WO2023151312A1 - Anticorps neutralisant à large spectre de betacoronavirus et son utilisation - Google Patents

Anticorps neutralisant à large spectre de betacoronavirus et son utilisation Download PDF

Info

Publication number
WO2023151312A1
WO2023151312A1 PCT/CN2022/126628 CN2022126628W WO2023151312A1 WO 2023151312 A1 WO2023151312 A1 WO 2023151312A1 CN 2022126628 W CN2022126628 W CN 2022126628W WO 2023151312 A1 WO2023151312 A1 WO 2023151312A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
seq
acid sequence
sequence shown
deletions
Prior art date
Application number
PCT/CN2022/126628
Other languages
English (en)
Chinese (zh)
Inventor
谢晓亮
曹云龙
Original Assignee
北京昌平实验室
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 北京昌平实验室 filed Critical 北京昌平实验室
Priority to CN202280091594.4A priority Critical patent/CN118679181A/zh
Publication of WO2023151312A1 publication Critical patent/WO2023151312A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • 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
    • A61K39/42Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum viral
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens
    • 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
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • 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
    • C07K2317/52Constant 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/165Coronaviridae, e.g. avian infectious bronchitis virus

Definitions

  • the invention relates to the field of immunology, in particular to the B-coronavirus broad-spectrum neutralizing antibody and its application.
  • the present invention relates to B-coronavirus broad-spectrum neutralizing antibodies or antigen-binding fragments thereof, and the application of the antibodies or fragments in the diagnosis, prevention and treatment of various diseases caused by B-coronavirus infection.
  • Coronaviruses are the largest RNA viruses identified so far, which can infect mammalian humans, mice, pigs, cats, dogs, bats, cattle, and avian vertebrates, causing respiratory, intestinal, liver, and nervous system damage in hosts. disease. Coronaviridae is divided into 4 genera, namely ⁇ , ⁇ , ⁇ and ⁇ coronaviruses, among which ⁇ is the most harmful to humans. Betacoronavirus is divided into ⁇ coronavirus genus and five subgenus, namely Embecovirus, Sarbecovirus, Merbecovirus, Nobecovirus and Hibecovirus.
  • beta coronavirus The subgenus of beta coronavirus (Sarbecovirus) includes the new coronavirus (SARS-CoV-2) and its variants, severe acute respiratory syndrome coronavirus (SARS-CoV) and its variants, SARS-related Coronavirus (SARSr-CoV), subdivided as shown in Figure 1 (DOI: 10.1038/s41586-020-2294-9).
  • Betacoronavirus as a single-stranded RNA virus, translates and replicates directly in cells, and is more prone to mutation than DNA viruses. Since the outbreak of SARS-CoV-2 at the end of 2019, a variety of mutant strains have been produced, among which the World Health Organization has listed as Variant of concern (VOC), which has reached 5 types: Alpha (Alpha, B. 1.1.7), Beta (Beta, B.1.351), Gamma (Gamma, P.1), Delta (Delta, B.1.617.2) and Omicron (B.1.1.529).
  • VOC Variant of concern
  • the present invention applies techniques in many fields such as immunology, molecular biology, virology, protein structure analysis, etc., develops a set of unique technical routes, and obtains a series of B-coronavirus broad-spectrum neutralizing antibodies.
  • These antibodies come from the memory B cells of SARS-CoV-infected recovered patients who have been vaccinated with SARS-CoV-2 vaccines. They have the characteristics of broad-spectrum high-binding activity and broad-spectrum high neutralization of B-coronaviruses, and can efficiently neutralize novel coronaviruses, including novel coronaviruses.
  • Viruses, atypical coronaviruses and other pathogenic viruses are especially suitable for the diagnosis, prevention and treatment of various diseases caused by B-coronavirus infection, and have important clinical value.
  • the present invention successfully predicts the binding epitope and escape map of these neutralizing antibodies on B-coronavirus, and finds antibody pairs based on this information, which is verified by cryo-electron microscopy. Thus the sequences of these antibodies were reported for the first time.
  • the invention provides an antibody or antigen-binding fragment thereof comprising a heavy chain variable region and a light chain variable region comprising a complementarity determining sequence of specific amino acids. district.
  • the heavy and light chain variable regions comprise specific amino acid sequences.
  • the antibody or antigen-binding fragment thereof comprises a constant region derived from a human immunoglobulin, eg, a heavy chain constant region and a light chain constant region comprising specific amino acid sequences.
  • the antigen-binding fragment is selected from Fab, Fab', (Fab') 2 , Fv, disulfide-linked Fv, scFv, diabody, single domain antibody (sdAb), chimeric Antibodies, bispecific antibodies or multispecific antibodies.
  • the invention also provides an isolated nucleic acid molecule encoding an antibody of the invention or an antigen-binding fragment thereof, or a heavy chain variable region and/or a light chain variable region thereof.
  • the nucleic acid molecule is operably linked to an expression control sequence.
  • the present invention also provides an expression vector comprising the nucleic acid molecule of the present invention.
  • the present invention also provides a host cell transformed with the nucleic acid molecule of the present invention or the expression vector of the present invention.
  • the present invention also provides a method for preparing an antibody or an antigen-binding fragment thereof, comprising:
  • the present invention also provides a pharmaceutical composition, which comprises the antibody or antigen-binding fragment thereof of the present invention, and a pharmaceutically acceptable carrier and/or excipient.
  • the present invention also provides a method for preventing and/or treating diseases caused by B-coronavirus infection, the method comprising administering to a subject an effective amount of the antibody of the present invention or its antigen-binding fragment or the present invention.
  • the pharmaceutical composition of the invention is preferably a mammal, more preferably a human.
  • the present invention also provides the use of the antibody or antigen-binding fragment thereof of the present invention for preparing a medicament for preventing and/or treating diseases caused by B-coronavirus infection.
  • the beta coronavirus includes novel coronavirus (SARS-CoV-2) and variants thereof, severe acute respiratory syndrome coronavirus (SARS-CoV) and variants thereof, SARS-related coronaviruses (SARSr-CoV).
  • SARS-CoV-2 novel coronavirus
  • SARS-CoV severe acute respiratory syndrome coronavirus
  • SARSr-CoV SARS-related coronaviruses
  • the mutant strain of SARS-CoV-2 is selected from Alpha (Alpha, B.1.1.7), Beta (Beta, B.1.351), Gamma (Gamma, P.1), Delta ( Delta, B.1.617.2), Omicron (B.1.1.529) or any combination thereof.
  • the present invention also provides a conjugate comprising an antibody or antigen-binding fragment thereof of the present invention, and a detectable label linked to said antibody or antigen-binding fragment thereof.
  • the detectable label is selected from enzymes (such as horseradish peroxidase or alkaline phosphatase), chemiluminescent reagents (such as acridinium esters, luminol and derivatives thereof, or ruthenium derivatives), fluorescent dyes (such as fluorescein or fluorescent protein), radionuclides or biotin.
  • enzymes such as horseradish peroxidase or alkaline phosphatase
  • chemiluminescent reagents such as acridinium esters, luminol and derivatives thereof, or ruthenium derivatives
  • fluorescent dyes such as fluorescein or fluorescent protein
  • the present invention also provides a kit comprising an antibody or antigen-binding fragment thereof of the present invention or a conjugate of the present invention.
  • the kit comprises a conjugate of the invention.
  • the kit comprises an antibody or antigen-binding fragment thereof of the invention, and a second antibody that specifically recognizes the antibody or antigen-binding fragment thereof.
  • the second antibody further includes a detectable label, such as an enzyme (such as horseradish peroxidase or alkaline phosphatase), a chemiluminescent reagent (such as acridinium esters, luminol, and derivatives thereof, or ruthenium derivatives), fluorescent dyes (such as fluorescein or fluorescent protein), radionuclides or biotin.
  • a detectable label such as an enzyme (such as horseradish peroxidase or alkaline phosphatase), a chemiluminescent reagent (such as acridinium esters, luminol, and derivatives thereof, or ruthenium derivatives), fluorescent dyes (such as fluorescein or fluorescent protein), radionuclides or biotin.
  • an enzyme such as horseradish peroxidase or alkaline phosphatase
  • a chemiluminescent reagent such as acridinium esters, luminol, and derivatives thereof, or
  • the present invention also provides a method for detecting the presence or level of B-coronavirus in a sample, comprising:
  • detection of the binding represents the presence of B-coronavirus in the sample.
  • the detection of the strength of the binding represents the level of the B-coronavirus in the sample.
  • the sample is a blood sample (eg, whole blood, plasma, or serum), feces, oral or nasal secretions, or alveolar lavage fluid from the subject.
  • a blood sample eg, whole blood, plasma, or serum
  • feces e.g., feces, oral or nasal secretions, or alveolar lavage fluid from the subject.
  • the subject is a mammal, such as a human.
  • the sample is not a sample from a subject, eg, the sample is from a vaccine sample.
  • the present invention also provides the use of the antibody of the present invention or its antigen-binding fragment or the conjugate of the present invention in the preparation of a kit for detecting the presence or level of B-coronavirus in a sample .
  • Antibodies or antigen-binding fragments thereof in the present invention should be understood as more than one, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12 kinds, 13 kinds, 14 kinds, 15 kinds, 16 kinds, 17 kinds, 18 kinds, 19 kinds, 20 kinds, 21 kinds of combinations.
  • the antibodies or antigen-binding fragments thereof of the present invention can also be combined with other antibodies or antigen-binding fragments thereof.
  • the antibody or antigen-binding fragment thereof is a combination of 2 specific antibodies or antigen-binding fragments thereof, such as a combination of BD55-5840 and BD55-5514.
  • Figure 1 shows the sub-genre breakdown of Sarbecovirus.
  • Figure 2 shows the body weight change curve of mice in the SARS-CoV-2 (Omicron BA.1) challenge experiment.
  • FIG. 3 shows the viral load of mouse tissues and organs in the SARS-CoV-2 (Omicron BA.1) challenge experiment.
  • Figure 4 shows the body weight change curve of mice in the SARS-CoV-2 (Omicron BA.5) challenge experiment.
  • Figure 5 shows the virus load of mouse tissues and organs in the SARS-CoV-2 (Omicron BA.5) challenge experiment.
  • Figure 6 shows the cryo-EM structure of BD55-5514+BD55-5840 combined with BA.1 Spike.
  • Figure 7 shows the binding epitope of BD55-5514 on the BA.1 RBD.
  • Figure 8 shows the binding epitope of BD55-5840 on the BA.1 RBD.
  • antibody refers to immunoglobulins and immunoglobulin fragments, whether native or partially or wholly synthetically (e.g., recombinantly) produced, including retained full-length immunoglobulins comprising at least part of the variable region of an immunoglobulin molecule. Any fragment of the binding specificity of a globulin.
  • an antibody includes any protein having a binding domain that is homologous or substantially homologous to an immunoglobulin antigen binding domain (antibody combining site).
  • Antibodies include antibody fragments.
