Nothing Special   »   [go: up one dir, main page]

WO2022242758A1 - Anticorps anti-cd73 et son utilisation - Google Patents

Anticorps anti-cd73 et son utilisation Download PDF

Info

Publication number
WO2022242758A1
WO2022242758A1 PCT/CN2022/094182 CN2022094182W WO2022242758A1 WO 2022242758 A1 WO2022242758 A1 WO 2022242758A1 CN 2022094182 W CN2022094182 W CN 2022094182W WO 2022242758 A1 WO2022242758 A1 WO 2022242758A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
antigen
amino acid
binding fragment
Prior art date
Application number
PCT/CN2022/094182
Other languages
English (en)
Chinese (zh)
Inventor
黄贤明
岳睿
张慧
陈振埕
梁世德
Original Assignee
百奥泰生物制药股份有限公司
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 百奥泰生物制药股份有限公司 filed Critical 百奥泰生物制药股份有限公司
Publication of WO2022242758A1 publication Critical patent/WO2022242758A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • 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
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705

Definitions

  • the invention belongs to the field of biomedicine, and in particular relates to an anti-CD73 antibody and its application.
  • CD73 (cluster of differentiation 73) is also known as extracellular 5'-nucleotidase, which in humans is encoded by the NT5E gene.
  • CD73 is a glycoprotein fixed on the cell surface by glycosylphosphatidylinositol, which has enzymatic activity and can convert AMP into adenosine.
  • CD73 is expressed on various cell types such as endothelial cells, lymphocyte subsets, stromal cells and tumor cells.
  • CD73 catalyzes the generation of adenosine from AMP, which can bind to the adenosine receptor on the surface of T cells, and inhibit the expansion and immune activity of T cells, thus forming an immunosuppressive microenvironment.
  • knocking out CD73 or targeting CD73 enzymatic activity will show certain anti-tumor effects; at the same time, targeting CD73 can significantly improve the efficacy of monoclonal antibodies such as PD-L1/PD1, CTLA4 or 4-1BB. Antitumor effect.
  • Target CD73 to inhibit its enzymatic activity and relieve the adenosine-mediated immunosuppressive microenvironment to achieve anti-tumor effects. Studies have shown that certain anti-CD73 antibodies can increase the activity of B cells after binding to B cells, and contribute to the production of antibodies and the formation of immune memory.
  • CD73 is a potential target for cancer therapy.
  • the present invention provides anti-CD73 antibodies or antigen-binding fragments. These antibodies or antigen-binding fragments can specifically bind to CD73, block the immunosuppressive signaling pathway downstream of CD73, and help the immune system to clear tumor cells.
  • Some embodiments provide antibodies or antigen-binding fragments that specifically bind CD73 and that comprise one or more of the following amino acid sequences:
  • HCDR1 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO: 1 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 1;
  • HCDR2 comprising, or consisting of, the amino acid sequence shown in SEQ ID NO: 2 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 2;
  • HCDR3 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO:3 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO:3;
  • LCDR1 comprising, or consisting of, the amino acid sequence shown in SEQ ID NO: 4 or an amino acid sequence with a single site substitution, deletion or insertion compared to SEQ ID NO: 4;
  • LCDR2 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO:5 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO:5;
  • LCDR3 which comprises an amino acid sequence as shown in any one of SEQ ID NO:6-9 or has a single site substitution, deletion or An inserted amino acid sequence, or consisting of it.
  • the antibody or antigen-binding fragment specifically binds CD73 and comprises:
  • HCDR1 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO: 1 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 1;
  • HCDR2 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 2 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 2;
  • HCDR3 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO: 3 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 3.
  • HCDR1 comprises or consists of the amino acid sequence shown in SEQ ID NO:1
  • HCDR2 comprises or consists of the amino acid sequence shown in SEQ ID NO:2
  • HCDR3 comprises the amino acid sequence shown in SEQ ID NO:3 An amino acid sequence or consisting of it.
  • the antibody or antigen-binding fragment specifically binds CD73 and comprises:
  • LCDR1 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO:4 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO:4;
  • LCDR2 which comprises or consists of the amino acid sequence shown in SEQ ID NO: 5 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 5;
  • LCDR3 which comprises an amino acid sequence as shown in any one of SEQ ID NO:6-9 or has a single site substitution, deletion or An inserted amino acid sequence, or consisting of it.
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:4 or consists of it
  • LCDR2 comprises the amino acid sequence shown in SEQ ID NO:5 or consists of it
  • LCDR3 comprises the amino acid sequence shown in SEQ ID NO:6-9 The amino acid sequence shown in any one or consists of it.
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:4 or consists of it
  • LCDR2 comprises the amino acid sequence shown in SEQ ID NO:5 or consists of it
  • LCDR3 comprises the amino acid sequence shown in SEQ ID NO:6 An amino acid sequence or consisting of it.
  • LCDR1 comprises or consists of the amino acid sequence shown in SEQ ID NO:4, LCDR2 comprises or consists of the amino acid sequence shown in SEQ ID NO:5, LCDR3 comprises the amino acid sequence shown in SEQ ID NO:7 An amino acid sequence or consisting of it. In some embodiments, LCDR1 comprises or consists of the amino acid sequence shown in SEQ ID NO:4, LCDR2 comprises or consists of the amino acid sequence shown in SEQ ID NO:5, LCDR3 comprises the amino acid sequence shown in SEQ ID NO:8 An amino acid sequence or consisting of it.
  • LCDR1 comprises the amino acid sequence shown in SEQ ID NO:4 or consists of it
  • LCDR2 comprises the amino acid sequence shown in SEQ ID NO:5 or consists of it
  • LCDR3 comprises the amino acid sequence shown in SEQ ID NO:9 An amino acid sequence or consisting of it.
  • the antibody or antigen-binding fragment specifically binds CD73 and comprises:
  • HCDR1 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO: 1 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 1;
  • HCDR2 comprising, or consisting of, the amino acid sequence shown in SEQ ID NO: 2 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 2;
  • HCDR3 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO:3 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO:3;
  • LCDR1 comprising, or consisting of, an amino acid sequence as shown in SEQ ID NO:4 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO:4;
  • LCDR2 which comprises or consists of the amino acid sequence shown in SEQ ID NO: 5 or an amino acid sequence having a single site substitution, deletion or insertion compared to SEQ ID NO: 5;
  • LCDR3 which comprises an amino acid sequence as shown in any one of SEQ ID NO:6-9 or has a single site substitution, deletion or An inserted amino acid sequence, or consisting of it.
  • the substitutions are conservative amino acid substitutions.
  • the antibody or antigen-binding fragment comprises HCDR1 as set forth in SEQ ID NO:1, HCDR2 as set forth in SEQ ID NO:2, HCDR3 as set forth in SEQ ID NO:3, HCDR3 as set forth in SEQ ID NO:3, LCDR1 shown in NO:4, LCDR2 as shown in SEQ ID NO:5 and LCDR3 as shown in SEQ ID NO:6.
  • the antibody or antigen-binding fragment comprises HCDR1 as set forth in SEQ ID NO:1, HCDR2 as set forth in SEQ ID NO:2, HCDR3 as set forth in SEQ ID NO:3, HCDR3 as set forth in SEQ ID NO:3, LCDR1 shown in NO:4, LCDR2 as shown in SEQ ID NO:5 and LCDR3 as shown in SEQ ID NO:7.
  • the antibody or antigen-binding fragment comprises HCDR1 as set forth in SEQ ID NO:1, HCDR2 as set forth in SEQ ID NO:2, HCDR3 as set forth in SEQ ID NO:3, HCDR3 as set forth in SEQ ID NO:3, LCDR1 shown in NO:4, LCDR2 as shown in SEQ ID NO:5 and LCDR3 as shown in SEQ ID NO:8.
  • the antibody or antigen-binding fragment comprises HCDR1 as set forth in SEQ ID NO:1, HCDR2 as set forth in SEQ ID NO:2, HCDR3 as set forth in SEQ ID NO:3, HCDR3 as set forth in SEQ ID NO:3, LCDR1 shown in NO:4, LCDR2 as shown in SEQ ID NO:5 and LCDR3 as shown in SEQ ID NO:9.
  • the heavy chain variable region of the antibody or antigen-binding fragment comprises the amino acid sequence set forth in SEQ ID NO: 10, a sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 10 , or an amino acid sequence having one or more conservative amino acid substitutions compared to the sequence shown in SEQ ID NO: 10, or consisting of; and/or
  • the light chain variable region of the antibody or antigen-binding fragment comprises the amino acid sequence shown in any one of SEQ ID NO: 11-14, compared with the sequence shown in any one of SEQ ID NO: 11-14 has at least A sequence with 80% identity, or an amino acid sequence having one or more conservative amino acid substitutions compared with the sequence shown in any one of SEQ ID NO: 11-14, or consists of it.
  • the heavy chain variable region of the antibody or antigen-binding fragment comprises the sequence shown in SEQ ID NO: 10
  • the light chain variable region of the antibody or antigen-binding fragment comprises SEQ ID NO : the sequence shown in any one of 11-14.
  • the heavy chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 10, and the light chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 11 sequence shown. In some embodiments, the heavy chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 10, and the light chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 12 sequence shown. In some embodiments, the heavy chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 10, and the light chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 13 sequence shown. In some embodiments, the heavy chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 10, and the light chain variable region of the antibody or antigen-binding fragment comprises the sequence set forth in SEQ ID NO: 14 sequence shown.
  • the antibody or antigen-binding fragment further comprises a heavy chain constant region, a light chain constant region, an Fc region, or a combination thereof.
  • the light chain constant region is a kappa or lambda chain constant region.
  • the antibody or antigen-binding fragment is an isotype of IgG, IgM, IgA, IgE, or IgD.
  • the isotype is IgGl, IgG2, IgG3 or IgG4.
  • the antibody or antigen-binding fragment is a murine antibody, a chimeric antibody, a humanized antibody, or a fully human antibody.
  • the Fc is a variant Fc region.
  • the variant Fc region has one or more amino acid modifications, such as substitutions, deletions or insertions, relative to the parental Fc region.
  • the amino acid modification of the Fc region alters effector function activity relative to the activity of the parental Fc region.
  • the variant Fc region may have altered (i.e., increased or decreased) antibody-dependent cellular cytotoxicity (ADCC), complement-mediated cytotoxicity (CDC), phagocytosis, opsonization, or cell binding .
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-mediated cytotoxicity
  • phagocytosis opsonization
  • cell binding i.e., phagocytosis
  • amino acid modifications of the Fc region can alter the affinity of the variant Fc region for Fc ⁇ R (Fc ⁇ receptor) relative to the parent Fc region.
  • the Fc region is derived from IgGl or IgG4.
  • the Fc region mutation is N297A, L234A, or L235A (Eu numbering).
  • the Fc region mutation is E345R or S440Y (Eu numbering).
  • the antibody or antigen-binding fragment is a scFv, Fab, Fab' or F(ab) 2 . In some embodiments, the antibody or antigen-binding fragment is a monoclonal antibody.
  • the heavy chain constant region of the antibody or antigen-binding fragment comprises an amino acid sequence as shown in SEQ ID NO: 15 or 16, or has at least an amino acid sequence compared to the sequence shown in SEQ ID NO: 15 or 16 A sequence of 80% identity, or an amino acid sequence having one or more conservative amino acid substitutions compared to the sequence shown in SEQ ID NO: 15 or 16, or consisting of it; and/or
  • the light chain constant region of the antibody or antigen-binding fragment comprises an amino acid sequence as shown in SEQ ID NO: 17, or a sequence with at least 80% identity compared with the sequence shown in SEQ ID NO: 17, or a sequence with SEQ ID NO: 17 ID NO: 17 refers to an amino acid sequence having one or more conservative amino acid substitutions compared to the sequence, or consisting of it.
