CN116063464A - Antibodies or antigen binding fragments thereof to coronaviruses - Google Patents

Antibodies or antigen binding fragments thereof to coronaviruses Download PDF

Info

Publication number
CN116063464A
CN116063464A CN202210841040.3A CN202210841040A CN116063464A CN 116063464 A CN116063464 A CN 116063464A CN 202210841040 A CN202210841040 A CN 202210841040A CN 116063464 A CN116063464 A CN 116063464A
Authority
CN
China
Prior art keywords
antibody
sequence
seq
binding fragment
antigen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210841040.3A
Other languages
Chinese (zh)
Inventor
黄竞荷
吴凡
刘梅
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Super Extraordinary Shanghai Medical Technology Co ltd
Original Assignee
SHANGHAI PUBLIC HEALTH CLINICAL CENTER
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 SHANGHAI PUBLIC HEALTH CLINICAL CENTER filed Critical SHANGHAI PUBLIC HEALTH CLINICAL CENTER
Priority to CN202210841040.3A priority Critical patent/CN116063464A/en
Publication of CN116063464A publication Critical patent/CN116063464A/en
Pending legal-status Critical Current

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
    • 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
    • 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
    • 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
    • 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
    • C07K2317/56Immunoglobulins 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2469/00Immunoassays for the detection of microorganisms
    • G01N2469/10Detection of antigens from microorganism in sample from host

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

Abstract

The invention relates to an antibody or antigen binding fragment thereof of coronavirus, a nucleic acid molecule encoding the antibody or antigen binding fragment thereof, a vector comprising the nucleic acid molecule, a host cell comprising the vector, and the use of the antibody or antigen binding fragment thereof for preparing a medicament for treating or preventing diseases caused by coronavirus, and for detecting products; the inventor obtains a series of antibodies and antigen binding fragments thereof of coronaviruses by utilizing B cell in-vitro monoclonal culture and high-flux antibody screening technology, the antibodies and antigen binding fragments have strong binding capacity and neutralization capacity for SARS-CoV-2 viruses, and can recognize and bind S1 protein of SARS-CoV-2 viruses and RBD thereof, and have very strong affinity, so that the antibodies and antigen binding fragments can be presumed to have binding capacity and neutralization capacity for other coronaviruses and coronaviruses possibly appearing in the future, and have good clinical application prospect in the future.

