CN111153988A - Broad-spectrum neutralizing monoclonal antibody against enterovirus D68 - Google Patents

Broad-spectrum neutralizing monoclonal antibody against enterovirus D68 Download PDF

Info

Publication number
CN111153988A
CN111153988A CN201811326695.7A CN201811326695A CN111153988A CN 111153988 A CN111153988 A CN 111153988A CN 201811326695 A CN201811326695 A CN 201811326695A CN 111153988 A CN111153988 A CN 111153988A
Authority
CN
China
Prior art keywords
seq
ser
antibody
variable region
heavy chain
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
CN201811326695.7A
Other languages
Chinese (zh)
Other versions
CN111153988B (en
Inventor
黄忠
张超
代文龙
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.)
Shanghai Institute of Immunology and Infection, Chinese Academy of Sciences
Original Assignee
Institut Pasteur of Shanghai of CAS
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 Institut Pasteur of Shanghai of CAS filed Critical Institut Pasteur of Shanghai of CAS
Priority to CN201811326695.7A priority Critical patent/CN111153988B/en
Publication of CN111153988A publication Critical patent/CN111153988A/en
Application granted granted Critical
Publication of CN111153988B publication Critical patent/CN111153988B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C07K16/1009Picornaviridae, e.g. hepatitis A 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
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention discloses a neutralizing monoclonal antibody for resisting enterovirus D68. Specifically, the invention discloses two monoclonal antibodies aiming at enterovirus D68, and the antibodies have obvious binding and neutralizing activities on enterovirus D68.

Description

Broad-spectrum neutralizing monoclonal antibody against enterovirus D68
Technical Field
The invention belongs to the field of biological medicines, and particularly relates to two neutralizing monoclonal antibodies for resisting enterovirus D68.
Background
Enterovirus D68(EVD68) is a small non-enveloped virus belonging to the D family of enteroviruses within the picornaviridae family. Similar to other picornaviruses, it has an icosahedral capsid of-30 nm, consisting of 60 protomers, each composed of four subunit proteins, VP1, VP2, VP3 and VP 4. Based on the evolutionary analysis of VP1 nucleic acid sequences, the circulating EVD68 strain can be divided into three major evolutionary lineages, namely A, B and C. EVD68 is increasingly associated with acute respiratory and/or severe neurological diseases in children, primarily acute relaxant myelitis (AFM). Over the past decade, EVD68 has spread widely throughout the world, continuously resulting in outbreaks and sporadic cases, which pose a serious threat to global public health.
To date, there is no commercial vaccine or antiviral drug against EVD68 infection. Recently, we have reported that recombinant virus-like particles (VLPs) of EVD68 can be produced in baculovirus/insect cells and pichia pastoris expression systems, respectively, and that VLPs induce high levels of neutralizing antibodies in mice and protect mice against lethal infection. Monoclonal antibodies (mabs) are a promising approach to the treatment of infectious diseases, as demonstrated by the successful commercialization of the humanized monoclonal antibody palivizumab against respiratory syncytial virus.
Therefore, there is a need in the art to develop therapeutic neutralizing antibodies against enterovirus type D68 with good affinity to address the need for prevention and control of EVD 68.
Disclosure of Invention
The invention aims to provide a neutralizing monoclonal antibody of enterovirus D68 and application thereof.
In a first aspect of the invention, there is provided a heavy chain variable region of an antibody, said heavy chain variable region having complementarity determining regions CDRs selected from the group consisting of:
VH-CDR1 shown in SEQ ID NO. 2,
VH-CDR2 shown in SEQ ID NO. 4,
VH-CDR3 shown in SEQ ID NO 6,
VH-CDR1 shown in SEQ ID NO:22,
VH-CDR1 shown in SEQ ID NO:24, and
VH-CDR1 shown in SEQ ID NO. 26;
preferably, the heavy chain variable region has the amino acid sequence shown in SEQ ID NO 16 and/or SEQ ID NO 36.
In a second aspect of the invention, there is provided a heavy chain of an antibody, said heavy chain having a heavy chain variable region and a heavy chain constant region as described in the first aspect of the invention.
In another preferred embodiment, the constant region of the heavy chain is of human or murine origin.
In another preferred embodiment, the heavy chain has the amino acid sequence shown in SEQ ID NO. 15 and/or SEQ ID NO. 35.
In a third aspect of the present invention, there is provided a light chain variable region of an antibody, said light chain variable region having Complementarity Determining Regions (CDRs) selected from the group consisting of:
VL-CDR1 shown in SEQ ID NO. 8,
VL-CDR2 shown in SEQ ID NO. 10,
VL-CDR3 shown in SEQ ID NO. 12;
VL-CDR1 shown in SEQ ID NO 28,
VL-CDR2 shown in SEQ ID NO. 30, and
VL-CDR3 shown in SEQ ID NO. 32;
preferably, the light chain variable region has the amino acid sequence shown in SEQ ID NO. 20 and/or SEQ ID NO. 40.
In a fourth aspect of the invention, there is provided a light chain of an antibody, said light chain having a light chain variable region and a light chain constant region as described in the third aspect of the invention.
In another preferred embodiment, the light chain constant region is of human or murine origin.
In another preferred embodiment, the light chain has the amino acid sequence shown in SEQ ID NO 19 and/or SEQ ID NO 39.
In a fifth aspect of the invention, there is provided an antibody having:
(1) a heavy chain variable region according to the first aspect of the invention; and/or
(2) A light chain variable region according to the third aspect of the invention.
In another preferred embodiment, the antibody has: a heavy chain according to the second aspect of the invention; and/or a light chain according to the fourth aspect of the invention.
In another preferred embodiment, the antibody is specific anti-enterovirus D68 antibody.
In another preferred embodiment, the antibody comprises: a single chain antibody (scFv), a diabody, a monoclonal antibody, a chimeric antibody (e.g., a human-murine chimeric antibody), a murine antibody, or a humanized antibody.
In a sixth aspect of the present invention, there is provided a recombinant protein having:
(i) the sequence of a heavy chain variable region according to the first aspect of the invention, the sequence of a heavy chain according to the second aspect of the invention, the sequence of a light chain variable region according to the third aspect of the invention, the sequence of a light chain according to the fourth aspect of the invention, or the sequence of an antibody according to the fifth aspect of the invention; and
(ii) optionally a tag sequence to facilitate expression and/or purification.
In another preferred embodiment, the tag sequence is selected from the group consisting of: 6 × His tag, GGGS sequence, FLAG tag.
In another preferred embodiment, the recombinant protein specifically binds enterovirus type D68.
In a seventh aspect of the invention, there is provided a polynucleotide encoding a polypeptide selected from the group consisting of:
(1) a heavy chain variable region according to the first aspect of the invention, a heavy chain according to the second aspect of the invention, a light chain variable region according to the third aspect of the invention, a light chain according to the fourth aspect of the invention, or an antibody according to the fifth aspect of the invention; or
(2) A recombinant protein according to the sixth aspect of the invention.
In another preferred embodiment, the polynucleotide has the sequence shown in SEQ ID No.1, 3, 5, 7, 9, 11, 13, 14, 17, 18, 21, 23, 25, 27, 29, 31, 33, 34, 37 or 38.
In an eighth aspect of the invention, there is provided a vector comprising a polynucleotide according to the seventh aspect of the invention.
In another preferred embodiment, the carrier comprises: bacterial plasmids, bacteriophages, yeast plasmids, plant cell viruses, mammalian cell viruses such as adenoviruses, retroviruses, or other vectors.
According to a ninth aspect of the invention, there is provided a genetically engineered host cell comprising a vector or genome according to the eighth aspect of the invention into which has been integrated a polynucleotide according to the seventh aspect of the invention.
In a tenth aspect of the invention, there is provided an immunoconjugate comprising:
(a) a heavy chain variable region according to the first aspect of the invention, a heavy chain according to the second aspect of the invention, a light chain variable region according to the third aspect of the invention, a light chain according to the fourth aspect of the invention, or an antibody according to the fifth aspect of the invention, or a recombinant protein according to the sixth aspect of the invention; and
(b) a coupling moiety selected from the group consisting of: a detectable label, a drug, a toxin, a cytokine, a radionuclide, or an enzyme.
In another preferred embodiment, the conjugate is selected from the group consisting of: fluorescent or luminescent labels, radioactive labels, MRI (magnetic resonance imaging) or CT (computed tomography) contrast agents, or enzymes capable of producing detectable products, radionuclides, biotoxins, cytokines (e.g., IL-2, etc.), antibodies, antibody Fc fragments, antibody scFv fragments, gold nanoparticles/nanorods, viral particles, liposomes, nanomagnetic particles, prodrug-activating enzymes (e.g., DT-diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)), chemotherapeutic agents (e.g., cisplatin), or any form of nanoparticles, and the like.
In an eleventh aspect of the present invention, there is provided a pharmaceutical composition comprising:
(i) a heavy chain variable region according to the first aspect of the invention, a heavy chain according to the second aspect of the invention, a light chain variable region according to the third aspect of the invention, a light chain according to the fourth aspect of the invention, or an antibody according to the fifth aspect of the invention, or a recombinant protein according to the sixth aspect of the invention, or an immunoconjugate according to the tenth aspect of the invention; and
(ii) a pharmaceutically acceptable carrier.
In another preferred embodiment, the pharmaceutical composition is in the form of injection.
In another preferred embodiment, the pharmaceutical composition is used for preparing a medicament for treating enterovirus D68 type infection.
In a twelfth aspect of the invention, there is provided a use of the heavy chain variable region according to the first aspect of the invention, the heavy chain according to the second aspect of the invention, the light chain variable region according to the third aspect of the invention, the light chain according to the fourth aspect of the invention, the antibody according to the fifth aspect of the invention, the recombinant protein according to the sixth aspect of the invention, or the immunoconjugate according to the tenth aspect of the invention for the manufacture of a medicament, a reagent, a detection plate or a kit.
In another preferred embodiment, the reagent, test plate or kit is used for the detection of enterovirus type D68.
In another preferred embodiment, the medicament is for the treatment or prevention of an enterovirus type D68 infection.
In another preferred embodiment, the reagent comprises a chip and immune microparticles coated with antibodies.
In a thirteenth aspect of the present invention, there is provided a method for detecting enterovirus type D68 in a sample, the method comprising the steps of:
(1) contacting the sample with an antibody according to the fifth aspect of the invention;
(2) detecting the formation of an antigen-antibody complex, wherein the formation of the complex is indicative of the presence of enterovirus type D68 in the sample.
In a fourteenth aspect of the present invention, there is provided a method for producing a recombinant polypeptide, the method comprising:
(a) culturing a host cell according to the ninth aspect of the invention under conditions suitable for expression;
(b) isolating a recombinant polypeptide from the culture, said recombinant polypeptide being an antibody according to the fifth aspect of the invention or a recombinant protein according to the sixth aspect of the invention.
In a fifteenth aspect of the present invention, there is provided a pharmaceutical combination comprising:
(i) a first active ingredient comprising an antibody 1 according to the fifth aspect of the invention, or a recombinant protein according to the sixth aspect of the invention, or an antibody conjugate according to the tenth aspect of the invention, or a pharmaceutical composition according to the eleventh aspect of the invention, or a combination thereof;
(ii) a second active ingredient comprising a second antibody, or an antiviral drug.