  • antibody includes synthetic antibodies, recombinantly produced antibodies, multispecific antibodies (e.g., bispecific antibodies), human antibodies, non-human antibodies, humanized antibodies, chimeric antibodies, intrabodies, and antibody fragments, For example, but not limited to, Fab fragments, Fab' fragments, F(ab') 2 fragments, Fv fragments, disulfide-linked Fv (dsFv), Fd fragments, Fd' fragments, single chain Fv (scFv), single chain Fab( scFab), diabodies, anti-idiotypic (anti-Id) antibodies, or antigen-binding fragments of any of the foregoing.
  • Fab fragments Fab' fragments, F(ab') 2 fragments, Fv fragments, disulfide-linked Fv (dsFv), Fd fragments, Fd' fragments, single chain Fv (scFv), single chain Fab( scFab), diabodies, anti-idiotypic (anti-Id) antibodies,
  • Antibodies provided herein include any immunoglobulin class (e.g., IgG, IgM, IgD, IgE, IgA, and IgY), any class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2) or subclass (e.g., IgG2a and IgG2b) members.
  • immunoglobulin class e.g., Ig., IgG, IgM, IgD, IgE, IgA, and IgY
  • any class e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2
  • subclass e.g., IgG2a and IgG2b
  • an "antibody fragment” or “antigen-binding fragment” of an antibody refers to any portion of a full-length antibody that is less than full-length but includes at least a portion of the variable region (e.g., one or more CDRs and/or one or more antibody combining sites), and thus retain binding specificity and at least part of the specific binding ability of the full-length antibody.
  • an antigen-binding fragment refers to an antibody fragment that comprises an antigen-binding portion that binds the same antigen as the antibody from which the antibody fragment is derived.
  • Antibody fragments include antibody derivatives produced by enzymatic treatment of full-length antibodies, as well as derivatives produced synthetically, eg, recombinantly.
  • Antibodies include antibody fragments.
  • antibody fragments include, but are not limited to, Fab, Fab', F(ab') 2 , single chain Fv (scFv), Fv, dsFv, diabodies, Fd and Fd' fragments and other fragments, including modified fragments (see, For example, Methods in Molecular Biology, Vol 207: Recombinant Antibodies for Cancer Therapy Methods and Protocols (2003); Chapter 1; p 3-25, Kipriyanov).
  • the fragments may comprise multiple chains linked together, for example by disulfide bonds and/or by peptide linkers.
  • Antibody fragments generally comprise at least or about 50 amino acids, and typically at least or about 200 amino acids.
  • Antigen-binding fragments include any fragment of an antibody which, when inserted into an antibody framework (e.g., by replacing the corresponding region), results in an antibody that immunospecifically binds (i.e. exhibits a Ka of at least or at least about 10 7 -10 8 M ⁇ 1 ) an antigen .
  • monoclonal antibody refers to a population of identical antibodies, meaning that each individual antibody molecule in the population of monoclonal antibodies is identical to the other antibody molecules. This property is in contrast to that of polyclonal populations of antibodies, which comprise antibodies with a variety of different sequences.
  • Monoclonal antibodies can be prepared by many well-known methods (Smith et al. (2004) J. Clin. Pathol. 57, 912-917; and Nelson et al., J Clin Pathol (2000), 53, 111-117) .
  • monoclonal antibodies can be prepared by immortalizing B cells, eg, by fusing with myeloma cells to generate hybridoma cell lines or by infecting B cells with a virus such as EBV.
  • Recombinant techniques can also be used to prepare antibodies in vitro from clonal populations of host cells by transforming the host cells with plasmids carrying artificial sequences of nucleotides encoding the antibodies.
  • hybridomas refers to a cell or cell line (typically a myeloma or lymphoma cell) resulting from the fusion of antibody-producing lymphocytes and non-antibody-producing cancer cells.
  • hybridomas can proliferate and sustain production of specific monoclonal antibodies. Methods for producing hybridomas are known in the art (see, eg, Harlow & Lane, 1988).
  • hybridodoma or “hybridoma cell” is referred to, it also includes subclones and progeny cells of the hybridoma.
  • inventional antibody refers to an antibody comprising two heavy chains (which may be designated as H and H') and two light chains (which may be designated as L and L') and two antigen binding sites, wherein Each heavy chain may be a full-length immunoglobulin heavy chain or any functional region thereof that retains antigen binding ability (e.g., heavy chains include, but are not limited to, VH chains, VH-CH1 chains, and VH-CH1-CH2-CH3 chains), and Each light chain can be a full-length light chain or any functional region (eg, light chains include, but are not limited to, VL chains and VL-CL chains). Each heavy chain (H and H') is paired with one light chain (L and L', respectively).
  • a full-length antibody is one that has two full-length heavy chains (e.g., VH-CH1-CH2-CH3 or VH-CH1-CH2-CH3-CH4) and two full-length light chains (VL-CL) and a hinge region.
  • Antibodies such as those produced naturally by antibody-secreting B cells, as well as antibodies produced synthetically with the same domain.
  • dsFv refers to a Fv with an engineered intermolecular disulfide bond that stabilizes the VH-VL pair.
  • a Fab fragment is an antibody fragment obtained by papain digestion of a full-length immunoglobulin, or a fragment having the same structure produced synthetically, for example, by recombinant methods.
  • the Fab fragment comprises a light chain (comprising VL and CL) and another chain comprising the variable domain (VH) of the heavy chain and one constant region domain (CH1) of the heavy chain.
  • an F(ab') 2 fragment is an antibody fragment resulting from pepsin digestion of an immunoglobulin at pH 4.0-4.5, or a fragment having the same structure produced synthetically, eg, by recombinant methods.
  • An F(ab')2 fragment essentially consists of two Fab fragments, where each heavy chain portion contains an additional few amino acids, including a cysteine that forms a disulfide bond linking the two fragments.
  • a Fab' fragment is a fragment comprising half (one heavy chain and one light chain) of an F(ab') 2 fragment.
  • a scFv fragment refers to an antibody fragment comprising a variable light chain ( VL ) and a variable heavy chain ( VH ) covalently linked by a polypeptide linker in any order.
  • the length of the linker is such that the two variable domains are bridged substantially without interference.
  • An exemplary linker is (Gly-Ser) n residues interspersed with some Glu or Lys residues to increase solubility.
  • chimeric antibody refers to an antibody in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody antibodies.
  • Humanized antibodies refer to non-human (e.g., mouse) antibody forms that are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (e.g., Fv, Fab, Fab', F(ab') 2 or Antigen-binding subsequences of antibodies), containing minimal sequence derived from non-human immunoglobulins.
  • the humanized antibody is a human immunoglobulin (recipient antibody) in which residues from the complementarity determining regions (CDRs) of the recipient antibody are derived from a non-human species having the desired specificity, affinity and capacity ( Donor antibody) such as mouse, rat or rabbit CDR residue substitutions.
  • CDRs complementarity determining regions
  • the humanized antibody of the present invention also covers an antibody containing 1 or 2 two amino acid mutations in the CDR.
  • epitopic determinants refers to any antigenic determinant on an antigen to which the paratope of an antibody binds.
  • Epitopic determinants usually comprise chemically active surface types of molecules, such as amino acids or sugar side chains, and usually have specific three-dimensional structural characteristics as well as specific charge characteristics.
  • variable domain or variable region is a specific Ig domain of an antibody heavy or light chain comprising an amino acid sequence that varies from antibody to antibody.
  • Each light chain and each heavy chain has one variable region domain, VL and VH, respectively.
  • the variable domains provide antigen specificity and are thus responsible for antigen recognition.
  • Each variable region comprises CDRs and framework regions (FRs), the CDRs being part of the antigen binding site domain.
  • antigen-binding domain and "antigen-binding site” are used synonymously to refer to the domain within an antibody that recognizes and physically interacts with a cognate antigen.
  • a natural conventional full-length antibody molecule has two conventional antigen binding sites, each comprising a heavy chain variable region portion and a light chain variable region portion.
  • the conventional antigen binding site comprises loops linking antiparallel beta strands within the variable region domains.
  • the antigen binding site may comprise other portions of the variable region domain.
  • Each conventional antigen binding site contains 3 hypervariable regions from the heavy chain and 3 hypervariable regions from the light chain. Hypervariable regions are also known as complementarity determining regions (CDRs).
  • variable region domain contains 3 CDRs, named CDR1, CDR2 and CDR3.
  • the light chain variable region domain contains 3 CDRs, named VL CDR1, VL CDR2, and VL CDR3;
  • the heavy chain variable region domain contains 3 CDRs, named VH CDR1, VH CDR2, and VH CDR3.
  • the three CDRs in the variable region are not contiguous along the linear amino acid sequence, but are close together in the folded polypeptide.
  • the CDRs are located within loops connecting the parallel strands of the beta sheets of the variable domains.
  • those skilled in the art know and can identify CDRs based on Kabat or Chothia numbering (see, e.g., Kabat, E.A. et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, and Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917).
  • the framework regions are the domains within the antibody variable region domains located within the beta sheet; the FR regions are relatively more conserved than the hypervariable regions in terms of amino acid sequence.
  • a "constant region” domain is a domain in an antibody heavy or light chain that comprises a relatively more conserved amino acid sequence than that of a variable region domain.
  • each light chain has a single light chain constant region (CL) domain, while each heavy chain contains one or more heavy chain constant region (CH) domains, including CH1, CH2, CH3 and CH4.
  • Full-length IgA, IgD and IgG isotypes comprise CH1, CH2, CH3 and the hinge region, while IgE and IgM comprise CH1, CH2, CH3 and CH4.
  • the CH1 and CL domains extend the Fab arms of the antibody molecule, thus facilitating interaction with the antigen as well as turning the antibody arms.
  • Antibody constant regions may serve effector functions such as, but not limited to, clearance of antigens, pathogens and toxins to which the antibody specifically binds, such as by interacting with various cells, biomolecules and tissues.
  • a functional region of an antibody is a portion of an antibody comprising at least the VH, VL, CH (eg, CH1, CH2 or CH3), CL or hinge domain of the antibody, or at least a functional region thereof.
  • a functional region of a VH domain is at least a portion of a complete VH domain that retains at least a portion of the binding specificity of the complete VH domain (e.g., by retaining one or more CDRs of the complete VH domain), such that the VH A functional region of a domain binds antigen alone or in combination with another antibody domain (eg a VL domain) or region thereof.
  • exemplary functional regions of the VH domain are regions comprising CDR1, CDR2 and/or CDR3 of the VH domain.
  • a functional region of a VL domain is at least a portion of a complete VL domain that retains at least a portion of the binding specificity of the complete VL domain (e.g., by retaining one or more CDRs of the complete VL domain), such that the VL domain A functional region of a domain binds antigen alone or in combination with another antibody domain (eg a VH domain) or region thereof.
  • exemplary functional regions of the VL domain are regions comprising CDR1, CDR2 and/or CDR3 of the VL domain.
