WO2024078558A1 - Anticorps anti-cd100 et son utilisation - Google Patents

Anticorps anti-cd100 et son utilisation Download PDF

Info

Publication number
WO2024078558A1
WO2024078558A1 PCT/CN2023/124146 CN2023124146W WO2024078558A1 WO 2024078558 A1 WO2024078558 A1 WO 2024078558A1 CN 2023124146 W CN2023124146 W CN 2023124146W WO 2024078558 A1 WO2024078558 A1 WO 2024078558A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
sequence shown
amino acid
acid sequence
antibody
Prior art date
Application number
PCT/CN2023/124146
Other languages
English (en)
Chinese (zh)
Inventor
闫鑫甜
李双琦
郎国竣
方小鹏
高雪
韩洋洋
任雨琪
徐培芳
邓强强
Original Assignee
三优生物医药(上海)有限公司
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 三优生物医药(上海)有限公司 filed Critical 三优生物医药(上海)有限公司
Publication of WO2024078558A1 publication Critical patent/WO2024078558A1/fr

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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • the present invention belongs to the field of biomedicine. Specifically, the present invention relates to anti-CD100 antibodies and uses thereof.
  • CD100 also known as semaphorin 4D (SEMA4D)
  • SEMA4D semaphorin 4D
  • CD100 is expressed as a homodimer on the cell surface and can be released from the cell surface by proteolysis to produce an active soluble form of CD100.
  • CD100 is strongly expressed mainly in human lymphoid tissue, skeletal muscle and brain (at lower levels). Its biological activity is mainly characterized in the immune system: for example, as a receptor, CD100 can enhance T cell proliferation; as a ligand, it can promote the aggregation and survival of B cells and the activation and maturation of antigen-presenting cells (dendritic cells and macrophages). Through the high-affinity receptor Plexin-B1, CD100 can inhibit the migration of monocytes and B cells.
  • CD100 is widely expressed in many human tumors, and its expression is associated with aggressive disease in humans.
  • expression of CD100 by inflammatory and tumor cells regulates the infiltration, spatial distribution, and activity of myeloid and lymphocytes.
  • CD100 binds to Plexin receptors located on myeloid cells in the tumor microenvironment.
  • the CD100 protein is blocked, the CD100 barrier is eliminated. Once the barrier is breached, inflammatory dendritic cells and proinflammatory antigen-presenting cells migrate and infiltrate the tumor.
  • blocking CD100 with antibodies delays tumor growth and promotes durable tumor rejection.
  • CD100 Some antibodies against CD100 have been developed in the prior art, such as Pepinemab (Vaccinex, Inc.), but there is still a need in the art for anti-CD100 antibodies that can specifically bind to CD100 and block the binding of CD100 to its Plexin receptor.
  • Pepinemab Vaccinex, Inc.
  • One aspect of the present invention provides an antibody against CD100 (anti-CD100 antibody) or an antigen-binding fragment thereof, wherein the antibody or the antigen-binding fragment thereof specifically recognizes and binds to CD100.
  • the antibody or antigen-binding fragment thereof against CD100 comprises a heavy chain variable region, wherein the heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 sequences, and wherein the HCDR1 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:1, 7, 19, 33, 41, 47 or 52 by no more than 2 amino acids in addition, deletion or substitution; the HCDR2 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:2, 8, 14, 20, 26, 42, 48 or 53 by no more than 2 amino acids in addition, deletion or substitution; and/or the HCDR3 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:3, 9, 15, 21, 27, 30, 35, 38, 43, 49 or 54 by no more than 2 amino acids in addition, deletion or substitution.
  • the antibody or antigen-binding fragment thereof against CD100 comprises a light chain variable region, wherein the light chain variable region comprises LCDR1, LCDR2 and LCDR3 sequences, and wherein the LCDR1 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO: 4, 10, 16, 36, 44 or 50 by no more than 2 amino acids in addition, deletion or substitution; the LCDR2 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO: 5, 11 or 45 by no more than 2 amino acids in addition, deletion or substitution; and/or LCDR3 The sequence differs in amino acid sequence from the sequence shown in SEQ ID NO: 6, 12, 18, 24, 37, 46, 51 or 56 by no more than 2 amino acids being added, deleted or substituted.
  • the heavy chain variable region comprises a HCDR1 sequence shown in SEQ ID NO: 1, 7, 13, 19, 25, 33, 41, 47, 52 or 57; a HCDR2 sequence shown in SEQ ID NO: 2, 8, 14, 20, 26, 34, 42, 48, 53 or 58; and a HCDR3 sequence shown in SEQ ID NO: 3, 9, 15, 21, 27, 30, 35, 38, 43, 49, 54, 60, 63 or 66.
  • the light chain variable region comprises a LCDR1 sequence shown in SEQ ID NO:4, 10, 16, 22, 31, 36, 39, 44, 50, 61 or 64; a LCDR2 sequence shown in SEQ ID NO:5, 11, 17, 23, 28, 45, 55 or 59; and a LCDR3 sequence shown in SEQ ID NO:6, 12, 18, 24, 29, 32, 37, 40, 46, 51, 56, 62 or 65.
  • the heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 sequences
  • the light chain variable region comprises LCDR1, LCDR2 and LCDR3 sequences
  • the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3 sequences are selected from any one of (1)-(15): (1) the HCDR1 sequence shown in SEQ ID NO:1; the HCDR2 sequence shown in SEQ ID NO:2; the HCDR3 sequence shown in SEQ ID NO:3; the LCDR1 sequence shown in SEQ ID NO:4; the LCDR2 sequence shown in SEQ ID NO:5; and the LCDR3 sequence shown in SEQ ID NO:6; (2) the HCDR1 sequence shown in SEQ ID NO:7; the HCDR2 sequence shown in SEQ ID NO:8; the HCDR3 sequence shown in SEQ ID NO:9; the LCDR1 sequence shown in SEQ ID NO:10; the LCDR2 sequence shown in SEQ ID NO:11; and the LCDR3 sequence shown in SEQ ID
  • the heavy chain variable region comprises 1) an amino acid sequence as shown in SEQ ID NO: 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93 or 95; 2) an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, or more identical to the amino acid sequence as shown in SEQ ID NO: 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93 or 95.
  • the light chain variable region comprises 1) an amino acid sequence as shown in SEQ ID NO: 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94 or 96; 2) an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, or more identical to the amino acid sequence as shown in SEQ ID NO: 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94 or 96.
  • the heavy chain variable region and the light chain variable region are selected from any one of (1) to (15): (1) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 67; and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 68; (2) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 69; and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 70; (3) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 71; and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 72; (4) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 73; and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 74; (5) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 75; and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 76; (6) a heavy chain variable region comprising
  • the antibody or antigen-binding fragment thereof comprises a heavy chain comprising 1) an amino acid sequence as shown in SEQ ID NO: 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123 or 125; 2) an amino acid sequence that has at least 80%, at least 85%, at least 90% affinity, or at least 50% affinity with the amino acid sequence as shown in SEQ ID NO: 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123 or 125.
  • the antibody or antigen-binding fragment thereof comprises a light chain, wherein the light chain comprises 1) an amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126; 2) an amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126 that has at least 80%, at least 85%, at least 90% affinity, or at least 10% affinity with the amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126 0%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity;
  • the heavy chain and the light chain are selected from any one of (1) to (15): (1) a heavy chain comprising the amino acid sequence of SEQ ID NO: 97; a light chain comprising the amino acid sequence of SEQ ID NO: 98; (2) a heavy chain comprising the amino acid sequence of SEQ ID NO: 99; a light chain comprising the amino acid sequence of SEQ ID NO: 100; (3) a heavy chain comprising the amino acid sequence of SEQ ID NO: 101; a light chain comprising the amino acid sequence of SEQ ID NO: 102; (4) a heavy chain comprising the amino acid sequence of SEQ ID NO: 103; a light chain comprising the amino acid sequence of SEQ ID NO: 104; (5) a heavy chain comprising the amino acid sequence of SEQ ID NO: 105; a light chain comprising the amino acid sequence of SEQ ID NO: 106; (6) a heavy chain comprising the amino acid sequence of SEQ ID NO: 107; a light chain comprising the amino acid sequence of SEQ ID NO: 108
  • amino acid sequence shown in SEQ ID NO: 116 is shown in FIG. 117; the amino acid sequence shown in SEQ ID NO: 118 is shown in FIG. 119; the amino acid sequence shown in SEQ ID NO: 120 is shown in FIG. 121; the amino acid sequence shown in SEQ ID NO: 122 is shown in FIG. 123; the amino acid sequence shown in SEQ ID NO: 124 is shown in FIG. 125; the amino acid sequence shown in SEQ ID NO: 126 is shown in FIG. 127; the amino acid sequence shown in SEQ ID NO: 128 is shown in FIG. 129; the amino acid sequence shown in SEQ ID NO: 130 is shown in FIG. 131; the amino acid sequence shown in SEQ ID NO: 132 is shown in FIG. 133; the amino acid sequence shown in SEQ ID NO: 134 is shown in FIG. 135
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the anti-CD100 antibody or antigen-binding fragment thereof of the present invention and a pharmaceutically acceptable carrier.
  • the anti-CD100 antibody comprises antibody B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, or B13-e-2.
  • the present invention provides a pharmaceutical combination comprising the anti-CD100 antibody or antigen-binding fragment thereof of the present invention and an anti-PD-L1 antibody or antigen-binding fragment thereof.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof specifically recognizes and binds to PD-L1, wherein the anti-PD-L1 antibody or antigen-binding fragment thereof comprises an immunoglobulin single variable domain;
  • the immunoglobulin single variable domain comprises CDR1, which comprises the amino acid sequence shown in SEQ ID NO:130; CDR2, which comprises the amino acid sequence shown in SEQ ID NO:131; and CDR3, which comprises the amino acid sequence shown in SEQ ID NO:132.
  • the immunoglobulin single variable domain comprises: 1) the amino acid sequence shown in SEQ ID NO:133; or 2) an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:133.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof further comprises an Fc fragment of human IgG1.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises an amino acid sequence as shown in SEQ ID NO: 134 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO: 134.
  • the pharmaceutical combination is a pharmaceutical composition or a kit.
  • the present invention also provides use of the antibody or antigen-binding fragment thereof of the present invention, the pharmaceutical composition of the present invention, or the pharmaceutical combination of the present invention in the preparation of a medicament for treating cancer.
  • the present invention provides a method for treating cancer, comprising administering the antibody or antigen-binding fragment thereof, the pharmaceutical composition or the pharmaceutical combination of the present invention to a subject in need thereof.
  • the present invention relates to the antibody or antigen-binding fragment thereof of the present invention, the pharmaceutical composition of the present invention, or the pharmaceutical combination of the present invention for use in treating cancer.
  • the present invention also provides an isolated nucleic acid molecule encoding an anti-CD100 antibody or antigen-binding fragment thereof of the present invention.
  • the present invention also relates to an expression vector comprising the nucleic acid molecule of the present invention.
  • the present invention also relates to a host cell comprising the nucleic acid molecule or expression vector of the present invention.
  • the present invention also relates to a method for producing an anti-CD100 antibody or an antigen-binding fragment thereof, the method comprising:
  • FIG1 shows the ELISA test results of Pepinemab antibody and positive control 2D5 and 5D8 antibodies.
  • FIG. 2 shows the ELISA test results of antigens from different species.
  • FIG. 3 shows the FACS detection results of overexpressing cell lines of different species.
  • Figures 4A-4D show the FACS test results of Plexin receptor overexpression cell lines of different species.
  • Figure 4A is the test result of HuPlexin-B1-HEK293 overexpression cell line
  • Figure 4B is the test result of MusPlexin-B1-HEK293 overexpression cell line
  • Figure 4C is the test result of CynoPlexin-B1-HEK293 overexpression cell line
  • Figure 4D is the test result of HuPlexin-B2-HEK293 overexpression cell line.
  • Figures 5A-5X show the results of ELISA for detecting the binding activity of candidate antibodies to antigen proteins of different species.
  • Figures 5A-5H show the results of the binding activity of antibody molecules A14, A15, B13, H5, H12, H21, H74, H96, C-C081, C-C171 and C-B71 to HuCD100-His antigen protein;
  • Figures 5I-5P show the results of the binding activity of antibody molecules A14, A15, B13, H5, H12, H21, H74, H96, C-C081, C-C171 and C-B71 to MusCD100-His antigen protein;
  • Figures 5Q-5X show the results of the binding activity of antibody molecules A14, A15, B13, H5, H12, H21, H74, H96, C-C081, C-C171 and C-B71 to CynoCD100-His antigen protein.
  • FIG. 6A to FIG. 6O show the results of detecting the binding activity of candidate antibodies to overexpressing cell lines of different species using FACS.
  • Figures 6A-6E show the results of the binding activity of antibody molecules H5, H21, A14, A15, B13, H74, H96, H12, C-C081, C-C171 and C-B71 to HuCD100-HEK293 overexpressing cell lines;
  • Figures 6F-6J show the results of the binding activity of antibody molecules H5, H21, A14, A15, B13, H74, H96, H12, C-C081, C-C171 and C-B71 to MusCD100-HEK293 overexpressing cell lines;
  • Figures 6K-6O show the results of the binding activity of antibody molecules H5, H21, A14, A15, B13, H74, H96, H12, C-C081, C-C171 and C-B71 to CynoCD100-HEK293 overexpressing cell lines.
  • Figures 7A-7H show the results of FACS detection of candidate antibodies for HuPBMC and Jurkat cell binding activity.
  • Figures 7A-7D are the results of antibody molecules A14, A15, B13, H5, H21, H74, H96, H12, C-C081, C-C171 and C-B71 for HuPBMC cell binding activity;
  • Figures 7E-7H are the results of antibody molecules A14, A15, B13, H5, H21, H74, H96, H12, C-C081, C-C171 and C-B71 for Jurkat cell binding activity.
  • Figures 8A-8O show the results of candidate antibodies blocking the binding of CD100 to Plexin receptors of different species.
  • Figures 8A-8D show the results of antibody molecules A14, A15, B13, H5, H21, H74, H96, H12, C-C081, C-C171 and C-B71 blocking the binding of HuCD100 to HuPlexin-B1-HEK293 cells;
  • Figures 8E-8I show the results of antibody molecules A14, H5, A15, B13, H21, H74, H96, H12, C-C081, C-C171 and C-B71 blocking the binding of HuCD100 to HuPlexin-B2-HEK293 cells;
  • Figure 8J -8L shows the results of antibody molecules H5, A14, A15, B13, H12, H21, H74, H96, C-C081, C-C171 and C-B71 blocking the binding of MusCD100 to MusPlexin-B1-HEK293 cells;
  • Figures 9A-9C show candidate antibodies A14, A15, B13, H12, H21, H74, H96, H5, C-C081, MDSC proliferation inhibition assay results of C-C171 and C-B71.
  • Figures 10A-10B show the results of FACS detection of the binding activity of anti-PD-L1 antibody m18 to human PD-L1-CHO cells overexpressing cell lines and PD-L1-positive cells HCC827 cells.
  • Figure 10A shows the results of binding activity to human PD-L1-CHO cells;
  • Figure 10B shows the results of binding activity to HCC827 cells.
  • Figures 11A-11B show the results of FACS detection of the binding activity of anti-PD-L1 antibody m18 to overexpressing cell lines of different species.
  • Figure 11A shows the results of the binding activity to mouse PD-L1-CHO cells;
  • Figure 11B shows the results of the binding activity to cynomolgus monkey PD-L1-CHO cells.
  • FIG. 12 shows the results of ELISA detection of the specific binding activity of the anti-PD-L1 antibody m18 to B7-H1 and homologous proteins.
  • Figures 13A-13D show the results of FACS detection of the binding activity of the affinity-matured modified antibodies H5-h-7, H5-a-2, B13-c-5, and B13-e-2 of the candidate antibodies H5 and B13 to CD100-overexpressing cell lines of different species and CD100-positive PBMC cells.
  • Figure 13A shows the results of the binding activity to HuCD100-HEK293 cells;
  • Figure 13B shows the results of the binding activity to human PBMC cells;
  • Figure 13C shows the results of the binding activity to MusCD100-HEK 293 cells;
  • Figure 13D shows the results of the binding activity to CynoCD100-HEK 293 cells.
  • Figures 14A-14D show the results of affinity-matured antibodies H5 and B13 after H5-h-7, H5-a-2, B13-c-5, and B13-e-2 blocking the binding of CD100 of different species to Plexin receptors.
  • Figure 14A shows the results of candidate antibodies blocking the binding of HuCD100 to HuPlexin-B1-HEK293 cells;
  • Figure 14B shows the results of candidate antibodies blocking the binding of HuCD100 to HuPlexin-B2-HEK293 cells;
  • Figure 14C shows the results of candidate antibodies blocking the binding of MusCD100 to MusPlexin-B1-HEK293 cells;
  • Figure 14D shows the results of candidate antibodies blocking the binding of CynoCD100 to CynoPlexin-B1-HEK293 cells.
  • FIG. 15 shows the results of MDSC proliferation inhibition assays of the candidate antibodies H5 and affinity-matured modified antibodies H5-h-7, H5-a-2, B13-c-5, and B13-e-2.
  • Figures 16A and 16B show the results of tumor inhibition effect detection.
  • Figure 16A shows the results of tumor volume change;
  • Figure 16B shows the results of body weight change.
  • At least one or “one or more” may mean 1, 2, 3, 4, 5, 6, 7, 8 or more.
  • the expressions “comprises,””comprising,””containing,” and “having” are open ended, meaning the inclusion of the listed elements, steps, or components but not the exclusion of other unlisted elements, steps, or components.
  • the expression “consisting of” excludes any element, step, or component not specified.
  • the expression “consisting essentially of” means that the scope is limited to the specified elements, steps, or components, plus optional elements, steps, or components that do not significantly affect the basic and novel properties of the claimed subject matter. It should be understood that the expressions “consisting essentially of” and “consisting of” are encompassed within the meaning of the expression “comprising.”
  • connection term "and/or” between multiple mentioned elements should be understood to include individual and combined options.
  • “and/or” includes “and” and “or”.
  • a and/or B includes A, B and A+B.
  • A, B and/or C include A, B, C and any combination thereof, such as A+B, A+C, B+C and A+B+C. More elements defined by “and/or” are understood in a similar manner and include any one of them and any combination thereof.
  • any numerical value or numerical range such as concentration or concentration range, is understood to be modified by the term "about” in any case. Therefore, numerical values generally include ⁇ 10% of the value. For example, a concentration of 1 mg/mL includes 0.9 mg/mL to 1.1 mg/mL. Similarly, a concentration range of 1% to 10% (w/v) includes 0.9% (w/v) to 11% (w/v). As used herein, the use of numerical ranges explicitly includes all possible subranges, all individual numerical values within the range, including integers and fractions within the range, unless the context clearly indicates otherwise.
  • antibody refers to an immunoglobulin or a fragment thereof, which specifically binds to an antigenic epitope through at least one antigen binding site. Antibodies encompass antibody fragments.
  • the term “antibody” includes synthetic antibodies, recombinantly produced antibodies, multispecific antibodies (e.g., bispecific antibodies), human antibodies, non-human antibodies, humanized antibodies, chimeric antibodies, intracellular antibodies, and antibody fragments, such as but not limited to Fab fragments, Fab' fragments, F(ab') 2 fragments, Fv fragments, disulfide-linked Fv (dsFv), Fd fragments, Fd' fragments, single-chain Fv (scFv), single-chain Fab (scFab), double antibodies, anti-idiotype (anti-Id) antibodies, or antigen-binding fragments of any of the above antibodies.
  • Antibodies provided herein include any immunoglobulin type (e.g., IgG, IgM, IgD, IgE, IgA, and IgY), any class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2) or subclass (e.g., IgG2a and IgG2b) members.
  • the antibody of the present invention is a human antibody.
  • an "antibody fragment” or “antigen-binding fragment” of an antibody refers to any portion of a full-length antibody that is less than full-length but contains at least a portion of the variable region of the antibody that binds to an antigen (e.g., one or more CDRs and/or one or more antibody binding sites), and thus retains binding specificity and at least a portion of the specific binding ability of the full-length antibody.
