CN116751289A - anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof - Google Patents

anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof Download PDF

Info

Publication number
CN116751289A
CN116751289A CN202310967602.3A CN202310967602A CN116751289A CN 116751289 A CN116751289 A CN 116751289A CN 202310967602 A CN202310967602 A CN 202310967602A CN 116751289 A CN116751289 A CN 116751289A
Authority
CN
China
Prior art keywords
antibody
west nile
nile virus
expression vector
seq
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.)
Granted
Application number
CN202310967602.3A
Other languages
Chinese (zh)
Other versions
CN116751289B (en
Inventor
肖鹤
冯健男
沈倍奋
郝相君
朱婉露
陈国江
王晶
乔春霞
李新颖
刘成华
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.)
Academy of Military Medical Sciences AMMS of PLA
Original Assignee
Academy of Military Medical Sciences AMMS of PLA
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 Academy of Military Medical Sciences AMMS of PLA filed Critical Academy of Military Medical Sciences AMMS of PLA
Priority to CN202310967602.3A priority Critical patent/CN116751289B/en
Publication of CN116751289A publication Critical patent/CN116751289A/en
Application granted granted Critical
Publication of CN116751289B publication Critical patent/CN116751289B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

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
    • C07K16/1081Togaviridae, e.g. flavivirus, rubella virus, hog cholera virus
    • 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
    • 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
    • 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
    • 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
    • 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
    • G01N2333/18Togaviridae; Flaviviridae
    • G01N2333/183Flaviviridae, e.g. pestivirus, mucosal disease virus, bovine viral diarrhoea virus, classical swine fever virus (hog cholera virus) or border disease 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention discloses an anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof, and provides an anti-West Nile virus neutralizing antibody and a derivative containing the antibody, wherein the derivative comprises a nucleic acid molecule for encoding the antibody, an expression vector containing the nucleic acid molecule, a host cell containing the expression vector, a pharmaceutical composition, a detection reagent, a detection kit and the like.