In another preferred embodiment, the second antibody is another anti-EVD 68 antibody (e.g., a 6-1).
In another preferred embodiment, the antiviral drug is an anti-EVD 68 viral drug (e.g., pleconaril).
In a sixteenth aspect of the invention there is provided the use of an antibody according to the fifth aspect of the invention, or a recombinant protein according to the sixth aspect of the invention, or an antibody conjugate according to the tenth aspect of the invention, or a pharmaceutical composition according to the eleventh aspect of the invention, in combination with a second antibody or an antiviral medicament, in the manufacture of a medicament for the treatment of a disease associated with infection with EVD 68.
In another preferred embodiment, the second antibody is another anti-EVD 68 antibody (e.g., a 6-1).
In another preferred embodiment, the antiviral drug is an anti-EVD 68 viral drug (e.g., pleconaril).
In a seventeenth aspect of the invention, there is provided a method of treating a disease associated with infection by EVD68 by administering to a subject in need thereof an effective amount of an antibody according to the fifth aspect of the invention, or a recombinant protein according to the sixth aspect of the invention, or an antibody conjugate according to the tenth aspect of the invention, or a pharmaceutical composition according to the eleventh aspect of the invention, or a combination thereof.
In another preferred example, the disease associated with EVD68 infection includes EVD68 single virus infection and mixed infection.
In another preferred example, the mixed infection comprises EVD68 and EV71 mixed infection.
In another preferred example, the method further comprises: administering to the subject a safe and effective amount of a second antibody before, during and/or after administration of the first active ingredient.
In another preferred embodiment, the second antibody is another anti-EVD 68 antibody (e.g., A6-1).
In another preferred example, the method further comprises: administering to the subject a safe and effective amount of an anti-EVD 68 virus drug before, during, and/or after administration of the first active ingredient.
In another preferred embodiment, the antiviral drug is an anti-EVD 68 viral drug (e.g., pleconaril).
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1 shows the neutralizing activity of monoclonal antibodies against EVD 68. 100TCID50 of EVD68 strain US/MO/14-18947 with two-fold serial dilutions of purified antibodies 2G10, 2H12, 4C11 and 8F12, respectively, were incubated for 1 hour and then added to RD cells. Cell viability was determined 3 days post infection by CellTiter-Glo 2.0 kit and half maximal inhibitory concentration (IC50) was calculated for each antibody using GraphPad Prism software. ZIKV-specific mab 1C11 and HCV-specific mab 1F4 were used as isotype controls. Data are presented as mean ± standard error of mean of triplicate wells.
FIG. 2 shows the specificity and affinity of monoclonal antibody for EVD68 antigen binding. (A-C) the reactivity of the mAbs (2G10, 2H12, 4C11 and 8F12) to EVD68VLP (A), EV71VLP (B) and CVA16VLP (C) was determined by ELISA. mAbs 1C11 and 1F were used as isotype controls. anti-EV 71 mAb D5 and anti-CVA 16 mAb 9B5 were used as positive controls in the detection of EV71VLP and CVA16VLP, respectively. (D and E) BLI determination of the binding affinity of mAbs to EVD68VLP (D) and EVD68US/MO/14-18947 virus (E). Binding affinity at steady state is expressed as the equilibrium dissociation constant (KD).
FIG. 3 shows an alignment of the complete amino acid sequences of the variable regions of EVD68 mab. The coding sequences for the variable regions of the antibody heavy and light chains were determined by 5' RACE, respectively. (A and B) heavy chain variable region of 2G10, 2H12, 4C11 and 8F12 antibodies (V)H) (A) and light chain variable region (V)L) The amino acid sequence of (B). The positions of Complementarity Determining Regions (CDRs) were identified and marked using IgBLAST tools. Dots represent amino acid residues identical to those of the variable region of mAb 2G10, and red lines represent amino acid deletions compared to mAb 2G 10.
Figure 4 shows the prophylactic efficacy of mabs 2H12 and 8F12 on EVD68 infection in neonatal mice. Groups of ICR mice (age < 24H; n-12-14/group) were intraperitoneally injected with PBS, 10. mu.g/g of 2H12, 8F12, or isotype control mab (1C11 or 1F4) and infected with US/MO/14-18947 one day later. Infected mice were monitored daily for (a) survival and (B) clinical symptoms. Clinical symptoms were graded as follows: 0, health; 1, lethargy and bradykinesia; 2, weakness of limbs; 3, paralysis of limbs; and 4, death. Survival of mice in each antibody-treated group was compared to PBS control group. Statistical significance was as follows: ns, no significant difference, P is more than or equal to 0.05; p < 0.01; p < 0.001.
Figure 5 shows the efficacy of mabs in treating EVD68 infection in mice. One day large ICR mice (n-13-27/group) were infected with US/MO/14-18947(a-D) or US/KY/14-18953(E and F). Mice were given PBS, 10. mu.g/g of 2H12, 10. mu.g/g of 8F12, or a cocktail of the two mabs (10. mu.g/g of each mab) 1 day (A and B) or 3 days (C to F) post-infection. Survival and (B, D and F) clinical symptoms were then monitored daily (A, C and E). Clinical symptoms were graded as follows: 0, health; 1, lethargy and bradykinesia; 2, weakness of limbs; 3, paralysis of limbs; and 4, death. The survival of the antibody-treated mice was compared to mice in the PBS control group. Statistical significance was as follows: ns, no significant difference, P is more than or equal to 0.05; p < 0.05; p < 0.01; p < 0.001.
Detailed Description
The present inventors have made extensive and intensive studies to unexpectedly obtain two monoclonal antibodies against enterovirus type D68(EVD 68). Specifically, the inventors prepared mabs with strong neutralizing activity against EVD68 and evaluated their therapeutic efficacy in EVD68 infected neonatal mice. The results show that 2 neutralizing antibodies against EVD68 (designated BSD01 and BSD02) with significant binding activity against enterovirus type D68 were prepared from EVD68VLP immunized mice. In addition, the combination of mab BSD01 and BSD02 can provide effective broad-spectrum neutralization and protection against each tested strain of EVD 68. The monoclonal antibodies BSD01 and BSD02 have the functions of preventing and treating EVD68 infection. On the basis of this, the present invention has been completed.
Before the present invention is described, it is to be understood that this invention is not limited to the particular methodology and experimental conditions described, as such methodologies and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
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. As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now exemplified.
Enterovirus D68
In the past 10 years, the prevalence of enterovirus type D68 (enterovirus D68, EVD68) has become more and more prevalent around the world and has been linked to severe respiratory infections, acute delayed myelitis (AFM), and there is an urgent need to develop effective antiviral drugs.
EVD68 was first isolated in 1962 in four pediatric patients with pneumonia, bronchitis in the United states. Since then, EVD68 infection has only occurred in sporadic worldwide. For example, there are only 26 reports in the united states during the 36 th year from 1970 to 2005. However, during the last 10 years, the number of clinical cases of EVD68 infection has increased dramatically worldwide. In 2014, the united states reported the largest outbreak of EVD68 infection with a history of 1153 total infections from 8 months in 2014 to 1 month in 2015, with a large number of patients requiring treatment in intensive care units and 14 patients dying. This number of infections was significantly underestimated because most clinical laboratories at that time lacked a rapid molecular detection method specific for EVD 68. This outbreak quickly passed to canada, europe, and asia, with over 2000 cases of EVD68 infection reported in 20 countries in 2014. After the major outbreak in 2014, EVD68 continued to be popular in many countries in america, asia, and europe. These epidemiological investigations suggest that EVD68 is becoming an important pathogen for severe respiratory infections. In addition, 120 pediatric patients identified acute delayed paralysis (AFM) during the us outbreak of EVD68 in 2014, the major clinical features of which include muscle weakness and paralysis, with spinal motor neuron damage. This AFM outbreak suggested an association between EVD68 infection and AFM, a hypothesis further confirmed by many subsequent clinical cases and epidemiological investigations. Based on the evolutionary analysis of VP1, EVD68 can be divided into three lineages (clade) A, B and C, and the original Fermon line. Pedigree B can further be divided into 3 sublines B1, B2 and B3. In recent years, the EVD68 infection is reported more and more in China, and severe pneumonia, severe nervous system complications and death cases are reported. Wang Jianwei and his team investigated the seropositivity rate of neutralizing antibodies between the years beijing 2004 and 2011, who found that neutralizing antibody titers at EVD68 were generally lower in 2004 but significantly higher in 2009 for all age groups in the sampled population. Neutralizing antibodies against EVD68 increased significantly over time from 2007 to 2011. These findings indicate that EVD68 has spread widely in the chinese population in recent years. Beijing reported severe cases of pneumonia associated with EVD68 in 2014. The first death cases related to EVD68 infection in China were reported in hong Kong in the same year. Taiwan detected the first acute delayed paralysis cases related to EVD68 infection in 2016 in 8 months in our country. Since our country has not incorporated EVD68 into the national legislated infectious disease monitoring system, the public health burden due to EVD68 infection is underestimated. Indeed, EVD68 has become a significant threat to public health, particularly in children. However, there is currently no effective vaccine or drug to prevent or treat EVD68 infection. There is an urgent need to develop corresponding vaccines and drugs to cope with the possible future epidemic of EVD 68. Therefore, screening of neutralizing antibodies against enterovirus D68 is of great significance for the prevention and control thereof.
BSD01 and BSD02
As used herein, the term "BSD 01" is used interchangeably with "2H 12", 2G10 ".
As used herein, the term "BSD 02" is used interchangeably with "8F 12", "4C 11".
Antibodies
As used herein, the term "antibody" or "immunoglobulin" is an heterotetrameric glycan protein of about 150000 daltons with the same structural features, consisting of two identical light chains (L) and two identical heavy chains (H). Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide bonds varies between heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bonds. Each heavy chain has at one end a variable region (VH) followed by a plurality of constant regions. Each light chain has a variable domain (VL) at one end and a constant domain at the other end; the constant region of the light chain is opposite the first constant region of the heavy chain, and the variable region of the light chain is opposite the variable region of the heavy chain. Particular amino acid residues form the interface between the variable regions of the light and heavy chains.
As used herein, the term "variable" means that certain portions of the variable regions in an antibody differ in sequence, which form the binding and specificity of each particular antibody for its particular antigen, however, the variability is not evenly distributed throughout the antibody variable region it is concentrated in three segments called Complementarity Determining Regions (CDRs) or hypervariable regions in the light and heavy chain variable regions the more conserved portions of the variable regions are called Framework Regions (FRs). The variable regions of the native heavy and light chains each contain four FR regions, roughly in a β -fold configuration, connected by three CDRs forming a connecting loop, and in some cases may form a partial β fold structure.
The "light chains" of vertebrate antibodies (immunoglobulins) can be classified into one of two distinct classes (termed kappa and lambda) according to the amino acid sequence of their constant regions of the heavy chains, the immunoglobulins can be classified into different classes.5 classes of immunoglobulins, IgA, IgD, IgE, IgG and IgM, some of which can be further classified into subclasses (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA and IgA 2. the constant regions of the heavy chains corresponding to different classes of immunoglobulins are respectively termed α, delta, epsilon, gamma, and mu. the subunit structures and three-dimensional configurations of the different classes of immunoglobulins are well known to those skilled in the art.
