conventional limiting dilution method.
  • FACS fluorescence activated cell sorting
  • Preferred cells for screening are mammalian cells that have been forced to express the antigen.
  • untransformed mammalian cells that have been used as host cells as a control, the binding activity of the antibody to the antigen on the cell surface can be selectively detected.
  • hybridomas that produce the desired monoclonal antibody can be obtained.
  • cells expressing a target antigen can be immobilized, and the binding activity of an antibody to the antigen-expressing cells can be evaluated based on the principles of ELISA.
  • antigen-expressing cells are immobilized in the wells of an ELISA plate.
  • Hybridoma culture supernatant is contacted with the immobilized cells in the wells, and antibodies binding to the immobilized cells are detected.
  • the monoclonal antibody is derived from a mouse, the antibody bound to the cells can be detected with an anti-mouse immunoglobulin antibody.
  • Hybridomas producing the desired antibody capable of binding to the antigen, selected by these screening methods can be cloned by limiting dilution or other methods.
  • the hybridomas producing the monoclonal antibodies thus prepared can be subcultured in a conventional culture medium and can be stored for a long period of time in liquid nitrogen.
  • the hybridoma can be cultured according to conventional methods, and the desired monoclonal antibody can be obtained from the culture supernatant.
  • the hybridoma can be administered to a compatible mammal to grow, and the monoclonal antibody can be obtained from the ascites fluid.
  • the former method is suitable for obtaining highly pure antibodies.
  • Antibodies encoded by antibody genes cloned from antibody-producing cells such as hybridomas can also be suitably used.
  • the cloned antibody gene is incorporated into a suitable vector and introduced into a host, whereby the antibody encoded by the gene is expressed.
  • Methods for isolating antibody genes, introducing them into vectors, and transforming host cells have already been established, for example, by Vandamme et al. (Eur. J. Biochem. (1990) 192 (3), 767-775). Methods for producing recombinant antibodies are also known, as described below.
  • RNA is usually extracted from the hybridoma.
  • mRNA can be extracted from cells using the following methods, for example. - Guanidine ultracentrifugation method (Biochemistry (1979) 18 (24), 5294-5299) -AGPC method (Anal. Biochem. (1987) 162 (1), 156-159)
  • the extracted mRNA can be purified using, for example, an mRNA Purification Kit (manufactured by GE Healthcare Biosciences).
  • kits for extracting total mRNA directly from cells such as the QuickPrep mRNA Purification Kit (manufactured by GE Healthcare Biosciences) are also commercially available.
  • mRNA can be obtained from hybridomas.
  • cDNA encoding the antibody V region can be synthesized from the obtained mRNA using reverse transcriptase.
  • cDNA can be synthesized using an AMV Reverse Transcriptase First-Strand cDNA Synthesis Kit (manufactured by Seikagaku Corporation) or the like.
  • SMART RACE cDNA Amplification Kit (Clontech) and the 5'-RACE method using PCR (Proc. Natl. Acad. Sci. USA (1988) 85 (23), 8998-9002, Nucleic Acids Res. (1989) 17 (8), 2919-2932) can be used appropriately to synthesize and amplify cDNA.
  • appropriate restriction enzyme sites can be introduced at both ends of the cDNA, as described below.
  • the desired cDNA fragment is purified from the resulting PCR product and then ligated to vector DNA.
  • a recombinant vector is thus created, introduced into E. coli or other bacteria, and colonies are selected.
  • the desired recombinant vector can then be prepared from the E. coli that formed the colonies. Whether the recombinant vector contains the base sequence of the desired cDNA is then confirmed using known methods, such as the dideoxynucleotide chain termination method.
  • the easiest way to obtain genes encoding variable regions is to use the 5'-RACE method, which uses primers for amplifying variable region genes.
  • cDNA is synthesized using RNA extracted from hybridoma cells as a template, and a 5'-RACE cDNA library is obtained.
  • a commercially available kit such as the SMART RACE cDNA Amplification Kit, can be used to synthesize the 5'-RACE cDNA library.
  • antibody genes are amplified by PCR.
  • Primers for amplifying mouse antibody genes can be designed based on known antibody gene sequences. These primers have different base sequences for each immunoglobulin subclass. Therefore, it is desirable to determine the subclass in advance using a commercially available kit such as the IsoStrip Mouse Monoclonal Antibody Isotyping Kit (Roche Diagnostics).
  • primers can be used that are capable of amplifying genes encoding ⁇ 1, ⁇ 2a, ⁇ 2b, and ⁇ 3 heavy chains, and ⁇ and ⁇ light chains.
  • the 3' primer generally anneals to a region corresponding to the constant region close to the variable region.
  • the 5' primer used is one that comes with the 5' RACE cDNA library construction kit.
  • the PCR products thus amplified can be used to reconstitute immunoglobulins consisting of a combination of heavy and light chains.
  • the antigen-binding activity of the reconstituted immunoglobulins can be used as an index to screen for the desired antibody. For example, screening can be performed as follows: (1) contacting an antibody containing a V region encoded by a cDNA obtained from a hybridoma with a cell expressing a desired antigen; (2) detecting the binding of the antigen-expressing cells to the antibody; and (3) selecting an antibody that binds to the antigen-expressing cells.
  • Methods for detecting the binding between an antibody and the antigen-expressing cells are known. Specifically, the binding between an antibody and the antigen-expressing cells can be detected by techniques such as the FACS mentioned above. Fixed specimens of the antigen-expressing cells can be used as appropriate to evaluate the binding activity of the antibody.
  • Panning methods using phage vectors are also suitable as a screening method for antibodies using binding activity as an indicator.
  • antibody genes are obtained as a library of heavy and light chain subclasses from a polyclonal antibody-expressing cell population, screening methods using phage vectors are advantageous.
  • Genes encoding the variable regions of the heavy and light chains can be linked with an appropriate linker sequence to form single-chain Fvs (scFvs).
  • scFvs single-chain Fvs
  • DNA encoding an scFv with the desired binding activity can be recovered by recovering the phage that has bound to the antigen. By repeating this procedure as necessary, scFvs with the desired binding activity can be concentrated.
  • the cDNA is digested with a restriction enzyme that recognizes restriction enzyme sites inserted at both ends of the cDNA.
  • Preferred restriction enzymes recognize and digest nucleotide sequences that appear infrequently in the nucleotide sequences constituting the antibody gene.
  • a restriction enzyme that generates a cohesive end it is preferable to insert a restriction enzyme that generates a cohesive end.
  • An antibody expression vector can be obtained by inserting the cDNA encoding the V region of the antibody digested as described above into an appropriate expression vector.
  • a chimeric antibody refers to an antibody in which the constant region and variable region are derived from different sources. Therefore, in addition to heterogeneous chimeric antibodies such as mouse-human, human-human allogeneic chimeric antibodies are also included in the chimeric antibodies of the present invention.
  • a chimeric antibody expression vector can be constructed by inserting the V region gene into an expression vector that already contains the constant region. Specifically, for example, a restriction enzyme recognition sequence for a restriction enzyme that digests the V region gene can be appropriately placed on the 5' side of an expression vector carrying DNA encoding the desired antibody constant region (C region).
  • a chimeric antibody expression vector is constructed by fusing the two genes in frame after digestion with the same combination of restriction enzymes.
  • the antibody gene is incorporated into an expression vector so that it is expressed under the control of an expression control region.
  • Expression control regions for antibody expression include, for example, enhancers and promoters.
  • an appropriate signal sequence can be added to the amino terminus so that the expressed antibody is secreted extracellularly. The signal sequence is cleaved from the carboxyl terminal portion of the expressed polypeptide, allowing the antibody to be secreted extracellularly.
  • recombinant cells expressing DNA encoding the antibody can be obtained.
  • DNA encoding the antibody heavy chain (H chain) and light chain (L chain) are each incorporated into separate expression vectors.
  • H chain and L chain By simultaneously transforming (co-transfecting) the same host cell with vectors incorporating the H chain and L chain, antibody molecules equipped with both H chains and L chains can be expressed.
  • host cells can be transformed by incorporating DNA encoding the H chain and L chain into a single expression vector (see International Publication WO 94/11523).
  • animal cells When eukaryotic cells are used as host cells, animal cells, plant cells, or fungal cells can be used as appropriate.
  • animal cells include the following: (1) Mammalian cells: CHO, COS, myeloma, BHK (baby hamster kidney), Hela, Vero, etc. (2) Amphibian cells: Xenopus oocytes, etc. (3) Insect cells: sf9, sf21, Tn5, etc.
  • an antibody gene expression system using plant cells derived from the genus Nicotiana such as Nicotiana tabacum
  • Nicotiana tabacum a plant cells derived from the genus Nicotiana, such as Nicotiana tabacum.
  • callus cultured cells can be used as appropriate.
  • fungal cells can be used: - Yeasts: Saccharomyces genus such as Saccharomyces cerevisiae, Pichia genus such as Pichia pastoris - Filamentous fungi: Aspergillus genus such as Aspergillus niger
  • Antibody gene expression systems using prokaryotic cells are also known.
  • bacterial cells such as Escherichia coli (E. coli) and Bacillus subtilis can be used as appropriate.
  • An expression vector containing the desired antibody gene is introduced into these cells by transformation. By culturing the transformed cells in vitro, the desired antibody can be obtained from the culture of the transformed cells.
  • transgenic animals can also be used to produce recombinant antibodies. That is, the antibody can be obtained from an animal into which a gene encoding the desired antibody has been introduced.
  • an antibody gene can be constructed as a fusion gene by inserting it in-frame into a gene encoding a protein specifically produced in milk. Examples of proteins secreted into milk include goat beta-casein.
  • a DNA fragment containing the fusion gene with the antibody gene inserted is injected into a goat embryo, and the injected embryo is then introduced into a female goat.
  • the transgenic goat (or its offspring) born to the goat that received the embryo produces milk from which the desired antibody can be obtained as a fusion protein with a milk protein.
  • hormones can be administered to transgenic goats to increase the amount of milk containing the desired antibody produced by the transgenic goat (Bio/Technology (1994), 12 (7), 699-702).
  • antigen-binding domains derived from recombinant antibodies that have been artificially modified to reduce heterologous antigenicity to humans can be appropriately used.
  • Recombinant antibodies include, for example, humanized antibodies. These modified antibodies can be appropriately produced using known methods.
  • the antibody variable regions used to create the various binding domains in the antigen-binding molecules described herein are typically composed of three complementarity-determining regions (CDRs) sandwiched between four framework regions (FRs).
  • CDRs are essentially the regions that determine the binding specificity of an antibody.
  • the amino acid sequences of CDRs are highly diverse. On the other hand, the amino acid sequences that make up FRs often show high identity even between antibodies with different binding specificities. Therefore, it is generally believed that the binding specificity of one antibody can be transferred to another antibody by CDR grafting.
  • selecting a human FR that is highly identical to the mouse FR is considered advantageous in terms of maintaining CDR function.
  • the base sequences to be linked are also designed to be connected in-frame to each other.
  • Human FRs are synthesized individually using each primer.
  • products are obtained in which DNA encoding mouse CDRs is added to each FR.
  • the base sequences encoding the mouse CDRs of each product are designed to overlap with each other.
  • the overlapping CDR portions of the products synthesized using the human antibody gene as a template are annealed to each other to carry out a complementary strand synthesis reaction. Through this reaction, the human FRs are linked via the mouse CDR sequences.
  • transgenic animals carrying the entire repertoire of human antibody genes can be used as immunized animals, and the desired human antibodies can be obtained by DNA immunization.
  • a technique for obtaining human antibodies by panning using a human antibody library is also known.
  • the V region of a human antibody is expressed on the surface of a phage as a single-chain antibody (scFv) using phage display.
  • Phages expressing scFvs that bind to an antigen can be selected.
  • the DNA sequence encoding the V region of a human antibody that binds to the antigen can be determined.
  • an expression vector can be created by fusing the V region sequence in frame with the sequence of the C region of the desired human antibody and then inserting it into an appropriate expression vector.
  • the expression vector is introduced into a suitable expression cell such as those listed above, and the gene encoding the human antibody is expressed to obtain the human antibody.
  • techniques for obtaining human antibodies by panning using a human antibody library include techniques using cell-free translation systems, techniques that display antigen-binding molecules on the surface of cells or viruses, and techniques that use emulsions.
  • techniques that use cell-free translation systems include ribosome display, which forms a complex between mRNA and the translated protein via ribosomes by removing stop codons, cDNA display, which covalently binds a gene sequence to the translated protein using compounds such as puromycin, and mRNA display, and CIS display, which forms a complex between a gene and the translated protein using a nucleic acid-binding protein.
  • E. coli display In addition to phage display, other techniques for displaying antigen-binding molecules on the surface of cells or viruses include E. coli display, Gram-positive bacteria display, yeast display, mammalian cell display, and virus display. Techniques that use emulsions include in vitro virus display, which involves encapsulating genes and translation-related molecules in an emulsion. These methods are already known (Nat Biotechnol. 2000 Dec;18(12):1287-92, Nucleic Acids Res. 2006;34(19):e127, Proc Natl Acad Sci U S A. 2004 Mar 2;101(9):2806-10, Proc Natl Acad Sci U S A. 2004 Jun 2 2;101(25):9193-8, Protein Eng Des Sel.
  • One preferred embodiment of the antigen-binding molecules of the present invention is a multispecific antibody.
  • an Fc region with reduced Fc ⁇ receptor-binding activity is used as the Fc region of a multispecific antibody of the present invention
  • an Fc region derived from a known multispecific antibody can also be used as appropriate.
  • Bispecific antibodies are particularly preferred as multispecific antibodies of the present invention.
  • a technique can be applied that suppresses undesired association between heavy chains by introducing charge repulsion at the interface of the second constant region (CH2) or third constant region (CH3) of the antibody heavy chain (WO2006/106905).
  • examples of amino acid residues that come into contact at the interface of other H chain constant regions include the regions corresponding to residues 356 (EU numbering), 439 (EU numbering), 357 (EU numbering), 370 (EU numbering), 399 (EU numbering), and 409 (EU numbering) in the CH3 region.
  • an antibody containing two types of H chain CH3 regions can be an antibody in which one to three pairs of amino acid residues selected from the following pairs of amino acid residues (1) to (3) in the first H chain CH3 region have the same charge: (1) amino acid residues contained in the H chain CH3 region, which are amino acid residues at positions 356 and 439 (EU numbering), (2) amino acid residues contained in the H chain CH3 region, which are amino acid residues at positions 357 and 370 (EU numbering), and (3) amino acid residues contained in the H chain CH3 region, which are amino acid residues at positions 399 and 409 (EU numbering).
  • the antibody may have a second H chain CH3 region different from the first H chain CH3 region, and the second H chain CH3 region has a set of amino acid residues selected from the sets of amino acid residues shown in (1) to (3), wherein one to three sets of amino acid residues corresponding to the sets of amino acid residues shown in (1) to (3) that have the same charge in the first H chain CH3 region have an opposite charge to the corresponding amino acid residues in the first H chain CH3 region.
  • amino acid residues described in (1) to (3) above are close to each other when associated. Those skilled in the art can find the positions corresponding to the amino acid residues described in (1) to (3) above for the desired H chain CH3 region or H chain constant region by homology modeling using commercially available software, and can modify the amino acid residues at those positions as appropriate.
  • the "charged amino acid residue” is preferably selected from amino acid residues included in either group (a) or (b) below: (a) glutamic acid (E), aspartic acid (D), (b) Lysine (K), arginine (R), histidine (H).
  • having the same charge means, for example, that two or more amino acid residues all have amino acid residues included in either group (a) or (b) above.
  • Having opposite charges means, for example, that when at least one amino acid residue among two or more amino acid residues has an amino acid residue included in either group (a) or (b) above, the remaining amino acid residues have amino acid residues included in a different group.
  • the antibody may have the first H chain CH3 region and the second H chain CH3 region cross-linked by a disulfide bond.
  • amino acid residues that can be modified in the present invention are not limited to those in the antibody variable region or constant region described above. Those skilled in the art can identify amino acid residues that form an interface between polypeptide mutants or heteromultimers by homology modeling using commercially available software, and can modify the amino acid residues at those sites to control association.
  • knob By substituting a larger side chain (knob) for an amino acid side chain in the variable region of one of the antibody's heavy chains and a smaller side chain (hole) for an amino acid side chain in the opposing variable region of the other heavy chain, the knob can be positioned in the hole, thereby efficiently facilitating aggregation between polypeptides with different amino acids that have Fc domains (WO1996/027011, Ridgway JB et al., Protein Engineering (1996) 9, 617-621, Merchant AM et al., Nature Biotechnology (1998) 16, 677-681, US20130336973).
  • Other methods for forming multispecific antibodies include antibody production techniques that utilize the association of antibody CH1 and CL, and VH and VL, as described in WO2011/028952, WO2014/018572, and Nat Biotechnol. 2014 Feb;32(2):191-8.; techniques for producing bispecific antibodies using separately prepared monoclonal antibodies (Fab Arm Exchange), as described in WO2008/119353 and WO2011/131746; and techniques described in WO2012/058768 and Other techniques that can be used include the technology described in WO2013/063702 for controlling the association between CH3s of antibody heavy chains, the technology described in WO2012/023053 for producing bispecific antibodies consisting of two types of light chains and one type of heavy chain, and the technology described in Christoph et al. (Nature Biotechnology Vol. 31, pp. 753-758 (2013)) for producing bispecific antibodies using two bacterial cell lines that each express one half of an antibody chain consisting of one H chain and one L chain.
  • one embodiment of multispecific antibody formation involves mixing two types of monoclonal antibodies in the presence of a reducing agent, cleaving the disulfide bonds in the core hinge, and then reassociating them to obtain a heterodimerized bispecific antibody (FAE).
  • FEE heterodimerized bispecific antibody
  • electrostatic interactions WO2006/106905
  • heterodimerization can be induced more efficiently during reassociation (WO2015/046467).
  • the multispecific antibody of the present invention by isolating and purifying it from the produced antibodies.
  • a method has been reported in which amino acid substitutions are introduced into the variable regions of two types of H chains to impart a difference in isoelectric point, making it possible to purify two types of homoantibodies and the desired heteroantibody by ion exchange chromatography (WO2007114325).
  • a common L chain capable of conferring binding ability to multiple different H chains can be obtained and used as the common L chain for a multispecific antibody.
  • IgG By expressing IgG by introducing such a common L chain and multiple different H chain genes into cells, it becomes possible to efficiently express multispecific IgG (Nature Biotechnology (1998) 16, 677-681).
  • a method can also be used to select a common L chain that corresponds to any different H chain and exhibits high binding ability (WO2004/065611).
  • an Fc region in which C-terminal heterogeneity has been reduced can be used as appropriate. More specifically, an Fc region in which glycine at position 446 and lysine at position 447, as specified according to EU numbering, in the amino acid sequences of two polypeptides that constitute an Fc region originating from IgG1, IgG2, IgG3, or IgG4, are deleted, is provided.
  • the antigen-binding molecule of the present invention may also be an antigen-binding molecule having the same amino acid sequence as the antigen-binding molecule having the above-mentioned modifications added thereto, which has been separately prepared.
  • the structure of a multispecific antigen-binding molecule of the present invention is not limited, as long as it comprises: (1) a first antigen-binding domain comprising the antibody H-chain variable region and L-chain variable region and having CD3-binding activity; (2) a second antigen-binding domain comprising an amino acid sequence different from that of the first antigen-binding domain; and, optionally, (3) a domain comprising an Fc region with reduced Fc ⁇ receptor-binding activity.
  • the above-mentioned domains can be directly linked via peptide bonds.
  • the linked polypeptide forms an antibody structure.
  • the antibody can be purified from the culture medium of the above-mentioned hybridoma, or from the culture medium of a desired host cell in which a polynucleotide encoding the polypeptide constituting the antibody is stably maintained.
  • Preferred antibody variable regions of the present invention that have CD3-binding activity include antibody variable regions that have binding activity to CD3 ⁇ .
  • antibody variable regions include the heavy chain variable regions of antibody Nos. 2, 14, 25, 29, 30-32, and 34 listed in Table 5 below, or antibody H chain variable regions having the amino acid sequences of heavy chain CDR1, CDR2, and CDR3; and the light chain variable regions of antibody Nos. 2, 14, 25, 29, 30-32, and 34 listed in Table 5, or antibody L chain variable regions having the amino acid sequences of light chain CDR1, CDR2, and CDR3; or antibody H chain variable regions and L chain variable regions functionally equivalent to these variable regions.
  • binding affinity to an antigen under specified conditions is equivalent, or that when used as a multispecific antigen-binding molecule, the cytotoxic activity against cells expressing the target antigen or tissues containing said cells is equivalent.
  • Binding affinity and cytotoxic activity can be measured based on the descriptions herein.
  • the cells used to measure cytotoxic activity may be desired cells expressing the target antigen or desired tissues containing said cells, but for example, human cancer cell lines expressing the target antigen can be used.
  • the reduction in binding activity to Fc ⁇ receptors may also be equivalent.
  • an antibody H-chain variable region functionally equivalent to an antibody H-chain variable region described herein means that when combined with the antibody L-chain variable region described herein as the pair of the original H-chain, it has equivalent binding affinity, or when used as a multispecific antigen-binding molecule, it has equivalent cytotoxic activity against cells expressing the target antigen or tissues containing said cells.
  • an antibody L-chain variable region functionally equivalent to an antibody L-chain variable region described herein means that when combined with the antibody H-chain variable region described herein as the pair of the original L-chain, it has equivalent binding affinity, or when used as a multispecific antigen-binding molecule, it has equivalent cytotoxic activity against cells expressing the target antigen or tissues containing said cells.
  • “equivalent” does not necessarily mean the same level of activity; it may also mean that the activity is enhanced, and therefore can also be expressed as “equivalent or greater.”
  • “equivalent” refers to a value (KD value/parent KD value) of 2.0 or less compared to the binding affinity (parent KD value) of a control antibody variable region.
  • the KD value/parent KD value is preferably 1.5 or less, more preferably 1.3 or less, 1.2 or less, 1.1 or less, 1.0 or less, 0.9 or less, 0.8 or less, 0.7 or less, 0.6 or less, or 0.5 or less.
  • the KD value/parent KD value is preferably 10 -6 to 2.0, more preferably 10 -3 to 1.5, more preferably 10 -1 to 1.3, more preferably 0.8 to 1.2, more preferably 0.9 to 1.1, and most preferably 1.0.
  • "equivalent” refers to a value (cell growth inhibition rate/parent cell growth inhibition rate) of 0.7 or higher compared to the cell growth inhibition rate of a control multispecific antigen-binding molecule (parent cell growth inhibition rate).
  • the concentration of the added multispecific antigen-binding molecule is determined appropriately, but is preferably measured at, for example, 0.01 nM, 0.05 nM, 0.1 nM, 0.5 nM, or 1 nM, preferably 0.05 nM or 0.1 nM.
  • the value of cell proliferation inhibition rate/parent cell proliferation inhibition rate is preferably 0.8 or more, more preferably 0.9 or more, 1.0 or more, 1.2 or more, 1.5 or more, 2 or more, 3 or more, 5 or more, 10 or more, or 20 or more. There is no upper limit, but it may be, for example, 10, 102 , 103 , 104 , 105 , or 106 .
  • examples of such activity include a value (concentration for 50% cell growth inhibition/concentration for 50% parent cell growth inhibition) of 1.5 or less, relative to the concentration of the original multispecific antigen-binding molecule that inhibits 50% cell growth (concentration for 50% parent cell growth inhibition).
  • concentration of multispecific antigen-binding molecule required to reduce the cell growth rate by half compared to when the multispecific antigen-binding molecule is not added.
  • concentration for 50% cell growth inhibition/concentration for 50% parent cell growth inhibition is preferably 1.3 or less, and more preferably 1.2 or less, 1.1 or less, 1.0 or less, 0.9 or less, 0.8 or less, 0.7 or less, 0.6 or less, or 0.5 or less.
  • 10-1 , 10-2 , 10-3 , 10-4 , 10-5 , or 10-6 is preferably 10 ⁇ 6 to 1.5 ⁇ 10 ⁇ 0 , more preferably 10 ⁇ 6 to 10 ⁇ 1 , more preferably 10 ⁇ 6 to 10 ⁇ 2 , and even more preferably 10 ⁇ 6 to 10 ⁇ 3 .
  • Cytotoxic activity can be assessed by in vitro TDCC activity as described in Example 4, and in this case, "equivalent” refers to a value (TDCC activity/parent TDCC activity) of 0.7 or greater compared to the TDCC activity of a control multispecific antigen-binding molecule.
  • the TDCC activity/parent TDCC activity value is preferably 0.8 or greater, and more preferably 0.9 or greater, 1.0 or greater, 1.2 or greater, 1.5 or greater, 2 or greater, 3 or greater, 5 or greater, 10 or greater, or 20 or greater.
  • the KD value of a second antigen-binding domain having binding activity to a target antigen may be, for example, 5x10 -9 M or less, preferably 4x10 -9 M or less, for example, 3x10 -9 M or less, 2x10 -9 M or less, 1x10 -9 M or less, 8x10 -10 M or less, 5x10 -10 M or less, 4x10 -10 M or less, 3x10 -10 M or less, 2x10 -10 M or less, 1x10 -10 M or less, 8x10 -11 M or less, 5x10 -11 M or less, 4x10 -11 M or less, 3x10 -11 M or less, 2x10 -11 M or less, 1x10 -11 M or less, 8x10 -12 M or less, 5x10 -12 M or less, 4x10 -12 M or less, 3x10 -12 M or less, 2x10 -12 M or less, 1x10 -12 M or less, 8x10 -13 M or less, 5x10
  • the KD value for CD3, for example, human CD3, more specifically, for example, human CD3 ⁇ chain may be, for example, 5x10 ⁇ 7 M or less, preferably 2x10 ⁇ 7 M or less, for example, 1.5x10 ⁇ 7 M or less, 1.4x10 ⁇ 7 M or less, 1.3x10 ⁇ 7 M or less, 1.2x10 ⁇ 7 M or less, 1x10 ⁇ 7 M or less, 3x10 ⁇ 8 M or less, 2x10 ⁇ 8 M or less, 1x10 ⁇ 8 M or less, 8x10 ⁇ 9 M or less, 5x10 ⁇ 9 M or less, 4x10 ⁇ 9 M or less, 3x10 ⁇ 9 M or less, 2x10 ⁇ 9 M or less, 1x10 ⁇ 9 M or less, 8x10 ⁇ 10 M or less, 5x10 ⁇ 10 M or less, 4x10 ⁇ 10 M or less, 3x10 ⁇ 9 M or less, 2x10 ⁇ 9 M or less, 1x10 ⁇ 9
  • the multispecific antigen-binding molecules of the present invention preferably have KD values for the target antigen and human CD3 (e.g., human CD3 ⁇ chain) of 5x10-9 M or less and 5x10-7 M or less, respectively, and more preferably 1x10-9 M or less and 5x10-8 M or less, respectively.
  • human CD3 e.g., human CD3 ⁇ chain
  • a "functionally equivalent" antibody variable region is not particularly limited as long as it is an antibody heavy chain variable region and/or antibody light chain variable region that meets the above-mentioned conditions.
  • Such antibody variable regions may, for example, have one or more amino acids (e.g., 1, 2, 3, 4, 5, or 10 amino acids) substituted, deleted, added, and/or inserted into the amino acid sequence of the variable regions of antibodies Nos. 2, 14, 25, 29, 30-32, and 34 listed in Table 5.
  • Methods for substituting, deleting, adding, and/or inserting one or more amino acids into an amino acid sequence that are well known to those skilled in the art include methods for introducing mutations into proteins.
  • amino acid side chain properties include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), amino acids with aliphatic side chains (G, A, V, L, I, P), amino acids with hydroxyl-containing side chains (S, T, Y), amino acids with sulfur-containing side chains (C, M), amino acids with carboxylic acid- and amide-containing side chains (D, N, E, Q), amino acids with base-containing side chains (R, K, H), and amino acids with aromatic-containing side chains (H, F, Y, W) (the numbers in parentheses represent the single-letter symbols of the amino acids).
  • hydrophobic amino acids A, I, L, M, F, P, W, Y, V
  • hydrophilic amino acids R, D, N, C, E, Q, G, H, K, S, T
  • amino acids with aliphatic side chains G,
  • Substitution of amino acids within each of these groups is referred to as a conservative substitution. It is already known that polypeptides having modified amino acid sequences by deletion, addition, and/or substitution of one or more amino acid residues with other amino acids can retain their biological activity (Mark, D. F. et al., Proc. Natl. Acad. Sci. USA (1984) 81:5662-6; Zoller, M. J. and Smith, M., Nucleic Acids Res. (1982) 10:6487-500; Wang, A. et al., Science (1984) 224:1431-3; Dalbadie-McFarland, G. et al., Proc. Natl. Acad. Sci. USA (1982) 79:6409-13).
  • Variable regions of the present invention containing such amino acid modifications have at least 70%, more preferably at least 75%, more preferably at least 80%, even more preferably at least 85%, even more preferably at least 90%, and most preferably at least 95% amino acid sequence identity with the CDR sequence, FR sequence, or entire variable region amino acid sequence before modification.
  • sequence identity is defined as the percentage of residues identical to those in the original H-chain variable region or L-chain variable region amino acid sequence, after aligning the sequences as necessary to maximize sequence identity and introducing gaps as appropriate. Amino acid sequence identity can be determined by the method described below.
  • “functionally equivalent antibody variable regions” can also be obtained from nucleic acids that hybridize under stringent conditions to nucleic acids consisting of nucleotide sequences encoding the amino acid sequences of the variable regions of antibodies Nos. 2, 14, 25, 29, 30 to 32, and 34 listed in Table 5, for example.
  • Examples of stringent hybridization conditions for isolating nucleic acids that hybridize under stringent conditions to nucleic acids consisting of nucleotide sequences encoding the amino acid sequences of the variable regions include 6 M urea, 0.4% SDS, 0.5 x SSC, and 37°C, or hybridization conditions of equivalent stringency.
  • washing conditions after hybridization include, for example, 0.5xSSC (1xSSC is 0.15 M NaCl, 0.015 M sodium citrate, pH 7.0) and 0.1% SDS at 60°C, more preferably 0.2xSSC and 0.1% SDS at 60°C, more preferably 0.2xSSC and 0.1% SDS at 62°C, more preferably 0.2xSSC and 0.1% SDS at 65°C, and more preferably 0.1xSSC and 0.1% SDS at 65°C.
  • the sequence of the isolated nucleic acid can be determined by known methods described below.
  • the homology of the isolated nucleic acid is at least 50% or more, more preferably 70% or more, and even more preferably 90% or more (e.g., 95%, 96%, 97%, 98%, 99% or more) sequence identity across the entire base sequence.
  • PCR polymerase chain reaction
  • nucleotide sequences and amino acid sequences can be determined using the BLAST algorithm by Karlin and Altschul (Proc. Natl. Acad. Sci. USA (1993) 90:5873-7). Programs called BLASTN and BLASTX have been developed based on this algorithm (Altschul et al., J. Mol. Biol. (1990) 215:403-10).
  • the present invention relates to nucleic acids (polynucleotides) encoding the antigen-binding molecules of the present invention.
  • the antigen-binding molecules of the present invention can be incorporated into any expression vector.
  • An appropriate host can be transformed with the expression vector to create cells expressing the antigen-binding molecules.
  • the antigen-binding molecules encoded by the nucleic acids (polynucleotides) can be obtained by culturing the cells expressing the antigen-binding molecules and recovering the expression product from the culture supernatant.
  • the present invention relates to vectors containing nucleic acids (polynucleotides) encoding the antigen-binding molecules of the present invention, cells harboring the vectors, and methods for producing antigen-binding molecules, which include culturing the cells and recovering the antigen-binding molecules from the culture supernatant.
  • nucleic acids polynucleotides
  • methods for producing antigen-binding molecules which include culturing the cells and recovering the antigen-binding molecules from the culture supernatant.
  • the present invention provides pharmaceutical compositions comprising, as an active ingredient, an antigen-binding molecule of the present invention (particularly a multispecific antigen-binding molecule).
  • the present invention provides pharmaceutical compositions that induce cytotoxicity, comprising, as an active ingredient, the antigen-binding molecule.
  • the pharmaceutical compositions of the present invention induce cytotoxicity, particularly T-cell-dependent cytotoxicity, and are preferably administered to subjects suffering from or at risk of recurrence of a disease for which such cytotoxicity is necessary for prevention or treatment.
  • the pharmaceutical compositions of the present invention are for use in the treatment or prevention of cancer.
  • compositions containing an antigen-binding molecule of the present invention can also be described as cytotoxicity inducers and cytostatic agents containing the antigen-binding molecule as an active ingredient; methods for inducing cytotoxicity and methods for inhibiting cell proliferation comprising the step of administering the antigen-binding molecule to a subject; the antigen-binding molecule for use in inducing cytotoxicity and inhibiting cell proliferation; or use of the antigen-binding molecule in the production of cytotoxicity inducers and cytostatic agents.
  • compositions containing an antigen-binding molecule of the present invention can also be described as a cancer therapeutic or preventive agent containing the antigen-binding molecule as an active ingredient, a method for treating or preventing cancer which comprises administering the antigen-binding molecule to a subject, the antigen-binding molecule for use in cancer therapeutic or preventive purposes, or use of the antigen-binding molecule in the manufacture of a cancer therapeutic or preventive agent.
  • containing an antigen-binding molecule as an active ingredient means containing the antigen-binding molecule as the main active ingredient, and does not limit the content of the antigen-binding molecule.
  • the antigen-binding molecule of the present invention can be administered by administering or incorporating a nucleic acid encoding the antigen-binding molecule of the present invention into a living body using a vector or the like, thereby directly expressing the antigen-binding molecule of the present invention in the living body; however, the antigen-binding molecule may also be administered without using a vector.
  • vectors include viral vectors and plasmid vectors, and further examples include adenovirus vectors and adeno-associated virus vectors.
  • the nucleic acid encoding the antigen-binding molecule of the present invention may be administered directly to a living body, or cells into which a nucleic acid encoding the antigen-binding molecule of the present invention has been introduced may be administered to a living body.
  • the antigen-binding molecule of the present invention can be administered by chemically modifying mRNA encoding the antigen-binding molecule of the present invention to increase mRNA stability in the living body, and then directly administering the mRNA to a human to express the antigen-binding molecule of the present invention in the living body (see EP2101823B and WO2013/120629).
  • B cells into which a nucleic acid encoding the antigen-binding molecule of the present invention has been introduced may be administered (Sci Immunol. (2019) 4(35), eaax0644).
  • bacteria into which nucleic acids encoding the antigen-binding molecules of the present invention have been introduced may be administered (Nature Reviews Cancer (2016) 18, 727-743).
  • the antigen-binding molecules of the present invention can be encapsulated in microcapsules (such as microcapsules made of hydroxymethylcellulose, gelatin, or poly(methyl methacrylate)) to form colloid drug delivery systems (such as liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules) (see, for example, "Remington's Pharmaceutical Science 16th edition,” Oslo Ed. (1980)).
  • methods for producing sustained-release drugs are also known, and these methods can be applied to the antigen-binding molecules of the present invention (J. Biomed. Mater. Res. (1981) 15, 267-277; Chemtech. (1982) 12, 98-105; U.S. Pat. No. 3,773,719; European Patent Publications EP 58481 and EP 133988; Biopolymers (1983) 22, 547-556).
  • the pharmaceutical composition, cytotoxicity inducer, cytostatic agent, or cancer treatment or prevention agent (hereinafter collectively referred to as the pharmaceutical composition of the present invention) of the present invention can be administered to a patient either orally or parenterally.
  • Parenteral administration is preferred.
  • Specific examples of such administration methods include injection, nasal administration, pulmonary administration, and transdermal administration.
  • Injection methods include, for example, intravenous injection, intramuscular injection, intraperitoneal injection, and subcutaneous injection.
  • the pharmaceutical composition of the present invention can be administered systemically or locally by injection.
  • an appropriate administration method can be selected depending on the patient's age and symptoms.
  • the dosage can be selected, for example, from a range of 0.0001 mg to 1000 mg per kg of body weight per administration.
  • the dosage can be selected, for example, from a range of 0.001 mg/body to 100,000 mg/body per patient.
  • the pharmaceutical composition of the present invention is not limited to these dosages.
  • compositions of the present invention can be formulated in accordance with conventional methods (e.g., Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, U.S.A.) and may contain pharmaceutically acceptable carriers and additives.
  • pharmaceutically acceptable carriers and additives examples include surfactants, excipients, colorants, flavorings, preservatives, stabilizers, buffers, suspending agents, isotonicity agents, binders, disintegrants, lubricants, flow enhancers, and flavoring agents.
  • other commonly used carriers can be used as appropriate.
  • carriers include light anhydrous silicic acid, lactose, crystalline cellulose, mannitol, starch, carmellose calcium, carmellose sodium, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinyl acetal diethylaminoacetate, polyvinylpyrrolidone, gelatin, medium-chain fatty acid triglycerides, polyoxyethylene hydrogenated castor oil 60, sucrose, carboxymethyl cellulose, corn starch, and inorganic salts.
  • a non-limiting example of a technique that can be combined with the antigen-binding molecules of the present invention is the creation of T cells that secrete T cell-redirecting antibodies using a CD3-binding domain (Trends Immunol. (2019) 40(3) 243-257).
  • One non-limiting creation method involves introducing nucleic acid encoding a bispecific antibody comprising the CD3-binding domain described herein and a cancer antigen-binding domain into effector cells such as T cells using gene modification techniques.
  • the present invention provides a method for inducing (inducing) damage to a target cell or a method for inhibiting the proliferation of the cell, the method comprising the step of contacting the target cell with a multispecific antigen-binding molecule of the present invention that binds to an antigen specifically expressed on the surface of the cell.
  • the present invention provides a method for inducing (inducing) damage to a cell expressing a target cancer-specific antigen or a tumor tissue containing the expressing cell, or a method for inhibiting the proliferation of the cell or tumor tissue, the method comprising the step of contacting the cell expressing the target cancer-specific antigen with a multispecific antigen-binding molecule of the present invention that binds to the antigen.
  • the present invention provides a method for inducing (inducing) damage to a cell or a method for inhibiting the proliferation of the cell, the method comprising the step of contacting a cell having the function of suppressing an immune response with a multispecific antigen-binding molecule of the present invention that binds to an antigen expressed on the surface of the cell.
  • the multispecific antigen-binding molecule that binds to the antigen is as described above for the antigen-binding molecule of the present invention contained in the cytotoxicity-inducing agent and cytostatic agent of the present invention.
  • the cells to which the multispecific antigen-binding molecule of the present invention that binds to the antigen are not particularly limited, as long as the antigen is expressed.
  • contact is performed, for example, by adding a multispecific antigen-binding molecule of the present invention that binds to the antigen to the culture medium of target antigen-expressing cells cultured in a test tube.
  • the antigen-binding molecule to be added may be in the form of a solution or a solid obtained by lyophilization or the like, as appropriate.
  • aqueous solution When added as an aqueous solution, it may be an aqueous solution containing only the multispecific antigen-binding molecule of the present invention, or it may be a solution containing, for example, the surfactants, excipients, colorants, flavorings, preservatives, stabilizers, buffers, suspending agents, isotonicity agents, binders, disintegrants, lubricants, flow enhancers, flavoring agents, etc. described above.
  • concentration to be added is not particularly limited, but a final concentration in the culture medium of preferably 1 pg/ml to 1 g/ml, more preferably 1 ng/ml to 1 mg/ml, and even more preferably 1 ⁇ g/ml to 1 mg/ml, is suitable.
  • "contact" in the present invention can also be achieved by administering to a non-human animal into which cells expressing the target antigen have been transplanted, or to an animal that has cells endogenously expressing the antigen.
  • Administration can be performed either orally or parenterally.
  • Parenteral administration is particularly preferred, and specific examples of such administration methods include injection, intranasal administration, pulmonary administration, and transdermal administration.
  • Injection can be, for example, intravenous injection, intramuscular injection, intraperitoneal injection, and subcutaneous injection.
  • the pharmaceutical composition of the present invention, or the cytotoxicity inducer and cell proliferation inhibitor can be administered systemically or locally by injection.
  • An appropriate administration method can be selected depending on the age and symptoms of the subject animal.
  • the solution When administered as an aqueous solution, the solution may contain only the multispecific antigen-binding molecule of the present invention, or may contain, for example, the above-mentioned surfactants, excipients, colorants, flavorings, preservatives, stabilizers, buffers, suspending agents, isotonicity agents, binders, disintegrants, lubricants, flow enhancers, and flavoring agents.
  • the dosage can be selected, for example, from the range of 0.0001 mg to 1000 mg per kg of body weight per administration. Alternatively, the dosage can be selected, for example, from the range of 0.001 to 100,000 mg/body per patient.
  • the dosage of the multispecific antigen-binding molecules of the present invention is not limited to these dosages.
  • the following methods are preferably used to evaluate or measure the cytotoxicity induced in target cells (particularly cells expressing a target antigen other than CD3, to which the second antigen-binding domain of the multispecific antigen-binding molecule of the present invention binds) by contact with the multispecific antigen-binding molecule of the present invention.
  • Methods for evaluating or measuring the cytotoxic activity in vitro include methods for measuring cytotoxic T cell activity. Whether or not the multispecific antigen-binding molecule of the present invention has T cell mediated cytotoxic activity can be measured by known methods (e.g., Current Protocols in Immunology, Chapter 7. Immunologic Studies in Humans, Editor, John E., Coligan et al., John Wiley & Sons, Inc., (1993)).
  • an antigen-binding molecule that binds to an antigen different from the target antigen and that is not expressed by the cells used in the test is used as a control in the same manner as the multispecific antigen-binding molecule of the present invention, and activity can be determined if the multispecific antigen-binding molecule of the present invention exhibits stronger cytotoxic activity than the antigen-binding molecule used as the control.
  • cytotoxic activity in vivo, cells expressing the target antigen are transplanted intradermally or subcutaneously into a non-human test animal, and the test antigen-binding molecule is then administered intravenously or intraperitoneally daily or at intervals of several days from the same day or the following day. By measuring the size of the tumor over time, the difference in the change in size of the tumor can be defined as cytotoxic activity.
  • a control antigen-binding molecule is administered, and cytotoxic activity can be determined if the tumor size in the group administered with the antigen-binding molecule of the present invention is significantly smaller than the tumor size in the group administered with the control antigen-binding molecule.
  • Methods for assessing or measuring the inhibitory effect on the proliferation of cells expressing a target antigen include measuring the uptake of isotope-labeled thymidine into cells and the MTT assay. Furthermore, methods for assessing or measuring cell proliferation inhibitory activity in vivo can be suitably the same as the methods for assessing or measuring cytotoxic activity in vivo described above.
  • kits for use in the methods of the present invention which comprise the antigen-binding molecules of the present invention or antigen-binding molecules produced by the production methods of the present invention.
  • the kits may also contain other packaged components such as pharmaceutically acceptable carriers, vehicles, and instructions for use.
  • the present invention also relates to the antigen-binding molecules of the present invention or antigen-binding molecules produced by the production methods of the present invention, for use in the methods of the present invention.
  • Example 1 Preparation of CDR-modified antibodies with improved binding stability to CD3 antigen Bispecific antibodies that damage target cells by recognizing the T cell marker CD3 and an antigen on the target cell have been reported, but there was still room for improvement in the binding stability of the reported CD3-binding antibody variable regions. Improving this profile was expected to have benefits such as sustained in vivo efficacy and ease of in vitro evaluation. Specifically, the antibody variable region described in WO2015174439 lost its binding ability over time in buffer, and further modification to improve binding stability was considered necessary.
  • the antibody heavy chain gene TR01H113-F760mnP17 (SEQ ID NO: 30) was constructed, which contains the heavy chain variable region of an antibody against human CD3 and the constant region sequence of human IgG1, as described in WO2015174439.
  • the antibody light chain gene L0011-KT0 (SEQ ID NO: 34) contained the light chain variable region of an antibody against human CD3 and the constant region sequence of human ⁇ chain. Plasmids encoding CDR-modified antibodies were obtained by introducing modifications into the bases encoding the CDR amino acid sequences of this antibody gene using methods known to those skilled in the art.
  • the heavy and light chain-encoding plasmids were mixed and transfected into human embryonic kidney cell-derived HEK293 cells.
  • the supernatant was cultured for 4 days and purified using methods known to those skilled in the art to obtain antibodies.
  • the absorbance of the purified antibody solution at 280 nm was measured using a spectrophotometer.
  • the concentration of the purified antibody was calculated from the obtained measurement values using the extinction coefficient calculated by the PACE method (Protein Science (1995) 4, 2411-2423).
  • Example 2 Evaluation of binding of antibodies with altered CDRs to CD3 antigen
  • a polynucleotide encoding an antigen consisting of human CD3 ⁇ and CD3 ⁇ and a FLAG tag was introduced into HEK293 cells, and the culture supernatant was purified by ion exchange chromatography, FLAG tag affinity chromatography, and gel filtration chromatography to obtain the antigen (hereinafter referred to as CD3eg-linker).
  • CD3eg-linker The interaction analysis between the prepared antibodies and CD3eg-linker was performed using Octet HTX as follows. Phosphate-buffered saline (pH 7.4) containing 0.05% TWEEN 20 was used as the buffer.
  • Protein A chips (Sartorius) were used as the sensor chip.
  • the antibodies listed in Table 1 were prepared in buffer at 10 ⁇ g/mL, and human CD3eg-linker was serially diluted to 900, 300, 100, and 33 nM.
  • the antibodies were loaded onto the sensor chip for 60 seconds after a 30-second baseline.
  • Antigen binding characteristics were measured by a 30-second baseline step followed by 180-second antigen binding and dissociation steps.
  • the binding value was defined as the magnitude of the response at 295 seconds into the measurement.
  • the equilibrium dissociation constant (KD) was determined by global fitting of the response curve at multiple concentrations (Octet BLI Analysis Version: 12.2.2.4). In this global fitting, the 1:1 binding model was used for all regions of the association and dissociation phases.
  • Example 3 Evaluation of binding stability of antibodies with modified CDRs to CD3eg-linker The prepared antibodies were diluted to 0.1 mg/mL and stored at 4°C for 3 days or at 50°C for 3 days, and antigen binding was evaluated using the same method as in Example 2.
  • binding H/I ratio in the table is the ratio of the binding measurement values for each antibody after storage at 4°C to those after storage at 50°C, and indicates the extent to which each antibody maintains its binding activity at 50°C.
  • Antibodies Nos. 2, 14, 25, 29, 30-32, and 34 created in the present invention had binding values at 50°C that were equal to or greater than 0.078, the value of the previously reported control antibody 1. Furthermore, the binding H/I values for these antibodies were 75% or greater, exceeding the 61% value of the previously reported control antibody 1. Therefore, it was confirmed that these modified antibodies showed a significant improvement over the decrease in binding ability at 50°C in buffer.
  • Example 4 Evaluation of antibodies with modified CDRs by TDCC Reporter Bioassay.
  • Bispecific antibodies were prepared using each antibody prepared in the present invention and an anti-Marvel D3 antibody using methods known to those skilled in the art.
  • the prepared antibodies were diluted to 0.044 mg/mL and stored at 4°C or 50°C for 3 days, as in Example 3.
  • the in vitro TDCC activity of these antibodies was measured to assess differences in TDCC activity depending on storage temperature.
  • In vitro TDCC activity was measured using TCR/CD3 Effector Cells (NFAT) and the Bio-Glo luciferase assay system (Promega), and TDCC activity was quantified by luminescence from the reporter gene luciferase.
  • the reaction was initiated by adding 10 ⁇ L of NFAT-RE-luc2 Jurkat cells (prepared in medium at a concentration of 3 ⁇ 10 6 /mL) and LS 174T cells (ATCC) (prepared at a concentration of 5 ⁇ 10 5 mL) to each well of a 384-well plate.
  • LS 174T is a cell line expressing the MarvelD3 antigen.
  • the reaction was carried out overnight at 37°C.
  • reporter gene expression was quantified by adding luciferase substrate to each well and measuring luminescence using a plate reader.
  • the luciferase response represents the average of the values measured for the 10 nM and 100 nM antibody solutions.
  • the “activation H/I ratio” in the table is the ratio of the luciferase response values for each antibody after storage at 4°C to those after storage at 50°C, and indicates the extent to which each antibody lost its binding activity at 50°C.
  • Antibody Nos. 2, 14, 25, 29, 30-32, and 34 selected in Example 3 exhibited luciferase responses of 4.3.E+05 or higher when stored at 4°C, which corresponds to the response of the previously reported control antibody 1 when stored at 50°C.
  • antibody Nos. 14, 25, 29, 30-32, and 34 exhibited activated H/I TDCC activity ratios of 67% or higher when stored at 50°C. This was superior to the previously reported value of control antibody 1, indicating that these antibodies were modified antibodies that maintained TDCC activity while improving the activated H/I ratio.
  • the amino acid sequences of antibody Nos. 1, 2, 14, 25, 29, 30-32, and 34 are shown in Table 5.
  • the present invention provides a novel CD3-binding domain that has superior stability compared to CD3-binding domains used in conventional T cell-redirecting antibodies, and an antigen-binding molecule containing said CD3-binding domain.
  • Bispecific antigen-binding molecules prepared by combining the CD3-binding domain of the present invention with an antigen-binding domain that binds to an antigen expressed on the surface of target cells such as cancer cells can induce T cell-dependent cytotoxicity against the target cells and can be used to treat or prevent various cancers.
  • Antigen-binding molecules containing the CD3-binding domain of the present invention show only a small decrease in CD3-binding activity at room temperature or above, and are expected to maintain their T cell-dependent cytotoxic activity for a long period of time when administered to patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Plant Pathology (AREA)
  • Mycology (AREA)
  • Physics & Mathematics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present disclosure provides a novel CD3-binding domain with superior stability compared to CD3-binding domains used in conventional T cell redirecting antibodies, and an antigen-binding molecule comprising the CD3-binding domain. A bispecific antigen-binding molecule prepared by combining the CD3-binding domain of the present disclosure with an antigen-binding domain binding to an antigen expressed on the surface of a target cell such as a cancer cell can induce T-cell-dependent cytotoxicity to the target cell and can be used for the treatment or prevention of various cancers.

Description

安定性が改善されたCD3標的抗原結合分子CD3-targeted antigen-binding molecules with improved stability

 本開示は、安定性が改善されたCD3結合ドメインを含む抗原結合分子、当該抗原結合分子を含む医薬組成物、およびその使用等に関する。 The present disclosure relates to an antigen-binding molecule comprising a CD3-binding domain with improved stability, a pharmaceutical composition comprising the antigen-binding molecule, and uses thereof.

 抗体は血漿中での安定性が高く、副作用が少ないことから医薬品として注目されている(非特許文献1および非特許文献2)。抗体は、抗原に結合する作用、アゴニスト作用やアンタゴニスト作用だけでなく、ADCC(Antibody Dependent Cytotoxicity:抗体依存性傷害活性), ADCP(Antibody Dependent Cell phagocytosis:抗体依存性細胞食作用), CDC(補体依存性細胞傷害活性)といったエフェクター細胞による細胞傷害活性(エフェクター機能とも言う)を誘導し、がん細胞に対する抗腫瘍効果を発揮することが知られている(非特許文献3)。抗体のFc領域がNK細胞やマクロファージなどのエフェクター細胞上に存在するFcレセプターに結合することにより、抗体が結合した標的のがん細胞に対してこれらのエフェクター細胞が発揮する細胞傷害がADCCである。 Antibodies are attracting attention as pharmaceuticals due to their high stability in plasma and minimal side effects (Non-Patent Documents 1 and 2). Antibodies not only bind to antigens and have agonistic and antagonistic effects, but are also known to exert antitumor effects against cancer cells by inducing cytotoxic activity (also known as effector function) by effector cells, such as ADCC (Antibody Dependent Cytotoxicity), ADCP (Antibody Dependent Cell phagocytosis), and CDC (Complement-Dependent Cytotoxicity) (Non-Patent Document 3). ADCC is the cytotoxicity exerted by effector cells against antibody-bound target cancer cells when the Fc region of an antibody binds to Fc receptors present on effector cells such as NK cells and macrophages.

 天然型の免疫グロブリンは、可変領域で抗原と結合し、定常領域でFcγR、FcRn、FcαR、FcεRといったレセプターや補体と結合する。通常の天然型のIgG型の抗体は、その可変領域(Fab)により1つのエピトープを認識して結合することから、1つの抗原にしか結合することが出来ない。一方で、がんや炎症においては多種類のタンパク質が関与することが知られており、タンパク質同士がクロストークしていることがある。 Natural immunoglobulins bind to antigens through their variable regions, and to receptors such as FcγR, FcRn, FcαR, and FcεR, or to complements, through their constant regions. Normal natural IgG antibodies recognize and bind to a single epitope through their variable region (Fab), and can therefore only bind to a single antigen. However, it is known that many different proteins are involved in cancer and inflammation, and crosstalk between these proteins can occur.