  • an antibody that immunospecifically binds (or specifically binds ) an antigen has an affinity constant Ka (or 1 x 10 -7 M or A dissociation constant (K d ) of 1 ⁇ 10 -8 M or lower) binds the antigen.
  • Affinity constants can be determined by standard kinetic methods of antibody reactions, for example, immunoassays, surface plasmon resonance (SPR) (Rich and Myszka (2000) Curr. Opin. Biotechnol 11:54; Englebienne (1998) Analyst. 123: 1599), isothermal titration calorimetry (ITC), or other kinetic interaction assays known in the art (see, for example, Paul, ed., Fundamental Immunology, 2nd ed., Raven Press, New York, pages 332-336 (1989); see also US Patent No. 7,229,619 describing exemplary SPR and ITC methods for calculating the binding affinity of antibodies).
  • SPR surface plasmon resonance
  • ITC isothermal titration calorimetry
  • the term "compete" with respect to antibodies means that a first antibody or antigen-binding fragment thereof binds an epitope in a manner sufficiently similar to a second antibody or antigen-binding fragment thereof such that the first antibody binds to its cognate epitope. Binding results are detectably reduced in the presence of the second antibody compared to the absence of the second antibody. Alternatively, this may, but need not be the case, where the binding of the second antibody to its epitope is also detectably reduced in the presence of the primary antibody. That is, a first antibody can inhibit the binding of a second antibody to its epitope without the second antibody inhibiting the binding of the first antibody to its respective epitope.
  • each antibody detectably inhibits the binding of the other antibody to its cognate epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to "cross-compete" binding with each other their respective epitopes.
  • Both competing and cross-competing antibodies are encompassed by the invention. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope or fragment thereof), those skilled in the art will appreciate that such competing and/or cross-competing antibodies are based on the teachings provided herein. are contemplated by the present invention and may be used in the methods disclosed herein.
  • polypeptide refers to two or more amino acids covalently linked.
  • polypeptide and protein are used interchangeably herein.
  • isolated protein means a protein, polypeptide, or antibody that: (1) is not associated with its naturally associated components in its native state, (2) is free from the same Other proteins of the species, (3) are expressed by cells from a different species, or (4) do not occur in nature.
  • a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which the polypeptide is naturally derived will be “isolated” from its naturally associated components. Proteins can also be rendered substantially free of naturally associated components by isolation, ie, using protein purification techniques well known in the art.
  • Suitable conservative amino acid substitutions are known to those skilled in the art and can generally be made without altering the biological activity of the resulting molecule.
  • those skilled in the art recognize that single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al., Molecular Biology of the Gene, 4th Edition, 1987, The Benjamin/Cummings Pub .co., p. 224).
  • conservative substitution means an amino acid substitution that does not adversely affect or alter the expected properties of the protein/polypeptide comprising the amino acid sequence.
  • conservative substitutions can be introduced by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • Conservative amino acid substitutions include substitutions for amino acid residues with amino acid residues that have similar side chains, e.g., are physically or functionally similar (e.g., have similar size, shape, charge, chemical properties, including ability to form covalent or hydrogen bonds, etc.) Families of amino acid residues having similar side chains have been defined in the art.
  • These families include those with basic side chains (e.g., lysine, arginine, and histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine) , asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), non-polar side chains (such as alanine, valine, leucine, isoleucine amino acid, proline, phenylalanine, methionine), beta branched side chains (e.g. threonine, valine, isoleucine) and aromatic side chains (e.g.
  • basic side chains e.g., lysine, arginine, and histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine
  • non-polar side chains such as
  • amino acids are generally represented by single-letter and three-letter abbreviations known in the art.
  • alanine can be represented by A or Ala.
  • polynucleotide and “nucleic acid molecule” refer to an oligomer or polymer comprising at least two linked nucleotides or nucleotide derivatives, including nucleotides linked together usually by phosphodiester bonds.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • an isolated nucleic acid molecule is a nucleic acid molecule that is separated from other nucleic acid molecules that exist in the natural source of the nucleic acid molecule.
  • An "isolated" nucleic acid molecule such as a cDNA molecule, may be substantially free of other cellular material or culture medium when prepared by recombinant techniques, or substantially free of chemical precursors or other chemical components when chemically synthesized.
  • Exemplary isolated nucleic acid molecules provided herein include isolated nucleic acid molecules encoding provided antibodies or antigen-binding fragments.
  • Sequence "identity” has an art-recognized meaning, and the percent sequence identity between two nucleic acid or polypeptide molecules or regions can be calculated using published techniques. Sequence identity can be measured along the entire length of a polynucleotide or polypeptide, or along a region of the molecule.
  • identity is well known to those skilled in the art (Carrillo, H. & Lipman, D., SIAM J Applied Math 48:1073 (1988) ).
  • operably linked with reference to nucleic acid sequences, regions, elements or domains means that the nucleic acid regions are functionally related to each other.
  • a promoter can be operably linked to a nucleic acid encoding a polypeptide such that the promoter regulates or mediates transcription of the nucleic acid.
  • a "vector" is a replicable nucleic acid from which one or more heterologous proteins can be expressed when the vector is transformed into an appropriate host cell.
  • Reference to vectors includes those into which a nucleic acid encoding a polypeptide or fragment thereof can be introduced, typically by restriction digestion and ligation. Reference to vectors also includes those vectors comprising a nucleic acid encoding a polypeptide.
  • Vectors are used to introduce nucleic acids encoding polypeptides into host cells for amplifying nucleic acids or expressing/displaying polypeptides encoded by nucleic acids. Vectors usually remain episomal, but can be designed to allow integration of a gene, or part thereof, into the chromosome of the genome. Also contemplated are vectors for artificial chromosomes, such as yeast artificial vectors and mammalian artificial chromosomes. The selection and use of such vehicles are well known to those skilled in the art.
  • vector also includes "viral vector” or “viral vector”.
  • viral vectors are engineered viruses that are operably linked to foreign genes to transfer (either as vehicles or shuttles) foreign genes into cells.
  • expression refers to the process by which a polypeptide is produced by the transcription and translation of a polynucleotide. Expression levels of a polypeptide can be assessed by any method known in the art, including, for example, methods for determining the amount of polypeptide produced by a host cell. Such methods may include, but are not limited to, quantification of polypeptides in cell lysates by ELISA, Coomassie blue staining after gel electrophoresis, Lowry protein assay, and Bradford protein assay.
  • expression vector includes a vector capable of expressing DNA operably linked to regulatory sequences, such as a promoter region, capable of affecting the expression of such DNA fragments. Such additional fragments may include promoter and terminator sequences, and optionally may include one or more origins of replication, one or more selectable markers, enhancers, polyadenylation signals, and the like. Expression vectors are generally derived from plasmid or viral DNA, or may contain elements of both. Thus, an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid, phage, recombinant virus or other vector, which, when introduced into an appropriate host cell, results in the expression of cloned DNA. Appropriate expression vectors are well known to those skilled in the art and include expression vectors replicable in eukaryotic cells and/or prokaryotic cells as well as expression vectors that remain episomal or integrate into the host cell genome.
  • Codon optimization refers to the replacement of at least one codon of a native sequence (e.g. about or more than about 1, 2, 3, 4, 5 , 10, 15, 20, 25, 50 or more codons while maintaining the native amino acid sequence and modifying the nucleic acid sequence so as to enhance expression in the host cell of interest.
  • a native sequence e.g. about or more than about 1, 2, 3, 4, 5 , 10, 15, 20, 25, 50 or more codons
  • Codon bias differs in codon usage between organisms
  • mRNA messenger RNA
  • tRNA transfer RNA
  • Codon Usage Database Codon Usage Database
  • Codon Usage Database available at www.kazusa.orjp/codon/
  • these tables can be accessed through various Modal adjustments apply. See, Nakamura Y. et al., "Codon usage tabulated from the international DNA sequence databases: status for the year 2000. Nucl. Acids Res., 28: 292 (2000).
  • a "host cell” is a cell for receiving, maintaining, replicating and amplifying a vector.
  • Host cells can also be used to express polypeptides encoded by vectors. When the host cell divides, the nucleic acid contained in the vector replicates, thereby amplifying the nucleic acid.
  • Host cells can be eukaryotic or prokaryotic. Suitable host cells include, but are not limited to, CHO cells, various COS cells, HeLa cells, HEK cells such as HEK 293 cells.
  • the term "pharmaceutically acceptable carrier and/or excipient” refers to a carrier and/or excipient compatible with the subject and the active ingredient pharmacologically and/or physiologically, These are well known in the art (see, e.g., Remington's Pharmaceutical Sciences. Edited by Gennaro AR, 19th ed. Pennsylvania: Mack Publishing Company, 1995), and include, but are not limited to: pH adjusters, surfactants, adjuvants, ions Strength enhancers, diluents, agents to maintain osmotic pressure, agents to delay absorption, preservatives.
  • pH adjusting agents include, but are not limited to, phosphate buffers.
  • Surfactants include but are not limited to cationic, anionic or nonionic surfactants such as Tween-80.
  • Ionic strength enhancers include, but are not limited to, sodium chloride.
  • Preservatives include, but are not limited to, various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Agents to maintain osmotic pressure include, but are not limited to, sugars, NaCl, and the like.
  • Agents that delay absorption include, but are not limited to, monostearates and gelatin.
  • Diluents include, but are not limited to, water, aqueous buffers (eg, buffered saline), alcohols and polyols (eg, glycerol), and the like.
  • Preservatives include, but are not limited to, various antibacterial and antifungal agents, such as thimerosal, 2-phenoxyethanol, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Stabilizer has the meaning generally understood by those skilled in the art, and it can stabilize the desired activity of the active ingredient in the medicine, including but not limited to sodium glutamate, gelatin, SPGA, sugars (such as sorbitol, mannitol, starch, sucrose , lactose, dextran, or glucose), amino acids (such as glutamic acid, glycine), proteins (such as dry whey, albumin or casein) or their degradation products (such as lactalbumin hydrolyzate), etc.
  • the pharmaceutically acceptable carrier or excipient comprises a sterile injectable liquid (eg, aqueous or non-aqueous suspension or solution).