Description

anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to an anti-West Nile virus neutralizing antibody capable of reducing ADE effect and application thereof.
Background
West Nile Virus (WNV) is a enveloped single-stranded positive-stranded RNA virus with a particle diameter of 45-50. 50 nm. WNV belongs to the family flaviviridae, members of the genus flaviviridae. Other members of the genus also include ZiKV, dengue (DENV), yellow Fever (YFV), epidemic encephalitis B (JEV), and the like. The single open reading frame (open reading frame, ORF) in the WNV viral genome encodes 10 mature proteins co-translated and post-translationally processed by viral and intracellular proteases, including 3 structural proteins (capsid protein C, membrane protein prM/M and envelope protein E) and 7 non-structural proteins (NS 1, NS2a, NS2b, NS3, NS4a, NS4b and NS 5), wherein the structural proteins are primarily involved in the formation of WNV viral particles, the non-structural proteins are involved in WNV viral RNA replication, and the functional region of the E protein is composed of 3 domains (dii, dii and dii).
West Nile fever (West Nile fever) is a zoonotic disease caused by WNV. WNV is transmitted mainly by mosquitoes, which can cause nervous system diseases and even death of human beings, and also can cause morbidity of animals such as horses, birds, cats and the like. WNV is widely prevalent in africa, europe, middle east, north america, and west asia. At present, no specific therapeutic drug aiming at the West Nile virus exists. Antibody-dependent enhancement (ADE-dependent enhancement) is believed to be one important cause of exacerbation of a patient's condition following secondary infection, and ADE effects are generally mediated by non-neutralizing antibodies, but neutralizing antibodies also induce ADE at sub-neutralizing concentrations, particularly cross-reactive antibodies to the flavivirus envelope (E) protein which may enhance infection by heterologous flaviviruses, inducing ADE. Since ADE may amplify the severity of west nile virus infection, lowering ADE has become a major focus of development of antibody-based therapies.
Disclosure of Invention
In view of the above, the present invention aims to provide an anti-west nile virus neutralizing antibody that reduces ADE effect and uses thereof.
In order to achieve the above purpose, the present invention adopts the following technical scheme:
in a first aspect, the invention provides an antibody against west nile virus.
Further, the amino acid sequences of HCDR1, HCDR2 and HCDR3 in the heavy chain variable region of the antibody are CDR1, CDR2 and CDR3 in the heavy chain variable region as shown in SEQ ID NO. 4;
the amino acid sequences of LCDR1, LCDR2 and LCDR3 in the light chain variable region of the antibody are CDR1, CDR2 and CDR3 in the light chain variable region shown in SEQ ID NO. 8.
Further, the amino acid sequences of the HCDR1, the HCDR2 and the HCDR3 are respectively shown as SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3 or are respectively amino acid sequences with at least 90 percent of homology with the amino acid sequences corresponding to the SEQ ID NO. 1, the SEQ ID NO. 2 and the SEQ ID NO. 3;
the amino acid sequences of LCDR1, LCDR2 and LCDR3 are respectively shown as SEQ ID NO. 5, SEQ ID NO. 6 and SEQ ID NO. 7 or are respectively the amino acid sequences with at least 90% homology with the amino acid sequences corresponding to SEQ ID NO. 5, SEQ ID NO. 6 and SEQ ID NO. 7.
In some embodiments, amino acid sequences having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence homology to the amino acid sequences corresponding to HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, LCDR3 of the invention are also within the scope of the invention, the amino acid sequences having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence homology comprising being obtained by one or more amino acid or nucleotide deletion, insertion or substitution mutations of the parent sequence.
In some embodiments, the amino acid sequences corresponding to HCDR1, HCDR2 and HCDR3 of the present invention are not limited to the amino acid sequences shown in SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, the amino acid sequences corresponding to LCDR1, LCDR2 and LCDR3 of the present invention are not limited to the amino acid sequences shown in SEQ ID NO. 5, SEQ ID NO. 6 and SEQ ID NO. 7, and the amino acid sequences or nucleotide sequences corresponding to CDR1, CDR2 and CDR3 in the heavy chain variable region shown in SEQ ID NO. 4, and CDR1, CDR2 and CDR3 in the light chain variable region shown in SEQ ID NO. 8 are all within the scope of the present invention by using any CDR numbering scheme (existing CDR numbering scheme or new CDR numbering scheme to be generated in the future).
In some embodiments, the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, LCDR3 are defined according to any one of an IMGT numbering scheme, chothia numbering scheme, kabat numbering scheme, martin (enhanced Chothia) numbering scheme, abM numbering scheme, aho numbering scheme, or any combination of any plurality of (two or more of) HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, LCDR3 defined by the above definition are also included in the scope of the invention.
The mutation of the nucleotide sequence corresponding to the antibody of the present invention can be easily performed by one of ordinary skill in the art using a known method, for example, a method using directed point mutation. Those artificially modified nucleotides having 75% or more homology to the nucleotide sequence corresponding to the antibody of the present invention are derived from the nucleotide sequence of the present invention and are equivalent to the sequences of the present invention, as long as they encode the antibody of the first aspect of the present invention, and are also included in the scope of the present invention.
In a second aspect, the invention provides an anti-west nile virus neutralizing antibody that reduces the ADE effect.
Further, the HCDR1, HCDR2, HCDR3 in the heavy chain variable region of the antibody is HCDR1, HCDR2, HCDR3 as described in the first aspect of the invention;
the LCDR1, LCDR2, LCDR3 in the light chain variable region of the antibody is LCDR1, LCDR2, LCDR3 as described in the first aspect of the invention;
the amino acid sequence of the Fc segment of the antibody is shown as SEQ ID NO. 9 or the amino acid sequence with at least 90% homology with the amino acid sequence corresponding to SEQ ID NO. 9.
In a specific embodiment of the invention, the anti-West Nile virus neutralizing antibody reducing the ADE effect is obtained by subjecting the Fc segment 234 and 235 of the antibody according to the first aspect of the invention to point mutation (both L mutation to A). The heavy chain variable region and the light chain variable region of the anti-West Nile virus neutralizing antibody for reducing the ADE effect are as described in the first aspect of the invention, the amino acid sequence of the Fc segment of the anti-West Nile virus neutralizing antibody for reducing the ADE effect is shown as SEQ ID NO. 9, and the nucleotide sequence of the Fc segment is shown as SEQ ID NO. 10.
In a third aspect, the invention provides a nucleic acid molecule.
Further, the nucleic acid molecule encodes an antibody according to the first aspect of the invention or an antibody according to the second aspect of the invention.
In a specific embodiment of the invention, the nucleotide sequence of said nucleic acid molecule encoding the Fc segment of the antibody according to the second aspect of the invention is shown in SEQ ID NO. 9.
In some embodiments, the nucleic acid molecule is isolated or purified. The sequence of the nucleic acid molecule may be obtained using conventional techniques or using hybridoma techniques. Once the relevant sequences are obtained, recombinant methods can be used to obtain the relevant sequences in large quantities. This is usually done by cloning it into a vector, transferring it into a cell, and isolating the relevant sequence from the propagated host cell by conventional methods. Furthermore, the sequences concerned, in particular fragments of short length, can also be synthesized by artificial synthesis. Generally, fragments of longer sequence are obtained by first synthesizing a plurality of small fragments and then ligating them.
In a fourth aspect, the invention provides an expression vector.
Further, the expression vector comprises the nucleic acid molecule of the third aspect of the invention.
In some embodiments, the vector includes a DNA vector (e.g., a plasmid), an RNA vector (e.g., a vector of viral origin). In some embodiments, the viral-derived vectors include, but are not limited to: lentiviral vectors, retroviral vectors, adenoviral vectors, adeno-associated viral vectors, poxviral vectors, herpesviral vectors.
In some embodiments, the expression vector may be constructed using methods well known to those of skill in the art. These methods include, but are not limited to: recombinant DNA techniques, DNA synthesis techniques, and the like. The DNA encoding the antibody may be operably linked to multiple cloning sites in a vector to direct mRNA synthesis to express the protein, or for homologous recombination.
In a fifth aspect, the invention provides an engineered host cell.
Further, the engineered host cell comprises the expression vector of the fourth aspect of the invention.