 複数のターゲットを阻害する分子として、1分子で2種類以上の抗原と結合する抗体(Bispecific抗体、二重特異性抗体)が研究されている。天然型のIgG型の抗体を改良することにより、異なる2つの抗原(第1の抗原と第2の抗原)への結合活性を付与することが可能である(非特許文献4)。そのため、2種類以上の抗原を1つの分子で中和する、または細胞傷害活性をもつ細胞とがん細胞をクロスリンクすることで抗腫瘍活性を高めることが可能である。 Bispecific antibodies, which bind to two or more antigens with a single molecule, are being researched as molecules that inhibit multiple targets. By improving natural IgG antibodies, it is possible to confer binding activity to two different antigens (a first antigen and a second antigen) (Non-Patent Document 4). Therefore, it is possible to neutralize two or more antigens with a single molecule, or to enhance anti-tumor activity by cross-linking cytotoxic cells with cancer cells.

 Bispecific抗体の一つとして、T細胞をエフェクター細胞として動員する細胞傷害をその抗腫瘍効果のメカニズムとする抗体であるT細胞リダイレクティング抗体(T cell-redirecting抗体)が1980年代から知られている(非特許文献5、6、7)。NK細胞やマクロファージをエフェクター細胞として動員するADCCをその抗腫瘍効果のメカニズムとする抗体とは異なり、T細胞リダイレクティング抗体は、T細胞上のT細胞レセプター(TCR)複合体の構成サブユニットのいずれかに対する抗体、特にCD3ε鎖に結合する抗体と、標的であるがん細胞上の抗原に結合する抗体を含むbi-specific抗体である。T細胞リダイレクティング抗体がCD3ε鎖とがん抗原に同時に結合することにより、T細胞ががん細胞に接近する。その結果、T細胞の持つ細胞傷害作用によりがん細胞に対する抗腫瘍効果が発揮されると考えられている。 T cell-redirecting antibodies, a type of bispecific antibody known since the 1980s, are antibodies whose antitumor effect is mediated by cytotoxicity, which recruits T cells as effector cells (Non-Patent Documents 5, 6, 7). Unlike antibodies whose antitumor effect is mediated by ADCC, which recruits NK cells and macrophages as effector cells, T cell-redirecting antibodies are bispecific antibodies that contain an antibody against one of the constituent subunits of the T cell receptor (TCR) complex on T cells, specifically an antibody that binds to the CD3ε chain, and an antibody that binds to an antigen on the target cancer cell. The simultaneous binding of T cell-redirecting antibodies to the CD3ε chain and the cancer antigen allows T cells to approach the cancer cells. As a result, it is believed that the cytotoxic activity of T cells exerts an antitumor effect on cancer cells.

 T細胞リダイレクティング抗体の一つとして知られているCatumaxomabは、2つのFabでそれぞれがん抗原(EpCAM)とT細胞に発現しているCD3ε鎖に結合する。Catumaxomabは、がん抗原およびCD3εと同時に結合することによってT細胞による細胞傷害活性を誘導し、かつ、がん抗原およびFcγRと同時に結合することによってNK細胞やマクロファージ等の抗原提示細胞による細胞傷害活性を誘導するtrifunctional抗体である。しかし、trifunctional抗体は、がん抗原が存在しない場合でもCD3εおよびFcγRと同時に結合するため、がん細胞が存在しない環境でもCD3εを発現しているT細胞とFcγRを発現している細胞がクロスリンクされ、各種サイトカインが大量に産生される。このようながん抗原非依存的な各種サイトカインの産生誘導により、trifunctional抗体の投与は現状、腹腔内に限定されており(非特許文献8)、重篤なサイトカインストーム様の副作用により全身投与は極めて困難である。 Catumaxomab, a T cell-redirecting antibody, uses two Fab fragments to bind to a cancer antigen (EpCAM) and the CD3ε chain expressed on T cells. Catumaxomab is a trifunctional antibody that simultaneously binds to both a cancer antigen and CD3ε, thereby inducing cytotoxic activity by T cells, and to both a cancer antigen and FcγR, thereby inducing cytotoxic activity by antigen-presenting cells such as NK cells and macrophages. However, because trifunctional antibodies simultaneously bind to CD3ε and FcγR even in the absence of cancer antigens, CD3ε-expressing T cells and FcγR-expressing cells are cross-linked, resulting in the mass production of various cytokines, even in an environment where cancer cells are not present. Due to this cancer antigen-independent induction of cytokine production, trifunctional antibody administration is currently limited to intraperitoneal administration (Non-Patent Document 8), and systemic administration is extremely difficult due to the severe cytokine storm-like side effects.

 一方、BiTE(bispecific T-cell engager)はcatumaxomabとは異なりFcγ受容体に対する結合部位を持たないため、がん抗原非依存的にT細胞とNK細胞やマクロファージなどに発現する受容体が架橋されることはない。そのため、catumaxomabが投与された場合に観察されたがん抗原非依存的なサイトカインの誘導は起こらないことが示されている。しかしながら、BiTEはFc領域を欠く低分子量型の改変抗体分子であるために、治療用抗体として通常用いられるIgG型の抗体に比較して、患者に投与されたBiTEの血中半減期は著しく短いという問題点が存在する。 On the other hand, unlike catumaxomab, BiTE (bispecific T-cell engager) does not have a binding site for Fcγ receptors, and therefore does not cross-link T cells with receptors expressed on NK cells, macrophages, etc. in a cancer antigen-independent manner. As a result, it has been shown that the cancer antigen-independent cytokine induction observed when catumaxomab is administered does not occur. However, because BiTE is a low-molecular-weight modified antibody molecule lacking the Fc region, there is a problem in that the half-life of BiTE administered to patients in the blood is significantly shorter than that of IgG antibodies, which are commonly used as therapeutic antibodies.

 近年、FcγRに対する結合活性を低減させたFc領域を用いることで、T細胞を標的がん細胞に近接させることにより発揮する抗腫瘍活性と、がん抗原非依存的にサイトカインストームなどを誘導しないという安全性上の優れた性質と、長い血中半減期とを併せ持つ、新たなポリペプチド会合体が提供されている(特許文献1)。このような有利な効果を有する新たなT細胞リダイレクティング抗体は、がん細胞を標的とするだけでなく、がん微小環境に存在する免疫応答を抑制する機能を有する細胞である制御性T細胞や疲弊T細胞を標的とする場合にも有用であることが報告されている(特許文献2、3)。 Recently, new polypeptide complexes have been provided that use an Fc region with reduced FcγR-binding activity to bring T cells into close proximity with target cancer cells, exhibiting excellent safety characteristics by not inducing cytokine storms or other conditions in a cancer antigen-independent manner, and have a long blood half-life (Patent Document 1). It has been reported that new T cell redirecting antibodies with such advantageous effects are useful not only for targeting cancer cells, but also for targeting regulatory T cells and exhausted T cells, which are cells present in the cancer microenvironment that function to suppress immune responses (Patent Documents 2 and 3).

WO2012/073985WO2012/073985 WO2016/047722WO2016/047722 WO2015/174439WO2015/174439

Nat. Biotechnol. (2005) 23, 1073-1078Nat. Biotechnol. (2005) 23, 1073-1078 Eur J Pharm Biopharm. (2005) 59 (3), 389-396Eur J Pharm Biopharm. (2005) 59 (3), 389-396 Drug Des Devel Ther (2009) 3, 7-16Drug Des Devel Ther (2009) 3, 7-16 MAbs. (2012) Mar 1, 4(2)MAbs. (2012) Mar 1, 4(2) Nature (1985) 314 (6012), 628-31Nature (1985) 314 (6012), 628-31 Int J Cancer (1988) 41 (4), 609-15.Int J Cancer (1988) 41 (4), 609-15. Proc Natl Acad Sci USA (1986) 83 (5), 1453-7Proc Natl Acad Sci USA (1986) 83 (5), 1453-7 Cancer Immunol Immunother. (2007) 56(9), 1397-406Cancer Immunol Immunother. (2007) 56(9), 1397-406

 本発明は上記の情況に鑑みてなされたものであり、特にT細胞リダイレクティング抗体で用いられるCD3結合ドメインに着目し、従来のT細胞リダイレクティング抗体で用いられているCD3結合ドメインよりも優れた安定性を有する新規CD3結合ドメイン、当該CD3結合ドメインを含む抗原結合分子、当該抗原結合分子の製造方法、および当該抗原結合分子を有効成分として含む医薬組成物、ならびにそれらの使用を提供することを目的とする。 The present invention was made in light of the above circumstances, and focuses particularly on the CD3-binding domain used in T cell redirecting antibodies. It aims to provide a novel CD3-binding domain that has greater stability than the CD3-binding domains used in conventional T cell redirecting antibodies, an antigen-binding molecule containing said CD3-binding domain, a method for producing said antigen-binding molecule, a pharmaceutical composition containing said antigen-binding molecule as an active ingredient, and uses thereof.

 本発明者らは、WO2015174439に記載されている二重特異性抗体に含まれるCD3結合FabのCDRに様々な改変を導入し、常温以上の環境下で保存した際のCD3結合活性の低下の程度が親Fabと比べて小さい改変体を得ることに成功した。本発明者らはさらに、それらの改変されたCD3結合ドメインを、がん特異的抗原結合ドメイン等の他の抗原結合ドメインと組み合わせて二重特異性抗原結合分子を調製したところ、それらを常温以上の環境下で保存した際のT細胞依存性細胞傷害活性の低下の程度も小さいことを見出した。本発明者らは、かかる発見に基づいて、本発明に係る多重特異性抗原結合分子が、がん特異的抗原等の標的抗原を発現する標的細胞を含む組織を傷害し得ることを明らかにした。 The present inventors introduced various modifications into the CDRs of the CD3-binding Fab contained in the bispecific antibody described in WO2015174439 and succeeded in obtaining modified antibodies that showed a smaller degree of decline in CD3-binding activity when stored at room temperature or above than the parent Fab. The present inventors further combined these modified CD3-binding domains with other antigen-binding domains, such as a cancer-specific antigen-binding domain, to prepare bispecific antigen-binding molecules and found that the degree of decline in T cell-dependent cytotoxicity when stored at room temperature or above was also small. Based on these findings, the present inventors have demonstrated that the multispecific antigen-binding molecules of the present invention can damage tissues containing target cells expressing a target antigen, such as a cancer-specific antigen.

 本開示はこのような知見に基づくものであり、具体的には以下に例示的に記載する実施態様を包含するものである。
〔1〕 第一の抗原結合ドメインおよび第二の抗原結合ドメインを含む抗原結合分子であって、第一の抗原結合ドメインが、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域を含み、
 前記H鎖可変領域が、
  アミノ酸配列NAWMH(配列番号:1)を含むH鎖CDR1;
  アミノ酸配列QIX1DKSQNYATX2VAESVKG(配列番号:2)を含むH鎖CDR2であって、X1がKまたはRであり、X2がYまたはFである、H鎖CDR2;及び
  アミノ酸配列VHYX3AGYGVDX4(配列番号:3)を含むH鎖CDR3であって、X3がAまたはPであり、X4がI、MまたはLである、H鎖CDR3
を含み、
 前記L鎖可変領域が、
  アミノ酸配列RSX5X6X7VVHENRX8TYLH(配列番号:4)を含むL鎖CDR1であって、X5がSまたはTであり、X6がQまたはMであり、X7がSまたはTであり、X8がQまたはNである、L鎖CDR1;
  アミノ酸配列KVSNRFS(配列番号:5)を含むL鎖CDR2;及び
  アミノ酸配列GQGTQVPYT(配列番号:6)を含むL鎖CDR3
を含む、抗原結合分子。
〔2〕 CD3に対する結合活性を有する前記抗体H鎖可変領域及びL鎖可変領域が、以下の(a1)~(a8)から選ばれるH鎖CDR1、CDR2及びCDR3、並びに、L鎖CDR1、CDR2及びCDR3の組合せのうちのいずれかを含む、〔1〕に記載の抗原結合分子:
 (a1) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、配列番号:10に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a2) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a3) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:16に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a4) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a5) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:13に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a6) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:17に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a7) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:18に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a8) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:19に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ。
〔3〕 CD3に対する結合活性を有する前記抗体H鎖可変領域及びL鎖可変領域が、以下の(a1)~(a8)から選ばれるH鎖可変領域及びL鎖可変領域の組合せのうちのいずれかを含む、〔1〕または〔2〕に記載の抗原結合分子:
 (a1)配列番号:20に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a2) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a3) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:26に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a4) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a5) 配列番号:23に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a6) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:27に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a7) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:28に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a8) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:29に示されるアミノ酸配列を含むL鎖可変領域の組合せ。
〔4-1〕 Fc領域をさらに含む、〔1〕~〔3〕のいずれかに記載の抗原結合分子。
〔4-2〕 Fcγ受容体に対する結合活性が低下しているFc領域をさらに含む、〔1〕~〔3〕のいずれかに記載の抗原結合分子。
〔4-3〕抗原結合分子が、以下の(a1)~(a8)から選ばれる重鎖及び軽鎖の組合せのうちのいずれかを含む、〔4-1〕または〔4-2〕に記載の抗原結合分子:
 (a1)配列番号:30に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a2) 配列番号:31に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a3) 配列番号:31に示されるアミノ酸配列を含む重鎖及び配列番号:36に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a4) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a5) 配列番号:33に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a6) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:37に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a7) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:38に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a8) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:39に示されるアミノ酸配列を含む軽鎖の組合せ。
〔5-1〕 単一特異性抗原結合分子である、〔1〕~〔4〕のいずれかに記載の抗原結合分子。
〔5-2〕 第一の抗原結合ドメインおよび第二の抗原結合ドメインが、同じアミノ酸配列を含む、〔1〕~〔4〕のいずれかに記載の抗原結合分子。
〔5-3〕 第一の抗原結合ドメインおよび第二の抗原結合ドメインが、互いに異なるアミノ酸配列を含む、〔1〕~〔4〕のいずれかに記載の抗原結合分子。
〔6-1〕 多重特異性抗原結合分子である、〔1〕~〔4〕のいずれかに記載の抗原結合分子。
〔6-2〕 二重特異性抗原結合分子である、〔1〕~〔4〕のいずれかに記載の抗原結合分子。
〔6-3〕 三重以上の特異性を有する抗原結合分子である、〔1〕~〔4〕のいずれかに記載の抗原結合分子。
〔7-1〕 第二の抗原結合ドメインが、がん抗原に対する結合活性を有する抗体可変領域を含む、〔6〕に記載の抗原結合分子。
〔7-2〕 第二の抗原結合ドメインが、免疫応答を抑制する機能を有する細胞の表面に発現する分子に対する結合活性を有する抗体可変領域を含む、〔6〕に記載の抗原結合分子。
〔8〕 抗体である、〔1〕~〔7〕のいずれかに記載の抗原結合分子。
〔9〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子をコードする核酸。
〔10〕 〔9〕に記載された核酸が導入されたベクター。
〔11〕 〔9〕に記載の核酸または〔10〕に記載のベクターを含む、細胞。
〔12〕 〔11〕に記載の細胞を培養する工程を含む、〔1〕~〔8〕のいずれかに記載の抗原結合分子を製造する方法。
〔13〕 〔12〕に記載の方法によって製造された、抗原結合分子。
〔14〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子と、薬学的に許容される担体とを含む、医薬組成物。
〔15-1〕 細胞傷害を誘導するための、〔14〕に記載の医薬組成物。
〔15-2〕 T細胞依存的細胞傷害を誘導するための、〔14〕に記載の医薬組成物。
〔15-3〕 がんの治療または予防に使用するための、〔14〕に記載の医薬組成物。
〔16-1〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子または〔14〕に記載の医薬組成物を投与する工程を含む、細胞傷害を誘導する方法。
〔16-2〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子または〔14〕に記載の医薬組成物を投与する工程を含む、T細胞依存的細胞傷害を誘導する方法。
〔16-3〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子または〔14〕に記載の医薬組成物を投与する工程を含む、がんを治療または予防する方法。
〔17-1〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子または〔14〕に記載の医薬組成物と、使用のための説明書とを含む、細胞傷害を誘導するためのキット。
〔17-2〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子または〔14〕に記載の医薬組成物と、使用のための説明書とを含む、T細胞依存的細胞傷害を誘導するためのキット。
〔17-3〕 〔1〕~〔8〕のいずれかに記載の抗原結合分子または〔14〕に記載の医薬組成物と、使用のための説明書とを含む、がんの治療または予防のためのキット。
〔18-1〕 細胞傷害の誘導における使用のための、〔1〕~〔8〕のいずれかに記載の抗原結合分子。
〔18-2〕 T細胞依存的細胞傷害の誘導における使用のための、〔1〕~〔8〕のいずれかに記載の抗原結合分子。
〔18-3〕 がんの治療または予防における使用のための、〔1〕~〔8〕のいずれかに記載の抗原結合分子。
〔19-1〕 細胞傷害誘導剤の製造における、〔1〕~〔8〕のいずれかに記載の抗原結合分子の使用。
〔19-2〕 T細胞依存的細胞傷害誘導剤の製造における、〔1〕~〔8〕のいずれかに記載の抗原結合分子の使用。
〔19-3〕 がんの治療剤または予防剤の製造における、〔1〕~〔8〕のいずれかに記載の抗原結合分子の使用。
〔A1〕単一の抗原結合ドメインを含む一価の抗原結合分子であって、前記単一の抗原結合ドメインが、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域を含み、
 前記H鎖可変領域が、
  アミノ酸配列NAWMH(配列番号:1)を含むH鎖CDR1;
  アミノ酸配列QIX1DKSQNYATX2VAESVKG(配列番号:2)を含むH鎖CDR2であって、X1がKまたはRであり、X2がYまたはFである、H鎖CDR2;及び
  アミノ酸配列VHYX3AGYGVDX4(配列番号:3)を含むH鎖CDR3であって、X3がAまたはPであり、X4がI、MまたはLである、H鎖CDR3
を含み、
 前記L鎖可変領域が、
  アミノ酸配列RSX5X6X7VVHENRX8TYLH(配列番号:4)を含むL鎖CDR1であって、X5がSまたはTであり、X6がQまたはMであり、X7がSまたはTであり、X8がQまたはNである、L鎖CDR1;
  アミノ酸配列KVSNRFS(配列番号:5)を含むL鎖CDR2;及び
  アミノ酸配列GQGTQVPYT(配列番号:6)を含むL鎖CDR3
を含む、抗原結合分子。
〔A2〕 CD3に対する結合活性を有する前記抗体H鎖可変領域及びL鎖可変領域が、以下の(a1)~(a8)から選ばれるH鎖CDR1、CDR2及びCDR3、並びに、L鎖CDR1、CDR2及びCDR3の組合せのうちのいずれかを含む、〔A1〕に記載の抗原結合分子:
 (a1) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、配列番号:10に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a2) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a3) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:16に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a4) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a5) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:13に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a6) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:17に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a7) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:18に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a8) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:19に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ。
〔A3〕 CD3に対する結合活性を有する前記抗体H鎖可変領域及びL鎖可変領域が、以下の(a1)~(a8)から選ばれるH鎖可変領域及びL鎖可変領域の組合せのうちのいずれかを含む、〔A1〕または〔A2〕に記載の抗原結合分子:
 (a1)配列番号:20に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a2) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a3) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:26に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a4) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a5) 配列番号:23に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a6) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:27に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a7) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:28に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a8) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:29に示されるアミノ酸配列を含むL鎖可変領域の組合せ。
〔A4-1〕 Fc領域をさらに含む、〔A1〕~〔A3〕のいずれかに記載の抗原結合分子。
〔A4-2〕 Fcγ受容体に対する結合活性が低下しているFc領域をさらに含む、〔A1〕~〔A3〕のいずれかに記載の抗原結合分子。
〔A4-3〕抗原結合分子が、以下の(a1)~(a8)から選ばれる重鎖及び軽鎖の組合せのうちのいずれかを含む、〔A4-1〕または〔A4-2〕に記載の抗原結合分子:
 (a1)配列番号:30に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a2) 配列番号:31に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a3) 配列番号:31に示されるアミノ酸配列を含む重鎖及び配列番号:36に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a4) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a5) 配列番号:33に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a6) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:37に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a7) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:38に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a8) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:39に示されるアミノ酸配列を含む軽鎖の組合せ。
〔A5〕 〔A1〕~〔A4〕のいずれかに記載の抗原結合分子をコードする核酸。
〔A6〕 〔A5〕に記載された核酸が導入されたベクター。
〔A7〕 〔A5〕に記載の核酸または〔A6〕に記載のベクターを含む、細胞。
〔A8〕 〔A7〕に記載の細胞を培養する工程を含む、〔A1〕~〔A4〕のいずれかに記載の抗原結合分子を製造する方法。
〔A9〕 〔A8〕に記載の方法によって製造された、抗原結合分子。
The present disclosure is based on these findings and specifically includes the embodiments exemplified below.
[1] An antigen-binding molecule comprising a first antigen-binding domain and a second antigen-binding domain, wherein the first antigen-binding domain comprises an antibody H chain variable region and L chain variable region that have binding activity to CD3;
the heavy chain variable region
H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1);
an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3), wherein X3 is A or P, and X4 is I, M, or L.
Including,
the light chain variable region
an L chain CDR1 comprising the amino acid sequence RSX5X6X7VVHENRX8TYLH ( SEQ ID NO: 4 ), wherein X5 is S or T, X6 is Q or M, X7 is S or T, and X8 is Q or N;
L chain CDR2 comprising the amino acid sequence KVSNRFS (SEQ ID NO: 5); and L chain CDR3 comprising the amino acid sequence GQGTQVPYT (SEQ ID NO: 6).
An antigen-binding molecule comprising:
[2] The antigen-binding molecule of [1], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain CDR1, CDR2, and CDR3, and L chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8):
(a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a3) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 16, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a4) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a5) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 13, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a6) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 17, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a7) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 18, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a8) A combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 19, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6.
[3] The antigen-binding molecule of [1] or [2], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain variable region and L chain variable region selected from the following (a1) to (a8):
(a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26;
(a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a5) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 23 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a6) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 27;
(a7) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 28;
(a8) A combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 29.
[4-1] The antigen-binding molecule of any of [1] to [3], further comprising an Fc region.
[4-2] the antigen-binding molecule of any of [1] to [3], further comprising an Fc region with reduced binding activity to an Fcγ receptor;
[4-3] The antigen-binding molecule of [4-1] or [4-2], which comprises any one of the combinations of heavy chains and light chains selected from the following (a1) to (a8):
(a1) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 30 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a2) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a3) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 36;
(a4) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a5) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 33 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a6) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 37;
(a7) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 38;
(a8) A combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 39.
[5-1] The antigen-binding molecule of any of [1] to [4], which is a monospecific antigen-binding molecule.
[5-2] the antigen-binding molecule of any one of [1] to [4], wherein the first antigen-binding domain and the second antigen-binding domain comprise the same amino acid sequence;
[5-3] the antigen-binding molecule of any one of [1] to [4], wherein the first antigen-binding domain and the second antigen-binding domain comprise amino acid sequences that are different from each other;
[6-1] The antigen-binding molecule of any of [1] to [4], which is a multispecific antigen-binding molecule.
[6-2] The antigen-binding molecule of any of [1] to [4], which is a bispecific antigen-binding molecule.
[6-3] The antigen-binding molecule of any of [1] to [4], which is an antigen-binding molecule with triple or higher specificity.
[7-1] the antigen-binding molecule of [6], wherein the second antigen-binding domain comprises an antibody variable region having binding activity against a cancer antigen;
[7-2] the antigen-binding molecule of [6], wherein the second antigen-binding domain comprises an antibody variable region that has binding activity toward a molecule expressed on the surface of a cell that has the function of suppressing an immune response;
[8] The antigen-binding molecule of any one of [1] to [7], which is an antibody.
[9] A nucleic acid encoding the antigen-binding molecule of any one of [1] to [8].
[10] A vector into which the nucleic acid according to [9] has been introduced.
[11] A cell comprising the nucleic acid of [9] or the vector of [10].
[12] A method for producing the antigen-binding molecule of any one of [1] to [8], comprising the step of culturing the cell of [11].
[13] An antigen-binding molecule produced by the method according to [12].
[14] A pharmaceutical composition comprising the antigen-binding molecule of any of [1] to [8] and a pharmaceutically acceptable carrier.
[15-1] The pharmaceutical composition according to [14], for inducing cell damage.
[15-2] The pharmaceutical composition according to [14], for inducing T cell-dependent cytotoxicity.
[15-3] The pharmaceutical composition according to [14], for use in the treatment or prevention of cancer.
[16-1] A method for inducing cytotoxicity, comprising the step of administering the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14].
[16-2] A method for inducing T cell-dependent cytotoxicity, comprising the step of administering the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14].
[16-3] A method for treating or preventing cancer, comprising the step of administering the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14].
[17-1] A kit for inducing cytotoxicity, comprising the antigen-binding molecule of any one of [1] to [8] or the pharmaceutical composition of [14], and instructions for use.
[17-2] A kit for inducing T cell-dependent cytotoxicity, comprising the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14], and instructions for use.
[17-3] A kit for treating or preventing cancer, comprising the antigen-binding molecule of any of [1] to [8] or the pharmaceutical composition of [14], and instructions for use.
[18-1] The antigen-binding molecule of any one of [1] to [8], for use in inducing cytotoxicity.
[18-2] The antigen-binding molecule of any one of [1] to [8], for use in inducing T cell-dependent cytotoxicity.
[18-3] the antigen-binding molecule of any one of [1] to [8], for use in the treatment or prevention of cancer.
[19-1] Use of the antigen-binding molecule of any of [1] to [8] in the production of a cytotoxicity-inducing agent.
[19-2] Use of the antigen-binding molecule of any of [1] to [8] in the production of an inducer of T cell-dependent cytotoxicity.
[19-3] Use of the antigen-binding molecule of any of [1] to [8] in the manufacture of a cancer treatment or prevention agent.
[A1] A monovalent antigen-binding molecule comprising a single antigen-binding domain, wherein the single antigen-binding domain comprises an antibody H chain variable region and an L chain variable region that have binding activity to CD3;
the heavy chain variable region
H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1);
an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3), wherein X3 is A or P, and X4 is I, M, or L.
Including,
the light chain variable region
an L chain CDR1 comprising the amino acid sequence RSX5X6X7VVHENRX8TYLH ( SEQ ID NO: 4 ), wherein X5 is S or T, X6 is Q or M, X7 is S or T, and X8 is Q or N;
L chain CDR2 comprising the amino acid sequence KVSNRFS (SEQ ID NO: 5); and L chain CDR3 comprising the amino acid sequence GQGTQVPYT (SEQ ID NO: 6).
An antigen-binding molecule comprising:
[A2] The antigen-binding molecule of [A1], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain CDR1, CDR2, and CDR3, and L chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8):
(a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a3) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 16, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a4) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a5) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 13, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a6) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 17, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a7) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 18, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a8) A combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 19, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6.
[A3] The antigen-binding molecule of [A1] or [A2], wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain variable region and L chain variable region selected from the following (a1) to (a8):
(a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26;
(a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a5) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 23 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a6) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 27;
(a7) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 28;
(a8) A combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 29.
[A4-1] The antigen-binding molecule of any of [A1] to [A3], which further comprises an Fc region.
[A4-2] The antigen-binding molecule of any of [A1] to [A3], further comprising an Fc region with reduced binding activity to an Fcγ receptor.
[A4-3] The antigen-binding molecule of [A4-1] or [A4-2], which comprises any one of the combinations of heavy chains and light chains selected from the following (a1) to (a8):
(a1) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 30 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a2) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a3) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 36;
(a4) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a5) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 33 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a6) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 37;
(a7) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 38;
(a8) A combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 39.
[A5] A nucleic acid encoding the antigen-binding molecule of any of [A1] to [A4].
[A6] A vector into which the nucleic acid described in [A5] has been introduced.
[A7] A cell comprising the nucleic acid according to [A5] or the vector according to [A6].
[A8] A method for producing the antigen-binding molecule of any of [A1] to [A4], comprising the step of culturing the cell of [A7].
[A9] An antigen-binding molecule produced by the method described in [A8].

I.定義
 以下の定義は、本明細書において説明する本発明の理解を容易にするために提供される。
I. Definitions The following definitions are provided to facilitate understanding of the invention described herein.

 用語「抗原結合分子」は、その抗原に充分なアフィニティで結合することのできる分子のことをいう。一態様において、その抗原とは無関係なタンパク質への抗原結合分子の結合の程度は、(例えば、放射免疫測定法 (radioimmunoassay: RIA) により)測定したとき、当該抗原結合分子の当該抗原への結合の約10%未満である。特定の態様において、抗原結合分子は、その抗原に対して、≦1μM、≦100nM、≦10nM、≦1nM、≦0.1nM、≦0.01nM、または≦0.001nM(例えば、10-8M以下、例えば10-8M~10-13M、例えば、10-9M~10-13M)の解離定数 (Kd) を有する。特定の態様において、抗原結合分子は、異なる種からのその抗原の間で保存されている当該抗原のエピトープに結合する。いくつかの態様において、抗原結合分子は抗体である。 The term "antigen-binding molecule" refers to a molecule capable of binding to its antigen with sufficient affinity. In one embodiment, the extent of binding of an antigen-binding molecule to proteins unrelated to the antigen is less than about 10% of the binding of the antigen-binding molecule to the antigen, as measured (e.g., by radioimmunoassay (RIA)). In certain embodiments, the antigen-binding molecule has a dissociation constant (Kd) for the antigen of ≦1 μM, ≦100 nM, ≦10 nM, ≦1 nM, ≦0.1 nM, ≦0.01 nM, or ≦0.001 nM (e.g., 10 −8 M or less, e.g., 10 −8 M to 10 −13 M, e.g., 10 −9 M to 10 −13 M). In certain embodiments, the antigen-binding molecule binds to an epitope of the antigen that is conserved among the antigens from different species. In some embodiments, the antigen-binding molecule is an antibody.

 本明細書において、「抗体」とは、天然のものであるかまたは部分的もしくは完全合成により製造された免疫グロブリンをいう。抗体はそれが天然に存在する血漿や血清等の天然資源や抗体を産生するハイブリドーマ細胞の培養上清から単離され得るし、または遺伝子組換え等の手法を用いることによって部分的にもしくは完全に合成され得る。抗体の例としては免疫グロブリンのアイソタイプおよびそれらのアイソタイプのサブクラスが好適に挙げられる。ヒトの免疫グロブリンとして、IgG1、IgG2、IgG3、IgG4、IgA1、IgA2、IgD、IgE、IgMの9種類のクラス(アイソタイプ)が知られている。本発明の抗体には、これらのアイソタイプのうちIgG1、IgG2、IgG3、IgG4が含まれ得る。 As used herein, the term "antibody" refers to an immunoglobulin that is natural or partially or fully synthetically produced. Antibodies can be isolated from natural sources such as plasma or serum where they occur, or from the culture supernatant of antibody-producing hybridoma cells, or can be partially or fully synthesized using techniques such as genetic recombination. Suitable examples of antibodies include immunoglobulin isotypes and their isotype subclasses. Nine known classes (isotypes) of human immunoglobulins are IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgD, IgE, and IgM. Of these isotypes, antibodies of the present invention may include IgG1, IgG2, IgG3, and IgG4.

 抗体の「クラス」は、抗体の重鎖に備わる定常ドメインまたは定常領域のタイプのことをいう。抗体には5つの主要なクラスがある:IgA、IgD、IgE、IgG、およびIgMである。そして、このうちいくつかはさらにサブクラス(アイソタイプ)に分けられてもよい。例えば、IgG1、IgG2、IgG3、IgG4、IgA1、およびIgA2である。異なるクラスの免疫グロブリンに対応する重鎖定常ドメインを、それぞれ、α、δ、ε、γ、およびμと呼ぶ。 An antibody's "class" refers to the type of constant domain or constant region present in its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM. Some of these may be further divided into subclasses (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. The heavy-chain constant domains corresponding to the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively.

 「天然型抗体」は、天然に生じる様々な構造を伴う免疫グロブリン分子のことをいう。例えば、天然型IgG抗体は、ジスルフィド結合している2つの同一の軽鎖と2つの同一の重鎖から構成される約150,000ダルトンのヘテロ四量体糖タンパク質である。N末端からC末端に向かって、各重鎖は、可変重鎖ドメインまたは重鎖可変ドメインとも呼ばれる可変領域 (VH) を有し、それに3つの定常ドメイン(CH1、CH2、およびCH3)が続く。同様に、N末端からC末端に向かって、各軽鎖は、可変軽鎖ドメインまたは軽鎖可変ドメインとも呼ばれる可変領域 (VL) を有し、それに定常軽鎖 (CL) ドメインが続く。抗体の軽鎖は、その定常ドメインのアミノ酸配列に基づいて、カッパ(κ)およびラムダ(λ)と呼ばれる、2つのタイプの1つに帰属させられてよい。 "Native antibodies" refer to immunoglobulin molecules with a variety of naturally occurring structures. For example, native IgG antibodies are heterotetrameric glycoproteins of approximately 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From the N-terminus to the C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3). Similarly, from the N-terminus to the C-terminus, each light chain has a variable region (VL), also called a variable light domain or light chain variable domain, followed by a constant light (CL) domain. The light chains of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of their constant domains.

 用語「可変領域」または「可変ドメイン」は、抗体を抗原へと結合させることに関与する、抗体の重鎖または軽鎖のドメインのことをいう。天然型抗体の重鎖および軽鎖の可変ドメイン(それぞれVHおよびVL)は、通常、各ドメインが4つの保存されたフレームワーク領域 (FR) および3つの超可変領域 (HVR) を含む、類似の構造を有する。(例えば、Kindt et al. Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007) 参照。)1つのVHまたはVLドメインで、抗原結合特異性を与えるに充分であろう。さらに、ある特定の抗原に結合する抗体は、当該抗原に結合する抗体からのVHまたはVLドメインを使ってそれぞれVLまたはVHドメインの相補的ライブラリをスクリーニングして、単離されてもよい。例えばPortolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991) 参照。 The terms "variable region" or "variable domain" refer to the domains of an antibody heavy or light chain that are involved in binding the antibody to antigen. The heavy and light chain variable domains (VH and VL, respectively) of natural antibodies typically have a similar structure, with each domain containing four conserved framework regions (FR) and three hypervariable regions (HVR). (See, e.g., Kindt et al., Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007)). A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind to a particular antigen may be isolated by screening a complementary library of VL or VH domains, respectively, with a VH or VL domain from an antibody that binds to that antigen. See, for example, Portolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).

 本明細書で用いられる用語「超可変領域」または「HVR」は、配列において超可変であり(「相補性決定領域」または「CDR」(complementarity determining region))、および/または構造的に定まったループ(「超可変ループ」)を形成し、および/または抗原接触残基(「抗原接触」)を含む、抗体の可変ドメインの各領域のことをいう。通常、抗体は6つのHVRを含む:VHに3つ(H1、H2、H3)、およびVLに3つ(L1、L2、L3)である。本明細書での例示的なHVRは、以下のものを含む:
 (a) アミノ酸残基26-32 (L1)、50-52 (L2)、91-96 (L3)、26-32 (H1)、53-55 (H2)、および96-101 (H3)のところで生じる超可変ループ (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987));
 (b) アミノ酸残基24-34 (L1)、50-56 (L2)、89-97 (L3)、31-35b (H1)、50-65 (H2)、 および95-102 (H3)のところで生じるCDR (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991));
 (c) アミノ酸残基27c-36 (L1)、46-55 (L2)、89-96 (L3)、30-35b (H1)、47-58 (H2)、および93-101 (H3) のところで生じる抗原接触 (MacCallum et al. J. Mol. Biol. 262: 732-745 (1996));ならびに、
 (d) HVRアミノ酸残基46-56 (L2)、47-56 (L2)、48-56 (L2)、49-56 (L2)、26-35 (H1)、26-35b (H1)、49-65 (H2)、93-102 (H3)、および94-102 (H3)を含む、(a)、(b)、および/または(c)の組合せ。
 別段示さない限り、HVR残基および可変ドメイン中の他の残基(例えば、FR残基)は、本明細書では上記のKabatらにしたがって番号付けされる。
As used herein, the term "hypervariable region" or "HVR" refers to each region of an antibody variable domain that is hypervariable in sequence (the "complementarity determining region" or "CDR") and/or forms structurally defined loops (the "hypervariable loops") and/or contains antigen-contacting residues (the "antigen contacts"). Typically, antibodies contain six HVRs: three in the VH (H1, H2, H3) and three in the VL (L1, L2, L3). Exemplary HVRs herein include the following:
(a) hypervariable loops occurring at amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3) (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987));
(b) CDRs occurring at amino acid residues 24-34 (L1), 50-56 (L2), 89-97 (L3), 31-35b (H1), 50-65 (H2), and 95-102 (H3) (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991));
(c) antigen contacts occurring at amino acid residues 27c-36 (L1), 46-55 (L2), 89-96 (L3), 30-35b (H1), 47-58 (H2), and 93-101 (H3) (MacCallum et al. J. Mol. Biol. 262: 732-745 (1996)); and
(d) A combination of (a), (b), and/or (c), comprising HVR amino acid residues 46-56 (L2), 47-56 (L2), 48-56 (L2), 49-56 (L2), 26-35 (H1), 26-35b (H1), 49-65 (H2), 93-102 (H3), and 94-102 (H3).
Unless otherwise indicated, HVR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Kabat et al., supra.

 「フレームワーク」または「FR」は、超可変領域 (HVR) 残基以外の、可変ドメイン残基のことをいう。可変ドメインのFRは、通常4つのFRドメイン:FR1、FR2、FR3、およびFR4からなる。それに応じて、HVRおよびFRの配列は、通常次の順序でVH(またはVL)に現れる:FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4。 "Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain typically consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences typically appear in VH (or VL) in the following order: FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.

 本明細書で用語「Fc領域」は、少なくとも定常領域の一部分を含む免疫グロブリン重鎖のC末端領域を定義するために用いられる。この用語は、天然型配列のFc領域および変異体Fc領域を含む。一態様において、ヒトIgG重鎖Fc領域はCys226から、またはPro230から、重鎖のカルボキシル末端まで延びる。ただし、Fc領域のC末端のリジン (Lys447) またはグリシン‐リジン(Gly446-Lys447)は、存在していてもしていなくてもよい。一態様において、Fc領域のC末端のリジン (Lys447) またはグリシン‐リジン(Gly446-Lys447)は、分解によって除去されてもよい。本明細書では別段特定しない限り、Fc領域または定常領域中のアミノ酸残基の番号付けは、Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD 1991 に記載の、EUナンバリングシステム(EUインデックスとも呼ばれる)にしたがう。 The term "Fc region" is used herein to define the C-terminal region of an immunoglobulin heavy chain comprising at least a portion of the constant region. This term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226 or from Pro230 to the carboxyl terminus of the heavy chain, except that the C-terminal lysine (Lys447) or glycine-lysine (Gly446-Lys447) residue at the Fc region may or may not be present. In one embodiment, the C-terminal lysine (Lys447) or glycine-lysine (Gly446-Lys447) residue at the Fc region may be removed by degradation. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system (also known as the EU index) as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD 1991.

 「天然型配列Fc領域」は、自然界で見出されるFc領域のアミノ酸配列と同一のアミノ酸配列を含む。天然型配列ヒトFc領域は、天然型配列ヒトIgG1 Fc領域(非AおよびAアロタイプ);天然型配列ヒトIgG2 Fc領域;天然型配列ヒトIgG3 Fc領域;および天然型配列ヒトIgG4 Fc領域、ならびに天然に存在するそれらの変異体を含む。  A "native sequence Fc region" comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature. Native sequence human Fc regions include native sequence human IgG1 Fc regions (non-A and A allotypes); native sequence human IgG2 Fc regions; native sequence human IgG3 Fc regions; and native sequence human IgG4 Fc regions, as well as naturally occurring variants thereof.

 「変異Fc領域」は、少なくとも1つのアミノ酸修飾、好ましくは1つまたは複数のアミノ酸置換によって天然型配列Fc領域のそれと相違するアミノ酸配列を含む。好ましくは、変異Fc領域は、天然型配列Fc領域または親ポリペプチドのFc領域と比較して、天然型配列Fc領域中または親ポリペプチドのFc領域中に少なくとも1つのアミノ酸置換、例えば約1~約10個のアミノ酸置換、好ましくは約1~約5個のアミノ酸置換を有する。本明細書の変異Fc領域は、好ましくは、天然型配列Fc領域および/または親ポリペプチドのFc領域と少なくとも約80%の配列同一性、より好ましくはそれらと少なくとも約90%の配列同一性、最も好ましくはそれらと少なくとも約95%の配列同一性を備える。 A "variant Fc region" comprises an amino acid sequence that differs from that of a native-sequence Fc region by at least one amino acid modification, preferably one or more amino acid substitutions. Preferably, the variant Fc region has at least one amino acid substitution, e.g., about 1 to about 10 amino acid substitutions, preferably about 1 to about 5 amino acid substitutions, in the native-sequence Fc region or in the Fc region of the parent polypeptide compared to the native-sequence Fc region or the Fc region of the parent polypeptide. The variant Fc region herein preferably has at least about 80% sequence identity with the native-sequence Fc region and/or the Fc region of the parent polypeptide, more preferably at least about 90% sequence identity thereto, and most preferably at least about 95% sequence identity thereto.

 「Fc受容体」または「FcR」は、抗体のFc領域に結合する受容体のことをいう。いくつかの態様において、FcRは、天然型ヒトFcRである。いくつかの態様において、FcRは、IgG抗体に結合するもの(ガンマ受容体)であり、FcγRI、FcγRII、およびFcγRIIIサブクラスの受容体を、これらの受容体の対立遺伝子変異体および選択的スプライシングによる形態を含めて、含む。FcγRII受容体は、FcγRIIA(「活性化受容体」)およびFcγRIIB(「阻害受容体」)を含み、これらは主としてその細胞質ドメインにおいて相違する類似のアミノ酸配列を有する。活性化受容体FcγRIIAは、その細胞質ドメインに免疫受容体チロシン活性化モチーフ (immunoreceptor tyrosine-based activation motif: ITAM) を含む。阻害受容体FcγRIIBは、その細胞質ドメインに免疫受容体チロシン阻害モチーフ(immunoreceptor tyrosine-based inhibition motif: ITIM)を含む。(例えば、Daeron, Annu. Rev. Immunol. 15:203-234 (1997) を参照のこと。)FcRは、例えば、Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991);Capel et al., Immunomethods 4:25-34 (1994);およびde Haas et al., J. Lab. Clin. Med 126:330-41 (1995)において総説されている。将来同定されるものを含む他のFcRも、本明細書の用語「FcR」に包含される。 "Fc receptor" or "FcR" refers to a receptor that binds to the Fc region of an antibody. In some embodiments, the FcR is a native human FcR. In some embodiments, the FcR is one that binds IgG antibodies (gamma receptors) and includes receptors of the FcγRI, FcγRII, and FcγRIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcγRII receptors include FcγRIIA ("activating receptors") and FcγRIIB ("inhibiting receptors"), which have similar amino acid sequences that differ primarily in their cytoplasmic domains. Activating receptor FcγRIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain. Inhibiting receptor FcγRIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain. (See, e.g., Daeron, Annu. Rev. Immunol. 15:203-234 (1997).) FcRs are reviewed, e.g., in Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991); Capel et al., Immunomethods 4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med 126:330-41 (1995). Other FcRs, including those identified in the future, are also encompassed by the term "FcR" herein.

 用語「Fc受容体」または「FcR」はまた、母体のIgGの胎児への移動(Guyer et al., J. Immunol. 117:587 (1976)およびKim et al., J. Immunol. 24:249 (1994))ならびに免疫グロブリンのホメオスタシスの調節を担う、新生児型受容体FcRnを含む。FcRnへの結合を測定する方法は公知である(例えば、Ghetie and Ward., Immunol. Today 18(12):592-598 (1997); Ghetie et al., Nature Biotechnology, 15(7):637-640 (1997); Hinton et al., J. Biol. Chem. 279(8):6213-6216 (2004); WO2004/92219 (Hinton et al.)を参照のこと)。 The term "Fc receptor" or "FcR" also includes the neonatal receptor FcRn, which is responsible for regulating maternal IgG transfer to the fetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)) and immunoglobulin homeostasis. Methods for measuring binding to FcRn are known (see, e.g., Ghetie and Ward., Immunol. Today 18(12):592-598 (1997); Ghetie et al., Nature Biotechnology, 15(7):637-640 (1997); Hinton et al., J. Biol. Chem. 279(8):6213-6216 (2004); WO2004/92219 (Hinton et al.)).

 インビボでのヒトFcRnへの結合およびヒトFcRn高アフィニティ結合ポリペプチドの血漿半減期は、例えばヒトFcRnを発現するトランスジェニックマウスもしくはトランスフェクトされたヒト細胞株においてまたは変異Fc領域を伴うポリペプチドが投与される霊長類において測定され得る。血漿半減期の代わりに、血中半減期または血清半減期が測定され得る。WO2000/42072 (Presta) は、FcRに対する結合が増加したまたは減少した抗体変異体を記載している。例えば、Shields et al. J. Biol. Chem. 9(2):6591-6604 (2001) も参照のこと。 In vivo binding to human FcRn and plasma half-life of human FcRn high-affinity binding polypeptides can be measured, for example, in transgenic mice or transfected human cell lines expressing human FcRn, or in primates to which polypeptides with mutant Fc regions are administered. Instead of plasma half-life, blood half-life or serum half-life can be measured. WO2000/42072 (Presta) describes antibody variants with increased or decreased binding to FcRs. See also, e.g., Shields et al. J. Biol. Chem. 9(2):6591-6604 (2001).

 用語「Fc領域含有抗体」は、Fc領域を含む抗体のことをいう。Fc領域のC末端リジン(EUナンバリングシステムにしたがえば残基447)またはFc領域のC末端グリシン-リジン(残基446-447)は、例えば抗体の精製の間にまたは抗体をコードする核酸の組み換え操作によって除去され得る。したがって、本発明によるFc領域を有する抗体を含む組成物は、G446-K447を伴う抗体、G446を伴いK447を伴わない抗体、G446-K447が完全に除去された抗体(IgGの場合、C末端がP445である抗体若しくはP445にアミド基(NH2)が付加したプロリンアミド(Pro-NH2)となっている抗体)、または上記3つのタイプの抗体の混合物を含み得る。 The term "Fc region-containing antibody" refers to an antibody that contains an Fc region. The C-terminal lysine (residue 447 according to the EU numbering system) or the C-terminal glycine-lysine (residues 446-447) of the Fc region can be removed, for example, during antibody purification or by recombinant manipulation of the nucleic acid encoding the antibody. Thus, a composition containing an antibody with an Fc region according to the present invention can include an antibody with G446-K447, an antibody with G446 but no K447, an antibody from which G446-K447 have been completely removed (in the case of IgG, an antibody whose C-terminus is P445 or an antibody with an amide group (NH2) added to P445, resulting in proline amide (Pro-NH2)), or a mixture of the above three types of antibodies.

 「エフェクター機能」は、抗体のFc領域に起因する、抗体のアイソタイプによって異なる生物学的活性のことをいう。抗体のエフェクター機能の例には次のものが含まれる:C1q結合および補体依存性細胞傷害(complement dependent cytotoxicity:CDC);Fc受容体結合;抗体依存性細胞介在性細胞傷害(antibody-dependent cell-mediated cytotoxicity: ADCC);貪食作用;細胞表面受容体(例えば、B細胞受容体)の下方制御;および、B細胞活性化。 "Effector function" refers to a biological activity attributable to the Fc region of an antibody, which varies depending on the antibody isotype. Examples of antibody effector functions include: C1q binding and complement-dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down-regulation of cell surface receptors (e.g., B cell receptors); and B cell activation.

 「抗体依存性細胞介在性細胞傷害」または「ADCC」は、分泌されたIgが特定の細胞傷害性細胞(例えば、NK細胞、好中球およびマクロファージ)上に存在するFc受容体 (FcR) に結合しそれによってこれらの細胞傷害性エフェクター細胞が抗原を有する標的細胞に特異的に結合することができそしてその後にその標的細胞を細胞毒によって殺傷することができるようになる、細胞傷害の一形態のことをいう。ADCCを媒介するプライマリ細胞であるNK細胞はFcγRIIIのみを発現し、単球はFcγRI、FcγRII、およびFcγRIIIを発現する。造血細胞上のFcRの発現は、Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991) の第464頁の表3にまとめられている。目的の分子のADCC活性を評価するために、インビトロADCC測定法、例えば米国特許第5,500,362号もしくは第5,821,337号または米国特許第6,737,056号 (Presta) に記載のものが実施され得る。そのような測定法に有用なエフェクター細胞は、PBMCおよびNK細胞を含む。あるいはまたは加えて、目的の分子のADCC活性は、例えばClynes et al. PNAS (USA) 95:652-656 (1998)に開示される動物モデルのような動物モデルにおいて、インビボで評価されてもよい。 "Antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to a form of cytotoxicity in which secreted Ig binds to Fc receptors (FcRs) present on certain cytotoxic cells (e.g., NK cells, neutrophils, and macrophages), thereby enabling these cytotoxic effector cells to specifically bind to antigen-bearing target cells and subsequently kill them with cytotoxins. NK cells, the primary cells mediating ADCC, express only FcγRIII, whereas monocytes express FcγRI, FcγRII, and FcγRIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991). To assess ADCC activity of a molecule of interest, in vitro ADCC assays, such as those described in U.S. Pat. Nos. 5,500,362 or 5,821,337 or U.S. Pat. No. 6,737,056 (Presta), may be performed. Effector cells useful for such assays include PBMCs and NK cells. Alternatively, or additionally, ADCC activity of a molecule of interest may be assessed in vivo in an animal model, such as that disclosed in Clynes et al. PNAS (USA) 95:652-656 (1998).

 本明細書でいう用語「モノクローナル抗体」は、実質的に均一な抗体の集団から得られる抗体のことをいう。すなわち、その集団を構成する個々の抗体は、生じ得る変異抗体(例えば、自然に生じる変異を含む変異抗体、またはモノクローナル抗体調製物の製造中に発生する変異抗体。そのような変異体は通常若干量存在している。)を除いて、同一でありおよび/または同じエピトープ(決定基)に結合する。異なるエピトープに対する異なる抗体を典型的に含むポリクローナル抗体調製物とは対照的に、モノクローナル抗体調製物に含まれる各モノクローナル抗体は、抗原上の単一のエピトープに対するものである。したがって、修飾語「モノクローナル」は、実質的に均一な抗体の集団から得られるものである、という抗体の特徴を示し、何らかの特定の方法による抗体の製造を求めるものと解釈されるべきではない。例えば、本発明にしたがって用いられるモノクローナル抗体は、これらに限定されるものではないが、ハイブリドーマ法、組換えDNA法、ファージディスプレイ法、ヒト免疫グロブリン遺伝子座の全部または一部を含んだトランスジェニック動物を利用する方法を含む、様々な手法によって作製されてよい。 As used herein, the term "monoclonal antibody" refers to an antibody obtained from a population of substantially homogeneous antibodies. That is, the individual antibodies comprising the population are identical and/or bind the same epitope (determinant), except for possible variants (e.g., variants containing naturally occurring mutations or variants that arise during the production of a monoclonal antibody preparation; such variants are typically present in small amounts). In contrast to polyclonal antibody preparations, which typically contain different antibodies directed against different epitopes, each monoclonal antibody in a monoclonal antibody preparation is directed against a single epitope on an antigen. Thus, the modifier "monoclonal" indicates the character of the antibody as being obtained from a population of substantially homogeneous antibodies, and should not be construed as requiring production of the antibody by any particular method. For example, monoclonal antibodies for use in accordance with the present invention may be produced by a variety of techniques, including, but not limited to, hybridoma technology, recombinant DNA technology, phage display technology, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci.

 用語「キメラ」抗体は、重鎖および/または軽鎖の一部分が特定の供給源または種に由来する一方で、重鎖および/または軽鎖の残りの部分が異なった供給源または種に由来する抗体のことをいう。 The term "chimeric" antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.