  • such sterile injectable liquids are selected from the group consisting of water for injection (WFI), bacteriostatic water for injection (BWFI), sodium chloride solution (eg 0.9% (w/v) NaCl), dextrose solutions (eg, 5% dextrose), solutions containing surfactants (eg, 0.01% polysorbate 20), pH buffered solutions (eg, phosphate buffered saline), Ringer's solutions, and any combination thereof.
  • WFI water for injection
  • BWFI bacteriostatic water for injection
  • sodium chloride solution eg 0.9% (w/v) NaCl
  • dextrose solutions eg, 5% dextrose
  • solutions containing surfactants eg, 0.01% polysorbate 20
  • pH buffered solutions eg, phosphate buffered saline
  • Ringer's solutions e.g, Ringer's solutions, and any combination thereof.
  • prevention refers to methods performed to prevent or delay the occurrence of a disease or disorder or symptom (eg, SARS-CoV-2 infection) in a subject.
  • treatment refers to a method performed to obtain a beneficial or desired clinical result.
  • a beneficial or desired clinical outcome includes, but is not limited to, relief of symptoms, reduction of the extent of the disease, stabilization (i.e., no longer worsening) of the disease state, delay or slowing of the progression of the disease, amelioration or palliation of the disease status, and relief of symptoms (whether partial or total), whether detectable or undetectable.
  • treating can also refer to prolonging survival as compared to expected survival if not receiving treatment.
  • therapeutic effect means the effect resulting from a treatment in an individual that alters, usually ameliorates or ameliorate the symptoms of, or cures a disease or condition.
  • prophylactically effective amount or “prophylactically effective dose” refers to the amount of a substance, compound, material, or composition comprising a compound that, when administered to a subject, will have the desired prophylactic effect, e.g., prevent or delay a disease or symptom occurrence or recurrence of the disease or symptoms, and to reduce the likelihood of occurrence or recurrence of the disease or symptoms.
  • a full prophylactically effective dose does not have to occur by administering one dose, and can only occur after administering a series of doses.
  • a prophylactically effective amount can be administered in one or more administrations.
  • terapéuticaally effective amount or “therapeutically effective dose” refers to an amount of a substance, compound, material, or composition comprising a compound that is at least sufficient to produce a therapeutic effect when administered to a subject. Thus, it is the amount necessary to prevent, cure, ameliorate, arrest or partially arrest the symptoms of a disease or disorder.
  • the term "subject” preferably refers to a mammal, such as a human.
  • the subject e.g., a human
  • SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
  • SARS-CoV-2 contains at least three membrane proteins, including surface spike protein (S), integral membrane protein (M) and membrane protein (E).
  • S surface spike protein
  • M integral membrane protein
  • E membrane protein
  • the receptor of SARS-CoV-2 is the same as that of SARS-CoV, through the receptor binding domain (RBD) on the S protein to specifically bind to angiotensin transferase 2 (ACE2) on the host cell After receiving the membrane fusion and cell entry of the virus, it plays a vital role in the process of virus infection of cells.
  • ACE2 angiotensin transferase 2
  • the term "SARS-CoV-2" includes various known isolates, such as both the original strain (such as the first sequenced isolate GenBank: MN908947.3), and the subsequently discovered mutant strain.
  • the term “SARS-CoV-2” encompasses both isolates whose S protein does not contain a mutation (for example, compared to the reference strain MN908947.3) and includes a mutation in its S protein (for example, compared to Amino acid substitutions compared to the reference strain MN908947.3, eg K417N, E484K, N501Y, L452R, T478K or any combination thereof).
  • the variant strain is preferably selected from isolates comprising mutations (eg amino acid substitutions such as K417N, E484K, N501Y, L452R, T478K or any combination thereof) in their S protein.
  • the mutant strain is selected from Alpha (Alpha, B.1.1.7), Beta (Beta, B.1.351), Gamma (Gamma, P.1), Delta (Delta, B.1.617.2) and Omicron (Omicron, B.1.1.529).
  • new coronavirus pneumonia and “COVID-19” refer to pneumonia caused by SARS-CoV-2 infection, both have the same meaning and can be used interchangeably.
  • neutralizing activity means that the antibody or antibody fragment has the ability to bind to the antigenic protein on the virus, thereby preventing the virus from infecting the cells and/or the maturation of the virus progeny and/or the release of the virus progeny
  • the functional activity of the antibody or antibody fragment with neutralizing activity can prevent the amplification of the virus, thereby inhibiting or eliminating the infection of the virus.
  • the present invention provides an antibody or an antigen-binding fragment thereof comprising a heavy chain variable region and a light chain variable region comprising complementarity determining regions (CDRs) of specific amino acid sequences, Or an amino acid sequence having 1 or 2 amino acid residue substitutions, deletions or additions relative to a specific amino acid sequence.
  • CDRs complementarity determining regions
  • the heavy and light chain variable regions comprise specific amino acid sequences or variants thereof. wherein said variant has a substitution, deletion or addition of one or several amino acids, such as 1, 2, 3, 4 or 5 amino acid substitutions, deletions or additions, compared to the sequence from which it is derived, or have at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% Sequences with sequence identity; preferably, said substitutions are conservative substitutions.
  • the antibody or antigen-binding fragment thereof comprises a constant region derived from a human immunoglobulin, eg, a heavy chain constant region and a light chain constant region comprising specific amino acid sequences.
  • the information of the partial sequences involved in the present invention is provided in the following Table 1, specifically related to 21 kinds of antibodies (numbering respectively BD55-1239, BD55-3372, BD55-3500, BD55-3546, BD55-4637, BD55-5242, BD55 -5263, BD55-5300, BD55-5386, BD55-5477, BD55-5483, BD55-5484, BD55-5514, BD55-5549, BD55-5558, BD55-5585, BD55-5591, BD55-5640, BD55-5697 , BD55-5700, BD55-5840) VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, VL CDR3 sequences and VH, VL sequences.
  • These 21 antibodies have common CH (SEQ ID NO: 169) and CL (SEQ ID NO: 170) sequences.
  • the antibodies of the present invention can be prepared by various methods known in the art, for example, by genetic engineering and recombination techniques. For example, DNA molecules encoding the heavy and light chain genes of the antibodies of the present invention are obtained by chemical synthesis or PCR amplification. The resulting DNA molecule is inserted into an expression vector and then transfected into a host cell. Then, the transfected host cells are cultured under specific conditions, and express the antibody of the present invention.
  • Antigen-binding fragments of the present invention can be obtained by hydrolyzing intact antibody molecules (see Morimoto et al., J. Biochem. Biophys. Methods 24: 107-117 (1992) and Brennan et al., Science 229: 81 (1985)) . In addition, these antigen-binding fragments can also be directly produced by recombinant host cells (reviewed in Hudson, Curr. Opin. Immunol. 11: 548-557 (1999); Little et al., Immunol. Today, 21: 364-370 (2000 )).
  • Fab' fragments can be obtained directly from host cells; Fab' fragments can be chemically coupled to form F(ab') 2 fragments (Carter et al., Bio/Technology, 10:163-167 (1992)).
  • Fv, Fab or F(ab') 2 fragments can also be directly isolated from the culture medium of recombinant host cells. Other techniques for preparing these antigen-binding fragments are well known to those of ordinary skill in the art.
  • the invention provides an isolated nucleic acid molecule comprising nucleotides encoding an antibody of the invention or an antigen-binding fragment thereof, or a heavy chain variable region and/or a light chain variable region thereof sequence.
  • the isolated nucleic acid molecule encodes an antibody or antigen-binding fragment thereof of the invention, or a heavy chain variable region and/or a light chain variable region thereof.
  • the isolated nucleic acid molecule comprises a first nucleotide sequence encoding a heavy chain or a heavy chain variable region of an antibody or antigen-binding fragment thereof of the invention and a first nucleotide sequence encoding the antibody or antigen-binding fragment thereof.
  • the isolated nucleic acid molecule of the present invention comprises a second nucleotide sequence comprising the first nucleotide sequence.
  • the invention provides a vector (eg, a cloning vector or an expression vector) comprising an isolated nucleic acid molecule as described above.
  • vectors of the invention are, for example, plasmids, cosmids, phage, and the like.
  • the vector comprises a first nucleotide sequence encoding a heavy chain or a heavy chain variable region of an antibody of the invention or an antigen-binding fragment thereof and a light chain or a light chain encoding the antibody or an antigen-binding fragment thereof
  • the second nucleotide sequence of the light chain variable region wherein the first nucleotide sequence and the second nucleotide sequence are present on the same or different vectors.
  • the vector of the present invention comprises a first vector containing the first nucleotide sequence and a vector containing the A second vector of the second nucleotide sequence.
  • the vector comprises a first nucleotide sequence encoding a heavy chain variable region of an antibody of the invention or an antigen-binding fragment thereof, and/or encoding a light chain of an antibody of the invention or an antigen-binding fragment thereof A second nucleotide sequence of the variable region; wherein said first and second nucleotide sequences are provided on the same or different vectors.
  • the vector comprises a first nucleotide sequence encoding a heavy chain of an antibody of the invention or an antigen-binding fragment thereof, and/or a second nucleotide sequence encoding a light chain of an antibody or an antigen-binding fragment thereof of the invention A nucleotide sequence; wherein said first nucleotide sequence and second nucleotide sequence are provided on the same or different vectors.
  • the present invention provides a host cell transformed with a nucleic acid molecule of the present invention or an expression vector of the present invention.
  • host cells include, but are not limited to, prokaryotic cells such as bacterial cells (such as E. coli cells), and eukaryotic cells such as fungal cells (such as yeast cells), insect cells, plant cells, and animal cells (such as mammalian cells, such as small mouse cells, human cells, etc.).
  • prokaryotic cells such as bacterial cells (such as E. coli cells), and eukaryotic cells such as fungal cells (such as yeast cells), insect cells, plant cells, and animal cells (such as mammalian cells, such as small mouse cells, human cells, etc.).
  • the host cells of the invention are mammalian cells, such as CHO cells, various COS cells, HeLa cells, HEK cells such as HEK 293 cells. .
  • a method for preparing the antibody of the present invention or an antigen-binding fragment thereof comprising culturing the host cell of the present invention under the expression of a nucleic acid molecule or expression vector suitable for the present invention, and isolating and purifying the antibody obtained from the present invention.
  • pharmaceutically acceptable carrier and/or excipient includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, etc. that are physiologically compatible .
  • the carrier and/or excipient are suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (eg by injection or infusion).