  • an antigen-binding fragment refers to an antibody fragment that contains an antigen-binding portion that binds to the same antigen as the antibody from which the antibody fragment was derived.
  • Antibody fragments include antibody derivatives produced by enzymatic treatment of full-length antibodies, as well as synthetically produced derivatives, such as recombinantly produced derivatives.
  • Antibodies include antibody fragments. Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab') 2 , single-chain Fv (scFv), Fv, dsFv, diabodies, Fd and Fd' fragments and other fragments, including modified fragments (see, e.g., Methods in Molecular Biology, Vol 207: Recombinant Antibodies for Cancer Therapy Methods and Protocols (2003); Chapter 1; p 3-25, Kipriyanov).
  • the fragments can include multiple chains linked together, for example, by disulfide bonds and/or by peptide linkers.
  • Antibody fragments generally contain at least or about 50 amino acids, and typically at least or about 200 amino acids.
  • Antigen-binding fragments include any antibody fragment that, when inserted into an antibody framework (e.g., by replacing the corresponding region), results in an antibody that immunospecifically binds to an antigen.
  • immunoglobulin single variable domain or “single variable domain” refers to a single variable region (variable domain) with antigen binding activity. Unlike conventional antibodies that consist of a pair of VH and VL that form a functional antigen binding unit, a single variable domain can form a functional antigen binding unit on its own.
  • a single variable domain can be derived from naturally occurring light chain-free antibodies, such as the variable domain of heavy chain of heavy-chain antibodies (VHH) of camelids (such as camels and alpacas) and the single variable domain of the new antigen receptor of sharks (IgNAR variable single-domain, VNAR), or it can be screened from a full-length antibody, such as a light chain variable domain and a heavy chain variable domain with antigen binding activity in a human antibody.
  • VHH usually contains three highly variable "complementarity determining regions (CDR)" and four relatively conserved "framework regions (FR)", and from the N-terminus to The C-terminus is linked in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4.
  • single domain antibody refers to an antibody that contains a single immunoglobulin variable domain (single variable domain) as a functional antigen-binding fragment. Similar to the variable region of a full-length antibody, a single variable domain generally contains CDR1, CDR2, and CDR3 that form an antigen-binding site and a supporting framework region. Unlike full-length antibodies that generally contain two heavy chains and two light chains, single domain antibodies generally contain a single peptide chain consisting of a single variable domain with a molecular weight of only about 15 kDa.
  • the single variable domain can be, for example, a variable domain of a heavy-chain antibody (VHH) of an alpaca, an IgNAR variable domain of a shark, or a human light-chain antibody variable domain.
  • VHH heavy-chain antibody
  • heavy-chain-only antibody and “heavy-chain antibody” are used interchangeably and in their broadest sense to refer to an antibody that lacks conventional antibody light chains and contains only one VHH and a heavy chain constant region (e.g., Fc fragment) that does not contain CH1.
  • Fc fragment generally refers to a crystallizable fragment of a conventional antibody or heavy chain antibody digested with papain.
  • the Fc fragment of IgG and heavy chain antibodies can include a portion of the hinge region, CH2 and CH3.
  • the Fc fragment can include at least a portion of the hinge region (e.g., all or part of the hinge region), CH2 and CH3.
  • a "full-length antibody” generally comprises two heavy chains (which may be indicated as H and H') and two light chains (which may be indicated as L and L').
  • Each heavy chain may be a full-length immunoglobulin heavy chain or any functional region thereof that retains antigen binding ability, and each light chain may be a full-length light chain or any functional region.
  • Each light chain comprises a "light chain variable region (VL)” and a “light chain constant region (CL)” from the N-terminus (amino acid end) to the C-terminus (carboxyl end).
  • Each heavy chain comprises a "heavy chain variable region (VH)” and a “heavy chain constant region (CH)” from the N-terminus to the C-terminus.
  • the heavy chain constant region of a full-length antibody may comprise VH-CH1-hinge region (hinge) CH2-CH3 from the N-terminus to the C-terminus.
  • Each heavy chain (H and H') is paired with a light chain (L and L', respectively).
  • Light chains are classified as ⁇ or ⁇ (Kappa or Lambda). Each heavy chain category can be combined with a ⁇ or ⁇ light chain.
  • light chains and heavy chains are covalently bound to each other, and when immunoglobulins are produced by hybridomas, B cells, or genetically engineered host cells, the "tail" portions of the two heavy chains are bound together by covalent disulfide bonds or non-covalent linkages.
  • the amino acids are from the N-terminal at the forked end of the Y configuration to the C-terminal at the bottom of each chain.
  • the basic structure of some antibodies e.g., IgG antibodies
  • the basic structure of some antibodies includes two heavy chain subunits and two light chain subunits, which are covalently linked by disulfide bonds to form a "Y" structure.
  • variable and variable are used from a functional perspective.
  • variable region of the variable light (VL) chain or the variable heavy (VH) chain portion determines antigen recognition and specificity.
  • constant regions of the light chain (CL) and the heavy chain (CH1, CH2 or CH3) confer biological properties, such as secretion, transplacental movement, Fc receptor binding, complement binding, etc.
  • the numbering of the constant region domains increases as they move away from the antigen binding site or the N-terminus of the antibody.
  • the N-terminal portion is the variable region, and the C-terminal portion is the constant region; the CH3 and CL domains actually include the carboxyl termini of the heavy and light chains, respectively.
  • variable region i.e., "binding domain” allows the binding molecule to selectively recognize an epitope on an antigen and specifically bind to the epitope. That is, the VL domain and the VH domain of a binding molecule, such as an antibody, or a subset of these complementary determining regions (CDRs) combine to form a variable region that determines a three-dimensional antigen binding site. More specifically, the antigen binding site is determined by three CDRs on each VH and VL chain.
  • CDRs complementary determining regions
  • the remaining amino acids in the binding domain are called "framework (FR)" regions, which show smaller intermolecular bonds. Difference.
  • the binding domain formed by the positioned CDRs determines a surface complementary to the epitope on the immunoreactive antigen. This complementary surface promotes the non-covalent binding of the antibody to its complementary epitope.
  • the amino acids of the CDRs and framework regions respectively composed in any given heavy or light chain variable region can be identified by conventional methods (see, "Sequences of Proteins of Immunological Interest", Kabat, E. et al., U.S. Department of Health and Human Services, (1983); and Chothia and Lesk, J. Mol. Biol., 196: 901-917 (1987), which are incorporated herein by reference in their entirety).
  • the CDRs (CDRL or LCDR) of the light chain variable region can be referred to as LCDR1, LCDR2 and LCDR3, and the CDRs (CDRH or HCDR) of the heavy chain variable region can be referred to as HCDR1, HCDR2 and HCDR3.
  • the amino acid sequences of CDRs are shown according to the AbM definition rules (the sequences in the claims of the present invention are also shown according to the AbM definition rules).
  • the CDR of an antibody can be defined by a variety of methods in the art, such as Chothia based on the three-dimensional structure of the antibody and the topology of the CDR loop (see, for example, Chothia, C. et al., Nature, 342, 877-883 (1989); and Al-Lazikani, B. et al., J. Mol. Biol., 273, 927-948 (1997)), Kabat based on antibody sequence variability (see, for example, Kabat, E.A.
  • the scope of the antibodies also covers antibodies whose variable region sequences comprise the specific CDR sequences but whose declared CDR boundaries differ from the specific CDR boundaries defined herein due to the application of a different scheme (e.g., a different assignment system rule or combination).
  • framework region and “framework region” can be used interchangeably.
  • framework region refers to those amino acid residues in the antibody variable region other than the CDR sequences defined above.
  • the "Fv" fragment composed of a VH and a VL through non-covalent interaction is the smallest antigen-binding fragment containing an antigen-binding site.
  • a single variable domain also has antigen-binding ability.
  • VH and VL can be connected by a peptide linker to obtain a "single-chain Fv (scFv)".
  • a "disulfide-stabilized Fv (dsFv)” or a “single-chain disulfide-stabilized Fv (scdsFv or dsscFv)” can be obtained respectively.
  • a Fab fragment is an antibody fragment obtained by digesting a full-length immunoglobulin with papain, or a fragment having the same structure produced by, for example, recombinant synthesis.
  • VL-CL an antibody heavy chain variable region and a heavy chain constant region
  • Fd an antibody heavy chain constant region
  • Single-chain “Fab (scFab)” can be obtained by linking CL and CH1 in “Fab” with a peptide linker.
  • F(ab') 2 is an antibody fragment resulting from digestion of immunoglobulin with pepsin at pH 4.0-4.5, or a fragment having the same structure produced by, for example, recombinant synthesis.
  • Fab' is half of F(ab') 2 , which can be obtained by reducing the disulfide bonds in the hinge region of F(ab') 2 .
  • hinge region includes the portion of the heavy chain molecule that connects the CH1 domain and the CH2 domain.
  • the hinge region contains approximately 25 amino acids and is flexible, thus allowing the two N-terminal antigen binding regions to move independently.
  • disulfide bond includes a covalent bond formed between two sulfur atoms.
  • the amino acid cysteine contains a sulfhydryl group that can form a disulfide bond or bridge a second sulfhydryl group.
  • chimeric antibody refers to an antibody in which the immunoreactive region or site is obtained or derived from a first species, while the constant region (which may be complete, partial or modified) is obtained from a second species.
  • the target binding region or site will be from a non-human source (e.g., mouse or primate) and the constant region is human.
  • humanized antibody refers to an antibody in which a non-human antibody is modified to increase sequence homology with a human antibody.
  • Humanized antibodies generally retain the antigen binding ability of the non-human antibody from which they are derived and have lower immunogenicity to the human body.
  • Humanized antibodies can be obtained by engineering any non-human species antibody or an antibody (e.g., a chimeric antibody) comprising a sequence of a non-human species.
  • Non-human species may, for example, include mice, rats, rabbits, alpacas, sharks, or non-human primates. The technology of obtaining humanized antibodies from non-human antibodies is well known to those skilled in the art.
  • the CDR sequence of a non-human antibody is transplanted into a human antibody framework region.
  • a non-human antibody e.g., a mouse antibody
  • the key amino acid residues of the non-human antibody (e.g., mouse antibody) framework sequence can be retained in the human antibody framework region, i.e., "back mutation" is performed (see, e.g., Morrison et al. (1984) Proc. Natl. Acad. Sci. 81(21): 6851-6855; Neuberger et al. (1984) Nature 312: 604-608).
  • human antibody refers to an antibody produced by a human or an antibody prepared using any technique known in the art having an amino acid sequence corresponding to an antibody produced by a human.
  • the definition of a human antibody encompasses complete or full-length antibodies, fragments thereof, and/or antibodies comprising at least one human heavy chain and/or light chain polypeptide.
  • an "affinity matured" antibody comprises one or more modifications (e.g., substitutions of amino acid residues) in one or more CDRs such that the affinity matured antibody has improved affinity for the antigen compared to a parent antibody that does not comprise such modifications.
  • modifications e.g., substitutions of amino acid residues
  • Methods for affinity maturation of antibodies are known in the art, see, e.g., Marks et al., Bio/Technology 10:779-783 (1992); Barbas et al., Proc. Nat. Acad. Sci. USA 91:3809-3813 (1994); Scier et al., Gene 169:147-155 (1995); and Hawkins et al., J. Mol. Biol. 226:889-896 (1992).
  • percent (%) sequence identity and “sequence identity” of amino acid sequences have a definition recognized in the art, which refers to the percentage of identity between two polypeptide sequences determined by sequence alignment (e.g., by manual inspection or a publicly known algorithm). It can be determined using methods known to those skilled in the art, such as using publicly available computer software such as BLAST, BLAST-2, Clustal Omega and FASTA software.
  • an amino acid sequence "derived from” or “derived from” a reference amino acid sequence is identical or homologous to part or all of the reference amino acid sequence.
  • an amino acid sequence derived from the heavy chain constant region of a human immunoglobulin may have at least 80%, at least 85%, at least 90%, or at least 100% similarity to the wild-type sequence of the heavy chain constant region of a human immunoglobulin from which it is derived. At least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% sequence identity.
  • Affinity or "binding affinity” is a measure of the strength of the non-covalent binding between an antibody and an antigen. Affinity can be determined using conventional techniques known in the art, such as biofilm interferometry (e.g., using the Octet Fortebio assay system), radioimmunoassay, surface plasmon resonance, enzyme-linked immunosorbent assay (ELISA), or flow cytometry (FACS).
  • biofilm interferometry e.g., using the Octet Fortebio assay system
  • radioimmunoassay e.g., using the Octet Fortebio assay system
  • ELISA enzyme-linked immunosorbent assay
  • FACS flow cytometry
  • Specific binding generally refers to a binding molecule, such as an antibody or its fragment, variant or derivative, which binds to an epitope through its antigen binding domain, and the binding requires some complementarity between the antigen binding domain and the epitope.
  • a binding molecule binds to an epitope through its antigen binding domain more easily than it binds to a random, unrelated epitope
  • the binding molecule is said to "specifically bind” the epitope.
  • the term “specificity” is used herein to qualitatively analyze the relative affinity of a certain antibody in combination with a certain epitope.
  • binding molecule "A” has a higher specificity to a given epitope than binding molecule "B”, or it can be said that binding molecule "A” specifically binds to an epitope "C” with a higher specificity than its related epitope "D".
  • a binding molecule e.g., an antibody or fragment, variant, or derivative thereof, preferentially binds to an epitope to the extent that it blocks the binding of a reference antibody or antigen-binding fragment to the epitope
  • the binding molecule e.g., an antibody or fragment, variant, or derivative thereof
  • competitive inhibition can be determined by any method known in the art, e.g., a competition ELISA assay. It can be said that the binding molecule competitively inhibits at least 90%, at least 80%, at least 70%, at least 60%, or at least 50% of the binding of a reference antibody or antigen-binding fragment to a given epitope.
  • PD-L1 refers to programmed cell death ligand 1 (PD-L1, see, e.g., Freeman et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000 Oct 2; 192(7)).
  • PD-L1 belongs to the B7 family.
  • Alternative names or synonyms of PD-L1 include PDCD1L1, PDL1, B7 homolog 1 (B7-H1), surface antigen cluster of differentiation 274 (cluster of differentiation 274, CD274) or B7-H, etc.
  • a representative amino acid sequence of human PD-L1 is disclosed in NCBI accession number NP_054862.1, and a representative nucleic acid sequence encoding human PD-L1 is shown under NCBI accession number: NM_014143.4.
  • PD-L1 is expressed in the placenta, spleen, lymph nodes, thymus, heart, fetal liver, and is also found in many tumors or cancer cells.
  • PD-L1 binds to its receptor PD-1 or B7-1, which is expressed on activated T cells, B cells, and bone marrow cells. The binding of PD-L1 to its receptor induces signal transduction to inhibit TCR-mediated activation of cytokine production and T cell proliferation. Therefore, PD-L1 plays a major role in suppressing the immune system during specific events (e.g., pregnancy, autoimmune diseases, tissue allografts), and is thought to allow tumors or cancer cells to bypass immune checkpoints and escape immune responses.
  • specific events e
  • B7 family refers to a class of co-stimulatory factors in the immune process of the body with similar structures. It belongs to the immunoglobulin class and is related to the activation of T and B cells and the body's immunity.
  • polynucleotide and “nucleic acid” are used interchangeably to refer to a polymer of deoxyribonucleotides (deoxyribonucleic acid, DNA) or a polymer of ribonucleotides (ribonucleic acid, RNA).
  • Polynucleotide sequence and “nucleotide sequence” are used interchangeably to refer to the order of nucleotides in a polynucleotide.
  • DNA coding strand sense strand
  • RNA it encodes can be considered to have the same nucleotide sequence, and the deoxythymidylic acid in the DNA coding strand sequence corresponds to the uridine acid in the RNA sequence it encodes.
  • an isolated nucleic acid molecule is one that is separated from other nucleic acid molecules that are present in the natural source of the nucleic acid molecule.
  • isolated nucleic acid molecules such as cDNA molecules can be substantially free of other cellular materials or culture media when prepared by recombinant technology, or substantially free of chemical precursors or other chemical components when chemically synthesized.
  • Exemplary isolated nucleic acid molecules provided herein include isolated nucleic acid molecules encoding the antibody or Fab provided.
  • expression includes transcription and/or translation of a nucleotide sequence. Thus, expression may involve the production of transcripts and/or polypeptides.
  • vector is a medium for introducing exogenous polynucleotides into host cells, and when the vector is transformed into an appropriate host cell, the exogenous polynucleotides are amplified or expressed.
  • the vector usually remains free, but can be designed to integrate a gene or part thereof into a chromosome of the genome.
  • the definition of vector encompasses plasmids, linearized plasmids, viral vectors, cosmids, phage vectors, phagemids, artificial chromosomes (e.g., yeast artificial chromosomes and mammalian artificial chromosomes), etc.
  • Viral vectors include, but are not limited to, retroviral vectors (including lentiviral vectors), adenoviral vectors, adeno-associated viral vectors, herpes virus vectors, poxvirus vectors, and baculovirus vectors, etc.
  • host cell is a cell used to receive, maintain, replicate and amplify a vector. Host cells can also be used to express polypeptides encoded by the vector. When the host cell divides, the nucleic acid contained in the vector is replicated, thereby amplifying the nucleic acid.
  • the host cell can be a eukaryotic cell or a prokaryotic cell. Suitable host cells include, but are not limited to, CHO cells, various COS cells, HeLa cells, HEK cells such as HEK 293 cells.
  • treat or “treat” or “treat” or “alleviate” or “alleviate” refer to therapeutic measures that cure, alleviate, reduce the symptoms of an existing diagnosed pathological condition or disorder, and/or arrest or slow the progression of an existing diagnosed pathological condition or disorder.
  • Terms such as “prevent,” “prevent,” “avoid,” “contain,” and the like refer to preventative or prophylactic measures that prevent the progression of an undiagnosed target pathological condition or disorder.
  • a subject in need thereof may include a subject already suffering from a disease; a subject susceptible to a disease; and a subject in need of prevention of a disease.
  • therapeutic effect refers to the effect resulting from treatment of a subject that alters, typically ameliorates or improves the symptoms of a disease or condition, or cures the disease or condition.
  • a therapeutically effective amount refers to an amount of an antibody, polypeptide, polynucleotide, small organic molecule or other drug that is effective for "treating" a disease or condition in a subject or mammal.