 「ヒト化」抗体は、非ヒトHVRからのアミノ酸残基およびヒトFRからのアミノ酸残基を含む、キメラ抗体のことをいう。ある態様では、ヒト化抗体は、少なくとも1つ、典型的には2つの可変ドメインの実質的にすべてを含み、当該可変領域においては、すべてのもしくは実質的にすべてのHVR(例えばCDR)は非ヒト抗体のものに対応し、かつ、すべてのもしくは実質的にすべてのFRはヒト抗体のものに対応する。ヒト化抗体は、任意で、ヒト抗体に由来する抗体定常領域の少なくとも一部分を含んでもよい。抗体(例えば、非ヒト抗体)の「ヒト化された形態」は、ヒト化を経た抗体のことをいう。 A "humanized" antibody refers to a chimeric antibody that contains amino acid residues from non-human HVRs and human FRs. In certain embodiments, a humanized antibody contains substantially all of at least one, and typically two, variable domains, in which all or substantially all HVRs (e.g., CDRs) correspond to those of a non-human antibody and all or substantially all FRs correspond to those of a human antibody. A humanized antibody may optionally contain at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody (e.g., a non-human antibody) refers to an antibody that has undergone humanization.

 「ヒト抗体」は、ヒトもしくはヒト細胞によって産生された抗体またはヒト抗体レパートリーもしくは他のヒト抗体コード配列を用いる非ヒト供給源に由来する抗体のアミノ酸配列に対応するアミノ酸配列を備える抗体である。このヒト抗体の定義は、非ヒトの抗原結合残基を含むヒト化抗体を、明確に除外するものである。 A "human antibody" is an antibody with an amino acid sequence that corresponds to that of an antibody produced by a human or human cell, or an antibody derived from the human antibody repertoire or other non-human source that uses human antibody coding sequences. This definition of a human antibody specifically excludes humanized antibodies, which contain non-human antigen-binding residues.

 用語「全長抗体」、「完全抗体」、および「全部抗体」は、本明細書では相互に交換可能に用いられ、天然型抗体構造に実質的に類似した構造を有する、または本明細書で定義するFc領域を含む重鎖を有する抗体のことをいう。 The terms "full length antibody," "complete antibody," and "whole antibody" are used interchangeably herein and refer to an antibody having a structure substantially similar to that of a native antibody or having a heavy chain that includes an Fc region as defined herein.

 「抗体断片」は、完全抗体が結合する抗原に結合する当該完全抗体の一部分を含む、当該完全抗体以外の分子のことをいう。抗体断片の例は、これらに限定されるものではないが、Fv、Fab、Fab'、Fab’-SH、F(ab')2;ダイアボディ;線状抗体;単鎖抗体分子(例えば、scFv);および、抗体断片から形成された多重特異性抗原結合分子を含む。 An "antibody fragment" refers to a molecule other than a complete antibody that contains a portion of the complete antibody that binds to the antigen to which the complete antibody binds. Examples of antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabodies; linear antibodies; single-chain antibody molecules (e.g., scFv); and multispecific antigen-binding molecules formed from antibody fragments.

 本明細書において、「Fv(variable fragment)」という用語は、抗体の軽鎖可変領域(VL(light chain variable region))と抗体の重鎖可変領域(VH(heavy chain variable region))とのペアからなる抗体由来の抗原結合ドメインの最小単位を意味する。1988年にSkerraとPluckthunは、バクテリアのシグナル配列の下流に抗体の遺伝子を挿入し大腸菌中で当該遺伝子の発現を誘導することによって、Fvが均一でかつ活性を保持した状態で大腸菌のペリプラズム画分から調製されることを見出した(Science (1988) 240 (4855), 1038-1041)。ペリプラズム画分から調製されたFvは、抗原に対する結合を有する態様でVHとVLが会合していた。 As used herein, the term "Fv (variable fragment)" refers to the smallest unit of an antibody-derived antigen-binding domain consisting of a pair of an antibody light chain variable region (VL (light chain variable region)) and an antibody heavy chain variable region (VH (heavy chain variable region)). In 1988, Skerra and Pluckthun discovered that homogeneous and active Fv could be prepared from the periplasmic fraction of E. coli by inserting an antibody gene downstream of a bacterial signal sequence and inducing expression of the gene in E. coli (Science (1988) 240 (4855), 1038-1041). In the Fv prepared from the periplasmic fraction, VH and VL were associated in a manner that allowed them to bind to antigens.

 本明細書において、「scFv」、「単鎖抗体」、または「sc(Fv)2」という用語は、単一のポリペプチド鎖内に、重鎖および軽鎖の両方に由来する可変領域を含むが、定常領域を欠いている抗体断片を意味する。一般に、単鎖抗体は、抗原結合を可能にすると思われる所望の構造を形成するのを可能にする、VHドメインとVLドメインの間のポリペプチドリンカーをさらに含む。単鎖抗体は、The Pharmacology of Monoclonal Antibodies, 113巻, Rosenburg、及び、Moore編, Springer-Verlag, New York, 269~315(1994)においてPluckthunによって詳細に考察されている。同様に、国際特許出願公開WO1988/001649および米国特許第4,946,778号および同第5,260,203号を参照。特定の態様において、単鎖抗体はまた、二重特異性であるか、かつ/またはヒト化され得る。 As used herein, the term "scFv,""single-chainantibody," or "sc(Fv) 2 " refers to an antibody fragment that contains, in a single polypeptide chain, the variable regions from both the heavy and light chains, but lacks a constant region. Generally, single-chain antibodies further comprise a polypeptide linker between the VH and VL domains, which enables them to form the desired structure that may enable antigen binding. Single-chain antibodies are discussed in detail by Plückthun in *The Pharmacology of Monoclonal Antibodies*, Vol. 113, Rosenburg and Moore (eds.), Springer-Verlag, New York, pp. 269-315 (1994). See also International Patent Application Publication No. WO 1988/001649 and U.S. Pat. Nos. 4,946,778 and 5,260,203. In certain embodiments, single-chain antibodies may also be bispecific and/or humanized.

 scFvはFvを構成するVHとVLとがペプチドリンカーによって連結された抗原結合ドメインである(Proc. Natl. Acad. Sci. U.S.A. (1988) 85 (16), 5879-5883)。当該ペプチドリンカーによってVHとVLとが近接した状態に保持され得る。 scFv is an antigen-binding domain in which the VH and VL constituting the Fv are linked by a peptide linker (Proc. Natl. Acad. Sci. U.S.A. (1988) 85 (16), 5879-5883). The peptide linker allows the VH and VL to be held in close proximity.

 sc(Fv)2は二つのVLと二つのVHの4つの可変領域がペプチドリンカー等のリンカーによって連結され一本鎖を構成する単鎖抗体である(J Immunol. Methods (1999) 231 (1-2), 177-189)。この二つのVHとVLは異なるモノクローナル抗体から由来することもあり得る。例えば、Journal of Immunology (1994) 152 (11), 5368-5374に開示されるような同一抗原中に存在する二種類のエピトープを認識する二重特異性sc(Fv)2も好適に挙げられる。sc(Fv)2は、当業者に公知の方法によって作製され得る。例えば、scFvをペプチドリンカー等のリンカーで結ぶことによって作製され得る。 sc(Fv) 2 is a single-chain antibody in which four variable regions, two VL and two VH, are linked by a linker such as a peptide linker to form a single chain (J Immunol. Methods (1999) 231 (1-2), 177-189). The two VH and VL may be derived from different monoclonal antibodies. Suitable examples include bispecific sc(Fv) 2s that recognize two different epitopes present in the same antigen, as disclosed in Journal of Immunology (1994) 152 (11), 5368-5374. sc(Fv) 2s can be produced by methods known to those skilled in the art. For example, they can be produced by linking scFvs with a linker such as a peptide linker.

 本明細書におけるsc(Fv)2を構成する抗原結合ドメインの構成としては、二つのVH及び二つのVLが、一本鎖ポリペプチドのN末端側を基点としてVH、VL、VH、VL([VH]リンカー[VL]リンカー[VH]リンカー[VL])の順に並んでいることを特徴とする抗原結合分子が挙げられるが、二つのVHと2つのVLの順序は特に上記の構成に限定されず、どのような順序で並べられていてもよい。例えば以下のような、順序の構成も挙げることができる。
[VL]リンカー[VH]リンカー[VH]リンカー[VL]
[VH]リンカー[VL]リンカー[VL]リンカー[VH]
[VH]リンカー[VH]リンカー[VL]リンカー[VL]
[VL]リンカー[VL]リンカー[VH]リンカー[VH]
[VL]リンカー[VH]リンカー[VL]リンカー[VH]
As used herein, the configuration of the antigen-binding domain constituting sc(Fv) 2 includes antigen-binding molecules in which two VHs and two VLs are arranged in the following order, starting from the N-terminus of the single-chain polypeptide: VH, VL, VH, VL ([VH] linker-[VL] linker-[VH] linker-[VL]). However, the order of the two VHs and two VLs is not limited to the above configuration and may be arranged in any order. For example, the following order configurations are also possible:
[VL] linker [VH] linker [VH] linker [VL]
[VH] linker [VL] linker [VL] linker [VH]
[VH] linker [VH] linker [VL] linker [VL]
[VL] linker [VL] linker [VH] linker [VH]
[VL] linker [VH] linker [VL] linker [VH]

 sc(Fv)2の分子形態についてはWO2006/132352においても詳細に記載されており、当業者であればこれらの記載に基づいて、本明細書で開示される抗原結合分子の作製のために適宜所望のsc(Fv)2を作製することが可能である。 The molecular structure of sc(Fv) 2 is also described in detail in WO2006/132352, and those skilled in the art can prepare the desired sc(Fv) 2 appropriately for producing the antigen-binding molecules disclosed herein based on these descriptions.

 また本発明の抗原結合分子は、PEG等のキャリアー高分子や抗がん剤等の有機化合物をコンジュゲートしてもよい。また、本発明の抗原結合分子のアミノ酸配列に糖鎖付加配列を挿入することによって、糖鎖による所望の効果を得ることを目的として、好適に抗原結合分子に糖鎖が付加され得る。 The antigen-binding molecules of the present invention may also be conjugated to carrier polymers such as PEG or organic compounds such as anticancer drugs. Furthermore, by inserting a glycosylation sequence into the amino acid sequence of the antigen-binding molecules of the present invention, a glycosylation sequence can be suitably added to the antigen-binding molecules in order to obtain the desired effects of the glycosylation.

 抗体の可変領域を結合するリンカーとしては、遺伝子工学により導入し得る任意のペプチドリンカー、又は合成化合物リンカー(例えば、Protein Engineering, 9 (3), 299-305, 1996に開示されるリンカー等)を用いることができるが、本発明においてはペプチドリンカーが好ましい。ペプチドリンカーの長さは特に限定されず、目的に応じて当業者が適宜選択することが可能であるが、好ましい長さは5アミノ酸以上(上限は特に限定されないが、通常、30アミノ酸以下、好ましくは20アミノ酸以下)であり、特に好ましくは15アミノ酸である。sc(Fv)2に3つのペプチドリンカーが含まれる場合には、全て同じ長さのペプチドリンカーを用いてもよいし、異なる長さのペプチドリンカーを用いてもよい。ペプチドリンカーの例としては、グリシンとセリンで構成されるフレキシブルなポリペプチドリンカーが広く知られている。ペプチドリンカーの長さや配列は目的に応じて当業者が適宜選択することができる。 The linker linking the antibody variable regions can be any peptide linker that can be introduced by genetic engineering or a synthetic compound linker (e.g., the linkers disclosed in Protein Engineering, 9 (3), 299-305, 1996). In the present invention, however, a peptide linker is preferred. The length of the peptide linker is not particularly limited and can be selected appropriately by those skilled in the art depending on the purpose. A preferred length is 5 amino acids or more (the upper limit is not particularly limited, but typically 30 amino acids or less, preferably 20 amino acids or less), with 15 amino acids being particularly preferred. When sc(Fv) 2 contains three peptide linkers, all of the peptide linkers may be the same length, or peptide linkers of different lengths may be used. A widely known example of a peptide linker is a flexible polypeptide linker composed of glycine and serine. The length and sequence of the peptide linker can be selected appropriately by those skilled in the art depending on the purpose.

 合成化学物リンカー(化学架橋剤)は、ペプチドの架橋に通常用いられている架橋剤、例えばN-ヒドロキシスクシンイミド(NHS)、ジスクシンイミジルスベレート(DSS)、ビス(スルホスクシンイミジル)スベレート(BS3)、ジチオビス(スクシンイミジルプロピオネート)(DSP)、ジチオビス(スルホスクシンイミジルプロピオネート)(DTSSP)、エチレングリコールビス(スクシンイミジルスクシネート)(EGS)、エチレングリコールビス(スルホスクシンイミジルスクシネート)(スルホ-EGS)、ジスクシンイミジル酒石酸塩(DST)、ジスルホスクシンイミジル酒石酸塩(スルホ-DST)、ビス[2-(スクシンイミドオキシカルボニルオキシ)エチル]スルホン(BSOCOES)、ビス[2-(スルホスクシンイミドオキシカルボニルオキシ)エチル]スルホン(スルホ-BSOCOES)などであり、これらの架橋剤は市販されている。 Synthetic chemical linkers (chemical crosslinkers) are crosslinkers commonly used for crosslinking peptides, such as N-hydroxysuccinimide (NHS), disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl)suberate (BS3), dithiobis(succinimidyl propionate) (DSP), dithiobis(sulfosuccinimidyl propionate) (DTSSP), ethylene glycol bis(succinimidyl succinate) (EGS), ethylene glycol bis(sulfosuccinimidyl succinate) (sulfo-EGS), disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST), bis[2-(succinimidooxycarbonyloxy)ethyl]sulfone (BSOCOES), and bis[2-(sulfosuccinimidooxycarbonyloxy)ethyl]sulfone (sulfo-BSOCOES). These crosslinkers are commercially available.

 4つの抗体可変領域を結合する場合には、通常、3つのリンカーが必要となるが、全て同じリンカーを用いてもよいし、異なるリンカーを用いてもよい。 When linking four antibody variable regions, three linkers are usually required, but the same linker may be used for all three, or different linkers may be used.

 「Fab」は、一本の軽鎖、ならびに一本の重鎖のCH1領域および可変領域から構成される。Fab分子の重鎖は、別の重鎖分子とのジスルフィド結合を形成できない。 "Fab" consists of one light chain and the CH1 and variable regions of one heavy chain. The heavy chain of a Fab molecule cannot form disulfide bonds with another heavy chain molecule.

 「F(ab')2」及び「Fab'」とは、イムノグロブリン(モノクローナル抗体)をタンパク質分解酵素であるペプシンあるいはパパイン等で処理することにより製造され、ヒンジ領域中の2本のH鎖間に存在するジスルフィド結合の前後で消化されて生成される抗体フラグメントを意味する。例えば、IgGをパパインで処理することにより、ヒンジ領域中の2本のH鎖間に存在するジスルフィド結合の上流で切断されてVL(L鎖可変領域)とCL(L鎖定常領域)からなるL鎖、及びVH(H鎖可変領域)とCHγ1(H鎖定常領域中のγ1領域)とからなるH鎖フラグメントがC末端領域でジスルフィド結合により結合した相同な2つの抗体フラグメントが製造され得る。これら2つの相同な抗体フラグメントはそれぞれFab'といわれる。 The terms "F(ab') 2 " and "Fab'" refer to antibody fragments produced by treating an immunoglobulin (monoclonal antibody) with a proteolytic enzyme such as pepsin or papain, resulting in digestion across the disulfide bond between the two heavy chains in the hinge region. For example, treating IgG with papain cleaves it upstream of the disulfide bond between the two heavy chains in the hinge region, producing two homologous antibody fragments in which an light chain consisting of a VL (light chain variable region) and a CL (light chain constant region) and an heavy chain fragment consisting of a VH (heavy chain variable region) and a CHγ1 (the γ1 region of the heavy chain constant region) are linked by a disulfide bond at their C-terminal regions. These two homologous antibody fragments are each referred to as Fab'.

 「F(ab')2」は、二本の軽鎖、ならびに、鎖間のジスルフィド結合が2つの重鎖間で形成されるようにCH1ドメインおよびCH2ドメインの一部分の定常領域を含む二本の重鎖を含む。本明細書において開示される抗原結合分子を構成するF(ab')2は、所望の抗原結合ドメインを有する全長モノクローナル抗体等をペプシン等のタンパク質分解酵素にて部分消化した後に、Fc断片をプロテインAカラムに吸着させて除去することにより、好適に取得され得る。かかるタンパク質分解酵素としてはpH等の酵素の反応条件を適切に設定することにより制限的にF(ab')2を生じるように全長抗体を消化し得るものであれば特段の限定はされず、例えば、ペプシンやフィシン等が例示できる。 "F(ab') 2 " comprises two light chains and two heavy chains comprising constant regions, i.e., portions of the CH1 and CH2 domains, such that interchain disulfide bonds are formed between the two heavy chains. The F(ab') 2 constituting the antigen-binding molecule disclosed herein can be suitably obtained by partially digesting a full-length monoclonal antibody or the like having the desired antigen-binding domain with a protease such as pepsin, followed by removal of the Fc fragment by adsorption onto a protein A column. Such a protease is not particularly limited, as long as it can digest a full-length antibody so as to produce F(ab') 2 in a limited manner by appropriately setting the enzyme reaction conditions, such as pH, and examples thereof include pepsin and ficin.

 「アフィニティ」は、分子(例えば、抗体)の結合部位1個と、分子の結合パートナー(例えば、抗原)との間の、非共有結合的な相互作用の合計の強度のことをいう。別段示さない限り、本明細書で用いられる「結合アフィニティ」は、ある結合対のメンバー(例えば、抗体と抗原)の間の1:1相互作用を反映する、固有の結合アフィニティのことをいう。分子XのそのパートナーYに対するアフィニティは、一般的に、解離定数 (Kd) により表すことができる。アフィニティは、本明細書に記載のものを含む、当該技術分野において知られた通常の方法によって測定され得る。結合アフィニティを測定するための具体的な実例となるおよび例示的な態様については、下で述べる。 "Affinity" refers to the strength of the total non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and the molecule's binding partner (e.g., an antigen). Unless otherwise indicated, "binding affinity," as used herein, refers to the intrinsic binding affinity that reflects a 1:1 interaction between members of a binding pair (e.g., an antibody and an antigen). The affinity of a molecule X for its partner Y can generally be expressed by the dissociation constant (Kd). Affinity can be measured by conventional methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described below.

 「特異的結合」とは、特異的に結合する分子の一方の分子がその一または複数の結合する相手方の分子以外の分子に対しては何ら有意な結合を示さないことをいう。また、抗体可変領域を含むドメインが、ある抗原中に含まれる複数のエピトープのうち特定のエピトープに対して特異的である場合にも用いられる。また、抗体可変領域を含むドメインが結合するエピトープが複数の異なる抗原に含まれる場合には、当該抗体可変領域を含むドメインを有する抗原結合分子は当該エピトープを含む様々な抗原と結合することができる。 "Specific binding" refers to the state in which one of the specifically binding molecules does not exhibit any significant binding to any molecules other than the one or more molecules to which it binds. The term is also used when a domain containing an antibody variable region is specific to a particular epitope among multiple epitopes contained in an antigen. Furthermore, when the epitopes to which the domain containing an antibody variable region binds are contained in multiple different antigens, an antigen-binding molecule having the domain containing the antibody variable region can bind to various antigens containing that epitope.

 「エピトープ」という用語は、タンパク質性、非タンパク質性を問わず、抗原結合分子(例えば、抗体)が結合する抗原上の部位を指す。例えば、エピトープは、その構造によって定義され得る。また、当該エピトープを認識する抗原結合分子(例えば、抗体)の抗原に対する結合活性によっても当該エピトープが定義され得る。抗原がペプチド又はポリペプチドである場合には、エピトープを構成するアミノ酸残基によってエピトープを特定することも可能である。また、エピトープが糖鎖である場合には、特定の糖鎖構造によってエピトープを特定することも可能である。 The term "epitope" refers to a site on an antigen to which an antigen-binding molecule (e.g., an antibody), whether proteinaceous or non-proteinaceous, binds. For example, an epitope can be defined by its structure. Alternatively, an epitope can be defined by the binding activity of an antigen-binding molecule (e.g., an antibody) that recognizes the epitope. When the antigen is a peptide or polypeptide, the epitope can also be identified by the amino acid residues that make up the epitope. When the epitope is a glycan, the epitope can also be identified by a specific glycan structure.

 線状エピトープは、アミノ酸の一次構造が認識されたエピトープを含むエピトープである。線状エピトープには、典型的には、少なくとも3個、および最も普通には少なくとも5個、例えば約8個~約10個、または6個~20個のアミノ酸が固有の配列で含まれる。 Linear epitopes are epitopes that contain a recognized primary amino acid structure. Linear epitopes typically contain at least three, and most usually at least five, e.g., about 8 to about 10, or 6 to 20 amino acids in a unique sequence.

 立体構造エピトープは、線状エピトープとは対照的に、典型的にはエピトープを構成するアミノ酸は一次構造として連続的に存在しておらず、不連続のアミノ酸残基からなるエピトープである(不連続エピトープという場合もある)。立体構造エピトープは、線状エピトープと比べて増加した数のアミノ酸を包含する場合がある。立体構造エピトープの認識に関して、抗原結合分子(例えば、抗体)は、ペプチドまたはタンパク質の三次元構造を認識する。例えば、タンパク質分子が折り畳まれて三次元構造を形成する場合には、立体構造エピトープを形成するあるアミノ酸および/またはポリペプチド主鎖は並列となり、抗原結合分子(例えば、抗体)がエピトープを認識するのを可能にする。エピトープの立体構造を決定する方法には、X線結晶学、二次元核磁気共鳴分光学並びに部位特異的なスピン標識および電磁常磁性共鳴分光学が含まれるが、これらには限定されない。例えば、Epitope Mapping Protocols in Methods in Molecular Biology (1996)、第66巻、Morris(編)を参照。 In contrast to linear epitopes, conformational epitopes are epitopes in which the amino acids constituting the epitope are typically not contiguous as a primary structure but are composed of discontinuous amino acid residues (sometimes referred to as discontinuous epitopes). Conformational epitopes may encompass an increased number of amino acids compared to linear epitopes. In recognizing conformational epitopes, antigen-binding molecules (e.g., antibodies) recognize the three-dimensional structure of a peptide or protein. For example, when a protein molecule folds to form a three-dimensional structure, certain amino acids and/or polypeptide backbones that form a conformational epitope are juxtaposed, allowing antigen-binding molecules (e.g., antibodies) to recognize the epitope. Methods for determining the conformational structure of an epitope include, but are not limited to, X-ray crystallography, two-dimensional nuclear magnetic resonance spectroscopy, and site-directed spin labeling and electromagnetic paramagnetic resonance spectroscopy. See, for example, Epitope Mapping Protocols in Methods in Molecular Biology (1996), Vol. 66, Morris (ed.).

 エピトープに結合する抗原結合分子(例えば、抗体)の構造は、「パラトープ」、「抗原結合部位」、「抗原結合領域」、または「抗原結合ドメイン」と呼ばれる。エピトープとパラトープの間に作用する、水素結合、静電気力、ファンデルワールス力、疎水結合等により、エピトープとパラトープは安定して結合する。このエピトープとパラトープの間の結合力はアフィニティ(affinity)と呼ばれる。複数の抗原に対して複数の抗原結合分子(例えば、抗体)が結合するときの結合力の総和はアビディティ(avidity)と呼ばれる。例えば、複数の抗原結合ドメインを含む抗原結合分子(すなわち多価の抗原結合分子)が複数のエピトープに結合する際には、アフィニティが相乗的に働くため、アビディティはアフィニティよりも高くなる。 The structure of an antigen-binding molecule (e.g., an antibody) that binds to an epitope is called the "paratope," "antigen-binding site," "antigen-binding region," or "antigen-binding domain." The epitope and paratope bind stably due to hydrogen bonds, electrostatic forces, van der Waals forces, hydrophobic bonds, and other forces that act between them. The binding strength between these epitopes and paratopes is called affinity. When multiple antigen-binding molecules (e.g., antibodies) bind to multiple antigens, the sum of their binding strengths is called avidity. For example, when an antigen-binding molecule containing multiple antigen-binding domains (i.e., a multivalent antigen-binding molecule) binds to multiple epitopes, the affinities act synergistically, resulting in avidity being higher than affinity.

 参照抗原結合分子と「競合する抗原結合分子」は、競合アッセイにおいてその参照抗原結合分子の自身の抗原への結合を50%以上阻止する抗原結合分子のことをいい、また逆にいえば、参照抗原結合分子は、競合アッセイにおいて前述の抗原結合分子の自身の抗原への結合を50%以上阻止する。競合する抗原結合分子は参照抗原結合分子と同一又は重複するエピトープに結合する抗原結合分子、および参照抗原結合分子が結合するエピトープの十分近位にある隣接エピトープに結合し立体障害を起こす抗原結合分子を含む。例示的な競合アッセイが、本明細書で提供される。 An "antigen-binding molecule that competes with a reference antigen-binding molecule" refers to an antigen-binding molecule that inhibits the binding of the reference antigen-binding molecule to its own antigen by 50% or more in a competition assay; conversely, a reference antigen-binding molecule inhibits the binding of the aforementioned antigen-binding molecule to its own antigen by 50% or more in a competition assay. Competitive antigen-binding molecules include antigen-binding molecules that bind to the same or overlapping epitope as the reference antigen-binding molecule, and antigen-binding molecules that bind to an adjacent epitope that is sufficiently proximal to the epitope bound by the reference antigen-binding molecule and cause steric hindrance. An exemplary competition assay is provided herein.

 「単離された」抗体は、そのもともとの環境の成分から分離されたものである。いくつかの態様において、抗体は、例えば、電気泳動(例えば、SDS-PAGE、等電点分離法 (isoelectric focusing: IEF)、キャピラリー電気泳動)またはクロマトグラフ(例えば、イオン交換または逆相HPLC)で測定して、95%または99%を超える純度まで精製される。抗体の純度の評価のための方法の総説として、例えば、Flatman et al., J. Chromatogr. B 848:79-87 (2007) を参照のこと。 An "isolated" antibody is one that has been separated from components of its original environment. In some embodiments, the antibody is purified to greater than 95% or 99% purity, as measured, for example, by electrophoresis (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (e.g., ion exchange or reverse-phase HPLC). For a review of methods for assessing antibody purity, see, e.g., Flatman et al., J. Chromatogr. B 848:79-87 (2007).

 「単離された」核酸は、そのもともとの環境の成分から分離された核酸分子のことをいう。単離された核酸は、その核酸分子を通常含む細胞の中に含まれた核酸分子を含むが、その核酸分子は染色体外に存在しているかまたは本来の染色体上の位置とは異なる染色体上の位置に存在している。 "Isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its original environment. Isolated nucleic acid includes a nucleic acid molecule contained in a cell that normally contains that nucleic acid molecule, but where the nucleic acid molecule is present extrachromosomally or in a chromosomal location that is different from its natural chromosomal location.

 「抗原結合分子をコードする単離された核酸」は、好ましくは抗体の重鎖可変領域および軽鎖可変領域を含む抗原結合分子をコードする1つまたは複数の核酸分子のことをいい、1つのベクターまたは別々のベクターに乗っている核酸分子、および、宿主細胞中の1つまたは複数の位置に存在している核酸分子を含む。 "Isolated nucleic acid encoding an antigen-binding molecule" refers to one or more nucleic acid molecules that encode an antigen-binding molecule, preferably comprising an antibody heavy chain variable region and a light chain variable region, and includes nucleic acid molecules carried on a single vector or separate vectors, and nucleic acid molecules present at one or more locations in a host cell.

 本明細書で用いられる用語「ベクター」は、それが連結されたもう1つの核酸を増やすことができる、核酸分子のことをいう。この用語は、自己複製核酸構造としてのベクター、および、それが導入された宿主細胞のゲノム中に組み入れられるベクターを含む。あるベクターは、自身が動作的に連結された核酸の、発現をもたらすことができる。そのようなベクターは、本明細書では「発現ベクター」とも称される。ベクターはウイルスを用いる方法やエレクトロポレーション法などにより宿主細胞に導入することができるが、ベクターの導入は生体外での導入に限られるものではなく、ベクターを直接生体に導入することも可能である。 As used herein, the term "vector" refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. This term includes vectors as self-replicating nucleic acid structures and vectors that are integrated into the genome of a host cell into which they are introduced. Some vectors are capable of conferring expression of a nucleic acid to which they are operatively linked. Such vectors are also referred to herein as "expression vectors." Vectors can be introduced into host cells using methods such as viruses or electroporation, but vector introduction is not limited to ex vivo introduction; vectors can also be introduced directly into a living body.

 用語「宿主細胞」、「宿主細胞株」、および「宿主細胞培養物」は、相互に交換可能に用いられ、外来核酸を導入された細胞(そのような細胞の子孫を含む)のことをいう。宿主細胞は「形質転換体」および「形質転換細胞」を含み、これには初代の形質転換細胞および継代数によらずその細胞に由来する子孫を含む。子孫は、親細胞と核酸の内容において完全に同一でなくてもよく、変異を含んでいてもよい。オリジナルの形質転換細胞がスクリーニングされたまたは選択された際に用いられたものと同じ機能または生物学的活性を有する変異体子孫も、本明細書では含まれる。 The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells," which include the primary transformed cell and progeny derived from that cell regardless of the number of passages. The progeny may not be completely identical in nucleic acid content to the parent cell and may contain mutations. Mutant progeny that have the same function or biological activity as that for which the original transformed cell was screened or selected are also included herein.

 用語「薬学的製剤」および「医薬組成物」は、その中に含まれた有効成分の生物学的活性が効果を発揮し得るような形態にある調製物であって、かつ製剤が投与される対象に許容できない程度に毒性のある追加の要素を含んでいない、調製物のことをいう。 The terms "pharmaceutical formulation" and "pharmaceutical composition" refer to a preparation in a form that allows the biological activity of the active ingredient contained therein to be effective, and that does not contain additional components that are unacceptably toxic to the subject to whom the formulation is administered.

 「薬学的に許容される担体」は、対象に対して無毒な、薬学的製剤中または医薬組成物中の有効成分以外の成分のことをいう。薬学的に許容される担体は、これらに限定されるものではないが、緩衝液、賦形剤、安定化剤、または保存剤を含む。 "Pharmaceutically acceptable carrier" refers to any ingredient, other than an active ingredient, in a pharmaceutical formulation or composition that is non-toxic to a subject. Pharmaceutically acceptable carriers include, but are not limited to, buffers, excipients, stabilizers, or preservatives.

 「個体」または「対象」は哺乳動物である。哺乳動物は、これらに限定されるものではないが、飼育動物(例えば、ウシ、ヒツジ、ネコ、イヌ、ウマ)、霊長類(例えば、ヒト、およびサルなどの非ヒト霊長類)、ウサギ、ならびに、げっ歯類(例えば、マウスおよびラット)を含む。特定の態様では、個体または対象は、ヒトである。 An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domestic animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human.

 ある剤(例えば、薬学的製剤)の「有効量」は、所望の治療的または予防的結果を達成するために有効である、必要な用量におけるおよび必要な期間にわたっての、量のことをいう。 An "effective amount" of an agent (e.g., a pharmaceutical formulation) refers to the amount, at the dosage and for the period of time necessary, that is effective to achieve the desired therapeutic or prophylactic result.

 用語「添付文書」は、治療用品の商用パッケージに通常含まれ、そのような治療用品の使用に関する、適応症、用法、用量、投与方法、併用療法、禁忌、および/または警告についての情報を含む使用説明書のことをいうために用いられる。 The term "package insert" is used to refer to instructions typically included in commercial packaging for therapeutic products that contain information about the indications, usage, dosage, method of administration, concomitant therapy, contraindications, and/or warnings regarding the use of such therapeutic product.

 本明細書で用いられる「治療」(および、その文法上の派生語、例えば「治療する」、「治療すること」など)は、治療される個体の自然経過を改変することを企図した臨床的介入を意味し、予防のためにも、臨床的病態の経過の間にも実施され得る。治療の望ましい効果は、これらに限定されるものではないが、疾患の発生または再発の防止、症状の軽減、疾患による任意の直接的または間接的な病理的影響の減弱、転移の防止、疾患の進行速度の低減、疾患状態の回復または緩和、および寛解または改善された予後を含む。いくつかの態様において、本発明の抗体は、疾患の発症を遅らせる、または疾患の進行を遅くするために用いられる。 As used herein, "treatment" (and its grammatical derivatives, such as "treat" and "treating") refers to a clinical intervention intended to alter the natural course of the individual being treated and may be performed prophylactically or during the course of a clinical condition. Desirable effects of treatment include, but are not limited to, prevention of disease onset or recurrence, alleviation of symptoms, attenuation of any direct or indirect pathological effects of the disease, prevention of metastasis, reduction in the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, the antibodies of the invention are used to delay the onset of disease or slow the progression of disease.

 用語「細胞増殖性障害」および「増殖性障害」は、一定程度の異常な細胞増殖に関連する障害のことをいう。一態様において、細胞増殖性障害は、がんである。 The terms "cell proliferative disorder" and "proliferative disorder" refer to disorders associated with some degree of abnormal cell proliferation. In one embodiment, the cell proliferative disorder is cancer.

 用語「がん」および「がん性」は、調節されない細胞成長/増殖によって典型的に特徴づけられる哺乳動物における生理学的状態のことをいうまたは説明するものである。がんの例は、がん腫、リンパ腫(例えば、ホジキンおよび非ホジキンリンパ腫)、芽細胞腫、肉腫、および白血病を含むが、これらに限定されない。そのようながんのより詳細な例は、扁平上皮細胞がん、小細胞肺がん、非小細胞肺がん、肺の腺がん、肺の扁平上皮がん、腹膜のがん、肝細胞がん、消化管がん、膵がん、神経膠腫、子宮頸がん、卵巣がん、肝がん、膀胱がん、ヘパトーマ、乳がん、結腸がん、結腸直腸がん、子宮内膜または子宮がん、唾液腺がん、腎がん、肝がん、前立腺がん、外陰部がん、甲状腺がん、肝細胞がん、白血病および他のリンパ球増殖性障害、ならびに様々なタイプの頭頸部がんを含む。 The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation. Examples of cancer include, but are not limited to, carcinoma, lymphoma (e.g., Hodgkin's and non-Hodgkin's lymphoma), blastoma, sarcoma, and leukemia. More specific examples of such cancers include squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, cancer of the peritoneum, hepatocellular carcinoma, gastrointestinal cancer, pancreatic cancer, glioma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, liver cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatocellular carcinoma, leukemia and other lymphoproliferative disorders, and various types of head and neck cancer.

 用語「腫瘍」は、悪性か良性かによらず、すべての新生物性細胞成長および増殖ならびにすべての前がん性およびがん性細胞および組織のことをいう。用語「がん」、「がん性」、「細胞増殖性障害」、「増殖性障害」および「腫瘍」は、本明細書でいう場合、相互に排他的でない。 The term "tumor" refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms "cancer," "cancerous," "cell proliferative disorder," "proliferative disorder," and "tumor" are not mutually exclusive as used herein.

 「細胞成長または増殖の阻害」は、細胞の成長または増殖を少なくとも10%、20%、30%、40%、50%、60%、70%、80%、90%、95%、または100%だけ減少させることを意味し、細胞死を誘導することを含む。 "Inhibiting cell growth or proliferation" means reducing cell growth or proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, including inducing cell death.

 参照ポリペプチド配列に対する「パーセント (%) アミノ酸配列同一性」は、最大のパーセント配列同一性を得るように配列を整列させてかつ必要ならギャップを導入した後の、かつ、いかなる保存的置換も配列同一性の一部と考えないとしたときの、参照ポリペプチド配列中のアミノ酸残基と同一である候補配列中のアミノ酸残基の、百分率比として定義される。パーセントアミノ酸配列同一性を決める目的のアラインメントは、当該技術分野における技術の範囲内にある種々の方法、例えば、BLAST、BLAST-2、ALIGN、Megalign (DNASTAR) ソフトウェア、またはGENETYX(登録商標)(株式会社ゼネティックス)などの、公に入手可能なコンピュータソフトウェアを使用することにより達成することができる。当業者は、比較される配列の全長にわたって最大のアラインメントを達成するために必要な任意のアルゴリズムを含む、配列のアラインメントをとるための適切なパラメーターを決定することができる。 "Percent (%) amino acid sequence identity" to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical to amino acid residues in the reference polypeptide sequence, after aligning the sequences to achieve the maximum percent sequence identity and introducing gaps, if necessary, and after not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved by a variety of methods within the skill of the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN, Megalign (DNASTAR) software, or GENETYX® (Genetyx Corporation). Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms necessary to achieve maximum alignment over the entire length of the sequences being compared.

 ALIGN-2配列比較コンピュータプログラムは、ジェネンテック社の著作であり、そのソースコードは米国著作権庁 (U.S. Copyright Office, Wasington D.C., 20559) に使用者用書類とともに提出され、米国著作権登録番号TXU510087として登録されている。ALIGN-2プログラムは、ジェネンテック社 (Genentech, Inc., South San Francisco, California) から公に入手可能であるし、ソースコードからコンパイルしてもよい。ALIGN-2プログラムは、Digital UNIX V4.0Dを含むUNIXオペレーティングシステム上での使用のためにコンパイルされる。すべての配列比較パラメーターは、ALIGN-2プログラムによって設定され、変動しない。
 アミノ酸配列比較にALIGN-2が用いられる状況では、所与のアミノ酸配列Aの、所与のアミノ酸配列Bへの、またはそれとの、またはそれに対する%アミノ酸配列同一性(あるいは、所与のアミノ酸配列Bへの、またはそれとの、またはそれに対する、ある%アミノ酸配列同一性を有するまたは含む所与のアミノ酸配列A、ということもできる)は、次のように計算される:分率X/Yの100倍。ここで、Xは配列アラインメントプログラムALIGN-2によって、当該プログラムのAおよびBのアラインメントにおいて同一である一致としてスコアされたアミノ酸残基の数であり、YはB中のアミノ酸残基の全数である。アミノ酸配列Aの長さがアミノ酸配列Bの長さと等しくない場合、AのBへの%アミノ酸配列同一性は、BのAへの%アミノ酸配列同一性と等しくないことが、理解されるであろう。別段特に明示しない限り、本明細書で用いられるすべての%アミノ酸配列同一性値は、直前の段落で述べたとおりALIGN-2コンピュータプログラムを用いて得られるものである。
The ALIGN-2 sequence comparison computer program is the copyright of Genentech, Inc., and its source code, together with user documentation, has been filed with the U.S. Copyright Office, Washington, DC 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program is compiled for use on UNIX operating systems, including Digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
In situations where ALIGN-2 is used for amino acid sequence comparison, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (alternatively, one can say that a given amino acid sequence A has or contains a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: 100 times the fraction X/Y, where X is the number of amino acid residues scored by the sequence alignment program ALIGN-2 as identical matches in that program's alignment of A and B, and Y is the total number of amino acid residues in B. It will be understood that if the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless otherwise specified, all % amino acid sequence identity values used herein are obtained using the ALIGN-2 computer program as described in the immediately preceding paragraph.

 本明細書で用語「および/または」は、「および/または」の前後に記載される各対象またはそれらの任意の組合せを示すために用いられる。例えば、「A、Bおよび/またはC」は、「A」、「B」、「C」の各対象のほか、組み合わせ「AおよびB」、「AおよびC」、「BおよびC」、ならびに「AおよびBおよびC」も含む。 The term "and/or" is used herein to refer to each of the objects listed before and after "and/or" or any combination thereof. For example, "A, B and/or C" includes each of the objects "A," "B," and "C," as well as the combinations "A and B," "A and C," "B and C," and "A and B and C."

II.発明の詳細な説明
 本開示の発明は、概して、優れた安定性を有するCD3結合ドメインを含む抗原結合分子、それらを含む医薬組成物、およびそれらの使用に関する。具体的には、本発明の抗原結合分子は、従来のT細胞リダイレクティング抗体(例えば、WO2015174439に記載されている二重特異性抗体)に含まれるCD3結合ドメインと比べて安定性が改善されたCD3結合ドメインを含む。本発明の抗原結合分子は、安定性が改善された本発明のCD3結合ドメインを含む分子であればその構造は特に限定されず、天然型抗体構造を有する分子であってもよいし、他の人工的に設計された構造を有するポリペプチドであってもよい。
II. Detailed Description of the Invention The invention of this disclosure generally relates to antigen-binding molecules comprising a CD3-binding domain with excellent stability, pharmaceutical compositions comprising them, and uses thereof. Specifically, the antigen-binding molecules of the present invention comprise a CD3-binding domain with improved stability compared to the CD3-binding domain contained in conventional T cell-redirecting antibodies (e.g., the bispecific antibodies described in WO2015174439). The structure of the antigen-binding molecules of the present invention is not particularly limited, as long as they comprise the CD3-binding domain of the present invention with improved stability. The antigen-binding molecules may be molecules having a natural antibody structure or polypeptides having other artificially designed structures.

 いくつかの態様において、本発明の抗原結合分子は抗体である。いくつかの態様において、本発明の抗原結合分子は、キメラ、ヒト化、またはヒト抗体を含む、モノクローナル抗体である。一態様において、本発明の抗原結合分子は、例えば、Fv、Fab、Fab'、scFv、ダイアボディ、シングルドメイン抗体、またはF(ab')2断片などの、抗体断片である。別の態様において、抗体は、例えば、完全IgG1抗体、完全IgG2抗体、完全IgG3抗体、または完全IgG4抗体などの、全長抗体である。 In some embodiments, the antigen-binding molecule of the present invention is an antibody. In some embodiments, the antigen-binding molecule of the present invention is a monoclonal antibody, including a chimeric, humanized, or human antibody. In one embodiment, the antigen-binding molecule of the present invention is an antibody fragment, such as an Fv, Fab, Fab', scFv, diabody, single-domain antibody, or F(ab') 2 fragment. In another embodiment, the antibody is a full-length antibody, such as a complete IgG1 antibody, a complete IgG2 antibody, a complete IgG3 antibody, or a complete IgG4 antibody.

優れた安定性を有するCD3結合ドメインを含む抗原結合分子
 一局面において、本発明は、CD3に対する結合活性を有する第一の抗原結合ドメイン(CD3結合ドメイン)と、第二の抗原結合ドメインとを含む、抗原結合分子を提供する。別の局面において、本発明は、CD3に対する結合活性を有する単一の抗原結合ドメイン(CD3結合ドメイン)を含む、一価の抗原結合分子を提供する。本発明の抗原結合分子に含まれるCD3結合ドメインは、優れた安定性を有し、常温以上での保管後のCD3結合活性の低下が小さいことを特徴とする。一態様において、本発明の抗原結合分子に含まれるCD3結合ドメインは、所定の時間(例えば3日間)にわたって常温以上(例えば50℃)で保存した後のCD3結合活性が、低温(例えば4℃)で保存した後のCD3結合活性の65%以上、70%以上、または75%以上を維持する。いくつかの態様において、本発明の抗原結合分子に含まれるCD3結合ドメインは、CD3ε鎖に対する結合活性を有する。いくつかの態様において、CD3は哺乳動物のCD3である。特定の態様において、CD3はヒトCD3である。いくつかの態様において、本発明の抗原結合分子に含まれる抗原結合ドメインは、Fv、Fab、Fab'、またはscFvである。
Antigen-binding molecules comprising a CD3-binding domain with excellent stability . In one aspect, the present invention provides antigen-binding molecules comprising a first antigen-binding domain (CD3-binding domain) having binding activity to CD3 and a second antigen-binding domain. In another aspect, the present invention provides monovalent antigen-binding molecules comprising a single antigen-binding domain (CD3-binding domain) having binding activity to CD3. The CD3-binding domain contained in the antigen-binding molecules of the present invention is characterized by excellent stability and a small decrease in CD3-binding activity after storage at room temperature or higher. In one embodiment, the CD3-binding activity of the CD3-binding domain contained in the antigen-binding molecules of the present invention after storage at room temperature or higher (e.g., 50°C) for a predetermined period of time (e.g., 3 days) maintains 65% or more, 70% or more, or 75% or more of the CD3-binding activity after storage at a low temperature (e.g., 4°C). In some embodiments, the CD3-binding domain contained in the antigen-binding molecules of the present invention has binding activity to the CD3 epsilon chain. In some embodiments, the CD3 is mammalian CD3. In certain embodiments, the CD3 is human CD3. In some embodiments, the antigen-binding domain comprised in the antigen-binding molecule of the present invention is Fv, Fab, Fab', or scFv.

CD3結合ドメイン
 一態様において、本発明の抗原結合分子に含まれるCD3結合ドメインは、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域を含み、
 前記H鎖可変領域は、
  アミノ酸配列NAWMH(配列番号:1)を含むH鎖CDR1;
  アミノ酸配列QIX1DKSQNYATX2VAESVKG(配列番号:2)を含むH鎖CDR2であって、X1がKまたはRであり、X2がYまたはFである、H鎖CDR2;及び
  アミノ酸配列VHYX3AGYGVDX4(配列番号:3)を含むH鎖CDR3であって、X3がAまたはPであり、X4がI、MまたはLである、H鎖CDR3
を含み、
 前記L鎖可変領域は、
  アミノ酸配列RSX5X6X7VVHENRX8TYLH(配列番号:4)を含むL鎖CDR1であって、X5がSまたはTであり、X6がQまたはMであり、X7がSまたはTであり、X8がQまたはNである、L鎖CDR1;
  アミノ酸配列KVSNRFS(配列番号:5)を含むL鎖CDR2;及び
  アミノ酸配列GQGTQVPYT(配列番号:6)を含むL鎖CDR3
を含む。なお、本明細書において、H鎖及び重鎖は相互に交換可能に用いられ、L鎖と軽鎖は相互に交換可能に用いられる。
CD3-binding domain In one embodiment, the CD3-binding domain contained in the antigen-binding molecule of the present invention comprises an antibody H chain variable region and L chain variable region that have binding activity to CD3,
The H chain variable region
H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1);
an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3 ), wherein X3 is A or P, and X4 is I, M, or L.
Including,
The light chain variable region
an L chain CDR1 comprising the amino acid sequence RSX5X6X7VVHENRX8TYLH ( SEQ ID NO: 4 ), wherein X5 is S or T, X6 is Q or M, X7 is S or T, and X8 is Q or N;
L chain CDR2 comprising the amino acid sequence KVSNRFS (SEQ ID NO: 5); and L chain CDR3 comprising the amino acid sequence GQGTQVPYT (SEQ ID NO: 6).
In this specification, the terms H chain and heavy chain are used interchangeably, and the terms L chain and light chain are used interchangeably.

 特定の態様において、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域は、以下の(a1)~(a8)から選ばれるH鎖CDR1、CDR2及びCDR3、並びに、L鎖CDR1、CDR2及びCDR3の組合せのうちのいずれかを含む:
 (a1) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、配列番号:10に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a2) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a3) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:16に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a4) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a5) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:13に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a6) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:17に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a7) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:18に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a8) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:19に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ。
In certain embodiments, the antibody heavy chain variable region and light chain variable region having binding activity to CD3 comprise any combination of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8):
(a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a3) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 16, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a4) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a5) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 13, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a6) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 17, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a7) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 18, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a8) A combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 19, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6.

 一態様において、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域は、以下の(a1)~(a8)から選ばれるH鎖可変領域及びL鎖可変領域の組合せのうちのいずれかを含む:
 (a1)配列番号:20に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a2) 配列番号:21に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a3) 配列番号:21に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:26に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a4) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a5) 配列番号:23に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a6) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:27に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a7) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:28に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ、
 (a8) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するH鎖可変領域と、配列番号:29に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有するL鎖可変領域との組合せ。
In one embodiment, the antibody heavy chain variable region and light chain variable region having binding activity to CD3 comprise any combination of heavy chain variable region and light chain variable region selected from the following (a1) to (a8):
(a1) a combination of an H chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 20 and an L chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25;
(a2) a combination of a heavy chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 21 and a light chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25;
(a3) a combination of a heavy chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 21 and a light chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 26;
(a4) a combination of a heavy chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 22 and a light chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25;
(a5) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 23 and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25;
(a6) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 22 and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 27;
(a7) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 22 and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 28;
(a8) A combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 29.

 一態様において、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域は、以下の(a1)~(a8)から選ばれるH鎖可変領域と、L鎖可変領域との組合せのうちのいずれかを含む:
 (a1)配列番号:20に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:10に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a2) 配列番号:21に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:11に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a3) 配列番号:21に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:11に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:26に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:16に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a4) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a5) 配列番号:23に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:13に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:25に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a6) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:27に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:17に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a7) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:28に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:18に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ、
 (a8) 配列番号:22に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、H鎖可変領域と、配列番号:29に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:19に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、L鎖可変領域との組合せ。
In one embodiment, the antibody H chain variable region and L chain variable region having binding activity to CD3 comprise any combination of an H chain variable region and an L chain variable region selected from the following (a1) to (a8):
(a1) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 20, and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 25, and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a2) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 21 and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 25 and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a3) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 21 and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 26 and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 16, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a4) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 22 and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 25 and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a5) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 23 and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 13, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 25 and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a6) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 22 and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 27 and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 17, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a7) a combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 22 and comprising an H-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, and an H-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 28 and comprising an L-chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 18, an L-chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a8) A combination of an H-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 22 and comprising an H-chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H-chain CDR2 having the amino acid sequence shown in SEQ ID NO: 9, and an H-chain CDR3 having the amino acid sequence shown in SEQ ID NO: 12, and an L-chain variable region having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 29 and comprising an L-chain CDR1 having the amino acid sequence shown in SEQ ID NO: 19, an L-chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L-chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6.

 特定の態様において、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域は、以下の(a1)~(a8)から選ばれるH鎖可変領域及びL鎖可変領域の組合せのうちのいずれかを含む:
 (a1)配列番号:20に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a2) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a3) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:26に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a4) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a5) 配列番号:23に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a6) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:27に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a7) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:28に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a8) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:29に示されるアミノ酸配列を含むL鎖可変領域の組合せ。
In certain embodiments, the antibody heavy chain variable region and light chain variable region having binding activity to CD3 comprises any one of the combinations of heavy chain variable region and light chain variable region selected from the following (a1) to (a8):
(a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26;
(a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a5) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 23 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a6) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 27;
(a7) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 28;
(a8) A combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 29.

 一態様において、本発明の抗原結合分子は、CD3に対する結合活性を有する重鎖及び軽鎖として、以下の(a1)~(a8)から選ばれる重鎖及び軽鎖の組合せのうちのいずれかを含む:
 (a1)配列番号:30に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a2) 配列番号:31に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a3) 配列番号:31に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:36に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a4) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a5) 配列番号:33に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a6) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:37に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a7) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:38に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ、
 (a8) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する重鎖と、配列番号:39に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有する軽鎖との組合せ。
In one embodiment, the antigen-binding molecule of the present invention comprises, as heavy and light chains having binding activity to CD3, any of the combinations of heavy and light chains selected from the following (a1) to (a8):
(a1) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 30 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 35;
(a2) a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 31 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 35;
(a3) a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 31 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 36;
(a4) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 32 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 35;
(a5) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 33 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 35;
(a6) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 32 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 37;
(a7) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 32 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 38;
(a8) A combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 32 and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence set forth in SEQ ID NO: 39.

 一態様において、本発明の抗原結合分子は、CD3に対する結合活性を有する重鎖及び軽鎖として、以下の(a1)~(a8)から選ばれる重鎖と、軽鎖との組合せのうちのいずれかを含む:
 (a1)配列番号:30に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:10に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a2) 配列番号:31に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:11に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a3) 配列番号:31に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:11に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:36に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:16に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a4) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a5) 配列番号:33に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:13に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:35に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a6) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:37に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:17に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a7) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:38に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:18に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ、
 (a8) 配列番号:32に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:12に示されるアミノ酸配列を含むH鎖CDR3を含む、重鎖と、配列番号:39に示されるアミノ酸配列に対して少なくとも90%または少なくとも95%の配列同一性を有し、かつ配列番号:19に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む、軽鎖との組合せ。
In one embodiment, the antigen-binding molecule of the present invention comprises, as a heavy chain and a light chain having binding activity to CD3, any combination of a heavy chain and a light chain selected from the following (a1) to (a8):
(a1) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 30 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 7, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 10, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 35 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6;
(a2) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 31 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 8, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 11, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 35 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6;
(a3) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 31 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 8, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 11, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 36 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 16, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6;
(a4) A combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 32 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 9, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 12, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 35 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6,
(a5) A combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 33 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 9, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 13, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 35 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6,
(a6) a combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 32 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 9, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 12, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 37 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 17, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6;
(a7) A combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 32 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 9, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 12, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 38 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 18, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6,
(a8) A combination of a heavy chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 32 and comprising an H chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 having the amino acid sequence shown in SEQ ID NO: 9, and an H chain CDR3 having the amino acid sequence shown in SEQ ID NO: 12, and a light chain having at least 90% or at least 95% sequence identity to the amino acid sequence shown in SEQ ID NO: 39 and comprising an L chain CDR1 having the amino acid sequence shown in SEQ ID NO: 19, an L chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6.