  • the active compound ie, antibody molecule, immunoconjugate
  • compositions of the present invention may also contain pharmaceutically acceptable antioxidants.
  • pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, etc.; (2) oil-soluble antioxidants, such as ascorbic acid palmitate esters, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, ⁇ -tocopherol, etc.; and (3) metal chelating agents such as citric acid, ethylenediaminetetraacetic acid (EDTA ), sorbitol, tartaric acid, phosphoric acid, etc.
  • water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, etc.
  • oil-soluble antioxidants such as ascorbic acid palmitate esters, butylated hydroxyanisole (
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Prevention of the presence of microorganisms can be ensured by sterilization procedures or by the inclusion of various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol sorbic acid, and the like.
  • various antibacterial and antifungal agents for example, parabens, chlorobutanol, phenol sorbic acid, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol or sodium oxide in the composition.
  • Prolonged absorption of injectable drugs can be brought about by including in the composition agents which delay absorption, for example, monostearate salts and gelatin.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Conventional media or agents, to the extent they are incompatible with the active compounds, are possible in the pharmaceutical compositions of the present invention. Supplementary active compounds can also be incorporated into the compositions.
  • compositions generally must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • the carrier can be a solvent or dispersion containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • Proper fluidity can be maintained, for example, by the use of coatings such as lecithin, by maintaining the desired particle size in the case of dispersions, and by the use of surfactants.
  • Sterile injectable solutions can be prepared by mixing the active compound in the required amount in an appropriate solvent and, as required, adding one or a combination of ingredients enumerated above, followed by sterile microfiltration.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying (lyophilization) which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated and the particular mode of administration.
  • the amount of active ingredient which can be combined with carrier materials to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Generally, based on 100%, this amount will range from about 0.01% to about 99% of the active ingredient, preferably from about 0.1% to about 70%, most preferably from about 1% to about 30%, and pharmaceutically acceptable combination of carriers.
  • Dosage regimens can be adjusted to provide the optimum desired response (eg, a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to result in combination with the required pharmaceutical carrier. desired therapeutic effect.
  • dosages range from about 0.0001 to 100 mg/kg, more typically 0.01 to 20 mg/kg of recipient body weight.
  • dosages may be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight, 10 mg/kg body weight or 20 mg/kg body weight, or within the range of 1-20 mg/kg.
  • Exemplary treatment regimens entail weekly dosing, once every two weeks, once every three weeks, once every four weeks, once monthly, once every 3 months, once every 3-6 months, or an initial dosing interval Slightly shorter (eg once a week to once every three weeks) followed by longer dosing intervals (eg monthly to once every 3-6 months).
  • antibody molecules of the invention may also be administered as a sustained release formulation, in which case less frequent dosing is required. Dosage and frequency vary according to the half-life of the antibody molecule in the patient. In general, human antibodies exhibit the longest half-life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. Dosage and frequency of administration will vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, relatively lower doses are administered at infrequent intervals over a prolonged period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, it is sometimes desirable to administer higher doses at shorter intervals until the progression of the disease is reduced or stopped, preferably until the patient exhibits partial or complete amelioration of disease symptoms. Thereafter, the patient can be dosed in a prophylactic regimen.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied to obtain an amount of the active ingredient effective to achieve the desired therapeutic response for a particular patient, composition and mode of administration without being toxic to the patient.
  • the selected dosage level will depend on a variety of pharmacokinetic factors, including the activity of the particular composition of the invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound employed, the duration of treatment, and the particular Other drugs, compounds and/or materials with which the composition is used in conjunction, the age, sex, weight, condition, general health and medical history of the patient being treated, and similar factors well known in the medical field.
  • the antibody or antigen-binding fragment thereof of the present invention or the pharmaceutical composition of the present invention can be administered by one or more routes of administration using one or more methods known in the art. Those skilled in the art will appreciate that the route and/or manner of administration will vary depending on the desired result. Preferred routes of administration for antibodies of the invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, such as injection or infusion.
  • parenteral administration refers to modes of administration other than enteral and topical administration, usually injection, including but not limited to intravenous, intramuscular, intraarterial, intrathecal, intrasaccular, intraorbital, Intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcutaneous, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, and intrasternal injection and infusion.
  • the antibody or antigen-binding fragment thereof of the invention or the pharmaceutical composition of the invention may also be administered by non-parenteral routes, such as topical, epidermal or mucosal routes, for example, intranasally, orally, vaginally, rectally, Sublingual or topical.
  • non-parenteral routes such as topical, epidermal or mucosal routes, for example, intranasally, orally, vaginally, rectally, Sublingual or topical.
  • the active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controlled release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • therapeutic compositions can be administered using medical devices known in the art.
  • the therapeutic compositions of the present invention may be administered with a needle-free hypodermic injection device, such as the devices disclosed in US Patent Nos. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880;
  • a needle-free hypodermic injection device such as the devices disclosed in US Patent Nos. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880;
  • Examples of known implants and modules that can be used in the present invention include: U.S. Patent No. 4,487,603, which discloses an implantable microinfusion pump for dispersing drugs at a controlled rate; U.S. Patent No. 4,486,194, which discloses Patents disclose therapeutic devices for drug delivery through the skin; U.S. Patent No.
  • antibodies of the invention may be formulated to ensure proper distribution in vivo.
  • the blood-brain barrier (BBB) blocks many highly hydrophilic compounds.
  • BBB blood-brain barrier
  • therapeutic compounds of the invention are able to cross the BBB (if desired)
  • they can be formulated, eg, in liposomes.
  • liposomes For methods of preparing liposomes, see, eg, US Patents 4,522,811; 5,374,548 and 5,399,331.
  • Liposomes contain one or more targeting moieties that can be selectively transported into specific cells or organs, thereby enhancing targeted drug delivery (see, e.g., V.V. Ranade (1989) J. Clin. Pharmacol. 29:685 ).
  • targeting moieties include folic acid or biotin (see, e.g., U.S. Patent 5,416,016 to Low et al); mannosides (Umezawa et al (1988) Biochem. 1995) FEBS Lett.357:140; M.Owais et al. (1995) Antimicrob.Agents Chemother.39:180); Surfactant protein A receptor (Briscoe et al. (1995) Am.J.Physiol.1233:134); p120 (Schreier et al. (1994) J. Biol. Chem. 269:9090); see also K. Keinanen; M.L. Laukkanen (1994) FEBS Lett. 346:123; J.J. Killion; I.J. Fidler (1994) Immunomethods 4:273.
  • the pharmaceutically acceptable carrier and/or excipient comprises a sterile injectable liquid (eg, aqueous or non-aqueous suspension or solution).
  • a sterile injectable liquid eg, aqueous or non-aqueous suspension or solution.
  • sterile injectable liquids are selected from the group consisting of water for injection (WFI), bacteriostatic water for injection (BWFI), sodium chloride solution (eg 0.9% (w/v) NaCl), dextrose solutions (eg, 5% dextrose), solutions containing surfactants (eg, 0.01% polysorbate 20), pH buffered solutions (eg, phosphate buffered saline), Ringer's solutions, and any combination thereof.
  • WFI water for injection
  • BWFI bacteriostatic water for injection
  • sodium chloride solution eg 0.9% (w/v) NaCl
  • dextrose solutions eg, 5% dextrose
  • surfactants eg, 0.01% polysorb
  • the antibody of the present invention or its antigen-binding fragment can be used to neutralize B-coronavirus in vitro or in a subject, block or inhibit the infection of cells by B-coronavirus, so as to prevent and/or treat B-coronavirus in a subject beta-coronavirus infection or diseases associated with beta-coronavirus infection.
  • the present invention provides a method for preventing and/or treating diseases caused by B-coronavirus infection, the method comprising administering to a subject an effective amount of the antibody of the present invention or an antigen-binding fragment thereof or the antibody of the present invention pharmaceutical composition.
  • the beta coronavirus includes novel coronavirus (SARS-CoV-2) and variants thereof, severe acute respiratory syndrome coronavirus (SARS-CoV) and variants thereof, SARS-related coronaviruses (SARSr-CoV).
  • SARS-CoV-2 novel coronavirus
  • SARS-CoV severe acute respiratory syndrome coronavirus
  • SARSr-CoV SARS-related coronaviruses
  • the SARS-CoV-2 includes variant strains.
  • the S protein of the mutant strain contains mutations, such as amino acid substitutions, deletions or additions.
  • the S protein of the mutant strain comprises one or more amino acid substitutions selected from K417N, E484K, N501Y, L452R, and T478K.
  • the mutant strain of SARS-CoV-2 is selected from Alpha (Alpha, B.1.1.7), Beta (Beta, B.1.351), Gamma (Gamma, P.1), Delta ( Delta, B.1.617.2), Omicron (B.1.1.529) or any combination thereof.
  • the subject is preferably a mammal, more preferably a human.
  • the antibody or antigen-binding fragment thereof, or the pharmaceutical composition is used alone, or in combination with another pharmaceutically active agent (eg, another antiviral agent).
  • another pharmaceutically active agent eg, another antiviral agent
  • the antibody or antigen-binding fragment thereof of the invention, or the pharmaceutical composition of the invention and the additional pharmaceutically active agent may be administered simultaneously, separately or sequentially.
  • the present invention provides a method for neutralizing B-coronavirus, blocking or inhibiting the binding of B-coronavirus to ACE2 receptor, or blocking or inhibiting the infection of B-coronavirus to cells, which Included are the use of antibodies or antigen-binding fragments thereof or pharmaceutical compositions of the invention.
  • the method can be used in vitro or in a subject (such as a human) to neutralize B-coronavirus, block or inhibit the binding of B-coronavirus to ACE2 receptors, or block or inhibit the infection of B-coronavirus to cells .
  • the method is used to neutralize the virulence of B-coronavirus in the sample, block or inhibit the binding of B-coronavirus to ACE2 receptor, or block or inhibit the binding of B-coronavirus to cells Infect.