  • a therapeutically effective amount of a drug can reduce the number of cancer cells; block or stop cancer cell division, reduce or block the increase in tumor size; inhibit, for example, suppress, block, prevent, stop, delay or reverse cancer cell infiltration to peripheral organs, including, for example, cancer spread to soft tissue and bone; inhibit, for example, suppress, block, prevent, shrink, stop, delay or reverse tumor metastasis; inhibit, for example, suppress, block, prevent, stop, delay or reverse tumor growth; alleviate one or more symptoms associated with cancer to some extent, reduce morbidity and mortality; improve quality of life; or a combination of these effects.
  • the drug prevents growth and/or kills existing cancer cells, it can refer to cell growth inhibition and/or cytotoxicity.
  • the term "subject" refers to a mammal, such as a human.
  • the antibody numbers used herein are only used to distinguish or identify antibodies or products, and are not intended to indicate that such identification is a feature of the antibody or product of the present invention. It should be understood by those skilled in the art that, for example, for the purpose of distinction or identification, other antibodies or products may also use such identification, but it does not refer to the same or equivalent antibodies or products. Similarly, the similar numbers or identifications used in the embodiments are Also for the convenience of example only, the antibody or product of the invention is defined by the features described in the appended claims.
  • the present invention provides an antibody against CD100 (anti-CD100 antibody) or an antigen-binding fragment thereof, wherein the antibody or the antigen-binding fragment thereof specifically recognizes and binds to CD100.
  • the anti-CD100 antibody or antigen-binding fragment thereof of the present invention is a chimeric antibody, a humanized antibody, a human antibody, a scFv, a Fab, a Fab', a F(ab') 2 , a Fv fragment, a disulfide-stabilized Fv (dsFv) or a diabody.
  • the antibody or antigen-binding fragment thereof is a human antibody.
  • the antibody or antigen-binding fragment thereof has at least one of the following characteristics:
  • the antibodies or antigen-binding fragments thereof of the present invention can specifically bind to CD100 (eg, human CD100) and block its interaction with Plexin-B1 or Plexin-B2.
  • the tumors targeted include but are not limited to those described below for neoplastic diseases.
  • the antibodies or antigen-binding fragments thereof of the present invention can inhibit tumor growth by at least about 10%, preferably at least about 20%, more preferably at least about 30%, more preferably at least about 40%, more preferably at least about 50%, more preferably at least about 60%, more preferably at least about 70%, more preferably at least about 80%.
  • the antibody or antigen-binding fragment thereof against CD100 of the present invention comprises a heavy chain variable region, wherein the heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 sequences, and wherein the HCDR1 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:1, 7, 19, 33, 41, 47 or 52 by no more than 2 amino acids in addition, deletion or substitution; the HCDR2 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:2, 8, 14, 20, 26, 42, 48 or 53 by no more than 2 amino acids in addition, deletion or substitution; and/or the HCDR3 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:3, 9, 15, 21, 27, 30, 35, 38, 43, 49 or 54 by no more than 2 amino acids in addition, deletion or substitution.
  • the antibody or antigen-binding fragment thereof against CD100 of the present invention comprises a light chain variable region, wherein the light chain variable region comprises LCDR1, LCDR2 and LCDR3 sequences, and wherein the LCDR1 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:4, 10, 16, 36, 44 or 50 by no more than 2 amino acids in addition, deletion or substitution; the LCDR2 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:5, 11 or 45 by no more than 2 amino acids in addition, deletion or substitution; and/or the LCDR3 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:6, 12, 18, 24, 37, 46, 51 or 56 by no more than 2 amino acids in addition, deletion or substitution.
  • the antibody or antigen-binding fragment thereof against CD100 of the present invention comprises a heavy chain variable region and a light chain variable region, wherein
  • the heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 sequences, wherein the HCDR1 sequence has no more than 2 amino acid sequences identical to the sequence shown in SEQ ID NO: 1, 7, 19, 33, 41, 47 or 52.
  • the HCDR2 sequence has no more than 2 amino acid additions, deletions or substitutions from the sequence shown in SEQ ID NO: 2, 8, 14, 20, 26, 42, 48 or 53; and/or the HCDR3 sequence has no more than 2 amino acid additions, deletions or substitutions from the sequence shown in SEQ ID NO: 3, 9, 15, 21, 27, 30, 35, 38, 43, 49 or 54; and
  • the light chain variable region comprises LCDR1, LCDR2 and LCDR3 sequences, and wherein the LCDR1 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:4, 10, 16, 36, 44 or 50 by no more than 2 amino acids in addition, deletion or substitution; the LCDR2 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:5, 11 or 45 by no more than 2 amino acids in addition, deletion or substitution; and/or the LCDR3 sequence differs in amino acid sequence from the sequence shown in SEQ ID NO:6, 12, 18, 24, 37, 46, 51 or 56 by no more than 2 amino acids in addition, deletion or substitution.
  • the HCDR1 sequence comprises the amino acid sequence set forth in SEQ ID NO:7 (SEQ ID NO:7; GFTFSSYSMN), wherein the amino acid at position 3 is substituted. In one embodiment, the HCDR1 sequence comprises the amino acid sequence set forth in SEQ ID NO:7 (SEQ ID NO:7; GFTFSSYSMN), wherein the amino acid at position 8 is substituted. In one embodiment, the HCDR1 sequence comprises the amino acid sequence set forth in SEQ ID NO:7 (SEQ ID NO:7; GFTFSSYSMN), wherein the amino acid at position 3 and the amino acid at position 8 are substituted. In one embodiment, the amino acid at position 3 in SEQ ID NO:7 is substituted with P, and the amino acid at position 8 in SEQ ID NO:7 is substituted with E (SEQ ID NO:13; GFPFSSYEMN).
  • the HCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:1 (SEQ ID NO:1; SGSFSGYYWT), wherein the amino acid at position 1 is substituted. In one embodiment, the HCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:1 (SEQ ID NO:1; SGSFSGYYWT), wherein the amino acid at position 10 is substituted. In one embodiment, the HCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:1 (SEQ ID NO:1; SGSFSGYYWT), wherein the amino acid at position 1 and the amino acid at position 10 are substituted. In one embodiment, the amino acid at position 1 in SEQ ID NO:1 is substituted with G, and the amino acid at position 10 in SEQ ID NO:1 is substituted with S (SEQ ID NO:25; GGSFSGYYWS).
  • the HCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:33 (SEQ ID NO:33; GGSISSSNWWS), wherein the amino acid at position 6 is substituted. In one embodiment, the amino acid at position 6 in SEQ ID NO:33 is substituted with G (SEQ ID NO:57; GGSISGSNWWS).
  • the HCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:26 (SEQ ID NO:26; EINHSGSTN), wherein the amino acid at position 3 is substituted. In one embodiment, the amino acid at position 3 in SEQ ID NO:26 is substituted with Y (SEQ ID NO:34; EIYHSGSTN).
  • the HCDR2 sequence comprises the amino acid sequence set forth in SEQ ID NO:26 (SEQ ID NO:26; EINHSGSTN), wherein the amino acid at position 3 is substituted. In one embodiment, the HCDR2 sequence comprises the amino acid sequence set forth in SEQ ID NO:26 (SEQ ID NO:26; EINHSGSTN), wherein the amino acid at position 7 is substituted. In one embodiment, the HCDR2 sequence comprises the amino acid sequence set forth in SEQ ID NO:26 (SEQ ID NO:26; EINHSGSTN), wherein the amino acid at position 3 and the amino acid at position 7 are substituted. In one embodiment, the amino acid at position 3 in SEQ ID NO:26 is substituted with Y, and the amino acid at position 7 in SEQ ID NO:26 is substituted with E (SEQ ID NO:58; EIYHSGETN).
  • the HCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO: 35 (SEQ ID NO:35; YDFWSGSGLDY), wherein the amino acid at position 4 is substituted. In one embodiment, the amino acid at position 4 in SEQ ID NO:35 is substituted with E (SEQ ID NO:60; YDFESGSGLDY).
  • the HCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:3 (SEQ ID NO:3; GPAYYADALDGFDI), wherein the amino acid at position 6 is substituted. In one embodiment, the amino acid at position 6 in SEQ ID NO:3 is substituted with P (SEQ ID NO:63; GPAYYPDALDGFDI).
  • the HCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:3 (SEQ ID NO:3; GPAYYADALDGFDI), wherein the amino acid at position 5 is substituted. In one embodiment, the HCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:3 (SEQ ID NO:3; GPAYYADALDGFDI), wherein the amino acid at position 6 is substituted. In one embodiment, the HCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:3 (SEQ ID NO:3; GPAYYADALDGFDI), wherein the amino acid at position 5 and the amino acid at position 6 are substituted. In one embodiment, the amino acid at position 5 in SEQ ID NO:3 is substituted with L, and the amino acid at position 6 in SEQ ID NO:3 is substituted with P (SEQ ID NO:66; GPAYLPDALDGFDI).
  • the LCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO: 16 (SEQ ID NO: 16; SGDKLGDKYAY), wherein the amino acid at position 10 is substituted. In one embodiment, the amino acid at position 10 in SEQ ID NO: 16 is substituted with V (SEQ ID NO: 22; SGDKLGDKYVY).
  • the LCDR1 sequence comprises the amino acid sequence set forth in SEQ ID NO:16 (SEQ ID NO:16; SGDKLGDKYAY), wherein the amino acid at position 7 is substituted. In one embodiment, the LCDR1 sequence comprises the amino acid sequence set forth in SEQ ID NO:16 (SEQ ID NO:16; SGDKLGDKYAY), wherein the amino acid at position 11 is substituted. In one embodiment, the LCDR1 sequence comprises the amino acid sequence set forth in SEQ ID NO:16 (SEQ ID NO:16; SGDKLGDKYAY), wherein the amino acid at position 7 and the amino acid at position 11 are substituted. In one embodiment, the amino acid at position 7 in SEQ ID NO:16 is substituted with E, and the amino acid at position 11 in SEQ ID NO:16 is substituted with F (SEQ ID NO:31; SGDKLGEKYAF).
  • the LCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:16 (SEQ ID NO:16; SGDKLGDKYAY), wherein the amino acid at position 4 is substituted. In one embodiment, the LCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:16 (SEQ ID NO:16; SGDKLGDKYAY), wherein the amino acid at position 10 is substituted. In one embodiment, the LCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:16 (SEQ ID NO:16; SGDKLGDKYAY), wherein the amino acid at position 4 and the amino acid at position 10 are substituted. In one embodiment, the amino acid at position 4 in SEQ ID NO:16 is substituted with R, and the amino acid at position 10 in SEQ ID NO:16 is substituted with S (SEQ ID NO:39; SGDRLGDKYSY).
  • the LCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:36 (SEQ ID NO:36; SGDKLGDKFAS), wherein the amino acid at position 7 is substituted.
  • the amino acid at position 7 in SEQ ID NO:36 is substituted with E (SEQ ID NO:61; SGDKLGEKFAS).
  • the LCDR1 sequence comprises the amino acid sequence shown in SEQ ID NO:4 (SEQ ID NO:4; SGDKLGDKYVS), wherein the amino acid at position 6 is substituted.
  • the amino acid at position 6 in SEQ ID NO:4 is substituted with Q (SEQ ID NO:64; SGDKLQDKYVS).
  • the LCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:5 (SEQ ID NO:5; QDNKRPS), wherein the amino acid at position 3 is substituted. In one embodiment, the amino acid at position 3 in SEQ ID NO:5 is substituted with S (SEQ ID NO:17; QDSKRPS).
  • the LCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:5 (SEQ ID NO:5; QDNKRPS), wherein the amino acid at position 3 is substituted.
  • the amino acid at position 3 in SEQ ID NO:5 is substituted with R (SEQ ID NO:23; QDRKRPS).
  • the LCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:5 (SEQ ID NO:5; QDNKRPS), wherein the amino acid at position 3 is substituted.
  • the amino acid at position 3 in SEQ ID NO:5 is substituted with A (SEQ ID NO:28; QDAKRPS).
  • the LCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:11 (SEQ ID NO:11; AASSLQS), wherein the amino acid at position 1 is substituted. In one embodiment, the LCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:11 (SEQ ID NO:11; AASSLQS), wherein the amino acid at position 5 is substituted. In one embodiment, the LCDR2 sequence comprises the amino acid sequence shown in SEQ ID NO:11 (SEQ ID NO:11; AASSLQS), wherein the amino acid at position 1 and the amino acid at position 5 are substituted. In one embodiment, the amino acid at position 1 in SEQ ID NO:11 is substituted with T, and the amino acid at position 5 in SEQ ID NO:11 is substituted with V (SEQ ID NO:55; TASSVQS).
  • the LCDR2 sequence comprises the amino acid sequence set forth in SEQ ID NO:5 (SEQ ID NO:5; QDNKRPS), wherein the amino acid at position 3 is substituted. In one embodiment, the LCDR2 sequence comprises the amino acid sequence set forth in SEQ ID NO:5 (SEQ ID NO:5; QDNKRPS), wherein the amino acid at position 7 is substituted. In one embodiment, the LCDR2 sequence comprises the amino acid sequence set forth in SEQ ID NO:5 (SEQ ID NO:5; QDNKRPS), wherein the amino acid at position 3 and the amino acid at position 7 are substituted. In one embodiment, the amino acid at position 3 in SEQ ID NO:5 is substituted with R, and the amino acid at position 7 in SEQ ID NO:5 is substituted with N (SEQ ID NO:59; QDRKRPN).
  • the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:6 (SEQ ID NO:6; QAWDSSTKAYV), wherein the amino acid at position 8 is substituted. In one embodiment, the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:6 (SEQ ID NO:6; QAWDSSTKAYV), wherein the amino acid at position 9 is substituted. In one embodiment, the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:6 (SEQ ID NO:6; QAWDSSTKAYV), wherein the amino acid at position 8 and the amino acid at position 9 are substituted. In one embodiment, the amino acid at position 8 in SEQ ID NO:6 is substituted with A, and the amino acid at position 9 in SEQ ID NO:6 is substituted with G (SEQ ID NO:29; QAWDSSTAGYV).
  • the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO: 18 (SEQ ID NO: 18; QAWDSSTV), wherein an amino acid is added between amino acid position 7 and amino acid position 8.
  • the amino acid added between amino acid position 7 and amino acid position 8 in SEQ ID NO: 18 is Y (SEQ ID NO: 32; QAWDSSTYV).
  • the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO: 18 (SEQ ID NO: 18; QAWDSSTV), wherein the amino acid at position 2 is substituted. In one embodiment, the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO: 18 (SEQ ID NO: 18; QAWDSSTV), wherein an amino acid is added between the amino acid at position 7 and the amino acid at position 8. In one embodiment, the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO: 18 (SEQ ID NO: 18; QAWDSSTV), wherein the amino acid at position 2 is substituted, and an amino acid is added between the amino acid at position 7 and the amino acid at position 8.
  • amino acid at position 2 in SEQ ID NO: 18 is substituted with V, and the amino acid added between the amino acid at position 7 and the amino acid at position 8 in SEQ ID NO: 18 is A (SEQ ID NO: 40; QVWDSSTAV).
  • the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO: 37 (SEQ ID NO:37; QAWDSGTVI), wherein the amino acid at position 8 is substituted. In one embodiment, the amino acid at position 8 in SEQ ID NO:37 is substituted with A (SEQ ID NO:62; QAWDSGTAI).
  • the LCDR3 sequence comprises the amino acid sequence shown in SEQ ID NO:6 (SEQ ID NO:6; QAWDSSTKAYV), wherein the amino acid at position 6 is substituted.
  • the amino acid at position 6 in SEQ ID NO:6 is substituted with E (SEQ ID NO:65; QAWDSETKAYV).
  • the heavy chain variable region comprises a HCDR1 sequence shown in SEQ ID NO: 1, 7, 13, 19, 25, 33, 41, 47, 52 or 57; a HCDR2 sequence shown in SEQ ID NO: 2, 8, 14, 20, 26, 34, 42, 48, 53 or 58; and a HCDR3 sequence shown in SEQ ID NO: 3, 9, 15, 21, 27, 30, 35, 38, 43, 49, 54, 60, 63 or 66.
  • the light chain variable region comprises a LCDR1 sequence shown in SEQ ID NO:4, 10, 16, 22, 31, 36, 39, 44, 50, 61 or 64; a LCDR2 sequence shown in SEQ ID NO:5, 11, 17, 23, 28, 45, 55 or 59; and a LCDR3 sequence shown in SEQ ID NO:6, 12, 18, 24, 29, 32, 37, 40, 46, 51, 56, 62 or 65.
  • the heavy chain variable region comprises a HCDR1 sequence shown in SEQ ID NO: 1, 7, 13, 19, 25, 33, 41, 47, 52 or 57; a HCDR2 sequence shown in SEQ ID NO: 2, 8, 14, 20, 26, 34, 42, 48, 53 or 58; and a HCDR3 sequence shown in SEQ ID NO: 3, 9, 15, 21, 27, 30, 35, 38, 43, 49, 54, 60, 63 or 66; and
  • the light chain variable region comprises a LCDR1 sequence shown in SEQ ID NO:4, 10, 16, 22, 31, 36, 39, 44, 50, 61 or 64; a LCDR2 sequence shown in SEQ ID NO:5, 11, 17, 23, 28, 45, 55 or 59; and a LCDR3 sequence shown in SEQ ID NO:6, 12, 18, 24, 29, 32, 37, 40, 46, 51, 56, 62 or 65.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO: 1, the HCDR2 sequence shown in SEQ ID NO: 2, and the HCDR3 sequence shown in SEQ ID NO: 3.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO: 7, the HCDR2 sequence shown in SEQ ID NO: 8, and the HCDR3 sequence shown in SEQ ID NO: 9.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO: 13, the HCDR2 sequence shown in SEQ ID NO: 14, and the HCDR3 sequence shown in SEQ ID NO: 15.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO: 19, the HCDR2 sequence shown in SEQ ID NO: 20, and the HCDR3 sequence shown in SEQ ID NO: 21. In one embodiment, the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO: 25, the HCDR2 sequence shown in SEQ ID NO: 26, and the HCDR3 sequence shown in SEQ ID NO: 27. In one embodiment, the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:25, the HCDR2 sequence shown in SEQ ID NO:26, and the HCDR3 sequence shown in SEQ ID NO:30.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:33, the HCDR2 sequence shown in SEQ ID NO:34, and the HCDR3 sequence shown in SEQ ID NO:35. In one embodiment, the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:33, the HCDR2 sequence shown in SEQ ID NO:34, and the HCDR3 sequence shown in SEQ ID NO:38. In one embodiment, the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:41, the HCDR2 sequence shown in SEQ ID NO:42, and the HCDR3 sequence shown in SEQ ID NO:43.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:47, the HCDR2 sequence shown in SEQ ID NO:48, and the HCDR3 sequence shown in SEQ ID NO:49.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO: 52, the HCDR2 sequence shown in SEQ ID NO: 53 and the NO:54.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:57, the HCDR2 sequence shown in SEQ ID NO:58 and the HCDR3 sequence shown in SEQ ID NO:35.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:33, the HCDR2 sequence shown in SEQ ID NO:34 and the HCDR3 sequence shown in SEQ ID NO:60. In one embodiment, the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:1, the HCDR2 sequence shown in SEQ ID NO:2 and the HCDR3 sequence shown in SEQ ID NO:63. In one embodiment, the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:1, the HCDR2 sequence shown in SEQ ID NO:2 and the HCDR3 sequence shown in SEQ ID NO:66.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:4, the LCDR2 sequence shown in SEQ ID NO:5, and the LCDR3 sequence shown in SEQ ID NO:6.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:10, the LCDR2 sequence shown in SEQ ID NO:11, and the LCDR3 sequence shown in SEQ ID NO:12.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:16, the LCDR2 sequence shown in SEQ ID NO:17, and the LCDR3 sequence shown in SEQ ID NO:18.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:22, the LCDR2 sequence shown in SEQ ID NO:23, and the LCDR3 sequence shown in SEQ ID NO:24. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:16, the LCDR2 sequence shown in SEQ ID NO:28, and the LCDR3 sequence shown in SEQ ID NO:29. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:31, the LCDR2 sequence shown in SEQ ID NO:5, and the LCDR3 sequence shown in SEQ ID NO:32.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:36, the LCDR2 sequence shown in SEQ ID NO:23, and the LCDR3 sequence shown in SEQ ID NO:37. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:39, the LCDR2 sequence shown in SEQ ID NO:17, and the LCDR3 sequence shown in SEQ ID NO:40. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:44, the LCDR2 sequence shown in SEQ ID NO:45, and the LCDR3 sequence shown in SEQ ID NO:46.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:50, the LCDR2 sequence shown in SEQ ID NO:11, and the LCDR3 sequence shown in SEQ ID NO:51. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:50, the LCDR2 sequence shown in SEQ ID NO:55, and the LCDR3 sequence shown in SEQ ID NO:56. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:36, the LCDR2 sequence shown in SEQ ID NO:59, and the LCDR3 sequence shown in SEQ ID NO:37.