In some embodiments, the host cell comprises a eukaryotic cell, a prokaryotic cell. In some embodiments, the eukaryotic cells include, but are not limited to: mammalian cells, insect cells, plant cells, yeast cells. Such prokaryotic cells include, but are not limited to: bacteria, actinomycetes, cyanobacteria, mycoplasma, chlamydia, rickettsia. In some embodiments, host cells for expressing the antibodies of the invention include E.coli, yeast cells, insect cells, COS cells, CHO cells, immune cells, and the like. In some embodiments, the immune cells include T cells, B cells, NK cells, iNKT cells, CTL cells, dendritic cells, myeloid cells, monocytes, macrophages, or any combination thereof.
In some embodiments, the host cell is prepared by: introducing a nucleic acid molecule according to the third aspect of the invention or an expression vector according to the fourth aspect of the invention into a host cell, such methods of introducing include, but are not limited to: physical, chemical, biological methods. The physical methods include, but are not limited to: calcium phosphate precipitation, lipofection, particle bombardment, microinjection, and electroporation; such chemical methods include, but are not limited to: colloidal dispersion systems, lipid-based systems; the colloidal dispersion system includes, but is not limited to: macromolecular complexes, nanocapsules, microspheres, beads; the lipid-based system includes, but is not limited to: oil-in-water emulsions, micelles, mixed micelles, liposomes; the biological methods include, but are not limited to: DNA vectors, lentiviral vectors, poxviral vectors, herpes simplex viral vectors, adenoviral vectors, adeno-associated viral vectors.
In some embodiments, the nucleic acid molecules of the third aspect of the invention or the expression vectors of the fourth aspect of the invention may be introduced into host cells by a variety of suitable means, and are not limited to the methods listed in the invention, such as calcium phosphate transfection, DEAE-dextran mediated transfection, microinjection, electroporation, TALEN methods, ZFN methods, non-viral vector mediated transfection (e.g., liposomes) or viral vector mediated transfection (e.g., lentiviral infection, retroviral infection, adenoviral infection), and other physical, chemical or biological means for transferring into cells, such as transposon technology, CRISPR-Cas9, and the like.
In a sixth aspect, the invention provides a recombinant expression vector.
Further, the recombinant expression vector is pFRT-IgG1 kappa-FcM-WNV;
the recombinant expression vector is obtained by taking an antibody mammalian cell expression vector pFT-IgG 1 kappa as a template and mutating 234 and 235 sites of an Fc segment from L to A.
In some embodiments, the antibody mammalian cell expression vector pFT-IgG 1 kappa is a commercial expression vector, available via conventional purchase routes.
In a seventh aspect, the present invention provides any one of the following substances, comprising:
(1) An antibody derivative obtained by coupling an antibody according to the first aspect of the invention or an antibody according to the second aspect of the invention to a functional agent;
in some embodiments, the functional agent may be a therapeutic agent or a diagnostic agent. In some embodiments, the therapeutic agent includes, but is not limited to: ribavirin, acyclovir, oseltamivir, interferon alpha, arbidol, honeysuckle, fructus forsythiae, radix isatidis, dandelion and radix bupleuri. In some embodiments, the diagnostic agent includes, but is not limited to: radionuclides, chemiluminescent agents, bioluminescent agents, paramagnetic ions, enzymes, photosensitive diagnostic agents. In some embodiments, the radionuclide comprises: 110 In、 111 In、 177 Lu、 18 F、 52 Fe、 62 Cu、 64 Cu、 67 Cu、 67 Ga、 68 Ga、 86 Y、 90 Y、 89 Zr、 94m Tc、 94 Tc、 99m Tc、 120 I、 123 I、 124 I、 125 I、 131 I、 154-158 Gd、 32 F、 11 C、 13 N、 15 O、 186 Re、 188 Re (E); the chemiluminescent agent comprises luminol, isoluminol, aromatic acridinium ester, imidazole, acridinium salt and oxalate; the bioluminescent agent comprises fluorescein, luciferase and aequorin; the paramagnetic ion comprises chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II); the enzyme comprises horseradish peroxidase, alkaline phosphatase, glucose oxidase, beta-D-galactosidase, urease, catalase, or glucoamylase; the photosensitive diagnostic agent comprises dihydroxysilicon phthalocyanine, methylene blue, protoporphyrin, hematoporphyrin and photoporphyrin.
(2) A pharmaceutical composition comprising an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention and/or an antibody derivative according to the seventh aspect of the invention;
in some embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers and/or excipients, which are described in detail in Remington's Pharmaceutical Sciences (19 th ed., 1995), which are used as needed to aid stability of the formulation or to aid in enhancing the bioavailability of the active or active substance. In some embodiments, a safe and effective amount of the pharmaceutical composition of the invention is administered to a human when the pharmaceutical composition is used. The dosage and mode of administration of the pharmaceutical composition is not particularly limited, and the skilled practitioner will generally be able to readily determine the prescription and dosage of the prescription for the desired treatment and/or prophylaxis, for example, by injection or other means of treatment.
In some embodiments, the pharmaceutical composition may also be used in combination with other therapeutic agents having therapeutic and/or prophylactic effects on diseases associated with West Nile Virus infection, including any substance having therapeutic and/or prophylactic effects on diseases associated with West Nile Virus infection. Therefore, the technical scheme of combining the pharmaceutical composition and any other therapeutic agent with the therapeutic and/or prophylactic effects on diseases related to the West Nile virus infection is also included in the protection scope of the invention.
(3) A detection reagent comprising an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention and/or an antibody derivative according to the seventh aspect of the invention;
in some embodiments, diagnostic and/or auxiliary diagnostic uses of the detection reagent are achieved by the following methods: contacting a sample suspected of having the target analyte (west nile virus and/or west nile virus E protein) with a detection reagent according to the present invention, and analyzing the detected sample for the presence or level of the target analyte.
In some embodiments, the detection reagent may further comprise a detectable label including a fluorescent dye, avidin, a paramagnetic atom, a radioisotope. In some embodiments, the fluorescent dye is fluorescein, rhodamine, texas red, phycoerythrin, phycocyanin, allophycocyanin, polymannuin-chlorophyll protein; the avidin is biotin, ovalbumin, streptavidin, vitelline avidin or avidin-like; the radioisotope is radioiodine, radioactive cesium, radioactive iridium or radioactive cobalt.
(4) A test kit comprising the test reagent.
In some embodiments, the assay kit further comprises a container, instructions for use, buffers, etc., and in other embodiments, the kit further comprises a lysis medium for lysing the sample to be tested, universal reagents and buffers required for the assay, such as various buffers, assay labels, assay substrates, etc. The detection kit can be an in-vitro diagnosis device and is used for diagnosing and/or assisting in diagnosing diseases related to West Nile virus infection.
In an eighth aspect, the present invention provides any one of the following methods, the method comprising:
(1) A method of making an antibody according to the first aspect of the invention or an antibody according to the second aspect of the invention, the method comprising the steps of: culturing the engineered host cell according to the fifth aspect of the invention, and isolating and purifying the antibody according to the first aspect of the invention or the antibody according to the second aspect of the invention from the host cell culture product;
(2) A method of making an engineered host cell according to the fifth aspect of the invention, the method comprising the steps of: introducing a nucleic acid molecule according to the third aspect of the invention or an expression vector according to the fourth aspect of the invention into a host cell;
in some embodiments, the method of introducing includes a physical method, a chemical method, a biological method. In some embodiments, the physical methods include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation; the chemical method comprises a colloid dispersion system and a lipid-based system; the colloid dispersion system comprises macromolecular complexes, nanocapsules, microspheres and beads; the lipid-based system includes oil-in-water emulsions, micelles, mixed micelles, and liposomes; the biological method comprises a DNA vector and an RNA vector.
(3) A method for detecting west nile virus and/or west nile virus E protein for non-diagnostic and non-therapeutic purposes, the method comprising the steps of: contacting a sample to be tested with the antibody according to the first aspect of the invention, the antibody according to the second aspect of the invention, the antibody derivative according to the seventh aspect of the invention, and detecting the formation of an immune complex of west nile virus and/or west nile virus E protein and the antibody;
(4) A method of inhibiting west nile virus and/or west nile virus E protein activity in vitro, the method comprising the steps of: introducing an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention and/or an expression vector according to the fourth aspect of the invention into a cell of an organism, whereby the activity of a west nile virus and/or west nile virus E protein is inhibited by expression of the antibody according to the first aspect of the invention or the antibody according to the second aspect of the invention;