Description

Antibodies or antigen binding fragments thereof to coronaviruses
Technical Field
The invention relates to an antibody or antigen binding fragment thereof of coronavirus, a nucleic acid molecule encoding the antibody or antigen binding fragment thereof, a vector containing the nucleic acid molecule, a host cell containing the vector, application of the antibody or antigen binding fragment thereof in preparing a medicament for treating or preventing diseases caused by coronavirus, and application in detecting products, and belongs to the field of biological medicine.
Background
Novel coronavirus pneumonia (2019-nCOV) is an acute respiratory infectious disease caused by the SARS-COV-2 novel coronavirus.
SARS-CoV-2 virus belongs to coronaviridae, and has homology of 77.2% with SARS coronavirus in the same genus as the beta coronavirus in the fulminant of 2003. The major envelope protein of SARS-CoV-2 virus is its Spike protein (also called Spike protein, abbreviated as S protein) which is hydrolyzed into two parts S1 and S2 by intracellular proteases during viral infection. Where S2 is a transmembrane protein and S1 has a receptor binding domain (Receptor Binding domain, abbreviated RBD) that recognizes and binds the cellular receptor angiotensin converting enzyme-2 (ACE-2). The spike protein formed by S1 and S2 is the virus receptor which specifically recognizes and binds to the target cell receptor of SARS-CoV-2 virus and mediates virus infection, and is also the recognition target of the neutralizing antibody to be developed.
The research shows that the virus specific recovered human blood plasma is clinically used, can effectively neutralize the virus, prevent the virus from diffusing in various organs in the body, and plays an important role in the prognosis of the disease course of patients. However, polyclonal plasma is not only of limited origin, but its clinical use is also limited by conditions such as difficulty in quality control, differential blood group supply to the recipient, potential infectious agents, and the like. The fully human monoclonal antibody capable of neutralizing SARS-CoV-2 virus is separated from the new coronavirus rehabilitation person, which can effectively overcome the above problems and is one of the main directions of the current new coronavirus drug development.
Up to now, several research teams at home and abroad report that fully human monoclonal antibodies capable of combining SARS-CoV-2 virus S protein, such as BD-368-2, B38, etc., are isolated from peripheral blood of new coronal pneumonia healers, and are still in the experimental research and development stage at present. The technical method adopted by the research teams is to utilize S protein or S protein Receptor Binding Domain (RBD) of recombinant expressed SARS-CoV-2 virus as bait, screen and separate B cells (memory B cells) capable of binding the proteins from peripheral blood of a rehabilitee, obtain heavy chain and light chain pairing genes of antibodies expressed by single B cells by using a cell sequencing or single cell sequencing method, express the antibodies in an in vitro recombination mode, and then verify the capability of neutralizing the viruses. Since the method uses a marker protein (the S protein or S protein receptor binding region of the above-described recombinant expressed SARS-CoV-2 virus called bait) to screen and enrich B cells in advance before performing antibody gene sequencing, only antibodies specifically binding to the marker protein can be screened.
Huang Jinghe doctor (one of the inventors of the present application) initiated in 2013 in vitro monoclonal culture and high throughput antibody screening technology (Huang J et al nature Protocols 2013) of human B cells, isolated fully human monoclonal antibodies from peripheral blood of new coronatine rehabilitation, by the following procedure: firstly, detecting neutralizing antibodies of serum of a new coronapneumonia rehabilitation person by utilizing a SARS-CoV-2 and SARS-CoV pseudovirus neutralizing system, and screening rehabilitation persons with high neutralizing activity on the SARS-CoV-2 and the SARS-CoV simultaneously; then collecting peripheral blood lymphocytes of a rehabilitee, and sorting out memory B lymphocytes by using flow cells; single B cells were seeded in 384 well plates and cultured with the addition of cytokines and feeder cells, and the cultured B cells secreted antibodies into the supernatant after expansion and differentiation in vitro. Then, the neutralizing capacity of the antibody in the supernatant to SARS-CoV-2 and SARS-CoV viruses is detected by utilizing an in-vitro high-flux neutralizing experiment, positive clones which can neutralize the two viruses simultaneously are screened out, the heavy chain and light chain variable regions of the antibody are cloned by utilizing an RT-PCR method, and the monoclonal antibody is obtained by constructing the heavy chain and light chain expression vectors of the antibody and then transfecting 293T cells to express and purify the monoclonal antibody.
The antibodies reported by other groups at present have better neutralizing capacity on the tested SARS-CoV-2 virus strain, but because the SARS-CoV-2 virus is RNA virus, the genome sequence of the virus is easy to mutate in the process of spreading epidemic. When mutations occur in the non-conserved region sites recognized by these antibodies, new epidemic strains are generated, resulting in the antibodies losing their protective effect against the mutant virus.
Thus, it would still be desirable for those skilled in the art to develop new antibodies that have binding and neutralizing capabilities for coronaviruses, including the SARS-CoV-2 virus.
Disclosure of Invention
To solve the above technical problems, in one aspect, the present invention provides an antibody of coronavirus, or an antigen-binding fragment thereof, comprising a heavy chain variable region comprising three heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3 and a light chain variable region comprising three light chain complementarity determining regions LCDR1, LCDR2 and LCDR3; wherein:
the sequence general formula of the HCDR1 is as follows: GX 1 TVSSNY, where X 1 Is L, I or F;
the sequence general formula of the HCDR2 is as follows: x is X 2 YSGGSX 3 Wherein X is 2 Is any one amino acid of L or I, X 3 Is an amino acid of either A or T.
Preferably, the HCDR3 has the sequence formula: ARDLIX 4 YGMDV, wherein X 4 An amino acid which is either D or T;
the sequence of the LCDR1 is QGISSY, and the sequence of the LCDR2 is AAS;
the sequence general formula of the LCDR3 is as follows: QQLNSYPPX 5 T, where X 5 Is either L or Y.
In a preferred embodiment of the present invention, the sequence of the HCDR1 is shown as SEQ ID NO.1, the sequence of the HCDR2 is shown as SEQ ID NO.2, and the sequence of the HCDR3 is shown as SEQ ID NO. 3; the sequence of the LCDR1 is shown as SEQ ID NO.5, the sequence of the LCDR2 is shown as SEQ ID NO.6, and the sequence of the LCDR3 is shown as SEQ ID NO. 7; or the other one of the above-mentioned materials,
the sequence of the HCDR1 is shown as SEQ ID NO.11, the sequence of the HCDR2 is shown as SEQ ID NO.12, and the sequence of the HCDR3 is shown as SEQ ID NO. 13; and the sequence of LCDR1 is shown as SEQ ID NO.15, the sequence of LCDR2 is shown as SEQ ID NO.16, and the sequence of LCDR3 is shown as SEQ ID NO. 17.
In another preferred embodiment of the present invention, the heavy chain variable region has a sequence as shown in SEQ ID NO.4 or a sequence having 80% or more sequence homology with the sequence shown in SEQ ID NO.4, and the light chain variable region has a sequence as shown in SEQ ID NO.8 or a sequence having 80% or more sequence homology with the sequence shown in SEQ ID NO. 8; or alternatively, the process may be performed,
the heavy chain variable region has a sequence shown as SEQ ID NO.14 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO.14, and the light chain variable region has a sequence shown as SEQ ID NO.18 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO. 18.
In a preferred embodiment of the present invention, the sequence of the HCDR1 is shown as SEQ ID NO.21, the sequence of the HCDR2 is shown as SEQ ID NO.22, and the sequence of the HCDR3 is shown as SEQ ID NO. 23; the sequence of LCDR1 is shown as SEQ ID NO.25, the sequence of LCDR2 is shown as SEQ ID NO.26, and the sequence of LCDR3 is shown as SEQ ID NO. 27; or alternatively, the process may be performed,
the sequence of the HCDR1 is shown as SEQ ID NO.31, the sequence of the HCDR2 is shown as SEQ ID NO.32, and the sequence of the HCDR3 is shown as SEQ ID NO. 33; the sequence of the LCDR1 is shown as SEQ ID NO.35, the sequence of the LCDR2 is shown as SEQ ID NO.36, and the sequence of the LCDR3 is shown as SEQ ID NO. 37; or alternatively, the process may be performed,
the sequence of the HCDR1 is shown as SEQ ID NO.41, the sequence of the HCDR2 is shown as SEQ ID NO.42, and the sequence of the HCDR3 is shown as SEQ ID NO. 43; the sequence of LCDR1 is shown as SEQ ID NO.45, the sequence of LCDR2 is shown as SEQ ID NO.46, and the sequence of LCDR3 is shown as SEQ ID NO. 47; or alternatively, the process may be performed,
the sequence of the HCDR1 is shown as SEQ ID NO.51, the sequence of the HCDR2 is shown as SEQ ID NO.52, and the sequence of the HCDR3 is shown as SEQ ID NO. 53; the sequence of the LCDR1 is shown as SEQ ID NO.55, the sequence of the LCDR2 is shown as SEQ ID NO.56, and the sequence of the LCDR3 is shown as SEQ ID NO. 57; or alternatively, the process may be performed,
the sequence of the HCDR1 is shown as SEQ ID NO.61, the sequence of the HCDR2 is shown as SEQ ID NO.62, and the sequence of the HCDR3 is shown as SEQ ID NO. 63; and the sequence of LCDR1 is shown as SEQ ID NO.65, the sequence of LCDR2 is shown as SEQ ID NO.66, and the sequence of LCDR3 is shown as SEQ ID NO. 67.