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO: 61, the LCDR2 sequence shown in SEQ ID NO: 23, and the LCDR3 sequence shown in SEQ ID NO: 62. In one embodiment, the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO: 64, the LCDR2 sequence shown in SEQ ID NO: 5, and the LCDR3 sequence shown in SEQ ID NO: 65.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:1, the HCDR2 sequence shown in SEQ ID NO:2 and the HCDR3 sequence shown in SEQ ID NO:3;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:4, the LCDR2 sequence shown in SEQ ID NO:5 and the LCDR3 sequence shown in SEQ ID NO:6.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:7, the HCDR2 sequence shown in SEQ ID NO:8 and the HCDR3 sequence shown in SEQ ID NO:9;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:10, the LCDR2 sequence shown in SEQ ID NO:11 and the LCDR3 sequence shown in SEQ ID NO:12.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:13, the HCDR2 sequence shown in SEQ ID NO:14 and the HCDR3 sequence shown in SEQ ID NO:15;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:16, the LCDR2 sequence shown in SEQ ID NO:17 and the LCDR3 sequence shown in SEQ ID NO:18.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:19, the HCDR2 sequence shown in SEQ ID NO:20 and the HCDR3 sequence shown in SEQ ID NO:21;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:22, the LCDR2 sequence shown in SEQ ID NO:23 and the LCDR3 sequence shown in SEQ ID NO:24.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:25, the HCDR2 sequence shown in SEQ ID NO:26 and the HCDR3 sequence shown in SEQ ID NO:27;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:16, the LCDR2 sequence shown in SEQ ID NO:28 and the LCDR3 sequence shown in SEQ ID NO:29.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:25, the HCDR2 sequence shown in SEQ ID NO:26 and the HCDR3 sequence shown in SEQ ID NO:30;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:31, the LCDR2 sequence shown in SEQ ID NO:5 and the LCDR3 sequence shown in SEQ ID NO:32.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:33, the HCDR2 sequence shown in SEQ ID NO:34 and the HCDR3 sequence shown in SEQ ID NO:35;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:36, the LCDR2 sequence shown in SEQ ID NO:23 and the LCDR3 sequence shown in SEQ ID NO:37.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:33, the HCDR2 sequence shown in SEQ ID NO:34 and the HCDR3 sequence shown in SEQ ID NO:38;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:39, the LCDR2 sequence shown in SEQ ID NO:17 and the LCDR3 sequence shown in SEQ ID NO:40.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:41, the HCDR2 sequence shown in SEQ ID NO:42 and the HCDR3 sequence shown in SEQ ID NO:43;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:44, the LCDR2 sequence shown in SEQ ID NO:45 and the LCDR3 sequence shown in SEQ ID NO:46.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:47, the HCDR2 sequence shown in SEQ ID NO:48 and the HCDR3 sequence shown in SEQ ID NO:49;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:50, the LCDR2 sequence shown in SEQ ID NO:11 and the LCDR3 sequence shown in SEQ ID NO:51.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:52, the HCDR2 sequence shown in SEQ ID NO:53 and the HCDR3 sequence shown in SEQ ID NO:54;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:50, the LCDR2 sequence shown in SEQ ID NO:55 and the LCDR3 sequence shown in SEQ ID NO:56.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:57, the HCDR2 sequence shown in SEQ ID NO:58 and the HCDR3 sequence shown in SEQ ID NO:35;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:36, the LCDR2 sequence shown in SEQ ID NO:59 and the HCDR3 sequence shown in SEQ ID NO:37. LCDR3 sequences are shown.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:33, the HCDR2 sequence shown in SEQ ID NO:34 and the HCDR3 sequence shown in SEQ ID NO:60;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:61, the LCDR2 sequence shown in SEQ ID NO:23 and the LCDR3 sequence shown in SEQ ID NO:62.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:1, the HCDR2 sequence shown in SEQ ID NO:2 and the HCDR3 sequence shown in SEQ ID NO:63;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:64, the LCDR2 sequence shown in SEQ ID NO:5 and the LCDR3 sequence shown in SEQ ID NO:65.
  • the heavy chain variable region comprises the HCDR1 sequence shown in SEQ ID NO:1, the HCDR2 sequence shown in SEQ ID NO:2 and the HCDR3 sequence shown in SEQ ID NO:66;
  • the light chain variable region comprises the LCDR1 sequence shown in SEQ ID NO:4, the LCDR2 sequence shown in SEQ ID NO:5 and the LCDR3 sequence shown in SEQ ID NO:6.
  • the heavy chain variable region comprises 1) an amino acid sequence as shown in SEQ ID NO: 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93 or 95; 2) an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, or more identical to the amino acid sequence as shown in SEQ ID NO: 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93 or 95.
  • the light chain variable region comprises 1) an amino acid sequence as shown in SEQ ID NO: 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94 or 96; 2) an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, or more identical to the amino acid sequence as shown in SEQ ID NO: 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94 or 96.
  • the heavy chain variable region comprises 1) an amino acid sequence as shown in SEQ ID NO: 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93 or 95; 2) an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92% identical to the amino acid sequence as shown in SEQ ID NO: 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93 or 95.
  • the light chain variable region comprises 1) an amino acid sequence as shown in SEQ ID NO: 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94 or 96; 2) an amino acid sequence as shown in SEQ ID NO: 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94 or 96; 80, 82, 84, 86, 88, 90, 92, 94 or 96 has an amino acid sequence that has at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity; or 3) an amino acid sequence that has one or more amino acid substitutions, additions and/or deletions compared to the amino acid sequence shown in SEQ ID NO:68, 70, 72, 74,
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:67; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:68.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:69; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:70.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:71; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:72.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:73; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:74.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:75; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:76.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:77; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:78.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:79; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:80.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:81; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:82.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:83; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:84.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:85; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:86.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:87; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:88.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:89; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:90.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:91; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:92.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:93; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:94.
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:95; the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:96.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain comprising 1) an amino acid sequence as set forth in SEQ ID NO:97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123 or 125; 2) an amino acid sequence identical to that of SEQ ID NO:97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123 or 125; 1) an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity with the amino acid sequence shown in SEQ ID NO: 117, 119, 121, 123 or 125; or 3) an amino acid sequence having one or more amino acid substitutions, additions and/or deletions compared to the amino acid
  • the antibody or antigen-binding fragment thereof comprises a light chain, wherein the light chain comprises 1) an amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126; 2) an amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126 that has at least 80%, at least 85%, at least 90% affinity, or at least 10% affinity with the amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126 0%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity;
  • the antibody or antigen-binding fragment thereof comprises a heavy chain comprising 1) an amino acid sequence as shown in SEQ ID NO: 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123 or 125; 2) an amino acid sequence that has at least 80%, at least 85%, at least 90% affinity with the amino acid sequence as shown in SEQ ID NO: 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123 or 125; %, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity; or 3) an amino acid sequence having one or more amino acid substitutions, additions and/or deletions compared to the amino acid sequence shown in SEQ ID NO:97, 99, 101, 103, 105, 107, 111
  • the antibody or its antigen-binding fragment comprises a light chain, which comprises 1) an amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126; 2) an amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126 that has at least 80%, at least 85%, at least 90%, at least 100% or more of the amino acid sequence as shown in SEQ ID NO:98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124 or 126.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:97; the light chain comprises the amino acid sequence shown in SEQ ID NO:98.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:99; the light chain comprises the amino acid sequence shown in SEQ ID NO:100.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:101; the light chain comprises the amino acid sequence shown in SEQ ID NO:102.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO: 103; the light chain comprises The amino acid sequence shown in SEQ ID NO:104.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:105; the light chain comprises the amino acid sequence shown in SEQ ID NO:106.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:107; the light chain comprises the amino acid sequence shown in SEQ ID NO:108.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:109; the light chain comprises the amino acid sequence shown in SEQ ID NO:110.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:113; the light chain comprises the amino acid sequence shown in SEQ ID NO:114.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:115; the light chain comprises the amino acid sequence shown in SEQ ID NO:116.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:117; the light chain comprises the amino acid sequence shown in SEQ ID NO:118.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:119; the light chain comprises the amino acid sequence shown in SEQ ID NO:120.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:121; the light chain comprises the amino acid sequence shown in SEQ ID NO:122.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:123; the light chain comprises the amino acid sequence shown in SEQ ID NO:124.
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:125; the light chain comprises the amino acid sequence shown in SEQ ID NO:126.
  • the anti-CD100 antibody includes anti-CD100 antibody B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5 or B13-e-2, in particular, anti-CD100 antibody B13, C-C081 or B13-c-5.
  • the present invention provides an antibody B13 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 which comprises the amino acid sequence shown in SEQ ID NO:1,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:2,
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:3;
  • the light chain variable region comprises:
  • LCDR1 which comprises the amino acid sequence shown in SEQ ID NO:4,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:5, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:6.
  • the anti-CD100 antibody B13 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:67 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:67.
  • the anti-CD100 antibody B13 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:68 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:68.
  • the anti-CD100 antibody B13 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:97 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:97.
  • the anti-CD100 antibody B13 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:98 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:98.
  • the present invention provides an antibody C-C081 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:7,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:8, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:9;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:10,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO: 11, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:12.
  • the anti-CD100 antibody C-C081 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:69 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:69.
  • the anti-CD100 antibody C-C081 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:70 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:70.
  • the anti-CD100 antibody C-C081 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:99 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:99.
  • the anti-CD100 antibody C-C081 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:100 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:100.
  • the present invention provides an antibody A14 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:13,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:14, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:15;
  • the light chain variable region comprises:
  • LCDR1 which comprises the amino acid sequence shown in SEQ ID NO:16
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:17.
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:18.
  • the anti-CD100 antibody A14 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:71 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:71.
  • the anti-CD100 antibody A14 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:72 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:72.
  • the anti-CD100 antibody A14 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:101 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:101.
  • the anti-CD100 antibody A14 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:102 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:102.
  • the present invention provides an antibody A15 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:19,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:20, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:21;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:22,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:23, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:24.
  • the anti-CD100 antibody A15 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:73 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:73.
  • the anti-CD100 antibody A15 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:74 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:74.
  • the anti-CD100 antibody A15 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:103 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:103.
  • the anti-CD100 antibody A15 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:104 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:104.
  • the present invention provides an antibody H74 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:25,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:26, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:27;
  • the light chain variable region comprises:
  • LCDR1 which comprises the amino acid sequence shown in SEQ ID NO:16
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:28, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:29.
  • the anti-CD100 antibody H74 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:75 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:75.
  • the anti-CD100 antibody H74 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:76 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:76.
  • the anti-CD100 antibody H74 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:105 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:105.
  • the anti-CD100 antibody H74 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:106 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:106.
  • the present invention provides an antibody H96 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:25,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:26, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:30;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:31,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:5, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:32.
  • the anti-CD100 antibody H96 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:77 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:77.
  • the anti-CD100 antibody H96 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:78 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:78.
  • the anti-CD100 antibody H96 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:107 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:107.
  • the anti-CD100 antibody H96 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:108 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:108.
  • the present invention provides an antibody H5 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:33,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:34, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:35;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:36,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:23, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:37.
  • the anti-CD100 antibody H5 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:79 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:79.
  • the anti-CD100 antibody H5 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:80 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:80.
  • the anti-CD100 antibody H5 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:109 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:109.
  • the anti-CD100 antibody H5 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence of SEQ ID NO:110 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:110.
  • the present invention provides an antibody H12 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:33,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:34, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:38;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:39,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:17.
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:40.
  • the anti-CD100 antibody H12 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:81 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:81.
  • the anti-CD100 antibody H12 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:82 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:82.
  • the anti-CD100 antibody H12 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:111 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:111.
  • the anti-CD100 antibody H12 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:112 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:112.
  • the present invention provides an antibody H21 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 which comprises the amino acid sequence shown in SEQ ID NO:41,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:42, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:43;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:44,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:45, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:46.
  • the anti-CD100 antibody H21 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:83 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:83.
  • the anti-CD100 antibody H21 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:84 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:84.
  • the anti-CD100 antibody H21 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:113 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:113.
  • the anti-CD100 antibody H21 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:114 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:114.
  • the present invention provides an antibody C-C171 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 which comprises the amino acid sequence shown in SEQ ID NO:47,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:48, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:49;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:50,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO: 11, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:51.
  • the anti-CD100 antibody C-C171 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:85 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:85.