 特定の態様において、本発明の抗原結合分子は、CD3に対する結合活性を有する重鎖及び軽鎖として、以下の(a1)~(a8)から選ばれる重鎖及び軽鎖の組合せのうちのいずれかを含む:
 (a1)配列番号:30に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a2) 配列番号:31に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a3) 配列番号:31に示されるアミノ酸配列を含む重鎖及び配列番号:36に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a4) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a5) 配列番号:33に示されるアミノ酸配列を含む重鎖及び配列番号:35に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a6) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:37に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a7) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:38に示されるアミノ酸配列を含む軽鎖の組合せ、
 (a8) 配列番号:32に示されるアミノ酸配列を含む重鎖及び配列番号:39に示されるアミノ酸配列を含む軽鎖の組合せ。
In a specific embodiment, the antigen-binding molecule of the present invention comprises, as a heavy chain and a light chain having binding activity to CD3, any of the following combinations of heavy chains and light chains selected from (a1) to (a8):
(a1) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 30 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a2) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a3) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 31 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 36;
(a4) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a5) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 33 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 35;
(a6) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 37;
(a7) a combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 38;
(a8) A combination of a heavy chain comprising the amino acid sequence shown in SEQ ID NO: 32 and a light chain comprising the amino acid sequence shown in SEQ ID NO: 39.

第二の抗原結合ドメイン
 本発明の抗原結合分子において、第二の抗原結合ドメインは、標的とする抗原に対する結合活性を有する限り、その構造は特に限定されない。いくつかの態様において、第二の抗原結合ドメインは、抗体H鎖可変領域及びL鎖可変領域を含む。
Second antigen-binding domain In the antigen-binding molecules of the present invention, the structure of the second antigen-binding domain is not particularly limited, as long as it has binding activity to a target antigen. In some embodiments, the second antigen-binding domain comprises an antibody heavy chain variable region and light chain variable region.

単一特異性抗原結合分子
 いくつかの態様において、本発明の抗原結合分子は単一特異性抗原結合分子であり、第一の抗原結合ドメインおよび第二の抗原結合ドメインは、いずれもCD3(例えば、ヒトCD3)に対する結合活性を有する。一態様において、第一の抗原結合ドメインおよび第二の抗原結合ドメインは、CD3における同じエピトープに対する結合活性を有する。特定の態様において、第一の抗原結合ドメインおよび第二の抗原結合ドメインは、いずれもCD3ε鎖(例えば、ヒトCD3ε鎖)に対する結合活性を有する。
 特定の態様において、第一の抗原結合ドメインおよび第二の抗原結合ドメインは、同じアミノ酸配列を含む。別の特定の態様において、第一の抗原結合ドメインおよび第二の抗原結合ドメインは、互いに異なるアミノ酸配列を含む。
Monospecific antigen-binding molecules In some embodiments, the antigen-binding molecules of the present invention are monospecific antigen-binding molecules, and both the first and second antigen-binding domains have binding activity to CD3 (e.g., human CD3). In one embodiment, the first and second antigen-binding domains have binding activity to the same epitope on CD3. In a specific embodiment, both the first and second antigen-binding domains have binding activity to the CD3ε chain (e.g., human CD3ε chain).
In a particular embodiment, the first antigen-binding domain and the second antigen-binding domain comprise the same amino acid sequence. In another particular embodiment, the first antigen-binding domain and the second antigen-binding domain comprise different amino acid sequences from each other.

多重特異性抗原結合分子
 いくつかの態様において、本発明の抗原結合分子は、少なくとも2つの異なる部位に結合特異性を有する、多重特異性抗原結合分子である。特定の態様において、本発明の多重特異性抗原結合分子は二重特異性抗原結合分子である。別の特定の態様において、本発明の多重特異性抗原結合分子は三重以上の特異性を有する抗原結合分子であり、例えば、三重特異性抗原結合分子、四重特異性抗原結合分子、五重特異性抗原結合分子、または六重特異性抗原結合分子である。
Multispecific antigen-binding molecules In some embodiments, the antigen-binding molecules of the present invention are multispecific antigen-binding molecules having binding specificities at at least two different sites. In certain embodiments, the multispecific antigen-binding molecules of the present invention are bispecific antigen-binding molecules. In another specific embodiment, the multispecific antigen-binding molecules of the present invention are antigen-binding molecules with three or more specificities, such as trispecific, tetraspecific, pentaspecific, or hexaspecific antigen-binding molecules.

 本発明の多重特異性抗原結合分子の一態様において、第一の抗原結合ドメインはCD3に対する結合活性を有し、第二の抗原結合ドメインはCD3以外の抗原に対する結合活性を有する。このような態様において、第二の抗原結合ドメインが認識および結合する「CD3以外の抗原」は、好ましくはT細胞の持つ細胞傷害作用(T細胞依存的細胞傷害活性)の標的とされる細胞の表面に発現する分子であり、例えば、がん細胞の表面に特異的に発現する分子(本明細書において、がん抗原またはがん特異的抗原とも称される)、および免疫応答を抑制する機能を有する細胞の表面に発現する分子(本明細書において、免疫抑制細胞表面抗原とも称される)が挙げられる。 In one embodiment of the multispecific antigen-binding molecule of the present invention, the first antigen-binding domain has binding activity to CD3, and the second antigen-binding domain has binding activity to an antigen other than CD3. In this embodiment, the "antigen other than CD3" recognized and bound by the second antigen-binding domain is preferably a molecule expressed on the surface of cells that are targeted by the cytotoxic activity of T cells (T cell-dependent cytotoxicity), and examples include molecules specifically expressed on the surface of cancer cells (also referred to herein as cancer antigens or cancer-specific antigens) and molecules expressed on the surface of cells that have the function of suppressing immune responses (also referred to herein as immunosuppressive cell surface antigens).

 一態様において、第二の抗原結合ドメインは、がん抗原に対する結合活性を有する。このような態様において、本発明の多重特異性抗原結合分子は、第一の抗原結合ドメインを介してCD3に結合し、第二の抗原結合ドメインを介してがん抗原に結合することによって、T細胞を標的がん細胞に近接せしめ、T細胞による標的がん細胞を含む組織に対する細胞傷害活性を通じて、がんを治療または予防することができる。
 別の態様において、第二の抗原結合ドメインは、免疫抑制細胞表面抗原に対する結合活性を有する。このような態様において、本発明の多重特異性抗原結合分子は、第一の抗原結合ドメインを介してCD3に結合し、第二の抗原結合ドメインを介して免疫抑制細胞表面抗原に結合することによって、T細胞を、免疫応答を抑制する機能を有する細胞(本明細書において免疫抑制細胞とも称される)に近接せしめ、T細胞による免疫応答抑制細胞に対する細胞傷害活性を通じて、がんを治療または予防することができる。
 本発明の多重特異性抗原結合分子において、第二の抗原結合ドメインとして、がん抗原または免疫抑制細胞表面抗原に対する結合活性を有する、公知の抗原結合ドメインおよび将来同定されるものを含む他の抗原結合ドメインを用いることができる。
In one embodiment, the second antigen-binding domain has binding activity to a cancer antigen. In such an embodiment, the multispecific antigen-binding molecule of the present invention binds to CD3 via the first antigen-binding domain and binds to a cancer antigen via the second antigen-binding domain, thereby bringing T cells into proximity with target cancer cells and enabling the treatment or prevention of cancer through the cytotoxic activity of T cells against tissues containing the target cancer cells.
In another embodiment, the second antigen-binding domain has binding activity to an immunosuppressive cell surface antigen. In such an embodiment, the multispecific antigen-binding molecule of the present invention binds to CD3 via the first antigen-binding domain and binds to the immunosuppressive cell surface antigen via the second antigen-binding domain, thereby bringing T cells into proximity with cells that have the function of suppressing immune responses (also referred to herein as immunosuppressive cells), and enabling the treatment or prevention of cancer through the cytotoxic activity of T cells against the immune response-suppressing cells.
In the multispecific antigen-binding molecules of the present invention, known antigen-binding domains and other antigen-binding domains, including those to be identified in the future, that have binding activity for cancer antigens or immunosuppressive cell surface antigens can be used as the second antigen-binding domain.

 好ましい態様において、本発明の多重特異性抗原結合分子は、CD3に対する結合活性を有する第一の抗原結合ドメインとして、配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、及び配列番号:10に示されるアミノ酸配列を含むH鎖CDR3を含む抗体H鎖可変領域(例えば、配列番号:20に示されるアミノ酸配列を含むH鎖可変領域)と、配列番号:14に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3を含む抗体L鎖可変領域(例えば、配列番号:24に示されるアミノ酸配列を含むL鎖可変領域)とを含む対照多重特異性抗原結合分子の細胞傷害活性と比較して、同等又はそれ以上の細胞傷害活性を有する。そのような多重特異性抗原結合分子としては、例えば、後掲の表5に記載の抗体番号2, 14, 25, 29, 30~32, 34の重鎖および軽鎖CDR、重鎖および軽鎖可変領域、または重鎖および軽鎖を、CD3に対する結合活性を有する第一の抗原結合ドメインとして含む、多重特異性抗原結合分子(例えば二重特異性抗原結合分子、例えば二重特異性抗体)を挙げることができる。 In a preferred embodiment, the multispecific antigen-binding molecule of the present invention has a cytotoxic activity that is equivalent to or greater than that of a control multispecific antigen-binding molecule that comprises, as a first antigen-binding domain having binding activity to CD3, an antibody heavy chain variable region (e.g., an heavy chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20) comprising an heavy chain CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an heavy chain CDR2 having the amino acid sequence shown in SEQ ID NO: 7, and an heavy chain CDR3 having the amino acid sequence shown in SEQ ID NO: 10, and an antibody light chain variable region (e.g., an light chain variable region comprising the amino acid sequence shown in SEQ ID NO: 24) comprising an light chain CDR1 having the amino acid sequence shown in SEQ ID NO: 14, an light chain CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an light chain CDR3 having the amino acid sequence shown in SEQ ID NO: 6. Examples of such multispecific antigen-binding molecules include multispecific antigen-binding molecules (e.g., bispecific antigen-binding molecules, e.g., bispecific antibodies) that comprise, as a first antigen-binding domain, the heavy and light chain CDRs, heavy and light chain variable regions, or heavy and light chains of antibody Nos. 2, 14, 25, 29, 30 to 32, and 34 listed in Table 5 below.

CD3に対する結合活性を有する抗体可変領域を含むドメイン
 本明細書において「CD3に対する結合活性を有する抗体可変領域」及び「CD3に対する結合活性を有する抗体可変領域を含むドメイン」は相互に交換可能に用いられ、T細胞受容体とともにT細胞受容体複合体を構成するアダプター分子であるCD3の一部または全部に特異的に結合し且つ相補的である領域を含んで成る抗CD3抗体の部分をいう。好ましくは、当該ドメインは抗CD3抗体の軽鎖可変領域(VL)と抗CD3抗体の重鎖可変領域(VH)とを含む。こうしたドメインの例としては、「scFv(single chain Fv)」、「単鎖抗体(single chain antibody)」、「Fv」、「scFv2(single chain Fv 2)」、「Fab」または「F(ab')2」等が好適に挙げられる。
Domain containing an antibody variable region with binding activity to CD3. Herein, the terms "antibody variable region with binding activity to CD3" and "domain containing an antibody variable region with binding activity to CD3" are used interchangeably and refer to a portion of an anti-CD3 antibody comprising a region that specifically binds to and is complementary to part or all of CD3, an adaptor molecule that forms the T cell receptor complex together with the T cell receptor. Preferably, the domain contains the light chain variable region (VL) and heavy chain variable region (VH) of an anti-CD3 antibody. Suitable examples of such domains include "scFv (single chain Fv),""single chain antibody,""Fv,"" scFv2 (single chain Fv2),""Fab," and "F(ab') 2 ."

 本発明に係るCD3結合ドメイン(CD3に対する結合活性を有する抗体可変領域を含むドメイン)は、CD3を構成するγ鎖、δ鎖又はε鎖配列に存在するエピトープであればいずれのエピトープに結合するものであってもよい。本発明において、好ましくはヒトCD3複合体のε鎖の細胞外領域に存在するエピトープに結合する抗CD3抗体の軽鎖可変領域(VL)と重鎖可変領域(VH)とを含むドメインが好適に用いられる。こうしたドメインとしては、実施例に記載の抗CD3抗体の軽鎖可変領域(VL)と抗CD3抗体の重鎖可変領域(VH)が好適に用いられる。CD3結合活性を有する抗体可変領域を含むドメインの起源となる抗CD3抗体は前記のとおり適宜ヒト化された抗体やヒト抗体が適宜用いられる。CD3を構成するγ鎖、δ鎖、及びε鎖の構造は、そのポリヌクレオチド配列が、それぞれRefSeq登録番号NM_000073.2、NM_000732.4、及びNM_000733.3に、そのポリペプチド配列が、それぞれRefSeq登録番号NP_000064.1、NP_000723.1、及びNP_000724.1にて登録されている。 The CD3-binding domain of the present invention (a domain containing an antibody variable region having binding activity to CD3) may bind to any epitope present in the gamma, delta, or epsilon chain sequence that constitutes CD3. In the present invention, a domain containing the light chain variable region (VL) and heavy chain variable region (VH) of an anti-CD3 antibody that binds to an epitope present in the extracellular domain of the epsilon chain of the human CD3 complex is preferably used. The light chain variable region (VL) and heavy chain variable region (VH) of an anti-CD3 antibody described in the Examples are preferably used as such a domain. As described above, an appropriate humanized antibody or human antibody is used as the anti-CD3 antibody that is the source of the domain containing an antibody variable region having CD3-binding activity. The polynucleotide sequences of the gamma, delta, and epsilon chains that make up CD3 are registered under RefSeq accession numbers NM_000073.2, NM_000732.4, and NM_000733.3, respectively, and their polypeptide sequences are registered under RefSeq accession numbers NP_000064.1, NP_000723.1, and NP_000724.1, respectively.

CD3以外の抗原に対する結合活性を有する抗体可変領域を含むドメイン
 本明細書において「CD3以外の抗原に対する結合活性を有する抗体可変領域」及び「CD3以外の抗原に対する結合活性を有する抗体可変領域を含むドメイン」は相互に交換可能に用いられ、当該抗原(例えば、がん抗原:本明細書において、がん特異的抗原とも称される)の一部または全部に特異的に結合し且つ相補的である領域を含んで成る抗体の部分をいう。抗体可変領域を含むドメインは一または複数の抗体の可変ドメインより提供され得る。好ましくは、抗体可変領域を含むドメインは抗体軽鎖可変領域(VL)と抗体重鎖可変領域(VH)とを含む。こうした抗体可変領域を含むドメインの例としては、「scFv(single chain Fv)」、「単鎖抗体(single chain antibody)」、「Fv」、「scFv2(single chain Fv 2)」、「Fab」または「F(ab')2」等が好適に挙げられる。
Domains Comprising Antibody Variable Regions with Binding Activity to Antigens Other Than CD3: Herein, the terms "antibody variable region with binding activity to antigens other than CD3" and "domains comprising antibody variable regions with binding activity to antigens other than CD3" are used interchangeably and refer to portions of antibodies comprising a region that specifically binds to and is complementary to part or all of the antigen (e.g., a cancer antigen; also referred to herein as a cancer-specific antigen). A domain comprising an antibody variable region can be provided by one or more antibody variable domains. Preferably, a domain comprising an antibody variable region comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH). Suitable examples of such domains comprising antibody variable regions include "scFv (single chain Fv),""single chain antibody,""Fv,"" scFv2 (single chain Fv2),""Fab," and "F(ab') 2 ."

抗原
 本明細書において、「抗原」は、抗原結合ドメインが結合するエピトープを含むことのみで制限される。抗原の好適な例として、それだけに限定されないが、動物またはヒト由来のペプチド、ポリペプチド、およびタンパク質が挙げられる。標的組織に起因する疾患を治療するために用いられる抗原の好適な例として、それだけに限定されないが、標的細胞(例:がん細胞および炎症細胞)の表面に発現する分子、標的細胞を含む組織中の他の細胞表面に発現する分子、標的細胞と標的細胞を含む組織に対して免疫学的役割を持つ細胞の表面に発現する分子、ならびに標的細胞を含む組織の間質中に存在する大分子が挙げられる。
Antigen : As used herein, the term "antigen" is limited to including only the epitope to which the antigen-binding domain binds. Suitable examples of antigens include, but are not limited to, peptides, polypeptides, and proteins derived from animals or humans. Suitable examples of antigens used to treat diseases caused by target tissues include, but are not limited to, molecules expressed on the surface of target cells (e.g., cancer cells and inflammatory cells), molecules expressed on the surface of other cells in tissues containing target cells, molecules expressed on the surface of cells that play an immunological role in target cells and tissues containing target cells, and large molecules present in the interstitium of tissues containing target cells.

 一実施態様において、抗原は、任意の動物種(例えば、ヒト;または非ヒト動物、例えば、マウス、ラット、ハムスター、モルモット、ウサギ、サル、カニクイザル、アカゲザル、マントヒヒ、チンパンジー、ヤギ、ヒツジ、イヌ、ウマ、ブタ、ウシ、またはラクダ)、または任意の鳥類に由来してもよく;抗原は好ましくは、ヒト、ウサギ、サル、ラット、またはマウスに由来する。 In one embodiment, the antigen may be derived from any animal species (e.g., human; or non-human animal, e.g., mouse, rat, hamster, guinea pig, rabbit, monkey, cynomolgus monkey, rhesus monkey, hamadryas baboon, chimpanzee, goat, sheep, dog, horse, pig, cow, or camel), or any bird; preferably, the antigen is derived from human, rabbit, monkey, rat, or mouse.

 本発明の抗原結合分子が認識および結合する「抗原」(例えばCD3)は、別段示さない限り、霊長類(例えば、ヒト)およびげっ歯類(例えば、マウスおよびラット)などの哺乳動物を含む、任意の脊椎動物供給源からの任意の天然型の「抗原」(例えばCD3)のことをいう。この用語は、「全長」のプロセシングを受けていない「抗原」(例えばCD3)も、細胞中でのプロセシングの結果生じるいかなる形態の「抗原」(例えばCD3)も包含する。これらの用語はまた、自然に生じる「抗原」(例えばCD3)の変異体、例えば、スプライス変異体や対立遺伝子変異体も包含する。 Unless otherwise specified, the "antigen" (e.g., CD3) recognized and bound by the antigen-binding molecules of the present invention refers to any naturally occurring "antigen" (e.g., CD3) from any vertebrate source, including mammals such as primates (e.g., humans) and rodents (e.g., mice and rats). This term encompasses "full-length" unprocessed "antigen" (e.g., CD3) as well as any form of "antigen" (e.g., CD3) that results from processing within a cell. These terms also encompass naturally occurring variants of the "antigen" (e.g., CD3), such as splice variants and allelic variants.

 いくつかの態様において、抗原としては、17-IA、4-1BB、4Dc、6-ケト-PGF1a、8-イソ-PGF2a、8-オキソ-dG、A1 アデノシン受容体、A33、ACE、ACE-2、アクチビン、アクチビンA、アクチビンAB、アクチビンB、アクチビンC、アクチビンRIA、アクチビンRIA ALK-2、アクチビンRIB ALK-4、アクチビンRIIA、アクチビンRIIB、ADAM、ADAM10、ADAM12、ADAM15、ADAM17/TACE、ADAM8、ADAM9、ADAMTS、ADAMTS4、ADAMTS5、アドレシン、aFGF、ALCAM、ALK、ALK-1、ALK-7、アルファ-1-アンチトリプシン、アルファ-V/ベータ-1アンタゴニスト、ANG、Ang、APAF-1、APE、APJ、APP、APRIL、AR、ARC、ART、アルテミン、抗Id、ASPARTIC、心房性ナトリウム利尿因子、av/b3インテグリン、Axl、b2M、B7-1、B7-2、B7-H、B-リンパ球刺激因子(BlyS)、BACE、BACE-1、Bad、BAFF、BAFF-R、Bag-1、BAK、Bax、BCA-1、BCAM、Bcl、BCMA、BDNF、b-ECGF、bFGF、BID、Bik、BIM、BLC、BL-CAM、BLK、BMP、BMP-2 BMP-2a、BMP-3/オステオゲニン(Osteogenin)、BMP-4、BMP-2b、BMP-5、BMP-6、Vgr-1、BMP-7(OP-1)、BMP-8(BMP-8a、OP-2)、BMPR、BMPR-IA(ALK-3)、BMPR-IB(ALK-6)、BRK-2、RPK-1、BMPR-II(BRK-3)、BMP、b-NGF、BOK、ボンベシン、骨由来神経栄養因子、BPDE、BPDE-DNA、BTC、補体因子3(C3)、C3a、C4、C5、C5a、C10、CA125、CAD-8、カルシトニン、cAMP、癌胎児性抗原(CEA)、癌関連抗原、カテプシンA、カテプシンB、カテプシンC/DPPI、カテプシンD、カテプシンE、カテプシンH、カテプシンL、カテプシンO、カテプシンS、カテプシンV、カテプシンX/Z/P、CBL、CCI、CCK2、CCL、CCL1、CCL11、CCL12、CCL13、CCL14、CCL15、CCL16、CCL17、CCL18、CCL19、CCL2、CCL20、CCL21、CCL22、CCL23、CCL24、CCL25、CCL26、CCL27、CCL28、CCL3、CCL4、CCL5、CCL6、CCL7、CCL8、CCL9/10、CCR、CCR1、CCR10、CCR10、CCR2、CCR3、CCR4、CCR5、CCR6、CCR7、CCR8、CCR9、CD1、CD2、CD3、CD3E、CD4、CD5、CD6、CD7、CD8、CD10、CD11a、CD11b、CD11c、CD13、CD14、CD15、CD16、CD18、CD19、CD20、CD21、CD22、CD23、CD25、CD27L、CD28、CD29、CD30、CD30L、CD32、CD33(p67タンパク質)、CD34、CD38、CD40、CD40L、CD44、CD45、CD46、CD49a、CD52、CD54、CD55、CD56、CD61、CD64、CD66e、CD74、CD80(B7-1)、CD89、CD95、CD123、CD137、CD138、CD140a、CD146、CD147、CD148、CD152、CD164、CEACAM5、CFTR、cGMP、CINC、ボツリヌス菌毒素、ウェルシュ菌毒素、CKb8-1、クローディン6、CLC、CMV、CMV UL、CNTF、CNTN-1、COX、C-Ret、CRG-2、CT-1、CTACK、CTGF、CTLA-4、PD1、PDL1、LAG3、TIM3、galectin-9、CX3CL1、CX3CR1、CXCL、CXCL1、CXCL2、CXCL3、CXCL4、CXCL5、CXCL6、CXCL7、CXCL8、CXCL9、CXCL10、CXCL11、CXCL12、CXCL13、CXCL14、CXCL15、CXCL16、CXCR、CXCR1、CXCR2、CXCR3、CXCR4、CXCR5、CXCR6、サイトケラチン腫瘍関連抗原、DAN、DCC、DcR3、DC-SIGN、補体制御因子(Decay accelerating factor)、des(1-3)-IGF-I(脳IGF-1)、Dhh、ジゴキシン、DLL3、DNAM-1、Dnase、Dpp、DPPIV/CD26、Dtk、ECAD、EDA、EDA-A1、EDA-A2、EDAR、EGF、EGFR(ErbB-1)、EMA、EMMPRIN、ENA、エンドセリン受容体、エンケファリナーゼ、eNOS、Eot、エオタキシン1、EpCAM、エフリンB2/EphB4、EPO、ERCC、E-セレクチン、ET-1、第IIa因子、第VII因子、第VIIIc因子、第IX因子、線維芽細胞活性化タンパク質(FAP)、Fas、FcR1、FEN-1、フェリチン、FGF、FGF-19、FGF-2、FGF-3、FGF-8、FGFR、FGFR-3、フィブリン、FL、FLIP、Flt-3、Flt-4、卵胞刺激ホルモン、フラクタルカイン、FZD1、FZD2、FZD3、FZD4、FZD5、FZD6、FZD7、FZD8、FZD9、FZD10、G250、Gas6、GCP-2、GCSF、GD2、GD3、GDF、GDF-1、GDF-3(Vgr-2)、GDF-5(BMP-14、CDMP-1)、GDF-6(BMP-13、CDMP-2)、GDF-7(BMP-12、CDMP-3)、GDF-8(ミオスタチン)、GDF-9、GDF-15(MIC-1)、GDNF、GDNF、GFAP、GFRa-1、GFR-アルファ1、GFR-アルファ2、GFR-アルファ3、GITR、グルカゴン、Glut4、糖タンパク質IIb/IIIa(GPIIb/IIIa)、GM-CSF、gp130、gp72、GRO、成長ホルモン放出因子、ハプテン(NP-capまたはNIP-cap)、HB-EGF、HCC、HCMV gBエンベロープ糖タンパク質、HCMV gHエンベロープ糖タンパク質、HCMV UL、造血成長因子(HGF)、Hep B gp120、ヘパラナーゼ、Her2、Her2/neu(ErbB-2)、Her3(ErbB-3)、Her4(ErbB-4)、単純ヘルペスウイルス(HSV) gB糖タンパク質、HSV gD糖タンパク質、HGFA、高分子量黒色腫関連抗原(HMW-MAA)、HIV gp120、HIV IIIB gp 120 V3ループ、HLA、HLA-DR、HM1.24、HMFG PEM、HRG、Hrk、ヒト心臓ミオシン、ヒトサイトメガロウイルス(HCMV)、ヒト成長ホルモン(HGH)、HVEM、I-309、IAP、ICAM、ICAM-1、ICAM-3、ICE、ICOS、IFNg、Ig、IgA受容体、IgE、IGF、IGF結合タンパク質、IGF-1R、IGFBP、IGF-I、IGF-II、IL、IL-1、IL-1R、IL-2、IL-2R、IL-4、IL-4R、IL-5、IL-5R、IL-6、IL-6R、IL-8、IL-9、IL-10、IL-12、IL-13、IL-15、IL-18、IL-18R、IL-21、IL-23、IL-27、インターフェロン(INF)-アルファ、INF-ベータ、INF-ガンマ、インヒビン、iNOS、インスリンA鎖、インスリンB鎖、インスリン様増殖因子1、インテグリンアルファ2、インテグリンアルファ3、インテグリンアルファ4、インテグリンアルファ4/ベータ1、インテグリンアルファ4/ベータ7、インテグリンアルファ5(アルファV)、インテグリンアルファ5/ベータ1、インテグリンアルファ5/ベータ3、インテグリンアルファ6、インテグリンベータ1、インテグリンベータ2、インターフェロンガンマ、IP-10、I-TAC、JE、カリクレイン2、カリクレイン5、カリクレイン6、カリクレイン11、カリクレイン12、カリクレイン14、カリクレイン15、カリクレインL1、カリクレインL2、カリクレインL3、カリクレインL4、KC、KDR、ケラチノサイト増殖因子(KGF)、ラミニン5、LAMP、LAP、LAP(TGF-1)、潜在的TGF-1、潜在的TGF-1 bp1、LBP、LDGF、LECT2、レフティ、ルイス-Y抗原、ルイス-Y関連抗原、LFA-1、LFA-3、Lfo、LIF、LIGHT、リポタンパク質、LIX、LKN、Lptn、L-セレクチン、LT-a、LT-b、LTB4、LTBP-1、肺表面、黄体形成ホルモン、リンホトキシンベータ受容体、Mac-1、MAdCAM、MAG、MAP2、MARC、MCAM、MCAM、MCK-2、MCP、M-CSF、MDC、Mer、メタロプロテイナーゼ、MGDF受容体、MGMT、MHC(HLA-DR)、MIF、MIG、MIP、MIP-1-アルファ、MK、MMAC1、MMP、MMP-1、MMP-10、MMP-11、MMP-12、MMP-13、MMP-14、MMP-15、MMP-2、MMP-24、MMP-3、MMP-7、MMP-8、MMP-9、MPIF、Mpo、MSK、MSP、ムチン(Muc1)、MUC18、ミュラー管抑制物質、Mug、MuSK、NAIP、NAP、NCAD、N-Cアドヘリン、NCA 90、NCAM、NCAM、ネプリライシン、ニューロトロフィン-3、-4、または-6、ニュールツリン、神経成長因子(NGF)、NGFR、NGF-ベータ、nNOS、NO、NOS、Npn、NRG-3、NT、NTN、OB、OGG1、OPG、OPN、OSM、OX40L、OX40R、p150、p95、PADPr、副甲状腺ホルモン、PARC、PARP、PBR、PBSF、PCAD、P-カドヘリン、PCNA、PDGF、PDGF、PDK-1、PECAM、PEM、PF4、PGE、PGF、PGI2、PGJ2、PIN、PLA2、胎盤性アルカリホスファターゼ(PLAP)、PlGF、PLP、PP14、プロインスリン、プロレラキシン、プロテインC、PS、PSA、PSCA、前立腺特異的膜抗原(PSMA)、PTEN、PTHrp、Ptk、PTN、R51、RANK、RANKL、RANTES、RANTES、レラキシンA鎖、レラキシンB鎖、レニン、呼吸器多核体ウイルス(RSV)F、RSV Fgp、Ret、リウマトイド因子、RLIP76、RPA2、RSK、S100、SCF/KL、SDF-1、SERINE、血清アルブミン、sFRP-3、Shh、SIGIRR、SK-1、SLAM、SLPI、SMAC、SMDF、SMOH、SOD、SPARC、Stat、STEAP、STEAP-II、TACE、TACI、TAG-72(腫瘍関連糖タンパク質-72)、TARC、TCA-3、T細胞受容体(例えば、T細胞受容体アルファ/ベータ)、TdT、TECK、TEM1、TEM5、TEM7、TEM8、TERT、睾丸PLAP様アルカリホスファターゼ、TfR、TGF、TGF-アルファ、TGF-ベータ、TGF-ベータ Pan Specific、TGF-ベータRI(ALK-5)、TGF-ベータRII、TGF-ベータRIIb、TGF-ベータRIII、TGF-ベータ1、TGF-ベータ2、TGF-ベータ3、TGF-ベータ4、TGF-ベータ5、トロンビン、胸腺Ck-1、甲状腺刺激ホルモン、Tie、TIMP、TIQ、組織因子、TMEFF2、Tmpo、TMPRSS2、TNF、TNF-アルファ、TNF-アルファベータ、TNF-ベータ2、TNFc、TNF-RI、TNF-RII、TNFRSF10A(TRAIL R1 Apo-2、DR4)、TNFRSF10B(TRAIL R2 DR5、KILLER、TRICK-2A、TRICK-B)、TNFRSF10C(TRAIL R3 DcR1、LIT、TRID)、TNFRSF10D(TRAIL R4 DcR2、TRUNDD)、TNFRSF11A(RANK ODF R、TRANCE R)、TNFRSF11B(OPG OCIF、TR1)、TNFRSF12(TWEAK R FN14)、TNFRSF13B(TACI)、TNFRSF13C(BAFF R)、TNFRSF14(HVEM ATAR、HveA、LIGHT R、TR2)、TNFRSF16(NGFR p75NTR)、TNFRSF17(BCMA)、TNFRSF18(GITR AITR)、TNFRSF19(TROY TAJ、TRADE)、TNFRSF19L(RELT)、TNFRSF1A(TNF RI CD120a、p55-60)、TNFRSF1B(TNF RII CD120b、p75-80)、TNFRSF26(TNFRH3)、TNFRSF3(LTbR TNF RIII、TNFC R)、TNFRSF4(OX40 ACT35、TXGP1 R)、TNFRSF5(CD40 p50)、TNFRSF6(Fas Apo-1、APT1、CD95)、TNFRSF6B(DcR3 M68、TR6)、TNFRSF7(CD27)、TNFRSF8(CD30)、TNFRSF9(4-1BB CD137、ILA)、TNFRSF21(DR6)、TNFRSF22(DcTRAIL R2 TNFRH2)、TNFRST23(DcTRAIL R1 TNFRH1)、TNFRSF25(DR3 Apo-3、LARD、TR-3、TRAMP、WSL-1)、TNFSF10(TRAIL Apo-2リガンド、TL2)、TNFSF11(TRANCE/RANKリガンド ODF、OPGリガンド)、TNFSF12(TWEAK Apo-3リガンド、DR3リガンド)、TNFSF13(APRIL TALL2)、TNFSF13B(BAFF BLYS、TALL1、THANK、TNFSF20)、TNFSF14(LIGHT HVEMリガンド、LTg)、TNFSF15(TL1A/VEGI)、TNFSF18(GITRリガンド AITRリガンド、TL6)、TNFSF1A(TNF-a コネクチン(Conectin)、DIF、TNFSF2)、TNFSF1B(TNF-b LTa、TNFSF1)、TNFSF3(LTb TNFC、p33)、TNFSF4(OX40リガンド gp34、TXGP1)、TNFSF5(CD40リガンド CD154、gp39、HIGM1、IMD3、TRAP)、TNFSF6(Fasリガンド Apo-1リガンド、APT1リガンド)、TNFSF7(CD27リガンド CD70)、TNFSF8(CD30リガンド CD153)、TNFSF9(4-1BBリガンド CD137リガンド)、TP-1、t-PA、Tpo、TRAIL、TRAIL R、TRAIL-R1、TRAIL-R2、TRANCE、トランスフェリン受容体、TRF、Trk、TROP-2、TLR(Toll-like receptor)1、TLR2、TLR3、TLR4、TLR5、TLR6、TLR7、TLR8、TLR9、TLR10、TSG、TSLP、腫瘍関連抗原CA125、腫瘍関連抗原発現ルイスY関連炭水化物、TWEAK、TXB2、Ung、uPAR、uPAR-1、ウロキナーゼ、VCAM、VCAM-1、VECAD、VE-カドヘリン、VE-カドヘリン-2、VEFGR-1(flt-1)、VEGF、VEGFR、VEGFR-3(flt-4)、VEGI、VIM、ウイルス抗原、VLA、VLA-1、VLA-4、VNRインテグリン、フォン・ヴィレブランド因子、WIF-1、WNT1、WNT2、WNT2B/13、WNT3、WNT3A、WNT4、WNT5A、WNT5B、WNT6、WNT7A、WNT7B、WNT8A、WNT8B、WNT9A、WNT9A、WNT9B、WNT10A、WNT10B、WNT11、WNT16、XCL1、XCL2、XCR1、XCR1、XEDAR、XIAP、XPD、HMGB1、IgA、Aβ、CD81、CD97、CD98、DDR1、DKK1、EREG、Hsp90、IL-17/IL-17R、IL-20/IL-20R、酸化LDL、PCSK9、プレカリクレイン、RON、TMEM16F、SOD1、クロモグラニンA、クロモグラニンB、tau、VAP1、高分子キニノーゲン、IL-31、IL-31R、Nav1.1、Nav1.2、Nav1.3、Nav1.4、Nav1.5、Nav1.6、Nav1.7、Nav1.8、Nav1.9、EPCR、C1、C1q、C1r、C1s、C2、C2a、C2b、C3、C3a、C3b、C4、C4a、C4b、C5、C5a、C5b、C6、C7、C8、C9、B因子、D因子、H因子、プロパージン、スクレロスチン、フィブリノーゲン、フィブリン、プロトロンビン、トロンビン、組織因子、第V因子、第Va因子、VII因子、第VIIa因子、第VIII因子、第VIIIa因子、第IX因子、第IXa因子、第X因子、第Xa因子、第XI因子、第XIa因子、第XII因子、第XIIa因子、第XIII因子、第XIIIa因子、TFPI、アンチトロンビンIII、EPCR、トロンボモデュリン、TAPI、tPA、プラスミノーゲン、プラスミン、PAI-1、PAI-2、GPC3、シンデカン-1、シンデカン-2、シンデカン-3、シンデカン-4、LPA、S1Pなどが挙げられる。いくつかの態様において、抗原としては、ホルモンおよび成長因子のための受容体などが挙げられる。特定の態様において、抗原は、腫瘍組織に含まれる細胞(例えば腫瘍細胞、免疫細胞、間質細胞など)が発現または分泌する抗原分子、特に、がん細胞が特異的に発現する抗原(がん特異的抗原)で
ある。特定の態様において、抗原は、免疫応答を抑制する機能を有する細胞の表面に発現する抗原分子である。
In some embodiments, antigens include 17-IA, 4-1BB, 4Dc, 6-keto-PGF1a, 8-iso-PGF2a, 8-oxo-dG, A1 adenosine receptor, A33, ACE, ACE-2, activin, activin A, activin AB, activin B, activin C, activin RIA, activin RIA ALK-2, activin RIB ALK-4, activin RIIA, activin RIIB, ADAM, ADAM10, ADAM12, ADAM15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, addressin, aFGF, ALCAM, ALK, ALK-1, ALK-7, alpha-1-antitrypsin, alpha-V/beta-1 antagonist, ANG, Ang, APAF-1, APE, APJ, APP, APRIL, AR, A RC, ART, Artemin, Anti-Id, ASPARTIC, Atrial natriuretic factor, av/b3 integrin, Axl, b2M, B7-1, B7-2, B7-H, B-lymphocyte stimulatory factor (BlyS), BACE, BACE-1, Bad, BAFF, BAFF-R, Bag-1, BAK, Bax, BCA-1, BCAM, Bcl, BCMA, BDNF, b-ECGF, bFGF, BID, Bik, BIM, BLC, BL-CAM, BLK, BMP, BMP-2 BMP-2a, BMP-3/osteogenin, BMP-4, BMP-2b, BMP-5, BMP-6, Vgr-1, BMP-7 (OP-1), BMP-8 (BMP-8a, OP-2), BMPR, BMPR-IA (ALK-3), BMPR-IB (ALK-6), BRK-2, RPK-1, BMPR-II (BRK-3), BMP, b-NGF, BOK, bombesin, bone-derived neurotrophic factor, BPDE, BPDE-DNA, BTC, complement factor 3 (C3), C3a, C4, C5, C5a, C10, CA125, CAD-8, Calcitonin, cAMP, carcinoembryonic antigen (CEA), cancer-associated antigen, cathepsin A, cathepsin B, cathepsin C/DPPI, cathepsin D, cathepsin E, cathepsin H, cathepsin L, cathepsin O, cathepsin S, cathepsin V, cathepsin X/Z/P, CBL, CCI, CCK2, CCL, CCL1, CCL11, CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL2, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, C CL28, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9/10, CCR, CCR1, CCR10, CCR10, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CD1, CD2, CD3, CD3E, CD 4, CD5, CD6, CD7, CD8, CD10, CD11a, CD11b, CD11c, CD13, CD14, CD15, CD16, CD18, CD19, CD20, CD21, CD22, CD23, CD25, CD27L, CD28, CD29, CD30, CD30L, CD32, CD33 (p67 protein), CD34, CD38, CD40, CD40L, CD44, CD45, CD46, CD49a, CD52, CD54, CD55, CD56, CD61, CD64, CD66e, CD74, CD80 (B7-1), CD89, CD95, CD123, CD137, CD138, CD140a, CD146, CD147, CD148, CD152, CD164, CEACAM5, CFTR, cGMP, CINC, botulinum toxin, Clostridium perfringens toxin, CKb8-1, claudin 6, CLC, CMV, CMV UL, CNTF, CNTN-1, COX, C-Ret, CRG-2, CT-1, CTACK, CTGF, CTLA-4, PD1, PDL1, LAG3, TIM3, galectin-9, CX3CL1, CX3CR1, CXCL, CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CX CL8, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CXCL15, CXCL16, CXCR, CXCR1, CXC R2, CXCR3, CXCR4, CXCR5, CXCR6, cytokeratin tumor-associated antigen, DAN, DCC, DcR3, DC-SIGN, complement regulatory factor (Decay accelerating factor), des(1-3)-IGF-I (brain IGF-1), Dhh, digoxin, DLL3, DNAM-1, Dnase, Dpp, DPPIV/CD26, Dtk, ECAD, EDA, EDA-A1, EDA-A2, EDAR, EGF, EGFR (ErbB-1), EMA, EMMPRIN, ENA, endothelin receptor, enkephalinase, eNOS, Eot, eotaxin 1, EpCAM, Ef Phospholipase B2/EphB4, EPO, ERCC, E-selectin, ET-1, Factor IIa, Factor VII, Factor VIIIc, Factor IX, fibroblast activation protein (FAP), Fas, FcR1, FEN-1, ferritin, FGF, FGF-19, FGF-2, FGF-3, FGF-8, FGFR, FGFR-3, fibrin, FL, FLIP, Flt-3, Flt-4, follicle-stimulating hormone, fractal chi FZD1, FZD2, FZD3, FZD4, FZD5, FZD6, FZD7, FZD8, FZD9, FZD10, G250, Gas6, GCP-2, GCSF, GD2, GD3, GDF, GDF-1, GDF-3 (Vgr-2), GDF-5 (BMP-14, CDMP-1), GDF-6 (BMP-13, CDMP-2), GDF-7 (BMP-12, CDMP-3), GDF-8 (myostatin), GD F-9, GDF-15 (MIC-1), GDNF, GDNF, GFAP, GFRa-1, GFR-alpha1, GFR-alpha2, GFR-alpha3, GITR, glucagon, Glut4, glycoprotein IIb/IIIa (GPIIb/IIIa), GM-CSF, gp130, gp72, GRO, growth hormone-releasing factor, hapten (NP-cap or NIP-cap), HB-EGF, HCC, HCMV gB envelope glycoprotein, HCMV gH envelope glycoprotein, HCMV UL, hematopoietic growth factor (HGF), Hep B gp120, heparanase, Her2, Her2/neu (ErbB-2), Her3 (ErbB-3), Her4 (ErbB-4), herpes simplex virus (HSV) gB glycoprotein, HSV gD glycoprotein, HGFA, high-molecular-weight melanoma-associated antigen (HMW-MAA), HIV gp120, HIV IIIB gp 120 V3 loop, HLA, HLA-DR, HM1.24, HMFG PEM, HRG, Hrk, human cardiac myosin, human cytomegalovirus (HCMV), human growth hormone (HGH), HVEM, I-309, IAP, ICAM, ICAM-1, ICAM-3, ICE, ICOS, IFNg, Ig, IgA receptor, IgE, IGF, IGF-binding protein, IGF-1R, IGFBP, IGF-I, IGF-II, IL, IL-1, IL-1R, IL-2 , IL-2R, IL-4, IL-4R, IL-5, IL-5R, IL-6, IL-6R, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-18, IL-18R, IL-21, IL-23, IL-27, interferon (INF)-alpha, INF-beta, INF-gamma, inhibin, iNOS, insulin A chain, insulin B chain, insulin-like growth factor receptor 1 (IGFR), insulin-like growth factor receptor 2 (IGFR), insulin-like growth factor receptor 3 (IGFR), insulin-like growth factor receptor 4 (IGFR), insulin-like growth factor receptor 5 (IGFR), insulin-like growth factor receptor 6 (IGFR), insulin-like growth factor receptor 7 (IGFR), insulin-like growth factor receptor 8 (IGFR), insulin-like growth factor receptor 9 (IGFR), insulin-like growth factor receptor 1 ... factor 1, integrin alpha 2, integrin alpha 3, integrin alpha 4, integrin alpha 4/beta 1, integrin alpha 4/beta 7, integrin alpha 5 (alpha V), integrin alpha 5/beta 1, integrin alpha 5/beta 3, integrin alpha 6, integrin beta 1, integrin beta 2, interferon gamma, IP-10, I-TAC, JE, kallikrein 2, kallikrein 5, kallikrein 6, kallikrein 11, kallikrein 12, kallikrein 14, kallikrein 15, kallikrein L1, kallikrein L2, kallikrein L3, kallikrein L4, KC, KDR, keratinocyte growth factor (KGF), laminin 5, LAMP, LAP, LAP (TGF-1), latent TGF-1, latent TGF-1 bp1, LBP, LDGF, LECT2, Lefty, Lewis-Y antigen, Lewis-Y related antigen, LFA-1, LFA-3, Lfo, LIF, LIGHT, lipoprotein, LIX, LKN, Lptn, L-selectin, LT-a, LT-b, LTB4, LTBP-1, lung surface, luteinizing hormone, lymphotoxin beta receptor, Mac-1, MAdCAM, MAG, MAP2, MARC, MCAM, MCAM, MCK-2, MCP, M-CSF, MDC, Mer, metalloproteinase, MG DF receptor, MGMT, MHC (HLA-DR), MIF, MIG, MIP, MIP-1-alpha, MK, MMAC1, MMP, MMP-1, MMP-10, MMP-11, MMP-12, MMP-13, MMP-14, MMP-15, MMP-2, MMP-24, MMP-3, MMP-7, MMP-8, MMP-9, MPIF, Mpo, MSK, MSP, mucin (Muc1), MUC18, Müllerian inhibitory substance, Mug, MuSK, NAIP, NAP, NCAD, NCadherin, NCA 90, NCAM, NCAM, neprilysin, neurotrophin-3, -4, or -6, neurturin, nerve growth factor (NGF), NGFR, NGF-beta, nNOS, NO, NOS, Npn, NRG-3, NT, NTN, OB, OGG1, OPG, OPN, OSM, OX40L, OX40R, p150, p95, PADPr, parathyroid hormone, PARC, PARP, PBR, PBSF, PCAD, P-cadherin, PCNA, PDGF, PDK-1, P ECAM, PEM, PF4, PGE, PGF, PGI2, PGJ2, PIN, PLA2, placental alkaline phosphatase (PLAP), PlGF, PLP, PP14, proinsulin, prorelaxin, protein C, PS, PSA, PSCA, prostate-specific membrane antigen (PSMA), PTEN, PTHrp, Ptk, PTN, R51, RANK, RANKL, RANTES, relaxin A chain, relaxin B chain, renin, respiratory syncytial virus (RSV) F, RSV Fgp, Ret, rheumatoid factor, RLIP76, RPA2, RSK, S100, SCF/KL, SDF-1, SERINE, serum albumin, sFRP-3, Shh, SIGIRR, SK-1, SLAM, SLPI, SMAC, SMDF, SMOH, SOD, SPARC, Stat, STEAP, STEAP-II, TACE, TACI, TAG-72 (tumor-associated glycoprotein-72), TARC, TCA-3, T cell receptor (e.g., T cell receptor alpha/beta), TdT, TECK, TEM1, TEM5, TEM7, TEM8, TERT, testicular PLAP-like alkaline phosphatase, TfR, TGF, TGF-alpha, TGF-beta, TGF-beta Pan Specific, TGF-beta RI (ALK-5), TGF-beta RII, TGF-beta RIIb, TGF-beta RIII, TGF-beta 1, TGF-beta 2, TGF-beta 3, TGF-beta 4, TGF-beta 5, Thrombin, Thymic Ck-1, Thyroid-stimulating hormone, Tie, TIMP, TIQ, Tissue factor, TMEFF2, Tmpo, TMPRSS2, TNF, TNF-alpha, TNF-alphabeta, TNF-beta 2, TNFc, TNF-RI, TNF-RII, TNFRSF10A (TRAIL R1 Apo-2, DR4), TNFRSF10B (TRAIL R2 DR5, KILLER, TRICK-2A, TRICK-B), TNFRSF10C (TRAIL R3 DcR1, LIT, TRID), TNFRSF10D (TRAIL R4 DcR2, TRUNDD), TNFRSF11A (RANK ODF R, TRANCE R), TNFRSF11B (OPG OCIF, TR1), TNFRSF12 (TWEAK R FN14), TNFRSF13B (TACI), TNFRSF13C (BAFF R), TNFRSF14 (HVEM ATAR, HveA, LIGHT R, TR2), TNFRSF16 (NGFR p75NTR), TNFRSF17 (BCMA), TNFRSF18 (GITR AITR), TNFRSF19 (TROY TAJ, TRADE), TNFRSF19L (RELT), TNFRSF1A (TNF RI CD120a, p55-60), TNFRSF1B (TNF RII CD120b, p75-80), TNFRSF26 (TNFRH3), TNFRSF3 (LTbR TNF RIII, TNFC R), TNFRSF4 (OX40 ACT35, TXGP1 R), TNFRSF5 (CD40 p50), TNFRSF6 (Fas Apo-1, APT1, CD95), TNFRSF6B (DcR3 M68, TR6), TNFRSF7 (CD27), TNFRSF8 (CD30), TNFRSF9 (4-1BB CD137, ILA), TNFRSF21 (DR6), TNFRSF22 (DcTRAIL R2 TNFRH2), TNFRST23 (DcTRAIL R1 TNFRH1), TNFRSF25 (DR3 Apo-3, LARD, TR-3, TRAMP, WSL-1), TNFSF10 (TRAIL Apo-2 ligand, TL2), TNFSF11 (TRANCE/RANK ligand ODF, OPG ligand), TNFSF12 (TWEAK Apo-3 ligand, DR3 ligand), TNFSF13 (APRIL TALL2), TNFSF13B (BAFF BLYS, TALL1, THANK, TNFSF20), TNFSF14 (LIGHT HVEM ligand, LTg), TNFSF15 (TL1A/VEGI), TNFSF18 (GITR ligand, AITR ligand, TL6), TNFSF1A (TNF-α connectin, DIF, TNFSF2), TNFSF1B (TNF-β LTa, TNFSF1), TNFSF3 (LTb TNFC, p33), TNFSF4 (OX40 ligand gp34, TXGP1), TNFSF5 (CD40 ligand CD154, gp39, HIGM1, IMD3, TRAP), TNFSF6 (Fas ligand Apo-1 ligand, APT1 ligand), TNFSF7 (CD27 ligand CD70), TNFSF8 (CD30 ligand CD153), TNFSF9 (4-1BB ligand CD137 ligand), TP-1, t-PA, Tpo, TRAIL, TRAIL R, TRAIL-R1, TRAIL-R2, TRANCE, transferrin receptor, TRF, Trk, TROP-2, TLR (Toll-like receptor) 1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TSG, TSLP, tumor-associated antigen CA125, tumor-associated antigen expression Lewis Y-related carbohydrate, TWEAK, TXB2, Ung, uPAR, uPAR-1, urokinase, VCAM, VCAM-1, VECAD, VE-cadherin, VE-cadherin-2, VEFGR-1 (flt-1), VEGF, VEGFR, VEGFR-3 (flt-4), VEGI, VIM, viral antigen, VLA, VLA-1, VLA-4, VNR integrin, von・Willebrand factor, WIF-1, WNT1, WNT2, WNT2B/13, WNT3, WNT3A, WNT4, WNT5A, WNT5B, WNT6, WNT7A, WNT7B, WNT8A, WNT8B, WNT9A, WNT9A, WNT9B, WNT10A, WNT10B, WNT 11, WNT16, XCL1, XCL2, K9, prekallikrein, RON, TMEM16F, SOD1, chromogranin A, chromogranin B, tau, VAP1, high molecular weight kininogen, IL-31, IL-31R, Nav1.1, Nav1.2, Nav1.3, Nav1.4, Nav1.5, Nav1.6, Nav1.7, Nav1.8, Nav1.9, EPCR, C1, C1q, C1r, C1s, C2, C2a, C2b, C3, C3a, C3b, C4, C4a, C4b, C5, C5a, C5b, C6, C7, C8, C9, factor B, factor D, factor H, properdin, sclerostin, fibrinogen, Examples of antigens include fibrin, prothrombin, thrombin, tissue factor, factor V, factor Va, factor VII, factor VIIa, factor VIII, factor VIIIa, factor IX, factor IXa, factor X, factor Xa, factor XI, factor XIa, factor XII, factor XIIa, factor XIII, factor XIIIa, TFPI, antithrombin III, EPCR, thrombomodulin, TAPI, tPA, plasminogen, plasmin, PAI-1, PAI-2, GPC3, syndecan-1, syndecan-2, syndecan-3, syndecan-4, LPA, and S1P. In some embodiments, the antigen includes receptors for hormones and growth factors. In certain embodiments, the antigen is an antigen molecule expressed or secreted by cells contained in tumor tissue (e.g., tumor cells, immune cells, stromal cells, etc.), particularly an antigen specifically expressed by cancer cells (cancer-specific antigens). In certain embodiments, the antigen is an antigenic molecule expressed on the surface of a cell that functions to suppress an immune response.