In another preferred embodiment of the present invention, the heavy chain variable region has a sequence as shown in SEQ ID NO.24 or a sequence having 80% or more sequence homology with the sequence shown in SEQ ID NO.24, and the light chain variable region has a sequence as shown in SEQ ID NO.28 or a sequence having 80% or more sequence homology with the sequence shown in SEQ ID NO. 28; or alternatively, the process may be performed,
the heavy chain variable region has a sequence shown as SEQ ID NO.34 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO.34, and the light chain variable region has a sequence shown as SEQ ID NO.38 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO. 38; or alternatively, the process may be performed,
the heavy chain variable region has a sequence shown as SEQ ID NO.44 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO.44, and the light chain variable region has a sequence shown as SEQ ID NO.48 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO. 48; or alternatively, the process may be performed,
the heavy chain variable region has a sequence shown as SEQ ID NO.54 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO.54, and the light chain variable region has a sequence shown as SEQ ID NO.58 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO. 58; or alternatively, the process may be performed,
the heavy chain variable region has a sequence shown as SEQ ID NO.64 or a sequence with more than 80% sequence homology with the sequence shown as SEQ ID NO.64, and the light chain variable region has a sequence shown as SEQ ID NO.68 or a sequence with more than 80% sequence homology with the sequence shown as SEQ ID NO. 68.
With respect to the percentage of "sequence homology," the percentage of sequence identity is generated by determining the number of amino acid residues present in two sequences to produce the number of matched positions, dividing the number of matched positions by the total number of positions in the comparison window, and multiplying the structure by 100.
In a specific embodiment of the invention, the heavy chain variable region may be increased or decreased in amino acid sequence based on the first amino acid sequence or the light chain variable region may be increased or decreased in amino acid sequence based on the second amino acid sequence, such as a similar amino acid substitution or a small amino acid substitution, particularly an amino acid increase or decrease in the conserved sequence portion, resulting in variants of the antibody having higher homology (80% homology or more) and retaining the original antibody function, i.e., the function and properties of specifically binding to coronaviruses, which variants are also within the scope of the invention.
In a preferred embodiment of the invention, the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown in SEQ ID NO.9, and the light chain amino acid sequence is shown in SEQ ID NO. 10; or the other one of the above-mentioned materials,
the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown as SEQ ID NO.19, and the light chain amino acid sequence is shown as SEQ ID NO. 20; or alternatively, the process may be performed,
the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown as SEQ ID NO.29, and the light chain amino acid sequence is shown as SEQ ID NO. 30; or alternatively, the process may be performed,
the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown as SEQ ID NO.39, and the light chain amino acid sequence is shown as SEQ ID NO. 40; or alternatively, the process may be performed,
the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown as SEQ ID NO.49, and the light chain amino acid sequence is shown as SEQ ID NO. 50; or alternatively, the process may be performed,
the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown as SEQ ID NO.59, and the light chain amino acid sequence is shown as SEQ ID NO. 60; or alternatively, the process may be performed,
the heavy chain amino acid sequence of the antibody or antigen binding fragment thereof is shown as SEQ ID NO.69, and the light chain amino acid sequence is shown as SEQ ID NO. 70.
In a preferred embodiment of the invention, the antibody or antigen binding fragment thereof is a neutralizing antibody or antigen binding fragment thereof of a coronavirus.
The term "neutralizing antibody" is an antibody or antigen binding fragment that specifically binds to a viral receptor protein, which specifically binds to a protein that inhibits a biological function of the viral receptor protein, e.g., prevents the receptor protein from binding to its target cell receptor, which specifically reduces the ability of the virus to infect a target cell; in the present application, neutralizing antibody or antigen-binding fragment thereof of coronavirus refers to an antibody or antigen-binding fragment thereof that binds to the S protein of coronavirus.
The term "antibody" is used herein in its broadest sense to encompass natural and artificial antibodies of various structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), single chain antibodies, intact antibodies, and antibody fragments, antigen-binding proteins, fusion proteins, and the like, that exhibit the desired antigen-binding activity.
In a preferred embodiment of the invention, the antibody is a monoclonal antibody.
In a more preferred embodiment of the invention, the antibody is a fully human monoclonal antibody.
In a preferred embodiment of the invention, the Fc domain of the antibody is derived from an Fc domain of an immunoglobulin, including an Fc domain of a native sequence or a variant Fc domain.
In a preferred embodiment of the invention, the antibody is any one or a combination of several of IgG1, igG2, igG3 or IgG 4.
Preferably, the antibody may be an intact antibody selected from the group consisting of IgG1, igG2, igG3, or IgG 4.
In a preferred embodiment of the invention, the antigen binding fragment is Fv, fab, F (ab ') 2, fab', dsFv, scFv, sc (Fv) 2 or a single chain antibody.
In a preferred embodiment of the invention, the antibody or antigen-binding fragment thereof may be combined with other antibodies or antigen-binding fragments thereof to form an antibody or antigen-binding fragment thereof having at least two antigen-binding sites, i.e. a multispecific antibody or antigen-binding fragment thereof; the two antigen binding sites may be different epitopes of the same antigen or different epitopes of different antigens.
In a preferred embodiment of the invention, the above-described antibodies, or antigen binding fragments thereof, may be further chemically modified, e.g., one or more chemical groups may be attached to the antibody to increase one or more functional properties of the antibody. For example, common chemical modifications are glycosylation modifications, polyethylene glycol modifications, and the like. Among other things, glycosylation modifications can be made, for example, in the heavy or light chain variable regions, adding one or more glycosylation sites, to improve a portion of the function of an antibody, e.g., to enhance the immunogenicity of an antibody or to improve the pharmacokinetics of an antibody, etc. For example, the antibody or antigen-binding fragment thereof is subjected to an acylation reaction or an alkylation reaction with an active polyethylene glycol (e.g., an active ester or aldehyde derivative of polyethylene glycol) under suitable conditions to effect a pegylation modification to improve a partial function of the antibody, e.g., to increase the biological (e.g., serum) half-life of the antibody, etc. The above-described chemical modifications do not significantly alter the basic function and properties of the antibodies or antigen binding fragments thereof of the invention, i.e., the function and properties of binding specifically to coronaviruses; such chemically modified variants are also within the scope of the present invention.
In a preferred embodiment of the invention, the above-described antibodies or antigen-binding fragments thereof may be conjugated to other factors by chemical means or by genetic engineering means; for example, these factors may provide the effect of targeting the antibody to a desired functional site or other properties; for example, these factors may be one or more heterologous molecules, preferably, the heterologous molecules are cytotoxic agents. The above antibodies, or complexes formed by conjugation of antigen binding fragments thereof with other factors, fall within the scope of the present invention.
In another aspect, the invention provides a nucleic acid molecule, wherein the nucleic acid molecule encodes an antibody as described above, or an antigen binding fragment thereof.
In a preferred embodiment of the present invention, in the nucleic acid molecule, the nucleic acid sequence encoding the heavy chain variable region is shown as SEQ ID NO.71 and the nucleic acid sequence encoding the light chain variable region is shown as SEQ ID NO. 72; or alternatively, the process may be performed,
the nucleic acid sequence for encoding the heavy chain variable region is shown as SEQ ID NO.75, and the nucleic acid sequence for encoding the light chain variable region is shown as SEQ ID NO. 76; or alternatively, the process may be performed,
the nucleic acid sequence for encoding the heavy chain variable region is shown as SEQ ID NO.79, and the nucleic acid sequence for encoding the light chain variable region is shown as SEQ ID NO. 80; or alternatively, the process may be performed,
the nucleic acid sequence for encoding the heavy chain variable region is shown as SEQ ID NO.83, and the nucleic acid sequence for encoding the light chain variable region is shown as SEQ ID NO. 84; or alternatively, the process may be performed,