  • the anti-CD100 antibody C-C171 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:86 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:86.
  • the anti-CD100 antibody C-C171 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:115 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:115.
  • the anti-CD100 antibody C-C171 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:116 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:116.
  • the present invention provides an antibody C-B71 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:52,
  • HCDR2 comprising the amino acid sequence shown in SEQ ID NO:53, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:54;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:50,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:55, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:56.
  • the anti-CD100 antibody C-B71 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:87 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:87.
  • the anti-CD100 antibody C-B71 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:88 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:88.
  • the anti-CD100 antibody C-B71 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:117 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:117.
  • the anti-CD100 antibody C-B71 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:118 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:118.
  • the present invention provides an antibody H5-h-7 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:57,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:58, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:35;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:36,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:59, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:37.
  • the anti-CD100 antibody H5-h-7 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:89 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:89.
  • the anti-CD100 antibody H5-h-7 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:90 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:90.
  • the anti-CD100 antibody H5-h-7 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:119 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:119.
  • the anti-CD100 antibody H5-h-7 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:120 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:120.
  • the present invention provides an antibody H5-a-2 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:33,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:34, and
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:60;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:61,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:23, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:62.
  • the anti-CD100 antibody H5-a-2 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:91 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:91.
  • the anti-CD100 antibody H5-a-2 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:92 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:92.
  • the anti-CD100 antibody H5-a-2 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:121 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:121.
  • the anti-CD100 antibody H5-a-2 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:122 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:122.
  • the present invention provides an antibody B13-c-5 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 which comprises the amino acid sequence shown in SEQ ID NO:1,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:2,
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:63;
  • the light chain variable region comprises:
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:64,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:5, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:65.
  • the anti-CD100 antibody B13-c-5 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 93 or has a sequence similar to SEQ ID NO: 93. Amino acid sequences having at least 85%, at least 90%, at least 95% or more sequence identity.
  • the anti-CD100 antibody B13-c-5 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:94 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:94.
  • the anti-CD100 antibody B13-c-5 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:123 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:123.
  • the anti-CD100 antibody B13-c-5 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:124 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:124.
  • the present invention provides an antibody B13-e-2 or an antigen-binding fragment thereof against CD100,
  • the antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein
  • the heavy chain variable region comprises:
  • HCDR1 which comprises the amino acid sequence shown in SEQ ID NO:1,
  • HCDR2 which comprises the amino acid sequence shown in SEQ ID NO:2,
  • HCDR3 which comprises the amino acid sequence shown in SEQ ID NO:66;
  • the light chain variable region comprises:
  • LCDR1 which comprises the amino acid sequence shown in SEQ ID NO:4,
  • LCDR2 which comprises the amino acid sequence shown in SEQ ID NO:5, and
  • LCDR3 which comprises the amino acid sequence shown in SEQ ID NO:6.
  • the anti-CD100 antibody B13-e-2 or an antigen-binding fragment thereof comprises a heavy chain variable region (VH),
  • the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:95 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:95.
  • the anti-CD100 antibody B13-e-2 or an antigen-binding fragment thereof comprises a light chain variable region (VL),
  • the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:96 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:96.
  • the anti-CD100 antibody B13-e-2 or an antigen-binding fragment thereof comprises a heavy chain
  • the heavy chain comprises the amino acid sequence shown in SEQ ID NO:125 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:125.
  • the anti-CD100 antibody B13-e-2 or an antigen-binding fragment thereof comprises a light chain
  • the light chain comprises the amino acid sequence shown in SEQ ID NO:126 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:126.
  • the present invention provides a nucleic acid molecule comprising a polynucleotide sequence encoding an anti-CD100 antibody or antigen-binding fragment thereof of the present invention.
  • the nucleic acid molecule of the present invention encodes an anti-CD100 antibody or antigen-binding fragment thereof of the present invention.
  • Nucleic acid molecules of the present invention can be obtained using methods known in the art. For example, nucleic acid molecules of the present invention can be isolated from phage display libraries, yeast display libraries, immune animals, immortalized cells (e.g., mouse B cell hybridoma cells, EBV-mediated immortalized B cells) or chemical synthesis. Nucleic acid molecules of the present invention can be codon optimized for host cells used for expression.
  • the present invention also provides an expression vector comprising the nucleic acid molecule of the present invention.
  • the expression vector may further comprise additional polynucleotide sequences, such as regulatory sequences and antibiotic resistance genes.
  • the nucleic acid molecule of the present invention may be present in one or more expression vectors.
  • the nucleic acid molecule of the present invention is prepared as a recombinant nucleic acid. Recombinant nucleic acids may be prepared using techniques well known in the art, such as chemical synthesis, DNA recombination techniques (e.g., polymerase chain reaction (PCR) techniques), etc.
  • PCR polymerase chain reaction
  • the present invention also provides a host cell comprising the nucleic acid molecule or expression vector of the present invention.
  • the nucleic acid molecule or expression vector of the present invention can be introduced into a suitable host cell by various methods known in the art. Such methods include but are not limited to liposome transfection, electroporation, viral transduction, and calcium phosphate transfection.
  • a host cell is used to express an anti-CD100 antibody or antigen-binding fragment thereof of the present invention.
  • host cells include, but are not limited to, prokaryotic cells (e.g., bacteria, such as E. coli) and eukaryotic cells (e.g., yeast, insect cells, mammalian cells).
  • Mammalian host cells suitable for antibody expression include, but are not limited to, human cervical cancer cells (HeLa cells), human embryonic kidney cells (HEK cells, such as HEK 293 cells), Chinese hamster ovary (CHO) cells, and other mammalian cells suitable for antibody expression.
  • the present invention also provides a method for producing the anti-CD100 antibody or antigen-binding fragment thereof of the present invention, comprising the following steps:
  • the present invention provides a drug combination, by which two or more therapeutic agents or preventive agents can be administered to a subject.
  • the drug combination comprises an antibody or antigen-binding fragment thereof against CD100 and an antibody or antigen-binding fragment thereof against PD-L1.
  • the antibody against CD100 includes antibody B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 or a combination thereof.
  • the antibody or antigen-binding fragment thereof against PD-L1 specifically recognizes and binds to PD-L1, wherein the anti-PD-L1 antibody or antigen-binding fragment thereof comprises an immunoglobulin single variable domain.
  • the immunoglobulin single variable domain comprises:
  • CDR1 comprises the amino acid sequence shown in SEQ ID NO:130,
  • CDR2 which comprises the amino acid sequence shown in SEQ ID NO:131, and
  • CDR3 which comprises the amino acid sequence shown in SEQ ID NO:132.
  • the immunoglobulin single variable domain comprises: 1) the amino acid sequence shown in SEQ ID NO:133; or 2) an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:133.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof further comprises an Fc fragment of human IgG1.
  • the anti-PD-L1 antibody or its antigen-binding fragment comprises the amino acid sequence shown in SEQ ID NO:134 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:134.
  • the anti-PD-L1 antibody comprises anti-PD-L1 antibody m18-VHH or m18.
  • the present invention provides an antibody m18-VHH or an antigen-binding fragment thereof against PD-L1,
  • the antibody comprises a single variable domain of an immunoglobulin, wherein
  • the single variable domain comprises:
  • CDR1 comprises the amino acid sequence shown in SEQ ID NO:130,
  • CDR2 which comprises the amino acid sequence shown in SEQ ID NO:131, and
  • CDR3 which comprises the amino acid sequence shown in SEQ ID NO:132.
  • the anti-PD-L1 antibody m18-VHH comprises a single variable domain of an immunoglobulin
  • the single variable domain comprises the amino acid sequence shown in SEQ ID NO:133 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:133.
  • the present invention also provides an antibody m18 or an antigen-binding fragment thereof against PD-L1.
  • the antibody comprises a single variable domain of an immunoglobulin, wherein
  • the single variable domain comprises:
  • CDR1 comprises the amino acid sequence shown in SEQ ID NO:130,
  • CDR2 which comprises the amino acid sequence shown in SEQ ID NO:131, and
  • CDR3 which comprises the amino acid sequence shown in SEQ ID NO:132.
  • the anti-PD-L1 antibody m18 comprises a single variable domain of an immunoglobulin
  • the single variable domain comprises the amino acid sequence shown in SEQ ID NO:133 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:133.
  • the anti-PD-L1 antibody m18 further comprises an Fc fragment of human IgG1.
  • the antibody m18 comprises the amino acid sequence shown in SEQ ID NO:134 or an amino acid sequence having at least 85%, at least 90%, at least 95% or higher sequence identity with SEQ ID NO:134.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof of the present invention is a single domain antibody, a heavy chain antibody, a humanized antibody or a chimeric antibody.
  • the antibody or antigen-binding fragment thereof is a human antibody.
  • the antibody or antigen-binding fragment thereof has at least one of the following characteristics:
  • the tumors targeted include but are not limited to those described below for neoplastic diseases.
  • the antibodies or antigen-binding fragments thereof of the present invention can inhibit tumor growth by at least about 10%, preferably at least about 20%, more preferably at least about 30%, more preferably at least about 40%, more preferably at least about 50%, more preferably at least about 60%, more preferably at least about 70%, more preferably at least about 80%.
  • the drug combination comprises an antibody against CD100, which comprises antibodies B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 or a combination thereof; and the drug combination comprises an antibody against PD-L1, which comprises antibody m18-VHH or m18.
  • the drug combination comprises anti-CD100 antibodies B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 or a combination thereof and anti-PD-L1 antibodies m18-VHH or m18.
  • the drug combination comprises anti-CD100 antibody C-C081, B13 or B13-c-5 and anti-PD-L1 antibody m18-VHH or m18.
  • the pharmaceutical combination can be a pharmaceutical composition or a kit.
  • the present invention also provides a pharmaceutical composition comprising the anti-CD100 antibody or antigenic fragment thereof of the present invention and a pharmaceutically acceptable carrier.
  • the anti-CD100 antibody comprises anti-CD100 antibody B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 or a combination thereof, in particular anti-CD100 antibody B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5 or B13-e-2.
  • the pharmaceutical composition comprises a combination of anti-CD100 antibody B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 and anti-PD-L1 antibody m18 or m18-VHH.
  • the pharmaceutical composition comprises a combination of anti-CD100 antibody B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 and anti-PD-L1 antibody m18 or m18-VHH.
  • the pharmaceutical composition comprises a combination of anti-CD100 antibody B13, C-C081 or B13-c-5 and anti-PD-L1 antibody m18 or m18-VHH.
  • compositions provided herein can be in various dosage forms, including but not limited to solid, semisolid, liquid, powder or lyophilized forms.
  • preferred dosage forms can generally be, for example, injection solutions and lyophilized powders.
  • compositions provided herein can be administered to a subject by any method known in the art, such as by systemic or topical administration.
  • the route of administration includes, but is not limited to, parenteral (e.g., intravenous, intraperitoneal, intradermal, intramuscular, subcutaneous or intracavitary), topical (e.g., intratumoral), epidural or mucosal (e.g., intranasal, oral, vaginal, rectal, sublingual or topical).
  • parenteral e.g., intravenous, intraperitoneal, intradermal, intramuscular, subcutaneous or intracavitary
  • topical e.g., intratumoral
  • epidural or mucosal e.g., intranasal, oral, vaginal, rectal, sublingual or topical
  • the pharmaceutical composition is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion).
  • the dosage range of the anti-CD100 antibody or antigen-binding fragment thereof of the present invention is about 0.0001-100 mg/kg, more typically 0.01-20 mg/kg of the subject's body weight.
  • the dosage may be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight, 10 mg/kg body weight or 20 mg/kg body weight, or within the range of 1-20 mg/kg.
  • Exemplary treatment regimens require administration once a week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months, once every 3-6 months, or a slightly shorter initial dosing interval and a longer dosing interval in the later stages.
  • the dosage used may be 1200 mg administered every three weeks.
  • the administration method may be intravenous drip.
  • a "therapeutically effective dose” refers to a dose that results in a decrease in the severity of disease symptoms, an increase in the frequency and duration of disease-free periods, or the prevention of damage or disability caused by disease suffering.
  • the therapeutically effective dose may be a dose that inhibits, delays or reduces tumor and/or malignant cell growth and/or metastasis in cancer patients, and/or reduces the observed symptoms associated with the disease.
  • the therapeutically effective dose may vary according to many different factors, including the mode of administration, the target site, the physiological state of the patient, whether the patient is a human or other animal, other drugs administered, and whether the treatment is preventive or therapeutic.
  • the patient is a human, but non-human mammals, including transgenic animals, may also be treated.
  • the therapeutic dose may be titrated to optimize safety and efficacy using conventional methods known to those skilled in the art.
  • a "therapeutically effective dose" of an antibody or antigen-binding fragment thereof of the present invention preferably inhibits cell growth or tumor growth by at least about 10%, preferably at least about 20%, more preferably at least about 30%, more preferably at least about 40%, more preferably at least about 50%, more preferably at least about 60%, more preferably at least about 70%, more preferably at least about 80%.
  • the ability to inhibit tumor growth can be evaluated in an animal model system that predicts the efficacy of human tumors. Alternatively, it can also be evaluated by examining the ability to inhibit cell growth, which inhibition can be determined in vitro by experiments known to those skilled in the art.
  • An effective amount of an antibody or antigen-binding fragment thereof of the present invention can reduce tumor size, or otherwise alleviate symptoms of a subject such as preventing and/or treating metastasis or recurrence. Such an amount can be determined by those skilled in the art based on factors such as the size of the subject, the severity of the subject's symptoms, and the specific composition or route of administration selected.
  • the present invention relates to use of the anti-CD100 antibody or antigen-binding fragment thereof, pharmaceutical composition or pharmaceutical combination of the present invention in the preparation of a medicament for treating a disease in a subject.
  • the present invention also relates to the anti-CD100 antibody or antigen-binding fragment thereof, pharmaceutical composition or pharmaceutical combination of the present invention, which is used for treating diseases.
  • the present invention also provides a method for treating a disease in a subject, comprising administering to the subject a therapeutically effective amount of the anti-CD100 antibody or antigen-binding fragment thereof, pharmaceutical composition or pharmaceutical combination of the present invention.
  • the disease as described above is cancer.
  • Blockade of CD100 by the antibodies of the present invention can enhance the immune response to cancer cells in patients.
  • CD100 is widely expressed in many human tumors, and its expression is associated with human invasive diseases.
  • inflammatory cells and tumor cells express CD100 to regulate the infiltration, spatial distribution and activity of myeloid cells and lymphocytes.
  • CD100 binds to the Plexin receptor on myeloid cells in the tumor microenvironment.
  • the CD100 protein is blocked, the CD100 barrier can be eliminated. Once the barrier is breached, inflammatory dendritic cells and proinflammatory antigen presenting cells migrate and infiltrate the tumor.
  • blocking CD100 with antibodies can delay tumor growth and promote persistent tumor rejection.
  • cancer includes but is not limited to hematologic malignancies and solid tumors.
  • solid tumors include, for example, squamous cell carcinoma, adenocarcinoma, basal cell carcinoma, renal cell carcinoma, breast duct carcinoma, soft tissue sarcoma, osteosarcoma, melanoma, small cell lung cancer, non-small cell lung cancer, lung adenocarcinoma, peritoneal cancer, hepatocellular carcinoma, gastrointestinal cancer, gastric cancer, pancreatic cancer, neuroendocrine cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, brain cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial cancer or uterine cancer, esophageal cancer, salivary gland cancer, kidney cancer, liver cancer, prostate cancer, vulvar cancer, thyroid cancer, head and neck cancer, etc.
  • Hematologic malignancies include, for example, leukemia, lymphoma, myeloma, acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, hairy cell leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, multiple myeloma, etc. or its any combination. Cancer can also be metastatic. "Metastasis" refers to the spread of cancer cells from their original site to other parts of the body.
  • the anti-CD100 antibodies or antigen-binding fragments thereof, pharmaceutical compositions or drug combinations of the present invention can be used in combination with other treatment methods, including but not limited to: surgery, chemotherapy, radiotherapy, targeted therapy, immunotherapy, hormone therapy, angiogenesis inhibition and palliative care.