  • the heavy chain constant region of the antibody or antigen-binding fragment comprises an amino acid sequence as shown in SEQ ID NO: 15, and the light chain constant region of the antibody or antigen-binding fragment comprises an amino acid sequence such as SEQ ID The sequence shown in NO:17.
  • the heavy chain constant region of the antibody or antigen-binding fragment comprises an amino acid sequence as shown in SEQ ID NO: 16 and the light chain constant region of the antibody or antigen-binding fragment comprises an amino acid sequence such as SEQ ID The sequence shown in NO:17.
  • the heavy chain of the antibody comprises an amino acid sequence as set forth in SEQ ID NO: 18 or 19, a sequence having at least 80% identity compared to the sequence set forth in SEQ ID NO: 18 or 19, Or an amino acid sequence having one or more conservative amino acid substitutions compared to the sequence shown in SEQ ID NO: 18 or 19, or consisting of it; and/or
  • the light chain of the antibody comprises an amino acid sequence as shown in any one of SEQ ID NO:20-23, which has at least 80% identity compared with the sequence shown in any one of SEQ ID NO:20-23 sequence, or an amino acid sequence having one or more conservative amino acid substitutions compared to the sequence shown in any one of SEQ ID NO: 20-23, or consisting of.
  • the heavy chain of the antibody comprises or consists of an amino acid sequence as shown in SEQ ID NO: 18 or 19; and/or
  • the light chain of the antibody comprises an amino acid sequence as shown in any one of SEQ ID NO: 20-23, or consists of it.
  • the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 20. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 21. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 22. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 23.
  • the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 20. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 21. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 22. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO: 23.
  • the antibody or antigen-binding fragment is a monoclonal antibody (including a full-length monoclonal antibody), a multispecific antibody, or an antigen-binding fragment (eg, a bispecific antibody or antigen-binding fragment thereof).
  • the antibody has two heavy chains of identical sequence and two light chains of identical sequence, and the Fc regions pair to form disulfide bonds.
  • the antigen-binding fragment is Fab, Fab', Fv or scFv.
  • the antibody or antigen-binding fragment is an isolated antibody or antigen-binding fragment.
  • the invention also provides nucleic acids encoding said antibodies or antigen-binding fragments.
  • the nucleic acid is an isolated nucleic acid.
  • the nucleic acid sequence is selected from the nucleic acid sequences listed in Table 6.
  • the present invention also provides a vector comprising the nucleic acid.
  • the vector comprising the nucleic acid is a nucleic acid fragment, plasmid, phage, or virus.
  • the vector is an isolated vector.
  • the present invention also provides a host cell comprising the nucleic acid or vector.
  • the host cell is an isolated host cell.
  • the host cells are CHO cells, HEK cells (such as HEK293F cells), BHK cells, Cos1 cells, Cos7 cells, CV1 cells or murine L cells.
  • the present invention also provides a method for producing the antibody or antigen-binding fragment, which comprises culturing a host cell comprising a nucleic acid encoding the antibody or antigen-binding fragment in a culture medium.
  • the method further comprises purifying the antibody or antigen-binding fragment. Purification can be carried out by conventional methods, such as centrifuging the cell suspension first, collecting the supernatant, and centrifuging again to further remove impurities. Methods such as Protein A affinity column and ion exchange column can be used to purify antibody protein.
  • the present invention also provides a pharmaceutical composition, which comprises the antibody or antigen-binding fragment, and pharmaceutically acceptable auxiliary materials.
  • the present invention also provides methods and uses for preventing or treating tumors, cancers or infections.
  • a method for treating or ameliorating a tumor, cancer or infection comprising administering to a patient an effective dose of the antibody or antigen-binding fragment.
  • use of the antibody or antigen-binding fragment for treating or ameliorating tumors, cancers or infections is provided.
  • the use of the antibody or antigen-binding fragment in the preparation of a medicament for treating or improving tumor, cancer or infection is provided.
  • the cancers and tumors include, but are not limited to, breast cancer, endocrine cancer, neuroendocrine cancer, eye cancer, genitourinary cancer, germ cell cancer, gynecological cancer, head and neck cancer, hematology/blood cancer, musculoskeletal Cancer, nerve cancer, respiratory cancer/thorax cancer, bladder cancer, colon cancer, rectal cancer, lung cancer, endometrial cancer, kidney cancer, pancreatic cancer, liver cancer, stomach cancer, testicular cancer, esophagus cancer, prostate cancer, brain cancer, cervical cancer cancer, ovarian and thyroid cancer.
  • cancers and tumors include, but are not limited to, leukemias, melanomas, and lymphomas.
  • leukemia includes, but is not limited to, lymphocytic or myeloid leukemia, such as acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), acute myeloid (myelocytic) leukemia (AML) , chronic myelogenous leukemia (CML), hairy cell leukemia, T-cell prolymphocytic leukemia, large granular lymphocytic leukemia, or adult T-cell leukemia.
  • lymphomas include, but are not limited to, histiocytic lymphoma, follicular lymphoma, and Hodgkin's lymphoma.
  • infections include, but are not limited to, chronic infectious diseases such as HIV, HBV, HCV, COVID-19, and HSV, among others.
  • the invention also provides diagnostic methods and uses.
  • a method of detecting CD73 expression in a sample contacting the sample with the antibody or antigen-binding fragment such that the antibody or antigen-binding fragment binds to CD73, and detecting its binding, i.e., the expression of CD73 in the sample. content.
  • the use of the antibody or antigen-binding fragment in the preparation of a kit for diagnosing or prognosing cancer or tumor is provided.
  • a diagnostic or prognostic kit comprising the antibody or antigen-binding fragment
  • the kit further includes a second antibody that specifically recognizes the anti-CD73 antibody; any
  • the second antibody also includes detectable labels, such as radioactive isotopes, fluorescent substances, chemiluminescent substances, colored substances or enzymes; optionally, the kit is used to detect the presence of CD73 in the sample or its Level;
  • the kit further includes antibodies or antigen-binding fragments against other antigens, and/or cytotoxic agents, and optionally, instructions for use.
  • the invention provides an anti-CD73 antibody and its application.
  • the antibody or antigen-binding fragment of the invention can specifically bind to CD73, block the immunosuppressive signal pathway downstream of CD73, and help the immune system to clear tumor cells.
  • the antibody or antigen-binding fragment of the present invention is used for treating or improving tumor, cancer or infection, and also for the diagnosis and prognosis of cancer or tumor.
  • Figure 1 shows the binding of scFv to the antigen hCD73; wherein, BLK represents the control.
  • Figure 2 shows that anti-CD73 antibodies inhibit the enzymatic activity of cell membrane CD73.
  • Figure 3A shows the results of flow cytometric detection of CD73 (PE-labeled) in peripheral blood T cells, where A represents the peak area.
  • Figure 3B shows the results of flow cytometric detection of CD73 (PE-labeled) in peripheral blood B cells, where A represents the peak area.
  • Figure 4 shows that anti-CD73 antibody relieves the inhibition of T cell proliferation by AMP.
  • Figure 5 shows that the anti-CD73 antibody up-regulates the expression of B cell activation marker (CD69); wherein, the abscissa BLK represents the control, and the ordinate represents the average fluorescence intensity of CD69 in B cells.
  • CD69 B cell activation marker
  • an entity refers to one or more such entities, for example "an antibody” should be understood as one or more antibodies, therefore, the term “a” (or “an” ), “one or more” and “at least one” may be used interchangeably herein.
  • compositions, methods, etc. include the listed elements, such as components or steps, but not exclude others.
  • Consisting essentially of means that the compositions and methods exclude other elements that substantially affect the characteristics of the combination, but do not exclude elements that do not substantially affect the composition or method.
  • Consisting of means excluding elements not specifically recited.
  • polypeptide is intended to encompass the singular as well as the plural “polypeptides” and refers to a molecule formed of amino acid monomers linked linearly by amide bonds (also known as peptide bonds).
  • polypeptide refers to any chain or chains of two or more amino acids, and does not refer to a specific length of the product.
  • the definition of “polypeptide” includes peptide, dipeptide, tripeptide, oligopeptide, "protein”, “amino acid chain” or any other term used to refer to a chain of two or more amino acids, and the term “polypeptide” may Used in place of, or interchangeably with, any of the above terms.
  • polypeptide is also intended to refer to the products of post-expression modifications of the polypeptide, including but not limited to glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or non-natural Amino acid modifications that occur.
  • a polypeptide may be derived from natural biological sources or produced by recombinant techniques, but it need not be translated from a specified nucleic acid sequence, it may be produced by any means including chemical synthesis.
  • amino acid refers to an organic compound containing both amino and carboxyl groups, such as an ⁇ -amino acid, which can be encoded by a nucleic acid directly or in the form of a precursor.
  • a single amino acid is encoded by a nucleic acid consisting of three nucleotides (so-called codons or base triplets). Each amino acid is encoded by at least one codon. The fact that the same amino acid is encoded by different codons is called “degeneracy of the genetic code”.
  • Amino acids include natural amino acids and unnatural amino acids.
  • Natural amino acids include alanine (three-letter code: ala, one-letter code: A), arginine (arg, R), asparagine (asn, N), aspartic acid (asp, D), cysteine amino acid (cys, C), glutamine (gln, Q), glutamic acid (glu, E), glycine (gly, G), histidine (his, H), isoleucine (ile, I ), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y) and valine (val, V).
  • a “conservative amino acid substitution” refers to the replacement of one amino acid residue with another amino acid residue containing a side chain (R group) of similar chemical properties (eg, charge or hydrophobicity). In general, conservative amino acid substitutions are unlikely to substantially alter the functional properties of a protein.
  • classes of amino acids that contain chemically similar side chains include: 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic hydroxyl side chains: serine and threonine 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, Arginine and histidine; 6) acidic side chains: aspartic acid and glutamic acid.
  • the number of amino acids in the "conservative amino acid substitution of VL, VH” is about 1, about 2, about 3, about 4, about 5, about 6, about 8, about 9, about 10, about 11, about 13, about 14, about 15 conservative amino acid substitutions, or a range between any two of these values (inclusive), or any value therein.
  • the number of amino acids in the "heavy chain constant region, light chain constant region, heavy chain or light chain conservative amino acid substitution” is about 1, about 2, about 3, about 4, about 5, about 6, about 8, about 9, about 10, about 11, about 13, about 14, about 15, about 18, about 19, about 22, about 24, about 25, about 29 , about 31, about 35, about 38, about 41, about 45 conservative amino acid substitutions, or a range between any two of these values (inclusive) or any value therein.
  • isolated used in the present invention with respect to cells, nucleic acids, polypeptides, antibodies, etc., for example, "isolated" DNA, RNA, polypeptides, antibodies refers to the isolated components of the cell's natural environment, such as DNA or RNA. One or more of the isolated molecules.