(5) A method of inactivating west nile virus and/or west nile virus E protein, the method comprising the steps of: contacting a sample of interest with an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, an antibody derivative according to the seventh aspect of the invention.
In some embodiments, the sample may be selected from urine, blood, serum, plasma, saliva, ascites fluid, non-tissue associated cells, tissue, histological preparations, and the like, derived from a test subject, and the invention is not particularly limited to sample types.
In a ninth aspect, the present invention provides any one of the following applications, the applications comprising:
(1) Use of an antibody according to the first aspect of the invention and/or an antibody according to the second aspect of the invention for the preparation of an antibody derivative for the detection of west nile virus and/or west nile virus E protein, an antibody derivative for the treatment and/or prophylaxis of a disease associated with west nile virus infection;
(2) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention and/or an antibody derivative according to the seventh aspect of the invention for the preparation of a detection reagent for the detection of west nile virus and/or west nile virus E protein;
(3) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention, an antibody derivative according to the seventh aspect of the invention and/or a detection reagent according to the seventh aspect of the invention for the preparation of a detection kit for the detection of west nile virus and/or west nile virus E protein;
(4) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention, an antibody derivative according to the seventh aspect of the invention, a detection reagent according to the seventh aspect of the invention and/or a detection kit according to the seventh aspect of the invention for the preparation of a product for the diagnosis and/or co-diagnosis of a disease associated with a west nile virus infection;
(5) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention, an antibody derivative according to the seventh aspect of the invention, a detection reagent according to the seventh aspect of the invention and/or a detection kit according to the seventh aspect of the invention for the detection of west nile virus and/or west nile virus E protein for non-diagnostic purposes;
(6) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention and/or an expression vector according to the fourth aspect of the invention for the preparation of an engineered host cell for the treatment and/or prevention of diseases associated with West Nile Virus infection;
(7) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention and/or an antibody derivative according to the seventh aspect of the invention in the manufacture of a medicament for the treatment and/or prevention of a disease associated with a west nile virus infection;
(8) Use of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention, an antibody derivative according to the seventh aspect of the invention and/or a pharmaceutical composition according to the seventh aspect of the invention for the preparation of a biological agent for the treatment and/or prevention of diseases associated with a west nile virus infection;
(9) The use of a recombinant expression vector according to the sixth aspect of the invention for the preparation of antibodies that reduce the ADE effect.
In addition, the invention also provides a method for diagnosing and/or assisting in diagnosing diseases related to West Nile virus infection.
Further, the method comprises the following steps: detecting a test sample derived from a subject by using the antibody according to the first aspect of the present invention, the antibody according to the second aspect of the present invention, the antibody derivative according to the seventh aspect of the present invention, the detection reagent according to the seventh aspect of the present invention and/or the detection kit according to the seventh aspect of the present invention, and detecting the presence of west nile virus and/or west nile virus E protein in the test sample by antigen-antibody reaction to diagnose and/or aid in diagnosing whether the subject has west nile virus infection-related disease and/or risk of developing west nile virus infection-related disease.
In addition, the invention also provides a method for treating and/or preventing diseases related to West Nile virus infection.
Further, the method comprises the following steps: administering to a subject in need thereof a therapeutically and/or prophylactically effective amount of an antibody according to the first aspect of the invention, an antibody according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, an expression vector according to the fourth aspect of the invention, an engineered host cell according to the fifth aspect of the invention, an antibody derivative according to the seventh aspect of the invention and/or a pharmaceutical composition according to the seventh aspect of the invention.
In particular embodiments of the invention, the west nile virus infection-associated disease includes, but is not limited to: west Nile fever caused by West Nile virus infection, diseases related to West Nile virus infection (such as fever, rash, nausea, vomiting, neuroinvasive diseases, encephalitis, meningitis, acute delayed paralysis, etc.), and various symptoms or diseases caused by West Nile virus infection are within the scope of the invention.
Compared with the prior art, the invention has the following advantages and beneficial effects:
the invention provides a brand-new anti-West Nile virus neutralizing antibody for reducing ADE effect and a derivative containing the antibody, wherein the derivative comprises a nucleic acid molecule for encoding the antibody, an expression vector containing the nucleic acid molecule, a host cell containing the expression vector, a pharmaceutical composition, a detection reagent, a detection kit and the like. The antibody provided by the invention not only has stronger virus neutralization activity, but also can reduce ADE effect, lays a foundation for diagnosis and treatment of diseases related to West Nile virus infection, development of medicines for treating diseases related to West Nile virus infection and the like, can be used as candidates for clinical medicines for resisting West Nile virus, and has good application prospects.
Drawings
FIG. 1 is the identification of the recombinant expression plasmid pBRT-IgG 1 kappa-FcM-WNV by digestion, wherein M: DNA marker, a: light chain variable region enzyme digestion identification, 1 and 2: recombinant plasmid pFRT-IgG1 kappa-FcM-WNV was digested with BamHI/BsiWII, B: heavy chain variable region cleavage identification, 3 and 4: double digestion of recombinant plasmid pFRT-IgG1 kappa-FcM-WNV XhoI/NheI;
FIG. 2 is an SDS-PAGE electrophoresis of antibodies FcM-WNV and Wt-WNV, wherein M: protein markers, a and b: after reducing SDS-PAGE of FcM-WNV antibody and wild-type antibody Wt-WNV, 1M appears on the lanes r About 50X 10 3 Heavy chain band of KD and 1M r About 25 x 10 3 Light chain bands of KD, c and d: fcM-WNV antibody and wild-type antibody Wt-WNV were subjected to non-reducing SDS-PAGE and expressed as 1 specific antibody band, M r About 150 x 10 3 KD;
FIG. 3 is a graph showing the results of ELISA detection of binding between FcM-WNV and Wt-WNV and WNV EDIII;
FIG. 4 is a graph showing the results of detection of the expression of exogenous hCD64 gene;
FIG. 5 is a graph showing the results of binding assays for antibodies FcM-WNV and Wt-WNV and hCD 64;
FIG. 6 is a graph showing the results of the neutralization activity assays (K562) of antibodies FcM-WNV and Wt-WNV, wherein P <0.0001, P <0.001, and P <0.01;
FIG. 7 is a graph showing the results of the neutralization activity assays (BHK 21) of antibodies FcM-WNV and Wt-WNV, wherein P <0.0001, P <0.001, and P <0.01;
FIG. 8 is a graph showing the results of IC50 detection of antibodies FcM-WNV in K562 cells;
FIG. 9 is a graph showing the results of detection of the antibody FcM-WNV IC50 in BHK21 cells.
Detailed Description
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In order to facilitate an understanding of the present invention, the following terms referred to in the present invention are explained herein:
as used herein, the terms "comprises" or "comprising" are intended to include the stated element or component without excluding other elements or other components.
As used herein, the term "homology" refers to sequence similarity to an amino acid sequence or nucleotide sequence that is to be aligned. "homology" includes amino acid sequences having 75% or more, or 85% or more, or 90% or more, or 95% or more homology with the amino acid sequences provided herein. Homology can be assessed visually or by computer software. Using computer software, the identity between two or more sequences can be expressed in percent (%), which can be used to evaluate homology between related sequences. The 75% or more homology may be 75%, 80%, 85%, 90% or more than 95% homology.
In general, modification of one or more amino acids in a protein does not affect the function of the protein. Those skilled in the art will recognize that individual additions, deletions, insertions, substitutions to a single amino acid or a small percentage of amino acids or to an amino acid sequence are conservative modifications, where a change in protein results in a protein with similar function. Conservative substitution tables providing functionally similar amino acids are well known in the art.