  • anti-CD100 antibodies or antigen-binding fragments thereof, pharmaceutical compositions or drug combinations of the present invention may also be administered in combination with at least one or more therapeutic agents described herein.
  • the mode of combined administration is not limited.
  • the following therapeutic agents may be administered all at once or separately. When administered separately (in the case of using different administration regimens), they may be administered continuously without interruption or at predetermined intervals.
  • the anti-CD100 antibody or its antigen-binding fragment, pharmaceutical composition or drug combination of the present invention is further co-administered with one or more therapeutic agents selected from the following: chemotherapeutic agents, radioisotopes, immune checkpoint inhibitors and tumor antigen targeted drugs.
  • chemotherapeutic agents may include, for example, antimetabolites, alkylating agents, cytotoxic agents, topoisomerase inhibitors, microtubule inhibitors.
  • Tumor antigen targeted drugs include, but are not limited to, drugs targeting tumor-associated antigens and tumor-specific antigens.
  • therapeutic agents may include, for example, angiogenesis inhibitors, deacetylase (HDAC) inhibitors, Hedgehog signaling pathway blockers, mTOR inhibitors, p53/mdm2 inhibitors, PARP inhibitors, proteasome inhibitors (e.g., bortezomib, carfilzomib, ixazomib, Marizomib, Oprozomib) and tyrosine kinase inhibitors (e.g., BTK inhibitors).
  • HDAC deacetylase
  • anti-CD100 antibody B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 is administered in combination with anti-PD-L1 antibody m18 or m18-VHH.
  • the anti-CD100 antibody B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5 or B13-e-2 is administered in combination with the anti-PD-L1 antibody m18 or m18-VHH.
  • the anti-CD100 antibody C-C081, B13 or B13-c-5 is administered in combination with the anti-PD-L1 antibody m18 or m18-VHH.
  • the anti-CD100 antibody or its antigen binding fragment, pharmaceutical composition or drug combination of the present invention is administered in combination with a chemotherapeutic agent. In some embodiments, the anti-CD100 antibody or its antigen binding fragment, pharmaceutical composition or drug combination of the present invention is administered in combination with an immune checkpoint inhibitor. In some embodiments, the anti-CD100 antibody or its antigen binding fragment, pharmaceutical composition or drug combination of the present invention is administered in combination with a radioisotope. In some embodiments, the anti-CD100 antibody or its antigen binding fragment, pharmaceutical composition or drug combination of the present invention is administered in combination with a tumor targeting drug.
  • the present invention also provides a kit comprising an anti-CD100 antibody or antigen-binding fragment thereof, a pharmaceutical composition or a pharmaceutical combination of the present invention, and instructions for use.
  • the kit may also comprise a suitable container.
  • the kit also comprises a device for administration.
  • the kit generally includes a label indicating the intended use and/or method of use of the contents of the kit.
  • label includes any written or recorded material provided on or with the kit or otherwise provided with the kit.
  • the kit comprises anti-CD100 antibodies B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2, or a combination thereof.
  • the kit comprises anti-CD100 antibodies B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2, or a combination thereof.
  • the kit comprises anti-CD100 antibody C-C081, B13, or B13-c-5.
  • the kit comprises a combination of anti-CD100 antibodies B13, C-C081, A14, A15, H74, H96, H5, H12, H21, C-C171, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 and anti-PD-L1 antibody m18 or m18-VHH.
  • the kit comprises a combination of anti-CD100 antibodies B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2 and anti-PD-L1 antibody m18 or m18-VHH.
  • the kit comprises a combination of anti-CD100 antibody C-C081, B13 or B13-c-5 and anti-PD-L1 antibody m18 or m18-VHH.
  • the anti-CD100 antibody or antigen-binding fragment thereof of the present invention has at least one of the following beneficial effects: 1) having affinity activity for human, mouse or monkey CD100 protein; 2) having affinity activity for CD100-positive cells; 3) blocking the binding of CD100 to Plexin-B1 or Plexin-B2; 4) inhibiting MDSC cell proliferation; 5) inhibiting tumor growth.
  • the anti-CD100 antibody or its antigen-binding fragment in the present invention combined with the anti-PD-L1 antibody or its antigen-binding fragment has at least one of the following beneficial effects: 1) inhibiting or delaying tumor growth; 2) significantly improving the response rate to single medication; 3) prolonging the survival period; 4) enhancing the therapeutic effect of PD-L1 tumor immunotherapy; 5) high safety.
  • anti-CD100 control antibody The anti-CD100 antibody Pepinemab and the 2D5 and 5D8 antibodies of Shanghai Pioneer Pharmaceuticals were used as positive control antibodies. According to the sequences disclosed in WO2013148854A1 and patent WO2020011275, the coding gene sequences of Pepinemab monoclonal antibody and 2D5 and 5D8 antibodies were handed over to General Biotech Co., Ltd. for gene synthesis. Then, the eukaryotic expression vector pcDNA3.4 (Invitrogen) was constructed by homologous recombination.
  • pcDNA3.4 Invitrogen
  • the constructed recombinant protein expression vectors were transformed into Escherichia coli DH5 ⁇ , cultured overnight at 37°C, and then the plasmids were extracted using an endotoxin-free plasmid extraction kit (OMEGA, D6950-01) to obtain the desired expression plasmids for Pepinemab monoclonal antibody and 2D5 and 5D8 antibodies.
  • OEGA endotoxin-free plasmid extraction kit
  • the expression plasmid was transfected into CHO cells to express Pepinemab and 2D5 and 5D8 mAbs using the ExpiFectamine TM CHO transfection kit (Thermo Fisher, A29129) according to the manufacturer's instructions.
  • the cell culture supernatant was collected and centrifuged at 15,000 g for 10 min. The resulting supernatant was filtered through a 0.22 ⁇ m filter membrane and then purified by Protein A/G affinity chromatography. The antibody in the supernatant was affinity purified (MabSelect SuRe (Cytiva, 17543802)). The target antibody was eluted with 100 mM glycine (pH 3.0), and the eluted antibody was exchanged into PBS buffer through an ultrafiltration concentration tube (Millipore, UFC901096).
  • the specific method is as follows: use the purchased human CD100 protein (also known as HuCD100-Fc, 2 ⁇ g/mL, 30 ⁇ L/well) to coat the 96-well ELISA plate at 4°C overnight; wash the plate 3 times, block it with 5% skim milk prepared in PBS at room temperature for 1 hour; wash the plate 3 times, add the control antibody Pepinemab diluted in PBS gradient and incubate it at room temperature for 1 hour; after washing the plate, add the secondary antibody Anti-human-IgG-Kappa-HRP (Millipore, AP502P) and Anti-human-IgG-Lambda-HRP (Millipore, AP506P) diluted in PBS (1:6000) and incubate it at room temperature for 1 hour, wash the plate 6 times, add TMB (SurModics, TMBS-1000-01) to develop color for 5-20 minutes. After stopping the color development, read the data at OD450 using an enzyme reader (Molecular Devices,
  • Preparation of antigen protein Through genetic manipulation at the coding gene level, human Fc (SEQ ID NO: 135) or His tag was added to the C-terminus of the amino acid sequence of the 22-734th fragment of the ECD region of human CD100 protein (HuCD100, Uniprot ID: Q92854), the 24-733th fragment of the ECD region of mouse CD100 protein (MusCD100, Uniprot ID: O09126), and the 22-734th fragment of the ECD region of cynomolgus monkey CD100 protein (CynoCD100, Uniprot ID: A0A2K5TZC9).
  • the obtained nucleic acid sequences were constructed into pcDNA3.4 vectors, and then transformed into Escherichia coli DH5 ⁇ , cultured at 37°C overnight, and then the plasmid was extracted using an endotoxin-free plasmid extraction kit (OMEGA, D6950-01).
  • OEGA endotoxin-free plasmid extraction kit
  • the obtained plasmid was transiently transfected into HEK293 cells using ExpiFectamine TM 293 Transfection Kit (Gibco TM , A14524). CRL-1573 TM ). After 7 days of expression, the cell culture supernatant was collected. For proteins containing Fc tags, the cell suspension was centrifuged at high speed after the culture was completed and the supernatant was collected.
  • the obtained supernatant was filtered through a 0.22 ⁇ m filter membrane and purified by affinity chromatography using a Protein A/G column MabSelect SuRe (Cytiva, 17543802).
  • the target protein was eluted with 100mM glycine hydrochloride (pH 3.0), concentrated, buffer exchanged, packaged, SDS-PAGE identified and activity tested, and then stored frozen.
  • the protein containing the His tag was affinity purified using Ni Smart Beads 6FF (Changzhou Tiandi Renhe Biotechnology Co., Ltd., SA036050), and then the target protein was eluted with an imidazole gradient.
  • the eluted proteins were respectively exchanged into PBS buffer through ultrafiltration concentration tubes (Millipore, UFC901096), and finally human, mouse and monkey CD100 antigen proteins with Fc tags and His tags were obtained (HuCD100-Fc, MusCD100-Fc, CynoCD100-Fc, HuCD100-His, MusCD100-His, CynoCD100-His).
  • Antigen identification The prepared antigens (HuCD100-Fc, MusCD100-Fc, CynoCD100-Fc, HuCD100-His, MusCD100-His, CynoCD100-His) were detected using the antibody Pepinemab (IgG4SP) obtained in Example 1.1 that passed the quality inspection.
  • IgG4SP antibody Pepinemab
  • the specific method is as follows: ELISA plates were coated with 2 ⁇ g/mL antigen at 4°C overnight, and the purchased antigen protein Human Semaphorin 4D/SEMA4D/CD100Protein and Fc Tag antigen protein (Acro, CD0-H5257) were used as positive controls; after washing the plates three times, they were blocked with 5% skim milk prepared in PBS at room temperature for 1 hour; after washing the plates three times, Add the antibody Pepinemab diluted in PBS gradient and incubate at room temperature for 1 hour; after washing, add the secondary antibody Anti-human-IgG-Kappa-HRP (Millipore, AP502P) or Anti-human-IgG-Lambda-HRP (Millipore, AP506P) diluted in PBS (1:6000) and incubate at room temperature for 1 hour, wash the plate 6 times and then add TMB for color development for 5-20 minutes. Stop the color development reaction and read the data at OD450 with an
  • the affinity activity of the antibody Pepinemab for binding to the homemade antigen is comparable to that for binding to the purchased CD100 antigen protein, wherein the two HuCD100-Fcs are the homemade HuCD100 antigen and the commercially purchased HuCD100 antigen, respectively.
  • HuCD100-HEK293, Mus CD100-HEK293, CynoCD100-HEK293 The coding nucleic acid sequences of full-length human CD100 protein HuCD100 (Uniprot ID: Q92854), full-length mouse CD100 protein MusCD100 (Uniprot ID: O09126), and full-length cynomolgus monkey CD100 protein CynoCD100 (Uniprot ID: A0A2K5TZC9) were constructed into pLVX-puro plasmid (Clontech, catalog number 632164).
  • the obtained plasmid was electroporated into HEK293 cells (Invitrogen, NeonTM Transfection System, MP922947). CRL-1573 TM ). After electroporation, the obtained cells were transferred to DMEM medium (Gibco, 11995065) containing 10% FBS (Gibco, 15140-141) by volume and without antibiotics. The cells were then transferred to a 10 ⁇ 10 cm cell culture dish and cultured for 48 hours. The cells were then distributed to a 96-well cell culture plate at an average density of 10 4 cells/well, and puromycin with a final concentration of 2 ⁇ g/mL was added as a screening pressure. After about 2 weeks, the cell lines that formed clones were picked for identification.
  • Flow cytometric identification of HuCD100-HEK293, MusCD100-HEK293, and CynoCD100-HEK293 cells The cells of the above cell lines in the logarithmic growth phase were digested with trypsin and plated into 96-well plates. After washing with FACS buffer (1 ⁇ PBS buffer containing 2% FBS by volume), the primary antibody (Pepinemab) diluted in gradient dilution with PBS was added and incubated at 4°C for 30 min. After washing, the prepared fluorescent secondary antibody anti-human IgG Fc (abcam, 98596) was added and incubated at 4°C for 30 min. Finally, the cells were detected by flow cytometer (Beckman, CytoFLEXAOO-1-1102).
  • FIG3 The detection results are shown in FIG3 , which show that HuCD100-HEK293, MusCD100-HEK293, and CynoCD100-HEK293 cell lines highly express human, mouse, and monkey CD100 on their surfaces, respectively.
  • HuPlexin-B1-HEK293, MusPlexin-B1-HEK293, CynoPlexin-B1-HEK293, and HuPlexin-B2-HEK293) The nucleic acid sequences encoding the full-length human Plexin-B1 protein HuPlexin-B1 (Uniprot ID: O43157), the full-length mouse Plexin-B1 protein MusPlexin-B1 (Uniprot ID: Q8CJH3), the full-length cynomolgus monkey Plexin-B1 protein CynoPlexin-B1 (Uniprot ID: A0A1D5QMB8), and the full-length human Plexin-B2 protein HuPlexin-B2 (Uniprot ID: O15031) were constructed into the pLVX-puro plasmi
  • HuPlexin-B1-HEK293, MusPlexin-B1-HEK293, CynoPlexin-B1-HEK293, and HuPlexin-B2-HEK293 cells Digest the above cell lines in the logarithmic growth phase and plate them into 96-well plates.
  • the diluted antigen protein (HuCD100-Fc, MusCD100-Fc, CynoCD100-Fc prepared in Example 1.2 were obtained by biotin labeling HuCD100-Fc, MusCD100-Fc, CynoCD100-Fc-Biotin, for the biotin labeling method, please refer to the instructions of the biotin protein labeling kit of Roche, catalog number: 11418165001), incubated at 4°C for 30 min; after washing, the prepared eBioscience Streptavidin PE (Invitrogen, 2265658, 1:300) was added and incubated at 4°C for 30 min; finally, the protein was detected by flow cytometry (Beckman, CytoFLEXAOO-1-1102).
  • Figures 4A-4D The detection results are shown in Figures 4A-4D, wherein Figure 4A is a schematic diagram of the results of identifying HuPlexin-B1-HEK293 cell lines using human CD100 protein, Figure 4B is a schematic diagram of the results of identifying MusPlexin-B1-HEK293 cell lines using mouse CD100 protein, Figure 4C is a schematic diagram of the results of identifying CynoPlexin-B1-HEK293 cell lines using monkey CD100 protein, and Figure 4D is a schematic diagram of the results of identifying HuPlexin-B2-HEK293 cell lines using human CD100 protein.
  • an antibody gene phage display library was constructed, and the library was screened using the antigen proteins HuCD100-Fc, MusCD100-Fc, CynoCD100-Fc, HuCD100-His, MusCD100-His, and CynoCD100-His prepared in Example 1.2 as screening antigens to obtain multiple antibody molecules that specifically bind to human CD100.
  • Ficoll-Paque density gradient separation solution (purchased from GE, catalog number: 17144003S) was used to separate the peripheral blood mononuclear cells (PBMC) of normal human blood, and total RNA was extracted from the isolated PBMC cells by conventional methods.
  • Reverse transcription kit (purchased from TaKaRa, catalog number: 6210A) was used to reverse transcribe the extracted total RNA into cDNA according to the manufacturer's instructions. Based on the sequence similarity of the heavy chain and light chain germline genes, degenerate primers were designed at the front end of the V region of the heavy chain and the rear end of the first constant region, and the heavy chain variable region gene fragment and the light chain variable region gene fragment of the antibody were obtained after PCR.
  • the fragments containing the light chain variable region and the heavy chain variable region of the antibody were amplified by the fusion PCR method, and the PCR product and the phage display vector were digested, recovered and connected, and the connection product was recovered by a recovery kit (Omega, catalog number: D6492-02) (Li Xiaolin, Construction and preliminary screening of a large-capacity non-immune human Fab phage antibody library, Master's thesis of Peking Union Medical College, June 2007).
  • the library capacity was measured to be 3 ⁇ 10 11 cfu, i.e., an antibody gene library of 3 ⁇ 10 11 antibody genes (for the library capacity calculation method, see Example 2.2 in CN112250763A).
  • VSCM13 helper phage purchased from Stratagene was used to package the library to obtain an antibody gene phage display library (for the preparation of the antibody gene phage display library, see Example 2.3 in CN112250763A).
  • Magnetic bead screening is a panning process in which the antigen protein is labeled with biotin and then combined with magnetic beads coupled with streptavidin. The antigen-bound magnetic beads and the antibody gene phage display library are incubated, washed and eluted. Usually 3-4 rounds of panning are performed, whereby specific monoclonal antibodies against the antigen can be enriched in large quantities.
  • biotin-labeled antigen proteins HuCD100-Fc, CynoCD100-Fc, HuCD100-His, and MusCD100-His were used for phage display library screening, and after 3 rounds of panning, monoclonal antibody Fab against human CD100 was initially screened.
  • monoclonal antibody Fab against human CD100 was initially screened.
  • the immunotube screening is to coat the antigen protein HuCD100-Fc, CynoCD100-Fc, HuCD100-His, and MusCD100-His on the surface of an immunotube with high adsorption capacity, and then add the phage display antibody library to the immunotube and incubate, wash, and elute with the antigen protein adsorbed on the surface of the immunotube. After 2-4 rounds of panning, the specific monoclonal antibody Fab for the antigen is finally enriched. In this embodiment, the monoclonal antibody Fab for human CD100 is enriched after 3 rounds of panning.
  • the specific method is referred to Example 2.4.2 in CN112250763A.
  • the phage pool eluted in each round was tested by ELISA to evaluate the enrichment effect, and 10 clones were randomly selected from the phage pool in each round of screening for sequence analysis.
  • the enrichment effect and the repeatability ratio of the measured sequences were comprehensively analyzed to select the appropriate round for single clone selection.
  • the ELISA monoclonal primary screening used antigen proteins HuCD100-His, MusCD100-His, and CynoCD100-His.
  • the antibody Fabs binding to HuCD100-His, MusCD100-His, and CynoCD100-His obtained in the primary screening were prepared into Fab lysate, and then the overexpression cells HuCD100-HEK293, MusCD100-HEK293, and CynoCD100-HEK293 prepared in Example 2.1 were detected and verified by flow cytometry (FACS).
  • a total of 11 antibody Fab molecules that specifically bound to human CD100 were screened, and the 11 obtained antibody Fabs were named according to the corresponding clone numbers (A14, A15, H74, H96, H5, H12, H21, B13, C-C081, C-C171, and C-B71).
  • the obtained antibody variable region amino acid sequences are shown in Table 1.
  • the CDR sequences were determined using the AbM definition method for CDR.
  • the VH coding sequence in the Fab sequence of the screened monoclonal A14, A15, H74, H96, H5, H12, H21, B13, C-C081, C-C171, and C-B71 was connected to the coding sequence of the heavy chain constant region of human IgG4SP (SEQ ID NO: 127) to obtain the heavy chain coding sequence of the fully human antibody, and the VL coding sequence in the Fab sequence was connected to the coding sequence of the human light chain constant region (SEQ ID NO: 127).
  • the coding sequence of the Kappa type (SEQ ID NO: 128) or Lambda type (SEQ ID NO: 129) of the constant region (CL) of the chain was connected to obtain the light chain coding sequence of the fully human antibody.
  • the coding sequences of the antibody heavy chain and light chain were respectively inserted into the eukaryotic expression vector plasmid pcDNA3.4 (Invitrogen), transformed into Escherichia coli DH5 ⁇ , and cultured at 37°C overnight.
  • the plasmid was extracted using an endotoxin-free plasmid extraction kit (OMEGA, D6950-01) to obtain an endotoxin-free antibody plasmid for eukaryotic expression.
  • the full-length sequence of the antibody obtained above was expressed by the ExpiCHO transient expression system (Thermo Fisher, A29133) kit.
  • the specific method is as follows: On the day of transfection, the CHO cell density was confirmed to be about 7 ⁇ 10 6 to 1 ⁇ 10 7 viable cells/mL, and the cell survival rate was >98%. At this time, the cells were adjusted to a final concentration of 6 ⁇ 10 6 cells/mL with fresh ExpiCHO expression medium pre-warmed at 37°C.
  • the target plasmid was diluted with OptiPRO TM SFM pre-cooled at 4°C (1 ⁇ g of plasmid was added to 1 mL of the culture medium), and the ExpiFectamine TM CHO reagent was diluted with OptiPRO TM SFM at the same time, and then the two were mixed in equal volumes and gently pipetted to mix to prepare ExpiFectamine TM CHO/plasmid DNA mixed solution. Incubate the mixture at room temperature for 1-5 minutes, slowly add it to the prepared cell suspension while gently shaking, and finally place it in a cell culture shaker and culture it at 37°C and 8% CO2 .
  • ExpiCHO TM Enhancer reagent and ExpiCHO TM Feed reagent were added to the cell culture medium, and the shake flask was placed in a 32°C shaker and 5% CO 2 for continued culture. On the 5th day after transfection, the same volume of ExpiCHO TM Feed reagent was added, and the cell suspension was gently mixed while slowly adding. After 7 days of transfection, the cell culture supernatant expressing the target antibody protein was collected and centrifuged at 15000g for 10min.