As used herein, the term "monoclonal antibody (mab)" refers to an antibody obtained from a substantially homogeneous population, i.e., the individual antibodies contained in the population are identical, except for a few naturally occurring mutations that may be present. Monoclonal antibodies are directed against a single antigenic site with high specificity. Moreover, unlike conventional polyclonal antibody preparations (typically having different antibodies directed against different determinants), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, monoclonal antibodies are also advantageous in that they are synthesized by hybridoma culture and are not contaminated with other immunoglobulins. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
The invention also comprises a monoclonal antibody with the corresponding amino acid sequence of the anti-enterovirus D68 monoclonal antibody, a monoclonal antibody with the variable region chain of the anti-enterovirus D68 monoclonal antibody, and other proteins or protein conjugates and fusion expression products with the chains. Specifically, the invention includes any protein or protein conjugate and fusion expression product (i.e., immunoconjugate and fusion expression product) having light and heavy chains with hypervariable regions (complementarity determining regions, CDRs) so long as the hypervariable regions are identical or at least 90% homologous, preferably at least 95% homologous to the hypervariable regions of the light and heavy chains of the invention.
As known to those skilled in the art, immunoconjugates and fusion expression products include: drugs, toxins, cytokines (cytokines), radionuclides, enzymes, and other diagnostic or therapeutic molecules are conjugated to the anti-enterovirus D68 monoclonal antibody or fragment thereof. The invention also comprises a cell surface marker or antigen combined with the anti-enterovirus D68 monoclonal antibody or the fragment thereof.
The invention includes not only intact monoclonal antibodies, but also immunologically active antibody fragments, such as Fab or (Fab ")2A fragment; an antibody heavy chain; the light chain of the antibody.
As used herein, the terms "heavy chain variable region" and "VH"may be used interchangeably.
As used herein, the term "variable region" is used interchangeably with "Complementary Determining Region (CDR)".
In a preferred embodiment of the present invention, the heavy chain variable region of the antibody BSD01 includes the following three complementarity determining regions CDRs:
VH-CDR1, the amino acid sequence of which is SEQ ID NO. 2, and the coding nucleotide sequence of which is SEQ ID NO. 1;
VH-CDR2, whose amino acid sequence is SEQ ID NO. 4 and whose coding nucleotide sequence is SEQ ID NO. 3;
VH-CDR3, whose amino acid sequence is SEQ ID NO. 6 and whose coding nucleotide sequence is SEQ ID NO. 5.
In another preferred embodiment, the amino acid sequence of the heavy chain variable region is SEQ ID NO 16 and the coding nucleotide sequence is SEQ ID NO 14.
In another preferred embodiment of the present invention, the heavy chain variable region of the antibody BSD02 comprises the following three complementarity determining regions CDRs:
VH-CDR1, whose amino acid sequence is SEQ ID NO. 22 and whose coding nucleotide sequence is SEQ ID NO. 21;
VH-CDR2, the amino acid sequence of which is SEQ ID NO. 24, and the coding nucleotide sequence of which is SEQ ID NO. 23;
VH-CDR3, the amino acid sequence of which is SEQ ID NO:26, and the coding nucleotide sequence of which is SEQ ID NO: 25.
In another preferred embodiment, the amino acid sequence of the heavy chain variable region is SEQ ID NO:36 and the coding nucleotide sequence is SEQ ID NO: 34.
In a preferred embodiment of the invention, the heavy chain of the antibody comprises the above-described heavy chain variable region and a heavy chain constant region, which may be of murine or human origin.
As used herein, the terms "light chain variable region" and "VL"may be used interchangeably.
In a preferred embodiment of the present invention, the light chain variable region of the antibody BSD01 includes the following three complementarity determining regions CDRs:
VL-CDR1, the amino acid sequence of which is SEQ ID NO. 8, and the coding nucleotide sequence of which is SEQ ID NO. 7;
VL-CDR2, the amino acid sequence of which is SEQ ID NO. 10, and the coding nucleotide sequence of which is SEQ ID NO. 9;
VL-CDR3, the amino acid sequence of which is SEQ ID NO. 12 and the coding nucleotide sequence of which is SEQ ID NO. 11;
in another preferred embodiment, the variable region of the light chain has the amino acid sequence of SEQ ID NO. 20 and the coding nucleotide sequence of SEQ ID NO. 18.
In another preferred embodiment of the present invention, the light chain variable region of the antibody BSD02 comprises the following three complementarity determining regions CDRs:
VL-CDR1, having an amino acid sequence of SEQ ID NO. 28 and a coding nucleotide sequence of SEQ ID NO. 27;
VL-CDR2, the amino acid sequence of which is SEQ ID NO. 30, and the coding nucleotide sequence of which is SEQ ID NO. 29;
VL-CDR3, the amino acid sequence of which is SEQ ID NO:32, and the coding nucleotide sequence of which is SEQ ID NO: 31;
in another preferred embodiment, the variable region of the light chain has the amino acid sequence of SEQ ID NO. 40 and the coding nucleotide sequence of SEQ ID NO. 38.
In a preferred embodiment of the invention, the light chain of the antibody comprises the light chain variable region and a light chain constant region, which may be murine or human.
In the present invention, the terms "antibody of the invention", "protein of the invention", or "polypeptide of the invention" are used interchangeably and refer to an antibody that specifically binds enterovirus type D68, such as a protein or polypeptide having a heavy chain variable region (the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO:14 and/or SEQ ID NO: 34) and/or a light chain variable region (the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO:18 and/or SEQ ID NO: 38). They may or may not contain the initial methionine.
In another preferred embodiment, the antibody is a murine or human murine chimeric monoclonal antibody directed against enterovirus type D68, wherein the heavy chain constant region and/or the light chain constant region can be humanized. More preferably, the humanized heavy or light chain constant region is that of human IgG1, IgG2, or the like.
The invention also provides other proteins or fusion expression products having an antibody of the invention. In particular, the invention includes any protein or protein conjugate and fusion expression product (i.e., immunoconjugate and fusion expression product) having heavy and light chains with variable regions, provided that the variable regions are identical or at least about 90% homologous, preferably at least about 95% homologous, to the variable regions of the heavy and light chains of the antibodies of the invention.
In general, the antigen binding properties of an antibody can be described by 3 specific regions located in the variable regions of the heavy and light chains, called variable regions (CDRs), which are separated by 4 Framework Regions (FRs), the amino acid sequences of the 4 FRs being relatively conserved and not directly involved in the binding reaction.
The variable regions of the heavy and/or light chains of the antibodies of the invention are of particular interest, since at least some of them are involved in binding to an antigen. Thus, the invention includes those molecules having the light and heavy chain variable regions of a monoclonal antibody with CDRs that are more than 90% (preferably more than 95%, most preferably more than 98%) homologous to the CDRs identified herein.
The invention includes not only complete monoclonal antibodies, but also fragments of antibodies with immunological activity or fusion proteins of antibodies with other sequences. Accordingly, the invention also includes fragments, derivatives and analogs of the antibodies.
As used herein, the terms "fragment," "derivative," and "analog" refer to a polypeptide that retains substantially the same biological function or activity as an antibody of the invention. A polypeptide fragment, derivative or analogue of the invention may be (i) a polypeptide in which one or more conserved or non-conserved amino acid residues, preferably conserved amino acid residues, are substituted, and such substituted amino acid residues may or may not be encoded by the genetic code, or (ii) a polypeptide having a substituent group in one or more amino acid residues, or (iii) a polypeptide in which the mature polypeptide is fused to another compound, such as a compound that extends the half-life of the polypeptide, e.g. polyethylene glycol, or (iv) a polypeptide in which an additional amino acid sequence is fused to the sequence of the polypeptide (e.g. a leader or secretory sequence or a sequence used to purify the polypeptide or a proprotein sequence, or a fusion protein with a 6His tag). Such fragments, derivatives and analogs are within the purview of those skilled in the art in view of the teachings herein.
The antibody of the present invention refers to a polypeptide having binding activity of enterovirus type D68, comprising the CDR regions described above. The term also includes variants of the polypeptides comprising the CDR regions described above that have the same function as the antibodies of the invention. These variants include (but are not limited to): deletion, insertion and/or substitution of one or more (usually 1 to 50, preferably 1 to 30, more preferably 1 to 20, most preferably 1 to 10) amino acids, and addition of one or several (usually up to 20, preferably up to 10, more preferably up to 5) amino acids at the C-terminus and/or N-terminus. For example, in the art, substitutions with amino acids of similar or similar properties will not generally alter the function of the protein. Also, for example, the addition of one or several amino acids at the C-terminus and/or N-terminus does not generally alter the function of the protein. The term also includes active fragments and active derivatives of the antibodies of the invention.
Variants of the polypeptide include: homologous sequences, conservative variants, allelic variants, natural mutants, induced mutants, proteins encoded by DNA that hybridizes under high or low stringency conditions with DNA encoding an antibody of the invention, and polypeptides or proteins obtained using antisera raised against an antibody of the invention.
The invention also provides other polypeptides, such as fusion proteins comprising human antibodies or fragments thereof. In addition to almost full-length polypeptides, the invention also encompasses fragments of the antibodies of the invention. Typically, the fragment has at least about 50 contiguous amino acids of the antibody of the invention, preferably at least about 60 contiguous amino acids, more preferably at least about 80 contiguous amino acids, and most preferably at least about 100 contiguous amino acids.
In the present invention, "conservative variant of the antibody of the present invention" means that at most 10, preferably at most 8, more preferably at most 5, and most preferably at most 3 amino acids are substituted by amino acids having similar or similar properties as compared with the amino acid sequence of the antibody of the present invention to form a polypeptide. These conservative variant polypeptides are preferably generated by amino acid substitutions according to Table 1.
TABLE 1
Initial residue(s) Representative substitutions Preferred substitutions
Ala(A) Val;Leu;Ile Val
Arg(R) Lys;Gln;Asn Lys
Asn(N) Gln;His;Lys;Arg Gln
Asp(D) Glu Glu
Cys(C) Ser Ser
Gln(Q) Asn Asn
Glu(E) Asp Asp
Gly(G) Pro;Ala Ala
His(H) Asn;Gln;Lys;Arg Arg
Ile(I) Leu;Val;Met;Ala;Phe Leu
Leu(L) Ile;Val;Met;Ala;Phe Ile
Lys(K) Arg;Gln;Asn Arg
Met(M) Leu;Phe;Ile Leu
Phe(F) Leu;Val;Ile;Ala;Tyr Leu
Pro(P) Ala Ala
Ser(S) Thr Thr
Thr(T) Ser Ser
Trp(W) Tyr;Phe Tyr
Tyr(Y) Trp;Phe;Thr;Ser Phe
Val(V) Ile;Leu;Met;Phe;Ala Leu
The invention also provides polynucleotide molecules encoding the above antibodies or fragments or fusion proteins thereof. The polynucleotide of the present invention may be in the form of DNA or RNA. The form of DNA includes cDNA, genomic DNA or artificially synthesized DNA. The DNA may be single-stranded or double-stranded. The DNA may be the coding strand or the non-coding strand. The sequence of the coding region encoding the mature polypeptide may be identical to or a degenerate variant of the sequence of the coding region as shown in SEQ ID NO 1, 3, 5, 7, 9, 11, 13, 14, 17, 18, 21, 23, 25, 27, 29, 31, 33, 34, 37 or 38. As used herein, "degenerate variant" refers in the present invention to nucleic acid sequences which encode a nucleic acid sequence having the same amino acid sequence as a polypeptide of the present invention, but which differs from the sequence of the coding region as set forth in SEQ ID NO 1, 3, 5, 7, 9, 11, 13, 14, 17, 18, 21, 23, 25, 27, 29, 31, 33, 34, 37 or 38.