the nucleic acid sequence for encoding the heavy chain variable region is shown as SEQ ID NO.87, and the nucleic acid sequence for encoding the light chain variable region is shown as SEQ ID NO. 88; or alternatively, the process may be performed,
the nucleic acid sequence for encoding the heavy chain variable region is shown as SEQ ID NO.91, and the nucleic acid sequence for encoding the light chain variable region is shown as SEQ ID NO. 92; or alternatively, the process may be performed,
the nucleic acid sequence for encoding the heavy chain variable region is shown as SEQ ID NO.95, and the nucleic acid sequence for encoding the light chain variable region is shown as SEQ ID NO. 96.
In a more preferred embodiment of the invention, in the nucleic acid molecule,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.73, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 74; or alternatively, the process may be performed,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.77, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 78; or alternatively, the process may be performed,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.81, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 82; or alternatively, the process may be performed,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.85, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 86; or alternatively, the process may be performed,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.89, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 90; or alternatively, the process may be performed,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.93, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 94; or alternatively, the process may be performed,
the nucleic acid sequence for coding the heavy chain is shown as SEQ ID NO.97, and the nucleic acid sequence for coding the light chain is shown as SEQ ID NO. 98.
In a further aspect the invention provides a vector comprising a nucleic acid molecule as described above.
In a preferred embodiment of the invention, the vector further comprises an expression control sequence linked to the nucleic acid molecule described above.
The term "vector" refers to a nucleic acid vector into which a polynucleotide encoding a protein can be inserted and the protein expressed. The vector may be transformed, transduced or transfected into a host cell to allow expression of the genetic material elements carried thereby within the host cell. The vector may contain various elements for controlling expression, such as a promoter sequence, a transcription initiation sequence, an enhancer sequence, a selection element, a reporter gene, and the like. In addition, the vector may also contain a replication origin. It is also possible for the vector to include components that assist it in entering the cell, such as viral particles, liposomes or protein shells, but not just these. In embodiments of the present invention, the carrier may be selected from, but is not limited to: plasmids, phagemids, ke Sizhi, artificial chromosomes (e.g., yeast artificial chromosome YAC, bacterial artificial chromosome BAC, or P1-derived artificial chromosome PAC), phages (e.g., lambda phage or M13 phage), and animal viruses used as vectors, e.g., retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, herpesviruses (e.g., herpes simplex viruses), poxviruses, baculoviruses, papillomaviruses, papilloma viruses (e.g., SV 40).
In a further aspect the invention provides a host cell comprising the vector described above.
With respect to "host cells," one can choose, but is not limited to: prokaryotic cells such as Escherichia coli or Bacillus subtilis, fungal cells such as yeast cells or Aspergillus, insect cells such as S2 Drosophila cells or Sf9, or animal cell models such as fibroblasts, CHO cells, COS cells, NSO cells, heLa cells, BHK cells, HEK293 cells, etc.
Preferably, the host cell is a HEK293 cell.
In a further aspect, the invention provides a method of producing an antibody, or antigen-binding fragment thereof, as described above, wherein the host cell described above is cultured to produce the antibody, or antigen-binding fragment thereof.
In a further aspect, the invention provides a pharmaceutical composition comprising an antibody, or antigen-binding fragment thereof, as described above.
In a preferred embodiment of the invention, the pharmaceutical composition comprises a therapeutically effective amount of a neutralizing antibody, or antigen binding fragment thereof, and a pharmaceutically acceptable carrier or diluent. One skilled in the art can use a suitable pharmaceutical carrier or diluent in combination with a therapeutically effective amount of the neutralizing antibody, or antigen binding fragment thereof, for administration to a patient for the treatment or prevention of a disease caused by a coronavirus.
In a further aspect, the invention provides the use of an antibody, or antigen-binding fragment thereof, as described above, or a pharmaceutical composition as described above, in the manufacture of a medicament for the treatment or prophylaxis of a disease caused by a coronavirus.
In a preferred embodiment of the invention, the use refers to the use in the manufacture of a medicament for the treatment or prophylaxis of a disease caused by SARS-CoV-2, SARS-CoV or SARS-like coronavirus.
In one aspect, the invention also provides a method of treating or preventing a disease caused by a coronavirus by administering to a patient a therapeutically effective amount of an antibody, or antigen-binding fragment thereof, as described above; or administering to the patient a pharmaceutical composition comprising a therapeutically effective amount of an antibody, or antigen-binding fragment thereof, as described above. Preferably, the disease caused by coronavirus is a disease caused by SARS-CoV-2, SARS-CoV or SARS-like coronavirus.
In a further aspect the invention provides an assay product, wherein the assay product comprises an antibody as described above, or an antigen binding fragment thereof.
The detection product is used to detect the presence or level of coronavirus in a sample.
In one embodiment of the invention, the detection product includes, but is not limited to, a detection reagent, a detection kit, a detection chip or test paper, and the like.
The above-mentioned antibody or antigen-binding fragment thereof of the present invention may be labeled by a chemical method or a genetic engineering method, and the labeled antibody or antigen-binding fragment thereof may be used for detection; the labeled antibodies or antigen binding fragments thereof fall within the scope of the present invention.
The specific detection method can adopt the following steps, 1) providing a sample; 2) Contacting the sample with an antibody or antigen binding fragment thereof of the coronavirus of the invention described above; 3) An immune response between the sample and the antibody or antigen binding fragment thereof is detected.
The inventor obtains a series of antibodies and antigen binding fragments thereof of coronaviruses by utilizing B cell in-vitro monoclonal culture and high-flux antibody screening technology, the antibodies and antigen binding fragments thereof have strong binding capacity and neutralization capacity for SARS-CoV-2 viruses, and can recognize and bind S1 protein of SARS-CoV-2 viruses and RBD thereof, and have very strong affinity, so that the antibodies and antigen binding fragments thereof of a series of coronaviruses of the invention can be presumed to have binding capacity and neutralization capacity for other coronaviruses, and coronaviruses possibly occurring in the future, and have good clinical application prospect in the future.
Drawings
FIG. 1 shows the detection results of monoclonal antibody 4L12 recognizing the S1 protein and RBD thereof of SARS-CoV-2 virus, and the S2 protein;
FIG. 2 shows the detection results of the monoclonal antibody 12F5 recognizing the S1 protein and RBD thereof of SARS-CoV-2 virus, and the S2 protein;
FIG. 3 shows the detection results of monoclonal antibody 3D13 recognizing the S1 protein and RBD thereof, and the S2 protein of SARS-CoV-2 virus;
FIG. 4 shows the detection results of the monoclonal antibody 10C2 recognizing the S1 protein and RBD thereof of SARS-CoV-2 virus, and the S2 protein;
FIG. 5 shows the results of detection of the S1 protein and RBD thereof, and the S2 protein of the monoclonal antibody 16L9 recognizing SARS-CoV-2 virus;
FIG. 6 shows the detection results of the monoclonal antibody 20E21 recognizing the S1 protein and RBD thereof of SARS-CoV-2 virus, and the S2 protein;
FIG. 7 shows the detection results of the monoclonal antibody 22H22 recognizing the S1 protein and RBD thereof of SARS-CoV-2 virus, and the S2 protein;
FIG. 8 shows the results of affinity detection of RBD of monoclonal antibody 4L12 binding to S1 protein of SARS-CoV-2 virus;
FIG. 9 shows the results of affinity detection of RBD of monoclonal antibody 12F5 binding to S1 protein of SARS-CoV-2 virus;
FIG. 10 shows the results of affinity detection of RBD of monoclonal antibody 3D13 binding to S1 protein of SARS-CoV-2 virus;
FIG. 11 shows the results of affinity detection of RBD of monoclonal antibody 10C2 binding to S1 protein of SARS-CoV-2 virus;
FIG. 12 shows the results of affinity detection of RBD of monoclonal antibody 16L9 binding to S1 protein of SARS-CoV-2 virus;
FIG. 13 shows the results of affinity detection of RBD of monoclonal antibody 20E21 binding to S1 protein of SARS-CoV-2 virus;
FIG. 14 shows the results of affinity detection of RBD of monoclonal antibody 22H22 binding to S1 protein of SARS-CoV-2 virus.
Detailed Description