  • the obtained supernatant was affinity purified with MabSelect SuRe LX (GE, 17547403), and then the target antibody protein was eluted with 100mM sodium acetate (pH 3.0), followed by neutralization with 1M Tris-HCl, and finally the obtained antibody protein was exchanged into PBS buffer by ultrafiltration concentration tube (Millipore, UFC901096).
  • SDS-PAGE and SEC-HPLC were used to detect the relative molecular weight and purity of the candidate antibodies.
  • Preparation of reducing solution 2 ⁇ g of each obtained antibody and quality control product IPI were added to 5 ⁇ SDS loading buffer and 5 mM DTT, and heated in a 100°C dry bath for 10 min. After cooling the mixture to room temperature, centrifuge at 12000 rpm for 5 min to obtain the supernatant.
  • the supernatant was added to Bis-tris 4-15% gradient gel (GenScript) for gel electrophoresis and the protein bands were visualized by Coomassie Brilliant Blue staining.
  • the protein gel with the visualized protein bands was scanned using EPSON V550 color scanner (the gel was destained with destaining solution until its background was transparent), and the purity of the reduced and non-reduced bands was calculated by ImageJ according to the peak area normalization method.
  • Material preparation Mobile phase: 150mmol/L phosphate buffer, pH 7.4; dilute each antibody and quality control product IPI to 0.5mg/mL with mobile phase solution.
  • the binding of the expressed candidate antibodies (A14, A15, H74, H96, H5, H12, H21, B13, C-C081, C-C171, C-B71) to the CD100 antigen proteins HuCD100-His, MusCD100-His, and CynoCD100-His was detected based on the ELISA method, and the expressed antibodies (A14, A15, H74 , H96, H5, H12, H21, B13, C-C081, C-C171, C-B71) and CD100-overexpressing cells HuCD100-HEK293, MusCD100-HEK293, CynoCD100-HEK293 cells, human peripheral blood mononuclear cells naturally expressing human CD100 (hereinafter referred to as HuPBMC cells) and human T lymphocytic leukemia cells Jurkat cells.
  • HuPBMC cells human peripheral blood mononuclear cells naturally expressing human CD100
  • Jurkat cells human T lymphocytic leukemia cells
  • 96-well ELISA plates (30 ⁇ L/well) were coated with 2 ⁇ g/mL HuCD100-His, MusCD100-His, and CynoCD100-His at 4°C overnight. The next day, the plates were washed 3 times with PBST and blocked with 5% skim milk for 2 h. After washing the plates 3 times with PBST, gradient dilutions (3.0, 0.33, 0.11, 0.037, 0.012, 0.004, 0.0014, 0.0002 ⁇ g/mL) of each antibody and positive control antibody Pepinemab were added and incubated for 1 h.
  • the plates were washed 3 times with PBST and the secondary antibody Goat-anti-human Fc-HRP (abcam, ab97225) was added and incubated for 1 h. After the incubation, the plates were washed 6 times with PBST and TMB (SurModics, TMBS-1000-01) was added for color development. According to the color development results, 2 M stop solution was added to terminate the reaction, and the absorbance was read at OD450 using an enzyme reader (Molecular Devices, SpecterMax 190).
  • Goat-anti-human Fc-HRP abcam, ab97225
  • the binding activities of antibody molecule C-C081 to the antigen protein MusCD100-His were weaker than those of the positive control antibody and antibody molecule H1 2 and H96, whose binding activities with the antigen protein MusCD100-His were comparable to those of the positive control antibody, while the binding activities of the other antibody molecules with the antigen protein MusCD100-His were better than those of the positive control antibody; for the antigen protein CynoCD100-His ( Figures 5Q-5X), except for the antibody molecules H12 and H96, whose binding activities with the antigen protein CynoCD100-His were comparable to those of the positive control antibody, the binding activities of the other antibody molecules with the antigen protein CynoCD100-His were better than those of the positive control antibody.
  • three types of cells namely, human CD100 overexpressing cells HuCD100-HEK293, MusCD100-HEK293, and CynoCD100-HEK293, were used to evaluate the binding activity of the antibody.
  • HuCD100-HEK293, MusCD100-HEK293, and CynoCD100-HEK293 cells in the logarithmic growth phase were prepared into a single cell suspension, the density was adjusted to 1 ⁇ 10 6 cells/mL, and 100 ⁇ L was added to each well of a 96-well plate, centrifuged at 4°C, 300g, and the supernatant was removed.
  • Each antibody and the positive control antibody Pepinemab were added to the corresponding wells in each gradient dilution (20.0, 6.67, 2.22, 0.74, 0.25, 0.08, 0.027, 0.0027 ⁇ g/mL), mixed, and incubated at 4°C for 30 minutes.
  • the incubated cell mixture was washed three times and then 100 ⁇ L of 1:300 diluted secondary antibody Goat F(ab')2 Anti-Human IgG-Fc(PE) (abcam, ab98596) was added. The mixture was incubated at 4°C in the dark for 30 min. After washing three times, the cells were detected by flow cytometry (Beckman, CytoFLEX AOO-1-1102).
  • HuPBMC and Jurkat cells were used to evaluate the binding activity of the antibody.
  • HuPBMC AllCells, catalog number PB004F-C (Y1246)
  • Jurkat cells ATCC, TIB-152
  • HuPBMC AllCells, catalog number PB004F-C (Y1246)
  • Jurkat cells ATCC, TIB-152
  • HuPBMC AllCells, catalog number PB004F-C (Y1246)
  • Jurkat cells ATCC, TIB-152
  • HuPBMC AllCells, catalog number PB004F-C (Y1246)
  • Jurkat cells ATCC, TIB-152
  • Each antibody and the positive control antibody Pepinemab were added to the corresponding wells in each gradient dilution (5.00, 0.50, 0.17, 0.056, 0.019, 0.006, 0.002, 0.0002 ⁇ g/mL), mixed and incubated at 4°C for 30 minutes.
  • the incubated cell mixture was washed three times and then 100 ⁇ L of 1:300 diluted secondary antibody Goat F(ab') 2 Anti-Human IgG-Fc(PE) (abcam, ab98596) was added, incubated at 4°C in the dark for 30 min, washed three times and detected by flow cytometry (Beckman, CytoFLEX AOO-1-1102).
  • HuPlexin-B1-HEK293, MusPlexin-B1-HEK293, CynoPlexin-B1-HEK293, and HuPlexin-B2-HEK293, were used to evaluate the blocking activity of candidate antibodies (A14, A15, H74, H96, H5, H12, H21, B13, C-C081, C-C171, and C-B71).
  • the specific method is as follows: collect the cultured HuPlexin-B1-HEK293, MusPlexin-B1-HEK293, CynoPlexin-B1-HEK293, and HuPlexin-B2-HEK293 cells, centrifuge at 300 g to remove the supernatant, resuspend the cells in the prepared FACS buffer, count the cells, and adjust the cell suspension density to 1 ⁇ 10 6 cells/mL; HuPlexin-B1-HEK293, MusPlexin-B1-HEK293, CynoPlexin-B1-HEK293, and HuPlexin-B2-HEK293 cells were added to a 96-well plate at 100 ⁇ L per well, and the supernatant was removed by centrifugation at 300 g; each antibody and the positive control antibody Pepinemab were added to the corresponding wells of the 96-well plate in gradient dilutions (20.0, 2.00, 0.67, 0.22,
  • biotin labeling method refer to the instructions of the biotin protein labeling kit of Roche, catalog number: 11418165001), and the mixture was added to the cells and incubated together for 1 hour. The cell mixture was then washed 3 times. Resuspend the cells and incubate them at 4°C for 30 min; wash the incubated cell mixture three times, then add PE-labeled streptavidin (eBioscience, 12-4317-87) and incubate at 4°C for 30 min; wash the incubated cell mixture three times, then add FACS buffer to the wells, 200 ⁇ L per well, resuspend the cells, and detect by flow cytometer (Beckman, CytoFLEX AOO-1-1102).
  • the antibody molecules A14, A15, B13, C-C081 and C-C171 had better activities in blocking the binding of HuCD100 antigen protein to HuPlexin-B1-HEK293 than the positive control antibody; the antibody molecules A15, B13, H74, H96, H12 and C-C081 had better activities in blocking the binding of HuCD100 antigen protein to HuPlexin-B2-HEK293 than the positive control antibody; the antibody molecules H12, H74, H96 and C-C081 had better activities in blocking the binding of MusCD100 antigen protein to MusPlexin-B1-HEK293 than the positive control antibody; the antibody molecules A15, B13, H12, H96, C-C081, C-C171 and C-B71 had better activities in blocking the binding of CynoCD100 antigen protein to CynoPlexin-B1-HEK293 than the positive control antibody.
  • the in vitro pharmacodynamic function of the candidate antibodies was evaluated by examining their effects on the proliferation inhibition of myeloid-derived suppressor cells (MDSCs).
  • CD33+ cells were sorted from fresh PBMC using CD33 sorting magnetic beads (Miltenyi Biotech). After sorting, the cell density was adjusted to 1 ⁇ 10 6 /mL for standby use.
  • the antibody was diluted with 1640 complete medium in a gradient manner to 100.0, 33.33, 11.11, 3.704, 1.235, and 0.412 ⁇ g/mL.
  • the HuCD100-His antigen protein was diluted with 1640 complete medium to 400.0 ⁇ g/mL.
  • the antibody diluent and the HuCD100-His antigen protein diluent were evenly mixed at a ratio of 1:1 and incubated at room temperature for 30 minutes.
  • a 96-well cell culture plate was taken, and 100 ⁇ L of CD33 + cells with a density of 1 ⁇ 10 6 /mL (1 ⁇ 10 5 cells per well) were added to each well, and then 100uL of the antibody and HuCD100-His antigen protein mixture was added to each well, for a total of 200 ⁇ L. After the cell culture plate was placed in a 37°C cell culture incubator for 72 hours, the cells in the sample wells were transferred to a 96-well U-shaped plate, and the plate was washed twice with FACS buffer.
  • Three direct-labeled antibodies PE-anti-human CD33 (Biolegend, 303404), FITC-anti-human HLA-DR (Biolegend, 307604), and APC anti-human CD11b Antibody (Biolegend, 301310), were diluted at a ratio of 1:100 with FACS buffer. 100 ⁇ L of the three direct-labeled antibody mixture was added to each well and incubated at 4°C for 30 minutes. The plate was washed twice with FACS buffer. Detection was performed by flow cytometry (Beckman, CytoFLEX AOO-1-1102).
  • the antibody NB22D-21-huVH2 (see, for example, CN112745391A) was subjected to affinity maturation modification to improve antibody affinity and other biological activities.
  • Affinity maturation modification is based on M13 phage display technology, using codon-based primers (during primer synthesis, a single codon is composed of NNK) to introduce CDR region mutations, and construct four phage display libraries: Library 1 is a single-point combination mutation, Library 1 is a CDR1+CDR2+CDR3 combination mutation; Library 2, Library 3 and Library 4 are double-point combination mutations, Library 2 is a double-point combination mutation of CDR1+CDR3, Library 3 is a double-point combination mutation of CDR2+CDR3, and Library 4 is a double-point combination mutation of CDR1+CDR2.
  • Specific library construction method First, synthesize primers containing point mutations (Jinweizhi Biotechnology Co., Ltd.); secondly, use the coding sequence of the antibody to be modified (hereinafter referred to as the parent antibody) NB22D-21-huVH as a PCR amplification template to amplify the sequence containing the mutation in the CDR region, and combine the fragments containing different CDR mutations by the bridge PCR method, and then connect the point mutation antibody to the phage display vector by double enzyme digestion (HindIII and NotI) and double sticky end connection, and finally transfer the antibody sequence with the mutation site into Escherichia coli SS320 by electroporation.
  • phage library preparation and library screening see Example 3.
  • the obtained antibody is named m18-VHH.
  • the variable region amino acid sequence of the obtained antibody is shown in Table 7, and the CDR sequence is determined by using the AbM definition method.
  • a fusion expression vector was constructed by connecting the C-terminus of the VHH gene sequence to the N-terminus of the human IgG1Fc segment gene sequence to fuse the antibody m18-VHH and the human IgG1Fc segment (SEQ ID NO: 135), and the fusion expression vector plasmid was transformed into ExpiCHO cells and induced to express to obtain a VHH-Fc chimeric antibody protein fused with the Fc segment (SEQ ID NO: 134).
  • the VHH-Fc antibody is hereinafter named as antibody m18.
  • the antibody was expressed using the ExpiCHO transient expression system, using ExpiCHO TM expression medium (Gibco, A29100-01) and Gibco TM ExpiFectamine TM CHO transfection kit (Gibco, A29129). See Example 4 for specific methods.
  • the binding ability of the VHH-Fc antibody to PD-L1-overexpressing cells, human PD-L1-CHO cells, human non-small cell lung cancer cell line HCC827 cells, mouse PD-L1-CHO cells and cynomolgus monkey PD-L1-CHO cells was detected based on the FACS method.
  • the sources or preparation methods of the above cell lines please refer to CN112745391A.
  • human PD-L1-CHO cells were used to evaluate the binding activity of the antibody to human PD-L1 overexpressing cells.
  • Example 5 in CN112745391A, wherein the positive control antibody used is Avelumab (the preparation method is described in patent WO2013079174).
  • the binding activity of the antibody to the PD-L1 protein on human tumor cells was evaluated using the human non-small cell lung cancer cell line HCC827 cells (ATCC: CRL-2868).
  • Example 8 in CN112745391A, wherein the positive control antibody used is Avelumab (the preparation method is described in patent WO2013079174).
  • mouse PD-L1-CHO cells and cynomolgus monkey PD-L1-CHO cells were used to evaluate the cross-binding activity with monkey and mouse PD-L1.
  • the positive control antibody used is Avelumab (the preparation method is described in patent WO2013079174).
  • the results are shown in Figures 11A and 11B.
  • the antibody m18 has good binding activity with mouse PD-L1-CHO cells, while the parent antibody and the positive control antibody do not bind to mouse PD-L1-CHO cells ( Figure 11A). It can be expected that the antibody molecule m18 can be used for animal model experiments in Balb/C mice; the binding activity of the antibody m18 with crab-eating monkey PD-L1-CHO cells is better than that of the positive control antibody and is equivalent to that of the parent antibody ( Figure 11B).
  • Antibody m18 has no binding activity to B7 family molecules other than B7-H1 (i.e., PD-L1), and only has binding activity to B7-H1. This binding specificity is consistent with the parent antibody.
  • Antibodies B13 and H5 were subjected to affinity maturation to improve affinity and biological activity. Affinity maturation was based on M13 phage display technology, using codon-based primers (during primer synthesis, a single codon consisted of NNK) to introduce mutations in the CDR region, and four phage display libraries were constructed: Library 1 and Library 2 were single-point combination mutations, Library 1 was CDRL1+CDRL3+CDRH3 combination mutations, Library 2 was CDRL2+CDRH1+CDRH2 combination mutations; Library 3 and Library 4 were double-point saturation mutations, Library 3 was double-point saturation mutations of CDRL3, and Library 4 was double-point saturation mutations of CDRH3.
  • the specific library construction method is as follows: First, primers containing point mutations were synthesized (Genwizhi Biotechnology Co., Ltd.); secondly, the antibodies B13 and H5 to be modified were used as PCR amplification templates to amplify the sequences containing mutations in the CDR region, and fragments containing different CDR mutations were combined by the bridge PCR method, and then the point mutation antibodies were connected by double enzyme digestion (HindIII and NotI) and double sticky end ligation. The antibody sequence with the mutation site is transferred into the phage display vector, and finally the antibody sequence with the mutation site is transferred into E. coli SS320 by electroporation.
  • the specific process of library capacity calculation and phage library preparation is shown in Example 3.
  • Example 3 The specific operation method of library screening is shown in Example 3. After the library screening, primary screening, affinity sorting and sequence analysis, a total of 74 positive clones of H5 and 57 positive clones of B13 were obtained, and affinity sorting was performed respectively. Combined with the affinity sorting and sequence analysis data, 20 candidate molecules were finally selected from the H5 molecules and 28 candidate molecules were selected from the B13 molecules for construction, expression and functional screening.
  • B13 selected two anti-C100 affinity mature antibodies B13-c-5 and B13-e-2.
  • H5 selected two anti-C100 affinity mature antibodies H5-h-7 and H5-a-2.
  • the amino acid sequences of the obtained antibody variable regions are shown in Table 9, and the CDR sequences were determined by using the AbM definition method.
  • the expression level and in vitro physicochemical property test results are shown in Table 10.
  • the purity of the antibody was identified by SDS-PAGE, and the purity of the modified antibody was preferably greater than 95%.
  • the monomer purity of the antibody was identified by SEC-HPLC, and the monomer purity of the modified antibody was preferably greater than 98%.
  • Figure 13A shows the results of binding activity to HuCD100-HEK293 cells.
  • Figure 13B shows the results of binding activity to human PBMC cells.
  • Figure 13C shows the results of binding activity to MusCD100-HEK 293 cells.
  • Figure 13D shows the results of binding activity to CynoCD100-HEK 293 cells.
  • the affinity activities of the modified antibodies B13-c-5, B13-e-2, H5-h-7 and H5-a-2 for human CD100-overexpressing cells were significantly better than those of the positive control antibody Pepinemab, and were superior to their corresponding parent antibodies, respectively; and the affinity activities of the modified antibodies for mouse CD100-overexpressing cells (MusCD100-HEK 293) and crab-eating macaque CD100-overexpressing cells (CynoCD100-HEK 293) were significantly better than those of the positive control antibody Pepinemab, and were superior to their corresponding parent antibodies.
  • the modified antibodies can effectively block the binding of HuCD100 antigen protein to HuPlexin-B1-HEK293 and HuPlexin-B2-HEK293 cells, the binding of MusCD100 antigen protein to MusPlexin-B1-HEK293 cells, and the binding of CynoCD100 antigen protein to CynoPlexin-B1-HEK293 cells.
  • the activity of the modified antibody in blocking the binding of HuCD100 antigen protein to HuPlexin-B1-HEK293 cells was better than that of the positive control antibody and its corresponding parent antibody ( Figure 14A); the activity of the modified antibody in blocking the binding of HuCD100 antigen protein to HuPlexin-B2-HEK293 cells ( Figure 14B) and blocking the binding of MusCD100 antigen protein to HuPlexin-B1-HEK293 cells ( Figure 14C) was better than the positive control antibody but weaker than the corresponding parent antibody; the activity of H5-h-7 and H5-a-2 in blocking the binding of CynoCD100 antigen protein to CynoPlexin-B1-HEK293 cells was better than the positive control antibody and the parent antibody H5, and the activity of B13-c-5 and B13-e-2 in blocking the binding of CynoCD100 antigen protein to CynoPlexin-B1-HEK293 cells was better than the positive control antibody but weaker than the parent
  • results of MDSC proliferation inhibition assay are shown in FIG15 and Table 11.
  • the results show that anti-CD100 antibodies H5-h-7, H5-a-2, H5, B13, B13-c-5, B13-e-2 and the control antibody Pepinemab can effectively neutralize the induction of CD100 on MDSC cell populations.
  • 100 ⁇ g/mL of H5-h-7, H5-a-2, H5, B13, B13-c-5, B13-e-2 had significantly higher inhibitory activity on MDSC than the control antibody Pepinemab.
  • This example detects the tumor inhibition effect of 7 anti-CD100 antibodies (antibody B13, C-C081, C-B71, H5-h-7, H5-a-2, B13-c-5, B13-e-2) in combination with anti-PD-L1 antibody in animals.
  • the tumor cells used are CT-26 (Shanghai Cell Bank, Chinese Academy of Sciences, catalog number TCM37), and the antibody Pepinemab is used as a positive control.
  • the specific method is as follows: 6-8 week old female Balb/C mice weighing about 20 g (Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.) were used, and 5 ⁇ 10 5 CT-26 cells were injected subcutaneously unilaterally in each mouse. After two days of tumor bearing, the mice were randomly divided into groups and cages.
  • tumor-bearing nude mice There were 8 tumor-bearing nude mice in each group, for a total of 10 groups, including PBS negative control group, candidate antibody combination drug group (antibody B13 + antibody m18, antibody C-C081 + antibody m18, antibody C-B71 + antibody m18, antibody H5-h-7 + antibody m18, antibody H5-a-2 + antibody m18, antibody B13-c-5 + antibody m18, antibody B13-e-2 + antibody m18) and positive or reference control antibody group (Pepinemab + antibody m18, antibody m18).
  • the dosage of anti-CD100 antibody was 50 mpk
  • the dosage of anti-PD-L1 antibody was 5 mpk.
  • the administration method was intraperitoneal injection.
  • the drug was administered twice a week and the tumor volume was measured twice, for a total of 8 times/4 weeks (BIW*4).
  • the weight of mice was measured at the same time point.
  • L the tumor diameter
  • W the longest diameter of the tumor
  • W is the shortest diameter of the tumor
  • the combination of anti-CD100 antibody and anti-PD-L1 antibody can further synergistically inhibit or delay tumor growth compared with the single use of anti-PD-L1 antibody m18.
  • TGI% Tumor inhibition rate