  • isolated as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or cell culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • isolated nucleic acid is intended to include fragments of nucleic acid that do not occur in nature, and do not exist in nature.
  • isolated is also used herein to refer to cells or polypeptides that are separated from other cellular proteins or tissues.
  • Isolated polypeptide is intended to include purified and recombinant polypeptides.
  • Isolated polypeptides, antibodies, etc. will usually be prepared by at least one purification step.
  • the purity of the isolated nucleic acid, polypeptide, antibody, etc. is at least about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or some of these values The range (inclusive) between any two values of , or any value therein.
  • polypeptides or polynucleotides refers to polypeptides or polynucleotides, meaning forms of polypeptides or polynucleotides that do not occur in nature, non-limiting examples may be produced by combination of polynucleotides or polynucleotides that do not normally exist or peptide.
  • Homology refers to the sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing the alignable positions in each sequence. When a position in the sequences being compared is occupied by the same base or amino acid, then the molecules are homologous at that position. The degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences.
  • At least 80% identity is about 80% identity, about 81% identity, about 82% identity, about 83% identity, about 85% identity, about 86% identity, about 87% identity, About 88% identity, about 90% identity, about 91% identity, about 92% identity, about 94% identity, about 95% identity, about 98% identity, about 99% identity, or these A range (inclusive) between any two values in Numeric or any value therein.
  • a polynucleotide or polynucleotide sequence has a certain percentage (eg 90%, 95%, 98% or 99%) "identity or sequence identity" with another sequence
  • identity or sequence identity refers to the percentage of bases (or amino acids) that are identical in the two sequences being compared when the sequences are aligned. This alignment and percent identity or sequence identity can be determined using visual inspection or software programs known in the art, such as those described by Ausubel et al.eds. (2007) in Current Protocols in Molecular Biology. It is preferred to use the default parameters for the alignment.
  • Biologically equivalent polynucleotides are polynucleotides that share the above indicated percentages of identity and encode a polypeptide having the same or similar biological activity.
  • a polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A), cytosine (C), guanine (G), thymine (T), or when a polynucleotide In the case of RNA, thymine is replaced by uracil (U).
  • a "polynucleotide sequence” may be denoted by the letters of the polynucleotide molecule. This letter designation can be entered into a database in a computer with a central processing unit and used in bioinformatics applications such as for functional genomics and homology searches.
  • polynucleotide and “oligonucleotide” are used interchangeably to refer to a polymeric form of nucleotides of any length, whether deoxyribonucleotides or ribonucleotides or analogs thereof.
  • a polynucleotide can have any three-dimensional structure and can perform any function, known or unknown.
  • polynucleotides genes or gene fragments (e.g., probes, primers, EST or SAGE tags), exons, introns, messenger RNA (mRNA), transfer RNA, ribose Somatic RNA, ribozyme, cDNA, dsRNA, siRNA, miRNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes and primers.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs.
  • nucleotides can be made before or after assembly of the polynucleotide.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • Polynucleotides may be further modified after polymerization, for example by conjugation with labeling components.
  • the term also refers to double-stranded and single-stranded molecules. Unless otherwise stated or required, any embodiment of a polynucleotide of the present disclosure includes the double-stranded form and each of the two complementary single-stranded forms known or predicted to constitute the double-stranded form.
  • encoding when applied to a polynucleotide refers to a polynucleotide which is said to "encode” a polypeptide which, in its native state or when manipulated by methods well known to those skilled in the art, is transcribed and/or Or translation may result in the polypeptide and/or fragments thereof.
  • Antibodies and antigen-binding fragments disclosed in the present invention include but are not limited to polyclonal, monoclonal, multispecific, fully human, humanized, primatized, chimeric antibodies, single-chain antibodies, epitope-binding fragments (such as Fab, Fab' and F(ab') 2 , single chain Fv (scFv)).
  • Antibody and antigen-binding fragment refer to a polypeptide or polypeptide complex that specifically recognizes and binds to an antigen.
  • Antibodies can be whole antibodies and any antigen-binding fragments thereof or single chains thereof.
  • the term “antibody” thus includes any protein or peptide whose molecule contains at least a portion of an immunoglobulin molecule that has the biological activity to bind an antigen.
  • Antibodies and antigen-binding fragments include, but are not limited to, complementarity determining regions (CDRs), heavy chain variable regions (VH), light chain variable regions (VL), heavy chain constant regions of heavy or light chains or ligand-binding portions thereof (CH), light chain constant region (CL), framework region (FR) or any portion thereof, or at least a portion of a binding protein.
  • the CDR regions include the CDR regions of the light chain (LCDR1-3) and the CDR regions of the heavy chain (HCDR1-3).
  • Antibodies and antigen-binding fragments can specifically recognize and bind to one or more (eg, two) antigen polypeptides or polypeptide complexes.
  • Antibodies or antigen-binding fragments that specifically recognize and bind multiple (eg, two) antigens may be referred to as multispecific (eg, bispecific) antibodies or antigen-binding fragments.
  • antibody fragment refers to a part of an antibody, and the composition of the antibody fragment of the present invention may be similar to F(ab') 2 , F(ab) 2 , Fab', Fab in monospecific antibody fragments , Fv, scFv, etc. Regardless of their structure, antibody fragments bind to the same antigen recognized by the intact antibody.
  • antibody fragment includes aptamers, Spiegelmers and diabodies.
  • antiigen-binding fragment also includes any synthetic or genetically engineered protein that functions as an antibody by binding to a specific antigen to form a complex.
  • Single-chain variable fragment refers to a fusion protein of the variable regions of the heavy (VH) and light (VL) chains of an immunoglobulin. In some aspects, these regions are linked to short linker peptides of 10 to about 25 amino acids. Linkers can be rich in glycine for flexibility, and serine or threonine for solubility, and can connect the N-terminus of VH to the C-terminus of VL, or vice versa. Although the protein has had its constant regions removed and a linker introduced, it retains the specificity of the original immunoglobulin. scFv molecules are generally known in the art and are described, for example, in US Patent No. 5,892,019.
  • antibody includes a wide variety of polypeptides that can be distinguished biochemically.
  • classes of heavy chains include gamma, mu, alpha, delta or epsilon ( ⁇ , ⁇ , ⁇ , ⁇ , ⁇ ), with some subclasses (eg ⁇ 1- ⁇ 4).
  • the nature of this chain determines the "class” of the antibody as IgG, IgM, IgA, IgG or IgE, respectively.
  • the immunoglobulin subclasses (isotypes), eg, IgGl, IgG2, IgG3, IgG4, IgG5, etc., are well characterized and the functional specificities conferred are also known. All immunoglobulin classes are within the scope of the present disclosure. In some embodiments, the immunoglobulin molecule is of the IgG class.
  • Light chains can be classified as kappa ( ⁇ ) or lambda ( ⁇ ). Each heavy chain can be associated with a kappa or lambda light chain.
  • kappa
  • lambda
  • Each heavy chain can be associated with a kappa or lambda light chain.
  • immunoglobulins are produced by hybridomas, B cells, or genetically engineered host cells, their light and heavy chains are joined by covalent bonds, and the "tail" portions of the two heavy chains are linked by covalent disulfide bonds or non-covalent bonding.
  • the amino acid sequence extends from the N-terminus at the forked end of the Y configuration to the C-terminus at the bottom of each chain.
  • the variable region of the immunoglobulin kappa light chain is V ⁇ ; the variable region of the immunoglobulin lambda light chain is V ⁇ .
  • the terms “constant” and “variable” are used according to function.
  • the light chain variable (VL) and heavy chain variable (VH) portions determine antigen recognition and specificity.
  • the constant regions of the light and heavy chains confer important biological properties such as secretion, transplacental movement, Fc receptor binding, complement fixation, etc. By convention, the numbering of constant regions increases as they become farther away from the antigen-binding site or amino 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 comprise the carboxy-terminal ends of the heavy and light chains, respectively.
  • each antigen-binding domain In naturally occurring antibodies, the six “complementarity determining regions" or “CDRs” present in each antigen-binding domain are short, A non-contiguous sequence of amino acids that specifically binds to an antigen. The remaining other amino acids in the antigen-binding domain, referred to as the "framework" regions, show less inter-molecular variability.
  • the framework regions mostly adopt a ⁇ -sheet conformation, and the CDRs form loop structures attached to them, or in some cases form part of the ⁇ -sheet structure. Thus, the framework regions position the CDRs in the correct orientation by forming a scaffold through non-covalent interchain interactions.
  • the antigen-binding domain with the CDRs in specific positions forms a surface complementary to the epitope on the antigen that facilitates the non-covalent binding of the antibody to its antigenic epitope.
  • a surface complementary to the epitope on the antigen that facilitates the non-covalent binding of the antibody to its antigenic epitope.
  • those of ordinary skill in the art can identify the amino acids comprising CDR and framework regions by known methods (see Kabat, E., et al., U.S. Department of Health and Human Services, Sequences of Proteins of Immunological Interest, (1983) and Chothia and Lesk, J. Mol. Biol., 196:901-917 (1987)).
  • CDR complementarity determining regions
  • CDRs as defined by Kabat and Chothia include overlapping or subsets of amino acid residues when compared to each other. Nevertheless, it is within the scope of the invention to use either definition to refer to the CDRs of an antibody or variant thereof.
  • the exact residue numbers comprising a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can generally determine which specific residues are included in the CDRs based on the amino acid sequence of the variable region of the antibody.
  • Kabat et al. also defined a numbering system applicable to the variable region sequences of any antibody.
  • One of ordinary skill in the art can apply this "Kabat numbering" system to any variable region sequence independently of other experimental data other than the sequence itself.
  • “Kabat numbering” refers to the numbering system proposed by Kabat et al., U.S. Dept. of Health and Human Services in "Sequence of Proteins of Immunological Interest” (1983).
  • Antibodies can also use the EU or Chothia numbering system.
  • Antibodies disclosed herein may be derived from any animal, including birds and mammals.
  • the antibody is of human, murine, donkey, rabbit, goat, camel, llama, horse or chicken origin.
  • the variable regions may be of condricthoid origin (eg, from sharks).
  • a "heavy chain constant region” includes at least one of a CH1 domain, a hinge (eg, upper, middle, and/or lower hinge region) domain, a CH2 domain, a CH3 domain, or a variant or fragment.
  • the heavy chain constant regions of antibodies can be derived from different immunoglobulin molecules.
  • the heavy chain constant region of a polypeptide can include a CH1 domain derived from an IgG 1 molecule and a hinge region derived from an IgG 3 molecule.