  • the method comprises: contacting a sample comprising a betacoronavirus with an antibody or antigen-binding fragment thereof or a pharmaceutical composition of the invention.
  • the present invention relates to the use of the antibody of the present invention or its antigen-binding fragment for the preparation of a medicament for the prevention and/or treatment of diseases caused by B-coronavirus infection, and the medicament is used for one or more of the following item:
  • B-coronavirus infection or diseases related to B-coronavirus infection such as COVID-19
  • the antibody or antigen-binding fragment thereof of the present invention, or the pharmaceutical composition of the present invention can be formulated into any dosage form known in the medical field, for example, tablets, pills, suspensions, emulsions, solutions, gels, capsules, Powders, granules, elixirs, lozenges, suppositories, injections (including injections, sterile powders for injections and concentrated solutions for injections), inhalants, sprays, etc.
  • the preferred dosage form depends on the intended mode of administration and therapeutic use.
  • Antibodies or antigen-binding fragments thereof or pharmaceutical compositions of the invention should be sterile and stable under the conditions of manufacture and storage.
  • a preferred dosage form is injection. Such injections can be sterile injectable solutions.
  • sterile injectable solutions can be prepared by incorporating in an appropriate solvent the necessary dose of an antibody or antigen-binding fragment thereof of the present invention and, optionally, concomitantly incorporating other desired ingredients including, but not limited to pH adjusters, surfactants, adjuvants, ionic strength enhancers, isotonic agents, preservatives, diluents, or any combination thereof), followed by filter sterilization.
  • sterile injectable solutions can be prepared as sterile lyophilized powder (eg, by vacuum drying or freeze-drying) for ease of storage and use.
  • Such sterile lyophilized powders can be dispersed in suitable carriers before use, such as water for injection (WFI), bacteriostatic water for injection (BWFI), sodium chloride solution (such as 0.9% (w/v) NaCl), Dextrose solution (eg 5% glucose), surfactant containing solution (eg 0.01% polysorbate 20), pH buffer solution (eg phosphate buffer solution), Ringer's solution and any combination thereof.
  • WFI water for injection
  • BWFI bacteriostatic water for injection
  • sodium chloride solution such as 0.9% (w/v) NaCl
  • Dextrose solution eg 5% glucose
  • surfactant containing solution eg 0.01% polysorbate 20
  • pH buffer solution eg phosphate buffer solution
  • Ringer's solution any combination thereof.
  • the antibodies of the invention or antigen-binding fragments thereof, or the pharmaceutical compositions of the invention may be administered by any suitable method known in the art, including, but not limited to, oral, buccal, sublingual, ophthalmic, topical, parenteral, Rectal, intrathecal, intracytoplasmic reticulum, inguinal, intravesical, topical (eg, powder, ointment, or drops), or nasal routes.
  • the preferred route/mode of administration is parenteral (eg, intravenous or bolus injection, subcutaneous injection, intraperitoneal injection, intramuscular injection).
  • an antibody or antigen-binding fragment thereof or pharmaceutical composition of the invention is administered by intravenous injection or bolus injection.
  • dosing regimens may be adjusted to obtain the optimum desired response (eg, a therapeutic or prophylactic response). For example, a single dose can be administered, multiple doses can be administered over time or the dose can be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • An antibody or antigen-binding fragment thereof of the invention can be derivatized, eg, linked to another molecule (eg, another polypeptide or protein).
  • another molecule eg, another polypeptide or protein.
  • derivatization eg, labeling
  • the antibodies or antigen-binding fragments thereof of the invention are also intended to include such derivatized forms.
  • an antibody of the invention or an antigen-binding fragment thereof can be functionally linked (by chemical coupling, gene fusion, non-covalent linkage, or otherwise) to one or more other molecular moieties, such as another antibody (e.g., forming bispecific antibodies), detection reagents, pharmaceutical reagents, and/or proteins or polypeptides capable of mediating the binding of an antibody or antigen-binding fragment to another molecule (eg, an avidin or polyhistidine tag).
  • the antibodies or antigen-binding fragments thereof of the invention may also be derivatized with chemical groups, such as polyethylene glycol (PEG), methyl or ethyl groups, or sugar groups. These groups can be used to improve the biological properties of antibodies, such as increasing serum half-life.
  • an antibody or antigen-binding fragment thereof of the invention is detectably labeled.
  • the detectable label of the present invention can be any substance detectable by fluorescent, spectroscopic, photochemical, biochemical, immunological, electrical, optical or chemical means.
  • labels are well known in the art, examples of which include, but are not limited to, enzymes (e.g., horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, urease, glucose oxidase, etc.), radionuclide Chlorine (for example, 3 H, 125 I, 35 S, 14 C or 32 P), fluorescent dyes (for example, fluorescein isothiocyanate (FITC), fluorescein, tetramethylrhodamine isothiocyanate (TRITC) , phycoerythrin (PE), Texas Red, rhodamine, quantum dots or cyanine dye derivatives (such as Cy7, Alexa 750)), luminescent substances (such as chemiluminescent substances, such as acridine ester compounds,
  • the detectable label can be adapted for immunological detection (eg, enzyme-linked immunoassay, radioimmunoassay, fluorescent immunoassay, chemiluminescence immunoassay, etc.).
  • the detectable label may be selected from enzymes (such as horseradish peroxidase, alkaline phosphatase, or ⁇ -galactosidase), chemiluminescent reagents (such as acridinium esters, lucid Minol and its derivatives, or ruthenium derivatives), fluorescent dyes (such as fluorescein or fluorescent proteins, such as FITC, TRITC or PE), radionuclides or biotin.
  • enzymes such as horseradish peroxidase, alkaline phosphatase, or ⁇ -galactosidase
  • chemiluminescent reagents such as acridinium esters, lucid Minol and its derivatives, or ruthenium derivatives
  • fluorescent dyes
  • a detectable label as described above can be attached to an antibody or antigen-binding fragment thereof of the invention via linkers of various lengths to reduce potential steric hindrance.
  • the antibody of the present invention or its antigen-binding fragment can specifically bind to the RBD of the S protein of the B-coronavirus, so that it can be used to detect the RBD of the B-coronavirus or its S protein or S protein, and optionally according to the above detection results Diagnose whether the subject is infected with B-coronavirus.
  • the invention provides a kit comprising an antibody of the invention or an antigen-binding fragment thereof, or a conjugate of the invention.
  • the kit comprises a conjugate of the invention.
  • the kit comprises an antibody or antigen-binding fragment thereof of the invention. In some embodiments, the antibody or antigen-binding fragment thereof does not comprise a detectable label. In some embodiments, the kit further comprises a second antibody that specifically recognizes the antibody of the invention or an antigen-binding fragment thereof; optionally, the second antibody further comprises a detectable label, such as an enzyme (e.g., paprika root peroxidase or alkaline phosphatase), chemiluminescent reagents (such as acridinium esters, luminol and its derivatives, or ruthenium derivatives), fluorescent dyes (such as fluorescein or fluorescent protein), radioactive nuclei biotin or biotin.
  • an enzyme e.g., paprika root peroxidase or alkaline phosphatase
  • chemiluminescent reagents such as acridinium esters, luminol and its derivatives, or ruthenium derivatives
  • fluorescent dyes
  • the second antibody is specific for an antibody of the species (eg, human) from which the constant region comprised by the antibody or antigen-binding fragment thereof of the invention is derived.
  • the second antibody is an anti-immunoglobulin (eg, human immunoglobulin) antibody, eg, an anti-IgG antibody. In some embodiments, the second antibody is an anti-human IgG antibody.
  • kits of the invention may further comprise reagents for allowing the corresponding detectable label to be detected.
  • the detectable label is an enzyme
  • the kit may also comprise a chromogenic substrate for the corresponding enzyme, such as o-phenylenediamine (OPD), tetramethylbenzyldiamine (OPD) for horseradish peroxidase, Aniline (TMB), ABTS or luminol-like compounds, or p-nitrophenyl phosphate (p-NPP) or AMPPD for alkaline phosphatase.
  • OPD o-phenylenediamine
  • TMB Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • ABTS Aniline
  • the present invention provides a method for detecting the presence or level of a betacoronavirus or its S protein or the RBD of the S protein, or cells infected with a betacoronavirus in a sample, comprising making the sample contacting an antibody or antigen-binding fragment or conjugate thereof of the invention; detecting binding of said antibody or antigen-binding fragment or conjugate thereof to a target antigen in said sample; wherein detecting said binding represents said B-coronavirus exists in the sample, or the level of B-coronavirus in the sample is represented by detecting the strength of the combination.
  • the method is an immunological assay, such as an enzyme immunoassay (eg, ELISA), a chemiluminescent immunoassay, a fluorescent immunoassay, or a radioimmunoassay.
  • an enzyme immunoassay eg, ELISA
  • chemiluminescent immunoassay e.g. a chemiluminescent immunoassay
  • fluorescent immunoassay e.g., a fluorescent immunoassay
  • radioimmunoassay e.g., radioimmunoassay.
  • the method comprises using a conjugate of the invention.
  • the method comprises the use of an antibody or antigen-binding fragment thereof of the invention.
  • the antibody or antigen-binding fragment thereof does not comprise a detectable label.
  • the method further comprises using a detectable label (e.g., an enzyme (e.g., horseradish peroxidase or alkaline phosphatase), a chemiluminescent reagent (e.g., acridinium esters, luminol and derivatives thereof, or ruthenium derivatives), fluorescent dyes (such as fluorescein or fluorescent protein), radionuclides or biotin) to detect said antibodies or antigen-binding fragments thereof.
  • a detectable label e.g., an enzyme (e.g., horseradish peroxidase or alkaline phosphatase), a chemiluminescent reagent (e.g., acridinium esters, luminol and derivatives thereof, or ruthenium derivatives), fluorescent dyes (such as fluor
  • the second antibody is specific for an antibody of the species (eg, human) from which the constant region comprised by the antibody or antigen-binding fragment thereof of the invention is derived.
  • the second antibody is an anti-immunoglobulin (eg, human immunoglobulin) antibody, eg, an anti-IgG antibody. In some embodiments, the second antibody is an anti-human IgG antibody.
  • the method can be used for diagnostic purposes, for example, whether a subject is infected with B-coronavirus can be diagnosed based on the presence or level of B-coronavirus in a sample.
  • the sample can be a blood sample (eg, whole blood, plasma, or serum), feces, oral or nasal secretions, or alveolar lavage fluid from the subject.
  • the subject is a mammal, such as a human.
  • the method may be used for non-diagnostic purposes, eg, the sample is not from a subject, eg, a vaccine sample.
  • the beta coronavirus includes novel coronavirus (SARS-CoV-2) and variants thereof, severe acute respiratory syndrome coronavirus (SARS-CoV) and variants thereof, SARS-related coronaviruses (SARSr-CoV).
  • SARS-CoV-2 novel coronavirus
  • SARS-CoV severe acute respiratory syndrome coronavirus