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Mycology (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

L'invention concerne un anticorps anti-CD100 et son utilisation, une composition pharmaceutique ou une combinaison pharmaceutique comprenant l'anticorps, et une utilisation associée. L'invention concerne en outre un polynucléotide codant pour l'anticorps, un vecteur d'expression et un procédé de production de l'anticorps.
PCT/CN2023/124146 2022-10-12 2023-10-12 Anticorps anti-cd100 et son utilisation WO2024078558A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211246863.8 2022-10-12
CN202211246863.8A CN117866091A (zh) 2022-10-12 2022-10-12 抗cd100抗体及其用途

Publications (1)

Publication Number Publication Date
WO2024078558A1 true WO2024078558A1 (fr) 2024-04-18

Family

ID=90588839

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/124146 WO2024078558A1 (fr) 2022-10-12 2023-10-12 Anticorps anti-cd100 et son utilisation

Country Status (2)

Country Link
CN (1) CN117866091A (fr)
WO (1) WO2024078558A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060233793A1 (en) * 2003-01-31 2006-10-19 Marie-Francoise Belin Use of anti-cd100 antibodies
US20080219971A1 (en) * 2007-02-14 2008-09-11 Vaccinex, Inc. Human anti-cd100 antibodies
CN101312988A (zh) * 2005-10-11 2008-11-26 杜门蒂斯有限公司 抗体多肽文库筛选和选择的抗体多肽类
WO2010129917A2 (fr) * 2009-05-08 2010-11-11 Vaccinex, Inc. Anticorps anti-cd100 et leurs méthodes d'utilisation
CN110713537A (zh) * 2018-07-13 2020-01-21 上海开拓者生物医药有限公司 一种sema4d抗体及其制备方法和应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060233793A1 (en) * 2003-01-31 2006-10-19 Marie-Francoise Belin Use of anti-cd100 antibodies
CN101312988A (zh) * 2005-10-11 2008-11-26 杜门蒂斯有限公司 抗体多肽文库筛选和选择的抗体多肽类
US20080219971A1 (en) * 2007-02-14 2008-09-11 Vaccinex, Inc. Human anti-cd100 antibodies
WO2010129917A2 (fr) * 2009-05-08 2010-11-11 Vaccinex, Inc. Anticorps anti-cd100 et leurs méthodes d'utilisation
CN110713537A (zh) * 2018-07-13 2020-01-21 上海开拓者生物医药有限公司 一种sema4d抗体及其制备方法和应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
FISHER TERRENCE L., REILLY CHRISTINE A., WINTER LAURIE A., PANDINA TRACY, JONASON ALAN, SCRIVENS MARIA, BALCH LESLIE, BUSSLER HOLM: "Generation and preclinical characterization of an antibody specific for SEMA4D", MABS, LANDES BIOSCIENCE, US, vol. 8, no. 1, 2 January 2016 (2016-01-02), US , pages 150 - 162, XP055788166, ISSN: 1942-0862, DOI: 10.1080/19420862.2015.1102813 *

Also Published As

Publication number Publication date
CN117866091A (zh) 2024-04-12

Similar Documents

Publication Publication Date Title
JP6839772B2 (ja) 抗PD−1抗体との組み合わせのための抗Tim−3抗体
JP6839761B2 (ja) 抗PD−L1抗体との組み合わせのための抗Tim−3抗体
TWI754800B (zh) 新型抗ox40/pd-l1雙特異性抗體分子、新型抗vegf/gitr雙特異性抗體分子及其用途
AU2018274919A1 (en) Bispecific heterodimeric diabodies and uses thereof
CN110366560B (zh) 抗b7-h4抗体、其抗原结合片段及其医药用途
JP2019503687A (ja) 抗pd−l1抗体およびその使用
TW201900679A (zh) 細胞毒性t淋巴球相關蛋白4 (ctla-4) 之新穎單株抗體
CN111196854A (zh) Ox40抗体及其制备方法和应用
EP4101867A1 (fr) Anticorps bispécifique anti-cd3 et anti-cd123 et son utilisation
WO2021219048A1 (fr) Anticorps bispécifique ciblant nkg2a et pd-l1, et utilisation
TW202132351A (zh) 抗cd47/抗pd-l1抗體及其應用
JP2022514693A (ja) Muc18に特異的な抗体
EP4292611A1 (fr) Anticorps anti-cd112r et son utilisation
WO2022105811A1 (fr) Anticorps cd19 humanisé et son utilisation
WO2021143914A1 (fr) Anticorps anti-ox40, son procédé de production et son application
WO2019192493A1 (fr) Anticorps monoclonal anti-lag-3 humain et son utilisation
JP2022514786A (ja) Muc18に特異的な抗体
JP2021501569A (ja) Pdl1を標的とする抗体及びそれを用いる方法
TW202317639A (zh) 抗人cd3抗體及其應用
CN117751143A (zh) 抗pvrig/抗tigit双特异性抗体和应用
WO2024078558A1 (fr) Anticorps anti-cd100 et son utilisation
CN113637075A (zh) 双特异性抗原结合分子及其医药用途
WO2024114244A1 (fr) Anticorps anti-cd100 et son utilisation
WO2024061272A1 (fr) Anticorps anti-pd-l1 et son utilisation
TWI833227B (zh) 靶向pd-l1和cd73的特異性結合蛋白及其應用

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: 23876758

Country of ref document: EP

Kind code of ref document: A1