The final derivative or variant may be achieved using substitutions, deletions, insertions or any combination thereof. Typically, these changes are made at several amino acids to minimize molecular changes, particularly the immunogenicity and specificity of antigen binding proteins. However, in some cases greater variation may be tolerated. Amino acid substitutions are typically single base; the insertion will typically be on the order of about one to about twenty amino acid residues, although significantly larger insertions may be tolerated. Deletions range from about one to about twenty amino acid residues, although in some cases the deletions may be much larger.
As used herein, the term "nucleic acid molecule" refers to DNA molecules and RNA molecules. The nucleic acid molecule may be single-stranded or double-stranded, but is preferably double-stranded DNA. A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the coding sequence.
As used herein, the term "expression vector" may be, for example, a cloning vector, a binary vector, or an integrative vector. Expression includes transcription of the nucleic acid molecule, e.g., into translatable mRNA.
Non-limiting examples of carriers include, but are not limited to: pQE-12, pUC-series, pBluescript (Stratagene), pET-series expression vector (Novagen) or pCRTOPO (Invitrogen), λgt11, pJOE, pBBR1-MCS series, pJB861, pBSMuL, pBC2, pUCPKS, pTACT1, pTRE, pCAL-n-EK, pESP-1, pOP13CAT, E-027 pCAG Kosak-Cherry (L45 a) vector system, pREP (Invitrogen), pCEP4 (Invitrogen), pMC1neo (Stratagene). Non-limiting examples of plasmid vectors suitable for Pichia pastoris include, for example, plasmids pAO815, pPIC9K and pPIC3.5K (all Invitrogen). Another vector suitable for expression of proteins in Xenopus (Xenopus) embryos, zebra fish embryos, and a wide variety of mammalian and avian cells is the multipurpose expression vector pCS2+.
In general, vectors may contain one or more origins of replication (ori) and genetic systems for cloning or expression, one or more markers for selection in a host (e.g., antibiotic resistance), and one or more expression cassettes. In addition, the coding sequences contained in the vectors can be linked to transcriptional regulatory elements and/or to other amino acid coding sequences using established methods. Such regulatory sequences are well known to those skilled in the art and include, but are not limited to, regulatory sequences that ensure transcription initiation, internal Ribosome Entry Sites (IRES), and optionally regulatory elements that ensure transcription termination and transcript stabilization. Non-limiting examples of such regulatory elements that ensure transcription initiation include promoters, translation initiation codons, enhancers, insulators, and/or regulatory elements that ensure transcription termination, which are included downstream of the nucleic acid molecules of the invention. Further examples include Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing, nucleotide sequences encoding secretion signals, or signal sequences depending on the expression system used, which are capable of directing the expressed protein to a cellular compartment or culture medium. The vector may also contain additional expressible polynucleotides encoding one or more chaperones to facilitate correct protein folding.
Additional examples of suitable origins of replication include, but are not limited to: full-length ColE1, truncated ColEI, SV40 virus, and M13 origins of replication, while additional examples of suitable promoters include, but are not limited to: cytomegalovirus (CMV) promoter, SV 40-promoter, RSV-promoter (Rous sarcoma virus), lacZ promoter, tetracycline promoter/operator (tetp/o), chicken beta-actin promoter, CAG-promoter (combination of chicken beta-actin promoter and cytomegalovirus immediate early enhancer), gai10 promoter, human elongation factor 1 alpha-promoter, AOX1 promoter, GAL1 promoter CaM-kinase promoter, lac, trp or tac promoter, T7 or T5 promoter, lacUV5 promoter, alfalfa silver vein moth (Autographa californica) polynuclein polyhedrin (AcMNPV) polyhedrin promoter or globin intron in mammalian and other animal cells. An example of an enhancer is, for example, the SV 40-enhancer. Non-limiting additional examples of regulatory elements that ensure transcription termination include SV 40-polyadenylation sites, tk-polyadenylation sites, rho-factor independent lpp terminators or AcMNPV polyhedrin polyadenylation signals. Further non-limiting examples of selectable markers include dhfr, which confers resistance to methotrexate, npt, which confers resistance to the aminoglycosides neomycin, kanamycin and paromycin (paromycin), and hygro, which confers resistance to hygromycin. Additional selection genes have been described, namely trpB, which allow cells to use indole instead of tryptophan; hisD, which allows cells to replace histidine with histidinol (histidinol); mannose 6-phosphate isomerase, which allows cells to utilise mannose and ODC (ornithine decarboxylase), which confers resistance to the ornithine decarboxylase inhibitor 2- (difluoromethyl) -DL-ornithine DFMO or deaminase from aspergillus terreus (Aspergillus terreus) which confers resistance to blasticidin S.
To facilitate purification of the nucleic acid molecules of the invention, tag (tag) sequences may be inserted into the expression vector. Examples of tags include, but are not limited to: six histidine tags, myc tags or FLAG tags. Any tag known to those skilled in the art to facilitate purification may be used in the present invention.
As used herein, the term "therapeutically and/or prophylactically effective amount" refers to an amount of an antibody, pharmaceutical composition, and/or biological agent described herein that is effective to inhibit, prevent, retard, or treat symptoms of a particular disease, disorder, or side-effect. Such diseases, disorders and side effects include, but are not limited to, those pathological conditions associated with West Nile Virus infection (e.g., pathological conditions associated with the treatment and/or prevention of West Nile Virus infection-related diseases), wherein treatment or prevention includes inhibiting the activity of a cell, tissue or receptor, e.g., by contacting it with an antibody, pharmaceutical composition and/or biological agent of the invention. The therapeutically effective amount may vary depending on the route and dosage form of administration, the age and weight of the subject and/or the disease or condition being treated.
As used herein, the term "pharmaceutical composition" or "biological agent" may have any one of the agents selected from the group consisting of: tablets, pills, powders, granules, capsules, suspensions, solutions, emulsions, syrups, sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized formulations and suppositories. Furthermore, one or more administrations may be performed. At this time, the pharmaceutical composition and/or the biological agent is administered in the form of a liquid formulation, powder, aerosol, capsule, vaginal tablet, capsule or suppository. Routes of administration may include, but are not limited to: intraperitoneal, intravenous, intramuscular, subcutaneous, intradermal, oral, topical, intranasal, intrapulmonary, intrarectal, and the like. When administered orally, a coating may be formulated that protects the active ingredient in the pharmaceutical composition from degradation in the stomach. In addition, the active ingredient may be administered by any device capable of being transferred to the target tissue. In particular embodiments, the pharmaceutical compositions provided herein can be formulated into various dosage forms according to actual needs, and the dosage beneficial to the patient can be determined by the clinician based on the type, age, weight and general disease condition of the subject, mode of administration, and the like. The mode of administration may be, for example, injection or any other suitable mode of administration known to those skilled in the art.
As used herein, the term "subject" includes humans and non-human animals. Non-human animals include all vertebrates (e.g., mammals and non-mammals) such as non-human primates (e.g., cynomolgus monkeys), sheep, dogs, cows, chickens, amphibians, and reptiles. In certain embodiments, the "subject" is preferably a human.
The invention is further illustrated below in conjunction with specific examples, which are provided solely to illustrate the invention and are not to be construed as limiting the invention. One of ordinary skill in the art can appreciate that: many changes, modifications, substitutions and variations may be made to the embodiments without departing from the spirit and principles of the invention, the scope of which is defined by the claims and their equivalents. The experimental procedure, in which no specific conditions are noted in the examples below, is generally carried out according to conventional conditions or according to the conditions recommended by the manufacturer.
Example expression and in vitro Activity validation of WNV neutralizing antibodies that reduce the ADE Effect
1. Experimental method
(1) Plasmid construction
According to the point mutation kit of Beijing Tiangen Biochemical technology Co., ltd, the point mutation-L234A and L235A are carried out on 234 and 235 sites of Fc fragment in antibody mammalian cell expression vector pFRT-IgG1 kappa, the mutated plasmid is confirmed by sequencing, and the mutated plasmid is named pFRT-IgG1 kappa-FcM. PCR amplified WNV antibody heavy and light chain variable region sequences. After the PCR products are purified and recovered, the PCR products are respectively digested and sequentially connected to pFT-IgG 1 kappa-FcM to construct recombinant expression plasmids pFT-IgG 1 kappa-FcM-WNV. pFT-IgG 1 kappa-FcM-WNV was identified by digestion and sequencing.
(2) Antibody expression purification