The present invention will be described in detail below with reference to specific embodiments shown in the drawings. However, these embodiments are not intended to limit the present invention, and structural, methodological, or functional modifications of these embodiments are intended to be included within the scope of the present invention.
Example embodiments will now be described more fully with reference to the accompanying drawings. However, the exemplary embodiments can be embodied in many forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the concept of the example embodiments to those skilled in the art.
Materials, reagents and the like used in the following examples are commercially available unless otherwise specified. The specific techniques or conditions are not noted in the examples, and are carried out according to techniques or conditions described in the literature in the art (for example, refer to J. Sam Brookfield et al, third edition, scientific Press, et al, translation of molecular cloning Experimental guidelines, huang Peitang, et al) or according to the product specifications.
Example 1: screening and detection of antibodies to coronaviruses
The inventor carries out pseudo-virus neutralization experimental screening on the blood plasma of novel coronavirus pneumonia patients (follow-up visit after two weeks of recovery discharge) collected at the unit of the inventor (Shanghai city public health clinical center) from the 1 st month of 2020 to the 2 nd month of 2020, finds that the blood serum of three light patients has strong neutralization activity on SARS-CoV-2 pseudo-virus, and extracts the peripheral blood for research through the written consent of the unit ethics committee of the inventor and the patient.
1. Sorting of peripheral blood memory B cells
1) Isolation of peripheral blood lymphocytes: peripheral blood lymphocytes of the patient in the recovery period are isolated by mixing the peripheral blood with an equal amount of physiological saline and using lymphocyte separation liquid Lymphoprep (Stemcell Technologies, cat# 07851), and the operation process is described in the specification of lymphocyte separation liquid.
2) Sorting peripheral blood memory B cells: staining the peripheral blood lymphocytes isolated in step 1) above with an antibody mixture at 4 ℃ and in the dark for 30min, wherein the antibody mixture is a mixture of anti-CD 19-PE-Cy7 (BD Bioscience), igA-APC (Jackson Immunoresearch), igD-FITC (BD Bioscience) and IgM-PE (Jackson Immunoresearch); after staining, washing with 10ml PBS-BSA buffer and re-suspending in 500. Mu.l PBS-BSA; finally, CD19+ IgA-IgD-IgM-memory B cells were sorted out using a FACSaria III cell sorter (Becton Dickinson).
2. Incubation of peripheral blood memory B cells
Resuspending the selected CD19+ IgA-IgD-IgM-memory B cells in medium containing 10% FBS and 100U/ml IL-2, 50ng/ml IL-21 and irradiated 3T3-msCD40L feeder cells; memory B cells were seeded at a density of 4 cells/well in 384 well microtiter plates (final volume 50 μl) and incubated for 13 days; the growth factors IL-2 and IL-21 stimulate memory B cells to divide and grow, and secrete antibodies into the culture medium after incubation. Specific culturing methods are described in reference Huang J et al Nature Protocols 2013,8 (10): 1907-15.
3. Production of SARS-CoV-2 and SARS-CoV pseudoviruses
SARS-CoV-2 and SARS-CoV pseudoviruses are non-replication defective retroviral particles having SARS-CoV-2 and SARS-CoV Spike membrane proteins (Spike, S) on their surfaces and carrying a luciferase reporter gene, which can mimic the infection process of a host cell (e.g., human hepatoma cell line Huh-7, 293T cell line 293T-ACE2 stably expressing human ACE2 receptor) by SARS-CoV-2 and SARS-CoV viruses, respectively, and express the luciferase reporter gene in the infected cell. Since pseudovirus infection does not produce mature viral particles, the relevant procedures can be safely performed in biosafety secondary laboratories.
SARS-CoV-2 and SARS-CoV pseudoviruses are obtained by cotransfection of 293T cells with respective S protein expression plasmids and HIV Env-deficient backbone plasmid (pNL 4-3.Luc. R-E-) with luciferase reporter gene, respectively. The S gene sequences of SARS-CoV-2 and SARS-CoV are designed according to NCBI GenBank sequences NC_045512 and ABD72979.1, and the gene sequences are synthesized by Nanjing Jinsrui company after codon optimization and are connected to pcDNA3 1 eukaryotic expression vectors to construct SARS-CoV-2 and SARS-CoV S protein expression plasmids. The pNL4-3.Luc. R-E-backbone plasmid was derived from U.S. NIH AIDS ReagentProgram. All plasmids were expanded by transformation of DH 5. Alpha. Competent cells and purified using the plasmid purification kit from Mey biosystems, the purification procedure being as described in the kit instructions.
293T cells were cultured in DMEM medium containing 10% fetal bovine serum (Gibco) and plated onto 10cm cell plates prior to transfection. After 24 hours of incubation, 293T cells were co-transfected with the backbone plasmid (pNL 4-3.Luc. R-E-) and SARS-CoV-2-expressing plasmid at a ratio of 3:1 using EZ Trans cell transfection reagent (Lissajous organism), see instructions for use of EZ Trans cell transfection reagent for detailed transfection methods. After 48 hours of transfection, the supernatant containing pseudoviruses was collected, centrifuged at 1500 rpm for 10 minutes to remove cell debris and sub-packaged for detection of neutralizing antibodies by freezing in a-80℃freezer.
4. Neutralization screening
After 13 days of in vitro culture of peripheral blood memory B cells, 40. Mu.l of culture supernatant was collected per well for detection of SARS-CoV-2 and SARS-CoV neutralizing antibodies. The detection method comprises the following steps: mu.l of the culture supernatant was mixed with 20. Mu.l of the pseudovirus supernatant obtained by the above production in 384-well cell culture plates, and after incubation at room temperature for 30 minutes, 50. Mu.l of 5000 293T-ACE2 cells were added to each well and the culture was continued in a cell culture incubator. After 48 hours, cells were lysed using a luciferase assay kit (Luciferase Assay System, promega cat.#e1500) and luciferase activity was measured per well, for specific assay methods, see kit instructions. The chemiluminescent RLU values per well were measured using a multifunctional enzyme-labeled instrument (Perkin Elmer). And calculating the neutralization inhibition percentage of the culture supernatant to the pseudo virus according to the ratio of the culture supernatant to the virus control RLU value, and screening out holes with the inhibition percentage of more than 90 percent as virus neutralization positive holes.
5. RT-PCR amplification of heavy and light chain genes
Virus neutralizes B cells of the positive well and variable regions of heavy and light chains of immunoglobulin genes are amplified using RT-PCR. Primer design and specific operation procedures of RT-PCR are disclosed in reference Tiller, T.et al J.Immunol Methods 2018, 329:112-124. the amplified antibody heavy chain and light chain variable region genes are purified and recovered by agarose gel electrophoresis, cloned into a PMD19-T vector by using a PMD19-T vector cloning kit (Takara 6013), and subjected to gene sequencing by selecting a monoclonal.
6. Expression and purification of monoclonal antibodies
The heavy chain variable region gene of the antibody and the pCMV/R-10E8 heavy chain gene (NIHAIDS ReagentProgram Cat 12290) which are sequenced correctly are respectively subjected to enzyme digestion by Age I and Sal I, and then the target fragment after purification and recovery of the connecting gel is connected and DH5 alpha competent cells are transformed to construct an antibody expression heavy chain plasmid; sequencing the correct antibody Lambda or Kappa light chain variable region gene and pCMV/R-10E8 Lambda light chain gene expression plasmid (NIHAIDS Reagent Program Cat 12291) or pCMV/R-N6 Kappa light chain gene expression plasmid (NIH AIDS Reagent Program Cat 12966) respectively through enzyme digestion of Age I and Xho I or Age I and BsiwI, connecting the recovered target fragment after gel purification and transforming DH5 alpha competent cells to construct antibody expression light chain plasmid; antibody heavy and light chain plasmids were purified by a plasmid purification kit (Meiy organism) (see FIG. 1 for SDS-PAGE detection of expressed purified antibodies) and co-transfected with 293T cell expression using EZ Trans cell transfection reagent (Lissajous organism) at a 1:1 ratio. After 72 hours, the cell transfected supernatant was collected and antibody IgG was purified from the supernatant using protein-G column (Tiandi and Biotech, changzhou) according to the instructions for use of protein-G column. The antibody IgG obtained was purified, the absorbance at 280nm was measured using Nanodrop 2000 (Thermo Fisher) and the antibody concentration was calculated.
Through the above sections 1-6, the inventors of the present application obtained several IgG antibodies, 7 of which were disclosed herein (designations: 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 in this order).
Amino acid sequence numbering information for 7 antibodies is given in table 1 below:
TABLE 1
Figure BDA0003750406910000141
Figure BDA0003750406910000151
Nucleotide sequence numbering information for 7 antibodies is given in table 2 below:
TABLE 2
Figure BDA0003750406910000152
7. Detection of neutralizing Activity of 7 MAbs (4L 12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H 22) of the present application on SARS-CoV-2 coronavirus
Different concentrations of mab were tested on 96-well cell plates to inhibit pseudovirus infection of Huh-7 cells to test the neutralizing capacity of mab against SARS-CoV-2 coronavirus.