  • SARSr-CoV SARS-related coronaviruses
  • the SARS-CoV-2 includes variant strains.
  • the S protein of the mutant strain contains mutations, such as amino acid substitutions, deletions or additions.
  • the S protein of the mutant strain comprises one or more amino acid substitutions selected from K417N, E484K, N501Y, L452R, and T478K.
  • the mutant strain of SARS-CoV-2 is selected from Alpha (Alpha, B.1.1.7), Beta (Beta, B.1.351), Gamma (Gamma, P.1), Delta ( Delta, B.1.617.2), Omicron (B.1.1.529) or any combination thereof.
  • the method is an immunological assay, such as an enzyme immunoassay (eg, ELISA), a chemiluminescent immunoassay, a fluorescent immunoassay, or a radioimmunoassay.
  • an enzyme immunoassay eg, ELISA
  • chemiluminescent immunoassay e.g. a chemiluminescent immunoassay
  • fluorescent immunoassay e.g., a fluorescent immunoassay
  • radioimmunoassay e.g., radioimmunoassay.
  • the kit detects B-coronavirus or its S protein or the RBD of S protein, or the presence or level of cells infected with B-coronavirus in the sample by the detection method as described above , and optionally diagnosing whether the subject is infected with B-coronavirus according to the test result.
  • the sample is a blood sample (e.g., whole blood, plasma or serum), feces, oral or nasal secretions, or alveolar lavage fluid from a subject (e.g., a mammal, preferably a human) .
  • a blood sample e.g., whole blood, plasma or serum
  • feces e.g., feces, oral or nasal secretions
  • alveolar lavage fluid from a subject (e.g., a mammal, preferably a human) .
  • the beta coronavirus includes novel coronavirus (SARS-CoV-2) and variants thereof, severe acute respiratory syndrome coronavirus (SARS-CoV) and variants thereof, SARS-related coronaviruses (SARSr-CoV).
  • SARS-CoV-2 novel coronavirus
  • SARS-CoV severe acute respiratory syndrome coronavirus
  • SARSr-CoV SARS-related coronaviruses
  • the SARS-CoV-2 includes variant strains.
  • the S protein of the mutant strain contains mutations, such as amino acid substitutions, deletions or additions.
  • the S protein of the mutant strain comprises one or more amino acid substitutions selected from K417N, E484K, N501Y, L452R, and T478K.
  • the mutant strain of SARS-CoV-2 is selected from Alpha (Alpha, B.1.1.7), Beta (Beta, B.1.351), Gamma (Gamma, P.1), Delta ( Delta, B.1.617.2), Omicron (B.1.1.529) or any combination thereof.
  • antibodies or antigen-binding fragments thereof should be understood as Can be more than one, such as 2 types, 3 types, 4 types, 5 types, 6 types, 7 types, 8 types, 9 types, 10 types, 11 types, 12 types, 13 types, 14 types, 15 types, 16 kinds, 17 kinds, 18 kinds, 19 kinds, 20 kinds, 21 kinds of combinations.
  • Antibodies or antigen-binding fragments thereof of the present invention may also be combined with other antibodies or antigen-binding fragments thereof.
  • the antibody or antigen-binding fragment thereof may be a combination of two specific antibodies or antigen-binding fragments thereof, such as a combination of BD55-5840 and BD55-5514. Moreover, the combination of antibodies or antigen-binding fragments thereof has more excellent binding activity and neutralizing effect on B-coronavirus.
  • the molecular biology experiment methods and immunoassay methods used in the present invention are basically with reference to J.Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, 1989, and F.M.Ausubel et al., Compiled Molecular Biology Experimental Guide, 3rd Edition, John Wiley & Sons, Inc., 1995 by the method described; restriction endonucleases were used in accordance with the conditions recommended by the product manufacturer.
  • restriction endonucleases were used in accordance with the conditions recommended by the product manufacturer.
  • Enriched B cells were stained with the following human antigens and antibodies in FACS buffer (1 ⁇ PBS, 2% FBS, 1 mM EDTA): FITC anti-CD19 antibody (Biolegend), FITC anti-CD20 antibody (Biolegend), Brilliant Violet 421 anti-CD27 Antibody (Biolegend), PE/Cyanine7 anti-IgM and fluorophore-labeled RBD (SARS340 CoV-2 and SARS-CoV RBD, Sino Biological Inc.) and ovalbumin (Ova) were placed on ice for 30 minutes.
  • FACS buffer 1 ⁇ PBS, 2% FBS, 1 mM EDTA
  • FITC anti-CD19 antibody Biolegend
  • FITC anti-CD20 antibody Biolegend
  • Brilliant Violet 421 anti-CD27 Antibody Biolegend
  • PE/Cyanine7 anti-IgM and fluorophore-labeled RBD SARS340 CoV-2 and SARS-CoV RBD, Sino Biological Inc.
  • Raw FASTQ files were processed with reference to the human sequence GRCh38 database using Cell Ranger (version 6.1.1) software. Generate sequences using 'cellranger multi' or 'cellranger vdj' with default parameters. Protein sequences were then extracted and processed by IMGT/DomainGapAlign (version 4.10.2) to obtain annotations of V(D)J, CDR regions and mutation frequency.
  • V gene amino acid mutation rate mutation count/length of V gene peptide.
  • Paired immunoglobulin heavy and light chain genes obtained from 10X Genomics V(D)J sequencing and analysis were submitted for recombinant monoclonal antibody synthesis.
  • the heavy and light chain sequences were cloned separately into expression vectors based on Gibson assembly, and the two plasmids were subsequently co-transfected into HEK293F cells.
  • the secreted monoclonal antibody in the cell culture medium is then purified by Protein A affinity chromatography.
  • the specific binding capacity of these antibodies or plasma was analyzed by ELISA binding determination. Briefly, after coating, blocking and washing the RBDs of 22 currently known sarbecovirus viruses (see Table 2) with 0.03 ⁇ g/mL and 1 ⁇ g/mL, 1 ⁇ g/mL antibody or serially diluted plasma samples were added to the ELISA plate.
  • the 96-well plate was incubated with diluted goat anti-human IgG 373(H+L)/HRP antibody (JACKSON). Then tetramethylbenzidine (TMB, Solarbio) was added for color development for 10 minutes, and 2M H2SO4 was added to stop the development reaction, and the absorbance was measured at 450 nm using a PerkinElmer Ensight HH3400 model microplate reader, and the EC50 was calculated by GraphPad Prism 8.0 software ( See Table 3).
  • TMB tetramethylbenzidine
  • the protein analysis system (Fortebio) was used for the biolayer interference analysis kinetic experiment, and the sensor used was protein A biosensor (Fortebio 18-5010).
  • the sensors were first soaked in buffer for 10 minutes (buffer ForteBio 18-1105) to complete the sensor self-test step, followed by 30 seconds in the buffer to complete the baseline.
  • the antibody capture step the sensor was immersed in antibody at a concentration of 2 ⁇ g/ml for 300 s, and the threshold was set at 0.4 nm.
  • the sensor was then soaked in buffer for 120 seconds only to complete the baseline.
  • the sensors were soaked for 60 s in serial dilutions of the RBD protein.
  • the senor was switched to soak in buffer for 600 s. Finally, soak the sensor in the regeneration buffer (10 mM glycine hydrochloride, pH 1.5) for 30 seconds, then soak in the buffer for 30 seconds, and repeat 2 times to complete the regeneration step.
  • Neutralization assays were performed using the pseudovirus method to screen broadly binding antibodies to assess the neutralizing capacity of antibodies and plasma.
  • a series of two-fold dilutions of antibodies were incubated with seven VsV pseudoviruses of the sarbecovirus spike protein (labeled with luciferase) for 1 hr, and the mixture was incubated with Huh-7 cells. After culturing in a 37 °C incubator for 24 h, the cells were collected and lysed with a luciferase substrate (PerkinElmer), and then the luminescence intensity was measured by a microplate reader. IC50 was determined by a four-parameter nonlinear regression model (see Table 5).
  • Embodiment 6 Mouse challenge experiment--SARS-CoV-2 (Omicron BA.1) virus
  • Human ACE2 (hACE2) transgenic mice were divided into five groups with 5 mice in each group.
  • SARS-CoV-2 (Omicron BA.1) virus was used to infect mice with a dose of 1*10 5 TCID 50 per mouse via intranasal route, and the prevention group was given intraperitoneal or intramuscular injection of drugs 24 hours before challenge, with a dose of 20 mg/ kg.
  • the treatment group was given intraperitoneal injection of drugs 2 hours after challenge, the dose was 20 mg/kg.
  • the control group was intraperitoneally injected with an equal volume of PBS 2 hours after challenge.
  • the body weight of the mice was monitored at 0, 1, 2, and 3 days after the challenge, and the analysis of the body weight change curve (Fig. 2) showed that the body weight of the mice in each group decreased slightly on the 1 day after the challenge, and the weight of the mice in each group decreased slightly on the 2-3 days after the challenge. The body weight showed a rising trend, and all returned to the original body weight on the third day. There was no statistical difference in the weight change of mice among the experimental groups compared with the control group, which may be due to the low pathogenicity of BA.1 to hACE2 transgenic mice.
  • RNA viral ribonucleic acid
  • the viral load of the lung tissue samples in the muscle prevention group was low, and the statistical results showed that it was significantly different from that of the control group (P ⁇ 0.0001), and almost no viral RNA was detected in the trachea samples of the muscle prevention group.
  • the viral load in the lung tissue and trachea samples of the control drug DXP-593 treatment group was reduced individually, but there was no statistical difference from the control group as a whole.
  • the above experimental results showed that, compared with the control group, the treatment group, the abdominal cavity prevention group and the muscle prevention group could effectively reduce the viral load in the lung tissue and trachea of mice.
  • Embodiment 7 Mouse challenge experiment--SARS-CoV-2 (Omicron BA.5) virus
  • Human ACE2 (hACE2) transgenic mice were divided into four groups with 5 mice in each group.
  • the SARS-CoV-2 (Omicron BA.5) virus was used to infect mice with a dose of 1*10 5 TCID 50 /mouse via intranasal route, and the prevention group was injected intraperitoneally or intramuscularly with a dose of 20mg/ kg.
  • the treatment group was given intraperitoneal injection of drugs 2 hours after challenge, the dose was 20 mg/kg.
  • the control group was intraperitoneally injected with an equal volume of PBS 2 hours after challenge.
  • RNA viral ribonucleic acid
  • the viral load in the lung tissue and trachea of the abdominal cavity treatment group decreased by an average of 2.21 and 1.11 lg, and there was also a significant difference between the two groups (P ⁇ 0.05).
  • the average viral load in the lung tissue of the muscle prevention group decreased by 2.89 lg compared with the control group (P ⁇ 0.01), while the viral load in the trachea of the muscle prevention group decreased individually, but there was no statistical difference between the group and the control group.
  • the above experimental results show that, compared with the control group, both the intraperitoneal prevention group and the intraperitoneal treatment group can effectively reduce the viral load in the lung tissue and trachea of mice, while the muscle prevention group has a certain effect on reducing the viral load in the lung tissue .
  • the binding epitope of BD55-5514 is similar to that of ADG20 (Adintrevimab).
  • the sites on BA.1 RBD that participate in the binding of BD55-5514 mainly include Asp405, Asn437, Asn439, Gly502, Val503, Gly504, Tyr508, etc., and the sites such as Pro373-Thr376, Val407-Arg408, Pro499, and Tyr501 are also close to the binding interface.
  • the neutralization of BD55-5514 was not affected by Thr376Ala, Asp405Asn, Arg408Ser contained in BA.2, but was only affected by the mutations of Val503 and Gly504. These two sites are highly conserved and hardly occur in nature.
  • the binding epitope of BD55-5840 on RBD is similar to that of Ser309 (Sotrovimab), but the key binding site is different.
  • the RBD residues appearing on the interface include Asp339, Glu340, Thr345, Arg346, Lys440, Leu441, Ser443, Lys444, etc.
  • the mutations that mainly affect their binding are concentrated on Asp339-Glu340, Thr345-Arg346.