pFT-IgG 1 kappa-FcM-WNV was transfected into mammalian cells, expiCHO, 125 r/min,37℃and 5% CO using an ExpiCHO transfection kit according to kit instructions 2 The cell is cultured by a shaking table,8-10 d, and collecting cell culture supernatant, and purifying Protein A FF Protein column. Eluting with citric acid buffer solution at pH3.0, collecting the effluent, neutralizing with 1 mol/L TRIS-HCL buffer solution at pH8.5, dialyzing with 0.01 mol/L PBS at pH7.2 for 72 h, and filtering with 0.22 μm filter membrane for sterilization. The BCA method detects the concentration of the purified antibody, and SDS-PAGE electrophoresis detects the purity of the antibody.
(3) ELISA test
The coating solution was used to dilute the WNV DIII-Fc fusion protein to 1. Mu.g/mL, 100. Mu.L per well was added to the ELISA plate overnight at 4 ℃; after 3 PBST washes, 5% milk was blocked and incubated at 37 ℃ for 1 h; absorbing and discarding the blocking solution, adding the corresponding antibody diluted by the gradient of the blocking solution, and incubating at 37 ℃ for 1 h, wherein the initial concentration is 1 mug/mL; after PBST is washed for 3 times, goat anti-human (Fab') 2-HRP secondary antibody is added for reaction for 45 min at room temperature; after PBST was washed 3 times, TMB was developed. The absorbance (A450) value at 450 nm was measured after termination of 1 mol/L sulfuric acid.
(4) Flow cytometry detection
293T cells were seeded in 24-well plates (2X 10) 5 Per well), cells were cultured to 80% confluence, and pCMV3-FCGR1-GFPSpark was transfected into 293T cells according to Lipo3000 transfection reagent protocol. 24 Transfected cells were collected by pancreatin digestion after h, incubated with corresponding concentrations of antibody (5. Mu.g/mL) respectively, after washing with FACS wash for 1 pass, antibody label (PE-anti-human kappa chain) was added, after washing with FACS wash for 2 passes, incubated at 4℃in the absence of light for 30 min, resuspended in 1% paraformaldehyde, and flow cytometry detected.
(5) Preparation of WNV pseudoviruses
293T cells were seeded in 6-well plates (1X 10) 6 /well), and the cells were incubated overnight at 37 ℃. After culturing to 80% confluence of cells, prWNV-Rluc was combined with pcDNA3.1-CME at 1 according to Lipo3000 transfection reagent instructions: 1 were co-transfected into 293T cells. 72 After h, the cell culture supernatant was collected. 3000 Centrifuging at 4deg.C for 10 min, filtering the supernatant with 0.45 μm filter membrane, adding 20% serum, and storing in-80deg.C refrigerator.
(6) Infection test
The supernatant of the WNV pseudovirus prepared and stored at-80 ℃ was thawed in an ice bath, 10 2 After dilution by multiple times, the mixture is pre-diluted with the corresponding concentrationIs used, 37 ℃,1 h. The pseudo-virus supernatant + antibody mixture was then used to infect BHK21 or K562 cells, while the non-antibody-added pseudo-virus supernatant was set as a control. 48 After h, the lysed cells were assayed for luciferase activity (RLU). Experiments were repeated 3 times, with 4 duplicate wells per experiment.
(7) Statistical method
Statistical plots were drawn using GraphPad primer 8.3.0 software. The multiple groups are compared by adopting single-factor analysis of variance, the difference is statistically significant for P < 0.05, and the difference is statistically significant for P < 0.01.
2. Experimental results
(1) Construction of recombinant expression vector pFRT-IgG1 kappa-FcM-WNV
The pFT-IgG 1 kappa of antibody mammalian cell expression vector is used as template, the 234 site and 235 site of Fc region are point mutated, both the L site and 235 site are mutated into A, the mutated plasmid is confirmed by sequencing, the mutated plasmid is mutated into GCC, namely Lys-Ala, and the mutated plasmid is named pFT-IgG 1 kappa-FcM. The heavy and light chain variable region sequences of an antibody with significant ADE effect (herein designated Wt-WNV) obtained in the preliminary work of the present invention were cloned into pFRT-IgG1 kappa-FcM to construct a recombinant expression plasmid pFRT-IgG1 kappa-FcM-WNV, cleavage identification showed cleavage of the band of interest at the corresponding molecular weight (see fig. 1), and further confirmation of correct recombinant expression plasmid construction by sequencing. The sequence information of the antibodies Wt-WNV is shown in Table 1 below.
TABLE 1 sequence information for antibodies Wt-WNV
(2) Expression purification of antibody FcM-WNV
The expression plasmid pFRT-IgG1 kappa-FcM-WNV was transiently transfected into the high efficiency expression system ExpiCHO cells, and cell culture supernatants were collected at 8-10 d. Antibody proteins in the culture supernatant were purified using Protein a affinity purification techniques. SDS-PAGE result shows that the purity of the purified FcM-WNV antibody and the unmutated wild-type antibody Wt-WNV protein is higher than 90%, and the purified FcM-WNV antibody and the unmutated wild-type antibody Wt-WNV protein are specific under non-reducing condition Sexual protein band, relative molecular mass (M r ) About 150X 10 3 KD, similar to normal IgG size; under reducing conditions, disulfide bonds between the light and heavy chains of the antibody are opened, and the antibody molecule is reduced to a monomeric form, which is expressed as relative molecular masses of about 25×10, respectively 3 KD and 50X 10 3 Bands of KD (see fig. 2). It was suggested that LALA mutation of the Fc region of the antibody expression system did not affect the expression of the antibody. The amino acid sequences of the heavy chain variable region and the light chain variable region of the antibody FcM-WNV are shown in table 1, the nucleotide sequences of the heavy chain variable region and the light chain variable region of the antibody FcM-WNV are shown in SEQ ID NO. 11 and SEQ ID NO. 12 respectively, and the amino acid sequence and the nucleotide sequence of the Fc segment of the antibody FcM-WNV are shown in SEQ ID NO. 9 and SEQ ID NO. 10 respectively.
Amino acid sequence of Fc segment of antibody FcM-WNV: ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:9);
Nucleotide sequence of the Fc segment of antibody FcM-WNV: GCTAGCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACTGTGCCCTCTAGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAGCAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGTGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAA GCCGCCGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCAGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAAGAGATGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA (SEQ ID NO:10)。
(3) Binding detection of antibodies FcM-WNV to target antigen
Binding of FcM-WNV to the target antigen West Nile Virus E protein DIII was detected by ELISA. As shown in FIG. 3, fcM-WNV recognizes binding to WNV DIII and exhibits a concentration-dependent effect as does Wt-WNV. Whereas the unrelated control antibody did not bind to WNV iii. Indicating that LALA mutation of the Fc region of the antibody expression system did not affect antigen recognition of the expressed antibody.
(4) Binding assays for antibodies FcM-WNV to hFcgammaR I (CD 64)
ADE refers to antibody-mediated effects that enhance viral infection of target cells, and is currently believed to be associated with Fc receptors expressed on the surface of target cells, human Fc receptors include one high affinity IgG receptor hfcyril (CD 64) and two low affinity IgG receptors hfcyrilia, iib and iic (CD 32) and hfcyrilia and iiib (CD 16). Of these, only high affinity hfcyril binds monomeric IgG. Thus, in the following, the present invention compares the binding of antibody FcM-WNV to hCD64 with the unmutated wild-type antibody Wt-WNV. First, the present invention transiently transfects exogenous hCD64 gene into 293T cells, and after flow assay determines that the transfected 293T cells express hCD64 on the surface (see FIG. 4), immunofluorescence assay FcM-WNV and Wt-WNV bind to 293T-hCD64 cells. As shown in FIG. 5, both antibodies FcM-WNV and Wt-WNV can bind to hCD64, but compared with the unmutated wild-type antibody Wt-WNV, the binding force between the antibody FcM-WNV obtained by the expression of the antibody expression system after the mutation of the Fc region and hCD64 is obviously reduced, and the fluorescence intensity is obviously reduced (the average fluorescence intensity is reduced from 495 to 48).
(5) Neutralization assay (BHK 21 and K562 cells)
The antibody Wt-WNV is an antibody with obvious ADE effect obtained by the inventor of the invention in the screening work of a prophase phage antibody library, repeated experiments show that, consistent with the prophase experimental result, the antibody Wt-WNV has the effect of obviously enhancing the WNV pseudovirus infection K562 at the concentration of 5 mug/mL, and the RLU value of the intracellular reporter gene Renilla luciferase of the K562 cells after the infection of the antibody Wt-WNV-pseudovirus supernatant mixture is obviously increased compared with the control without the antibody (see figure 6); at the same concentration, however, 5. Mu.g/mL of antibody Wt-WNV did not completely block pseudovirus infection of BHK21 cells (see FIG. 7). While the antibody FcM-WNV obtained by expression of the antibody expression system after LALA mutation in the Fc region established by the invention can almost completely block pseudovirus infection of target cells at a concentration of 5 mug/mL, whether K562 or BHK21 (see FIG. 6 and FIG. 7), and the IC50 is 0.1571 mug/mL and 0.6304 mug/mL respectively (see FIG. 8 and FIG. 9). It was shown that antibodies FcM-WNV obtained by expression after mutation are capable of reversing the ADE effect by attenuating binding to FcgammaR, showing a neutralizing effect, compared to the non-mutated wild-type antibody Wt-WNV.