Polynucleotides encoding the mature polypeptides of the invention include: a coding sequence encoding only the mature polypeptide; the coding sequence for the mature polypeptide and various additional coding sequences; the coding sequence (and optionally additional coding sequences) as well as non-coding sequences for the mature polypeptide.
The term "polynucleotide encoding a polypeptide" may include a polynucleotide encoding the polypeptide, and may also include additional coding and/or non-coding sequences.
The present invention also relates to polynucleotides which hybridize to the sequences described above and which have at least 50%, preferably at least 70%, and more preferably at least 80% identity between the two sequences. The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the polynucleotides of the present invention. In the present invention, "stringent conditions" mean: (1) hybridization and elution at lower ionic strength and higher temperature, such as 0.2 XSSC, 0.1% SDS,60 ℃; or (2) adding denaturant during hybridization, such as 50% (v/v) formamide, 0.1% calf serum/0.1% Ficoll, 42 deg.C, etc.; or (3) hybridization occurs only when the identity between two sequences is at least 90% or more, preferably 95% or more. Moreover, the polypeptides encoded by the hybridizable polynucleotides have the same biological functions and activities as the mature polypeptides shown in SEQ ID NO 16 and/or SEQ ID NO 36, SEQ ID NO 20 and/or SEQ ID NO 40.
The full-length nucleotide sequence of the antibody of the present invention or a fragment thereof can be obtained by a PCR amplification method, a recombinant method, or an artificial synthesis method. One possibility is to use synthetic methods to synthesize the sequence of interest, especially when the fragment length is short. Generally, fragments with long sequences are obtained by first synthesizing a plurality of small fragments and then ligating them. Alternatively, the coding sequence for the heavy chain and an expression tag (e.g., 6His) can be fused together to form a fusion protein.
Once the sequence of interest has been obtained, it can be obtained in large quantities by recombinant methods. 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. The biomolecules (nucleic acids, proteins, etc.) to which the present invention relates include biomolecules in an isolated form.
At present, DNA sequences encoding the proteins of the present invention (or fragments or derivatives thereof) have been obtained completely by chemical synthesis. The DNA sequence may then be introduced into various existing DNA molecules (or vectors, for example) and cells known in the art. Furthermore, mutations can also be introduced into the protein sequences of the invention by chemical synthesis.
The invention also relates to a vector comprising a suitable DNA sequence as described above and a suitable promoter or control sequence. These vectors may be used to transform an appropriate host cell so that it can express the protein.
The host cell may be a prokaryotic cell, such as a bacterial cell; or lower eukaryotic cells, such as yeast cells; or higher eukaryotic cells, such as mammalian cells. Representative examples are: escherichia coli, streptomyces; bacterial cells of salmonella typhimurium; fungal cells such as yeast; insect cells of Drosophila S2 or Sf 9; CHO, COS7, 293 cells, etc.
Transformation of a host cell with recombinant DNA can be carried out using conventional techniques well known to those skilled in the art. When the host is prokaryotic, e.g., E.coli, competent cells capable of DNA uptake can be harvested after exponential growth phase using CaCl2Methods, the steps used are well known in the art. Another method is to use MgCl2. If desired, transformation can also be carried out by electroporation. When the host is a eukaryote, the following DNA transfection methods may be used: calcium phosphate coprecipitation, conventional mechanical methods such as microinjection, electroporation, liposome encapsulation, and the like.
The obtained transformant can be cultured by a conventional method to express the polypeptide encoded by the gene of the present invention. The medium used in the culture may be selected from various conventional media depending on the host cell used. The culturing is performed under conditions suitable for growth of the host cell. After the host cells have been grown to an appropriate cell density, the selected promoter is induced by suitable means (e.g., temperature shift or chemical induction) and the cells are cultured for an additional period of time.
The recombinant polypeptide in the above method may be expressed intracellularly or on the cell membrane, or secreted extracellularly. If necessary, the recombinant protein can be isolated and purified by various separation methods using its physical, chemical and other properties. These methods are well known to those skilled in the art. Examples of such methods include, but are not limited to: conventional renaturation treatment, treatment with a protein precipitant (such as salt precipitation), centrifugation, cell lysis by osmosis, sonication, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, High Performance Liquid Chromatography (HPLC), and other various liquid chromatography techniques, and combinations thereof.
The antibodies of the invention may be used alone or in combination or conjugated with detectable labels (for diagnostic purposes), therapeutic agents, PK (protein kinase) modifying moieties or combinations of any of the above.
Detectable labels for diagnostic purposes include, but are not limited to: a fluorescent or luminescent label, a radioactive label, an MRI (magnetic resonance imaging) or CT (computed tomography) contrast agent, or an enzyme capable of producing a detectable product.
Couplable therapeutic agents include, but are not limited to: insulin, IL-2, interferon, calcitonin, GHRH peptides, gut peptide analogs, albumin, antibody fragments, cytokines, and hormones.
Therapeutic agents that may also be conjugated or conjugated to the antibodies of the invention include, but are not limited to: 1. radionuclides (Koppe et al, 2005, Cancer metastasis reviews (Cancer metastasis) 24, 539); 2. biotoxicity (Chaudhary et al, 1989, Nature 339, 394; Epel et al, 2002, Cancer immunology and Immunotherapy (Cancer immunology) 51, 565); 3. cytokines such as IL-2 and the like (Gillies et al, 1992, Proc. Natl. Acad. Sci. USA (PNAS)89, 1428; Card et al, 2004, Cancer Immunology and immunotherapy (Cancer Immunology and Immunotherapy)53, 345; Halin et al, 2003, Cancer Research (Cancer Research)63, 3202); 4. gold nanoparticles/nanorods (Lapotko et al, 2005, Cancer letters 239, 36; Huang et al, 2006, Journal of the American Chemical Society 128, 2115); 5. viral particles (Peng et al, 2004, Gene therapy 11, 1234); 6. liposomes (Mamot et al, 2005, Cancer research 65, 11631); 7. nano magnetic particles; 8. prodrug activating enzymes (e.g., DT-diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)); 10. chemotherapeutic agents (e.g., cisplatin) or nanoparticles in any form, and the like.
The invention also provides a composition. In a preferred embodiment, the composition is a pharmaceutical composition comprising the above-described antibody or active fragment thereof or fusion protein thereof, and a pharmaceutically acceptable carrier. Generally, these materials will be formulated in a non-toxic, inert and pharmaceutically acceptable aqueous carrier medium, wherein the pH is generally from about 5 to about 8, preferably from about 6 to about 8, although the pH will vary depending on the nature of the material being formulated and the condition being treated. The formulated pharmaceutical compositions may be administered by conventional routes including, but not limited to: oral, respiratory, intratumoral, intraperitoneal, intravenous, or topical administration.
The pharmaceutical composition of the present invention can be directly used for binding enterovirus D68 type, and thus can be used for prolonging the half-life of the drug, and in addition, other therapeutic agents can be used simultaneously.
The pharmaceutical composition of the present invention comprises a safe and effective amount (e.g., 0.001-99 wt%, preferably 0.01-90 wt%, more preferably 0.1-80 wt%) of the monoclonal antibody (or conjugate thereof) of the present invention as described above and a pharmaceutically acceptable carrier or excipient. Such vectors include (but are not limited to): saline, buffer, glucose, water, glycerol, ethanol, and combinations thereof. The pharmaceutical preparation should be compatible with the mode of administration. The pharmaceutical composition of the present invention can be prepared in the form of an injection, for example, by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants. Pharmaceutical compositions such as injections, solutions are preferably manufactured under sterile conditions. The amount of active ingredient administered is a therapeutically effective amount, for example from about 1 microgram per kilogram of body weight to about 10 milligrams per kilogram of body weight per day. In addition, the polypeptides of the invention may also be used with other therapeutic agents.
In the case of pharmaceutical compositions, a safe and effective amount of the immunoconjugate is administered to the mammal, wherein the safe and effective amount is typically at least about 10 micrograms/kg body weight, and in most cases no more than about 8 mg/kg body weight, preferably the dose is from about 10 micrograms/kg body weight to about 1 mg/kg body weight. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
Hybridoma cell strain
The invention also provides a hybridoma cell strain capable of producing the enterovirus D68 monoclonal antibody; preferably, the invention provides a hybridoma cell strain with high titer aiming at the enterovirus D68 monoclonal antibody.
After obtaining the hybridoma producing the enterovirus D68 monoclonal antibody of the present invention, one skilled in the art can readily prepare an antibody using the hybridoma cell line. In addition, the structure of the antibody of the present invention (e.g., the heavy chain variable region and the light chain variable region of the antibody) can be easily known by those skilled in the art, and then the monoclonal antibody of the present invention can be prepared by recombinant methods.
Preparation of monoclonal antibodies
The antibodies of the invention can be prepared by a variety of techniques known to those skilled in the art. For example, an antigen of the invention (e.g., EVD68 Virus Like Particles (VLPs)) can be administered to an animal to induce the production of monoclonal antibodies. For Monoclonal Antibodies, they can be prepared using hybridoma technology (see Kohler et al, Nature 256; 495, 1975; Kohler et al, Eur. J. Immunol.6:511,1976; Kohler et al, Eur. J. Immunol.6:292,1976; Hammerling et al, In Monoclonal Antibodies and T Cell hybrids, Elsevier, N.Y.,1981) or can be prepared using recombinant DNA methods (U.S. Pat. No. 4,816,567).
Representative myeloma cells are those that fuse efficiently, support stable high-level production of antibody by selected antibody-producing cells, and are sensitive to medium (HAT medium matrix), including myeloma Cell lines, such as murine myeloma Cell lines, including those derived from MOPC-21 and MPC-11 mouse tumors (available from salk institute Cell Distribution Center, san diego, california, usa), and SP-2, NZ0, or X63-Ag8-653 cells (available from American Type Culture Collection, rockwell, maryland, usa). Human myeloma and mouse-human hybrid myeloma cell lines have also been described for the production of human monoclonal antibodies [ Kozbor, j.immunol., 133: 3001 (1984); brodeur et al, Techniques for the Production and use of monoclonal antibodies (monoclonal antibodies Production Techniques and Applications), pp 51-63 (Marcel Dekker, Inc., New York, 1987).
The medium in which the hybridoma cells are grown is assayed to detect the production of monoclonal antibodies of the desired specificity, e.g., by in vitro binding assays such as enzyme-linked immunosorbent assay (ELISA) or Radioimmunoassay (RIA). The location of the antibody-expressing cells can be detected by FACS. The hybridoma clones can then be subcloned by limiting dilution procedures (subcloned) and grown by standard methods (Goding, monoclonal antibodies): Principles and Practice (Principles and Practice), Academic Press (1986) pp 59-103). Suitable media for this purpose include, for example, DMEM or RPMI-1640 medium. In addition, hybridoma cells can grow in animals as ascites tumors.
The monoclonal antibodies secreted by the subclones are suitably isolated from the culture medium, ascites fluid or serum by conventional immunoglobulin purification procedures, such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis or affinity chromatography.
The invention provides a monoclonal antibody aiming at enterovirus D68 type, in particular to a monoclonal antibody aiming at enterovirus D68 type. In a preferred embodiment of the invention, the monoclonal antibody is prepared by a method for producing the monoclonal antibody from ascites of a BALB/C mouse.