がん特異的抗原
 標的細胞上の抗原の非限定的な例としての「がん特異的抗原」は、がん細胞と健常細胞を区別することを可能とする、がん細胞が発現する抗原を意味し、例えば、細胞の悪性化に伴って発現する抗原、細胞が、がん化した際に細胞表面やタンパク質分子上に現れる異常な糖鎖が含まれる。具体的には、ALK、プレイオトロフィン(PTN)、EpCAM、CA125、前立腺酸性フォスファターゼ(PAP)、前立腺特異的抗原(PSA)、TYRP1、HMW-MAA、前立腺特異的膜抗原(PSMA)、CEA、MUC1、HMFG1、TAG-72、GICA(CA19-9)、NY-ESO-1、LEA、CD15、CD17、CD19、CD20、CD22、CD30、CD33、CD38、CD77、CD79b、CD147、CD228、GD2、GD3、GM2、GM3、TSTA、ウイルスにより誘導される腫瘍抗原(例えばDNA腫瘍ウイルスおよびRNA腫瘍ウイルスのエンベロープ抗原)、α-フェトプロテイン(AFP)、5T4、分化抗原(例えば、L6およびL20抗原)、CD165、EGFR、ANKRD17、ErbB2、APO-1、SSEA-1、SCP-1、LeY、オリゴ糖抗原、SSEA-3、SSEA-4、CTAGE1、MART-1、シアリルTn(STn)、NY-CO-45、NY-LU-12、ART1、MA2、NOVA2、TSPAN8、MAGE-C1、MAGE-B1、MAGE-B2、MAGE-4A、 MAGE-X2、YKL-40、EREG、CA15-3、CLEC12A、Nectin4、Trop2、BCMA、Tissue factor、FRα (FOLR1)、ErbB3、Claudin18 (Claudin18.2)、B7-H3 (CD276)、MET、PSCA、PTK7、MSLN (Mesothelin)、CCR4、CDH6、IL13RA2、GPC3、XPR1、NOX1、MARVELD3 isoform 1、MARVELD3 isoform 2、SPINT2、MANSC1、SLC12A2、CDCP1、SEZ6L2、FLVCR1、SLC7A5、STEAP1、MMP14、TNFRSF21、TMPRSS4、クローディン6、DLL3ならびにこれらのポリペプチドの任意の断片、ならびにその改変された構造が挙げられる。
" Cancer-specific antigens " as non-limiting examples of antigens on target cells refer to antigens expressed by cancer cells that enable the distinction between cancer cells and healthy cells. Examples include antigens expressed as cells become malignant, and abnormal sugar chains that appear on the cell surface or protein molecules when cells become cancerous. Specific examples include ALK, pleiotrophin (PTN), EpCAM, CA125, prostatic acid phosphatase (PAP), prostate-specific antigen (PSA), TYRP1, HMW-MAA, prostate-specific membrane antigen (PSMA), CEA, MUC1, HMFG1, TAG-72, GICA (CA19-9), NY-ESO-1, LEA, CD15, CD17, CD19, CD20, CD22, CD30, CD33, CD38, CD77, CD79b, CD147, CD228, GD2, GD3, GM2, GM3, TSTA, and virus-induced tumors. Antigens (e.g., envelope antigens of DNA tumor viruses and RNA tumor viruses), alpha-fetoprotein (AFP), 5T4, differentiation antigens (e.g., L6 and L20 antigens), CD165, EGFR, ANKRD17, ErbB2, APO-1, SSEA-1, SCP-1, LeY, oligosaccharide antigens, SSEA-3, SSEA-4, CTAGE1, MART-1, sialyl Tn (STn), NY-CO-45, NY-LU-12, ART1, MA2, NOVA2, TSPAN8, MAGE-C1, MAGE-B1, MAGE-B2, MAGE-4A, Examples of polypeptides include MAGE-X2, YKL-40, EREG, CA15-3, CLEC12A, Nectin4, Trop2, BCMA, Tissue factor, FRα (FOLR1), ErbB3, Claudin18 (Claudin18.2), B7-H3 (CD276), MET, PSCA, PTK7, MSLN (Mesothelin), CCR4, CDH6, IL13RA2, GPC3, XPR1, NOX1, MARVELD3 isoform 1, MARVELD3 isoform 2, SPINT2, MANSC1, SLC12A2, CDCP1, SEZ6L2, FLVCR1, SLC7A5, STEAP1, MMP14, TNFRSF21, TMPRSS4, claudin 6, and DLL3, as well as any fragments of these polypeptides and modified structures thereof.

免疫応答を抑制する機能を有する細胞の表面に発現する抗原
 「免疫応答を抑制する機能を有する細胞」は、免疫応答を抑制する機能を有していれば特に限定されず、例えば、制御性T細胞(regulatory T cell (Treg))、疲弊T細胞(exhausetd T cell)、MDSC(myeloma derived stromal cell)、TAM (tumor associated macrophage)、Tr1 (induced regulatory T cell)、TADC(tumor-associated dendritic cell)、TAN(tumor-associated neutrophil)、CAF (cancer-associated fibroblast)、Breg (regulatory B cell)等が挙げられ、特に制御性T細胞及び疲弊T細胞が本発明の対象細胞として好ましい。これら免疫応答を抑制する機能を有する細胞の表面に発現する分子としては、具体的には、例えば、CTLA4、PD1、TIM3、LAG-3、CD244 (2B4)、CD160、GARP、OX40、CD137 (4-1BB)、CD25、VISTA、VISATA、BTLA、TNFR25、CD57、KLRG1、CCR2、CCR5、CCR6、CD39、CD73、CD4、CD18、CD49b、CD1d、CD5、CD21、TIM1、CD19、CD20、CD23、CD24、CD38、CD93、IgM、B220(CD45R)、CD317、PD-L1、CD11b、Ly6G、ICAM-1、FAP、PDGFR、Podoplanin、TIGIT等が挙げられる。これらの分子のうち、本発明の結合ドメインの対象となる好ましい分子としては、例えば、免疫応答抑制機能が高いと報告されている細胞画分(CD4+, CD25high, CD45RA-)に特異的に発現している細胞表面分子であるCTLA4, TIM3, LAG3, CD137(4-1BB), CD25, CCR5, CCR6, CD38, 及びTIGITが挙げられる。本発明の結合ドメインの対象となる好ましい分子としては、特に、CTLA4、LAG3、OX40があげられる。
Antigens expressed on the surface of cells that have the function of suppressing an immune response "cells that have the function of suppressing an immune response" are not particularly limited as long as they have the function of suppressing an immune response, and examples include regulatory T cells (Treg), exhausted T cells, myeloma-derived stromal cells (MDSC), tumor-associated macrophages (TAM), induced regulatory T cells (Tr1), tumor-associated dendritic cells (TADC), tumor-associated neutrophils (TAN), cancer-associated fibroblasts (CAF), and regulatory B cells (Breg). Regulatory T cells and exhausted T cells are particularly preferred as target cells of the present invention. Specific examples of molecules expressed on the surface of cells that have the function of suppressing these immune responses include CTLA4, PD1, TIM3, LAG-3, CD244 (2B4), CD160, GARP, OX40, CD137 (4-1BB), CD25, VISTA, VISATA, BTLA, TNFR25, CD57, KLRG1, CCR2, CCR5, CCR6, CD39, CD73, CD4, CD18, CD49b, CD1d, CD5, CD21, TIM1, CD19, CD20, CD23, CD24, CD38, CD93, IgM, B220 (CD45R), CD317, PD-L1, CD11b, Ly6G, ICAM-1, FAP, PDGFR, podoplanin, and TIGIT. Among these molecules, preferred target molecules for the binding domains of the present invention include, for example, CTLA4, TIM3, LAG3, CD137 (4-1BB), CD25, CCR5, CCR6, CD38, and TIGIT, which are cell surface molecules specifically expressed on cell fractions (CD4 + , CD25 high , CD45RA - ) that have been reported to have high immune response suppression function. Preferred target molecules for the binding domains of the present invention include, in particular, CTLA4, LAG3, and OX40.

抗原に対する結合活性
 下記にCD3に対して結合活性を有する抗体可変領域を含むドメインを有する被験抗原結合分子によるエピトープへの結合の確認方法が例示されるが、他の抗原に対して結合活性を有する抗体可変領域を含むドメインを有する被験抗原結合分子によるエピトープへの結合の確認方法も下記の例示に準じて適宜実施され得る。
Antigen-binding activity: Methods for confirming epitope binding by test antigen-binding molecules having domains containing antibody variable regions with binding activity to CD3 are exemplified below. Methods for confirming epitope binding by test antigen-binding molecules having domains containing antibody variable regions with binding activity to other antigens can also be appropriately performed in accordance with the examples below.

 例えば、CD3に対して結合活性を有する抗体可変領域を含むドメインを含む被験抗原結合分子が、CD3分子中に存在する線状エピトープを認識することは、たとえば次のようにして確認することができる。上記の目的のためにCD3の細胞外ドメインを構成するアミノ酸配列からなる線状のペプチドが合成される。当該ペプチドは、化学的に合成され得る。あるいは、CD3のcDNA中の、細胞外ドメインに相当するアミノ酸配列をコードする領域を利用して、遺伝子工学的手法により得られる。次に、細胞外ドメインを構成するアミノ酸配列からなる線状ペプチドと、CD3に対して結合活性を有する抗体可変領域を含むドメインを有する被験抗原結合分子との結合活性が評価される。たとえば、固定化された線状ペプチドを抗原とするELISAによって、当該ペプチドに対する当該抗原結合分子の結合活性が評価され得る。あるいは、CD3発現細胞に対する当該抗原結合分子の結合における、線状ペプチドによる阻害のレベルに基づいて、線状ペプチドに対する結合活性が明らかにされ得る。これらの試験によって、線状ペプチドに対する当該抗原結合分子の結合活性が明らかにされ得る。 For example, whether a test antigen-binding molecule containing a domain comprising an antibody variable region with binding activity to CD3 recognizes a linear epitope present in the CD3 molecule can be confirmed, for example, as follows. For this purpose, a linear peptide consisting of the amino acid sequence constituting the extracellular domain of CD3 is synthesized. This peptide can be chemically synthesized. Alternatively, it can be obtained by genetic engineering techniques using a region in CD3 cDNA that encodes the amino acid sequence corresponding to the extracellular domain. Next, the binding activity of the linear peptide consisting of the amino acid sequence constituting the extracellular domain to a test antigen-binding molecule containing a domain comprising an antibody variable region with binding activity to CD3 is assessed. For example, the binding activity of the antigen-binding molecule to the peptide can be assessed by ELISA using immobilized linear peptide as the antigen. Alternatively, the binding activity of the antigen-binding molecule to the linear peptide can be determined based on the level of inhibition by the linear peptide of binding of the antigen-binding molecule to CD3-expressing cells. These tests can determine the binding activity of the antigen-binding molecule to the linear peptide.

 また、CD3に対して結合活性を有する抗体可変領域を含むドメインを有する被験抗原結合分子が立体構造エピトープを認識することは、次のようにして確認され得る。上記の目的のために、CD3を発現する細胞が調製される。CD3に対して結合活性を有する抗体可変領域を含むドメインを有する被験抗原結合分子がCD3発現細胞に接触した際に当該細胞に強く結合する一方で、当該抗原結合分子が固定化されたCD3の細胞外ドメインを構成するアミノ酸配列からなる線状ペプチドに対して実質的に結合しないとき等が挙げられる。ここで、実質的に結合しないとは、ヒトCD3発現細胞に対する結合活性の80%以下、通常50%以下、好ましくは30%以下、特に好ましくは15%以下の結合活性をいう。 Furthermore, whether a test antigen-binding molecule having a domain containing an antibody variable region that has binding activity to CD3 recognizes a conformational epitope can be confirmed as follows. For this purpose, CD3-expressing cells are prepared. Examples of such cases include when a test antigen-binding molecule having a domain containing an antibody variable region that has binding activity to CD3 binds strongly to CD3-expressing cells upon contact with the cells, but does not substantially bind to a linear peptide consisting of the amino acid sequence that constitutes the extracellular domain of immobilized CD3. Here, "not substantially binding" refers to a binding activity that is 80% or less, typically 50% or less, preferably 30% or less, and particularly preferably 15% or less of the binding activity toward human CD3-expressing cells.

 CD3に対する抗原結合ドメインを含む被験抗原結合分子のCD3発現細胞に対する結合活性を測定する方法としては、例えば、Antibodies A Laboratory Manual記載の方法(Ed Harlow, David Lane, Cold Spring Harbor Laboratory (1988) 359-420)が挙げられる。即ち、CD3発現細胞を抗原とするELISAやFACS(fluorescence activated cell sorting)の原理によって評価され得る。 Methods for measuring the binding activity of a test antigen-binding molecule containing a CD3 antigen-binding domain to CD3-expressing cells include, for example, the method described in Antibodies: A Laboratory Manual (Ed Harlow, David Lane, Cold Spring Harbor Laboratory (1988) 359-420). Specifically, evaluation can be performed using ELISA or FACS (fluorescence activated cell sorting) principles, using CD3-expressing cells as antigens.

 ELISAフォーマットにおいて、CD3に対する抗原結合ドメインを含む被験抗原結合分子のCD3発現細胞に対する結合活性は、酵素反応によって生成するシグナルレベルを比較することによって定量的に評価される。すなわち、CD3発現細胞を固定化したELISAプレートに被験抗原結合分子を加え、細胞に結合した被験抗原結合分子が、被験抗原結合分子を認識する酵素標識抗体を利用して検出される。あるいはFACSにおいては、被験抗原結合分子の希釈系列を作成し、CD3発現細胞に対する抗体結合力価(titer)を決定することにより、CD3発現細胞に対する被験抗原結合分子の結合活性が比較され得る。 In the ELISA format, the binding activity of a test antigen-binding molecule containing a CD3 antigen-binding domain for CD3-expressing cells is quantitatively assessed by comparing the signal levels generated by the enzymatic reaction. That is, the test antigen-binding molecule is added to an ELISA plate on which CD3-expressing cells have been immobilized, and the test antigen-binding molecule bound to the cells is detected using an enzyme-labeled antibody that recognizes the test antigen-binding molecule. Alternatively, in FACS, a dilution series of the test antigen-binding molecule is prepared, and the antibody binding titer for CD3-expressing cells is determined, allowing the binding activity of the test antigen-binding molecule for CD3-expressing cells to be compared.

 緩衝液等に懸濁した細胞表面上に発現している抗原に対する被験抗原結合分子の結合は、フローサイトメーターによって検出することができる。フローサイトメーターとしては、例えば、次のような装置が知られている。
FACSCantoTM II
FACSAriaTM
FACSArrayTM
FACSVantageTM SE
FACSCaliburTM (いずれもBD Biosciences社の商品名)
EPICS ALTRA HyPerSort
Cytomics FC 500
EPICS XL-MCL ADC EPICS XL ADC
Cell Lab Quanta / Cell Lab Quanta SC(いずれもBeckman Coulter社の商品名)
The binding of a test antigen-binding molecule to an antigen expressed on the surface of cells suspended in a buffer solution or the like can be detected using a flow cytometer. Known flow cytometers include, for example, the following:
FACSCanto II
FACSAria
FACSArray
FACSVantage SE
FACSCalibur (both are trade names of BD Biosciences)
EPICS ALTRA HyperSort
Cytomics FC 500
EPICS XL-MCL ADC EPICS XL ADC
Cell Lab Quanta / Cell Lab Quanta SC (both are trade names of Beckman Coulter)

 例えば、CD3に対する抗原結合ドメインを含む被験抗原結合分子の抗原に対する結合活性の好適な測定方法の一例として、次の方法が挙げられる。まず、CD3を発現する細胞と反応させた被験抗原結合分子を認識するFITC標識した二次抗体で染色する。被験抗原結合分子を適宜好適な緩衝液によって希釈することによって、当該会合体が所望の濃度に調製して用いられる。例えば、10μg/mlから10 ng/mlまでの間のいずれかの濃度で使用され得る。次に、FACSCalibur(BD社)により蛍光強度と細胞数が測定される。当該細胞に対する抗体の結合量は、CELL QUEST Software(BD社)を用いて解析することにより得られた蛍光強度、すなわちGeometric Meanの値に反映される。すなわち、当該Geometric Meanの値を得ることにより、被験抗原結合分子の結合量によって表される被験抗原結合分子の結合活性が測定され得る。 For example, the following method is an example of a suitable method for measuring the antigen-binding activity of a test antigen-binding molecule containing a CD3 antigen-binding domain. First, the test antigen-binding molecule is reacted with CD3-expressing cells and stained with an FITC-labeled secondary antibody that recognizes the test antigen-binding molecule. The test antigen-binding molecule is diluted with an appropriate buffer solution to prepare the complex at the desired concentration. For example, it can be used at a concentration between 10 μg/ml and 10 ng/ml. Next, the fluorescence intensity and cell count are measured using a FACSCalibur (BD). The amount of antibody binding to the cells is reflected in the fluorescence intensity, i.e., the Geometric Mean value, obtained by analysis using CELL QUEST Software (BD). In other words, by obtaining the Geometric Mean value, the binding activity of the test antigen-binding molecule, represented by the amount of binding of the test antigen-binding molecule, can be measured.

 CD3に対する抗原結合ドメインを含む被験抗原結合分子が、ある抗原結合分子とエピトープを共有することは、両者の同じエピトープに対する競合によって確認され得る。抗原結合分子間の競合は、交叉ブロッキングアッセイなどによって検出される。例えば競合ELISAアッセイは、好ましい交叉ブロッキングアッセイである。 Whether a test antigen-binding molecule containing a CD3 antigen-binding domain shares an epitope with another antigen-binding molecule can be confirmed by competition between the two for the same epitope. Competition between antigen-binding molecules can be detected by cross-blocking assays, for example. A competitive ELISA assay is a preferred cross-blocking assay.

 具体的には、交叉ブロッキングアッセイにおいては、マイクロタイタープレートのウェル上にコートしたCD3タンパク質が、候補となる競合抗原結合分子の存在下、または非存在下でプレインキュベートされた後に、被験抗原結合分子が添加される。ウェル中のCD3タンパク質に結合した被験抗原結合分子の量は、同じエピトープへの結合に対して競合する候補となる競合抗原結合分子の結合能に間接的に相関している。すなわち同一エピトープに対する競合抗原結合分子の親和性が大きくなればなる程、被験抗原結合分子のCD3タンパク質をコートしたウェルへの結合活性は低下する。 Specifically, in a cross-blocking assay, CD3 protein coated on the wells of a microtiter plate is pre-incubated in the presence or absence of a candidate competing antigen-binding molecule, after which the test antigen-binding molecule is added. The amount of test antigen-binding molecule bound to CD3 protein in the well is indirectly correlated with the binding ability of the candidate competing antigen-binding molecule that competes for binding to the same epitope. In other words, the greater the affinity of the competing antigen-binding molecule for the same epitope, the lower the binding activity of the test antigen-binding molecule to CD3 protein-coated wells.

 CD3タンパク質を介してウェルに結合した被験抗原結合分子の量は、予め抗原結合分子を標識しておくことによって、容易に測定され得る。たとえば、ビオチン標識された抗原結合分子は、アビジンペルオキシダーゼコンジュゲートと適切な基質を使用することにより測定される。ペルオキシダーゼなどの酵素標識を利用した交叉ブロッキングアッセイは、特に競合ELISAアッセイといわれる。抗原結合分子は、検出あるいは測定が可能な他の標識物質で標識され得る。具体的には、放射標識あるいは蛍光標識などが公知である。 The amount of test antigen-binding molecules bound to the wells via CD3 protein can be easily measured by labeling the antigen-binding molecules in advance. For example, biotin-labeled antigen-binding molecules can be measured using an avidin-peroxidase conjugate and an appropriate substrate. Cross-blocking assays using enzyme labels such as peroxidase are specifically referred to as competitive ELISA assays. Antigen-binding molecules can also be labeled with other detectable or measurable labeling substances. Specific examples include radiolabels and fluorescent labels.

 候補の競合抗原結合分子の非存在下で実施されるコントロール試験において得られる結合活性と比較して、競合抗原結合分子が、CD3に対する抗原結合ドメインを含む被験抗原結合分子の結合を少なくとも20%、好ましくは少なくとも20~50%、さらに好ましくは少なくとも50%ブロックできるならば、当該被験抗原結合分子は競合抗原結合分子と実質的に同じエピトープに結合するか、又は同じエピトープへの結合に対して競合する抗原結合分子である。 If a competitor antigen-binding molecule can block the binding of a test antigen-binding molecule comprising an antigen-binding domain to CD3 by at least 20%, preferably at least 20-50%, and more preferably at least 50%, compared to the binding activity obtained in a control test performed in the absence of the candidate competitor antigen-binding molecule, the test antigen-binding molecule binds to substantially the same epitope as the competitor antigen-binding molecule, or is an antigen-binding molecule that competes for binding to the same epitope.

 CD3に対する抗原結合ドメインを含む被験抗原結合分子が結合するエピトープの構造が同定されている場合には、被験抗原結合分子と対照抗原結合分子とがエピトープを共有することは、当該エピトープを構成するペプチドにアミノ酸変異を導入したペプチドに対する両者の抗原結合分子の結合活性を比較することによって評価され得る。 If the structure of the epitope to which a test antigen-binding molecule containing a CD3 antigen-binding domain binds has been identified, whether the test antigen-binding molecule and a control antigen-binding molecule share the same epitope can be assessed by comparing the binding activity of both antigen-binding molecules to a peptide in which amino acid mutations have been introduced into the peptide constituting the epitope.

 こうした結合活性を測定する方法としては、例えば、前記のELISAフォーマットにおいて変異を導入した線状のペプチドに対する被験抗原結合分子及び対照抗原結合分子の結合活性を比較することによって測定され得る。ELISA以外の方法としては、カラムに結合した当該変異ペプチドに対する結合活性を、当該カラムに被検抗原結合分子と対照抗原結合分子を流下させた後に溶出液中に溶出される抗原結合分子を定量することによっても測定され得る。変異ペプチドを例えばGSTとの融合ペプチドとしてカラムに吸着させる方法は公知である。 Such binding activity can be measured, for example, by comparing the binding activity of test and control antigen-binding molecules to a mutated linear peptide in the ELISA format described above. As a method other than ELISA, the binding activity to the mutant peptide bound to a column can also be measured by passing the test and control antigen-binding molecules down the column and then quantifying the antigen-binding molecules eluted in the eluate. Methods for adsorbing mutant peptides to a column, for example as fusion peptides with GST, are known.

 また、同定されたエピトープが立体構造エピトープの場合には、被験抗原結合分子と対照抗原結合分子とがエピトープを共有することは、次の方法で評価され得る。まず、CD3を発現する細胞とエピトープに変異が導入されたCD3を発現する細胞が調製される。これらの細胞がPBS等の適切な緩衝液に懸濁された細胞懸濁液に対して被験抗原結合分子と対照抗原結合分子が添加される。次いで、適宜緩衝液で洗浄された細胞懸濁液に対して、被験抗原結合分子と対照抗原結合分子を認識することができるFITC標識された抗体が添加される。標識抗体によって染色された細胞の蛍光強度と細胞数がFACSCalibur(BD社)によって測定される。被験抗原結合分子と対照抗原結合分子の濃度は好適な緩衝液によって適宜希釈することによって所望の濃度に調製して用いられる。例えば、10μg/mlから10 ng/mlまでの間のいずれかの濃度で使用される。当該細胞に対する標識抗体の結合量は、CELL QUEST Software(BD社)を用いて解析することにより得られた蛍光強度、すなわちGeometric Meanの値に反映される。すなわち、当該Geometric Meanの値を得ることにより、標識抗体の結合量によって表される被験抗原結合分子と対照抗原結合分子の結合活性を測定することができる。 Furthermore, if the identified epitope is a conformational epitope, whether the test and control antigen-binding molecules share the same epitope can be assessed using the following method. First, CD3-expressing cells and cells expressing CD3 with an epitope mutation introduced are prepared. These cells are suspended in an appropriate buffer solution such as PBS, and the test and control antigen-binding molecules are added to the cell suspension. Next, an FITC-labeled antibody that can recognize the test and control antigen-binding molecules is added to the cell suspension after washing with an appropriate buffer solution. The fluorescence intensity and cell count of cells stained with the labeled antibody are measured using a FACSCalibur (BD). The test and control antigen-binding molecules are diluted with an appropriate buffer solution to the desired concentration before use. For example, they are used at a concentration between 10 μg/ml and 10 ng/ml. The amount of labeled antibody bound to the cells is reflected in the fluorescence intensity, i.e., the geometric mean value, obtained by analysis using CELLQUEST Software (BD). In other words, by obtaining the Geometric Mean value, it is possible to measure the binding activity of the test antigen-binding molecule and the control antigen-binding molecule, which is represented by the amount of bound labeled antibody.

 本方法において、例えば「変異CD3発現細胞に実質的に結合しない」ことは、以下の方法によって判断することができる。まず、変異CD3を発現する細胞に対して結合した被験抗原結合分子と対照抗原結合分子を、標識抗体で染色する。次いで細胞の蛍光強度を検出する。蛍光検出にフローサイトメトリーとしてFACSCaliburを用いた場合、得られた蛍光強度はCELL QUEST Softwareを用いて解析され得る。抗原結合分子存在下および非存在下でのGeometric Meanの値から、この比較値(ΔGeo-Mean)を下記の計算式に基づいて算出することにより、抗原結合分子の結合による蛍光強度の増加割合を求めることができる。 In this method, for example, "substantially no binding to mutant CD3-expressing cells" can be determined by the following method. First, test and control antigen-binding molecules bound to cells expressing mutant CD3 are stained with a labeled antibody. The fluorescence intensity of the cells is then detected. When a FACSCalibur is used for flow cytometry to detect fluorescence, the obtained fluorescence intensity can be analyzed using CELL QUEST Software. The percentage increase in fluorescence intensity due to antigen-binding molecule binding can be determined by calculating the comparative value (ΔGeo-Mean) from the Geometric Mean values in the presence and absence of the antigen-binding molecule using the formula below.

 ΔGeo-Mean=Geo-Mean(抗原結合分子存在下)/Geo-Mean(抗原結合分子非存在下) ΔGeo-Mean = Geo-Mean (in the presence of antigen-binding molecules) / Geo-Mean (in the absence of antigen-binding molecules)

 解析によって得られる被験抗原結合分子の変異CD3発現細胞に対する結合量が反映されたGeometric Mean比較値(変異CD3分子ΔGeo-Mean値)を、被験抗原結合分子のCD3発現細胞に対する結合量が反映されたΔGeo-Mean比較値と比較する。この場合において、変異CD3発現細胞及びCD3発現細胞に対するΔGeo-Mean比較値を求める際に使用する被験抗原結合分子の濃度は互いに同一又は実質的に同一の濃度で調整されることが特に好ましい。予めCD3中のエピトープを認識していることが確認された抗原結合分子が、対照抗原結合分子として利用される。 The Geometric Mean comparison value (mutant CD3 molecule ΔGeo-Mean value) obtained by analysis, which reflects the binding amount of the test antigen-binding molecule to mutant CD3-expressing cells, is compared with the ΔGeo-Mean comparison value, which reflects the binding amount of the test antigen-binding molecule to CD3-expressing cells. In this case, it is particularly preferable that the concentrations of the test antigen-binding molecule used to determine the ΔGeo-Mean comparison values for mutant CD3-expressing cells and CD3-expressing cells are adjusted to be identical or substantially identical to each other. An antigen-binding molecule that has been previously confirmed to recognize an epitope in CD3 is used as a control antigen-binding molecule.

 被験抗原結合分子の変異CD3発現細胞に対するΔGeo-Mean比較値が、被験抗原結合分子のCD3発現細胞に対するΔGeo-Mean比較値の、少なくとも80%、好ましくは50%、更に好ましくは30%、特に好ましくは15%より小さければ、「変異CD3発現細胞に実質的に結合しない」ものとする。Geo-Mean値(Geometric Mean)を求める計算式は、CELL QUEST Software User's Guide(BD biosciences社)に記載されている。比較値を比較することによってそれが実質的に同視し得る程度であれば、被験抗原結合分子と対照抗原結合分子のエピトープは同一であると評価され得る。 If the ΔGeo-Mean comparison value of a test antigen-binding molecule for mutant CD3-expressing cells is at least 80%, preferably 50%, more preferably 30%, and especially preferably 15% of the ΔGeo-Mean comparison value of the test antigen-binding molecule for CD3-expressing cells, the antigen-binding molecule is deemed to "not substantially bind to mutant CD3-expressing cells." The formula for calculating the Geo-Mean value (Geometric Mean) is described in the CELL QUEST Software User's Guide (BD biosciences). If the comparison values are substantially equivalent, the epitopes of the test antigen-binding molecule and the control antigen-binding molecule can be determined to be identical.

Fc領域に含まれるFcRn結合ドメインを含む抗原結合分子
 本発明の抗原結合分子の好ましい例として、抗体のFc領域に含まれるFcRn結合ドメインを含む抗原結合分子を挙げることができる。生体内に投与されたタンパク質の血中半減期を延ばす方法として、目的タンパク質に抗体のFcRn結合ドメインを付加し、FcRnを介したリサイクリング機能を利用する方法が良く知られている。
Antigen-binding molecules containing an FcRn-binding domain contained in the Fc region Preferred examples of the antigen-binding molecules of the present invention include antigen-binding molecules containing an FcRn-binding domain contained in the Fc region of an antibody. A well-known method for extending the blood half-life of a protein administered to the body involves adding an antibody FcRn-binding domain to the protein of interest and utilizing the FcRn-mediated recycling function.

 本発明において、「FcRn結合ドメイン」は、FcRnに対して結合活性を有するものであれば特に限定されず、FcRnに直接結合するドメインであってもよいし、間接的に結合するドメインであってもよい。FcRnに直接結合するドメインとしては、例えば、FcRnを抗原とする抗体の可変領域、Fab、抗体のFc領域、これらの断片、アルブミン、アルブミンドメイン3、ヒト血清アルブミン(HSA)、トランスフェリン等が挙げられる。また、FcRnと間接的に結合するドメインとしては、例えば、上述のFcRnに直接結合するドメインに対して結合活性を有するドメインが挙げられる。本発明との関連において、FcRn結合ドメインの態様の1つとして、抗体のFc領域、或いは、Fc領域中のFcRn結合領域を含む断片が挙げられる。ここで、「Fc領域」としては、例えばIgA1、IgA2、IgD、IgE、IgG1、IgG2、IgG3、IgG4、IgMタイプのFc領域が存在し、例えば天然型IgG由来のFc領域を用いることができる。天然型IgGとは、天然に見出されるIgGと同一のアミノ酸配列を包含し、免疫グロブリンガンマ遺伝子により実質的にコードされる抗体のクラスに属するポリペプチドを意味する。例えば天然型ヒトIgGとは天然型ヒトIgG1、天然型ヒトIgG2、天然型ヒトIgG3、天然型ヒトIgG4などを意味する。天然型IgGにはそれから自然に生じる変異体等も含まれる。ヒトIgG1、ヒトIgG2、ヒトIgG3、ヒトIgG4抗体の定常領域としては、遺伝子多型による複数のアロタイプ配列がSequences of proteins of immunological interest, NIH Publication No.91-3242に記載されているが、本発明においてはそのいずれであっても良い。特にヒトIgG1の配列としては、EUナンバリング356~358番目のアミノ酸配列がDELであってもEEMであってもよい。 In the present invention, the term "FcRn-binding domain" is not particularly limited as long as it has binding activity to FcRn, and may be a domain that directly binds to FcRn or a domain that indirectly binds to FcRn. Examples of domains that directly bind to FcRn include the variable region of an antibody whose antigen is FcRn, Fab, the Fc region of an antibody, fragments thereof, albumin, albumin domain 3, human serum albumin (HSA), and transferrin. Examples of domains that indirectly bind to FcRn include domains that have binding activity to the above-mentioned domains that directly bind to FcRn. In the context of the present invention, one embodiment of an FcRn-binding domain is the Fc region of an antibody, or a fragment of the Fc region that contains the FcRn-binding region. Examples of "Fc region" include IgA1, IgA2, IgD, IgE, IgG1, IgG2, IgG3, IgG4, and IgM Fc regions, and an Fc region derived from a native IgG can be used. Native IgG refers to a polypeptide that includes the same amino acid sequence as IgG found in nature and belongs to the class of antibodies substantially encoded by immunoglobulin gamma genes. For example, native human IgG refers to native human IgG1, native human IgG2, native human IgG3, native human IgG4, etc. Native IgG also includes naturally occurring variants thereof. For the constant regions of human IgG1, human IgG2, human IgG3, and human IgG4 antibodies, multiple allotype sequences due to genetic polymorphisms are described in Sequences of proteins of immunological interest, NIH Publication No. 91-3242, and any of these may be used in the present invention. In particular, for the sequence of human IgG1, the amino acid sequence at EU numbering positions 356 to 358 may be DEL or EEM.

 いくつかの態様において、本発明の抗原結合分子は、抗体のFc領域を含む。本発明の抗原結合分子が単一の抗原結合ドメインを含む場合、当該抗原結合ドメインは、二量体Fc領域を構成する2つのポリペプチドのうちの一つに連結されていてもよい。本発明の抗原結合分子が第一の抗原結合ドメインおよび第二の抗原結合ドメインを含む場合、二量体Fc領域を構成する2つのポリペプチドのうちの一方に第一の抗原結合ドメインが連結され、他方に第二の抗原結合ドメインが連結されていてもよいが、そのような態様に限定されない。特定の態様において、本発明の抗原結合分子は、Fcγ受容体に対する結合活性が低下している抗体のFc領域を含む。本発明の多重特異性抗原結合分子が抗体のFc領域を含む場合、catumaxomab等のtrifunctional抗体において見られるような標的抗原非依存的な各種サイトカインの産生誘導が生じるのを避けるため、Fcγ受容体に対する結合活性が低下している抗体のFc領域を用いることが好ましい。
 抗体のFc領域における、Fcγ受容体に対する結合活性の低下をもたらす様々なアミノ酸変異(改変)が知られており、そのような公知のアミノ酸変異(改変)または将来的に同定される他の変異(改変)を有するFc領域を、本発明の抗原結合分子に用いることができる。また、抗体のFc領域とFcγ受容体との結合を介して作用するエフェクター機能の強さは、IgGサブクラスによって異なり、ヒトではIgG1およびIgG3で高く、IgG2およびIgG4で低いことが知られている。そのため、本発明の抗原結合分子に含まれる、Fcγ受容体に対する結合活性が低下している抗体のFc領域として、ヒトIgG2またはIgG4の天然型Fc領域を用いることもできる。
In some embodiments, the antigen-binding molecules of the present invention comprise an antibody Fc region. When the antigen-binding molecules of the present invention comprise a single antigen-binding domain, the antigen-binding domain may be linked to one of the two polypeptides constituting the dimeric Fc region. When the antigen-binding molecules of the present invention comprise a first antigen-binding domain and a second antigen-binding domain, the first antigen-binding domain may be linked to one of the two polypeptides constituting the dimeric Fc region, and the second antigen-binding domain may be linked to the other, but this is not a limitation. In certain embodiments, the antigen-binding molecules of the present invention comprise an antibody Fc region with reduced Fcγ receptor-binding activity. When the multispecific antigen-binding molecules of the present invention comprise an antibody Fc region, it is preferable to use an antibody Fc region with reduced Fcγ receptor-binding activity to avoid the target antigen-independent induction of various cytokine production, as seen in trifunctional antibodies such as catumaxomab.
Various amino acid mutations (modifications) in the Fc region of an antibody that result in reduced binding activity to Fcγ receptors are known, and Fc regions bearing such known amino acid mutations (modifications) or other mutations (modifications) that will be identified in the future can be used in the antigen-binding molecules of the present invention. Furthermore, the strength of the effector function acting through the binding of the antibody Fc region to the Fcγ receptor varies depending on the IgG subclass, and in humans, it is known to be high in IgG1 and IgG3 and low in IgG2 and IgG4. Therefore, native Fc regions of human IgG2 or IgG4 can also be used as the Fc region of an antibody with reduced binding activity to Fcγ receptors that is included in the antigen-binding molecules of the present invention.

Fcγ受容体
 Fcγ受容体とは、IgG1、IgG2、IgG3、IgG4モノクローナル抗体のFc領域に結合し得る受容体をいい、実質的にFcγ受容体遺伝子にコードされるタンパク質のファミリーのいかなるメンバーをも意味する。ヒトでは、このファミリーには、アイソフォームFcγRIa、FcγRIbおよびFcγRIcを含むFcγRI(CD64);アイソフォームFcγRIIa(アロタイプH131およびR131を含む)、FcγRIIb(FcγRIIb-1およびFcγRIIb-2を含む)およびFcγRIIcを含むFcγRII(CD32);およびアイソフォームFcγRIIIa(アロタイプV158およびF158を含む)およびFcγRIIIb(アロタイプFcγRIIIb-NA1およびFcγRIIIb-NA2を含む)を含むFcγRIII(CD16)、並びにいかなる未発見のヒトFcγR類またはFcγRアイソフォームまたはアロタイプも含まれるが、これらに限定されるものではない。FcγRは、ヒト、マウス、ラット、ウサギおよびサルを含むが、これらに限定されるものではない、いかなる生物由来でもよい。マウスFcγR類には、FcγRI(CD64)、FcγRII(CD32)、FcγRIII(CD16)およびFcγRIII-2(CD16-2)、並びにいかなる未発見のマウスFcγR類またはFcγRアイソフォームまたはアロタイプも含まれるが、これらに限定されない。こうしたFcγ受容体の好適な例としてはヒトFcγRI(CD64)、FcγRIIA(CD32)、FcγRIIB(CD32)、FcγRIIIA(CD16)及び/又はFcγRIIIB(CD16)が挙げられる。FcγRIのポリヌクレオチド配列及びアミノ酸配列はそれぞれRefSeq登録番号NM_000566.3及びNP_000557.1にて、FcγRIIAのポリヌクレオチド配列及びアミノ酸配列はそれぞれRefSeq登録番号BC020823.1及びAAH20823.1にて、FcγRIIBのポリヌクレオチド配列及びアミノ酸配列はそれぞれRefSeq登録番号BC146678.1及びAAI46679.1にて、FcγRIIIAのポリヌクレオチド配列及びアミノ酸配列はそれぞれRefSeq登録番号BC033678.1及びAAH33678.1にて、FcγRIIIBのポリヌクレオチド配列及びアミノ酸配列は、それぞれRefSeq登録番号BC128562.1及びAAI28563.1にて登録されている。Fcγ受容体が、IgG1、IgG2、IgG3、IgG4モノクローナル抗体のFc領域に結合活性を有するか否かは、上記に記載されるFACSやELISAフォーマットのほか、ALPHAスクリーン(Amplified Luminescent Proximity Homogeneous Assay)や表面プラズモン共鳴(SPR)現象を利用したBIACORE法等によって確認され得る(Proc.Natl.Acad.Sci.USA (2006) 103 (11), 4005-4010)。
Fcγ Receptor An Fcγ receptor refers to a receptor that can bind to the Fc region of an IgG1, IgG2, IgG3, or IgG4 monoclonal antibody, and refers to any member of a family of proteins substantially encoded by the Fcγ receptor gene. In humans, this family includes, but is not limited to, FcγRI (CD64), which includes the isoforms FcγRIa, FcγRIb, and FcγRIc; FcγRII (CD32), which includes the isoforms FcγRIIa (including allotypes H131 and R131), FcγRIIb (including FcγRIIb-1 and FcγRIIb-2), and FcγRIIc; and FcγRIII (CD16), which includes the isoforms FcγRIIIa (including allotypes V158 and F158) and FcγRIIIb (including allotypes FcγRIIIb-NA1 and FcγRIIIb-NA2), as well as any unidentified human FcγRs or FcγR isoforms or allotypes. FcγRs may be derived from any organism, including, but not limited to, humans, mice, rats, rabbits, and monkeys. Mouse FcγRs include, but are not limited to, FcγRI (CD64), FcγRII (CD32), FcγRIII (CD16), and FcγRIII-2 (CD16-2), as well as any unidentified mouse FcγRs or FcγR isoforms or allotypes. Preferred examples of such Fcγ receptors include human FcγRI (CD64), FcγRIIA (CD32), FcγRIIB (CD32), FcγRIIIA (CD16), and/or FcγRIIIB (CD16). The polynucleotide and amino acid sequences of FcγRI are registered under RefSeq accession numbers NM_000566.3 and NP_000557.1, respectively; the polynucleotide and amino acid sequences of FcγRIIA are registered under RefSeq accession numbers BC020823.1 and AAH20823.1, respectively; the polynucleotide and amino acid sequences of FcγRIIB are registered under RefSeq accession numbers BC146678.1 and AAI46679.1, respectively; the polynucleotide and amino acid sequences of FcγRIIIA are registered under RefSeq accession numbers BC033678.1 and AAH33678.1, respectively; and the polynucleotide and amino acid sequences of FcγRIIIB are registered under RefSeq accession numbers BC128562.1 and AAI28563.1, respectively. Whether an Fcγ receptor has binding activity to the Fc region of an IgG1, IgG2, IgG3, or IgG4 monoclonal antibody can be confirmed by the FACS or ELISA formats described above, as well as by ALPHA screen (Amplified Luminescent Proximity Homogeneous Assay) and the BIACORE method using surface plasmon resonance (SPR) (Proc. Natl. Acad. Sci. USA (2006) 103 (11), 4005-4010).

Fcγ受容体に対する結合活性
 Fc領域がFcγI、FcγIIA、FcγIIB、FcγIIIA及び/又はFcγIIIBのいずれかのFcγ受容体に対する結合活性が低下していることは、上記に記載されるFACSやELISAフォーマットのほか、ALPHAスクリーン(Amplified Luminescent Proximity Homogeneous Assay)や表面プラズモン共鳴(SPR)現象を利用したBIACORE法等によって確認することができる(Proc.Natl.Acad.Sci.USA (2006) 103 (11), 4005-4010)。
Fcγ Receptor-Binding Activity: Reduced binding activity of an Fc region to any of the Fcγ receptors FcγI, FcγIIA, FcγIIB, FcγIIIA, and/or FcγIIIB can be confirmed by the FACS and ELISA formats described above, as well as ALPHA screen (Amplified Luminescent Proximity Homogeneous Assay) and the BIACORE method using surface plasmon resonance (SPR) (Proc. Natl. Acad. Sci. USA (2006) 103 (11), 4005-4010).

 ALPHAスクリーンは、ドナーとアクセプターの2つのビーズを使用するALPHAテクノロジーによって下記の原理に基づいて実施される。ドナービーズに結合した分子が、アクセプタービーズに結合した分子と生物学的に相互作用し、2つのビーズが近接した状態の時にのみ、発光シグナルが検出される。レーザーによって励起されたドナービーズ内のフォトセンシタイザーは、周辺の酸素を励起状態の一重項酸素に変換する。一重項酸素はドナービーズ周辺に拡散し、近接しているアクセプタービーズに到達するとビーズ内の化学発光反応を引き起こし、最終的に光が放出される。ドナービーズに結合した分子とアクセプタービーズに結合した分子が相互作用しないときは、ドナービーズの産生する一重項酸素がアクセプタービーズに到達しないため、化学発光反応は起きない。 The ALPHA screen is performed using ALPHA technology, which uses two beads, donor and acceptor, based on the following principle: A luminescent signal is detected only when a molecule bound to the donor bead biologically interacts with a molecule bound to the acceptor bead and the two beads are in close proximity. A photosensitizer inside the donor bead, excited by a laser, converts surrounding oxygen into excited singlet oxygen. The singlet oxygen diffuses around the donor bead and, when it reaches a nearby acceptor bead, triggers a chemiluminescent reaction within the bead, ultimately resulting in the emission of light. If the molecules bound to the donor bead and the molecules bound to the acceptor bead do not interact, the singlet oxygen produced by the donor bead does not reach the acceptor bead, and no chemiluminescent reaction occurs.

 例えば、ドナービーズにビオチン標識された抗原結合分子が結合され、アクセプタービーズにはグルタチオンSトランスフェラーゼ(GST)でタグ化されたFcγ受容体が結合される。競合する変異Fc領域を有する抗原結合分子の非存在下では、野生型Fc領域を有する抗原結合分子とFcγ受容体とは相互作用し520~620 nmのシグナルを生ずる。タグ化されていない変異Fc領域を有する抗原結合分子は、野生型Fc領域を有する抗原結合分子とFcγ受容体間の相互作用と競合する。競合の結果表れる蛍光の減少を定量することによって相対的な結合親和性が決定され得る。抗体等の抗原結合分子をSulfo-NHS-ビオチン等を用いてビオチン化することは公知である。Fcγ受容体をGSTでタグ化する方法としては、Fcγ受容体をコードするポリヌクレオチドとGSTをコードするポリヌクレオチドをインフレームで融合した融合遺伝子を発現可能なベクターに保持した細胞等において発現し、グルタチオンカラムを用いて精製する方法等が適宜採用され得る。得られたシグナルは例えばGRAPHPAD PRISM(GraphPad社、San Diego)等のソフトウェアを用いて非線形回帰解析を利用する一部位競合(one-site competition)モデルに適合させることにより好適に解析される。 For example, biotin-labeled antigen-binding molecules are bound to donor beads, and glutathione S-transferase (GST)-tagged Fcγ receptors are bound to acceptor beads. In the absence of competing antigen-binding molecules with mutant Fc regions, antigen-binding molecules with wild-type Fc regions interact with Fcγ receptors, generating a signal at 520-620 nm. Antigen-binding molecules with untagged mutant Fc regions compete with the interaction between antigen-binding molecules with wild-type Fc regions and Fcγ receptors. Relative binding affinity can be determined by quantifying the decrease in fluorescence that occurs as a result of competition. It is known to biotinylate antigen-binding molecules such as antibodies using sulfo-NHS-biotin or similar. Methods for tagging Fcγ receptors with GST include expressing a fusion gene in which a polynucleotide encoding the Fcγ receptor and a polynucleotide encoding GST are fused in-frame in cells harboring an expression vector, followed by purification using a glutathione column. The resulting signals are suitably analyzed by fitting them to a one-site competition model using nonlinear regression analysis, for example, using software such as GRAPHPAD PRISM (GraphPad, San Diego).

 相互作用を観察する物質の一方(リガンド)をセンサーチップの金薄膜上に固定し、センサーチップの裏側から金薄膜とガラスの境界面で全反射するように光を当てると、反射光の一部に反射強度が低下した部分(SPRシグナル)が形成される。相互作用を観察する物質の他方(アナライト)をセンサーチップの表面に流しリガンドとアナライトが結合すると、固定化されているリガンド分子の質量が増加し、センサーチップ表面の溶媒の屈折率が変化する。この屈折率の変化により、SPRシグナルの位置がシフトする(逆に結合が解離するとシグナルの位置は戻る)。Biacoreシステムは上記のシフトする量、すなわちセンサーチップ表面での質量変化を縦軸にとり、質量の時間変化を測定データとして表示する(センサーグラム)。センサーグラムのカーブからカイネティクス:結合速度定数(ka)と解離速度定数(kd)が、当該定数の比からアフィニティー(KD)が求められる。BIACORE法では阻害測定法も好適に用いられる。阻害測定法の例はProc.Natl.Acad.Sci.USA (2006) 103 (11), 4005-4010において記載されている。 One of the substances (ligand) whose interaction is to be observed is immobilized on the gold film of a sensor chip. When light is shone from the back of the sensor chip so that it is totally reflected at the interface between the gold film and the glass, a portion of the reflected light shows a decrease in reflection intensity (SPR signal). When the other substance (analyte) whose interaction is to be observed is poured over the surface of the sensor chip and the ligand and analyte bind, the mass of the immobilized ligand molecule increases, changing the refractive index of the solvent on the sensor chip surface. This change in refractive index shifts the position of the SPR signal (conversely, when the bond dissociates, the signal position returns). The Biacore system plots the amount of shift, i.e., the change in mass on the sensor chip surface, on the vertical axis, and displays the change in mass over time as measurement data (sensorgram). The kinetics (binding rate constant (ka) and dissociation rate constant (kd)) can be calculated from the sensorgram curve, and affinity (KD) can be calculated from the ratio of these constants. Inhibition measurement methods are also suitable for use with the BIACORE method. An example of an inhibition assay is described in Proc. Natl. Acad. Sci. USA (2006) 103 (11), 4005-4010.

 本明細書において、Fcγ受容体に対する結合活性が低下しているとは、例えば、上記の解析方法に基づいて、対照とするFc領域を有する抗原結合分子の結合活性に比較して被検抗原結合分子の競合活性が、50%以下、好ましくは45%以下、40%以下、35%以下、30%以下、20%以下、15%以下、特に好ましくは10%以下、9%以下、8%以下、7%以下、6%以下、5%以下、4%以下、3%以下、2%以下、1%以下の結合活性を示すことをいう。 As used herein, "decreased binding activity to an Fcγ receptor" means, for example, that the competitive activity of a test antigen-binding molecule is 50% or less, preferably 45% or less, 40% or less, 35% or less, 30% or less, 20% or less, or 15% or less, and particularly preferably 10% or less, 9% or less, 8% or less, 7% or less, 6% or less, 5% or less, 4% or less, 3% or less, 2% or less, or 1% or less, compared to the binding activity of a control antigen-binding molecule having an Fc region, based on the above-mentioned analytical method.

 対照とする抗原結合分子としては、IgG1、IgG2、IgG3又はIgG4モノクローナル抗体のFc領域を有する抗原結合分子が適宜使用され得る。これらのFc領域として、それぞれ、RefSeq登録番号AAC82527.1、AAB59393.1、CAA27268.1、AAB59394.1にて登録されているアミノ酸配列のN末にAを付加した構造が使用され得る。一態様において、ある特定のアイソタイプの抗体のFc領域の変異体を有する抗原結合分子を被検物質として使用する場合には、当該特定のアイソタイプの抗体のFc領域を有する抗原結合分子を対照として用いることによって、当該変異体が有する変異によるFcγ受容体への結合活性に対する効果を検証することができる。上記のようにして、Fcγ受容体に対する結合活性が低下していることが検証されたFc領域の変異体を有する抗原結合分子が適宜作製される。別の態様において、Fcγ受容体への結合活性について、ある特定のアイソタイプの抗体のFc領域を、別の特定のアイソタイプの抗体のFc領域と比較し、Fcγ受容体に対する結合活性が低いFc領域を、本発明の抗原結合分子に用いることができる。また、そのようにFcγ受容体に対する結合活性が低いFc領域にアミノ酸変異を導入し、Fcγ受容体に対する結合活性がさらに低いFc領域を取得することもできる。 Antigen-binding molecules having the Fc region of an IgG1, IgG2, IgG3, or IgG4 monoclonal antibody can be used as a control, as appropriate. These Fc regions may have structures in which an A is added to the N-terminus of the amino acid sequences registered under RefSeq accession numbers AAC82527.1, AAB59393.1, CAA27268.1, and AAB59394.1, respectively. In one embodiment, when an antigen-binding molecule having a mutant Fc region of an antibody of a certain isotype is used as a test substance, the effect of the mutation in the mutant on the binding activity to Fcγ receptors can be verified by using an antigen-binding molecule having the Fc region of the antibody of that specific isotype as a control. As described above, antigen-binding molecules having Fc region mutants verified to have reduced binding activity to Fcγ receptors can be appropriately prepared. In another embodiment, the Fc region of an antibody of a certain isotype is compared with the Fc region of an antibody of another certain isotype in terms of Fcγ receptor-binding activity, and the Fc region with lower Fcγ receptor-binding activity can be used in the antigen-binding molecules of the present invention. Furthermore, amino acid mutations can be introduced into such an Fc region with low Fcγ receptor-binding activity to obtain an Fc region with even lower Fcγ receptor-binding activity.

 このようなFc領域における変異の例としては、EUナンバリングに従って特定されるアミノ酸である231A-238Sの欠失(WO 2009/011941)、C226S, C229S, P238S, (C220S)(J.Rheumatol (2007) 34, 11)、C226S, C229S(Hum.Antibod.Hybridomas (1990) 1(1), 47-54)、C226S, C229S, E233P, L234V, L235A(Blood (2007) 109, 1185-1192)等の変異が公知である。 Known examples of such mutations in the Fc region include deletion of amino acids 231A-238S, identified according to EU numbering (WO 2009/011941), C226S, C229S, P238S, (C220S) (J. Rheumatol (2007) 34, 11), C226S, C229S (Hum. Antibod. Hybridomas (1990) 1(1), 47-54), and C226S, C229S, E233P, L234V, L235A (Blood (2007) 109, 1185-1192).