  • the heavy chain constant region may comprise a hinge region derived in part from an IgG 1 molecule and in part from an IgG 3 molecule.
  • part of the heavy chain may comprise a chimeric hinge region derived partly from an IgG 1 molecule and partly from an IgG4 molecule .
  • a “light chain constant region” includes a portion of the amino acid sequence from an antibody light chain.
  • the light chain constant region comprises at least one of a constant kappa domain or a constant lambda domain.
  • a “light chain-heavy chain pair” refers to a collection of light and heavy chains that can form dimers through disulfide bonds between the CL domain of the light chain and the CH1 domain of the heavy chain. The four chains are linked by disulfide bonds in a "Y" configuration, with the light chain starting at the mouth of the "Y” and continuing through the variable region surrounding the heavy chain.
  • a "VH domain” includes the amino-terminal variable domain of an immunoglobulin heavy chain, and a "CH1 domain” includes the first (mostly amino-terminal) constant region of an immunoglobulin heavy chain.
  • N297 in the two CH2 domains of the complete natural IgG molecule is connected to a branched carbohydrate chain.
  • the CH3 domain extends from the CH2 domain to the C-terminus of the IgG molecule and contains approximately 108 residues.
  • a "hinge region” includes part of the heavy chain region connecting the CH1 domain and the CH2 domain.
  • the hinge region comprises approximately 25 residues and is flexible, allowing the two N-terminal antigen-binding regions to move independently.
  • the hinge region can be subdivided into three distinct domains: upper, middle, and lower hinge domains (Roux et al., J. Immunol 161:4083 (1998)).
  • Disulfide bond refers to a covalent bond formed between two sulfur atoms.
  • a thiol group of cysteine can form a disulfide bond or bridge with a second thiol group.
  • the CH1 and CL regions are linked by disulfide bonds.
  • a “chimeric antibody” refers to any antibody whose variable regions are obtained or derived from a first species and whose constant regions (which may be complete, partial or modified) are derived from a second species.
  • the variable regions are of non-human origin (eg, mouse or primate) and the constant regions are of human origin.
  • Specific binding or “specific for” generally refers to the complementary binding of an antibody or antigen-binding fragment to a specific antigen through its antigen-binding domain and epitope to form a relatively stable complex.
  • Specificity can be expressed in terms of the relative affinity with which an antibody or antigen-binding fragment binds to a particular antigen or epitope. For example, antibody “A” may be said to have a higher specificity for that antigen than antibody “B” if it has a greater relative affinity for the same antigen than antibody "B”.
  • Specific binding can be described by an equilibrium dissociation constant ( KD ), with a smaller KD implying a tighter binding.
  • An antibody that "specifically binds" antigen a includes an equilibrium dissociation constant K D of antigen a of less than or equal to about 100 nM, less than or equal to about 10 nM, less than or equal to about 5 nM, less than or equal to about 1 nM.
  • Treatment means therapeutic treatment and prophylactic or preventive measures, the purpose of which is to prevent, slow down, ameliorate or stop an undesirable physiological change or disorder, such as the progression of a disease, including but not limited to the following whether detectable or undetectable Relief of symptoms, reduction of disease extent, stabilization of disease state (i.e. not worsening), delay or slowing of disease progression, amelioration, remission, alleviation or disappearance of disease state (whether partial or total), prolongation and Expected survival without treatment, etc.
  • Patients in need of treatment include those who already have a condition or disorder, are susceptible to having a condition or disorder, or are in need of prevention of the condition or disorder, and can or are expected to benefit from the administration of an antibody or pharmaceutical composition disclosed herein for detection , patients who benefit from the diagnostic process and/or treatment.
  • Patient refers to any mammal in need of diagnosis, prognosis, or treatment, including humans, dogs, cats, rabbits, mice, horses, cattle, and the like.
  • EC 50 means half maximum effect concentration (concentration for 50% of maximal effect, EC 50 ) refers to the concentration that can cause 50% of the maximum effect.
  • IC50 means 50% inhibitory concentration, ie the concentration of drug or inhibitor required to inhibit a given biological process by half.
  • the "parental Fc region" in the present invention can be a naturally occurring Fc region, and the gene encoding the Fc region can be from human, mouse, rabbit, camel, monkey, preferably human and mouse; for example, the parental Fc region is SEQ ID NO: 18 Or the Fc region of 19.
  • the present invention provides antibodies or antigen-binding fragments with high affinity for CD73 protein.
  • the anti-CD73 antibody or antigen-binding fragment of the present invention exhibits effective binding activity and biological activity, and is used for treatment and diagnosis.
  • these antibodies or antigen-binding fragments effectively block inhibitory immune checkpoints, activate lymphocytes to release cytokines, and are used to treat various types of cancer, tumor or infection and other related diseases.
  • the antigen-binding fragment is a scFv.
  • the VH in the scFv comprises an amino acid sequence as shown in SEQ ID NO:10, and the VL in the scFv comprises an amino acid sequence as shown in SEQ ID NO:11.
  • the VH in the scFv comprises an amino acid sequence as shown in SEQ ID NO:10, and the VL in the scFv comprises an amino acid sequence as shown in SEQ ID NO:12.
  • the VH in the scFv comprises an amino acid sequence as shown in SEQ ID NO:10, and the VL in the scFv comprises an amino acid sequence as shown in SEQ ID NO:13.
  • the VH in the scFv comprises an amino acid sequence as shown in SEQ ID NO:10, and the VL in the scFv comprises an amino acid sequence as shown in SEQ ID NO:14.
  • the linker connecting the variable region of the heavy chain and the variable region of the light chain in the scFv fragment is (G 4 S) n . In some embodiments, n is 1, 2, 3, 4 or 5.
  • the antibody is IgG1 or IgG4.
  • the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:20. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:21. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:22. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:18, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:23.
  • the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:20. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:21. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:22. In some embodiments, the heavy chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:19, and the light chain of the antibody comprises an amino acid sequence as shown in SEQ ID NO:23.
  • antibodies of the invention contain two heavy chains of identical sequence and two light chains of identical sequence.
  • the sequences of the antibodies or antigen-binding fragments disclosed in the present invention can be replaced, and the amino acid sequence after replacement is different from the naturally occurring amino acid sequence of the antibody.
  • the substituted amino acid sequence may be similar to the original sequence, such as having a certain proportion of identity with the original sequence, for example, it may be about 80%, about 85%, or about 90% identical to the original sequence. , about 95%, about 98%, about 99%, or a range between any two of these values (inclusive), or any value therein.
  • an antibody or antigen-binding fragment comprises an amino acid sequence with one or more modification groups.
  • an antibody or antigen-binding fragment disclosed herein may contain a flexible linker sequence, or may be modified to add functional groups (eg, PEG, drug, toxin, or tag).
  • the antibodies, antigen-binding fragments disclosed herein include derivatives that are modified, that is, modified by covalent linkage of any type of molecule to the antibody or antigen-binding fragment, wherein the covalent linkage does not prevent the antibody or antigen-binding fragment from binding to the epitope combined.
  • Examples including, but not limited to, antibodies or antigen-binding fragments may be glycosylated, acetylated, pegylated, phosphorylated, amidated, derivatized by known protecting/blocking groups, proteolytically cleaved, linked to Cell ligands or other proteins, etc.
  • the antibodies, antigen-binding fragments and modified derivatives thereof disclosed in the present invention include their salts with acids and/or bases.
  • an antibody or antigen-binding fragment can be conjugated to a therapeutic agent, prodrug, peptide, protein, enzyme, virus, lipid, biological response modifier, pharmaceutical agent, or PEG.
  • Antibodies or antigen-binding fragments can be detectably labeled by coupling them to chemiluminescent compounds. The presence of the chemiluminescently labeled antibody or antigen-binding fragment is then determined by detecting the luminescence that occurs during the course of the chemical reaction.
  • chemiluminescent labeling compounds include luminol, isoluminol, aromatic acridinium esters, imidazoles, acridinium salts, and oxalate esters.
  • the invention also discloses polynucleotides or nucleic acid molecules encoding the antibodies, antigen-binding fragments, and derivatives thereof of the invention.
  • the polynucleotide disclosed in the present invention can encode heavy chain variable region, light chain variable region, Fc region, part of heavy chain variable region, part of light chain variable region, heavy chain or light chain, etc. Methods of making antibodies are well known in the art and described herein.
  • antibodies are produced that do not elicit an adverse immune response in the animal (eg, human) to be treated.
  • the antibodies, antigen-binding fragments, or derivatives disclosed herein are modified to reduce their immunogenicity using art-recognized techniques.
  • antibodies can be humanized, primatized, deimmunized or chimeric antibodies can be prepared. These types of antibodies are derived from non-human antibodies, usually murine or primate antibodies, which retain or substantially retain the antigen-binding properties of the parent antibody but are less immunogenic in humans.
  • framework residues in the human framework regions will be replaced by corresponding residues from the CDR donor antibody, such as residues that improve antigen binding.
  • framework substitutions can be identified by methods known in the art, such as by modeling the interaction of CDRs and framework residues to identify framework residues important for antigen binding and by sequence alignment to identify abnormal framework residues at specific positions. (Refer to US Patent 5,585,089; Riechmann et al., Nature 332:323 (1988); the entire contents of which are incorporated herein by reference).
  • Antibodies can be humanized using a variety of techniques known in the art, such as CDR grafting (EP 239,400; WO 91/09967; US Patents 5,225,539, 5,530,101 and 5,585,089), repair or surface rearrangement (EP 592,106; EP 519,596; Padlan, et al., Molecular Immunology 28(4/5):489-498(1991); Studnicka et al., Protein Engineering 7(6):805-814(1994); Roguska, et al., Proc.Natl . Sci. USA 91:969-973 (1994)), and chain rearrangements (US Patent 5,565,332), the entire contents of which are incorporated herein by reference.
  • CDR grafting EP 239,400; WO 91/09967; US Patents 5,225,539, 5,530,101 and 5,585,089)
  • repair or surface rearrangement EP 592,106; EP 519,596; Padlan,
  • Deimmunization can also be used to reduce the immunogenicity of antibodies.
  • the term "deimmunization” includes altering antibodies to modify T cell epitopes (see eg WO/9852976 A1 and WO/0034317 A2).
  • the heavy and light chain variable region sequences from a starting antibody are analyzed and a human T cell epitope "map" from each variable region is generated, showing the epitopes relative to the complementarity determining regions (CDRs) and the positions of other key residues within the sequence.
  • CDRs complementarity determining regions
  • Individual T-cell epitopes from T-cell epitope maps are analyzed to identify alternative amino acid substitutions with lower risk of altering antibody activity.
  • a series of alternative heavy chain variable region sequences and light chain variable region sequences comprising combinations of amino acid substitutions are designed and these sequences are subsequently incorporated into a series of binding polypeptides.