The main advantages of the invention are:
(1) the invention provides two monoclonal antibodies BSD01 and BSD02 which can be specifically combined with enterovirus D68;
(2) the combination of the monoclonal antibodies BSD01 and BSD02 shows broad-spectrum and extremely high neutralizing activity on various subtypes of the enterovirus D68;
(3) the monoclonal antibody of the invention is easy to express and purify;
(4) the combination of the monoclonal antibodies BSD01 and BSD02 of the invention can effectively prevent and treat the infection of the enterovirus D68 on a mouse model.
The present invention will be described in further detail with reference to the following examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures for conditions not specified in detail in the following examples are generally carried out under conventional conditions such as those described in molecular cloning, A laboratory Manual (Huang Petang et al, Beijing: scientific Press, 2002) by Sambrook. J, USA, or under conditions recommended by the manufacturer. Unless otherwise indicated, percentages and parts are by weight. The test materials and reagents used in the following examples are commercially available without specific reference.
Materials and methods
The experimental materials used in the examples of the present invention were obtained from commercial sources unless otherwise specified.
1.1 cells and viruses
Human Rhabdomyosarcoma (RD) cells were cultured as described in the literature1. The mouse myeloma cell line SP2/0 was cultured in RPMI 1640 medium (Life technologies, USA) supplemented with 10% Fetal Bovine Serum (FBS). The EVD68 strain comprises a prototype strain Fermon and two clinical isolates, namely US/MO/14-18947 and US/KY/14-189532. All viruses were expanded in RD cells and titrated by the 50% tissue culture infectious dose (TCID50) method3
1.2 antigens
EVD68 VLPs are produced in insect cells, as described in the literature4. Methods for amplification and purification of EVD68US/MO/14-18947 virus particles are described in the literature5
1.3 monoclonal antibody preparation
Animal studies were approved by the animal welfare and use committee of the shanghai pasteur institute. All mice were purchased from Shanghai laboratory animal center (SLAC, China).
Purified EVD68VLP (5. mu.g/dose) was mixed well with aluminum hydroxide adjuvant (500. mu.g/dose; Invivogen, USA) prior to immunization. Aluminum-adsorbed VLP antigen was intraperitoneally injected into 6-8 week old female BALB/c mice at weeks 0, 2 and 4. Then 10 μ g of VLP was injected via tail vein for boosting. Three days after the boost, splenocytes were harvested and fused with SP2/0 myeloma cells under the action of polyethylene glycol (PEG)1450(Sigma, USA). FusionCells were cultured for 8 days in hypoxanthine, aminopterin and thymidine (HAT; Sigma) selective growth medium. The hybridoma supernatants were tested for their ability to neutralize EVD68 infection by neutralization as described below. Positive hybridoma cells were cloned 2 to 4 times by limiting dilution to obtain a monoclonal cell line. Selected hybridoma clones were amplified and subsequently injected intraperitoneally into liquid paraffin-induced BALB/c mice. Ascites were then collected and HiTrap was usedTMThe mAb was purified on a rProtein A FF affinity column (GEHealthcare, USA).
Similarly, ZIKV-specific mab 1C11 was prepared from mice immunized with the recombinant ZIKV virus (ZIKV) E protein extracellular domain (E80), and HCV-specific mab 1F4 was prepared from mice immunized with the soluble Hepatitis C Virus (HCV) E2 protein (sE2) (unpublished data). Both mabs served as isotype controls.
1.4 neutralization assay
The neutralizing activity of hybridoma supernatants and purified mabs against EVD68 was determined by a microneutralization assay, as described in the literature2. The neutralizing concentration of the mab is defined as the lowest antibody concentration that can completely inhibit the cytopathic effect. Immediately after the cytopathic effect was observed, cell viability was determined using the CellTiter-Glo 2.0 kit (Promega, USA) according to the manufacturer's instructions. The percent neutralization was calculated according to the following formula: 100 × (fluorescence of given sample-fluorescence of the simplexvirus control sample)/(fluorescence of the simplexcell control sample-luminescence of the simplexvirus control sample). The median inhibitory concentration (IC50) for each mab was calculated by non-linear regression using GraphPad Prism software. IC50 is defined as the concentration of antibody required to inhibit 50% of viral infection compared to infection with a simple viral control sample.
1.5 enzyme-linked immunosorbent assay (ELISA)
The antibody type of the mAb was identified by ELISA using the SBA Clonotyping System-HRP kit (southern Biotech, USA) according to the manufacturer's instructions.
To determine the specificity of monoclonal antibody binding, 50 ng/well of EVD68VLP, Enterovirus 71(EV71) VLP were coated in a miniprep ELISA plate (Nunc, USA)6Or coxsackievirus A16(CVA16)VLP6Overnight at 4 ℃. Blocking was then performed with 5% skim milk powder in PBS-Tween20 (PBST). After washing with PBST, purified mAb was added at 50. mu.L/well in two-fold serial dilutions, followed by incubation at 37 ℃ for 2 hours. After washing, horseradish peroxidase (HRP) -conjugated anti-mouse IgG (1: 10,000 dilution; Sigma-Aldrich, USA) was added and incubated at 37 ℃ for 1 hour. After development, the absorbance at 450nm was monitored using a microplate reader.
1.6 biofilm interferometry (BLI) assay
Prior to the BLI assay, purified EVD68VLP and US/MO/14-18947 virus particles were labeled with EZ-Link TM Sulfo-NHS-LC-LC-biotin kit (Thermo Fisher Scientific, USA) according to the manufacturer's instructions, respectively, and then used with ZebaTMThe desalting column (Thermo Fisher Scientific) was centrifuged to remove excess unreacted biotin. To determine the binding affinity of the antibodies to EVD68, BLI assays were performed on the Octet RED96 machine (PallFort Bio, usa) according to the manufacturer's instructions. Briefly, biotinylated EVD68VLP or US/MO/14-18947 was immobilized on a streptavidin-coated biosensor (PallForte Bio) until saturation. The antigen-bound biosensor was placed in a well containing a series of diluted mab samples to allow antigen-antibody binding, and then dissociated by immersion in dissociation buffer (0.01M PBS supplemented with 0.1% bovine serum albumin and 0.02% tween 20). The equilibrium dissociation constant (KD) was calculated using Octet data analysis software (pallfortebio).
1.7 determination and analysis of monoclonal antibody sequences
To identify the antibody sequences, total RNA was isolated from hybridoma cells using TRIzol reagent (Invitrogen, usa). First strand cDNA was then synthesized, purified and tailed using the 5'RACE System (Invitrogen) according to the manufacturer's instructions. The tailed cDNA was amplified by PCR using ExTaq enzyme (Takara, Japan) and then cloned into pMD19-T vector (Takara) for sequencing to obtain the 5' terminal sequences of the heavy and light chains of the mAb.
Determination of Complementarity Determining Region (CDR) positions using IgBLAST tools7
1.8 in vivo protective Capacity assay
Evaluation of the prophylactic and therapeutic efficacy of anti-EVD 68 neutralizing mab on a mouse model of EVD68 infection5. For the prevention experiments, groups of ICR mice (age)<24h) Intraperitoneal injection of PBS, 10. mu.g/g anti-EVD 68 monoclonal antibody (2H12 or 8F12) or 10. mu.g/g isotype control monoclonal antibody (1C11 or 1F4) respectively, and infection of the abdominal cavity by 8.0X 10 after one day4TCID50US/MO/14-18947. For the treatment experiment, multiple groups of ICR mice aged one day were injected intraperitoneally with 8.0 × 104TCID50US/MO/14-18947 or 1.0X 106TCID50US/KY/14-18953, injected intraperitoneally 24 hours or 72 hours after infection with PBS, 10. mu.g/g of 2H12, 10. mu.g/g of 8F12, or a mixture of mAbs 2H12 and 8F12 (10. mu.g/g of each mAb), respectively. Survival and clinical symptoms of all mice were observed daily for two weeks post infection. The clinical scoring grading criteria were as follows: 0, health; 1, lethargy and mobility difficulties; 2, weakness of limbs; 3, paralysis of limbs; and 4, death.
Results
2.1 preparation and neutralization of anti-EVD 68 monoclonal antibody
To prepare monoclonal antibodies against EVD68, spleen cells from EVD68VLP immunized mice were fused with SP2/0 myeloma cells to obtain hybridomas. Hybridoma cells were screened by a neutralization assay to test their ability to inhibit infection by the EVD68 clinical strain US/MO/14-18947(EVD68B type). Positive hybridoma cells were subcloned by limiting dilution. Finally, 4 stable clones (2G10, 2H12, 4C11 and 8F12) were obtained. Mabs 2G10 and 2H12 were of the IgG2a subtype, whereas mabs 4C11 and 8F12 were of the IgG2b subtype (table 2). The neutralizing effect of the four mabs on strain US/MO/14-18947 was determined by a neutralization assay. As shown in Table 2, mAbs 2G10, 2H12, 4C11 and 8F12 were all able to neutralize US/MO/14-18947 at neutralizing concentrations (the lowest antibody concentration required to completely inhibit the cytopathic effect) of 1.95, 0.12 and 0.06. mu.g/mL, respectively.
Half maximal inhibitory concentrations (IC 50; the concentration of antibody required to reduce cytopathic lesions by 50%) of 2G10, 2H12, 4C11 and 8F12 were determined to be 0.337, 0.412, 0.004. mu.g/mL, respectively (FIG. 1). These results indicate that mAbs 4C11 and 8F12 neutralize the US/MO/14-18947 strain more effectively than mAbs 2G10 and 2H 12. In contrast, the Zika virus (ZIKV) specific mAb 1C11(IgG2a isotype control) and the Hepatitis C Virus (HCV) specific mAb 1F4(IgG2b isotype control) did not yet exhibit any neutralization at the maximum tested concentration (500. mu.g/mL) (Table 2).
TABLE 2 characterization of anti-EVD 68 mAbs
Figure BDA0001858933080000231
aAndb: KD (equilibrium) values for the interaction of mAbs with EVD68VLP (a) or US/MO/14-18947 virus (b) were determined by BLI (see FIG. 2).
cThe neutralizing concentration of the mab is defined as the minimum antibody concentration required to completely inhibit the appearance of cytopathic effects.
danti-EVD 68 mabs 2H12 and 8F12 were mixed at 1: 1 in combination.
The results show that: the combination of BSD01 and BSD02 can effectively neutralize all tested EVD68 strains (see section 2.4 below).
2.2 binding Properties of anti-EVD 68 monoclonal antibodies
The ability of the mabs to recognize different antigens was tested by ELISA, including EVD68 VLPs, enterovirus 71(EV71) VLPs and coxsackievirus a16(CVA16) VLPs. The results showed that mabs 2G10, 2H12, 4C11 and 8F12 were able to react with EVD68 VLPs, but not with other proteins, indicating that these 4 mabs are specific for EVD 68; while the corresponding isotype control antibodies 1C11 and 1F4 were completely non-reactive (fig. 2A, 2B, 2C). In addition, mabs 2G10 and 2H12 showed stronger binding activity to EVD68 VLPs than mabs 4C11 and 8F12 (fig. 2A).