 すなわち、特定のアイソタイプの抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれかのアミノ酸;220位、226位、229位、231位、232位、233位、234位、235位、236位、237位、238位、239位、240位、264位、265位、266位、267位、269位、270位、295位、296位、297位、298位、299位、300位、325位、327位、328位、329位、330位、331位、332位が置換されているFc領域を有する抗原結合分子が好適に挙げられる。Fc領域の起源である抗体のアイソタイプとしては特に限定されず、IgG1、IgG2、IgG3又はIgG4モノクローナル抗体を起源とするFc領域が適宜利用され得る。 Preferred examples of such antigen-binding molecules include those having an Fc region in which any of the following amino acids, as specified by EU numbering, have been substituted among the amino acids constituting the Fc region of an antibody of a specific isotype: 220, 226, 229, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 264, 265, 266, 267, 269, 270, 295, 296, 297, 298, 299, 300, 325, 327, 328, 329, 330, 331, or 332. The antibody isotype from which the Fc region originates is not particularly limited, and Fc regions derived from IgG1, IgG2, IgG3, or IgG4 monoclonal antibodies can be used as appropriate.

 例えば、IgG1抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれかの置換(数字がEUナンバリングに従って特定されるアミノ酸残基の位置、数字の前に位置する一文字のアミノ酸記号が置換前のアミノ酸残基、数字の後に位置する一文字のアミノ酸記号が置換前のアミノ酸残基をそれぞれ表す);
(a)L234F、L235E、P331S、
(b)C226S、C229S、P238S、
(c)C226S、C229S、
(d)C226S、C229S、E233P、L234V、L235A
(e)L234A、L235A又はL235R、N297A
(f)L235A又はL235R、S239K、N297A
が施されているFc領域、又は、231位から238位のアミノ酸配列が欠失したFc領域を有する抗原結合分子も適宜使用され得る。
For example, any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG1 antibody (the numbers indicate the positions of the amino acid residues specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution):
(a) L234F, L235E, P331S,
(b) C226S, C229S, P238S,
(c) C226S, C229S,
(d) C226S, C229S, E233P, L234V, L235A
(e) L234A, L235A or L235R, N297A
(f) L235A or L235R, S239K, N297A
Alternatively, antigen-binding molecules having an Fc region in which the amino acid sequence at positions 231 to 238 has been deleted may also be used as appropriate.

 また、IgG2抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれかの置換(数字がEUナンバリングに従って特定されるアミノ酸残基の位置、数字の前に位置する一文字のアミノ酸記号が置換前のアミノ酸残基、数字の後に位置する一文字のアミノ酸記号が置換前のアミノ酸残基をそれぞれ表す);
(g)H268Q、V309L、A330S、P331S
(h)V234A
(i)G237A
(j)V234A、G237A
(k)A235E、G237A
(l)V234A、A235E、G237A
が施されているFc領域を有する抗原結合分子も適宜使用され得る。
Furthermore, any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG2 antibody (the numbers indicate the amino acid residue positions specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution):
(g) H268Q, V309L, A330S, P331S
(h) V234A
(i) G237A
(j) V234A, G237A
(k) A235E, G237A
(l) V234A, A235E, G237A
Antigen-binding molecules having an Fc region modified with α-glucan may also be used appropriately.

 また、IgG3抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれかの置換(数字がEUナンバリングに従って特定されるアミノ酸残基の位置、数字の前に位置する一文字のアミノ酸記号が置換前のアミノ酸残基、数字の後に位置する一文字のアミノ酸記号が置換前のアミノ酸残基をそれぞれ表す);
(m)F241A
(n)D265A
(o)V264A
が施されているFc領域を有する抗原結合分子も適宜使用され得る。
Furthermore, any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG3 antibody (the numbers indicate the amino acid residue positions specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution):
(m) F241A
(n) D265A
(o) V264A
Antigen-binding molecules having an Fc region modified with α-glucan may also be used appropriately.

 また、IgG4抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれかの置換(数字がEUナンバリングに従って特定されるアミノ酸残基の位置、数字の前に位置する一文字のアミノ酸記号が置換前のアミノ酸残基、数字の後に位置する一文字のアミノ酸記号が置換前のアミノ酸残基をそれぞれ表す);
(p)L235A、G237A、E318A
(q)L235E
(r)F234A、L235A
が施されているFc領域を有する抗原結合分子も適宜使用され得る。
Furthermore, any of the following substitutions, specified according to EU numbering, among the amino acids constituting the Fc region of an IgG4 antibody (the numbers indicate the amino acid residue positions specified according to EU numbering, the single-letter amino acid code preceding the number indicates the amino acid residue before substitution, and the single-letter amino acid code following the number indicates the amino acid residue before substitution):
(p) L235A, G237A, E318A
(q) L235E
(r) F234A, L235A
Antigen-binding molecules having an Fc region modified with α-glucan may also be used appropriately.

 その他の好ましい例として、IgG1抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれかのアミノ酸;233位、234位、235位、236位、237位、327位、330位、331位が、対応するIgG2またはIgG4においてそのEUナンバリングが対応するアミノ酸に置換されているFc領域を有する抗原結合分子が挙げられる。 Other preferred examples include antigen-binding molecules having an Fc region in which any of the following amino acids, identified according to EU numbering, that constitute the Fc region of an IgG1 antibody are substituted with amino acids corresponding to the corresponding EU numbering in the corresponding IgG2 or IgG4: 233, 234, 235, 236, 237, 327, 330, and 331.

 その他の好ましい例として、IgG1抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される下記のいずれか一つ又はそれ以上のアミノ酸;234位、235位、297位が他のアミノ酸によって置換されているFc領域を有する抗原結合分子が好適に挙げられる。置換後に存在するアミノ酸の種類は特に限定されないが、234位、235位、297位のいずれか一つ又はそれ以上のアミノ酸がアラニンに置換されているFc領域を有する抗原結合分子が特に好ましい。 Other preferred examples include antigen-binding molecules having an Fc region in which one or more of the amino acids at positions 234, 235, and 297, identified according to EU numbering, among the amino acids constituting the Fc region of an IgG1 antibody, have been substituted with other amino acids. The type of amino acid present after substitution is not particularly limited, but antigen-binding molecules having an Fc region in which one or more of the amino acids at positions 234, 235, and 297 have been substituted with alanine are particularly preferred.

 その他の好ましい例として、IgG1抗体のFc領域を構成するアミノ酸のうち、EUナンバリングに従って特定される265位のアミノ酸が他のアミノ酸によって置換されているFc領域を有する抗原結合分子が好適に挙げられる。置換後に存在するアミノ酸の種類は特に限定されないが、265位のアミノ酸がアラニンに置換されているFc領域を有する抗原結合分子が特に好ましい。 Another preferred example is an antigen-binding molecule having an Fc region in which the amino acid at position 265, as specified by EU numbering, among the amino acids constituting the Fc region of an IgG1 antibody, has been substituted with another amino acid. The type of amino acid present after substitution is not particularly limited, but antigen-binding molecules having an Fc region in which the amino acid at position 265 has been substituted with alanine are particularly preferred.

 特定の態様において、本発明の抗原結合分子は、Fcγ受容体に対する結合活性が低下しているFc領域を含む。そのような態様の例として、Fc領域に起因するエフェクター機能が減少した抗体が挙げられ、当該抗体は、特定のエフェクター機能(補体およびADCCなど)は不要または有害である場合に望ましい。減少したエフェクター機能を伴う抗体の例としては、Fc領域残基238、265、269、270、297、327、および329の1つまたは複数の置換を伴うものが挙げられる(米国特許第6,737,056号)。このようなFc変異体は、残基265および297のアラニンへの置換を伴ういわゆる「DANA」Fc変異体(米国特許第7,332,581号)を含む、アミノ酸ポジション265、269、270、297、および327の2つ以上の置換を伴うFc変異体を含む。 In certain embodiments, the antigen-binding molecules of the present invention comprise an Fc region with reduced binding activity to Fcγ receptors. Examples of such embodiments include antibodies with reduced effector functions attributable to the Fc region, which are desirable when certain effector functions (such as complement and ADCC) are unnecessary or deleterious. Examples of antibodies with reduced effector functions include those with one or more substitutions at Fc region residues 238, 265, 269, 270, 297, 327, and 329 (U.S. Patent No. 6,737,056). Such Fc variants include Fc variants with two or more substitutions at amino acid positions 265, 269, 270, 297, and 327, including the so-called "DANA" Fc variant with substitutions of residues 265 and 297 to alanine (U.S. Patent No. 7,332,581).

 Fc領域変異体の他の例については、米国特許第5,648,260号;米国特許第5,624,821号;およびWO94/29351も参照のこと。 For other examples of Fc region variants, see also U.S. Patent No. 5,648,260; U.S. Patent No. 5,624,821; and WO94/29351.

抗体の作製方法
 本発明の抗原結合分子の好ましい態様の1つとして、本発明の抗体の可変領域を含む、抗体を挙げることができる。
Method for Producing Antibodies One preferred embodiment of the antigen-binding molecule of the present invention is an antibody comprising the variable region of the antibody of the present invention.

 所望の結合活性を有する抗体を作製する方法は当業者において公知であり、ポリクローナルまたはモノクローナル抗体として取得され得る。本発明の抗体としては、哺乳動物由来のモノクローナル抗体が好適に作製され得る。哺乳動物由来のモノクローナル抗体には、ハイブリドーマにより産生されるもの、および遺伝子工学的手法により抗体遺伝子を含む発現ベクターで形質転換した宿主細胞によって産生されるもの等が含まれる。 Methods for producing antibodies with the desired binding activity are known to those skilled in the art, and they can be obtained as polyclonal or monoclonal antibodies. Monoclonal antibodies derived from mammals are preferably produced as antibodies of the present invention. Mammalian-derived monoclonal antibodies include those produced by hybridomas and those produced by host cells transformed by genetic engineering techniques with an expression vector containing an antibody gene.

 抗体取得のために免疫される哺乳動物としては、特定の動物に限定されるものではないが、ハイブリドーマ作製のための細胞融合に使用する親細胞との適合性を考慮して選択するのが好ましい。一般的にはげっ歯類の動物、例えば、マウス、ラット、ハムスター、あるいはウサギ、サル等が好適に使用される。 The mammal to be immunized to obtain antibodies is not limited to a specific animal, but it is preferable to select it taking into consideration its compatibility with the parent cells used in cell fusion to produce hybridomas. Generally, rodents such as mice, rats, hamsters, rabbits, and monkeys are preferably used.

 公知の方法にしたがって上記の動物が感作抗原により免疫される。例えば、一般的な方法として、感作抗原が哺乳動物の腹腔内または皮下に注射によって投与されることにより免疫が実施される。具体的には、PBS(Phosphate-Buffered Saline)や生理食塩水等で適当な希釈倍率で希釈された感作抗原が、所望により通常のアジュバント、例えばフロイント完全アジュバントと混合され、乳化された後に、該感作抗原が哺乳動物に4から21日毎に数回投与される。また、感作抗原の免疫時には適当な担体が使用され得る。特に分子量の小さい部分ペプチドが感作抗原として用いられる場合には、アルブミン、キーホールリンペットヘモシアニン等の担体タンパク質と結合した該感作抗原ペプチドを免疫することが望ましい場合もある。 The above-mentioned animals are immunized with the sensitizing antigen according to known methods. For example, a common method is to administer the sensitizing antigen to the mammal by intraperitoneal or subcutaneous injection. Specifically, the sensitizing antigen is diluted at an appropriate dilution ratio with PBS (Phosphate-Buffered Saline) or physiological saline, and if desired, mixed with a conventional adjuvant, such as Freund's complete adjuvant, and emulsified. The sensitizing antigen is then administered to the mammal several times every 4 to 21 days. An appropriate carrier may also be used when immunizing with the sensitizing antigen. In particular, when a partial peptide with a small molecular weight is used as the sensitizing antigen, it may be desirable to immunize with the sensitizing antigen peptide bound to a carrier protein such as albumin or keyhole limpet hemocyanin.

 また、所望の抗体を産生するハイブリドーマは、DNA免疫を使用し、以下のようにしても作製され得る。DNA免疫とは、免疫動物中で抗原タンパク質をコードする遺伝子が発現され得るような態様で構築されたベクターDNAが投与された当該免疫動物中で、感作抗原が当該免疫動物の生体内で発現されることによって、免疫刺激が与えられる免疫方法である。タンパク質抗原が免疫動物に投与される一般的な免疫方法と比べて、DNA免疫には、次のような優位性が期待される。
-膜タンパク質の構造を維持して免疫刺激が与えられ得る
-免疫抗原を精製する必要が無い
Hybridomas producing the desired antibodies can also be prepared using DNA immunization as follows. DNA immunization is an immunization method in which a vector DNA constructed in such a manner that a gene encoding an antigen protein can be expressed in the immunized animal is administered to the immunized animal, and a sensitizing antigen is expressed in the immunized animal's body, thereby conferring immune stimulation. Compared to general immunization methods in which a protein antigen is administered to the immunized animal, DNA immunization is expected to have the following advantages:
- Immunostimulation can be achieved by maintaining the structure of membrane proteins - No need to purify the immunizing antigen

 DNA免疫によって本発明のモノクローナル抗体を得るために、まず、抗原タンパク質を発現するDNAが免疫動物に投与される。抗原タンパク質をコードするDNAは、PCRなどの公知の方法によって合成され得る。得られたDNAが適当な発現ベクターに挿入され、免疫動物に投与される。発現ベクターとしては、たとえばpcDNA3.1などの市販の発現ベクターが好適に利用され得る。ベクターを生体に投与する方法として、一般的に用いられている方法が利用され得る。たとえば、発現ベクターが吸着した金粒子が、gene gunで免疫動物個体の細胞内に導入されることによってDNA免疫が行われる。 To obtain the monoclonal antibody of the present invention by DNA immunization, first, DNA that expresses the antigen protein is administered to the animal to be immunized. DNA encoding the antigen protein can be synthesized by known methods such as PCR. The obtained DNA is inserted into an appropriate expression vector and administered to the animal to be immunized. As the expression vector, commercially available expression vectors such as pcDNA3.1 can be suitably used. Commonly used methods can be used to administer the vector into the living body. For example, DNA immunization is performed by introducing gold particles to which the expression vector has been adsorbed into the cells of the animal to be immunized using a gene gun.

 このように哺乳動物が免疫され、血清中における抗原に結合する抗体力価の上昇が確認された後に、哺乳動物から免疫細胞が採取され、細胞融合に供される。好ましい免疫細胞としては、特に脾細胞が使用され得る。 After a mammal is immunized in this manner and an increase in the antibody titer that binds to the antigen in the serum is confirmed, immune cells are collected from the mammal and subjected to cell fusion. Splenocytes, in particular, are preferred as immune cells.

 前記免疫細胞と融合される細胞として、哺乳動物のミエローマ細胞が用いられる。ミエローマ細胞は、スクリーニングのための適当な選択マーカーを備えていることが好ましい。選択マーカーとは、特定の培養条件の下で生存できる(あるいはできない)形質を指す。選択マーカーには、ヒポキサンチン-グアニン-ホスホリボシルトランスフェラーゼ欠損(以下HGPRT欠損と省略する)、あるいはチミジンキナーゼ欠損(以下TK欠損と省略する)などが公知である。HGPRTやTKの欠損を有する細胞は、ヒポキサンチン-アミノプテリン-チミジン感受性(以下HAT感受性と省略する)を有する。HAT感受性の細胞はHAT選択培地中でDNA合成を行うことができず死滅するが、正常な細胞と融合すると正常細胞のサルベージ回路を利用してDNAの合成を継続することができるためHAT選択培地中でも増殖するようになる。 Mammalian myeloma cells are used as the cells to be fused with the immune cells. It is preferable that the myeloma cells have an appropriate selection marker for screening. A selection marker refers to a trait that allows (or prevents) survival under specific culture conditions. Known selection markers include hypoxanthine-guanine-phosphoribosyltransferase deficiency (hereinafter abbreviated as HGPRT deficiency) and thymidine kinase deficiency (hereinafter abbreviated as TK deficiency). Cells that are HGPRT or TK deficient are hypoxanthine-aminopterin-thymidine sensitive (hereinafter abbreviated as HAT sensitive). HAT-sensitive cells are unable to synthesize DNA in HAT selective medium and die, but when fused with normal cells, they can continue to synthesize DNA using the salvage pathway of normal cells, allowing them to grow even in HAT selective medium.

 HGPRT欠損やTK欠損の細胞は、それぞれ6チオグアニン、8アザグアニン(以下8AGと省略する)、あるいは5'ブロモデオキシウリジンを含む培地で選択され得る。これらのピリミジンアナログをDNA中に取り込む正常な細胞は死滅する。他方、これらのピリミジンアナログを取り込めないこれらの酵素を欠損した細胞は、選択培地の中で生存することができる。この他G418耐性と呼ばれる選択マーカーは、ネオマイシン耐性遺伝子によって2-デオキシストレプタミン系抗生物質(ゲンタマイシン類似体)に対する耐性を与える。細胞融合に好適な種々のミエローマ細胞が公知である。 HGPRT-deficient or TK-deficient cells can be selected on media containing 6-thioguanine, 8-azaguanine (hereafter abbreviated as 8AG), or 5'-bromodeoxyuridine, respectively. Normal cells that incorporate these pyrimidine analogs into their DNA die. On the other hand, cells lacking these enzymes and unable to incorporate these pyrimidine analogs can survive in selective media. Another selection marker, called G418 resistance, confers resistance to 2-deoxystreptamine antibiotics (gentamicin analogs) via the neomycin resistance gene. Various myeloma cell lines suitable for cell fusion are known.

 このようなミエローマ細胞として、例えば、P3(P3x63Ag8.653)(J. Immunol.(1979)123 (4), 1548-1550)、P3x63Ag8U.1(Current Topics in Microbiology and Immunology(1978)81, 1-7)、NS-1(C. Eur. J. Immunol.(1976)6 (7), 511-519)、MPC-11(Cell(1976)8 (3), 405-415)、SP2/0(Nature(1978)276 (5685), 269-270)、FO(J. Immunol. Methods(1980)35 (1-2), 1-21)、S194/5.XX0.BU.1(J. Exp. Med.(1978)148 (1), 313-323)、R210(Nature(1979)277 (5692), 131-133)等が好適に使用され得る。 Such myeloma cells include, for example, P3 (P3x63Ag8.653) (J. Immunol. (1979) 123 (4), 1548-1550), P3x63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (C. Eur. J. Immunol. (1976) 6 (7), 511-519), and MPC-11 (Cell ( 1976) 8 (3), 405-415), SP2/0 (Nature (1978) 276 (5685), 269-270), FO (J. Immunol. Methods (1980) 35 (1-2), 1-21), S194/5.XX0.BU.1 (J. Exp. Med. (1978) 148 (1), 313-323), R210 (Nature (1979) 277 (5692), 131-133), etc. can be suitably used.

 基本的には公知の方法、たとえば、ケーラーとミルステインらの方法(Methods Enzymol.(1981)73, 3-46)等に準じて、前記免疫細胞とミエローマ細胞との細胞融合が行われる。 The immune cells are fused with myeloma cells essentially according to known methods, such as the method of Köhler and Milstein et al. (Methods Enzymol. (1981) 73, 3-46).

 より具体的には、例えば細胞融合促進剤の存在下で通常の栄養培養液中で、前記細胞融合が実施され得る。融合促進剤としては、例えばポリエチレングリコール(PEG)、センダイウイルス(HVJ)等が使用され、更に融合効率を高めるために所望によりジメチルスルホキシド等の補助剤が添加されて使用される。 More specifically, the cell fusion can be carried out in a standard nutrient culture medium in the presence of a cell fusion promoter. Fusion promoters such as polyethylene glycol (PEG) and Sendai virus (HVJ) can be used, and if desired, an adjuvant such as dimethyl sulfoxide can be added to further enhance fusion efficiency.

 免疫細胞とミエローマ細胞との使用割合は任意に設定され得る。例えば、ミエローマ細胞に対して免疫細胞を1から10倍とするのが好ましい。前記細胞融合に用いる培養液としては、例えば、前記ミエローマ細胞株の増殖に好適なRPMI1640培養液、MEM培養液、その他、この種の細胞培養に用いられる通常の培養液が使用され、さらに、牛胎児血清(FCS)等の血清補液が好適に添加され得る。 The ratio of immune cells to myeloma cells can be set as desired. For example, a ratio of immune cells to myeloma cells of 1 to 10 is preferred. The culture medium used for the cell fusion may be, for example, RPMI1640 culture medium, MEM culture medium, or any other conventional culture medium used for this type of cell culture, which is suitable for growing the myeloma cell line. Furthermore, serum supplements such as fetal calf serum (FCS) may be suitably added.

 細胞融合は、前記免疫細胞とミエローマ細胞との所定量を前記培養液中でよく混合し、予め37℃程度に加温されたPEG溶液(例えば平均分子量1000から6000程度)が通常30から60%(w/v)の濃度で添加される。混合液が緩やかに混合されることによって所望の融合細胞(ハイブリドーマ)が形成される。次いで、上記に挙げた適当な培養液が逐次添加され、遠心して上清を除去する操作を繰り返すことによりハイブリドーマの生育に好ましくない細胞融合剤等が除去され得る。 Cell fusion is performed by thoroughly mixing a predetermined amount of the immune cells and myeloma cells in the culture medium, and then adding a PEG solution (for example, an average molecular weight of approximately 1000 to 6000) that has been preheated to approximately 37°C, usually at a concentration of 30 to 60% (w/v). The mixture is gently mixed to form the desired fused cells (hybridomas). Next, the appropriate culture medium listed above is added sequentially, and the mixture is centrifuged and the supernatant is removed repeatedly, allowing cell fusion agents and other substances that are undesirable for hybridoma growth to be removed.

 このようにして得られたハイブリドーマは、通常の選択培養液、例えばHAT培養液(ヒポキサンチン、アミノプテリンおよびチミジンを含む培養液)で培養することにより選択され得る。所望のハイブリドーマ以外の細胞(非融合細胞)が死滅するのに十分な時間(通常、係る十分な時間は数日から数週間である)上記HAT培養液を用いた培養が継続され得る。次いで、通常の限界希釈法によって、所望の抗体を産生するハイブリドーマのスクリーニングおよび単一クローニングが実施される。 The hybridomas obtained in this manner can be selected by culturing them in a conventional selective culture medium, such as HAT culture medium (a culture medium containing hypoxanthine, aminopterin, and thymidine). Culture can be continued using the above-mentioned HAT culture medium for a sufficient period of time (usually several days to several weeks) for cells other than the desired hybridoma (unfused cells) to die. Hybridomas producing the desired antibody are then screened and single-cloned using the conventional limiting dilution method.

 このようにして得られたハイブリドーマは、細胞融合に用いられたミエローマが有する選択マーカーに応じた選択培養液を利用することによって選択され得る。例えばHGPRTやTKの欠損を有する細胞は、HAT培養液(ヒポキサンチン、アミノプテリンおよびチミジンを含む培養液)で培養することにより選択され得る。すなわち、HAT感受性のミエローマ細胞を細胞融合に用いた場合、HAT培養液中で、正常細胞との細胞融合に成功した細胞が選択的に増殖し得る。所望のハイブリドーマ以外の細胞(非融合細胞)が死滅するのに十分な時間、上記HAT培養液を用いた培養が継続される。具体的には、一般に、数日から数週間の培養によって、所望のハイブリドーマが選択され得る。次いで、通常の限界希釈法によって、所望の抗体を産生するハイブリドーマのスクリーニングおよび単一クローニングが実施され得る。 Hybridomas obtained in this manner can be selected using a selective culture medium that corresponds to the selection marker possessed by the myeloma used in cell fusion. For example, cells lacking HGPRT or TK can be selected by culturing in HAT culture medium (a culture medium containing hypoxanthine, aminopterin, and thymidine). That is, when HAT-sensitive myeloma cells are used for cell fusion, cells that have successfully fused with normal cells can selectively grow in HAT culture medium. Culture in the above HAT culture medium is continued for a period of time sufficient for cells other than the desired hybridoma (non-fused cells) to die. Specifically, the desired hybridoma can generally be selected by culturing for several days to several weeks. Hybridomas producing the desired antibody can then be screened and single-cloned using the conventional limiting dilution method.

 所望の抗体のスクリーニングおよび単一クローニングが、公知の抗原抗体反応に基づくスクリーニング方法によって好適に実施され得る。所望の抗体は、例えば、FACS(fluorescence activated cell sorting)によってスクリーニングされ得る。FACSは、蛍光抗体と接触させた細胞をレーザー光で解析し、個々の細胞が発する蛍光を測定することによって細胞表面への抗体の結合を測定することを可能にするシステムである。 Screening and single cloning of the desired antibody can be suitably carried out by known screening methods based on antigen-antibody reactions. The desired antibody can be screened, for example, by FACS (fluorescence activated cell sorting). FACS is a system that analyzes cells that have been brought into contact with a fluorescent antibody using laser light, allowing the binding of the antibody to the cell surface to be measured by measuring the fluorescence emitted by individual cells.

 FACSによって本発明のモノクローナル抗体を産生するハイブリドーマをスクリーニングするためには、まず産生される抗体が結合する抗原を発現する細胞を調製する。スクリーニングのための好ましい細胞は、当該抗原を強制発現させた哺乳動物細胞である。宿主細胞として使用した形質転換されていない哺乳動物細胞を対照として用いることによって、細胞表面の抗原に対する抗体の結合活性が選択的に検出され得る。すなわち、宿主細胞に結合せず、抗原を強制発現させた細胞に結合する抗体を産生するハイブリドーマを選択することによって、所望のモノクローナル抗体を産生するハイブリドーマが取得され得る。 To screen hybridomas that produce the monoclonal antibodies of the present invention using FACS, first prepare cells that express the antigen to which the produced antibody binds. Preferred cells for screening are mammalian cells that have been forced to express the antigen. By using untransformed mammalian cells that have been used as host cells as a control, the binding activity of the antibody to the antigen on the cell surface can be selectively detected. In other words, by selecting hybridomas that produce antibodies that do not bind to host cells but that bind to cells that have been forced to express the antigen, hybridomas that produce the desired monoclonal antibody can be obtained.

 あるいは対象となる抗原を発現した細胞を固定化し、当該抗原発現細胞に対する抗体の結合活性がELISAの原理に基づいて評価され得る。たとえば、ELISAプレートのウェルに抗原発現細胞が固定化される。ハイブリドーマの培養上清をウェル内の固定化細胞に接触させ、固定化細胞に結合する抗体が検出される。モノクローナル抗体がマウス由来の場合、細胞に結合した抗体は、抗マウスイムノグロブリン抗体によって検出され得る。これらのスクリーニングによって選択された、抗原に対する結合能を有する所望の抗体を産生するハイブリドーマは、限界希釈法等によりクローニングされ得る。
 このようにして作製されるモノクローナル抗体を産生するハイブリドーマは通常の培養液中で継代培養され得る。また、該ハイブリドーマは液体窒素中で長期にわたって保存され得る。
Alternatively, cells expressing a target antigen can be immobilized, and the binding activity of an antibody to the antigen-expressing cells can be evaluated based on the principles of ELISA. For example, antigen-expressing cells are immobilized in the wells of an ELISA plate. Hybridoma culture supernatant is contacted with the immobilized cells in the wells, and antibodies binding to the immobilized cells are detected. If the monoclonal antibody is derived from a mouse, the antibody bound to the cells can be detected with an anti-mouse immunoglobulin antibody. Hybridomas producing the desired antibody capable of binding to the antigen, selected by these screening methods, can be cloned by limiting dilution or other methods.
The hybridomas producing the monoclonal antibodies thus prepared can be subcultured in a conventional culture medium and can be stored for a long period of time in liquid nitrogen.

 当該ハイブリドーマを通常の方法に従い培養し、その培養上清から所望のモノクローナル抗体が取得され得る。あるいはハイブリドーマをこれと適合性がある哺乳動物に投与して増殖せしめ、その腹水からモノクローナル抗体が取得され得る。前者の方法は、高純度の抗体を得るのに好適なものである。 The hybridoma can be cultured according to conventional methods, and the desired monoclonal antibody can be obtained from the culture supernatant. Alternatively, the hybridoma can be administered to a compatible mammal to grow, and the monoclonal antibody can be obtained from the ascites fluid. The former method is suitable for obtaining highly pure antibodies.

 当該ハイブリドーマ等の抗体産生細胞からクローニングされる抗体遺伝子によってコードされる抗体も好適に利用され得る。クローニングした抗体遺伝子を適当なベクターに組み込んで宿主に導入することによって、当該遺伝子によってコードされる抗体が発現する。抗体遺伝子の単離と、ベクターへの導入、そして宿主細胞の形質転換のための方法は例えば、Vandammeらによって既に確立されている(Eur.J. Biochem.(1990)192 (3), 767-775)。下記に述べるように組換え抗体の製造方法もまた公知である。 Antibodies encoded by antibody genes cloned from antibody-producing cells such as hybridomas can also be suitably used. The cloned antibody gene is incorporated into a suitable vector and introduced into a host, whereby the antibody encoded by the gene is expressed. Methods for isolating antibody genes, introducing them into vectors, and transforming host cells have already been established, for example, by Vandamme et al. (Eur. J. Biochem. (1990) 192 (3), 767-775). Methods for producing recombinant antibodies are also known, as described below.

 抗体の可変領域(V領域)をコードするcDNAを取得するためには、通常、まずハイブリドーマから全RNAが抽出される。細胞からmRNAを抽出するための方法として、たとえば次のような方法を利用することができる。
 -グアニジン超遠心法(Biochemistry (1979) 18 (24), 5294-5299)
 -AGPC法(Anal. Biochem. (1987) 162 (1), 156-159)
To obtain cDNA encoding the variable region (V region) of an antibody, total RNA is usually extracted from the hybridoma. mRNA can be extracted from cells using the following methods, for example.
- Guanidine ultracentrifugation method (Biochemistry (1979) 18 (24), 5294-5299)
-AGPC method (Anal. Biochem. (1987) 162 (1), 156-159)

 抽出されたmRNAは、例えばmRNA Purification Kit (GEヘルスケアバイオサイエンス製)等を使用して精製され得る。あるいは、QuickPrep mRNA Purification Kit (GEヘルスケアバイオサイエンス製)などのように、細胞から直接全mRNAを抽出するためのキットも市販されている。このようなキットを用いて、ハイブリドーマからmRNAが取得され得る。得られたmRNAから逆転写酵素を用いて抗体V領域をコードするcDNAが合成され得る。cDNAは、AMV Reverse Transcriptase First-strand cDNA Synthesis Kit(生化学工業社製)等によって合成され得る。また、cDNAの合成および増幅のために、SMART RACE cDNA 増幅キット(Clontech製)およびPCRを用いた5'-RACE法(Proc. Natl. Acad. Sci. USA (1988) 85 (23), 8998-9002、Nucleic Acids Res. (1989) 17 (8), 2919-2932)が適宜利用され得る。更にこうしたcDNAの合成の過程においてcDNAの両末端に後述する適切な制限酵素サイトが導入され得る。 The extracted mRNA can be purified using, for example, an mRNA Purification Kit (manufactured by GE Healthcare Biosciences). Alternatively, kits for extracting total mRNA directly from cells, such as the QuickPrep mRNA Purification Kit (manufactured by GE Healthcare Biosciences), are also commercially available. Using such kits, mRNA can be obtained from hybridomas. cDNA encoding the antibody V region can be synthesized from the obtained mRNA using reverse transcriptase. cDNA can be synthesized using an AMV Reverse Transcriptase First-Strand cDNA Synthesis Kit (manufactured by Seikagaku Corporation) or the like. Additionally, the SMART RACE cDNA Amplification Kit (Clontech) and the 5'-RACE method using PCR (Proc. Natl. Acad. Sci. USA (1988) 85 (23), 8998-9002, Nucleic Acids Res. (1989) 17 (8), 2919-2932) can be used appropriately to synthesize and amplify cDNA. Furthermore, during the cDNA synthesis process, appropriate restriction enzyme sites can be introduced at both ends of the cDNA, as described below.

 得られたPCR産物から目的とするcDNA断片が精製され、次いでベクターDNAと連結される。このように組換えベクターが作製され、大腸菌等に導入されコロニーが選択された後に、該コロニーを形成した大腸菌から所望の組換えベクターが調製され得る。そして、該組換えベクターが目的とするcDNAの塩基配列を有しているか否かについて、公知の方法、例えば、ジデオキシヌクレオチドチェインターミネーション法等により確認される。 The desired cDNA fragment is purified from the resulting PCR product and then ligated to vector DNA. A recombinant vector is thus created, introduced into E. coli or other bacteria, and colonies are selected. The desired recombinant vector can then be prepared from the E. coli that formed the colonies. Whether the recombinant vector contains the base sequence of the desired cDNA is then confirmed using known methods, such as the dideoxynucleotide chain termination method.

 可変領域をコードする遺伝子を取得するためには、可変領域遺伝子増幅用のプライマーを使った5'-RACE法を利用するのが簡便である。まずハイブリドーマ細胞より抽出されたRNAを鋳型としてcDNAが合成され、5'-RACE cDNAライブラリーが得られる。5'-RACE cDNAライブラリーの合成にはSMART RACE cDNA 増幅キットなど市販のキットが適宜用いられる。 The easiest way to obtain genes encoding variable regions is to use the 5'-RACE method, which uses primers for amplifying variable region genes. First, cDNA is synthesized using RNA extracted from hybridoma cells as a template, and a 5'-RACE cDNA library is obtained. A commercially available kit, such as the SMART RACE cDNA Amplification Kit, can be used to synthesize the 5'-RACE cDNA library.

 得られた5'-RACE cDNAライブラリーを鋳型として、PCR法によって抗体遺伝子が増幅される。公知の抗体遺伝子配列をもとにマウス抗体遺伝子増幅用のプライマーがデザインされ得る。これらのプライマーは、イムノグロブリンのサブクラスごとに異なる塩基配列である。したがって、サブクラスは予めIso Stripマウスモノクローナル抗体アイソタイピングキット(ロシュ・ダイアグノスティックス)などの市販キットを用いて決定しておくことが望ましい。 Using the resulting 5'-RACE cDNA library as a template, antibody genes are amplified by PCR. Primers for amplifying mouse antibody genes can be designed based on known antibody gene sequences. These primers have different base sequences for each immunoglobulin subclass. Therefore, it is desirable to determine the subclass in advance using a commercially available kit such as the IsoStrip Mouse Monoclonal Antibody Isotyping Kit (Roche Diagnostics).

 具体的には、たとえばマウスIgGをコードする遺伝子の取得を目的とするときには、重鎖としてγ1、γ2a、γ2b、γ3、軽鎖としてκ鎖とλ鎖をコードする遺伝子の増幅が可能なプライマーが利用され得る。IgGの可変領域遺伝子を増幅するためには、一般に3'側のプライマーには可変領域に近い定常領域に相当する部分にアニールするプライマーが利用される。一方5'側のプライマーには、5' RACE cDNAライブラリー作製キットに付属するプライマーが利用される。 Specifically, for example, when the goal is to obtain a gene encoding mouse IgG, primers can be used that are capable of amplifying genes encoding γ1, γ2a, γ2b, and γ3 heavy chains, and κ and λ light chains. To amplify the IgG variable region gene, the 3' primer generally anneals to a region corresponding to the constant region close to the variable region. Meanwhile, the 5' primer used is one that comes with the 5' RACE cDNA library construction kit.

 こうして増幅されたPCR産物を利用して、重鎖と軽鎖の組み合せからなるイムノグロブリンが再構成され得る。再構成されたイムノグロブリンの、抗原に対する結合活性を指標として、所望の抗体がスクリーニングされ得る。たとえば次のようにしてスクリーニングされ得る;
(1)ハイブリドーマから得られたcDNAによってコードされるV領域を含む抗体を所望の抗原発現細胞に接触させる工程、
(2)該抗原発現細胞と抗体との結合を検出する工程、および
(3)該抗原発現細胞に結合する抗体を選択する工程。
The PCR products thus amplified can be used to reconstitute immunoglobulins consisting of a combination of heavy and light chains. The antigen-binding activity of the reconstituted immunoglobulins can be used as an index to screen for the desired antibody. For example, screening can be performed as follows:
(1) contacting an antibody containing a V region encoded by a cDNA obtained from a hybridoma with a cell expressing a desired antigen;
(2) detecting the binding of the antigen-expressing cells to the antibody; and (3) selecting an antibody that binds to the antigen-expressing cells.

 抗体と該抗原発現細胞との結合を検出する方法は公知である。具体的には、先に述べたFACSなどの手法によって、抗体と該抗原発現細胞との結合が検出され得る。抗体の結合活性を評価するために該抗原発現細胞の固定標本が適宜利用され得る。 Methods for detecting the binding between an antibody and the antigen-expressing cells are known. Specifically, the binding between an antibody and the antigen-expressing cells can be detected by techniques such as the FACS mentioned above. Fixed specimens of the antigen-expressing cells can be used as appropriate to evaluate the binding activity of the antibody.

 結合活性を指標とする抗体のスクリーニング方法として、ファージベクターを利用したパニング法も好適に用いられる。ポリクローナルな抗体発現細胞群より抗体遺伝子を重鎖と軽鎖のサブクラスのライブラリーとして取得した場合には、ファージベクターを利用したスクリーニング方法が有利である。重鎖と軽鎖の可変領域をコードする遺伝子は、適当なリンカー配列で連結することによってシングルチェインFv(scFv)を形成することができる。scFvをコードする遺伝子をファージベクターに挿入することにより、scFvを表面に発現するファージが取得され得る。このファージと所望の抗原との接触の後に、抗原に結合したファージを回収することによって、目的の結合活性を有するscFvをコードするDNAが回収され得る。この操作を必要に応じて繰り返すことにより、所望の結合活性を有するscFvが濃縮され得る。 Panning methods using phage vectors are also suitable as a screening method for antibodies using binding activity as an indicator. When antibody genes are obtained as a library of heavy and light chain subclasses from a polyclonal antibody-expressing cell population, screening methods using phage vectors are advantageous. Genes encoding the variable regions of the heavy and light chains can be linked with an appropriate linker sequence to form single-chain Fvs (scFvs). By inserting a gene encoding an scFv into a phage vector, a phage that expresses scFv on its surface can be obtained. After contacting this phage with the desired antigen, DNA encoding an scFv with the desired binding activity can be recovered by recovering the phage that has bound to the antigen. By repeating this procedure as necessary, scFvs with the desired binding activity can be concentrated.

 目的とする抗体のV領域をコードするcDNAが得られた後に、当該cDNAの両末端に挿入した制限酵素サイトを認識する制限酵素によって該cDNAが消化される。好ましい制限酵素は、抗体遺伝子を構成する塩基配列に出現する頻度が低い塩基配列を認識して消化する。更に1コピーの消化断片をベクターに正しい方向で挿入するためには、付着末端を与える制限酵素の挿入が好ましい。上記のようにして消化された抗体のV領域をコードするcDNAを適当な発現ベクターに挿入することによって、抗体発現ベクターが取得され得る。このとき、抗体定常領域(C領域)をコードする遺伝子と、前記V領域をコードする遺伝子とがインフレームで融合されれば、キメラ抗体が取得される。ここで、キメラ抗体とは、定常領域と可変領域の由来が異なることをいう。したがって、マウス-ヒトなどの異種キメラ抗体に加え、ヒト-ヒト同種キメラ抗体も、本発明におけるキメラ抗体に含まれる。予め定常領域を有する発現ベクターに、前記V領域遺伝子を挿入することによって、キメラ抗体発現ベクターが構築され得る。具体的には、たとえば、所望の抗体定常領域(C領域)をコードするDNAを保持した発現ベクターの5'側に、前記V領域遺伝子を消化する制限酵素の制限酵素認識配列が適宜配置され得る。同じ組み合わせの制限酵素で消化された両者がインフレームで融合されることによって、キメラ抗体発現ベクターが構築される。 After obtaining cDNA encoding the V region of the desired antibody, the cDNA is digested with a restriction enzyme that recognizes restriction enzyme sites inserted at both ends of the cDNA. Preferred restriction enzymes recognize and digest nucleotide sequences that appear infrequently in the nucleotide sequences constituting the antibody gene. Furthermore, to insert one copy of the digested fragment into a vector in the correct orientation, it is preferable to insert a restriction enzyme that generates a cohesive end. An antibody expression vector can be obtained by inserting the cDNA encoding the V region of the antibody digested as described above into an appropriate expression vector. If the gene encoding the antibody constant region (C region) and the gene encoding the V region are fused in frame, a chimeric antibody can be obtained. Here, a chimeric antibody refers to an antibody in which the constant region and variable region are derived from different sources. Therefore, in addition to heterogeneous chimeric antibodies such as mouse-human, human-human allogeneic chimeric antibodies are also included in the chimeric antibodies of the present invention. A chimeric antibody expression vector can be constructed by inserting the V region gene into an expression vector that already contains the constant region. Specifically, for example, a restriction enzyme recognition sequence for a restriction enzyme that digests the V region gene can be appropriately placed on the 5' side of an expression vector carrying DNA encoding the desired antibody constant region (C region). A chimeric antibody expression vector is constructed by fusing the two genes in frame after digestion with the same combination of restriction enzymes.

 モノクローナル抗体の製造には、抗体遺伝子が発現制御領域による制御の下で発現するように発現ベクターに組み込まれる。抗体を発現するための発現制御領域とは、例えば、エンハンサーやプロモーターを含む。また、発現した抗体が細胞外に分泌されるように、適切なシグナル配列がアミノ末端に付加され得る。発現されたポリペプチドから、シグナル配列がそのカルボキシル末端部分から切断され、抗体が細胞外に分泌され得る。次いで、この発現ベクターによって適当な宿主細胞が形質転換されることによって、抗体をコードするDNAを発現する組換え細胞が取得され得る。 To produce a monoclonal antibody, the antibody gene is incorporated into an expression vector so that it is expressed under the control of an expression control region. Expression control regions for antibody expression include, for example, enhancers and promoters. In addition, an appropriate signal sequence can be added to the amino terminus so that the expressed antibody is secreted extracellularly. The signal sequence is cleaved from the carboxyl terminal portion of the expressed polypeptide, allowing the antibody to be secreted extracellularly. Next, by transforming an appropriate host cell with this expression vector, recombinant cells expressing DNA encoding the antibody can be obtained.

 抗体遺伝子の発現のために、抗体重鎖(H鎖)および軽鎖(L鎖)をコードするDNAは、それぞれ別の発現ベクターに組み込まれる。H鎖とL鎖が組み込まれたベクターによって、同じ宿主細胞に同時に形質転換(co-transfect)されることによって、H鎖とL鎖を備えた抗体分子が発現され得る。あるいはH鎖およびL鎖をコードするDNAが単一の発現ベクターに組み込まれることによって宿主細胞が形質転換され得る(国際公開WO 94/11523を参照のこと)。 To express antibody genes, DNA encoding the antibody heavy chain (H chain) and light chain (L chain) are each incorporated into separate expression vectors. By simultaneously transforming (co-transfecting) the same host cell with vectors incorporating the H chain and L chain, antibody molecules equipped with both H chains and L chains can be expressed. Alternatively, host cells can be transformed by incorporating DNA encoding the H chain and L chain into a single expression vector (see International Publication WO 94/11523).

 単離された抗体遺伝子を適当な宿主に導入することによって抗体を作製するための宿主細胞と発現ベクターの多くの組み合わせが公知である。これらの発現系は、いずれも本発明の抗原結合分子に含まれる抗原結合ドメインを単離するのに応用され得る。 Many combinations of host cells and expression vectors are known for producing antibodies by introducing isolated antibody genes into suitable hosts. All of these expression systems can be applied to isolating antigen-binding domains contained in the antigen-binding molecules of the present invention.

 真核細胞が宿主細胞として使用される場合、動物細胞、植物細胞、あるいは真菌細胞が適宜使用され得る。具体的には、動物細胞としては、次のような細胞が例示され得る。
 (1)哺乳類細胞、:CHO、COS、ミエローマ、BHK(baby hamster kidney)、Hela、Veroなど
 (2)両生類細胞:アフリカツメガエル卵母細胞など
 (3)昆虫細胞:sf9、sf21、Tn5など
When eukaryotic cells are used as host cells, animal cells, plant cells, or fungal cells can be used as appropriate. Specific examples of animal cells include the following:
(1) Mammalian cells: CHO, COS, myeloma, BHK (baby hamster kidney), Hela, Vero, etc. (2) Amphibian cells: Xenopus oocytes, etc. (3) Insect cells: sf9, sf21, Tn5, etc.

 あるいは植物細胞としては、ニコティアナ・タバカム(Nicotiana tabacum)などのニコティアナ(Nicotiana)属由来の細胞による抗体遺伝子の発現系が公知である。植物細胞の形質転換には、カルス培養した細胞が適宜利用され得る。 Alternatively, an antibody gene expression system using plant cells derived from the genus Nicotiana, such as Nicotiana tabacum, is known. For transformation of plant cells, callus cultured cells can be used as appropriate.

 更に真菌細胞としては、次のような細胞を利用することができる。
-酵母:サッカロミセス・セレビシエ(Saccharomyces cerevisiae)などのサッカロミセス(Saccharomyces)属、メタノール資化酵母(Pichia pastoris)などのPichia属
-糸状菌:アスペスギルス・ニガー(Aspergillus niger)などのアスペルギルス(Aspergillus)属
Furthermore, the following fungal cells can be used:
- Yeasts: Saccharomyces genus such as Saccharomyces cerevisiae, Pichia genus such as Pichia pastoris - Filamentous fungi: Aspergillus genus such as Aspergillus niger

 また、原核細胞を利用した抗体遺伝子の発現系も公知である。たとえば、細菌細胞を用いる場合、大腸菌(E. coli)、枯草菌などの細菌細胞が適宜利用され得る。これらの細胞中に、目的とする抗体遺伝子を含む発現ベクターが形質転換によって導入される。形質転換された細胞をin vitroで培養することにより、当該形質転換細胞の培養物から所望の抗体が取得され得る。 Antibody gene expression systems using prokaryotic cells are also known. For example, when using bacterial cells, bacterial cells such as Escherichia coli (E. coli) and Bacillus subtilis can be used as appropriate. An expression vector containing the desired antibody gene is introduced into these cells by transformation. By culturing the transformed cells in vitro, the desired antibody can be obtained from the culture of the transformed cells.

 組換え抗体の産生には、上記宿主細胞に加えて、トランスジェニック動物も利用され得る。すなわち所望の抗体をコードする遺伝子が導入された動物から、当該抗体を得ることができる。例えば、抗体遺伝子は、乳汁中に固有に産生されるタンパク質をコードする遺伝子の内部にインフレームで挿入することによって融合遺伝子として構築され得る。乳汁中に分泌されるタンパク質として、たとえば、ヤギβカゼインなどを利用され得る。抗体遺伝子が挿入された融合遺伝子を含むDNA断片はヤギの胚へ注入され、当該注入された胚が雌のヤギへ導入される。胚を受容したヤギから生まれるトランスジェニックヤギ(またはその子孫)が産生する乳汁からは、所望の抗体が乳汁タンパク質との融合タンパク質として取得され得る。また、トランスジェニックヤギから産生される所望の抗体を含む乳汁量を増加させるために、ホルモンがトランスジェニックヤギに対して投与され得る(Bio/Technology (1994), 12 (7), 699-702)。 In addition to the host cells described above, transgenic animals can also be used to produce recombinant antibodies. That is, the antibody can be obtained from an animal into which a gene encoding the desired antibody has been introduced. For example, an antibody gene can be constructed as a fusion gene by inserting it in-frame into a gene encoding a protein specifically produced in milk. Examples of proteins secreted into milk include goat beta-casein. A DNA fragment containing the fusion gene with the antibody gene inserted is injected into a goat embryo, and the injected embryo is then introduced into a female goat. The transgenic goat (or its offspring) born to the goat that received the embryo produces milk from which the desired antibody can be obtained as a fusion protein with a milk protein. Furthermore, hormones can be administered to transgenic goats to increase the amount of milk containing the desired antibody produced by the transgenic goat (Bio/Technology (1994), 12 (7), 699-702).

 本明細書において記載される抗原結合分子がヒトに投与される場合、例えば、当該分子における各種結合ドメインとして、抗体の可変領域を含むドメインを用いる場合は、ヒトに対する異種抗原性を低下させること等を目的として人為的に改変した遺伝子組換え型抗体由来の抗原結合ドメインが適宜採用され得る。遺伝子組換え型抗体には、例えば、ヒト化(Humanized)抗体等が含まれる。これらの改変抗体は、公知の方法を用いて適宜製造される。 When the antigen-binding molecules described herein are administered to humans, for example, when domains containing antibody variable regions are used as various binding domains in the molecules, antigen-binding domains derived from recombinant antibodies that have been artificially modified to reduce heterologous antigenicity to humans can be appropriately used. Recombinant antibodies include, for example, humanized antibodies. These modified antibodies can be appropriately produced using known methods.

 本明細書において記載される抗原結合分子における各種結合ドメインを作製するために用いられる抗体の可変領域は、通常、4つのフレームワーク領域(FR)にはさまれた3つの相補性決定領域(complementarity-determining region ; CDR)で構成されている。CDRは、実質的に、抗体の結合特異性を決定している領域である。CDRのアミノ酸配列は多様性に富む。一方FRを構成するアミノ酸配列は、異なる結合特異性を有する抗体の間でも、高い同一性を示すことが多い。そのため、一般に、CDRの移植によって、ある抗体の結合特異性を、他の抗体に移植することができるとされている。 The antibody variable regions used to create the various binding domains in the antigen-binding molecules described herein are typically composed of three complementarity-determining regions (CDRs) sandwiched between four framework regions (FRs). CDRs are essentially the regions that determine the binding specificity of an antibody. The amino acid sequences of CDRs are highly diverse. On the other hand, the amino acid sequences that make up FRs often show high identity even between antibodies with different binding specificities. Therefore, it is generally believed that the binding specificity of one antibody can be transferred to another antibody by CDR grafting.

 ヒト化抗体は、再構成(reshaped)ヒト抗体とも称される。具体的には、ヒト以外の動物、たとえばマウス抗体のCDRをヒト抗体に移植したヒト化抗体などが公知である。ヒト化抗体を得るための一般的な遺伝子組換え手法も知られている。具体的には、マウスの抗体のCDRをヒトのFRに移植するための方法として、たとえばOverlap Extension PCRが公知である。Overlap Extension PCRにおいては、ヒト抗体のFRを合成するためのプライマーに、移植すべきマウス抗体のCDRをコードする塩基配列が付加される。プライマーは4つのFRのそれぞれについて用意される。一般に、マウスCDRのヒトFRへの移植においては、マウスのFRと同一性の高いヒトFRを選択するのが、CDRの機能の維持において有利であるとされている。すなわち、一般に、移植すべきマウスCDRに隣接しているFRのアミノ酸配列と同一性の高いアミノ酸配列からなるヒトFRを利用するのが好ましい。 Humanized antibodies are also called reshaped human antibodies. Specifically, known humanized antibodies include those in which the CDRs of non-human animals, such as mouse antibodies, are grafted onto human antibodies. Common genetic recombination techniques for obtaining humanized antibodies are also known. Specifically, overlap extension PCR is a well-known method for grafting the CDRs of mouse antibodies onto human FRs. In overlap extension PCR, the base sequence encoding the CDR of the mouse antibody to be grafted is added to a primer used to synthesize the FR of the human antibody. Primers are prepared for each of the four FRs. In general, when grafting mouse CDRs onto human FRs, selecting a human FR that is highly identical to the mouse FR is considered advantageous in terms of maintaining CDR function. In other words, it is generally preferable to use a human FR whose amino acid sequence is highly identical to the amino acid sequence of the FR adjacent to the mouse CDR to be grafted.

 また連結される塩基配列は、互いにインフレームで接続されるようにデザインされる。それぞれのプライマーによってヒトFRが個別に合成される。その結果、各FRにマウスCDRをコードするDNAが付加された産物が得られる。各産物のマウスCDRをコードする塩基配列は、互いにオーバーラップするようにデザインされている。続いて、ヒト抗体遺伝子を鋳型として合成された産物のオーバーラップしたCDR部分を互いにアニールさせて相補鎖合成反応が行われる。この反応によって、ヒトFRがマウスCDRの配列を介して連結される。 The base sequences to be linked are also designed to be connected in-frame to each other. Human FRs are synthesized individually using each primer. As a result, products are obtained in which DNA encoding mouse CDRs is added to each FR. The base sequences encoding the mouse CDRs of each product are designed to overlap with each other. Next, the overlapping CDR portions of the products synthesized using the human antibody gene as a template are annealed to each other to carry out a complementary strand synthesis reaction. Through this reaction, the human FRs are linked via the mouse CDR sequences.