Landscapes

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

Abstract

La présente invention relève du domaine de l'immunologie, et concerne en particulier un anticorps neutralisant à large spectre de betacoronavirus et son utilisation. Plus particulièrement, la présente invention concerne un anticorps neutralisant à large spectre de betacoronavirus ou un fragment de liaison à l'antigène de celui-ci, et une utilisation de l'anticorps ou du fragment pour le diagnostic, la prévention et le traitement de diverses maladies provoquées par une infection à betacoronavirus.
PCT/CN2022/126628 2022-02-14 2022-10-21 Anticorps neutralisant à large spectre de betacoronavirus et son utilisation WO2023151312A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280091594.4A CN118679181A (zh) 2022-02-14 2022-10-21 乙型冠状病毒广谱中和抗体及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210131235.9A CN116621972A (zh) 2022-02-14 2022-02-14 乙型冠状病毒广谱中和抗体及其应用
CN202210131235.9 2022-02-14

Publications (1)

Publication Number Publication Date
WO2023151312A1 true WO2023151312A1 (fr) 2023-08-17

Family

ID=87563559

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/126628 WO2023151312A1 (fr) 2022-02-14 2022-10-21 Anticorps neutralisant à large spectre de betacoronavirus et son utilisation

Country Status (3)

Country Link
CN (2) CN116621972A (fr)
TW (1) TW202342513A (fr)
WO (1) WO2023151312A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024131774A1 (fr) * 2022-12-20 2024-06-27 北京昌平实验室 Anticorps neutralisants à large spectre dirigés contre une nouvelle souche mutante de coronavirus omicron et utilisation
WO2024131775A1 (fr) * 2022-12-20 2024-06-27 北京昌平实验室 Anticorps neutralisant à large spectre dirigé contre le nouveau coronavirus xbb.1.5, variants associés et utilisation correspondante

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111620946A (zh) * 2020-05-09 2020-09-04 江苏省疾病预防控制中心(江苏省公共卫生研究院) 分离的新型冠状病毒单克隆抗体或其抗原结合部分
CN111978395A (zh) * 2020-07-20 2020-11-24 四川大学 抗新型冠状病毒rbd结构域抗原的单克隆抗体
CN111995674A (zh) * 2020-09-03 2020-11-27 中国人民解放军军事科学院军事医学研究院 抗COVID-19病毒中和抗体mhC3及其人源化抗体与应用
CN112625125A (zh) * 2021-01-18 2021-04-09 中国人民解放军军事科学院军事医学研究院 一株中和新型冠状病毒感染的单抗
CN113264998A (zh) * 2021-01-28 2021-08-17 四川大学华西医院 抗新冠病毒SARS-CoV-2表面S1蛋白的单链抗体及其应用
CN113388029A (zh) * 2020-03-12 2021-09-14 中国科学院武汉病毒研究所 针对新型冠状病毒的中和性人源单克隆抗体及其应用
CN113480644A (zh) * 2020-08-10 2021-10-08 南开大学 一种抗冠状病毒的抗体及其应用
CN113683687A (zh) * 2020-05-19 2021-11-23 益科思特(北京)医药科技发展有限公司 抗新型冠状病毒Spike蛋白抗体及其应用
CN113943368A (zh) * 2021-10-15 2022-01-18 中国科学院微生物研究所 一种新型冠状病毒及其突变体的单克隆抗体及其应用

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113388029A (zh) * 2020-03-12 2021-09-14 中国科学院武汉病毒研究所 针对新型冠状病毒的中和性人源单克隆抗体及其应用
CN111620946A (zh) * 2020-05-09 2020-09-04 江苏省疾病预防控制中心(江苏省公共卫生研究院) 分离的新型冠状病毒单克隆抗体或其抗原结合部分
CN113683687A (zh) * 2020-05-19 2021-11-23 益科思特(北京)医药科技发展有限公司 抗新型冠状病毒Spike蛋白抗体及其应用
CN111978395A (zh) * 2020-07-20 2020-11-24 四川大学 抗新型冠状病毒rbd结构域抗原的单克隆抗体
CN113480644A (zh) * 2020-08-10 2021-10-08 南开大学 一种抗冠状病毒的抗体及其应用
CN111995674A (zh) * 2020-09-03 2020-11-27 中国人民解放军军事科学院军事医学研究院 抗COVID-19病毒中和抗体mhC3及其人源化抗体与应用
CN112625125A (zh) * 2021-01-18 2021-04-09 中国人民解放军军事科学院军事医学研究院 一株中和新型冠状病毒感染的单抗
CN113264998A (zh) * 2021-01-28 2021-08-17 四川大学华西医院 抗新冠病毒SARS-CoV-2表面S1蛋白的单链抗体及其应用
CN113943368A (zh) * 2021-10-15 2022-01-18 中国科学院微生物研究所 一种新型冠状病毒及其突变体的单克隆抗体及其应用

Also Published As

Publication number Publication date
CN118679181A (zh) 2024-09-20
CN116621972A (zh) 2023-08-22
TW202342513A (zh) 2023-11-01

Similar Documents

Publication Publication Date Title
CN111690059B (zh) 一种抗SARS-CoV-2的单克隆抗体1D7
CN111718411B (zh) 一种抗SARS-CoV-2的单克隆抗体1F2
WO2023151312A1 (fr) Anticorps neutralisant à large spectre de betacoronavirus et son utilisation
JP5833565B2 (ja) ヒトサイトメガロウイルスに対する結合メンバー
EP3105250B1 (fr) Molécules de liaison multispécifiques iga
US7879326B2 (en) Human neutralizing monoclonal antibodies to H5N1 influenza A virus
CN111333723B (zh) 针对狂犬病病毒g蛋白的单克隆抗体及其用途
CN106243218B (zh) 抗Flu B的广谱单克隆抗体及其用途
TWI785583B (zh) 一種抗新型冠狀病毒的單克隆抗體及其應用
CN107674123B (zh) 一种抗独特型抗体及其应用
EP4206224A1 (fr) Anticorps humain ou fragment de liaison à l'antigène de celui-ci dirigé contre une protéine de spicule de coronavirus
CN118388641A (zh) 针对SARS-CoV-2的抗体及其用途
TW202144405A (zh) 抗新型冠狀病毒(SARS-CoV-2)之新穎單株抗體及其應用
CN113549147B (zh) 抗柯萨奇a6型病毒的单克隆抗体及其应用
WO2023005805A1 (fr) Polypeptide épitope d'affinité générale pour rhinovirus humain, anticorps et leurs utilisations
WO2022148374A1 (fr) Anticorps 76e1 neutralisant entièrement humain à large spectre contre le coronavirus, et utilisation associée
US20230399385A1 (en) Neutralizing antibodies against sars-cov-2
WO2024131774A9 (fr) Anticorps neutralisants à large spectre dirigés contre une nouvelle souche mutante de coronavirus omicron et utilisation
WO2024131775A9 (fr) Anticorps neutralisant à large spectre dirigé contre le nouveau coronavirus xbb.1.5, variants associés et utilisation correspondante
CN118221808A (zh) 新型冠状病毒奥密克戎突变株广谱中和抗体及应用
US20220340644A1 (en) Influenza neutralizing antibodies and their uses
WO2022095996A1 (fr) Anticorps anti-sars-cov-2 et son utilisation
CN116693669A (zh) 针对冠状病毒的抗体及其用途
CN118406140A (zh) 针对SARS-CoV-2及其突变株的广谱单克隆抗体及其用途
CN113549146A (zh) 针对柯萨奇病毒b1型的单克隆抗体及其用途

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: 22925665

Country of ref document: EP

Kind code of ref document: A1