To quantify the binding affinity of the four anti-EVD 68 mabs, a biofilm interference assay (BLI) assay was performed. In this experiment, EVD68VLP was immobilized onto a sensor and then allowed to interact with different concentrations of anti-EVD 68 mab. 2G10 had an equilibrium dissociation constant (KD) of 5.4 nM; the KD value of 2H12 was 9.9 nM; the KD value of 4C11 was 22 nM; the KD value of 8F12 was 28nM (FIG. 2D). Apparently, the affinities of mabs 2G10 and 2H12 for EVD68 VLPs were higher than those of mabs 4C11 and 8F12, consistent with the ELISA results described above (fig. 2A).
There appears to be a contradiction between the relatively low binding affinity of mabs 4C11 and 8F12 to VLP immunogens and their highly potent neutralizing activity against US/MO/14-18947. To explain this discrepancy, we determined the binding affinity of the mAb to US/MO/14-18947 virus by BLI. As shown in FIG. 2E, the affinities of mAbs 4C11 and 8F12 for US/MO/14-18947 were significantly higher than for the VLP immunogen, with KD values below 5 nM; and monoclonal antibodies 2G10 and 2H12 show lower affinity to US/MO/14-18947, and KD values are both larger than 29 nM. Thus, differences in neutralizing activity of the mAbs may be explained in part by differences in the affinity of the mAbs for the US/MO/14-18947 virus.
2.3 sequence of anti-EVD 68 monoclonal antibody
To determine the sequence of the antibody, RNA was extracted from the hybridoma cells, followed by rapid amplification of the 5 'end of the cDNA (5' RACE). Sequencing results showed that mab 2G10 and 2H12 had the same sequence, whereas mab 4C11 and 8F12 had the same sequence (fig. 3). These results indicate that clones 2G10 and 2H12 were from the same parental hybridoma cell, while clones 4C11 and 8F12 were from another single clone, although all four were initially screened in different wells. For simplicity and convenience, mabs 2H12 and 8F12 were selected for subsequent analysis. Sequence comparison analysis showed that the heavy chain variable regions (V) of mAb 2H12 and mAb 8F12H) Has 79% similarity with the amino acid sequence of the monoclonal antibody 2H12, and the light chain variable region (V) of the monoclonal antibody 8F12L) Has a sequence similarity of 57%.
2.4 Cross-neutralization Capacity of monoclonal antibodies
The inventors also tested the cross-neutralizing potential of mabs against other EVD68 strains. As shown in Table 2, monoclonal antibodies 2G10 and 2H12 can effectively neutralize clinical strains US/KY/14-18953(EVD68A type), and the neutralizing concentration is less than 0.5 mu G/mL, but the neutralizing effect on prototype strains Fermon is very weak or not. In contrast, mAbs 4C11 and 8F12 weakly neutralized US/KY/14-18953, but strongly neutralized Fermon at a concentration of less than 1.0. mu.g/mL. The isotype control antibodies 1C11 and 1F4 did not show any neutralization at the maximum concentration tested (500. mu.g/mL) for both Fermon and US/KY/14-18953 strains.
To neutralize EVD68 broadly, the inventors selected antibodies 2H12 and 8F12 and used 1: 1 are combined together. As expected, this antibody combination showed a wider neutralization breadth than any single antibody (table 2). In addition, the neutralizing concentration of the antibody combination is 1/2 or 1/4 of the neutralizing concentration of the mab with the strongest neutralizing activity for a given strain. For example, the neutralization concentrations of mAbs 2H12 and 8F12 to US/KY/14-18953 were 0.12 and 125. mu.g/mL, respectively, while the neutralization concentration of the mixture of the two antibodies to the same strain was determined to be 0.24. mu.g/mL, which is half of the neutralization concentration of mAb 2H12, indicating that only one component of the mixture, mAb 2H12, was responsible for the major effect in US/KY/14-18953. These results indicate that there is no interference or synergy in neutralization when mabs 2H12 and 8F12 are combined.
2.5 preventive efficacy of monoclonal antibodies against EVD68 infection
Paralytic mouse model related to EVD68 infection previously established by the inventors5The efficacy of in vivo protection of monoclonal antibodies against EVD68 was evaluated above, and this animal model was based on the highly lethal strain US/MO/14-18947. To evaluate the prophylactic efficacy of the mabs, groups of neonatal ICR mice were injected with PBS, 10. mu.g/g of anti-EVD 68 mab (2H12 or 8F12), or 10. mu.g/g of isotype control mab (1C11 or 1F4), respectively. One day later, US/MO/14-18947 was infected. As shown in fig. 4, the suckling mice in the PBS group and the control antibody group began to develop clinical symptoms 3 days after infection, and 69-77% of these mice eventually died. In contrast, neonatal mice treated with mab 2H12 and 8F12 had 92% and 100% survival, respectively. These results indicate that mabs 2H12 and 8F12 were effective in preventing lethal EVD68 infection in mice.
2.6 therapeutic efficacy of monoclonal antibodies against EVD68 infection
To assess the therapeutic efficacy of the mAbs, one day old ICR mice were infected with US/MO/14-18947 and 24 or 72 hours later were injected with a single needle of PBS, 10. mu.g/g mAb 2H12, 10. mu.g/g mAb 8F12, or a mixture of the two mAbs (20. mu.g/g total, 10. mu.g/g of each mAb). Survival and clinical symptoms were monitored daily after infection. PBS-treated neonatal mice began to become sick 3 days after infection, and 74-86% of the mice eventually died of the infection (fig. 5A, 5B, 5C, 5D). In contrast, mab 2H12 treatment protected 100% (13/13) and 85% (11/13) mice from lethal challenge at 1 or 3 days post infection, respectively. Similarly, mice injected with 8F12 mab 1 or 3 days post-infection were well protected with 100% and 92% survival rates, respectively. Furthermore, mice treated with 8F12 showed less clinical symptoms than mice treated with 2H12 mab (fig. 5A, 5B, 5C, 5D). In addition, treatment with a mixture of antibodies 2H12 and 8F12 at 3 days post-infection protected 92% of the suckling mice from death, which was comparable to the effect of mab 8F12 treatment (fig. 5C and 5D). These results indicate that post-infection treatment with mab 2H12, 8F12, or a combination of the two may provide effective protection of mice against US/MO/14-18947 challenge.
The inventors also tested anti-EVD 68 monoclonal antibody against another lethal EVD68 strain US/KY/14-189535The protective effect of (1). 3 days after US/KY/14-18953 infection, the mice were injected with a single dose of PBS, mAb 2H12 (10. mu.g/g), mAb 8F12 (10. mu.g/g) or a mixture of both mAbs (10. mu.g/g of 2H12 and 10. mu.g/g of 8F 12). As shown in fig. 5E and 5F, most neonatal mice treated with PBS or mab 8F12 developed symptoms of paralysis or weakness of limbs, with a final mortality of 38% and 21% in both groups, respectively. In contrast, all suckling mice treated with mab 2H12 or the antibody cocktail survived without showing any signs of acroparalysis, indicating that mab 2H12 is more effective than mab 8F12 in protecting mice from US/KY/14-18953 infection. Taken together, these findings indicate that the combination of antibodies 2H12 and 8F12 is effective in treating infection by different EVD68 strains.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.
Sequence information of the antibodies of the invention
TABLE 3 amino acid and nucleotide sequences and sequence numbering thereof for BSD01, i.e., the heavy chain CDR1-3(VH-CDR1, VH-CDR2, VH-CDR3) and the light chain CDR1-3(VL-CDR1, VL-CDR2, VL-CDR3) of 2H12 (SEQ ID NO.:)
Figure BDA0001858933080000271
Nucleotide sequence of heavy chain of 13 monoclonal antibody 2H12 (SEQ ID NO.: 13)
Figure BDA0001858933080000272
Note: wherein the leader peptide sequence is underlined + italicized, and the constant region sequence is underlined
14 monoclonal antibody 2H12 heavy chain variable region nucleotide sequence
caggtccaactgcagcagcctggggctgagcttgtgatgcctggggcttcagtgaagatgtcctgcaaggcttctggctacac attcactgactactggatgcactgggtgaagcagaggcctggacaaggccttgagtggatcggagcgattgatacttctgata gttatactacctacaatcgaaagttcaagggcaaggccacattgactgtagacgaatcctccagcacagcctacatgcagct catcagcctgacatctgaggactctgcggtctattactgcgcaagaggaggggggggtaactctccttttgcttactggggcca agggactctggtcactgtctctgca
Amino acid sequence of heavy chain of 15 monoclonal antibody 2H12 (SEQ ID NO.: 15)
Figure BDA0001858933080000273
Note: wherein underlined + italicized indicates leader peptide sequences, underlined indicates constant region sequences,
is a stop codon. Bold + underlined are Complementarity Determining Regions (CDRs).
Amino acid sequence of heavy chain variable region of 16 monoclonal antibody 2H12
Figure BDA0001858933080000284
Nucleotide sequence of 17 monoclonal antibody 2H12 light chain
Figure BDA0001858933080000281
Note: wherein the leader peptide sequence is underlined + italicized, and the constant region sequence is underlined
Nucleotide sequence of light chain variable region of 18 monoclonal antibody 2H12 SEQ ID NO
gacatccagatgacccagtctccatcctccttatctgcctctctgggagaaagagtcagtctcacttgtcgggcaagtcaggac attggtagtagtttaaactggcttcagcaggaaccagatggaactattaaacgcctgatctacgccacatccagtttagattctg gtgtccccaaaaggttcagtggcagtaggtctgggtcagattattctctcaccatcagcagccttgaatctgaagattttgtaga ctattactgtctacaatatgctagttttccgctcacgttcggtgctgggaccaagctggagctgaaa
Amino acid sequence of 19 monoclonal antibody 2H12 light chain
Figure BDA0001858933080000282
Note: wherein underlined + italicized indicates leader peptide sequences, underlined indicates constant region sequences,
is a stop codon. Bold + underlined are Complementarity Determining Regions (CDRs).
SEQ ID NO. 20 monoclonal antibody 2H12 light chain variable region amino acid sequence
Figure BDA0001858933080000285
TABLE 4 heavy chain CDR1-3(VH-CDR1, VH-CDR2, VH-CDR3) and BSD02, 8F12
Amino acid sequence and nucleotide sequence of light chain CDR1-3(VL-CDR1, VL-CDR2, VL-CDR3) and its sequence number (SEQ ID NO.:)
Figure BDA0001858933080000283
Nucleotide sequence of 33 monoclonal antibody 8F12 heavy chain
Figure BDA0001858933080000291
Wherein the leader peptide sequence is underlined + italicized, and the constant region sequence is underlined
Nucleotide sequence of heavy chain variable region of 34 monoclonal antibody 8F12
caggtccaactgcagcagcctgggactgagctggtgaggcctggagcttcagtgaagctgtcctgcaaggcttctggctact ccttcaccagctactggatgaactgggtgaagcagaggcctggacaaggccttgagtggattggcatgattcatccttccgat agtgaaactaggttaaatcagaggttcaaggacaaggccacattgactgtagacaaatcctccaacacagcctacatgcaa ctcagcagcccgacatctgaggactctgcggtctattactgcgcaagagaggggtatgattacgacgggtttgcttattgggg ccaagggactctggtcactgtctctgca
Amino acid sequence of heavy chain of 35 monoclonal antibody 8F12 (SEQ ID NO.)
Figure BDA0001858933080000292
Note: wherein underlined + italicized indicates leader peptide sequences, underlined indicates constant region sequences,
is a stop codon. Bold + underlined are Complementarity Determining Regions (CDRs).