  • Genes for the complete heavy and light chains containing the modified variable and human constant regions are then cloned into expression vectors, and the plasmids are subsequently transformed into cell lines to produce complete antibodies.
  • Antibodies are then compared using appropriate biochemical and biological assays to identify the best antibody.
  • the binding specificity of the antibodies or antigen-binding fragments disclosed in the present invention can be detected by in vitro experiments, such as co-immunoprecipitation, radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA).
  • in vitro experiments such as co-immunoprecipitation, radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA).
  • scFv can refer to the technology of producing single chain unit (US Patent 4,694,778; Bird, Science 242:423-442 (1988), Huston et al., Proc.Natl.Acad.Sci.USA 55:5879-5883 (1988) and Ward et al., Nature 334:544-554 (1989) and Nie et al., Antibody Therapeutics 3(1):18-62 (2020)).
  • Single-chain fusion peptides are generated by amino acid bridging of the heavy and light chain fragments of the Fv region to form single-chain units.
  • the technique of assembling functional Fv fragments in E. coli can also be used (Skerra et al., Science 242:1038-1041 (1988)).
  • scFv single-chain Fv
  • antibodies include, for example, U.S. Patents 4,946,778 and 5,258,498, and Huston et al., Methods in Enzymology 203:46-88 (1991), Shu et al., Proc. Natl. Sci. USA 90: 1995-1999 (1993) and Skerra et al., Science 240: 1038-1040 (1988).
  • chimeric, humanized or fully human antibodies may be used.
  • Chimeric antibodies are molecules in which different parts of the antibody are derived from different animal species, such as antibodies that have the variable regions of a murine monoclonal antibody and the constant regions of a human immunoglobulin.
  • Methods for producing chimeric antibodies are known in the art, see Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., J. Immunol. Methods 125:191 -202 (1989); Neuberger et al., Nature 372:604-608 (1984); Takeda et al., Nature 314:452-454 (1985); and U.S. Patents 5,807,715, 4,816,567 and 4,816,397, the entire contents of which are incorporated by reference Incorporated into this article.
  • Antibodies can be prepared by a variety of methods known in the art, including phage display methods using antibody libraries derived from immunoglobulin sequences. See also U.S. Patents 4,444,887 and 4,716,111, and PCT Publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741, each The entire content of the patent is incorporated herein by reference.
  • DNA encoding the desired monoclonal antibody can be isolated and sequenced using conventional methods (e.g., using oligonucleotide probes capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). sequencing. Isolated and subcloned hybridoma cells can serve as a source of such DNA. Once isolated, the DNA can be placed into an expression vector and then transfected into prokaryotic or eukaryotic host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma that do not produce other immunoglobulins in cells.
  • prokaryotic or eukaryotic host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma that do not produce other immunoglobulins in cells.
  • Isolated DNA (which may be synthetic as described herein) can also be used to prepare the constant and variable region sequences of antibodies as described in US Pat. No. 5,658,570, the entire contents of which are incorporated herein by reference. This method extracts RNA from selected cells and converts it into cDNA, which is then amplified by PCR using Ig-specific primers. Suitable probes for this purpose are also mentioned in US Patent No. 5,658,570.
  • one or more CDRs of an antibody of the invention can be inserted into a framework region, eg, into a human framework region, to construct a humanized non-fully human antibody.
  • the framework regions may be naturally occurring or consensus framework regions, preferably human framework regions (see Chothia et al., J. Mol. Biol. 278:457-479 (1998) for a list of human framework regions).
  • Some polynucleotides may encode an antibody that specifically binds at least one epitope of an antigen of interest produced by a combination of framework regions and CDRs.
  • One or more amino acid substitutions may be made within the framework regions, and the amino acid substitutions may be selected to improve binding of the antibody to its antigen.
  • substitution or deletion of cysteine residues in one or more variable regions involved in interchain disulfide bond formation can be performed in this way, thereby producing antibody molecules lacking one or more interchain disulfide bonds.
  • Other modifications to polynucleotides within the skill of the art are also encompassed in the present invention.
  • Antibodies can be prepared using conventional recombinant DNA techniques. Antibody-producing vectors, cell lines, and the like can be selected, constructed, and cultured using techniques known to those skilled in the art. These techniques are described in various laboratory manuals and major publications, such as Recombinant DNA Technology for Production of Protein Therapeutics in Cultured Mammalian Cells, D.L. hacker, F.M. Wurm, in Reference Module in Life Sciences, 2017, which in its entirety includes The supplementary content is incorporated by reference in its entirety.
  • the DNA encoding the antibody can be designed and synthesized according to the amino acid sequence of the antibody described herein in a conventional manner, placed into an expression vector, and then transfected into a host cell, and cultured in a medium to produce the transfected host cell.
  • Monoclonal antibodies Monoclonal antibodies.
  • an antibody expression vector includes at least one promoter element, an antibody coding sequence, a transcription termination signal, and a polyA tail. Other elements include enhancers, Kozak sequences, and donor and acceptor sites for RNA splicing flanking the inserted sequence.
  • High-efficiency transcription can be obtained through the early and late promoters of SV40, long terminal repeats from retroviruses such as RSV, HTLV1, HIVI, and early promoters of cytomegalovirus, and other cellular promoters such as muscle Kinetin promoter.
  • Suitable expression vectors may include pIRES1neo, pRetro-Off, pRetro-On, PLXSN, or Plncx, pcDNA3.1(+/-), pcDNA/Zeo(+/-), pcDNA3.1/Hygro(+/-), PSVL, PMSG, pRSVcat, pSV2dhfr, pBC12MI and pCS2 etc.
  • Commonly used mammalian cells include 293 cells, Cos1 cells, Cos7 cells, CV1 cells, mouse L cells and CHO cells, etc.
  • the inserted gene fragment needs to contain selection markers, common selection markers include dihydrofolate reductase, glutamine synthetase, neomycin resistance, hygromycin resistance and other selection genes, so as to facilitate transfection Screening of successful cell isolation.
  • selection markers include dihydrofolate reductase, glutamine synthetase, neomycin resistance, hygromycin resistance and other selection genes, so as to facilitate transfection Screening of successful cell isolation.
  • the constructed plasmid is transfected into host cells without the above-mentioned genes, and cultured in a selective medium, the successfully transfected cells grow in large numbers and produce the desired target protein.
  • mutations can be introduced into the nucleotide sequence encoding the antibodies of the present invention using standard techniques known to those skilled in the art, including but not limited to site-directed mutagenesis and PCR-mediated mutations resulting in amino acid substitutions.
  • Variants include derivatives
  • mutations can be introduced randomly along all or part of the coding sequence, for example by saturation mutagenesis, and the resulting mutants can be screened for biological activity to identify mutants that retain activity.
  • the invention also provides treatment methods and uses.
  • a method for treating or improving various types of cancer, tumor or infection-related diseases comprising administering an effective dose of anti-CD73 antibody or antigen-binding fragment to a patient in need.
  • the application of anti-CD73 antibody or antigen-binding fragment in treating or improving cancer, tumor or infection and other related diseases is provided.
  • the application of the anti-CD73 antibody or antigen-binding fragment in the preparation of a medicament for treating or improving cancer, tumor or infection and other related diseases is provided.
  • the specific dosage and treatment regimen for any particular patient will depend on various factors, including the specific antibody or derivative used, the patient's age and weight, general health, sex, and diet, as well as the time of administration, frequency of excretion, drug combination, and the severity of the particular disease being treated. These factors are in the judgment of the medical caregiver, who is within the purview of those of ordinary skill in the art.
  • the dosage will also depend on the individual patient to be treated, the route of administration, the type of formulation, the nature of the compound employed, the severity of the disease and the effect desired.
  • the dosage used can be determined by principles of pharmacology and pharmacokinetics well known in the art.
  • the antibody of the present invention is administered to a patient at a dose of 0.01 mg/kg to 100 mg/kg of the patient's body weight each time. In some embodiments, the administration is every 1 week, 2 weeks, 3 weeks, or monthly. In some embodiments, the antibody or antigen-binding fragment of the present invention is administered to a patient at a dose of 0.01 mg/kg to 100 mg/kg of the patient's body weight, or 0.1 mg/kg to 20 mg/kg of the patient's body weight.
  • the initial dose may be followed by a second dose or doses of the antibody or antigen-binding fragment at about the same or less than the initial dose, wherein the subsequent doses may be separated by at least 1 to 3 days; or at least one week.
  • the dose and frequency of administration of the antibodies or antigen-binding fragments of the invention can be reduced by enhancing the uptake and tissue penetration (eg, into the brain) of the antibodies or antigen-binding fragments through modifications such as lipidation.
  • an anti-CD73 antibody (such as antibody P59-L17 or antibody P59-L17') is administered at an effective dose of about 15 mg to 1200 mg per dose.
  • the effective amount of anti-CD73 antibody (such as antibody P59-L17 or antibody P59-L17') administered is about 15 mg to 1200 mg per treatment cycle.
  • a treatment cycle is about 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values ( inclusive) or any value in it.
  • the effective amount of the anti-CD73 antibody administered to the patient per treatment cycle is about 15 mg, about 18 mg, about 90 mg, about 120 mg, about 160 mg, about 180 mg, about 200 mg, about 230 mg, about 250 mg, about 280 mg, About 300mg, about 310mg, about 334mg, about 350mg, about 360mg, about 370mg, about 380mg, about 390mg, about 400mg, about 500mg, about 600mg, about 720mg, about 800mg, about 900mg, about 1000mg, about 1100mg, about 1200mg , or a range between any two of these values (inclusive), or any value therein, or a formulation containing such an amount of anti-CD73 antibody.
  • a treatment cycle is administered once from 1 week to 7 weeks.
  • the effective amount of anti-CD73 antibody administered in each treatment cycle is about 100 mg to 300 mg, or a preparation containing this dose of anti-CD73 antibody; wherein, one treatment cycle is about 1 week, about 2 weeks, about 3 weeks weeks, about 4 weeks, or a range between any two of these values (inclusive), or any value therein.
  • the patient is administered an effective amount of anti-CD73 antibody of about 300 mg to 600 mg in each treatment cycle, or a preparation containing such a dose of anti-CD73 antibody; wherein, a treatment cycle is about 1 week, about 2 weeks, about 3 weeks, or about 4 weeks.
  • the effective amount of anti-CD73 antibody administered in each treatment cycle is about 700 mg to 1100 mg, or a preparation containing this dose of anti-CD73 antibody; wherein, one treatment cycle is about 1 week, about 2 weeks, about 3 weeks weeks, about 4 weeks, or a range between any two of these values (inclusive), or any value therein.
  • the effective amount of the anti-CD73 antibody administered to the patient per treatment cycle is about 60 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, About 190 mg, about 200 mg, about 360 mg, about 420 mg, about 720 mg, about 1080 mg, or a range between any two of these values (including endpoints) or any value therein, or a preparation containing such a dose of anti-CD73 antibody;