Claims (10)

1. An antibody for resisting west nile virus, characterized in that the amino acid sequences of HCDR1, HCDR2 and HCDR3 in the heavy chain variable region of the antibody are CDR1, CDR2 and CDR3 in the heavy chain variable region as shown in SEQ ID NO. 4;
the amino acid sequences of LCDR1, LCDR2 and LCDR3 in the light chain variable region of the antibody are CDR1, CDR2 and CDR3 in the light chain variable region shown in SEQ ID NO. 8.
2. The antibody of claim 1, wherein the amino acid sequences of HCDR1, HCDR2, and HCDR3 are respectively as shown in SEQ ID No. 1, SEQ ID No. 2, and SEQ ID No. 3 or are respectively amino acid sequences having at least 90% homology with the amino acid sequences corresponding to SEQ ID No. 1, SEQ ID No. 2, and SEQ ID No. 3;
the amino acid sequences of LCDR1, LCDR2 and LCDR3 are respectively shown as SEQ ID NO. 5, SEQ ID NO. 6 and SEQ ID NO. 7 or are respectively the amino acid sequences with at least 90% homology with the amino acid sequences corresponding to SEQ ID NO. 5, SEQ ID NO. 6 and SEQ ID NO. 7.
3. An anti-west nile virus neutralizing antibody that reduces the ADE effect, wherein HCDR1, HCDR2, HCDR3 in the heavy chain variable region of the antibody is HCDR1, HCDR2, HCDR3 as defined in claim 1 or 2;
The LCDR1, LCDR2, LCDR3 in the light chain variable region of the antibody is LCDR1, LCDR2, LCDR3 as recited in claim 1 or 2;
the amino acid sequence of the Fc segment of the antibody is shown as SEQ ID NO. 9 or the amino acid sequence with at least 90% homology with the amino acid sequence corresponding to SEQ ID NO. 9.
4. A nucleic acid molecule encoding the antibody of claim 1 or 2 or the antibody of claim 3.
5. An expression vector comprising the nucleic acid molecule of claim 4.
6. An engineered host cell comprising the expression vector of claim 5.
7. A recombinant expression vector, wherein the recombinant expression vector is pFRT-IgG1 kappa-FcM-WNV;
the recombinant expression vector is obtained by taking an antibody mammalian cell expression vector pFT-IgG 1 kappa as a template and mutating 234 and 235 sites of an Fc segment from L to A.
8. Any one of the following, including:
(1) An antibody derivative, wherein the antibody derivative is obtained by coupling the antibody of claim 1 or 2 or the antibody of claim 3 to a functional agent;
(2) A pharmaceutical composition comprising the antibody of claim 1 or 2, the antibody of claim 3, the nucleic acid molecule of claim 4, the expression vector of claim 5, the engineered host cell of claim 6, and/or the antibody derivative;
(3) A detection reagent comprising the antibody of claim 1 or 2, the antibody of claim 3, the nucleic acid molecule of claim 4, the expression vector of claim 5, the engineered host cell of claim 6, and/or the antibody derivative;
(4) A test kit, characterized in that the test kit comprises the test reagent.
9. A method of any one of the following, the method comprising:
(1) A method of making the antibody of claim 1 or 2 or the antibody of claim 3, comprising the steps of: culturing the engineered host cell of claim 6, and isolating and purifying the antibody of claim 1 or 2 or the antibody of claim 3 from the host cell culture product;
(2) A method of making the engineered host cell of claim 6, comprising the steps of: introducing the nucleic acid molecule of claim 4 or the expression vector of claim 5 into a host cell;
(3) A method for detecting west nile virus and/or west nile virus E protein for non-diagnostic and non-therapeutic purposes, comprising the steps of: contacting a sample to be tested with the antibody of claim 1 or 2, the antibody of claim 3, the antibody derivative of claim 8, and detecting the formation of an immune complex of west nile virus and/or west nile virus E protein and the antibody;
(4) A method of inhibiting west nile virus and/or west nile virus E protein activity in vitro, comprising the steps of: introducing the antibody of claim 1 or 2, the antibody of claim 3, the nucleic acid molecule of claim 4 and/or the expression vector of claim 5 into a cell of an organism, inhibiting the activity of west nile virus and/or west nile virus E protein by expressing the antibody of claim 1 or 2 or the antibody of claim 3;
(5) A method of inactivating west nile virus and/or west nile virus E protein, comprising the steps of: contacting a sample of interest with the antibody of claim 1 or 2, the antibody of claim 3, the antibody derivative of claim 8.
10. An application of any one of the following, the application comprising:
(1) Use of an antibody according to claim 1 or 2 and/or an antibody according to claim 3 for the preparation of an antibody derivative for the detection of west nile virus and/or west nile virus E protein, for the treatment and/or prophylaxis of diseases associated with west nile virus infection;
(2) Use of an antibody according to claim 1 or 2, an antibody according to claim 3, a nucleic acid molecule according to claim 4, an expression vector according to claim 5, an engineered host cell according to claim 6 and/or an antibody derivative according to claim 8 for the preparation of a detection reagent for the detection of west nile virus and/or west nile virus E protein;
(3) Use of an antibody according to claim 1 or 2, an antibody according to claim 3, a nucleic acid molecule according to claim 4, an expression vector according to claim 5, an engineered host cell according to claim 6, an antibody derivative according to claim 8 and/or a detection reagent according to claim 8 for the preparation of a detection kit for the detection of west nile virus and/or west nile virus E protein;
(4) Use of an antibody according to claim 1 or 2, an antibody according to claim 3, a nucleic acid molecule according to claim 4, an expression vector according to claim 5, an engineered host cell according to claim 6, an antibody derivative according to claim 8, a detection reagent according to claim 8 and/or a detection kit according to claim 8 for the preparation of a product for the diagnosis and/or co-diagnosis of diseases associated with west nile virus infection;
(5) Use of the antibody of claim 1 or 2, the antibody of claim 3, the nucleic acid molecule of claim 4, the expression vector of claim 5, the engineered host cell of claim 6, the antibody derivative of claim 8, the detection reagent of claim 8 and/or the detection kit of claim 8 for the detection of west nile virus and/or west nile virus E protein for non-diagnostic purposes;
(6) Use of an antibody according to claim 1 or 2, an antibody according to claim 3, a nucleic acid molecule according to claim 4 and/or an expression vector according to claim 5 for the preparation of an engineered host cell for the treatment and/or prevention of diseases associated with west nile virus infection;
(7) Use of an antibody according to claim 1 or 2, an antibody according to claim 3, a nucleic acid molecule according to claim 4, an expression vector according to claim 5, an engineered host cell according to claim 6 and/or an antibody derivative according to claim 8 for the preparation of a medicament for the treatment and/or prophylaxis of diseases associated with west nile virus infection;
(8) Use of an antibody according to claim 1 or 2, an antibody according to claim 3, a nucleic acid molecule according to claim 4, an expression vector according to claim 5, an engineered host cell according to claim 6, an antibody derivative according to claim 8 and/or a pharmaceutical composition according to claim 8 for the preparation of a biological agent for the treatment and/or prevention of diseases associated with west nile virus infection;
(9) Use of the recombinant expression vector of claim 7 for the preparation of an antibody that reduces the ADE effect.
CN202310967602.3A 2023-08-03 2023-08-03 anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof Active CN116751289B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310967602.3A CN116751289B (en) 2023-08-03 2023-08-03 anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310967602.3A CN116751289B (en) 2023-08-03 2023-08-03 anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof

Publications (2)

Publication Number Publication Date
CN116751289A true CN116751289A (en) 2023-09-15
CN116751289B CN116751289B (en) 2023-10-20

Family

ID=87955408

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310967602.3A Active CN116751289B (en) 2023-08-03 2023-08-03 anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof

Country Status (1)

Country Link
CN (1) CN116751289B (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2571404A1 (en) * 2004-06-21 2005-12-29 Washington University Antibodies against west nile virus and therapeutic and prophylactic uses thereof
CA2661042A1 (en) * 2006-08-18 2008-02-21 Armagen Technologies, Inc. Agents for blood-brain barrier delivery
CN112159470A (en) * 2020-10-09 2021-01-01 中国人民解放军军事科学院军事医学研究院 anti-WNV (WNV) infection binding molecule
CN115667506A (en) * 2021-02-10 2023-01-31 西雅图免疫公司 Recombinant ACE2-FC fusion molecules and methods of making and using the same

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2571404A1 (en) * 2004-06-21 2005-12-29 Washington University Antibodies against west nile virus and therapeutic and prophylactic uses thereof
CA2661042A1 (en) * 2006-08-18 2008-02-21 Armagen Technologies, Inc. Agents for blood-brain barrier delivery
CN112159470A (en) * 2020-10-09 2021-01-01 中国人民解放军军事科学院军事医学研究院 anti-WNV (WNV) infection binding molecule
CN115667506A (en) * 2021-02-10 2023-01-31 西雅图免疫公司 Recombinant ACE2-FC fusion molecules and methods of making and using the same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
OLIPHANT, T.等: "anti-West Nile virus monoclonal antibody heavy chain variable region, partial [Mus musculus]", GENBANK: AAY85980.1 *
OLIPHANT, T.等: "anti-West Nile virus monoclonal antibody light chain variable region, partial [Mus musculus]", GENBANK: AAY85981.1 *

Also Published As

Publication number Publication date
CN116751289B (en) 2023-10-20

Similar Documents

Publication Publication Date Title
CN107827990A (en) A kind of polypeptide, the nucleic acid for encoding it, the T lymphocytes of its modification and its application
JP6448554B2 (en) Methods for preventing and treating multiple sclerosis
CN115710311A (en) Antibodies or antigen-binding fragments thereof to coronaviruses
EA028433B1 (en) Antibody that binds to influenza b viruses and use thereof
Wu et al. Development of monoclonal antibodies against serum immunoglobulins from gibel carp (Carassius auratus gibelio) and their applications in serodiagnosis of inapparent infection and evaluation of vaccination strategies
CA2965327C (en) Vh4 antibodies against gray matter neuron and astrocyte
CN116751289B (en) anti-West Nile virus neutralizing antibody for reducing ADE effect and application thereof
CN116444661A (en) Neutralizing antibody P186-1H2 for broad spectrum neutralization of SARS-CoV-2 and application thereof
CN112159470B (en) anti-WNV (WNV) infection binding molecule
CN115286715A (en) CD 3-resistant nano antibody or antigen binding part thereof and preparation method thereof
CN109503712B (en) Monoclonal antibody ZKns2E11 and application thereof
CN109535249B (en) Monoclonal antibody ZKns3G2 and application thereof
CN109553680B (en) Monoclonal antibody ZKns4B8 and application thereof
CN117247451B (en) Single-domain antibody for human interleukin 6 protein and application thereof
KR100449906B1 (en) A diagnostic method of foot-and-mouth disease using recombinant FMDV 2C non-structural protein and monoclonal antibody
CN110655572A (en) Monoclonal antibody for resisting filovirus GP protein and application thereof
CN117106080B (en) Antibodies of human origin against yersinia pestis LcrV and related products and uses thereof
CN117343170B (en) anti-LcrV antibody for detecting Yersinia pestis and application thereof
CN116003582B (en) Antibody for detecting coronavirus and application thereof
US20070003551A1 (en) Immunoglobulin
US20230072640A1 (en) Human monoclonal antibodies against yellow fever virus and uses therefor
WO2021227138A1 (en) Monoclonal antibody for novel coronavirus, or antigen binding part thereof
Smith et al. Use of poxvirus display to select antibodies specific for complex membrane antigens
CN115960218A (en) Antibody for resisting novel coronavirus and application thereof
CN116178533A (en) Neutralizing antibody P5S-1D8 for broad spectrum neutralization of SARS-CoV-2 and application thereof

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
GR01 Patent grant
GR01 Patent grant