Amino acid sequence of heavy chain variable region of 36 monoclonal antibody 8F12
Figure BDA0001858933080000293
Nucleotide sequence of 37 monoclonal antibody 8F12 light chain
Figure BDA0001858933080000301
Wherein the leader peptide sequence is underlined + italicized, and the constant region sequence is underlined
38 monoclonal antibody 8F12 light chain variable region nucleotide sequence
caaattgttctcacccagtctccagcagccatgtctgcatctccaggggagaaggtcaccatgacctgcagtgccagctcaa gtgtgatttacatgtactggtaccagcagaagccaggatcctcccccagactcctgatttatgacacatccaacctggcttctg gagtccctgttcgcttcagtggcagtgggtctgggacctcttactctctcacaatcagccgaatggaggctgaagatgctgcca cttattactgccagcagtggagtagtttcccgtacacgttcggaggggggaccaagctggaaataaaa
Amino acid sequence of light chain of 39 monoclonal antibody 8F12
Figure BDA0001858933080000302
Note: wherein underlined + italicized indicates leader peptide sequences, underlined indicates constant region sequences,
is a stop codon. Bold + underlined are Complementarity Determining Regions (CDRs).
SEQ ID NO. 40 monoclonal antibody 8F12 light chain variable region amino acid sequence
Figure BDA0001858933080000303
Reference to the literature
1.Ku Z,Shi J,Liu Q,Huang Z.Development of murine monoclonalantibodies with potent neutralization effects on enterovirus 71.J VirolMethods 2012;186:193-7.
2.Zhang C et al.Enterovirus D68virus-like particles expressed inPichia pastoris potently induce neutralizing antibody responses and conferprotection against lethal viral infection in mice.Emerg Microbes Infect 2018;7:3.
3.Reed LJ,Muench H.A simple method of estimating fifty per centendpoints. Am J Epidemiol 1938;27:493-7.
4.Dai W et al.A virus-like particle vaccine confers protectionagainst enterovirus D68lethal challenge in mice.Vaccine 2018;36:653-9.
5.Zhang C et al.A Mouse Model of Enterovirus D68Infection forAssessment of the Efficacy of Inactivated Vaccine.Viruses 2018;10.
6.Zhang W et al.A virus-like particle-based tetravalent vaccine forhand,foot, and mouth disease elicits broad and balanced protectiveimmunity.Emerg Microbes Infect 2018;7:94.
7.Ye J,Ma N,Madden TL,Ostell JM.IgBLAST:an immunoglobulin variabledomain sequence analysis tool.Nucleic Acids Res 2013;41:W34-40.
Sequence listing
<110> Shanghai Pasteur institute of Chinese academy of sciences
<120> broad-spectrum neutralizing monoclonal antibody against enterovirus D68 type
<130>P2018-1895
<160>40
<170>PatentIn version 3.5
<210>1
<211>24
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>1
ggctacacat tcactgacta ctgg 24
<210>2
<211>8
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>2
Gly Tyr Thr Phe Thr Asp Tyr Trp
1 5
<210>3
<211>24
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>3
attgatactt ctgatagtta tact 24
<210>4
<211>8
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>4
Ile Asp Thr Ser Asp Ser Tyr Thr
1 5
<210>5
<211>36
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>5
gcaagaggag gggggggtaa ctctcctttt gcttac 36
<210>6
<211>12
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>6
Ala Arg Gly Gly Gly Gly Asn Ser Pro Phe Ala Tyr
1 5 10
<210>7
<211>18
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>7
caggacattg gtagtagt 18
<210>8
<211>6
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>8
Gln Asp Ile Gly Ser Ser
1 5
<210>9
<211>9
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>9
gccacatcc 9
<210>10
<211>3
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>10
Ala Thr Ser
1
<210>11
<211>27
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>11
ctacaatatg ctagttttcc gctcacg 27
<210>12
<211>9
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>12
Leu Gln Tyr Ala Ser Phe Pro Leu Thr
1 5
<210>13
<211>1407
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>13
atgagatgga gctgtatcat cctcttcttg gtagcaacag ctacaggtgt caactcccag 60
gtccaactgc agcagcctgg ggctgagctt gtgatgcctg gggcttcagt gaagatgtcc 120
tgcaaggctt ctggctacac attcactgac tactggatgc actgggtgaa gcagaggcct 180
ggacaaggcc ttgagtggat cggagcgatt gatacttctg atagttatac tacctacaat 240
cgaaagttca agggcaaggc cacattgact gtagacgaat cctccagcac agcctacatg 300
cagctcatca gcctgacatc tgaggactct gcggtctatt actgcgcaag aggagggggg 360
ggtaactctc cttttgctta ctggggccaa gggactctgg tcactgtctc tgcagccaaa 420
acaacagccc catcggtcta tccactggcc cctgtgtgtg gagatacaac tggctcctcg 480
gtgactctag gatgcctggt caagggttat ttccctgagc cagtgacctt gacctggaac 540
tctggatccc tgtccagtgg tgtgcacacc ttcccagctg tcctgcagtc tgacctctac 600
accctcagca gctcagtgac tgtaacctcg agcacctggc ccagccagtc catcacctgc 660
aatgtggccc acccggcaag cagcaccaag gtggacaaga aaattgagcc cagagggccc 720
acaatcaagc cctgtcctcc atgcaaatgc ccagcaccta acctcttggg tggaccatcc 780
gtcttcatct tccctccaaa gatcaaggat gtactcatga tctccctgag ccccatagtc 840
acatgtgtgg tggtggatgt gagcgaggat gacccagatg tccagatcag ctggtttgtg 900
aacaacgtgg aagtacacac agctcagaca caaacccata gagaggatta caacagtact 960
ctccgggtgg tcagtgccct ccccatccag caccaggact ggatgagtgg caaggagttc 1020
aaatgcaagg tcaacaacaa agacctccca gcgcccatcg agagaaccat ctcaaaaccc 1080
aaagggtcag taagagctcc acaggtatat gtcttgcctc caccagaaga agagatgact 1140
aagaaacagg tcactctgac ctgcatggtc acagacttca tgcctgaaga catttacgtg 1200
gagtggacca acaacgggaa aacagagcta aactacaaga acactgaacc agtcctggac 1260
tctgatggtt cttacttcat gtacagcaag ctgagagtgg aaaagaagaa ctgggtggaa 1320
agaaatagct actcctgttc agtggtccac gagggtctgc acaatcacca cacgactaag 1380
agcttctccc ggactccggg taaatga 1407
<210>14
<211>357
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>14
caggtccaac tgcagcagcc tggggctgag cttgtgatgc ctggggcttc agtgaagatg 60
tcctgcaagg cttctggcta cacattcact gactactgga tgcactgggt gaagcagagg 120
cctggacaag gccttgagtg gatcggagcg attgatactt ctgatagtta tactacctac 180
aatcgaaagt tcaagggcaa ggccacattg actgtagacg aatcctccag cacagcctac 240
atgcagctca tcagcctgac atctgaggac tctgcggtct attactgcgc aagaggaggg 300
gggggtaact ctccttttgc ttactggggc caagggactc tggtcactgt ctctgca 357
<210>15
<211>468
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>15
Met Arg Trp Ser Cys Ile Ile Leu Phe Leu Val Ala Thr Ala Thr Gly
1 5 10 15
Val Asn Ser Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Met
20 25 30
Pro Gly Ala Ser Val Lys Met Ser Cys Lys Ala Ser Gly Tyr Thr Phe
35 40 45
Thr Asp Tyr Trp Met His Trp Val Lys Gln Arg Pro Gly Gln Gly Leu
50 55 60
Glu Trp Ile Gly Ala Ile Asp Thr Ser Asp Ser Tyr Thr Thr Tyr Asn
65 70 75 80
Arg Lys Phe Lys Gly Lys Ala Thr Leu Thr Val Asp Glu Ser Ser Ser
85 90 95
Thr Ala Tyr Met Gln Leu Ile Ser Leu Thr Ser Glu Asp Ser Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Gly Gly Gly Gly Asn Ser Pro Phe Ala Tyr Trp
115 120 125
Gly Gln Gly Thr Leu Val Thr Val Ser Ala Ala Lys Thr Thr Ala Pro
130 135 140
Ser Val Tyr Pro Leu Ala Pro Val Cys Gly Asp Thr Thr Gly Ser Ser
145 150 155 160
Val Thr Leu Gly Cys Leu Val Lys Gly Tyr Phe Pro Glu Pro Val Thr
165 170 175
Leu Thr Trp Asn Ser Gly Ser Leu Ser Ser Gly Val His Thr Phe Pro
180 185 190
Ala Val Leu Gln Ser Asp Leu Tyr Thr Leu Ser Ser Ser Val Thr Val
195 200 205
Thr Ser Ser Thr Trp Pro Ser Gln Ser Ile Thr Cys Asn Val Ala His
210 215 220
Pro Ala Ser Ser Thr Lys Val Asp Lys Lys Ile Glu Pro Arg Gly Pro
225 230 235 240
Thr Ile Lys Pro Cys Pro Pro Cys Lys Cys Pro Ala Pro Asn Leu Leu
245 250 255
Gly Gly Pro Ser Val Phe Ile Phe Pro Pro Lys Ile Lys Asp Val Leu
260 265 270
Met Ile Ser Leu Ser Pro Ile Val Thr Cys Val Val Val Asp Val Ser
275 280 285
Glu Asp Asp Pro Asp Val Gln Ile Ser Trp Phe Val Asn Asn Val Glu
290 295 300
Val His Thr Ala Gln Thr Gln Thr His Arg Glu Asp Tyr Asn Ser Thr
305 310 315 320
Leu Arg Val Val Ser Ala Leu Pro Ile Gln His Gln Asp Trp Met Ser
325 330 335
Gly Lys Glu Phe Lys Cys Lys Val Asn Asn Lys Asp Leu Pro Ala Pro
340 345 350
Ile Glu Arg Thr Ile Ser Lys Pro Lys Gly Ser Val Arg Ala Pro Gln
355 360 365
Val Tyr Val Leu Pro Pro Pro Glu Glu Glu Met Thr Lys Lys Gln Val
370 375 380
Thr Leu Thr Cys Met Val Thr Asp Phe Met Pro Glu Asp Ile Tyr Val
385 390 395 400
Glu Trp Thr Asn Asn Gly Lys Thr Glu Leu Asn Tyr Lys Asn Thr Glu
405 410 415
Pro Val Leu Asp Ser Asp Gly Ser Tyr Phe Met Tyr Ser Lys Leu Arg
420 425 430
Val Glu Lys Lys Asn Trp Val Glu Arg Asn Ser Tyr Ser Cys Ser Val
435 440 445
Val His Glu Gly Leu His Asn His His Thr Thr Lys Ser Phe Ser Arg
450 455 460
Thr Pro Gly Lys
465
<210>16
<211>119
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>16
Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Met Pro Gly Ala
1 5 10 15
Ser Val Lys Met Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Trp Met His Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ala Ile Asp Thr Ser Asp Ser Tyr Thr Thr Tyr Asn Arg Lys Phe
50 55 60
Lys Gly Lys Ala Thr Leu Thr Val Asp Glu Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ile Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Gly Gly Asn Ser Pro Phe Ala Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ala