  • one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-CD73 antibody of about 50 mg to 80 mg in each treatment cycle, or a preparation containing this dose of anti-CD73 antibody; wherein, a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-CD73 antibody of about 60 mg per treatment cycle, or a preparation containing such a dose of anti-CD73 antibody; for example, about 60 mg is administered once.
  • the patient is administered an effective amount of anti-CD73 antibody of about 150 mg to 200 mg in each treatment cycle, or a preparation containing such a dose of anti-CD73 antibody; wherein, a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the effective amount of anti-CD73 antibody administered to the patient is about 180 mg per treatment cycle, or a preparation containing such a dose of anti-CD73 antibody; for example, about 180 mg is administered once.
  • the patient is administered an effective amount of anti-CD73 antibody of about 345 mg to 380 mg in each treatment cycle, or a preparation containing this dose of anti-CD73 antibody; wherein, a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-CD73 antibody of about 360 mg per treatment cycle, or a preparation containing such a dose of anti-CD73 antibody; for example, about 360 mg is administered once.
  • the patient is administered an effective amount of anti-CD73 antibody of about 693 mg to 730 mg in each treatment cycle, or a preparation containing this dose of anti-CD73 antibody; wherein, a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • a treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-CD73 antibody of about 720 mg per treatment cycle, or a preparation containing such a dose of anti-CD73 antibody; for example, about 720 mg is administered once.
  • the anti-CD73 antibody (such as antibody P59-L17 or antibody P59-L17') is about 0.3 mg/kg, about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4mg/kg, about 3mg/kg, about 3.6mg/kg, about 4mg/kg, about 4.8mg/kg, about 5mg/kg, about 5.5mg/kg, about 6mg/kg, about 6.9mg/kg, about 7 mg/kg, about 8.4 mg/kg, about 9 mg/kg, about 11 mg/kg, about 12 mg/kg, about 15 mg/kg, about 18 mg/kg, or the range between any two of these values (inclusive) ) or any value therein, or a preparation containing this dose of anti-CD73 antibody.
  • the effective amount of anti-CD73 antibody (such as antibody P59-L17 or antibody P59-L17') administered is about 0.3 mg/kg to 18 mg/kg once every 2 weeks or every 3 weeks. In some embodiments, the effective amount of anti-CD73 antibody (such as antibody P59-L17) administered is about 0.3 mg/kg, about 1 mg/kg, about 3 mg/kg, about 5 mg/kg, about 6 mg/kg, about 12 mg/kg kg, about 13mg/kg, or about 18mg/kg every 2 weeks or every 3 weeks.
  • the effective amount of anti-CD73 antibody (such as antibody P59-L17 or antibody P59-L17') administered is about 5 mg/kg every 2 weeks, about 6 mg/kg every 2 weeks, about 10 mg/kg kg once every 2 weeks, about 3mg/kg once every 3 weeks, about 5mg/kg once every 3 weeks, about 6mg/kg once every 3 weeks, about 7.5mg/kg once every 3 weeks, about 12mg /kg every 3 weeks, about 15mg/kg every 3 weeks, or about 18mg/kg every 3 weeks.
  • the patient is administered an anti-CD73 antibody once per treatment cycle.
  • the anti-CD73 antibody is administered multiple times, eg, 2, 3, 4, or 5 times, within each treatment cycle.
  • the patient can only be dosed 1 or 4 times per treatment cycle.
  • the patient is treated for one treatment cycle. In some embodiments, the patient is treated for multiple (eg, 2, 3, or 4) treatment cycles. In some embodiments, the patient is treated until the condition is in remission and no longer requires treatment.
  • compositions may be administered by any convenient route, such as by infusion or bolus injection, absorbed through the epithelium or mucous membranes (eg, oral mucosa, rectal and intestinal mucosa, etc.), and may be co-administered with other biologically active agents.
  • the pharmaceutical composition containing the antibody, antigen-binding fragment or derivative thereof of the present invention can be administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, externally (such as By powder, ointment, drops or transdermal patch), orally or by oral or nasal spray.
  • parenteral refers to modes of administration including intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • the mode of administration can be systemic administration or local administration.
  • compositions of the invention comprise a nucleic acid or polynucleotide encoding an antibody or antigen-binding fragment, which can be administered in vivo to facilitate its encoded expression by constructing it as part of a suitable nucleic acid expression vector.
  • a suitable nucleic acid expression vector Expression of the protein, followed by administration of the above-mentioned part of the vector to make it an intracellular part, for example by using a retroviral vector (see US Pat. No.
  • nucleic acid can be introduced into the cell by homologous recombination and integrated into the host cell DNA for expression.
  • Various known delivery systems can be used to administer the antibodies, antigen-binding fragments or derivatives thereof of the invention, or polynucleotides encoding them, such as encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compounds, Receptor-mediated endocytosis (see, eg, Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of nucleic acids as part of retroviruses or other vectors, etc.
  • an anti-CD73 antibody or antigen-binding fragment of the invention may be combined with other therapeutic or prophylactic regimens, comprising administering one or more antibodies or antigen-binding fragments of the invention together with one or more other therapeutic agents or methods use or in combination.
  • other treatment options include, but are not limited to, radiation therapy, chemotherapy, hormone therapy, and surgery, among others.
  • the antibodies can be administered simultaneously or separately from the other therapeutic agents.
  • the antibody or antigen-binding fragment of the invention can be administered before or after another other therapeutic agent is administered.
  • the antibody or antigen-binding fragment of the present invention can be used in combination with chemotherapeutic agents for the treatment of cancer or tumors, including but not limited to: camptothecin (camptothecin, CPT-11), 5-fluorouracil (5 -FU), cisplatin, doxorubicin, irinotecan, paclitaxel, gemcitabine, cisplatin, carboplatin, proteasome inhibitors (such as bortezomib (bortezomib) or MG132), Bcl-2 inhibitors (eg BH3I-2' (bcl-xl inhibitor), indoleamine dioxygenase-1 (IDO1) inhibitors (eg INCB24360), AT-101 (R -(-)-gossypol derivative), ABT-263 (small molecule), GX-15-070 (obatoclax), MCL-1 (myeloid leukemia cell differentiation protein-1 antagonist), iAP antagonists (e
  • the antibodies or antigen-binding fragments of the present invention can be used in combination with cytotoxic agents for the treatment of cancer or tumors, including but not limited to: uracil mustard, methine, cyclophosphamide (CYTOXANTM ), Ifosfamide, Melphalan (Melphalan), Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphamide, Busulfan (Busulfan), Carmustine ( Carmustine, Lomustine, Streptozocin, dacarbazine, and Temozolomide.
  • cytotoxic agents including but not limited to: uracil mustard, methine, cyclophosphamide (CYTOXANTM ), Ifosfamide, Melphalan (Melphalan), Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphamide, Busulfan (Busulfan), Carmustine ( Carmustine, Lomustine, Str
  • the antibodies or antigen-binding fragments of the present invention may be used in combination with antimetabolites for the treatment of cancer or tumors, including but not limited to: methotrexate, 5-fluorouracil, floxuridine, arabinosin Cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine and gemcitabine.
  • the antibodies or antigen-binding fragments of the present invention can be used in combination with anti-proliferative agents for the treatment of cancer or tumors, including but not limited to: docetaxel, docetaxel, DDM, Dick Statin (dictyostatin, DCT), Peloruside (Peloruside) A, epothilone, epothilone A, epothilone B, epothilone C, epothilone D, epothilone E , Epothilone F, Epothilone furan D, deoxyepothilone B1, spongolactone, patupilone (patupilone, EPO-906), ILX-651 (tasidotin hydrochloride (tasidotin hydrochloride) )), Halichondrin B, Eribulin mesylate (E-7389), Hemiasterlin (HTI-286), Cyrptophycin,
  • antibodies or antigen-binding fragments of the invention may be used in combination with agonists of costimulatory receptors and/or antagonists of inhibitory signaling on T cells for the treatment of cancer or tumors.
  • Targets for agonists or antagonists include, but are not limited to, CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, anti-galectin-9 antibodies, VEGF, BTLA, CD69, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, TIM-4, CD39, B7-1, B7-2, CD28, 4-1BB, 4-1BBL, GITR, GITRL, OX40, OX40L, CD70, CD27, CD40, DR3 and CD28H, etc.
  • the antibody that can be administered with an antibody or antigen-binding fragment of the invention is rituximab, trastuzumab, tositumomab, ibritumomab, Alemtuzumab, epratuzumab, bevacizumab, ipilimumab, galiximab, lucatumumab, Murromonab.
  • an antibody or antigen-binding fragment of the invention may be administered with an anti-PD-1 antibody, such as Nivolumab Pembrolizumab Toripalimab Sintilimab Camrelizumab Tislelizumab Penpulimab Zimberelimab Serplimab Or the anti-PD-1 antibody disclosed in WO2020207432, etc.
  • an anti-PD-1 antibody such as Nivolumab Pembrolizumab Toripalimab Sintilimab Camrelizumab Tislelizumab Penpulimab Zimberelimab Serplimab or the anti-PD-1 antibody disclosed in WO2020207432, etc.
  • the present invention also provides pharmaceutical compositions.
  • Such a composition comprises an effective dose of anti-CD73 antibody or antigen-binding fragment, and pharmaceutically acceptable auxiliary materials.
  • the pharmaceutical composition comprises 0.1%-90% anti-CD73 antibody or antigen-binding fragment.
  • the pharmaceutical composition further comprises an anti-cancer agent (eg, an immune checkpoint inhibitor).
  • the term "pharmaceutically acceptable” refers to a substance approved by a governmental regulatory agency or listed in a recognized pharmacopoeia for use in animals, especially in humans.
  • pharmaceutically acceptable excipients generally refer to any type of non-toxic solid, semi-solid or liquid fillers, diluents, encapsulating materials or formulation aids, etc.
  • adjuvant refers to a diluent, adjuvant, excipient or carrier with which the active ingredient can be administered to a patient.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, skim milk powder, glycerol, Propylene, ethylene glycol, water, ethanol, etc.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents such as acetates, citrates or phosphates.
  • Antibacterial agents such as benzyl alcohol or methylparaben, antioxidants such as ascorbic acid or sodium bisulfite, chelating agents such as ethylenediaminetetraacetic acid, and tonicity adjusting agents such as sodium chloride or dextrose are also contemplated.
  • These compositions may take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like.
  • compositions will contain a clinically effective dose of the antibody or antigen-binding fragment, preferably in a purified form, together with an appropriate amount of excipients to provide a dosage form suitable for the patient.
  • the formulation should be suitable for the mode of administration.
  • the parent formulation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • the composition is formulated into a pharmaceutical composition suitable for intravenous injection to human body according to conventional procedures.
  • Compositions for intravenous administration are generally solutions in sterile isotonic aqueous buffer.
  • the composition may also contain a solubilizer and a local anesthetic such as lidocaine to relieve pain at the injection site.
  • the active ingredients are presented alone or in combination in unit dosage form, eg, as a dry lyophilized powder or water-free concentrate, in a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
  • the composition may be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water or saline for injection can be used so that the active ingredient can be mixed before administration.
  • the compounds of the present invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include salts derived from anions such as hydrochloric acid, phosphoric acid, acetic acid, oxalic acid, tartaric acid, etc., and salts derived from such as sodium, potassium, ammonium, calcium, ferric hydroxide, isopropylamine, triethylamine, 2- Salts of ethylaminoethanol, histidine, procaine, etc. with cations.