 最終的に3つのCDRと4つのFRが連結されたV領域遺伝子は、その5'末端と3'末端にアニールし適当な制限酵素認識配列を付加されたプライマーによってその全長が増幅される。上記のように得られたDNAとヒト抗体C領域をコードするDNAとをインフレームで融合するように発現ベクター中に挿入することによって、ヒト型抗体発現用ベクターが作成できる。該組込みベクターを宿主に導入して組換え細胞を樹立した後に、該組換え細胞を培養し、該ヒト化抗体をコードするDNAを発現させることによって、該ヒト化抗体が該培養細胞の培養物中に産生される(欧州特許公開EP 239400、国際公開WO1996/002576参照)。 The V-region gene, which ultimately contains the three CDRs and four FRs, is amplified in its entirety using primers that anneal to its 5' and 3' ends and have appropriate restriction enzyme recognition sequences added. A human antibody expression vector can be created by inserting the DNA obtained as described above into an expression vector so that it is fused in-frame with DNA encoding the human antibody C-region. After introducing the integration vector into a host to establish recombinant cells, the recombinant cells are cultured and the DNA encoding the humanized antibody is expressed, resulting in the production of the humanized antibody in the cultured cell culture (see European Patent Publication EP 239400 and International Publication WO 1996/002576).

 上記のように作製したヒト化抗体の抗原への結合活性を定性的又は定量的に測定し、評価することによって、CDRを介して連結されたときに該CDRが良好な抗原結合部位を形成するようなヒト抗体のFRが好適に選択できる。必要に応じ、再構成ヒト抗体のCDRが適切な抗原結合部位を形成するようにFRのアミノ酸残基を置換することもできる。たとえば、マウスCDRのヒトFRへの移植に用いたPCR法を応用して、FRにアミノ酸配列の変異を導入することができる。具体的には、FRにアニーリングするプライマーに部分的な塩基配列の変異を導入することができる。このようなプライマーによって合成されたFRには、塩基配列の変異が導入される。アミノ酸を置換した変異型抗体の抗原への結合活性を上記の方法で測定し評価することによって所望の性質を有する変異FR配列が選択され得る(Sato, K.et al., Cancer Res, 1993, 53, 851-856)。 By qualitatively or quantitatively measuring and evaluating the antigen-binding activity of the humanized antibody prepared as described above, it is possible to select a suitable FR of a human antibody that will form a good antigen-binding site when linked via the CDR. If necessary, amino acid residues in the FR can be substituted so that the CDR of the reshaped human antibody forms a suitable antigen-binding site. For example, the PCR method used to graft mouse CDRs onto human FRs can be applied to introduce amino acid sequence mutations into the FR. Specifically, partial nucleotide sequence mutations can be introduced into primers that anneal to the FR. The nucleotide sequence mutations will be introduced into the FR synthesized using such primers. By measuring and evaluating the antigen-binding activity of mutant antibodies with amino acid substitutions using the above method, mutant FR sequences with desired properties can be selected (Sato, K. et al., Cancer Res, 1993, 53, 851-856).

 また、ヒト抗体遺伝子の全てのレパートリーを有するトランスジェニック動物(国際公開WO1993/012227、WO1992/003918、WO1994/002602、WO1994/025585、WO1996/034096、WO1996/033735参照)を免疫動物とし、DNA免疫により所望のヒト抗体が取得され得る。 In addition, transgenic animals carrying the entire repertoire of human antibody genes (see International Publications WO1993/012227, WO1992/003918, WO1994/002602, WO1994/025585, WO1996/034096, and WO1996/033735) can be used as immunized animals, and the desired human antibodies can be obtained by DNA immunization.

 さらに、ヒト抗体ライブラリーを用いて、パンニングによりヒト抗体を取得する技術も知られている。例えば、ヒト抗体のV領域が一本鎖抗体(scFv)としてファージディスプレイ法によりファージの表面に発現される。抗原に結合するscFvを発現するファージが選択され得る。選択されたファージの遺伝子を解析することにより、抗原に結合するヒト抗体のV領域をコードするDNA配列が決定できる。抗原に結合するscFvのDNA配列を決定した後、当該V領域配列を所望のヒト抗体C領域の配列とインフレームで融合させた後に適当な発現ベクターに挿入することによって発現ベクターが作製され得る。当該発現ベクターを上記に挙げたような好適な発現細胞中に導入し、該ヒト抗体をコードする遺伝子を発現させることにより当該ヒト抗体が取得される。これらの方法は既に公知である(国際公開WO1992/001047、WO1992/020791、WO1993/006213、WO1993/011236、WO1993/019172、WO1995/001438、WO1995/015388参照)。 Furthermore, a technique for obtaining human antibodies by panning using a human antibody library is also known. For example, the V region of a human antibody is expressed on the surface of a phage as a single-chain antibody (scFv) using phage display. Phages expressing scFvs that bind to an antigen can be selected. By analyzing the genes of the selected phage, the DNA sequence encoding the V region of a human antibody that binds to the antigen can be determined. After determining the DNA sequence of the scFv that binds to the antigen, an expression vector can be created by fusing the V region sequence in frame with the sequence of the C region of the desired human antibody and then inserting it into an appropriate expression vector. The expression vector is introduced into a suitable expression cell such as those listed above, and the gene encoding the human antibody is expressed to obtain the human antibody. These methods are already known (see International Publications WO1992/001047, WO1992/020791, WO1993/006213, WO1993/011236, WO1993/019172, WO1995/001438, and WO1995/015388).

 ファージディスプレイ法以外にも、ヒト抗体ライブラリを用いて、パンニングによりヒト抗体を取得する技術として、無細胞翻訳系を使用する技術、細胞またはウイルス表面に抗原結合分子を提示する技術、エマルジョンを使用する技術等が知られている。例えば、無細胞翻訳系を使用する技術としては、終止コドンの除去等によりリボゾームを介してmRNAと翻訳されたタンパク質の複合体を形成させるリボゾームディスプレイ法、ピューロマイシン等の化合物を用いて遺伝子配列と翻訳されたタンパク質を共有結合させるcDNAディスプレイ法、mRNAディスプレイ法や、核酸に対する結合タンパク質を用いて遺伝子と翻訳されたタンパク質の複合体を形成させるCISディスプレイ法等が使用され得る。また、細胞またはウイルス表面に抗原結合分子を提示する技術としては、ファージディスプレイ法以外にも、E. coliディスプレイ法、グラム陽性菌ディスプレイ法、酵母ディスプレイ法、哺乳類細胞ディスプレイ法、ウイルスディスプレイ法等が使用され得る。エマルジョンを使用する技術としては、エマルジョン中に遺伝子及び翻訳関連分子を内包させることによる、インビトロウイルスディスプレイ法等が使用され得る。これらの方法は既に公知である(Nat Biotechnol. 2000 Dec;18(12):1287-92、Nucleic Acids Res. 2006;34(19):e127、Proc Natl Acad Sci U S A. 2004 Mar 2;101(9):2806-10、Proc Natl Acad Sci U S A. 2004 Jun 22;101(25):9193-8、Protein Eng Des Sel. 2008 Apr;21(4):247-55、Proc Natl Acad Sci U S A. 2000 Sep 26;97(20):10701-5、MAbs. 2010 Sep-Oct;2(5):508-18、Methods Mol Biol. 2012;911:183-98)。 In addition to phage display, other known techniques for obtaining human antibodies by panning using a human antibody library include techniques using cell-free translation systems, techniques that display antigen-binding molecules on the surface of cells or viruses, and techniques that use emulsions. For example, techniques that use cell-free translation systems include ribosome display, which forms a complex between mRNA and the translated protein via ribosomes by removing stop codons, cDNA display, which covalently binds a gene sequence to the translated protein using compounds such as puromycin, and mRNA display, and CIS display, which forms a complex between a gene and the translated protein using a nucleic acid-binding protein. Furthermore, in addition to phage display, other techniques for displaying antigen-binding molecules on the surface of cells or viruses include E. coli display, Gram-positive bacteria display, yeast display, mammalian cell display, and virus display. Techniques that use emulsions include in vitro virus display, which involves encapsulating genes and translation-related molecules in an emulsion. These methods are already known (Nat Biotechnol. 2000 Dec;18(12):1287-92, Nucleic Acids Res. 2006;34(19):e127, Proc Natl Acad Sci U S A. 2004 Mar 2;101(9):2806-10, Proc Natl Acad Sci U S A. 2004 Jun 2 2;101(25):9193-8, Protein Eng Des Sel. 2008 Apr;21(4):247-55, Proc Natl Acad Sci U S A. 200 0 Sep 26;97(20):10701-5, MAbs. 2010 Sep-Oct;2(5):508-18, Methods Mol Biol. 2012;911:183-98).

多重特異性抗体の作製方法
 本発明の抗原結合分子の好ましい態様の1つとして、多重特異性抗体を挙げることができる。本発明の多重特異性抗体のFc領域として、Fcγ受容体に対する結合活性が低下しているFc領域を用いる場合、公知の多重特異性抗体を起源とするFc領域も適宜使用される。本発明の多重特異性抗体としては、特に二重特異性抗体が好ましい。
Method for Producing Multispecific Antibodies One preferred embodiment of the antigen-binding molecules of the present invention is a multispecific antibody. When an Fc region with reduced Fcγ receptor-binding activity is used as the Fc region of a multispecific antibody of the present invention, an Fc region derived from a known multispecific antibody can also be used as appropriate. Bispecific antibodies are particularly preferred as multispecific antibodies of the present invention.

 多重特異性抗体の会合化には、抗体H鎖の第二の定常領域(CH2)又はH鎖の第三の定常領域(CH3)の界面に電荷的な反発を導入して目的としないH鎖同士の会合を抑制する技術を適用することができる(WO2006/106905)。 To form multispecific antibodies, a technique can be applied that suppresses undesired association between heavy chains by introducing charge repulsion at the interface of the second constant region (CH2) or third constant region (CH3) of the antibody heavy chain (WO2006/106905).

 CH2又はCH3の界面に電荷的な反発を導入して意図しないH鎖同士の会合を抑制させる技術において、H鎖の他の定常領域の界面で接触するアミノ酸残基としては、例えばCH3領域におけるEUナンバリング356番目の残基、EUナンバリング439番目の残基、EUナンバリング357番目の残基、EUナンバリング370番目の残基、EUナンバリング399番目の残基、EUナンバリング409番目の残基に相対する領域を挙げることができる。 In the technology for suppressing unintended association between H chains by introducing charge repulsion at the CH2 or CH3 interface, examples of amino acid residues that come into contact at the interface of other H chain constant regions include the regions corresponding to residues 356 (EU numbering), 439 (EU numbering), 357 (EU numbering), 370 (EU numbering), 399 (EU numbering), and 409 (EU numbering) in the CH3 region.

 より具体的には、例えば、2種のH鎖CH3領域を含む抗体においては、第1のH鎖CH3領域における以下の(1)~(3)に示すアミノ酸残基の組から選択される1組ないし3組のアミノ酸残基が同種の電荷を有する抗体とすることができる; (1)H鎖CH3領域に含まれるアミノ酸残基であって、EUナンバリング356位および439位のアミノ酸残基、(2)H鎖CH3領域に含まれるアミノ酸残基であって、EUナンバリング357位および370位のアミノ酸残基、(3)H鎖CH3領域に含まれるアミノ酸残基であって、EUナンバリング399位および409位のアミノ酸残基。 More specifically, for example, an antibody containing two types of H chain CH3 regions can be an antibody in which one to three pairs of amino acid residues selected from the following pairs of amino acid residues (1) to (3) in the first H chain CH3 region have the same charge: (1) amino acid residues contained in the H chain CH3 region, which are amino acid residues at positions 356 and 439 (EU numbering), (2) amino acid residues contained in the H chain CH3 region, which are amino acid residues at positions 357 and 370 (EU numbering), and (3) amino acid residues contained in the H chain CH3 region, which are amino acid residues at positions 399 and 409 (EU numbering).

 更に、上記第1のH鎖CH3領域とは異なる第2のH鎖CH3領域における前記(1)~(3)に示すアミノ酸残基の組から選択されるアミノ酸残基の組であって、前記第1のH鎖CH3領域において同種の電荷を有する前記(1)~(3)に示すアミノ酸残基の組に対応する1組ないし3組のアミノ酸残基が、前記第1のH鎖CH3領域における対応するアミノ酸残基とは反対の電荷を有する抗体とすることができる。 Furthermore, the antibody may have a second H chain CH3 region different from the first H chain CH3 region, and the second H chain CH3 region has a set of amino acid residues selected from the sets of amino acid residues shown in (1) to (3), wherein one to three sets of amino acid residues corresponding to the sets of amino acid residues shown in (1) to (3) that have the same charge in the first H chain CH3 region have an opposite charge to the corresponding amino acid residues in the first H chain CH3 region.

 上記(1)~(3)に記載のそれぞれのアミノ酸残基は、会合した際に互いに接近している。当業者であれば、所望のH鎖CH3領域またはH鎖定常領域について、市販のソフトウェアを用いたホモロジーモデリング等により、上記(1)~(3)に記載のアミノ酸残基に対応する部位を見出すことができ、適宜、該部位のアミノ酸残基を改変に供することが可能である。 The amino acid residues described in (1) to (3) above are close to each other when associated. Those skilled in the art can find the positions corresponding to the amino acid residues described in (1) to (3) above for the desired H chain CH3 region or H chain constant region by homology modeling using commercially available software, and can modify the amino acid residues at those positions as appropriate.

 上記抗体において、「電荷を有するアミノ酸残基」は、例えば、以下の(a)または(b)のいずれかの群に含まれるアミノ酸残基から選択されることが好ましい;
(a)グルタミン酸(E)、アスパラギン酸(D)、
(b)リジン(K)、アルギニン(R)、ヒスチジン(H)。
In the above-described antibody, the "charged amino acid residue" is preferably selected from amino acid residues included in either group (a) or (b) below:
(a) glutamic acid (E), aspartic acid (D),
(b) Lysine (K), arginine (R), histidine (H).

 上記抗体において、「同種の電荷を有する」とは、例えば、2つ以上のアミノ酸残基のいずれもが、上記(a)または(b)のいずれか1の群に含まれるアミノ酸残基を有することを意味する。「反対の電荷を有する」とは、例えば、2つ以上のアミノ酸残基のなかの少なくとも1つのアミノ酸残基が、上記(a)または(b)のいずれか1の群に含まれるアミノ酸残基を有する場合に、残りのアミノ酸残基が異なる群に含まれるアミノ酸残基を有することを意味する。 In the above-mentioned antibodies, "having the same charge" means, for example, that two or more amino acid residues all have amino acid residues included in either group (a) or (b) above. "Having opposite charges" means, for example, that when at least one amino acid residue among two or more amino acid residues has an amino acid residue included in either group (a) or (b) above, the remaining amino acid residues have amino acid residues included in a different group.

 好ましい態様において上記抗体は、第1のH鎖CH3領域と第2のH鎖CH3領域がジスルフィド結合により架橋されていてもよい。  In a preferred embodiment, the antibody may have the first H chain CH3 region and the second H chain CH3 region cross-linked by a disulfide bond.

 本発明において改変に供するアミノ酸残基としては、上述した抗体の可変領域または抗体の定常領域のアミノ酸残基に限られない。当業者であれば、ポリペプチド変異体または異種多量体について、市販のソフトウェアを用いたホモロジーモデリング等により、界面を形成するアミノ酸残基を見出すことができ、会合を制御するように、該部位のアミノ酸残基を改変に供することが可能である。 The amino acid residues that can be modified in the present invention are not limited to those in the antibody variable region or constant region described above. Those skilled in the art can identify amino acid residues that form an interface between polypeptide mutants or heteromultimers by homology modeling using commercially available software, and can modify the amino acid residues at those sites to control association.

 また、本発明の多重特異性抗体の会合化には更に他の公知技術を用いることもできる。抗体の一方のH鎖の可変領域に存在するアミノ酸側鎖をより大きい側鎖(knob; 突起)に置換し、もう一方のH鎖の相対する可変領域に存在するアミノ酸側鎖をより小さい側鎖(hole; 空隙)に置換することによって、突起が空隙に配置され得るようにすることで効率的にFc領域を有する異なるアミノ酸を有するポリペプチド同士の会合化を起こすことができる(WO1996/027011、Ridgway JB et al., Protein Engineering (1996) 9, 617-621、Merchant AM et al. Nature Biotechnology (1998) 16, 677-681、US20130336973)。 Furthermore, other known techniques can also be used to aggregate the multispecific antibodies of the present invention. By substituting a larger side chain (knob) for an amino acid side chain in the variable region of one of the antibody's heavy chains and a smaller side chain (hole) for an amino acid side chain in the opposing variable region of the other heavy chain, the knob can be positioned in the hole, thereby efficiently facilitating aggregation between polypeptides with different amino acids that have Fc domains (WO1996/027011, Ridgway JB et al., Protein Engineering (1996) 9, 617-621, Merchant AM et al., Nature Biotechnology (1998) 16, 677-681, US20130336973).

 これに加えて、本発明の多重特異性抗体の形成には更に他の公知技術を用いることもできる。抗体の一方のH鎖のCH3の一部をその部分に対応するIgA由来の配列にし、もう一方のH鎖のCH3の相補的な部分にその部分に対応するIgA由来の配列を導入したstrand-exchange engineered domain CH3を用いることで、異なる配列を有するポリペプチドの会合化をCH3の相補的な会合化によって効率的に引き起こすことができる (Protein Engineering Design & Selection, 23; 195-202, 2010)。この公知技術を使っても効率的に目的の多重特異性抗体の形成させることができる。 In addition, other known techniques can also be used to form the multispecific antibodies of the present invention. By using a strand-exchange engineered domain CH3, in which part of the CH3 of one antibody H chain is replaced with a corresponding IgA-derived sequence, and the complementary part of the CH3 of the other H chain is replaced with a corresponding IgA-derived sequence, it is possible to efficiently induce association of polypeptides with different sequences through complementary association of CH3 (Protein Engineering Design & Selection, 23; 195-202, 2010). This known technique can also be used to efficiently form the desired multispecific antibodies.

 他にも多重特異性抗体の形成には、WO2011/028952やWO2014/018572やNat Biotechnol. 2014 Feb;32(2):191-8.に記載の抗体のCH1とCLの会合化、VH、VLの会合化を利用した抗体作製技術、WO2008/119353やWO2011/131746に記載の別々に調製したモノクローナル抗体同士を使用して二重特異性抗体を作製する技術(Fab Arm Exchange)、WO2012/058768やWO2013/063702に記載の抗体重鎖のCH3間の会合を制御する技術、WO2012/023053に記載の二種類の軽鎖と一種類の重鎖とから構成される二重特異性抗体を作製する技術、Christophら(Nature Biotechnology Vol. 31, p 753-758 (2013))に記載の1本のH鎖と1本のL鎖からなる抗体の片鎖をそれぞれ発現する2つのバクテリア細胞株を利用した二重特異性抗体を作製する技術等を用いることもできる。 Other methods for forming multispecific antibodies include antibody production techniques that utilize the association of antibody CH1 and CL, and VH and VL, as described in WO2011/028952, WO2014/018572, and Nat Biotechnol. 2014 Feb;32(2):191-8.; techniques for producing bispecific antibodies using separately prepared monoclonal antibodies (Fab Arm Exchange), as described in WO2008/119353 and WO2011/131746; and techniques described in WO2012/058768 and Other techniques that can be used include the technology described in WO2013/063702 for controlling the association between CH3s of antibody heavy chains, the technology described in WO2012/023053 for producing bispecific antibodies consisting of two types of light chains and one type of heavy chain, and the technology described in Christoph et al. (Nature Biotechnology Vol. 31, pp. 753-758 (2013)) for producing bispecific antibodies using two bacterial cell lines that each express one half of an antibody chain consisting of one H chain and one L chain.

 多重特異性抗体の形成の一態様としては、上述したように、二種類のモノクローナル抗体を還元剤存在下で混合し、コアヒンジのdisulfide結合を開裂させたのちに、再会合させてヘテロ二量化した二重特異性抗体を得る方法が挙げられるが(FAE)、CH3領域の相互作用界面に静電相互作用(WO2006/106905)を導入することにより、再会合時にさらに効率的にヘテロ二量化を誘起することができる(WO2015/046467)。天然型IgGを用いたFAEでは再会合がランダムに起こるため理論上50%の効率でしか二重特異性抗体が得られないが、当該方法では高収率で二重特異性抗体を製造することができる。 As mentioned above, one embodiment of multispecific antibody formation involves mixing two types of monoclonal antibodies in the presence of a reducing agent, cleaving the disulfide bonds in the core hinge, and then reassociating them to obtain a heterodimerized bispecific antibody (FAE). However, by introducing electrostatic interactions (WO2006/106905) into the interaction interface of the CH3 region, heterodimerization can be induced more efficiently during reassociation (WO2015/046467). With FAE using native IgG, reassociation occurs randomly, so theoretically bispecific antibodies can only be obtained with a 50% efficiency, but this method allows for the production of bispecific antibodies with a high yield.

 また、効率的に目的の多重特異性抗体を形成させることができない場合であっても、産生された抗体の中から目的の多重特異性抗体を分離、精製することによっても、本発明の多重特異性抗体を得ることが可能である。例えば、2種類のH鎖の可変領域にアミノ酸置換を導入し等電点の差を付与することで、2種類のホモ体と目的のヘテロ抗体をイオン交換クロマトグラフィーで精製可能にする方法が報告されている(WO2007114325)。また、ヘテロ体を精製する方法として、これまでに、プロテインAに結合するマウスIgG2aのH鎖と、プロテインAに結合しないラットIgG2bのH鎖からなるヘテロ二量化抗体をプロテインAを用いて精製する方法が報告されている(WO98050431、WO95033844)。更に、IgGとProteinAの結合部位であるEUナンバリング435番目および436番目のアミノ酸残基を、Tyr、HisなどのProteinAへの結合力の異なるアミノ酸に置換したH鎖を用いることで、各H鎖とProtein Aとの相互作用を変化させ、Protein Aカラムを用いることで、ヘテロ二量化抗体のみを効率的に精製することもできる。 Even if the desired multispecific antibody cannot be efficiently formed, it is still possible to obtain the multispecific antibody of the present invention by isolating and purifying it from the produced antibodies. For example, a method has been reported in which amino acid substitutions are introduced into the variable regions of two types of H chains to impart a difference in isoelectric point, making it possible to purify two types of homoantibodies and the desired heteroantibody by ion exchange chromatography (WO2007114325). Furthermore, a method for purifying heteroantibodies has been reported in which a heterodimerized antibody consisting of a mouse IgG2a H chain that binds to Protein A and a rat IgG2b H chain that does not bind to Protein A is purified using Protein A (WO98050431, WO95033844). Furthermore, by using H chains in which the amino acid residues at EU numbering positions 435 and 436, which are the binding sites between IgG and Protein A, have been replaced with amino acids such as Tyr and His that have different binding strengths to Protein A, the interaction between each H chain and Protein A can be changed, and by using a Protein A column, it is possible to efficiently purify only the heterodimerized antibody.

 また、異なる複数のH鎖に結合能を与え得る共通のL鎖を取得し、多重特異性抗体の共通L鎖として用いてもよい。このような共通L鎖と異なる複数のH鎖遺伝子を細胞に導入することによってIgGを発現させることで効率の良い多重特異性IgGの発現が可能となる(Nature Biotechnology (1998) 16, 677-681)。共通H鎖を選択する際に、任意の異なるH鎖に対応し高い結合能を示す共通L鎖を選択する方法も利用することができる(WO2004/065611)。 Alternatively, a common L chain capable of conferring binding ability to multiple different H chains can be obtained and used as the common L chain for a multispecific antibody. By expressing IgG by introducing such a common L chain and multiple different H chain genes into cells, it becomes possible to efficiently express multispecific IgG (Nature Biotechnology (1998) 16, 677-681). When selecting a common H chain, a method can also be used to select a common L chain that corresponds to any different H chain and exhibits high binding ability (WO2004/065611).

 また、本発明のFc領域として、Fc領域のC末端のヘテロジェニティーが改善されたFc領域が適宜使用され得る。より具体的には、IgG1、IgG2、IgG3又はIgG4を起源とするFc領域を構成する二つのポリペプチドのアミノ酸配列のうちEUナンバリングに従って特定される446位のグリシン、及び447位のリジンが欠失したFc領域が提供される。 Furthermore, as the Fc region of the present invention, an Fc region in which C-terminal heterogeneity has been reduced can be used as appropriate. More specifically, an Fc region in which glycine at position 446 and lysine at position 447, as specified according to EU numbering, in the amino acid sequences of two polypeptides that constitute an Fc region originating from IgG1, IgG2, IgG3, or IgG4, are deleted, is provided.

 これらの技術を複数、例えば2つ以上組合せて用いることもできる。また、これらの技術は、会合させたい2つのH鎖に適宜別々に適用させることもできる。さらに、これらの技術は、上述のFcγ受容体に対する結合活性が低下しているFc領域に組み合わせて用いることもできる。なお、本発明の抗原結合分子は、上記改変が加えられたものをベースにして、同一のアミノ酸配列を有する抗原結合分子を別途作製したものであってもよい。 Several of these techniques, for example, two or more in combination, can also be used. These techniques can also be applied separately, as appropriate, to the two H chains to be associated. Furthermore, these techniques can also be used in combination with the above-mentioned Fc region with reduced binding activity to Fcγ receptors. The antigen-binding molecule of the present invention may also be an antigen-binding molecule having the same amino acid sequence as the antigen-binding molecule having the above-mentioned modifications added thereto, which has been separately prepared.

 本発明に係る多重特異性抗原結合分子は、(1)前記の抗体H鎖可変領域及びL鎖可変領域を含む、CD3に対する結合活性を有する第一の抗原結合ドメイン、(2)第一の抗原結合ドメインとは異なるアミノ酸配列を含む第二の抗原結合ドメイン、ならびに任意で(3)Fcγ受容体に対する結合活性が低下しているFc領域を含むドメインを含むものであればよく、その構造は限定されない。
 本発明において、上記の各ドメインはペプチド結合で直接連結することができる。例えば、(1)及び(2)の抗原結合ドメインとしてF(ab')2を用い、(3)Fcγ受容体に対する結合活性が低下しているFc領域を含むドメインとしてこれらのFc領域を用いた場合に、(1)及び(2)に記載された抗原結合ドメインと(3)に記載されたFc領域を含むドメインとをペプチド結合で連結したときは、連結されたポリペプチドは抗体の構造を形成する。そのような抗体を作製するためには前述のハイブリドーマの培養液から精製する他、当該抗体を構成するポリペプチドをコードするポリヌクレオチドが安定に保持された所望の宿主細胞の培養液から当該抗体を精製することもできる。
The structure of a multispecific antigen-binding molecule of the present invention is not limited, as long as it comprises: (1) a first antigen-binding domain comprising the antibody H-chain variable region and L-chain variable region and having CD3-binding activity; (2) a second antigen-binding domain comprising an amino acid sequence different from that of the first antigen-binding domain; and, optionally, (3) a domain comprising an Fc region with reduced Fcγ receptor-binding activity.
In the present invention, the above-mentioned domains can be directly linked via peptide bonds. For example, when F(ab') 2 is used as the antigen-binding domain in (1) and (2), and these Fc regions are used as the domain containing an Fc region with reduced Fcγ receptor-binding activity in (3), when the antigen-binding domains described in (1) and (2) and the domain containing an Fc region in (3) are linked via peptide bonds, the linked polypeptide forms an antibody structure. To produce such an antibody, the antibody can be purified from the culture medium of the above-mentioned hybridoma, or from the culture medium of a desired host cell in which a polynucleotide encoding the polypeptide constituting the antibody is stably maintained.

 本発明の好ましいCD3結合活性を有する抗体可変領域としては、CD3εに対する結合活性を有する抗体可変領域が挙げられる。そのような抗体可変領域としては、例えば、後掲の表5に記載の抗体番号2, 14, 25, 29, 30~32, 34の重鎖可変領域、又は重鎖CDR1、CDR2及びCDR3のアミノ酸配列を有する抗体H鎖可変領域、ならびに、表5に記載の抗体番号2, 14, 25, 29, 30~32, 34の軽鎖可変領域、又は軽鎖CDR1、CDR2及びCDR3のアミノ酸配列を有する抗体L鎖可変領域、或いは、当該可変領域と機能的に同等の抗体H鎖可変領域及びL鎖可変領域が挙げられる。 Preferred antibody variable regions of the present invention that have CD3-binding activity include antibody variable regions that have binding activity to CD3ε. Examples of such antibody variable regions include the heavy chain variable regions of antibody Nos. 2, 14, 25, 29, 30-32, and 34 listed in Table 5 below, or antibody H chain variable regions having the amino acid sequences of heavy chain CDR1, CDR2, and CDR3; and the light chain variable regions of antibody Nos. 2, 14, 25, 29, 30-32, and 34 listed in Table 5, or antibody L chain variable regions having the amino acid sequences of light chain CDR1, CDR2, and CDR3; or antibody H chain variable regions and L chain variable regions functionally equivalent to these variable regions.

 ここで、本発明において「機能的に同等」とは、所定の条件下での抗原に対する結合親和性が同等である、或いは、多重特異性抗原結合分子として用いられた場合に、標的抗原を発現している細胞又は当該細胞を含む組織に対する細胞傷害活性が同等であることを意味する。結合親和性および細胞傷害活性は、本明細書の記載に基づいて測定することができる。細胞傷害活性を測定する細胞は、標的抗原を発現している所望の細胞又は当該細胞を含む所望の組織を用いてよいが、例えば標的抗原を発現するヒトがん細胞株を用いることができる。また抗体定常領域においては、Fcγ受容体に対する結合活性の低下が同等であることでもよい。 In the present invention, "functionally equivalent" means that the binding affinity to an antigen under specified conditions is equivalent, or that when used as a multispecific antigen-binding molecule, the cytotoxic activity against cells expressing the target antigen or tissues containing said cells is equivalent. Binding affinity and cytotoxic activity can be measured based on the descriptions herein. The cells used to measure cytotoxic activity may be desired cells expressing the target antigen or desired tissues containing said cells, but for example, human cancer cell lines expressing the target antigen can be used. Furthermore, with respect to antibody constant regions, the reduction in binding activity to Fcγ receptors may also be equivalent.

 例えば、本願明細書に記載された抗体H鎖可変領域(元となるH鎖可変領域)と機能的に同等な抗体H鎖可変領域は、その元となるH鎖の対として本願明細書に記載されている抗体L鎖可変領域と組み合わせた場合、結合親和性が同等であることを意味する、或いは、多重特異性抗原結合分子として用いられた場合に、標的抗原を発現している細胞又は当該細胞を含む組織に対する細胞傷害活性が同等であることを意味する。また本願明細書に記載された抗体L鎖可変領域(元となるL鎖可変領域)と機能的に同等な抗体L鎖可変領域は、その元となるL鎖の対として本願明細書に記載されている抗体H鎖可変領域と組み合わせた場合、結合親和性が同等である、或いは、多重特異性抗原結合分子として用いられた場合に、標的抗原を発現している細胞又は当該細胞を含む組織に対する細胞傷害活性が同等であることを意味する。 For example, an antibody H-chain variable region functionally equivalent to an antibody H-chain variable region described herein (the original H-chain variable region) means that when combined with the antibody L-chain variable region described herein as the pair of the original H-chain, it has equivalent binding affinity, or when used as a multispecific antigen-binding molecule, it has equivalent cytotoxic activity against cells expressing the target antigen or tissues containing said cells. Furthermore, an antibody L-chain variable region functionally equivalent to an antibody L-chain variable region described herein (the original L-chain variable region) means that when combined with the antibody H-chain variable region described herein as the pair of the original L-chain, it has equivalent binding affinity, or when used as a multispecific antigen-binding molecule, it has equivalent cytotoxic activity against cells expressing the target antigen or tissues containing said cells.

 また、「同等」とは、必ずしも同程度の活性である必要がなく、活性が増強されていてもよく、したがって「同等又はそれ以上」と表現することもできる。具体的には、抗原に対する結合親和性の場合は、「同等」とは、対照となる抗体可変領域の結合親和性(親KD値)と比較した値(KD値/親KD値)が2.0以下の場合を挙げることができる。KD値/親KD値の値は、好ましくは1.5以下であり、より好ましくは1.3以下、1.2以下、1.1以下、1.0以下、0.9以下、0.8以下、0.7以下、0.6以下、または0.5以下である。下限に制限はないが、例えば10-1、10-2、10-3、10-4、10-5、または10-6 であってよい。具体的には、本発明においてKD値/親KD値の値は、10-6~2.0が好ましく、より好ましくは10-3~1.5、より好ましくは10-1~1.3、より好ましくは0.8~1.2、より好ましくは0.9~1.1、最も好ましくは1.0である。細胞傷害活性の場合は、「同等」とは、対照となる多重特異性抗原結合分子の細胞増殖抑制率(親細胞増殖抑制率)と比較した値(細胞増殖抑制率/親細胞増殖抑制率)が0.7以上の場合を挙げることができる。添加する多重特異性抗原結合分子の濃度は適宜決定されるが、好ましくは、例えば0.01nM、0.05nM、0.1nM、0.5nM、または1nM、好ましくは0.05nMまたは0.1nMで測定する。細胞増殖抑制率/親細胞増殖抑制率の値は、好ましくは0.8以上であり、より好ましくは0.9以上、1.0以上、1.2以上、1.5以上、2以上、3以上、5以上、10以上、または20以上である。上限に制限はないが、例えば10、102、103、104、105、または106 であってよい。 Furthermore, "equivalent" does not necessarily mean the same level of activity; it may also mean that the activity is enhanced, and therefore can also be expressed as "equivalent or greater." Specifically, in the case of binding affinity to an antigen, "equivalent" refers to a value (KD value/parent KD value) of 2.0 or less compared to the binding affinity (parent KD value) of a control antibody variable region. The KD value/parent KD value is preferably 1.5 or less, more preferably 1.3 or less, 1.2 or less, 1.1 or less, 1.0 or less, 0.9 or less, 0.8 or less, 0.7 or less, 0.6 or less, or 0.5 or less. There is no lower limit, but it may be, for example, 10-1 , 10-2 , 10-3 , 10-4 , 10-5 , or 10-6 . Specifically, in the present invention, the KD value/parent KD value is preferably 10 -6 to 2.0, more preferably 10 -3 to 1.5, more preferably 10 -1 to 1.3, more preferably 0.8 to 1.2, more preferably 0.9 to 1.1, and most preferably 1.0. In the case of cytotoxic activity, "equivalent" refers to a value (cell growth inhibition rate/parent cell growth inhibition rate) of 0.7 or higher compared to the cell growth inhibition rate of a control multispecific antigen-binding molecule (parent cell growth inhibition rate). The concentration of the added multispecific antigen-binding molecule is determined appropriately, but is preferably measured at, for example, 0.01 nM, 0.05 nM, 0.1 nM, 0.5 nM, or 1 nM, preferably 0.05 nM or 0.1 nM. The value of cell proliferation inhibition rate/parent cell proliferation inhibition rate is preferably 0.8 or more, more preferably 0.9 or more, 1.0 or more, 1.2 or more, 1.5 or more, 2 or more, 3 or more, 5 or more, 10 or more, or 20 or more. There is no upper limit, but it may be, for example, 10, 102 , 103 , 104 , 105 , or 106 .

 また細胞傷害活性の場合は、元の多重特異性抗原結合分子の細胞に対する50%増殖抑制濃度(親細胞50%増殖抑制濃度)と比較した値(細胞50%増殖抑制濃度/親細胞50%増殖抑制濃度)が1.5以下の場合を挙げることができる。50%増殖抑制濃度とは、多重特異性抗原結合分子を添加しない場合に比べ、細胞増殖率を半減させるのに必要な多重特異性抗原結合分子の濃度のことである。「細胞50%増殖抑制濃度/親細胞50%増殖抑制濃度」の値は、好ましくは1.3以下であり、より好ましくは1.2以下、1.1以下、1.0以下、0.9以下、0.8以下、0.7以下、0.6以下、または0.5以下である。下限に制限はないが、例えば10-1、10-2、10-3、10-4、10-5、または10-6 であってよい。具体的には 10-6~1.5x10-0 が好ましく、より好ましくは10-6~10-1、より好ましくは10-6~10-2、より好ましくは10-6~10-3 である。 In the case of cytotoxic activity, examples of such activity include a value (concentration for 50% cell growth inhibition/concentration for 50% parent cell growth inhibition) of 1.5 or less, relative to the concentration of the original multispecific antigen-binding molecule that inhibits 50% cell growth (concentration for 50% parent cell growth inhibition). The concentration of multispecific antigen-binding molecule required to reduce the cell growth rate by half compared to when the multispecific antigen-binding molecule is not added. The value of "concentration for 50% cell growth inhibition/concentration for 50% parent cell growth inhibition" is preferably 1.3 or less, and more preferably 1.2 or less, 1.1 or less, 1.0 or less, 0.9 or less, 0.8 or less, 0.7 or less, 0.6 or less, or 0.5 or less. There is no lower limit, but it may be, for example, 10-1 , 10-2 , 10-3 , 10-4 , 10-5 , or 10-6 . Specifically, it is preferably 10 −6 to 1.5×10 −0 , more preferably 10 −6 to 10 −1 , more preferably 10 −6 to 10 −2 , and even more preferably 10 −6 to 10 −3 .

 細胞傷害活性は、実施例4に記載されているようにin vitroでのTDCC活性により評価することができ、その場合、「同等」とは、対照となる多重特異性抗原結合分子のTDCC活性と比較した値(TDCC活性/親TDCC活性)が0.7以上の場合を挙げることができる。TDCC活性/親TDCC活性の値は、好ましくは0.8以上であり、より好ましくは0.9以上、1.0以上、1.2以上、1.5以上、2以上、3以上、5以上、10以上、または20以上である。 Cytotoxic activity can be assessed by in vitro TDCC activity as described in Example 4, and in this case, "equivalent" refers to a value (TDCC activity/parent TDCC activity) of 0.7 or greater compared to the TDCC activity of a control multispecific antigen-binding molecule. The TDCC activity/parent TDCC activity value is preferably 0.8 or greater, and more preferably 0.9 or greater, 1.0 or greater, 1.2 or greater, 1.5 or greater, 2 or greater, 3 or greater, 5 or greater, 10 or greater, or 20 or greater.

 また、標的抗原に対する結合活性を有する第二の抗原結合ドメインについては、標的抗原に対するKD値は、例えば 5x10-9 M 以下であってよく、好ましくは 4x10-9M以下、例えば 3x10-9M以下、2x10-9M以下、1x10-9M以下、8x10-10M以下、5x10-10M以下、4x10-10M以下、3x10-10M以下、2x10-10M以下、1x10-10M以下、8x10-11M以下、5x10-11M以下、4x10-11M以下、3x10-11M以下、2x10-11M以下、1x10-11M以下、8x10-12M以下、5x10-12M以下、4x10-12M以下、3x10-12M以下、2x10-12M以下、1x10-12M以下、8x10-13M以下、5x10-13M以下、4x10-13M以下、3x10-13M以下、2x10-13M以下、または1x10-13M以下であってよい。 Furthermore, the KD value of a second antigen-binding domain having binding activity to a target antigen may be, for example, 5x10 -9 M or less, preferably 4x10 -9 M or less, for example, 3x10 -9 M or less, 2x10 -9 M or less, 1x10 -9 M or less, 8x10 -10 M or less, 5x10 -10 M or less, 4x10 -10 M or less, 3x10 -10 M or less, 2x10 -10 M or less, 1x10 -10 M or less, 8x10 -11 M or less, 5x10 -11 M or less, 4x10 -11 M or less, 3x10 -11 M or less, 2x10 -11 M or less, 1x10 -11 M or less, 8x10 -12 M or less, 5x10 -12 M or less, 4x10 -12 M or less, 3x10 -12 M or less, 2x10 -12 M or less, 1x10 -12 M or less, 8x10 -13 M or less, 5x10 -13 M or less, 4x10 -13 M or less, 3x10 -13 M or less, 2x10 -13 M or less, or 1x10 -13 M or less.

 また、CD3に対する結合活性を有する抗体可変領域を含む抗原結合ドメインについては、CD3、例えばヒトCD3、より具体的には例えばヒトCD3ε鎖に対するKD値は、例えば 5x10-7 M 以下であってよく、好ましくは2x10-7 M 以下、例えば1.5x10-7 M 以下、1.4x10-7M以下、1.3x10-7M以下、1.2x10-7M以下、1x10-7M以下、3x10-8M以下、2x10-8M以下、1x10-8M以下、8x10-9M以下、5x10-9M以下、4x10-9M以下、3x10-9M以下、2x10-9M以下、1x10-9M以下、8x10-10M以下、5x10-10M以下、4x10-10M以下、3x10-10M以下、2x10-10M以下、1x10-10M以下、8x10-11M以下、5x10-11M以下、4x10-11M以下、3x10-11M以下、2x10-11M以下、1x10-11M以下、8x10-12M以下、5x10-12M以下、4x10-12M以下、3x10-12M以下、2x10-12M以下、または1x10-12M以下である。 Furthermore, with regard to an antigen-binding domain comprising an antibody variable region that has binding activity to CD3, the KD value for CD3, for example, human CD3, more specifically, for example, human CD3ε chain, may be, for example, 5x10 −7 M or less, preferably 2x10 −7 M or less, for example, 1.5x10 −7 M or less, 1.4x10 −7 M or less, 1.3x10 −7 M or less, 1.2x10 −7 M or less, 1x10 −7 M or less, 3x10 −8 M or less, 2x10 −8 M or less, 1x10 −8 M or less, 8x10 −9 M or less, 5x10 −9 M or less, 4x10 −9 M or less, 3x10 −9 M or less, 2x10 −9 M or less, 1x10 −9 M or less, 8x10 −10 M or less, 5x10 −10 M or less, 4x10 −10 M or less, 3x10 −10 M or less, 2x10 −10 M or less, 1x10 -10 M or less, 8x10 -11 M or less, 5x10 -11 M or less, 4x10 -11 M or less, 3x10 -11 M or less, 2x10 -11 M or less, 1x10 -11 M or less, 8x10 -12 M or less, 5x10 -12 M or less, 4x10 -12 M or less, 3x10 -12 M or less, 2x10 -12 M or less, or 1x10 -12 M or less.

 本発明の多重特異性抗原結合分子は、好ましくは、標的抗原およびヒトCD3(例えばヒトCD3ε鎖)に対するKD値は、それぞれ5x10-9 M以下および 5x10-7 M以下であり、より好ましくは、それぞれ1x10-9 M以下および 5x10-8 M以下である。 The multispecific antigen-binding molecules of the present invention preferably have KD values for the target antigen and human CD3 (e.g., human CD3ε chain) of 5x10-9 M or less and 5x10-7 M or less, respectively, and more preferably 1x10-9 M or less and 5x10-8 M or less, respectively.

 本発明において「機能的に同等」の抗体可変領域とは、上述の条件を満たす抗体H鎖可変領域及び/又は抗体L鎖可変領域であれば特に限定されない。そのような抗体可変領域として、例えば、表5に記載の抗体番号2, 14, 25, 29, 30~32, 34の可変領域のアミノ酸配列に1または複数のアミノ酸(例えば1、2、3、4、5または10アミノ酸)が置換、欠失、付加及び/又は挿入されていてもよい。アミノ酸配列において、1または複数のアミノ酸が置換、欠失、付加及び/又は挿入するための、当業者によく知られた方法としては、タンパク質に変異を導入する方法が知られている。例えば、当業者であれば、部位特異的変異誘発法(Hashimoto-Gotoh, T, Mizuno, T, Ogasahara, Y, and Nakagawa, M. (1995) An oligodeoxyribonucleotide-directed dual amber method for site-directed mutagenesis. Gene 152, 271-275、Zoller, MJ, and Smith, M.(1983) Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors.Methods Enzymol. 100, 468-500、Kramer,W, Drutsa,V, Jansen,HW, Kramer,B, Pflugfelder,M, and Fritz,HJ(1984) The gapped duplex DNA approach to oligonucleotide-directed mutation construction. Nucleic Acids Res. 12, 9441-9456、Kramer W, and Fritz HJ(1987) Oligonucleotide-directed construction of mutations via gapped duplex DNA Methods. Enzymol. 154, 350-367、Kunkel,TA(1985) Rapid and efficient site-specific mutagenesis without phenotypic selection.Proc Natl Acad Sci U S A. 82, 488-492)などを用いてアミノ酸配列に適宜変異を導入することにより、上述の機能を有する抗体可変領域と機能的に同等な可変領域を調製することができる。 In the present invention, a "functionally equivalent" antibody variable region is not particularly limited as long as it is an antibody heavy chain variable region and/or antibody light chain variable region that meets the above-mentioned conditions. Such antibody variable regions may, for example, have one or more amino acids (e.g., 1, 2, 3, 4, 5, or 10 amino acids) substituted, deleted, added, and/or inserted into the amino acid sequence of the variable regions of antibodies Nos. 2, 14, 25, 29, 30-32, and 34 listed in Table 5. Methods for substituting, deleting, adding, and/or inserting one or more amino acids into an amino acid sequence that are well known to those skilled in the art include methods for introducing mutations into proteins. For example, those skilled in the art are familiar with site-directed mutagenesis methods (Hashimoto-Gotoh, T, Mizuno, T, Ogasahara, Y, and Nakagawa, M. (1995) An oligodeoxyribonucleotide-directed dual amber method for site-directed mutagenesis. Gene 152, 271-275; Zoller, MJ, and S mith, M. (1983) Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors.Methods Enzymo l. 100, 468-500, Kramer,W, Drutsa,V, Jansen,HW, Kramer,B, Pflugfelder,M, and Fritz,HJ(1984) The gapped duple x DNA approach to oligonucleotide-directed mutation construction. Nucleic Acids Res. 12, 9441-9456; Kramer W, and Fritz HJ (1987) Oligonucleotide-directed construction of mutations via gapped duplex DNA Methods. Enzymol. 154, 350-367; Kunkel, TA (1985) Rapid and efficient site-specific mutagenesis without phenotypic selection. Proc Natl Acad Sci U S A. 82, 488-492), etc., can be used to appropriately introduce mutations into the amino acid sequence, thereby preparing variable regions functionally equivalent to the antibody variable regions having the above-mentioned functions.

 アミノ酸残基を改変する場合には、アミノ酸側鎖の性質が保存されている別のアミノ酸に変異されることが望ましい。例えばアミノ酸側鎖の性質としては、疎水性アミノ酸(A、I、L、M、F、P、W、Y、V)、親水性アミノ酸(R、D、N、C、E、Q、G、H、K、S、T)、脂肪族側鎖を有するアミノ酸(G、A、V、L、I、P)、水酸基含有側鎖を有するアミノ酸(S、T、Y)、硫黄原子含有側鎖を有するアミノ酸(C、M)、カルボン酸及びアミド含有側鎖を有するアミノ酸(D、N、E、Q)、塩基含有側鎖を有するアミノ酸(R、K、H)、及び、芳香族含有側鎖を有するアミノ酸(H、F、Y、W)を挙げることができる(括弧内はいずれもアミノ酸の一文字標記を表す)。これらの各グループ内のアミノ酸の置換を保存的置換と称す。あるアミノ酸配列に対する1又は複数個のアミノ酸残基の欠失、付加及び/又は他のアミノ酸による置換により修飾されたアミノ酸配列を有するポリペプチドがその生物学的活性を維持することはすでに知られている(Mark, D. F. et al., Proc.Natl.Acad.Sci.USA (1984)81:5662-6; Zoller, M. J. and Smith, M., Nucleic Acids Res.(1982)10:6487-500; Wang, A. et al., Science(1984)224:1431-3; Dalbadie-McFarland, G. et al., Proc.Natl.Acad.Sci.USA (1982)79:6409-13)。このようなアミノ酸改変を含む本発明の可変領域は、改変前の可変領域のCDR配列、FR配列又は可変領域全体のアミノ酸配列と少なくとも70%、より好ましくは少なくとも75%、より好ましくは少なくとも80%、さらに好ましくは少なくとも85%、さらにより好ましくは少なくとも90%、そして、最も好ましくは少なくとも95%のアミノ酸配列の同一性を有する。本明細書において配列の同一性は、配列同一性が最大となるように必要に応じ配列を整列化し、適宜ギャップを導入した後、元となったH鎖可変領域又はL鎖可変領域のアミノ酸配列の残基と同一の残基の割合として定義される。アミノ酸配列の同一性は、後述の方法により決定することができる。 When modifying an amino acid residue, it is desirable to mutate it to another amino acid that preserves the properties of the amino acid side chain. For example, amino acid side chain properties include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), amino acids with aliphatic side chains (G, A, V, L, I, P), amino acids with hydroxyl-containing side chains (S, T, Y), amino acids with sulfur-containing side chains (C, M), amino acids with carboxylic acid- and amide-containing side chains (D, N, E, Q), amino acids with base-containing side chains (R, K, H), and amino acids with aromatic-containing side chains (H, F, Y, W) (the numbers in parentheses represent the single-letter symbols of the amino acids). Substitution of amino acids within each of these groups is referred to as a conservative substitution. It is already known that polypeptides having modified amino acid sequences by deletion, addition, and/or substitution of one or more amino acid residues with other amino acids can retain their biological activity (Mark, D. F. et al., Proc. Natl. Acad. Sci. USA (1984) 81:5662-6; Zoller, M. J. and Smith, M., Nucleic Acids Res. (1982) 10:6487-500; Wang, A. et al., Science (1984) 224:1431-3; Dalbadie-McFarland, G. et al., Proc. Natl. Acad. Sci. USA (1982) 79:6409-13). Variable regions of the present invention containing such amino acid modifications have at least 70%, more preferably at least 75%, more preferably at least 80%, even more preferably at least 85%, even more preferably at least 90%, and most preferably at least 95% amino acid sequence identity with the CDR sequence, FR sequence, or entire variable region amino acid sequence before modification. As used herein, sequence identity is defined as the percentage of residues identical to those in the original H-chain variable region or L-chain variable region amino acid sequence, after aligning the sequences as necessary to maximize sequence identity and introducing gaps as appropriate. Amino acid sequence identity can be determined by the method described below.

 また、「機能的に同等の抗体可変領域」には、例えば、表5に記載の抗体番号2, 14, 25, 29, 30~32, 34の可変領域のアミノ酸配列をコードする塩基配列からなる核酸にストリンジェントな条件下でハイブリダイズする核酸から得ることも可能である。可変領域のアミノ酸配列をコードする塩基配列からなる核酸にストリンジェントな条件下でハイブリダイズする核酸を単離するための、ストリンジェントなハイブリダイゼーション条件としては、6 M尿素、0.4% SDS、0.5 x SSC、37℃の条件またはこれと同等のストリンジェンシーのハイブリダイゼーション条件を例示できる。よりストリンジェンシーの高い条件、例えば、6 M尿素、0.4% SDS、0.1 x SSC、42℃の条件を用いれば、より相同性の高い核酸の単離を期待することができる。ハイブリダイゼーション後の洗浄条件は、例えば0.5xSSC(1xSSCは0.15 M NaCl、0.015M クエン酸ナトリウム、pH7.0)、および0.1% SDS、60℃における洗浄、より好ましくは 0.2xSSC、および0.1% SDS、60℃における洗浄、より好ましくは 0.2xSSC、および0.1% SDS、62℃における洗浄、より好ましくは 0.2xSSC、および0.1% SDS、65℃における洗浄、より好ましくは 0.1xSSC、および0.1% SDS、65℃における洗浄である。単離した核酸の配列の決定は、後述の公知の方法によって行うことが可能である。単離された核酸の相同性は、塩基配列全体で、少なくとも50%以上、さらに好ましくは70%以上、さらに好ましくは90%以上(例えば、95%、96%、97%、98%、99%以上)の配列の同一性を有する。
 上記ハイブリダイゼーション技術を利用する方法にかえて、可変領域のアミノ酸配列をコードする塩基配列情報を基に合成したプライマーを用いる遺伝子増幅法、例えば、ポリメラーゼ連鎖反応(PCR)法を利用して、可変領域のアミノ酸配列をコードする塩基配列からなる核酸とストリンジェントな条件下でハイブリダイズする核酸を単離することも可能である。
Furthermore, "functionally equivalent antibody variable regions" can also be obtained from nucleic acids that hybridize under stringent conditions to nucleic acids consisting of nucleotide sequences encoding the amino acid sequences of the variable regions of antibodies Nos. 2, 14, 25, 29, 30 to 32, and 34 listed in Table 5, for example. Examples of stringent hybridization conditions for isolating nucleic acids that hybridize under stringent conditions to nucleic acids consisting of nucleotide sequences encoding the amino acid sequences of the variable regions include 6 M urea, 0.4% SDS, 0.5 x SSC, and 37°C, or hybridization conditions of equivalent stringency. Higher stringency conditions, such as 6 M urea, 0.4% SDS, 0.1 x SSC, and 42°C, are expected to result in the isolation of nucleic acids with higher homology. Washing conditions after hybridization include, for example, 0.5xSSC (1xSSC is 0.15 M NaCl, 0.015 M sodium citrate, pH 7.0) and 0.1% SDS at 60°C, more preferably 0.2xSSC and 0.1% SDS at 60°C, more preferably 0.2xSSC and 0.1% SDS at 62°C, more preferably 0.2xSSC and 0.1% SDS at 65°C, and more preferably 0.1xSSC and 0.1% SDS at 65°C. The sequence of the isolated nucleic acid can be determined by known methods described below. The homology of the isolated nucleic acid is at least 50% or more, more preferably 70% or more, and even more preferably 90% or more (e.g., 95%, 96%, 97%, 98%, 99% or more) sequence identity across the entire base sequence.
Instead of the above-mentioned method utilizing hybridization techniques, it is also possible to isolate nucleic acids that hybridize under stringent conditions with nucleic acids consisting of a nucleotide sequence encoding the amino acid sequence of the variable region by using gene amplification methods, such as polymerase chain reaction (PCR), which use primers synthesized based on the nucleotide sequence information encoding the amino acid sequence of the variable region.