The detection method comprises the following steps: 1) Huh-7 cells were seeded in 96-well cell plates at 1X 10 per well 4 And 37 ℃,5% CO 2 Culturing the cells in a cell culture box for 24 hours; 2) Diluting the monoclonal antibody into different concentrations by using a cell culture medium, mixing the monoclonal antibody with an equal volume of pseudovirus diluent containing 100TCID50, and incubating the mixture at 37 ℃ for 1 hour; 3) Discarding the cell culture solution, adding 50 μl of virus antibody complex into each well, arranging multiple wells, and simultaneously arranging an antibody-free group, a virus-free group and a positive serum control group; 4) After 12 hours of cultivation, 150 μl of maintenance solution was added to each well, and cultivation was continued for 48 hours at 37deg.C; 5) Cells were lysed using a luciferase assay kit (LuciferaseAssay System, promega cat.#e1500) and luciferase activity per well was measured, for specific assay methods reference kit instructions; detecting the chemiluminescent RLU value of each hole by using a multifunctional enzyme-labeled instrument (Perkin Elmer); 6) The percent neutralization inhibition of pseudoviruses by different concentrations of mab was calculated from the ratio of mab to virus control RLU values, and the median inhibition dose IC50 of mab against viruses was calculated using PRISM7 software (GraphPad).
The results are shown in Table 3 below.
TABLE 3 Table 3
IC50(ng/mL)
4L12 4.5
12F5 11.1
3D13 16.1
10C2 30.1
16L9 4.1
20E21 2.3
22H22 60.6
As can be seen from Table 3, 7 monoclonal antibodies 4L12, 12F5, 3D13, 10C2, 16L9, 20E21, 22H22 can well neutralize SARS-CoV-2 virus at ng/ml concentration, and the neutralization activity is very strong. The stronger the neutralizing activity, the less the amount of antibody used, and the lower the cost. Therefore, the 7 antibodies of 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 have better clinical application prospect.
8. Detection of S1 protein and RBD protein of 7 monoclonal antibodies (4L 12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H 22) for recognizing SARS-CoV-2 virus
The recognition of S1 and RBD proteins of SARS-CoV-2 virus by the 7 monoclonal antibodies obtained by the purification is sequentially detected by an enzyme-linked immunosorbent assay (ELISA).
The detection method comprises the following steps: 1 μg/ml of antigen protein (Yinqiao Shenzhou) was coated in 96-well ELISA plates at 4℃overnight. The plate was washed 5 times with PBS-T solution (0.2% Tween-20) and blocked for 1 hour at room temperature by adding 300. Mu.l blocking solution (PBS, 1%FBS,5%milk) to each well. After washing the plate 3 times with PBS, the monoclonal antibody was serially diluted 5-fold with PBS diluent (PBS, 5%FBS,2%BSA,1%Tween-20), 100. Mu.l of the sample was added to the ELISA plate and incubated at 37℃for 1 hour. PBS-T plates were washed 5 times, 100 μl horseradish peroxidase-labeled goat anti-human IgG antibody (Jackson Immunoresearch) diluted 1:2500 in PBS diluent was added to each well, and incubated for 1 hour at room temperature. PBS-T was washed 5 times, 150. Mu.l of ABTS chromogenic substrate (Thermo Fisher) was added, and after development for 30 minutes at room temperature in the dark, the absorbance at 405nm was read by an ELISA reader.
Referring to FIG. 1, monoclonal antibody 4L12 recognizes the S1 protein of SARS-CoV-2 virus and its RBD, and the detection result of S2 protein;
referring to FIG. 2, monoclonal antibody 12F5 recognizes the S1 protein of SARS-CoV-2 virus and its RBD, and the detection result of the S2 protein;
referring to FIG. 3, the detection results of the monoclonal antibody 3D13 recognizing the S1 protein of SARS-CoV-2 virus and its RBD, and the S2 protein;
referring to FIG. 4, the detection results of the monoclonal antibody 10C2 recognizing the S1 protein of SARS-CoV-2 virus and its RBD, and the S2 protein;
referring to FIG. 5, the detection results of the monoclonal antibody 16L9 recognizing the S1 protein of SARS-CoV-2 virus and its RBD, and the S2 protein;
referring to FIG. 6, the detection results of the monoclonal antibody 20E21 recognizing the S1 protein of SARS-CoV-2 virus and its RBD, and the S2 protein;
referring to FIG. 7, monoclonal antibody 22H22 recognizes the S1 protein of SARS-CoV-2 virus and its RBD, and the detection result of the S2 protein.
1-7, monoclonal antibodies 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 are all capable of recognizing and binding to the S1 protein of SARS-CoV-2 virus and its RBD (conserved region); given that RBD of S1 protein of coronavirus is a region to which ACE2 receptor binds, it is highly conserved, and it is presumed that monoclonal antibodies 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 of the present application may have binding ability and neutralizing ability to other coronaviruses, and coronaviruses that may occur in the future, in addition to strong binding ability and neutralizing ability to SARS-CoV-2 virus.
8. Biological membrane layer interference technology for detecting binding capacity of 7 monoclonal antibodies (4L 12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H 22) and RBD of S1 protein of SARS-CoV-2 virus
In order to detect the interaction between 7 mabs of the present application and RBD of S1 protein of SARS-CoV-2 virus, the binding kinetics between them was detected using a biofilm layer interference technique, the detection procedure was performed on an OctetRED96 (Fortebio) instrument.
The detection method comprises the following steps: immersing the AHC probe in sterile water for 10 minutes in advance for balancing, and carrying out the detection process under the reaction condition of 30 ℃ in advance, wherein the detection process can be divided into the following five steps of 1) zeroing: immersing the probe in sterile water for 60 seconds to obtain a detection baseline; 2) Capture antibody: immersing the probe into a monoclonal antibody solution with the concentration of 10 mug/ml for 200 seconds to act as a capture antibody; 3) Zeroing again: immersing the probe in buffer solution (PBS solution added with 0.02% Tween 20) for 120 seconds to remove unbound antibody; 4) Combining with RBD: immersing the probe into RBD protein solution with initial concentration of 100nM and 3 times of gradient dilution, and reacting for 300 seconds to obtain a dynamic curve of the combination of the monoclonal antibody and the RBD; 5) Binding dissociation: the probe was placed in buffer for 300 seconds. The binding of the protein causes a change in the thickness of the biological membrane, resulting in a relative shift in the interference light wave, which is detected by the spectrometer, forming an interference spectrum, which is displayed as a real-time shift (nm) of the interference spectrum. The dynamic curve of RBD binding dissociation with the monoclonal antibodies of the application is detected. Subtracting the data of the buffer control wells from the data of the sample wells at the time of data analysis, subtracting the non-specific interference of the buffer solution, using 1:1, carrying out integral curve fitting on the combination of the monoclonal antibody and the RBD dilution concentration to obtain an average combination constant K on Dissociation constant K off Affinity constant K D Values.
The detection results are shown in FIGS. 8-14, and are the affinity detection results of RBD of monoclonal antibodies 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 binding to S1 protein of SARS-CoV-2 virus; five curves representing the dynamic binding dissociation curves of the mab with five different concentrations of RBD are shown in each figure.
As can be seen from fig. 8-14, the concentration gradient of RBD binding of 7 mabs 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 of the present application to S1 protein of SARS-CoV-2 virus; after combination, dissociation is carried out, and the number of dissociated RBDs is very small; k of 7 MAbs 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 D Values were (1.49+ -0.06) nM, (2.22+ -0.07) nM, (4.17+ -0.15) nM, (3.36+ -0.18) nM, (1.21+ -0.06) nM, (2.3+ -0.07) nM, (5.07+ -0.2) nM; the 7 mabs of this application are shown to have very strong affinity for the RBD conserved region of the S1 protein of SARS-CoV-2. From this it can be deduced that the 7 mabs 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 of the present application demonstrated in section 7 above have strong neutralizing activity against RBD of S1 protein of SARS-CoV-2 virus, as a result of the very strong affinity of the 7 mabs of the present application to RBD conserved regions of S1 protein of SARS-CoV-2 virus. From a combination of the results in Table 1 and FIGS. 1-14, it was further verified that 7 mabs 4L12, 12F5, 3D13, 10C2, 16L9, 20E21 and 22H22 of the present application may have binding and neutralizing capabilities for other coronaviruses, as well as coronaviruses that may occur in the future, in addition to having strong binding and neutralizing capabilities for SARS-CoV-2.
It should be understood that although the present disclosure describes embodiments, not every embodiment is provided with a separate embodiment, and that this description is for clarity only, and that the skilled artisan should recognize that the embodiments may be combined as appropriate to form other embodiments that will be understood by those skilled in the art.
The above list of detailed descriptions is only specific to practical embodiments of the present invention, and they are not intended to limit the scope of the present invention, and all equivalent embodiments or modifications that do not depart from the spirit of the present invention should be included in the scope of the present invention.