  • the fully human scFv phage display library was used to screen the antigen hCD73-biotin (ACROBiosystems, Cat. No. CD3-H82E3).
  • the screening method is: combine SA-magnetic beads (DynabeadsTM MyOneTM Streptavidin T1, catalog number 65602, Thermo fisher Scientific) with hCD73-biotin, room temperature for 2 hours or overnight at 4°C; add the blocked phage display library to PBS (Phosphate buffer saline) Wash the above-mentioned magnetic beads, perform screening, room temperature for 2 hours or overnight at 4°C; PBST (PBS buffer containing 0.05% Tween-20) was washed 15 times to wash off non-specifically bound phages; Use trypsin to digest at room temperature for 30 minutes, add 1/10 volume of FBS (fetal bovine serum) to terminate the digestion reaction, re-infect the digested phages into TG1 strains for
  • the construction method of the fully human scFv phage display library is as follows: amplify the VH gene and VL gene in unimmunized human peripheral blood lymphocytes (peripheral blood lymphocytes, PBLs) by polymerase chain reaction (polymerase chain reaction, PCR), and then Randomly combine heavy chain VH and light chain VL by overlapping polymerase chain reaction (overlapping polymerase chain reaction, overlap PCR) to form a single chain Fv (single chain Fv, scFv); scFv is constructed by inserting restriction sites into the phagemid vector A phage library (stock capacity greater than 10 11 ) was formed and displayed on the surface of the phage for antibody screening (for details, see SHEETS et al. (1998) Cell Biology.95:6157-6162).
  • IgG1 type monoclonal antibody design primers for VH and VL in the screened scFv
  • the DNA fragments were subjected to PCR, and the PCR products were digested with double enzymes (the restriction site endonucleases used by VH were ApaI/MfeI, and the restriction site endonucleases used by VL were HindIII/BsiWI), and the fragments were respectively linked to IgG1-containing complexes.
  • the nucleic acid sequences of the heavy chain and the light chain were sequence optimized (the optimized nucleic acid sequences are shown in the table 6) Link the optimized heavy chain and light chain nucleic acid sequences to the expression vector respectively, and after sequencing to confirm the correctness, obtain the light chain plasmid and antibody heavy chain plasmid for protein expression of the antibody.
  • the corresponding light chain plasmid and heavy chain plasmid were co-transfected into HEK293 cells (purchased from ATCC) through PEI (polyetherimide) for transient expression, and the supernatant was collected after 7 days of culture, and then Protein A column (GE Healthcare) was used to Purified by Immobilized Metal Affinity Chromatography (IMAC), the purity of the purified antibody protein was >95%.
  • PEI polyetherimide
  • IgG1-Fc that is, the constant region of IgG1, including CH1, hinge region, CH2 and CH3
  • pcDNA3.1 vector containing IgG1-Fc purchased from Invitrogen Company, V79020
  • HEK293 cells co-transfecting HEK293 cells with PEI
  • the supernatant was collected for purification after 7 days of culture.
  • the irrelevant antibody NR is an antibody that specifically binds to the new coronavirus. Its preparation method is: the corresponding light chain plasmid and heavy chain plasmid are co-transfected into HEK293 cells through PEI for transient expression, and the supernatant is collected after 7 days of culture for purification.
  • the heavy chain amino acid sequence of antibody NR (SEQ ID NO:30):
  • the anti-CD73 antibodies prepared in this example include antibodies P59-L1, P59-L6, P59-L12, and P59-L17.
  • the amino acid sequences and nucleic acid sequences related to the antibody examples are shown in Table 2-6; the Fc region of the heavy chain in Table 5 is used single underlined.
  • the purified antibody was sequenced, and the sequencing result was the same as the designed sequence.
  • the purified antibody is used for affinity detection and biological activity identification, etc.
  • Biacore T200 surface plasmon resonance instrument was used to measure the affinity constant (K D ) of the antibody.
  • the main test process was as follows (refer to the standard operating procedure of Biacore T200): -H08H) Samples were serially diluted with HBS-EP buffer (150 mM NaCl, 3 mM EDTA, 0.005% (v/v) surfactant P-20, and 10 mM HEPES, pH 7.4) (the initial concentration was 32 nM, then 2-fold dilution ); Dilute the antibody with sodium acetate solution (10mM, pH5.5) to a final concentration of about 20 ⁇ g/ml; use BiacoreT200, Protein A chip (GE healthcare, product number 29127556) for detection, the instrument settings are as follows: antibody capture binding The contact time is 180s, the flow rate is 30 ⁇ l/min; the sample contact time is 120s, the dissociation time is 300s, and the flow rate is 30 ⁇ l/min; the regeneration condition is: Glycine-HCl (
  • the data analysis software Evaluation Software3.1 was used to analyze the test results, and the sensor signals collected by the sample experimental flow path were subtracted from the reference flow path and the sample blank, and the kinetic "1:1" model was selected for fitting.
  • Kinetic parameters (Ka is the association rate, Kd is the dissociation rate; KD is the binding-dissociation equilibrium constant) are obtained.
  • the antibodies (P59-L1, P59-L6, P59-L12, P59-L17) had good binding ability to hCD73-His; especially the antibody P59-L17.
  • ELISA method was used for detection: first use 2 ⁇ g/ml human source hCD73-His (Beijing Sino Biological Science and Technology Co., Ltd., Catalog is 10904-H08H), cynomolgus monkey cCD73-His (near shore organisms, Catalog is CD3- C52H9) or mouse mCD73-His (near shore organisms, Catalog is CD3-M52H9) coated 96-well ELISA plate, overnight at 4°C; blocked with PBS buffer containing 5% BSA (bovine serum albumin) for 2 hours; blocked After the end, wash with PBS containing 0.05% TW-20, and then add antibody solution diluted with PBS containing 0.05% TW-20 and 0.5% BSA (the initial concentration of antibody is 4 ⁇ g/ml, 3-fold dilution, 10 gradients ); incubate at 37°C for 1 hour; after washing the plate, add HRP (horseradish peroxidase)-labele
  • antibody P59-L17 binds well to human and cynomolgus CD73, but does not bind to mouse CD73.
  • NA means no binding or substantially no binding.
  • MDA-MB-231 cells are human breast cancer cells with high expression of CD73 protein on the cell membrane surface (https://www.proteinatlas.org/ENSG00000101017-CD40/cell). In this example, MDA-MB-231 cells were used to detect the binding ability of anti-CD73 antibodies to CD73 molecules on the cell membrane surface.
  • the test method is: after the well-cultured MDA-MB-231 cells are collected and centrifuged, use isotype control IgG1-Fc (concentration is 10ug/ml, single concentration point) or anti-CD73 antibody (initial concentration is 10ug/ml, 2-fold dilution, 8 concentration gradients) for incubation; after 30 min, centrifuge to wash off the supernatant, wash once with PBS buffer, resuspend in PBS buffer and add fluorescent-labeled anti-human IgG flow antibody anti-humanFc-PE (purchased from Invitrogen, Cat. No. 12-4998-82); after incubation for 30 min, centrifuge to wash off the supernatant, wash twice with PBS buffer, resuspend the cells in PBS buffer, perform flow cytometry, count MFI and process data with SoftMax Pro.
  • IgG1-Fc concentration is 10ug/ml, single concentration point
  • anti-CD73 antibody initial concentration is 10ug
  • antibody P59-L17 bound to MDA-MB-231 cells, while negative control IgG1-Fc did not bind to MDA-MB-231 cells.
  • the detection principle of the detection kit (purchased from promega, the article number is G7570) is: excess AMP can inhibit ATP-dependent
  • the detection reagent emits fluorescence, and the cell membrane CD73 can decompose AMP to release the inhibitory effect of AMP.
  • the anti-CD73 antibody can inhibit the enzyme activity of CD73 so as to maintain the inhibition of AMP on the fluorescence process.
  • the final performance is: with the increase of antibody concentration, the fluorescence intensity corresponding weakened.
  • the inhibitory effect of anti-CD73 antibody on the enzymatic activity of CD73 on the cell membrane was detected.
  • the test method is: use DMEM medium containing 10% FBS to spread the well-growing MDA-MB-231 cells on a 96-well cell culture plate at 2.5 ⁇ 10 4 cells/well, and culture in a 37°C cell culture box overnight; Remove the supernatant of the culture medium the next day, add the antibody solution diluted in serum-free DMEM (initial concentration is 5ug/ml, 2-fold dilution, 8 concentration gradients) and AMP with a final concentration of 600 ⁇ M, and place in a 37°C cell culture incubator React for 3 hours; after the reaction, take 50 ⁇ l of the supernatant per well in a 96-well white plate, add 50 ⁇ l of ATP with a concentration of 200 ⁇ M, and finally add 100 ⁇ l of detection reagent, and immediately read the fluorescence value on a microplate reader.
  • T cells and B cells express CD73, and CD73 is involved in lymphocyte activity.
  • flow cytometry was performed to explore the expression of CD73 in peripheral blood T cells and B cells.
  • the test method is as follows: extract peripheral blood from healthy adults, and use lymphatic separation fluid (purchased from Beijing Dakwei Biotechnology Co., Ltd., article number is 7912011) to separate fresh PBMC (peripheral blood mononuclear cells); on ice, first use 1 mg/ml irrelevant antibody IgG1 (antibody NR) blocked the Fc receptor of PMBC for 30 min; centrifuged to wash off the blocking solution, added 10 ⁇ g/ml biotin-labeled anti-CD73 antibody or biotin-labeled IgG1-Fc, incubated on ice for 30 min; centrifuged Wash off the supernatant, add fluorescently labeled streptavidin SA-PE (purchased from Invitrogen, product number is 21627) and anti-CD3-APC (purchased from Elabscience, product number is FW2671) or anti-CD19-APC (purchased from Elabscience (Cat. No. FW1075) flow cytometric secondary antibody, incubated
  • the positive rate of T cells expressing CD73 was 47%; as shown in Figure 3B, the positive rate of B cells expressing CD73 was 80%.
  • CD73 on the cell membrane of CD73-positive T cells can decompose extracellular AMP into adenosine, and adenosine can mediate immunosuppression by binding to adenosine receptors on T cells, inhibiting T cell proliferation and cytokine secretion .
  • the function of anti-CD73 antibody to relieve the inhibition of T cell proliferation by AMP was detected.
  • the test method is as follows: extract peripheral blood from healthy adults, use lymphatic separation fluid to separate fresh PBMC (peripheral blood mononuclear cells), add CD3/CD28 to RPMI-1640 medium (purchased from Gibco) containing 10% FBS Antibody-coupled magnetic beads (Beijing Tongli Haiyuan Biotechnology Co., Ltd., catalog TL-601) stimulated culture of PBMC; the next day, 100IU/ML IL2 was added for expansion culture, and CD3 + with high purity was obtained after expansion culture for 6 days
  • T cells use a magnetic stand to remove the magnetic beads in the medium and centrifuge to wash the cells twice; spread T cells in 96-well cell culture plates at 2.5 ⁇ 10 4 cells/well, add gradiently diluted anti-CD73 antibody solution or negative Control IgG1-Fc (starting with an initial concentration of 1ug/ml, 3-fold dilution, and 7 concentration gradients), and at the same time, add AMP with a final concentration of 300 ⁇ M to each well, and place
  • the antibody P59-L17 relieved the inhibition of T cell proliferation by AMP in a concentration-gradient dependent manner.
  • anti-CD73 antibodies In addition to blocking adenosine receptor-mediated immunosuppression by inhibiting CD73 enzymatic activity, anti-CD73 antibodies have the function of upregulating the expression of B cell activation markers. In this example, the ability of the anti-CD73 antibody to up-regulate the expression of B cell activation marker CD69 was detected.
  • the test method is as follows: extract peripheral blood from healthy adults, use lymphatic separation fluid to separate fresh PBMC, culture PBMC with RPMI-1640 medium containing 10% FBS, add anti-CD73 antibody or IgG1-Fc, and culture overnight; the next day
  • the cells were centrifuged to remove the supernatant, CD19-APC (purchased from Elabscience, product number: FW1075) and CD69-PE (purchased from Elabscience, product number: E-AB-F1138D) flow cytometric secondary antibodies were added, incubated on ice for 30 min, and then detected.
  • the expression of CD69 on CD19 + B cells was analyzed.
  • antibody P59-17 significantly increased the expression of CD69 on B cell activation.