 塩基配列及びアミノ酸配列の同一性は、Karlin and AltschulによるアルゴリズムBLAST(Proc.Natl.Acad.Sci.USA(1993)90:5873-7)によって決定することができる。このアルゴリズムに基づいて、BLASTNやBLASTXと呼ばれるプログラムが開発されている(Altschul et al.,J.Mol.Biol.(1990)215:403-10)。BLASTに基づいてBLASTNによって塩基配列を解析する場合には、パラメーターは例えばscore = 100、wordlength = 12とする。また、BLASTに基づいてBLASTXによってアミノ酸配列を解析する場合には、パラメーターは例えばscore = 50、wordlength = 3とする。BLASTとGapped BLASTプログラムを用いる場合には、各プログラムのデフォルトパラメーターを用いる。これらの解析方法の具体的な手法は公知である(NCBI (National Center for Biotechnology Information)の BLAST(Basic Local Alignment Search Tool)のウェブサイトを参照;http://www.ncbi.nlm.nih.gov)。 The identity of nucleotide sequences and amino acid sequences can be determined using the BLAST algorithm by Karlin and Altschul (Proc. Natl. Acad. Sci. USA (1993) 90:5873-7). Programs called BLASTN and BLASTX have been developed based on this algorithm (Altschul et al., J. Mol. Biol. (1990) 215:403-10). When analyzing nucleotide sequences using BLASTN based on BLAST, the parameters are, for example, score = 100 and wordlength = 12. When analyzing amino acid sequences using BLASTX based on BLAST, the parameters are, for example, score = 50 and wordlength = 3. When using the BLAST and Gapped BLAST programs, the default parameters of each program are used. The specific techniques for these analysis methods are publicly known (see the NCBI (National Center for Biotechnology Information) BLAST (Basic Local Alignment Search Tool) website; http://www.ncbi.nlm.nih.gov).

核酸、ベクター、細胞
 一局面において、本発明は、本発明の抗原結合分子をコードする核酸(ポリヌクレオチド)に関する。本発明の抗原結合分子は、任意の発現ベクターに組み込むことができる。発現ベクターで適当な宿主を形質転換し、抗原結合分子の発現細胞とすることができる。抗原結合分子の発現細胞を培養し、培養上清から発現産物を回収すれば、当該核酸(ポリヌクレオチド)によってコードされる抗原結合分子を取得することができる。即ち本発明は、本発明の抗原結合分子をコードする核酸(ポリヌクレオチド)を含むベクター、当該ベクターを保持する細胞、および当該細胞を培養し培養上清から抗原結合分子を回収することを含む抗原結合分子の製造方法に関する。これらは例えば、前記組換え抗体と同様の手法により得ることができる。
Nucleic Acids, Vectors, and Cells In one aspect, the present invention relates to nucleic acids (polynucleotides) encoding the antigen-binding molecules of the present invention. The antigen-binding molecules of the present invention can be incorporated into any expression vector. An appropriate host can be transformed with the expression vector to create cells expressing the antigen-binding molecules. The antigen-binding molecules encoded by the nucleic acids (polynucleotides) can be obtained by culturing the cells expressing the antigen-binding molecules and recovering the expression product from the culture supernatant. Specifically, the present invention relates to vectors containing nucleic acids (polynucleotides) encoding the antigen-binding molecules of the present invention, cells harboring the vectors, and methods for producing antigen-binding molecules, which include culturing the cells and recovering the antigen-binding molecules from the culture supernatant. These can be obtained, for example, by techniques similar to those used for the recombinant antibodies described above.

医薬組成物
 一局面において、本発明は、本発明の抗原結合分子(特に多重特異性抗原結合分子)を有効成分として含有する医薬組成物を提供する。一態様において、本発明は、当該抗原結合分子を有効成分として含有する、細胞傷害を誘導する医薬組成物を提供する。本発明の医薬組成物は、細胞傷害、特にT細胞依存的細胞傷害を誘導し、当該細胞傷害が予防又は治療に必要な疾患を罹患している対象または再発する可能性がある対象に投与されることが好ましい。一態様において、本発明の医薬組成物は、がんの治療または予防に使用するためのものである。
Pharmaceutical Compositions In one aspect, the present invention provides pharmaceutical compositions comprising, as an active ingredient, an antigen-binding molecule of the present invention (particularly a multispecific antigen-binding molecule). In one embodiment, the present invention provides pharmaceutical compositions that induce cytotoxicity, comprising, as an active ingredient, the antigen-binding molecule. The pharmaceutical compositions of the present invention induce cytotoxicity, particularly T-cell-dependent cytotoxicity, and are preferably administered to subjects suffering from or at risk of recurrence of a disease for which such cytotoxicity is necessary for prevention or treatment. In one embodiment, the pharmaceutical compositions of the present invention are for use in the treatment or prevention of cancer.

 本発明の抗原結合分子(特に多重特異性抗原結合分子)を有効成分として含有する医薬組成物は、当該抗原結合分子を有効成分として含有する細胞傷害誘導剤および細胞増殖抑制剤、当該抗原結合分子を対象に投与する工程を含む細胞傷害を誘導する方法および細胞増殖を抑制する方法、細胞傷害の誘導および細胞増殖の抑制における使用のための当該抗原結合分子、または、細胞傷害誘導剤および細胞増殖抑制剤の製造における当該抗原結合分子の使用と表現することもできる。
 また、本発明の抗原結合分子(特に多重特異性抗原結合分子)を有効成分として含有する医薬組成物は、当該抗原結合分子を有効成分として含有する、がんの治療剤もしくは予防剤、当該抗原結合分子を対象に投与する工程を含む、がんを治療もしくは予防する方法、がんの治療もしくは予防における使用のための当該抗原結合分子、または、がんの治療剤もしくは予防剤の製造における当該抗原結合分子の使用と表現することもできる。
Pharmaceutical compositions containing an antigen-binding molecule of the present invention (particularly a multispecific antigen-binding molecule) as an active ingredient can also be described as cytotoxicity inducers and cytostatic agents containing the antigen-binding molecule as an active ingredient; methods for inducing cytotoxicity and methods for inhibiting cell proliferation comprising the step of administering the antigen-binding molecule to a subject; the antigen-binding molecule for use in inducing cytotoxicity and inhibiting cell proliferation; or use of the antigen-binding molecule in the production of cytotoxicity inducers and cytostatic agents.
Furthermore, pharmaceutical compositions containing an antigen-binding molecule of the present invention (particularly a multispecific antigen-binding molecule) as an active ingredient can also be described as a cancer therapeutic or preventive agent containing the antigen-binding molecule as an active ingredient, a method for treating or preventing cancer which comprises administering the antigen-binding molecule to a subject, the antigen-binding molecule for use in cancer therapeutic or preventive purposes, or use of the antigen-binding molecule in the manufacture of a cancer therapeutic or preventive agent.

 本発明において、「抗原結合分子を有効成分として含有する」とは、当該抗原結合分子を主要な活性成分として含むという意味であり、当該抗原結合分子の含有率を制限するものではない。 In the present invention, "containing an antigen-binding molecule as an active ingredient" means containing the antigen-binding molecule as the main active ingredient, and does not limit the content of the antigen-binding molecule.

 また、一態様において、本発明の抗原結合分子は、ベクター等を用いて本発明の抗原結合分子をコードする核酸を生体内に投与あるいは組み込み、本発明の抗原結合分子を直接生体内で発現させる方法によって投与され得るが、ベクターを用いることなく投与されてもよい。ベクターとして、ウイルスベクター、プラスミドベクターが例示され、さらにはアデノウイルスベクター、アデノ随伴ウイルスベクターが例示される。本発明の抗原結合分子をコードする核酸は直接生体に投与してもよく、また本発明の抗原結合分子をコードする核酸を導入した細胞を生体に投与してもよい。例えば、本発明の抗原結合分子をコードするmRNAに生体内でのmRNAの安定性を高めるための化学修飾を施し、当該mRNAをヒトに直接投与し、生体内で本発明の抗原結合分子を発現させる方法により本発明の抗原結合分子を投与しうる(EP2101823B、WO2013/120629参照)。また、本発明の抗原結合分子をコードする核酸を導入したB細胞を投与してもよい(Sci Immunol. (2019) 4(35), eaax0644)。また、本発明の抗原結合分子をコードする核酸を導入した細菌を投与してもよい(Nature Reviews Cancer (2018) 18, 727-743)。 In one embodiment, the antigen-binding molecule of the present invention can be administered by administering or incorporating a nucleic acid encoding the antigen-binding molecule of the present invention into a living body using a vector or the like, thereby directly expressing the antigen-binding molecule of the present invention in the living body; however, the antigen-binding molecule may also be administered without using a vector. Examples of vectors include viral vectors and plasmid vectors, and further examples include adenovirus vectors and adeno-associated virus vectors. The nucleic acid encoding the antigen-binding molecule of the present invention may be administered directly to a living body, or cells into which a nucleic acid encoding the antigen-binding molecule of the present invention has been introduced may be administered to a living body. For example, the antigen-binding molecule of the present invention can be administered by chemically modifying mRNA encoding the antigen-binding molecule of the present invention to increase mRNA stability in the living body, and then directly administering the mRNA to a human to express the antigen-binding molecule of the present invention in the living body (see EP2101823B and WO2013/120629). Alternatively, B cells into which a nucleic acid encoding the antigen-binding molecule of the present invention has been introduced may be administered (Sci Immunol. (2019) 4(35), eaax0644). Alternatively, bacteria into which nucleic acids encoding the antigen-binding molecules of the present invention have been introduced may be administered (Nature Reviews Cancer (2018) 18, 727-743).

 また、必要に応じ本発明の抗原結合分子は、マイクロカプセル(ヒドロキシメチルセルロース、ゼラチン、ポリ[メチルメタクリル酸]等のマイクロカプセル)に封入され、コロイドドラッグデリバリーシステム(リポソーム、アルブミンミクロスフェア、マイクロエマルジョン、ナノ粒子及びナノカプセル等)とされ得る("Remington's Pharmaceutical Science 16th edition", Oslo Ed. (1980)等参照)。さらに、薬剤を徐放性の薬剤とする方法も公知であり、当該方法は本発明の抗原結合分子に適用され得る(J.Biomed.Mater.Res. (1981) 15, 267-277、Chemtech. (1982) 12, 98-105、米国特許第3773719号、欧州特許公開公報EP58481号・EP133988号、Biopolymers (1983) 22, 547-556)。 If necessary, the antigen-binding molecules of the present invention can be encapsulated in microcapsules (such as microcapsules made of hydroxymethylcellulose, gelatin, or poly(methyl methacrylate)) to form colloid drug delivery systems (such as liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules) (see, for example, "Remington's Pharmaceutical Science 16th edition," Oslo Ed. (1980)). Furthermore, methods for producing sustained-release drugs are also known, and these methods can be applied to the antigen-binding molecules of the present invention (J. Biomed. Mater. Res. (1981) 15, 267-277; Chemtech. (1982) 12, 98-105; U.S. Pat. No. 3,773,719; European Patent Publications EP 58481 and EP 133988; Biopolymers (1983) 22, 547-556).

 本発明の医薬組成物、細胞傷害誘導剤、細胞増殖抑制剤、または、がんの治療剤もしくは予防剤(以下、本発明の医薬組成物と総称する)は、経口、非経口投与のいずれかによって患者に投与することができる。好ましくは非経口投与である。係る投与方法としては具体的には、注射投与、経鼻投与、経肺投与、経皮投与などが挙げられる。注射投与としては、例えば、静脈内注射、筋肉内注射、腹腔内注射、皮下注射などが挙げられる。例えば注射投与によって本発明の医薬組成物が全身または局部的に投与できる。また、患者の年齢、症状により適宜投与方法を選択することができる。投与量としては、例えば、一回の投与につき体重1 kgあたり0.0001 mgから1000 mgの範囲で投与量が選択できる。あるいは、例えば、患者あたり0.001 mg/bodyから100000 mg/bodyの範囲で投与量が選択され得る。しかしながら、本発明の医薬組成物はこれらの投与量に制限されるものではない。 The pharmaceutical composition, cytotoxicity inducer, cytostatic agent, or cancer treatment or prevention agent (hereinafter collectively referred to as the pharmaceutical composition of the present invention) of the present invention can be administered to a patient either orally or parenterally. Parenteral administration is preferred. Specific examples of such administration methods include injection, nasal administration, pulmonary administration, and transdermal administration. Injection methods include, for example, intravenous injection, intramuscular injection, intraperitoneal injection, and subcutaneous injection. For example, the pharmaceutical composition of the present invention can be administered systemically or locally by injection. Furthermore, an appropriate administration method can be selected depending on the patient's age and symptoms. The dosage can be selected, for example, from a range of 0.0001 mg to 1000 mg per kg of body weight per administration. Alternatively, the dosage can be selected, for example, from a range of 0.001 mg/body to 100,000 mg/body per patient. However, the pharmaceutical composition of the present invention is not limited to these dosages.

 本発明の医薬組成物は、常法に従って製剤化することができ(例えば、Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, U.S.A)、医薬的(薬学的)に許容される担体や添加物を共に含むものであってもよい。例えば界面活性剤、賦形剤、着色料、着香料、保存料、安定剤、緩衝剤、懸濁剤、等張化剤、結合剤、崩壊剤、滑沢剤、流動性促進剤、矯味剤等が挙げられる。更にこれらに制限されず、その他常用の担体が適宜使用できる。具体的には、軽質無水ケイ酸、乳糖、結晶セルロース、マンニトール、デンプン、カルメロースカルシウム、カルメロースナトリウム、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、ポリビニルアセタールジエチルアミノアセテート、ポリビニルピロリドン、ゼラチン、中鎖脂肪酸トリグリセライド、ポリオキシエチレン硬化ヒマシ油60、白糖、カルボキシメチルセルロース、コーンスターチ、無機塩類等を担体として挙げることができる。 The pharmaceutical compositions of the present invention can be formulated in accordance with conventional methods (e.g., Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, U.S.A.) and may contain pharmaceutically acceptable carriers and additives. Examples include surfactants, excipients, colorants, flavorings, preservatives, stabilizers, buffers, suspending agents, isotonicity agents, binders, disintegrants, lubricants, flow enhancers, and flavoring agents. Furthermore, without being limited to these, other commonly used carriers can be used as appropriate. Specific examples of carriers include light anhydrous silicic acid, lactose, crystalline cellulose, mannitol, starch, carmellose calcium, carmellose sodium, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinyl acetal diethylaminoacetate, polyvinylpyrrolidone, gelatin, medium-chain fatty acid triglycerides, polyoxyethylene hydrogenated castor oil 60, sucrose, carboxymethyl cellulose, corn starch, and inorganic salts.

 本発明の抗原結合分子と組み合わせ可能な技術の非限定な例として、CD3結合ドメインを利用した、T細胞リダイレクティング抗体を分泌するT細胞の作成が例示される(Trends Immunol.(2019) 40(3) 243-257)。非限定な作成手法の一つとしては、本明細書に記載のCD3結合ドメインと、がん抗原に対する結合ドメインとを含む二重特異性抗体をコードする核酸を遺伝子改変技術によりT細胞などのエフェクター細胞に導入する手法が挙げられる。 A non-limiting example of a technique that can be combined with the antigen-binding molecules of the present invention is the creation of T cells that secrete T cell-redirecting antibodies using a CD3-binding domain (Trends Immunol. (2019) 40(3) 243-257). One non-limiting creation method involves introducing nucleic acid encoding a bispecific antibody comprising the CD3-binding domain described herein and a cancer antigen-binding domain into effector cells such as T cells using gene modification techniques.

治療方法等
 一局面において、本発明は、標的細胞と当該細胞の表面に特異的に発現する抗原に結合する本発明の多重特異性抗原結合分子とを接触させる工程を含む、当該細胞に傷害を引き起こす(誘導する)方法、或いは、該細胞の増殖を抑制する方法を提供する。一態様において、本発明は、標的がん特異的抗原を発現する細胞と当該抗原に結合する本発明の多重特異性抗原結合分子とを接触させる工程を含む、当該抗原の発現細胞、又は、当該発現細胞を含む腫瘍組織に傷害を引き起こす(誘導する)方法、或いは、該細胞又は腫瘍組織の増殖を抑制する方法を提供する。別の態様において、本発明は、免疫応答を抑制する機能を有する細胞と、当該細胞の表面に発現する抗原に結合する本発明の多重特異性抗原結合分子とを接触させる工程を含む、当該細胞に傷害を引き起こす(誘導する)方法、或いは、該細胞の増殖を抑制する方法を提供する。当該抗原に結合する多重特異性抗原結合分子は、本発明の細胞傷害誘導剤および細胞増殖抑制剤に含有される当該抗原に結合する本発明の抗原結合分子として上述したとおりである。当該抗原に結合する本発明の多重特異性抗原結合分子が結合する細胞は、当該抗原が発現している細胞であれば特に限定されない。
In one aspect, such as a method for treatment , the present invention provides a method for inducing (inducing) damage to a target cell or a method for inhibiting the proliferation of the cell, the method comprising the step of contacting the target cell with a multispecific antigen-binding molecule of the present invention that binds to an antigen specifically expressed on the surface of the cell. In one aspect, the present invention provides a method for inducing (inducing) damage to a cell expressing a target cancer-specific antigen or a tumor tissue containing the expressing cell, or a method for inhibiting the proliferation of the cell or tumor tissue, the method comprising the step of contacting the cell expressing the target cancer-specific antigen with a multispecific antigen-binding molecule of the present invention that binds to the antigen. In another aspect, the present invention provides a method for inducing (inducing) damage to a cell or a method for inhibiting the proliferation of the cell, the method comprising the step of contacting a cell having the function of suppressing an immune response with a multispecific antigen-binding molecule of the present invention that binds to an antigen expressed on the surface of the cell. The multispecific antigen-binding molecule that binds to the antigen is as described above for the antigen-binding molecule of the present invention contained in the cytotoxicity-inducing agent and cytostatic agent of the present invention. The cells to which the multispecific antigen-binding molecule of the present invention that binds to the antigen are not particularly limited, as long as the antigen is expressed.

 本発明において「接触」は、例えば、試験管内で培養している標的抗原発現細胞の培養液に、当該抗原に結合する本発明の多重特異性抗原結合分子を添加することにより行われる。この場合において、添加される抗原結合分子の形状としては、溶液又は凍結乾燥等により得られる固体等の形状が適宜使用され得る。水溶液として添加される場合にあっては純粋に本発明の多重特異性抗原結合分子のみを含有する水溶液であり得るし、例えば上記記載の界面活性剤、賦形剤、着色料、着香料、保存料、安定剤、緩衝剤、懸濁剤、等張化剤、結合剤、崩壊剤、滑沢剤、流動性促進剤、矯味剤等を含む溶液でもあり得る。添加する濃度は特に限定されないが、培養液中の最終濃度として、好ましくは1 pg/mlから1 g/mlの範囲であり、より好ましくは1 ng/mlから1 mg/mlであり、更に好ましくは1μg/mlから1 mg/mlが好適に使用され得る。 In the present invention, "contact" is performed, for example, by adding a multispecific antigen-binding molecule of the present invention that binds to the antigen to the culture medium of target antigen-expressing cells cultured in a test tube. In this case, the antigen-binding molecule to be added may be in the form of a solution or a solid obtained by lyophilization or the like, as appropriate. When added as an aqueous solution, it may be an aqueous solution containing only the multispecific antigen-binding molecule of the present invention, or it may be a solution containing, for example, the surfactants, excipients, colorants, flavorings, preservatives, stabilizers, buffers, suspending agents, isotonicity agents, binders, disintegrants, lubricants, flow enhancers, flavoring agents, etc. described above. The concentration to be added is not particularly limited, but a final concentration in the culture medium of preferably 1 pg/ml to 1 g/ml, more preferably 1 ng/ml to 1 mg/ml, and even more preferably 1 μg/ml to 1 mg/ml, is suitable.

 また本発明において「接触」は更に、別の態様では、標的抗原の発現細胞を体内に移植した非ヒト動物や、内在的に当該抗原を発現する細胞を有する動物に投与することによっても行われる。投与方法は経口、非経口投与のいずれかによって実施できる。特に好ましくは非経口投与による投与方法であり、係る投与方法としては具体的には、注射投与、経鼻投与、経肺投与、経皮投与などが挙げられる。注射投与としては、例えば、静脈内注射、筋肉内注射、腹腔内注射、皮下注射などが挙げられる。例えば注射投与によって本発明の医薬組成物、あるいは細胞傷害誘導剤および細胞増殖阻害剤が全身または局部的に投与できる。また、被験動物の年齢、症状により適宜投与方法を選択することができる。水溶液として投与される場合にあっては純粋に本発明の多重特異性抗原結合分子のみを含有する水溶液であってもよいし、例えば上記記載の界面活性剤、賦形剤、着色料、着香料、保存料、安定剤、緩衝剤、懸濁剤、等張化剤、結合剤、崩壊剤、滑沢剤、流動性促進剤、矯味剤等を含む溶液であってもよい。投与量としては、例えば、一回の投与につき体重1 kgあたり0.0001 mgから1000 mgの範囲で投与量が選択できる。あるいは、例えば、患者あたり0.001から100000 mg/bodyの範囲で投与量が選択できる。しかしながら、本発明の多重特異性抗原結合分子の投与量はこれらの投与量に制限されるものではない。 In another embodiment, "contact" in the present invention can also be achieved by administering to a non-human animal into which cells expressing the target antigen have been transplanted, or to an animal that has cells endogenously expressing the antigen. Administration can be performed either orally or parenterally. Parenteral administration is particularly preferred, and specific examples of such administration methods include injection, intranasal administration, pulmonary administration, and transdermal administration. Injection can be, for example, intravenous injection, intramuscular injection, intraperitoneal injection, and subcutaneous injection. For example, the pharmaceutical composition of the present invention, or the cytotoxicity inducer and cell proliferation inhibitor can be administered systemically or locally by injection. An appropriate administration method can be selected depending on the age and symptoms of the subject animal. When administered as an aqueous solution, the solution may contain only the multispecific antigen-binding molecule of the present invention, or may contain, for example, the above-mentioned surfactants, excipients, colorants, flavorings, preservatives, stabilizers, buffers, suspending agents, isotonicity agents, binders, disintegrants, lubricants, flow enhancers, and flavoring agents. The dosage can be selected, for example, from the range of 0.0001 mg to 1000 mg per kg of body weight per administration. Alternatively, the dosage can be selected, for example, from the range of 0.001 to 100,000 mg/body per patient. However, the dosage of the multispecific antigen-binding molecules of the present invention is not limited to these dosages.

 本発明の多重特異性抗原結合分子の接触によって標的細胞(特に、本発明の多重特異性抗原結合分子の第二の抗原結合ドメインが結合する、CD3以外の標的抗原を発現する細胞)に引き起こされた細胞傷害を評価又は測定する方法として、以下の方法が好適に使用される。試験管内において該細胞傷害活性を評価又は測定する方法としては、細胞傷害性T細胞活性などの測定法を挙げることができる。本発明の多重特異性抗原結合分子がT細胞性傷害活性を有するか否か、公知の方法により測定することができる(例えば、Current protocols in Immunology, Chapter 7. Immunologic studies in humans, Editor, John E, Coligan et al., John Wiley & Sons, Inc.,(1993)等)。活性の測定に際しては、標的抗原とは異なる抗原であって試験に使用する細胞が発現していない抗原に結合する抗原結合分子を、本発明の多重特異性抗原結合分子と同様に対照として使用して、本発明の多重特異性抗原結合分子が対照として使用された抗原結合分子よりも強い細胞傷害活性を示すことにより活性が判定され得る。 The following methods are preferably used to evaluate or measure the cytotoxicity induced in target cells (particularly cells expressing a target antigen other than CD3, to which the second antigen-binding domain of the multispecific antigen-binding molecule of the present invention binds) by contact with the multispecific antigen-binding molecule of the present invention. Methods for evaluating or measuring the cytotoxic activity in vitro include methods for measuring cytotoxic T cell activity. Whether or not the multispecific antigen-binding molecule of the present invention has T cell mediated cytotoxic activity can be measured by known methods (e.g., Current Protocols in Immunology, Chapter 7. Immunologic Studies in Humans, Editor, John E., Coligan et al., John Wiley & Sons, Inc., (1993)). When measuring activity, an antigen-binding molecule that binds to an antigen different from the target antigen and that is not expressed by the cells used in the test is used as a control in the same manner as the multispecific antigen-binding molecule of the present invention, and activity can be determined if the multispecific antigen-binding molecule of the present invention exhibits stronger cytotoxic activity than the antigen-binding molecule used as the control.

 また、生体内で細胞傷害活性を評価又は測定するために、標的抗原を発現する細胞を、非ヒト被験動物の皮内又は皮下に移植後、当日又は翌日から毎日又は数日間隔で被験抗原結合分子を静脈又は腹腔内に投与される。腫瘍の大きさを経日的に測定することにより当該腫瘍の大きさの変化の差異が細胞傷害活性と規定され得る。試験管内での評価と同様に対照となる抗原結合分子が投与され、本発明の抗原結合分子の投与群における腫瘍の大きさが対照抗原結合分子の投与群における腫瘍の大きさよりも有意に小さいことが細胞傷害活性を有すると判定され得る。 Furthermore, to evaluate or measure cytotoxic activity in vivo, cells expressing the target antigen are transplanted intradermally or subcutaneously into a non-human test animal, and the test antigen-binding molecule is then administered intravenously or intraperitoneally daily or at intervals of several days from the same day or the following day. By measuring the size of the tumor over time, the difference in the change in size of the tumor can be defined as cytotoxic activity. As with in vitro evaluation, a control antigen-binding molecule is administered, and cytotoxic activity can be determined if the tumor size in the group administered with the antigen-binding molecule of the present invention is significantly smaller than the tumor size in the group administered with the control antigen-binding molecule.

 標的抗原を発現する細胞の増殖に対する抑制効果を評価又は測定する方法としては、アイソトープラベルしたthymidineの細胞へ取込み測定やMTT法が好適に用いられる。また、生体内で細胞増殖抑制活性を評価又は測定する方法として、上記記載の生体内において細胞傷害活性を評価又は測定する方法と同じ方法を好適に用いることができる。 Methods for assessing or measuring the inhibitory effect on the proliferation of cells expressing a target antigen include measuring the uptake of isotope-labeled thymidine into cells and the MTT assay. Furthermore, methods for assessing or measuring cell proliferation inhibitory activity in vivo can be suitably the same as the methods for assessing or measuring cytotoxic activity in vivo described above.

キット
 また本発明は、本発明の抗原結合分子または本発明の製造方法により製造された抗原結合分子を含む、本発明の方法に用いるためのキットを提供する。該キットには、その他、薬学的に許容される担体、媒体、使用方法を記載した指示書等をパッケージしておくことができる。
 また本発明は、本発明の方法に使用するための、本発明の抗原結合分子または本発明の製造方法により製造された抗原結合分子に関する。
The present invention also provides kits for use in the methods of the present invention, which comprise the antigen-binding molecules of the present invention or antigen-binding molecules produced by the production methods of the present invention. The kits may also contain other packaged components such as pharmaceutically acceptable carriers, vehicles, and instructions for use.
The present invention also relates to the antigen-binding molecules of the present invention or antigen-binding molecules produced by the production methods of the present invention, for use in the methods of the present invention.

 前述の発明は、明確な理解を助ける目的のもと、実例および例示を用いて詳細に記載したが、本明細書における記載および例示は、本発明の範囲を限定するものと解釈されるべきではない。本明細書で引用したすべての特許文献および科学文献の開示は、その全体にわたって、参照により明示的に本明細書に組み入れられる。 The foregoing invention has been described in detail by way of illustration and example for purposes of clarity of understanding, but the descriptions and illustrations herein should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated herein by reference in their entireties.

 以下に実施例により本発明をより詳細に説明するが、これらの実施例は本発明の範囲を制限するものではない。 The present invention will be described in more detail below using examples, but these examples are not intended to limit the scope of the present invention.

〔実施例1〕CD3抗原への結合安定性が向上したCDR改変体の作製
 これまでにT細胞マーカーであるCD3と標的細胞上の抗原を認識することにより標的細胞に障害を与える二重特異性抗体は報告されているが、報告されていたCD3結合抗体可変領域には結合安定性を改善する余地が残されていた。このプロファイルを改善することにより、in vivoでの薬効の持続やin vitroでの評価の容易さといった効果が期待された。具体的には、WO2015174439に記載されている抗体可変領域は、緩衝液中で経時的に結合能を失い、結合安定性の向上のためのさらなる改変が必要であると考えられた。
 まず、WO2015174439に記載されているヒトCD3に対する抗体の重鎖可変領域と、ヒトIgG1の定常領域配列とを有する抗体重鎖遺伝子TR01H113-F760mnP17(配列番号:30)、およびヒトCD3に対する抗体の軽鎖可変領域と、ヒトκ鎖の定常領域配列とを有する抗体軽鎖遺伝子L0011-KT0(配列番号:34)が作製された。この抗体遺伝子のCDRのアミノ酸配列を規定する塩基に対して、当業者公知の方法で改変を導入することによって、CDR改変抗体を規定するプラスミドが得られた。重鎖と軽鎖を規定するプラスミドを混合してヒト胎児腎細胞由来HEK293系細胞に導入した。4日間培養された上清から、当業者公知の方法で精製することにより、抗体が得られた。分光光度計を用いて、精製された抗体溶液の280 nmでの吸光度が測定された。得られた測定値から、PACE法により算出された吸光係数を用いて、精製された抗体の濃度が算出された(Protein Science (1995) 4, 2411-2423)。
[Example 1] Preparation of CDR-modified antibodies with improved binding stability to CD3 antigen Bispecific antibodies that damage target cells by recognizing the T cell marker CD3 and an antigen on the target cell have been reported, but there was still room for improvement in the binding stability of the reported CD3-binding antibody variable regions. Improving this profile was expected to have benefits such as sustained in vivo efficacy and ease of in vitro evaluation. Specifically, the antibody variable region described in WO2015174439 lost its binding ability over time in buffer, and further modification to improve binding stability was considered necessary.
First, the antibody heavy chain gene TR01H113-F760mnP17 (SEQ ID NO: 30) was constructed, which contains the heavy chain variable region of an antibody against human CD3 and the constant region sequence of human IgG1, as described in WO2015174439. The antibody light chain gene L0011-KT0 (SEQ ID NO: 34) contained the light chain variable region of an antibody against human CD3 and the constant region sequence of human κ chain. Plasmids encoding CDR-modified antibodies were obtained by introducing modifications into the bases encoding the CDR amino acid sequences of this antibody gene using methods known to those skilled in the art. The heavy and light chain-encoding plasmids were mixed and transfected into human embryonic kidney cell-derived HEK293 cells. The supernatant was cultured for 4 days and purified using methods known to those skilled in the art to obtain antibodies. The absorbance of the purified antibody solution at 280 nm was measured using a spectrophotometer. The concentration of the purified antibody was calculated from the obtained measurement values using the extinction coefficient calculated by the PACE method (Protein Science (1995) 4, 2411-2423).

〔実施例2〕改変されたCDRを有する抗体のCD3抗原への結合評価
 ヒトCD3εおよびCD3γならびにFLAGタグからなる抗原をコードするポリヌクレオチドをHEK293系細胞に導入し、培養上清をイオン交換クロマトグラフィー、FLAGタグによるアフィニティークロマトグラフィー、ゲル濾過クロマトグラフィーにより精製して抗原(以下CD3eg-linker)を得た。
 作製された抗体とCD3eg-linkerとの相互作用解析は、Octet HTXを用いて以下の方法で実施された。バッファーとして0.05% TWEEN20を含むリン酸緩衝生理食塩水(pH7.4)を使用した。センサーチップとしてProtein Aチップ(sartorius)が使用された。表1に記載の抗体は各10μg/mL、ヒトCD3eg-linkerは900, 300, 100, 33 nMに段階希釈でバッファー中にて調製された。抗体は30秒のベースラインののち、センサーチップに60秒間ローディングされ、その後30秒間のベースラインステップを経て各180秒間の抗原結合・解離のステップによって抗原結合特性が測定された。結合値として、測定295秒時点でのレスポンスの大きさが定義された。また、複数濃度でのレスポンスカーブへのグローバルフィッティング(Octet BLI Analysis Version: 12.2.2.4)により平衡解離定数KDが決定された。このグローバルフィッティングでは、結合相と解離相の全ての領域に対して1:1結合モデルによって算出された。
[Example 2] Evaluation of binding of antibodies with altered CDRs to CD3 antigen A polynucleotide encoding an antigen consisting of human CD3ε and CD3γ and a FLAG tag was introduced into HEK293 cells, and the culture supernatant was purified by ion exchange chromatography, FLAG tag affinity chromatography, and gel filtration chromatography to obtain the antigen (hereinafter referred to as CD3eg-linker).
The interaction analysis between the prepared antibodies and CD3eg-linker was performed using Octet HTX as follows. Phosphate-buffered saline (pH 7.4) containing 0.05% TWEEN 20 was used as the buffer. Protein A chips (Sartorius) were used as the sensor chip. The antibodies listed in Table 1 were prepared in buffer at 10 μg/mL, and human CD3eg-linker was serially diluted to 900, 300, 100, and 33 nM. The antibodies were loaded onto the sensor chip for 60 seconds after a 30-second baseline. Antigen binding characteristics were measured by a 30-second baseline step followed by 180-second antigen binding and dissociation steps. The binding value was defined as the magnitude of the response at 295 seconds into the measurement. The equilibrium dissociation constant (KD) was determined by global fitting of the response curve at multiple concentrations (Octet BLI Analysis Version: 12.2.2.4). In this global fitting, the 1:1 binding model was used for all regions of the association and dissociation phases.

〔実施例3〕改変されたCDRを有する抗体のCD3eg-linkerへの結合安定性の評価
 作成された抗体は0.1 mg/mLに希釈され、4℃で3日または50℃で3日の条件で保存され、実施例2と同様の方法で抗原結合が評価された。
[Example 3] Evaluation of binding stability of antibodies with modified CDRs to CD3eg-linker The prepared antibodies were diluted to 0.1 mg/mL and stored at 4°C for 3 days or at 50°C for 3 days, and antigen binding was evaluated using the same method as in Example 2.

 表中の「結合H/I比」とは、各抗体について4℃での保存の条件と50℃での保存の条件で保存後の結合測定の値の比をとったものであり、各抗体が50℃の条件でどの程度結合活性を維持するかを示す値である。本発明において作成された抗体番号2, 14, 25, 29, 30~32, 34は、50℃の保存条件時の結合値が既報のコントロール抗体1の0.078以上であった。また、これら抗体については、結合H/I値が75%以上と、既報のコントロール抗体1の61%を上回る数値であった。したがって、これら改変体は緩衝液中での50℃条件での結合能の低下が大きく改善されていることが確認された。 The "binding H/I ratio" in the table is the ratio of the binding measurement values for each antibody after storage at 4°C to those after storage at 50°C, and indicates the extent to which each antibody maintains its binding activity at 50°C. Antibodies Nos. 2, 14, 25, 29, 30-32, and 34 created in the present invention had binding values at 50°C that were equal to or greater than 0.078, the value of the previously reported control antibody 1. Furthermore, the binding H/I values for these antibodies were 75% or greater, exceeding the 61% value of the previously reported control antibody 1. Therefore, it was confirmed that these modified antibodies showed a significant improvement over the decrease in binding ability at 50°C in buffer.

〔実施例4〕改変されたCDRを有する抗体のTDCC Reporter Bioassayによる評価
 本発明において作成された各抗体と抗MarvelD3抗体を用いて、当業者公知の方法において二重特異性抗体が調製された。調製された抗体は各0.044mg/mLに希釈され、実施例3と同様に4℃または50℃で3日間保存された。これらの抗体について、In vitroでのTDCC活性が測定され、保存時の温度によるTDCC活性の違いが評価された。In vitro TDCC活性測定にはTCR/CD3 Effector Cells (NFAT)、Bio-Glo luciferase assay system(Promega)が用いられ、TDCC活性はレポーター遺伝子であるLuciferaseの発光により定量された。384ウェルプレートの各ウェルに、培地により3×106/mLに濃度を調製されたNFAT-RE-luc2 Jurkatと、5×105 mLに濃度を調製されたLS 174T細胞 (ATCC)、10 nMまたは100 nMの抗体溶液を各10μL添加することで混合し、反応を開始した。ここで、LS 174TはMarvelD3抗原を発現する細胞株である。反応は37℃で終夜行われ、インキュベートした各ウェルにLuciferase基質を添加してプレートリーダーで発光を測定することでレポーター遺伝子の発現が定量された。ここで、Luciferaseレスポンス(counts per second: CPS)は、10 nMと100 nMの抗体溶液でのそれぞれの測定値の平均値を表す。
Example 4: Evaluation of antibodies with modified CDRs by TDCC Reporter Bioassay. Bispecific antibodies were prepared using each antibody prepared in the present invention and an anti-Marvel D3 antibody using methods known to those skilled in the art. The prepared antibodies were diluted to 0.044 mg/mL and stored at 4°C or 50°C for 3 days, as in Example 3. The in vitro TDCC activity of these antibodies was measured to assess differences in TDCC activity depending on storage temperature. In vitro TDCC activity was measured using TCR/CD3 Effector Cells (NFAT) and the Bio-Glo luciferase assay system (Promega), and TDCC activity was quantified by luminescence from the reporter gene luciferase. The reaction was initiated by adding 10 μL of NFAT-RE-luc2 Jurkat cells (prepared in medium at a concentration of 3 × 10 6 /mL) and LS 174T cells (ATCC) (prepared at a concentration of 5 × 10 5 mL) to each well of a 384-well plate. LS 174T is a cell line expressing the MarvelD3 antigen. The reaction was carried out overnight at 37°C. After incubation, reporter gene expression was quantified by adding luciferase substrate to each well and measuring luminescence using a plate reader. The luciferase response (counts per second: CPS) represents the average of the values measured for the 10 nM and 100 nM antibody solutions.

NAは、測定が行われていないことを示す。 NA indicates that the measurement was not performed.

 表中の「活性化H/I比」とは、各抗体について4℃での保存の条件と50℃での保存の条件で保存後のLuciferaseレスポンスの値の比をとったものであり、各抗体が50℃の条件でどの程度結合活性を失ったかを示す値である。
 実施例3で選択された抗体番号2, 14, 25, 29, 30~32, 34は、4℃の条件で保存された場合のLuciferaseレスポンスにおいては50℃で保存された際の既報のコントロール抗体1のレスポンスに相当する4.3.E+05以上の値を示した。また、抗体番号14, 25, 29, 30~32, 34は、50℃の条件で保存された場合に67%以上の活性化H/I TDCC活性比を示した。これは既報のコントロール抗体1よりも優れた値であり、これらの抗体はTDCC活性を保ちながら活性化H/I比を改善した改変体であった。抗体番号1, 2, 14, 25, 29, 30~32, 34のアミノ酸配列を表5に示す。
The "activation H/I ratio" in the table is the ratio of the luciferase response values for each antibody after storage at 4°C to those after storage at 50°C, and indicates the extent to which each antibody lost its binding activity at 50°C.
Antibody Nos. 2, 14, 25, 29, 30-32, and 34 selected in Example 3 exhibited luciferase responses of 4.3.E+05 or higher when stored at 4°C, which corresponds to the response of the previously reported control antibody 1 when stored at 50°C. Furthermore, antibody Nos. 14, 25, 29, 30-32, and 34 exhibited activated H/I TDCC activity ratios of 67% or higher when stored at 50°C. This was superior to the previously reported value of control antibody 1, indicating that these antibodies were modified antibodies that maintained TDCC activity while improving the activated H/I ratio. The amino acid sequences of antibody Nos. 1, 2, 14, 25, 29, 30-32, and 34 are shown in Table 5.

 本発明によって、従来のT細胞リダイレクティング抗体で用いられているCD3結合ドメインと比べて優れた安定性を有する新規CD3結合ドメイン、および当該CD3結合ドメインを含む抗原結合分子が提供された。本発明のCD3結合ドメインを、がん細胞等の標的細胞の表面に発現する抗原に結合する抗原結合ドメインと組み合わせて調製した二重特異性抗原結合分子は、当該標的細胞に対してT細胞依存性細胞傷害を誘導することができ、様々ながんの治療または予防に用いることができる。本発明のCD3結合ドメインを含む抗原結合分子は、常温以上の環境下でのCD3に対する結合活性の低下の程度が小さく、患者に投与された場合、そのT細胞依存性細胞傷害活性が長期にわたって持続することが期待される。 The present invention provides a novel CD3-binding domain that has superior stability compared to CD3-binding domains used in conventional T cell-redirecting antibodies, and an antigen-binding molecule containing said CD3-binding domain. Bispecific antigen-binding molecules prepared by combining the CD3-binding domain of the present invention with an antigen-binding domain that binds to an antigen expressed on the surface of target cells such as cancer cells can induce T cell-dependent cytotoxicity against the target cells and can be used to treat or prevent various cancers. Antigen-binding molecules containing the CD3-binding domain of the present invention show only a small decrease in CD3-binding activity at room temperature or above, and are expected to maintain their T cell-dependent cytotoxic activity for a long period of time when administered to patients.

Claims (15)

 第一の抗原結合ドメインおよび第二の抗原結合ドメインを含む抗原結合分子であって、第一の抗原結合ドメインが、CD3に対する結合活性を有する抗体H鎖可変領域及びL鎖可変領域を含み、
 前記H鎖可変領域が、
  アミノ酸配列NAWMH(配列番号:1)を含むH鎖CDR1;
  アミノ酸配列QIX1DKSQNYATX2VAESVKG(配列番号:2)を含むH鎖CDR2であって、X1がKまたはRであり、X2がYまたはFである、H鎖CDR2;及び
  アミノ酸配列VHYX3AGYGVDX4(配列番号:3)を含むH鎖CDR3であって、X3がAまたはPであり、X4がI、MまたはLである、H鎖CDR3
を含み、
 前記L鎖可変領域が、
  アミノ酸配列RSX5X6X7VVHENRX8TYLH(配列番号:4)を含むL鎖CDR1であって、X5がSまたはTであり、X6がQまたはMであり、X7がSまたはTであり、X8がQまたはNである、L鎖CDR1;
  アミノ酸配列KVSNRFS(配列番号:5)を含むL鎖CDR2;及び
  アミノ酸配列GQGTQVPYT(配列番号:6)を含むL鎖CDR3
を含む、抗原結合分子。
An antigen-binding molecule comprising a first antigen-binding domain and a second antigen-binding domain, wherein the first antigen-binding domain comprises an antibody heavy chain variable region and an antibody light chain variable region having binding activity to CD3;
the heavy chain variable region
H chain CDR1 comprising the amino acid sequence NAWMH (SEQ ID NO: 1);
an H chain CDR2 comprising the amino acid sequence QIX1DKSQNYATX2VAESVKG (SEQ ID NO: 2 ), wherein X1 is K or R, and X2 is Y or F; and an H chain CDR3 comprising the amino acid sequence VHYX3AGYGVDX4 (SEQ ID NO: 3 ), wherein X3 is A or P, and X4 is I, M, or L.
Including,
the light chain variable region
an L chain CDR1 comprising the amino acid sequence RSX5X6X7VVHENRX8TYLH ( SEQ ID NO: 4 ), wherein X5 is S or T, X6 is Q or M, X7 is S or T, and X8 is Q or N;
L chain CDR2 comprising the amino acid sequence KVSNRFS (SEQ ID NO: 5); and L chain CDR3 comprising the amino acid sequence GQGTQVPYT (SEQ ID NO: 6).
An antigen-binding molecule comprising:
 CD3に対する結合活性を有する前記抗体H鎖可変領域及びL鎖可変領域が、以下の(a1)~(a8)から選ばれるH鎖CDR1、CDR2及びCDR3、並びに、L鎖CDR1、CDR2及びCDR3の組合せのうちのいずれかを含む、請求項1に記載の抗原結合分子:
 (a1) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:7に示されるアミノ酸配列を含むH鎖CDR2、配列番号:10に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a2) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a3) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:8に示されるアミノ酸配列を含むH鎖CDR2、配列番号:11に示されるアミノ酸配列を含むH鎖CDR3、配列番号:16に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a4) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a5) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:13に示されるアミノ酸配列を含むH鎖CDR3、配列番号:15に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a6) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:17に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a7) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:18に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ、
 (a8) 配列番号:1に示されるアミノ酸配列を含むH鎖CDR1、配列番号:9に示されるアミノ酸配列を含むH鎖CDR2、配列番号:12に示されるアミノ酸配列を含むH鎖CDR3、配列番号:19に示されるアミノ酸配列を含むL鎖CDR1、配列番号:5に示されるアミノ酸配列を含むL鎖CDR2、及び配列番号:6に示されるアミノ酸配列を含むL鎖CDR3の組み合わせ。
The antigen-binding molecule of claim 1, wherein the antibody H chain variable region and L chain variable region having binding activity to CD3 comprise any combination of H chain CDR1, CDR2, and CDR3, and L chain CDR1, CDR2, and CDR3 selected from the following (a1) to (a8):
(a1) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 7, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 10, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a2) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a3) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 8, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 11, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 16, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a4) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a5) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 13, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 15, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a6) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 17, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a7) a combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 18, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6;
(a8) A combination of an H chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1, an H chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 9, an H chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 12, an L chain CDR1 comprising the amino acid sequence shown in SEQ ID NO: 19, an L chain CDR2 comprising the amino acid sequence shown in SEQ ID NO: 5, and an L chain CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6.
 CD3に対する結合活性を有する前記抗体H鎖可変領域及びL鎖可変領域が、以下の(a1)~(a8)から選ばれるH鎖可変領域及びL鎖可変領域の組合せのうちのいずれかを含む、請求項1または2に記載の抗原結合分子:
 (a1)配列番号:20に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a2) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a3) 配列番号:21に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:26に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a4) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a5) 配列番号:23に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:25に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a6) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:27に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a7) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:28に示されるアミノ酸配列を含むL鎖可変領域の組合せ、
 (a8) 配列番号:22に示されるアミノ酸配列を含むH鎖可変領域及び配列番号:29に示されるアミノ酸配列を含むL鎖可変領域の組合せ。
The antigen-binding molecule of claim 1 or 2, wherein the antibody H chain variable region and L chain variable region having CD3-binding activity comprise any combination of H chain variable region and L chain variable region selected from the following (a1) to (a8):
(a1) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 20 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a2) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a3) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 21 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 26;
(a4) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a5) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 23 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 25;
(a6) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 27;
(a7) a combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 28;
(a8) A combination of an H-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 22 and an L-chain variable region comprising the amino acid sequence shown in SEQ ID NO: 29.
 Fc領域をさらに含む、請求項1~3のいずれか一項に記載の抗原結合分子。 The antigen-binding molecule of any one of claims 1 to 3, further comprising an Fc region.  単一特異性抗原結合分子である、請求項1~4のいずれか一項に記載の抗原結合分子。 The antigen-binding molecule of any one of claims 1 to 4, which is a monospecific antigen-binding molecule.  二重特異性抗原結合分子である、請求項1~4のいずれか一項に記載の抗原結合分子。 The antigen-binding molecule of any one of claims 1 to 4, which is a bispecific antigen-binding molecule.  第二の抗原結合ドメインが、がん抗原に対する結合活性を有する抗体可変領域を含む、請求項6に記載の抗原結合分子。 The antigen-binding molecule of claim 6, wherein the second antigen-binding domain comprises an antibody variable region having binding activity against a cancer antigen.  抗体である、請求項1~7のいずれか一項に記載の抗原結合分子。 The antigen-binding molecule of any one of claims 1 to 7, which is an antibody.  請求項1~8のいずれか一項に記載の抗原結合分子をコードする核酸。 A nucleic acid encoding the antigen-binding molecule of any one of claims 1 to 8.  請求項9に記載された核酸が導入されたベクター。 A vector incorporating the nucleic acid described in claim 9.  請求項10に記載のベクターを含む細胞。 A cell containing the vector described in claim 10.  請求項11に記載の細胞を培養する工程を含む、請求項1~8のいずれか一項に記載の抗原結合分子を製造する方法。 A method for producing the antigen-binding molecule of any one of claims 1 to 8, comprising the step of culturing the cell of claim 11.  請求項12に記載の方法によって製造された、抗原結合分子。 An antigen-binding molecule produced by the method of claim 12.  請求項1~8のいずれか一項に記載の抗原結合分子と、薬学的に許容される担体とを含む、医薬組成物。 A pharmaceutical composition comprising the antigen-binding molecule of any one of claims 1 to 8 and a pharmaceutically acceptable carrier.  がんの治療または予防に使用するための、請求項14に記載の医薬組成物。 The pharmaceutical composition of claim 14 for use in the treatment or prevention of cancer.
PCT/JP2025/003698 2024-02-06 2025-02-05 Cd3-targeting antigen-binding molecule with improved stability Pending WO2025169945A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2024016633 2024-02-06
JP2024-016633 2024-02-06

Publications (1)

Publication Number Publication Date
WO2025169945A1 true WO2025169945A1 (en) 2025-08-14

Family

ID=96700004

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2025/003698 Pending WO2025169945A1 (en) 2024-02-06 2025-02-05 Cd3-targeting antigen-binding molecule with improved stability

Country Status (1)

Country Link
WO (1) WO2025169945A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022529288A (en) * 2019-07-10 2022-06-20 中外製薬株式会社 Claudin 6-bonded molecule and its use
JP2024501653A (en) * 2020-12-23 2024-01-15 ベイジン マブワークス バイオテック カンパニー リミテッド Antibodies that bind to human and monkey CD3 and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022529288A (en) * 2019-07-10 2022-06-20 中外製薬株式会社 Claudin 6-bonded molecule and its use
JP2024501653A (en) * 2020-12-23 2024-01-15 ベイジン マブワークス バイオテック カンパニー リミテッド Antibodies that bind to human and monkey CD3 and uses thereof

Similar Documents

Publication Publication Date Title
US12359001B2 (en) Method for producing polypeptide hetero-oligomer
US20230174655A1 (en) Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY
JP7360242B2 (en) Mouse FcγRII-specific Fc antibody
US11932697B2 (en) Antigen-binding domain, and polypeptide including conveying section
TWI700292B (en) Manufacturing method of multi-peptide heteropolymer
JP7519990B2 (en) An antigen-binding molecule capable of binding to CD3 and CD137, but not simultaneously.
US10919953B2 (en) FcgammaRIIB-specific Fc region variant
US20220315909A1 (en) Protease substrate, and polypeptide including protease cleavage sequence
WO2025169945A1 (en) Cd3-targeting antigen-binding molecule with improved stability
RU2831840C2 (en) High-affinity anti-cd3 antibodies and methods for production and use thereof
HK1221963B (en) Method for producing polypeptide heteromultimer
HK1190166B (en) FCγRIIB-SPECIFIC FC ANTIBODY

Legal Events

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

Ref document number: 25752171

Country of ref document: EP

Kind code of ref document: A1