Claims (10)

1. An antibody or antigen-binding fragment thereof to a coronavirus comprising a heavy chain variable region comprising three heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3 and a light chain variable region comprising three light chain complementarity determining regions LCDR1, LCDR2 and LCDR3; the method is characterized in that:
the sequence of the HCDR1 is shown as SEQ ID NO.41, the sequence of the HCDR2 is shown as SEQ ID NO.42, and the sequence of the HCDR3 is shown as SEQ ID NO. 43; and the sequence of LCDR1 is shown as SEQ ID NO.45, the sequence of LCDR2 is shown as SEQ ID NO.46, and the sequence of LCDR3 is shown as SEQ ID NO. 47.
2. The antibody or antigen-binding fragment thereof of claim 1, wherein:
the heavy chain variable region has a sequence shown as SEQ ID NO.44 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO.44, and the light chain variable region has a sequence shown as SEQ ID NO.48 or a sequence with more than 80% of sequence homology with the sequence shown as SEQ ID NO. 48.
3. The antibody or antigen-binding fragment thereof of claim 1 or 2, wherein:
the antibody is a monoclonal antibody; preferably, the antibody is a fully human monoclonal antibody; preferably, the antibody is any one or a combination of a plurality of IgG1, igG2, igG3 or IgG 4; the antigen binding fragment is Fv, fab, F (ab ') 2, fab', dsFv, scFv or sc (Fv) 2.
4. A nucleic acid molecule characterized in that: the nucleic acid molecule encodes the antibody or antigen-binding fragment thereof of any one of claims 1 to 3.
5. A vector comprising the nucleic acid molecule of claim 4.
6. A host cell comprising the vector of claim 5.
7. A pharmaceutical composition characterized by: the pharmaceutical composition comprising the antibody or antigen-binding fragment thereof of any one of claims 1 to 3.
8. A test product, characterized by: the detection product comprising the antibody or antigen-binding fragment thereof of any one of claims 1 to 3.
9. A method of producing an antibody or antigen-binding fragment thereof according to any one of claims 1 to 3, wherein: culturing the host cell of claim 6 to produce the antibody or antigen-binding fragment thereof.
10. Use of an antibody or antigen-binding fragment thereof according to any one of claims 1 to 3 or a pharmaceutical composition according to claim 7 for the manufacture of a medicament for the treatment or prophylaxis of a disease caused by coronavirus.
CN202210841040.3A 2020-09-30 2020-09-30 Antibodies or antigen binding fragments thereof to coronaviruses Pending CN116063464A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210841040.3A CN116063464A (en) 2020-09-30 2020-09-30 Antibodies or antigen binding fragments thereof to coronaviruses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011065506.2A CN112159469B (en) 2020-09-30 2020-09-30 Antibodies or antigen-binding fragments thereof to coronaviruses
CN202210841040.3A CN116063464A (en) 2020-09-30 2020-09-30 Antibodies or antigen binding fragments thereof to coronaviruses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202011065506.2A Division CN112159469B (en) 2020-09-30 2020-09-30 Antibodies or antigen-binding fragments thereof to coronaviruses