Landscapes

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

Abstract

La présente invention concerne un anticorps anti-CD73 et son utilisation. L'anticorps de la présente invention peut se lier spécifiquement à CD73 pour bloquer l'interaction entre CD73 et un ligand de celui-ci. L'anticorps de la présente invention aide les cellules tumorales claires du système immunitaire et est également utilisé pour le diagnostic et le pronostic de tumeurs ou de cancers.
PCT/CN2022/094182 2021-05-21 2022-05-20 Anticorps anti-cd73 et son utilisation WO2022242758A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2021/095287 2021-05-21
CN2021095287 2021-05-21

Publications (1)

Publication Number Publication Date
WO2022242758A1 true WO2022242758A1 (fr) 2022-11-24

Family

ID=84060195

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/094182 WO2022242758A1 (fr) 2021-05-21 2022-05-20 Anticorps anti-cd73 et son utilisation

Country Status (2)

Country Link
CN (1) CN115368458A (fr)
WO (1) WO2022242758A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024040195A1 (fr) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditionnement pour l'ingénierie de cellules immunitaires in vivo

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021032173A1 (fr) * 2019-08-21 2021-02-25 Harbour Biomed (Shanghai) Co., Ltd. Anticorps anti-cd73 et son application

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021032173A1 (fr) * 2019-08-21 2021-02-25 Harbour Biomed (Shanghai) Co., Ltd. Anticorps anti-cd73 et son application

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024040195A1 (fr) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditionnement pour l'ingénierie de cellules immunitaires in vivo
WO2024040194A1 (fr) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditionnement pour l'ingénierie de cellules immunitaires in vivo

Also Published As

Publication number Publication date
CN115368458A (zh) 2022-11-22

Similar Documents

Publication Publication Date Title
US12037408B2 (en) Method of treating a tumor using anti-TfR antibodies
TWI564306B (zh) 雙特異性抗體
CN111138547A (zh) 针对cd19和cd3的同源二聚体型双特异性抗体及其制备方法和用途
JP2020504171A (ja) 抗PD−1抗体との組み合わせのための抗Tim−3抗体
JP2019505564A (ja) 抗bcmaポリペプチド及びタンパク質
EP3131917B1 (fr) Anticorps monoclonaux humanisés et chimériques contre cd99
WO2022143794A1 (fr) Anticorps anti-cldn18.2, son procédé de préparation et son utilisation
WO2021088927A1 (fr) Conjugués anticorps-médicament ciblant claudine 18,2
WO2019242619A1 (fr) Anticorps anti-lag-3 complètement humanisé et son application
WO2024012539A1 (fr) Anticorps anti-nectine-4 et son utilisation
CN116323676A (zh) 抗Claudin18.2和CD3的双特异性抗体以及其用途
WO2022242758A1 (fr) Anticorps anti-cd73 et son utilisation
TW201800418A (zh) 結合stat3之抗體治療劑
CN112794911B (zh) 人源化抗叶酸受体1抗体及其应用
EP4142793A1 (fr) Anticorps spécifiques à abcb5 et leurs utilisations
WO2022095970A1 (fr) Anticorps bispécifique et son utilisation
WO2023143547A1 (fr) Anticorps anti-cd28 et son utilisation
WO2022206677A1 (fr) Anticorps anti-vista et son application
WO2022028608A1 (fr) Anticorps anti pd-l1 et son utilisation
US20240270839A1 (en) Novel anti-claudin18 antibodies
TW202330627A (zh) 特異性結合cd47和her2的雙特異性抗體及其用途
CN114656567A (zh) 抗icos抗体及其应用
WO2022247933A1 (fr) ANTICORPS ANTI-SIRPα ET SON UTILISATION
WO2023193794A1 (fr) Application d'anticorps anti-vista dans un médicament combiné
WO2024153163A1 (fr) Anticorps anti-cd93 et son utilisation

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: 22804077

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22804077

Country of ref document: EP

Kind code of ref document: A1