Publications (1)

Publication Number Publication Date
CN116063464A true CN116063464A (en) 2023-05-05

Family

ID=73862418

Family Applications (3)

Application Number Title Priority Date Filing Date
CN202210890641.3A Pending CN115710311A (en) 2020-09-30 2020-09-30 Antibodies or antigen-binding fragments thereof to coronaviruses
CN202011065506.2A Active CN112159469B (en) 2020-09-30 2020-09-30 Antibodies or antigen-binding fragments thereof to coronaviruses
CN202210841040.3A Pending CN116063464A (en) 2020-09-30 2020-09-30 Antibodies or antigen binding fragments thereof to coronaviruses

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN202210890641.3A Pending CN115710311A (en) 2020-09-30 2020-09-30 Antibodies or antigen-binding fragments thereof to coronaviruses
CN202011065506.2A Active CN112159469B (en) 2020-09-30 2020-09-30 Antibodies or antigen-binding fragments thereof to coronaviruses

Country Status (1)

Country Link
CN (3) CN115710311A (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7116256B1 (en) 2020-04-02 2022-08-09 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Anti-SARS-COV-2-Spike Glycoprotein Antibodies and Antigen-Binding Fragments
CR20220660A (en) 2020-06-03 2023-02-17 Regeneron Pharma METHODS FOR TREATING OR PREVENTING SARS-CoV-2 INFECTIONS AND COVID-19 WITH ANTI-SARS-CoV-2 SPIKE GLYCOPROTEIN ANTIBODIES
WO2021249547A1 (en) * 2020-06-12 2021-12-16 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Anti-coronavirus antibodies and uses thereof
CN115141271B (en) * 2021-01-31 2024-06-11 中南大学湘雅医院 Novel coronavirus monoclonal antibody XY7 and application thereof
WO2022188829A1 (en) * 2021-03-10 2022-09-15 上海君实生物医药科技股份有限公司 Sars-cov-2 antibody and application thereof
CN113880947B (en) * 2021-07-26 2023-07-04 中国人民解放军军事科学院军事医学研究院 Small molecule antibody, coding gene thereof, preparation method and application thereof, and pharmaceutical composition
CN113563464B (en) * 2021-08-01 2023-02-03 中国疾病预防控制中心性病艾滋病预防控制中心 Humanized high-neutralization-activity anti-novel coronavirus monoclonal antibody and application thereof
CN114106191A (en) * 2021-12-20 2022-03-01 复旦大学 Bispecific antibody for neutralizing coronavirus
WO2023131262A1 (en) * 2022-01-10 2023-07-13 杰库(上海)生物医药研究有限公司 Antigen-binding protein specifically bound to sars-cov-2
CN114751986A (en) * 2022-01-27 2022-07-15 复旦大学 Multispecific antibodies for neutralizing novel coronaviruses
CN114409774B (en) * 2022-02-07 2024-04-02 浙江大学医学院附属第一医院 Broad-spectrum humanized anti-novel coronavirus monoclonal antibody and application thereof
CN115286712A (en) * 2022-03-18 2022-11-04 百斯医学诊断科技(北京)有限公司 Novel coronavirus Delta mutant strain specific antibody and application thereof
CN114957455B (en) * 2022-05-06 2023-05-16 深圳国家感染性疾病临床医学研究中心 Novel coronavirus monoclonal antibody and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111592594B (en) * 2020-03-13 2022-05-10 北京大学 Monoclonal antibody for resisting novel coronavirus and application thereof
CN111690058B (en) * 2020-03-30 2021-02-05 三优生物医药(上海)有限公司 Antibodies with neutralizing activity against coronaviruses and uses thereof
JP7116256B1 (en) * 2020-04-02 2022-08-09 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Anti-SARS-COV-2-Spike Glycoprotein Antibodies and Antigen-Binding Fragments
CN111647076B (en) * 2020-04-27 2021-02-26 南京医科大学 Neutralizing single-domain antibody for resisting novel coronavirus SARS-Cov-2 and application thereof
CN111592595B (en) * 2020-04-27 2021-02-19 南京医科大学 Neutralizing antibody against novel coronavirus SARS-Cov-2 and application thereof

Also Published As

Publication number Publication date
CN112159469A (en) 2021-01-01
CN115710311A (en) 2023-02-24
CN112159469B (en) 2022-08-02

Similar Documents

Publication Publication Date Title
CN116063464A (en) Antibodies or antigen binding fragments thereof to coronaviruses
CN116023478A (en) Neutralizing antibodies or antigen binding fragments thereof for coronaviruses
WO2022022445A1 (en) Antibody that specifically binds to coronavirus or antigen-binding fragment thereof
CN107847591B (en) Multivalent human immunodeficiency virus antigen binding molecules and uses thereof
KR100337069B1 (en) Anti-HIV Monoclonal Antibodies
US7736648B2 (en) Anti-RSV G protein antibodies
CN113354729B (en) Monoclonal antibody for resisting novel coronavirus and application thereof
WO2020038147A1 (en) Anti-bcma single domain antibodies and application thereof
US20170158753A1 (en) Monoclonal antibodies directed against envelope glycoproteins from multiple filovirus species
CN113354730B (en) Monoclonal antibody for resisting novel coronavirus and application thereof
TW202206455A (en) Monoclonal antibody against new coronavirus and use thereof
CN118184774A (en) Fully human antibody for neutralizing SARS-CoV-2 and variant strain and application thereof
AU2021326889A1 (en) Anti-CD73 antibody and use thereof
WO2021228135A1 (en) Method for preparing antigen-binding unit
JP2006514921A (en) Anti-TSG101 antibodies and their use for the treatment of viral infections
CN117430716B (en) Recombinant interferon drug targeting conserved HIV gp41 subunit near-membrane-end outer region and application thereof
US20240218074A1 (en) Anti-cd73 antibody and use thereof
US20240209065A1 (en) Secretory iga antibodies against covid infection
WO2022165434A2 (en) Fc-enhanced antibodies for prevention and treatment of ebola virus infection
WO2022161597A1 (en) Method for rapid identification of cross-reactive and/or rare antibodies
CN116574178A (en) Neutralizing humanized monoclonal antibody of Nipah virus and application thereof
WO2022162009A1 (en) Method for rapid identification of cross-reactive and/or rare antibodies
WO2022217048A1 (en) Oncolytic viruses expressing anti-ror1/anti-cd3 bispecific antibodies

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20230712

Address after: 201108 Room 411, Building 14, No. 3333, Huaning Road, Minhang District, Shanghai

Applicant after: Super Extraordinary (Shanghai) Medical Technology Co.,Ltd.

Address before: 201508 no.2901 Caolang Road, Jinshan District, Shanghai

Applicant before: SHANGHAI PUBLIC HEALTH CLINICAL CENTER

TA01 Transfer of patent application right