115
<210>17
<211>705
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>17
atgagggctc ctgcacagat ttttggcttc ttgttgctct tgtttccagg taccagatgt 60
gacatccaga tgacccagtc tccatcctcc ttatctgcct ctctgggaga aagagtcagt 120
ctcacttgtc gggcaagtca ggacattggt agtagtttaa actggcttca gcaggaacca 180
gatggaacta ttaaacgcct gatctacgcc acatccagtt tagattctgg tgtccccaaa 240
aggttcagtg gcagtaggtc tgggtcagat tattctctca ccatcagcag ccttgaatct 300
gaagattttg tagactatta ctgtctacaa tatgctagtt ttccgctcac gttcggtgct 360
gggaccaagc tggagctgaa acgggctgat gctgcaccaa ctgtatccat cttcccacca 420
tccagtgagc agttaacatc tggaggtgcc tcagtcgtgt gcttcttgaa caacttctac 480
cccaaagaca tcaatgtcaa gtggaagatt gatggcagtg aacgacaaaa tggcgtcctg 540
aacagttgga ctgatcagga cagcaaagac agcacctaca gcatgagcag caccctcacg 600
ttgaccaagg acgagtatga acgacataac agctatacct gtgaggccac tcacaagaca 660
tcaacttcac ccattgtcaa gagcttcaac aggaatgagt gttag 705
<210>18
<211>321
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>18
gacatccaga tgacccagtc tccatcctcc ttatctgcct ctctgggaga aagagtcagt 60
ctcacttgtc gggcaagtca ggacattggt agtagtttaa actggcttca gcaggaacca 120
gatggaacta ttaaacgcct gatctacgcc acatccagtt tagattctgg tgtccccaaa 180
aggttcagtg gcagtaggtc tgggtcagat tattctctca ccatcagcag ccttgaatct 240
gaagattttg tagactatta ctgtctacaa tatgctagtt ttccgctcac gttcggtgct 300
gggaccaagc tggagctgaa a 321
<210>19
<211>234
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>19
Met Arg Ala Pro Ala Gln Ile Phe Gly Phe Leu Leu Leu Leu Phe Pro
1 5 10 15
Gly Thr Arg Cys Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser
20 25 30
Ala Ser Leu Gly Glu Arg Val Ser Leu Thr Cys Arg Ala Ser Gln Asp
35 40 45
Ile Gly Ser Ser Leu Asn Trp Leu Gln Gln Glu Pro Asp Gly Thr Ile
50 55 60
Lys Arg Leu Ile Tyr Ala Thr Ser Ser Leu Asp Ser Gly Val Pro Lys
65 70 75 80
Arg Phe Ser Gly Ser Arg Ser Gly Ser Asp Tyr Ser Leu Thr Ile Ser
85 90 95
Ser Leu Glu Ser Glu Asp Phe Val Asp Tyr Tyr Cys Leu Gln Tyr Ala
100 105 110
Ser Phe Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys Arg
115 120 125
Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu Gln
130 135 140
Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe Tyr
145 150 155 160
Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg Gln
165 170 175
Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser Thr
180 185 190
Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu Arg
195 200 205
His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser Pro
210 215 220
Ile Val Lys Ser Phe Asn Arg Asn Glu Cys
225 230
<210>20
<211>107
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>20
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Leu Gly
1 5 10 15
Glu Arg Val Ser Leu Thr Cys Arg Ala Ser Gln Asp Ile Gly Ser Ser
20 25 30
Leu Asn Trp Leu Gln Gln Glu Pro Asp Gly Thr Ile Lys Arg Leu Ile
35 40 45
Tyr Ala Thr Ser Ser Leu Asp Ser Gly Val Pro Lys Arg Phe Ser Gly
50 55 60
Ser Arg Ser Gly Ser Asp Tyr Ser Leu Thr Ile Ser Ser Leu Glu Ser
65 70 75 80
Glu Asp Phe Val Asp Tyr Tyr Cys Leu Gln Tyr Ala Ser Phe Pro Leu
85 90 95
Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105
<210>21
<211>24
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>21
ggctactcct tcaccagcta ctgg 24
<210>22
<211>8
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>22
Gly Tyr Ser Phe Thr Ser Tyr Trp
1 5
<210>23
<211>24
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>23
attcatcctt ccgatagtga aact 24
<210>24
<211>8
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>24
Ile His Pro Ser Asp Ser Glu Thr
1 5
<210>25
<211>36
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>25
gcaagagagg ggtatgatta cgacgggttt gcttat 36
<210>26
<211>12
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>26
Ala Arg Glu Gly Tyr Asp Tyr Asp Gly Phe Ala Tyr
1 5 10
<210>27
<211>15
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>27
tcaagtgtga tttac 15
<210>28
<211>5
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>28
Ser Ser Val Ile Tyr
1 5
<210>29
<211>9
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>29
gacacatcc 9
<210>30
<211>3
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>30
Asp Thr Ser
1
<210>31
<211>27
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>31
cagcagtgga gtagtttccc gtacacg 27
<210>32
<211>9
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>32
Gln Gln Trp Ser Ser Phe Pro Tyr Thr
1 5
<210>33
<211>1425
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>33
atgggatgga gctctatcat cctcttcttg gtagcaacag ctacaggtgt ccactcccag 60
gtccaactgc agcagcctgg gactgagctg gtgaggcctg gagcttcagt gaagctgtcc 120
tgcaaggctt ctggctactc cttcaccagc tactggatga actgggtgaa gcagaggcct 180
ggacaaggcc ttgagtggat tggcatgatt catccttccg atagtgaaac taggttaaat 240
cagaggttca aggacaaggc cacattgact gtagacaaat cctccaacac agcctacatg 300
caactcagca gcccgacatc tgaggactct gcggtctatt actgcgcaag agaggggtat 360
gattacgacg ggtttgctta ttggggccaa gggactctgg tcactgtctc tgcagccaaa 420
acaacacccc catcagtcta tccactggcc cctgggtgtg gagatacaac tggttcctcc 480
gtgactctgg gatgcctggt caagggctac ttccctgagt cagtgactgt gacttggaac 540
tctggatccc tgtccagcag tgtgcacacc ttcccagctc tcctgcagtc tggactctac 600
actatgagca gctcagtgac tgtcccctcc agcacctggc caagtcagac cgtcacctgc 660
agcgttgctc acccagccag cagcaccacg gtggacaaaa aacttgagcc cagcgggccc 720
atttcaacaa tcaacccctg tcctccatgc aaggagtgtc acaaatgccc agctcctaac 780
ctcgagggtg gaccatccgt cttcatcttc cctccaaata tcaaggatgt actcatgatc 840
tccctgacac ccaaggtcac gtgtgtggtg gtggatgtga gcgaggatga cccagacgtc 900
cagatcagct ggtttgtgaa caacgtggaa gtacacacag ctcagacaca aacccataga 960
gaggattaca acagtactat ccgggtggtc agcaccctcc ccatccagca ccaggactgg 1020
atgagtggca aggagttcaa atgcaaggtc aacaacaaag acctcccatc acccatcgag 1080
agaaccatct caaaaattaa agggctagtc agagctccac aagtatacat cttgccgcca 1140
ccagcagagc agttgtccag gaaagatgtc agtctcactt gcctggtcgt gggcttcaac 1200
cctggagaca tcagtgtgga gtggaccagc aatgggcata cagaggagaa ctacaaggac 1260
accgcaccag tcctggactc tgacggttct tacttcatat atagcaagct caatatgaaa 1320
acaagcaagt gggagaaaac agattccttc tcatgcaacg tgagacacga gggtctgaaa 1380
aattactacc tgaagaagac catctcccgg tctccgggta aatga 1425
<210>34
<211>357
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>34
caggtccaac tgcagcagcc tgggactgag ctggtgaggc ctggagcttc agtgaagctg 60
tcctgcaagg cttctggcta ctccttcacc agctactgga tgaactgggt gaagcagagg 120
cctggacaag gccttgagtg gattggcatg attcatcctt ccgatagtga aactaggtta 180
aatcagaggt tcaaggacaa ggccacattg actgtagaca aatcctccaa cacagcctac 240
atgcaactca gcagcccgac atctgaggac tctgcggtct attactgcgc aagagagggg 300
tatgattacg acgggtttgc ttattggggc caagggactc tggtcactgt ctctgca 357
<210>35
<211>474
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>35
Met Gly Trp Ser Ser Ile Ile Leu Phe Leu Val Ala Thr Ala Thr Gly
1 5 10 15
Val His Ser Gln Val Gln Leu Gln Gln Pro Gly Thr Glu Leu Val Arg
20 25 30
Pro Gly Ala Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Ser Phe
35 40 45
Thr Ser Tyr Trp Met Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu
50 55 60
Glu Trp Ile Gly Met Ile His Pro Ser Asp Ser Glu Thr Arg Leu Asn
65 70 75 80
Gln Arg Phe Lys Asp Lys Ala Thr Leu Thr Val Asp Lys Ser Ser Asn
85 90 95
Thr Ala Tyr Met Gln Leu Ser Ser Pro Thr Ser Glu Asp Ser Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Glu Gly Tyr Asp Tyr Asp Gly Phe Ala Tyr Trp
115 120 125
Gly Gln Gly Thr Leu Val Thr Val Ser Ala AlaLys Thr Thr Pro Pro
130 135 140
Ser Val Tyr Pro Leu Ala Pro Gly Cys Gly Asp Thr Thr Gly Ser Ser
145 150 155 160
Val Thr Leu Gly Cys Leu Val Lys Gly Tyr Phe Pro Glu Ser Val Thr
165 170 175
Val Thr Trp Asn Ser Gly Ser Leu Ser Ser Ser Val His Thr Phe Pro
180 185 190
Ala Leu Leu Gln Ser Gly Leu Tyr Thr Met Ser Ser Ser Val Thr Val
195 200 205
Pro Ser Ser Thr Trp Pro Ser Gln Thr Val Thr Cys Ser Val Ala His
210 215 220
Pro Ala Ser Ser Thr Thr Val Asp Lys Lys Leu Glu Pro Ser Gly Pro
225 230 235 240
Ile Ser Thr Ile Asn Pro Cys Pro Pro Cys Lys Glu Cys His Lys Cys
245 250 255
Pro Ala Pro Asn Leu Glu Gly Gly Pro Ser Val Phe Ile Phe Pro Pro
260 265 270
Asn Ile Lys Asp Val Leu Met Ile Ser Leu Thr Pro Lys Val Thr Cys
275 280 285
Val Val Val Asp Val Ser Glu Asp Asp Pro Asp Val GlnIle Ser Trp
290 295 300
Phe Val Asn Asn Val Glu Val His Thr Ala Gln Thr Gln Thr His Arg
305 310 315 320
Glu Asp Tyr Asn Ser Thr Ile Arg Val Val Ser Thr Leu Pro Ile Gln
325 330 335
His Gln Asp Trp Met Ser Gly Lys Glu Phe Lys Cys Lys Val Asn Asn
340 345 350
Lys Asp Leu Pro Ser Pro Ile Glu Arg Thr Ile Ser Lys Ile Lys Gly
355 360 365
Leu Val Arg Ala Pro Gln Val Tyr Ile Leu Pro Pro Pro Ala Glu Gln
370 375 380
Leu Ser Arg Lys Asp Val Ser Leu Thr Cys Leu Val Val Gly Phe Asn
385 390 395 400
Pro Gly Asp Ile Ser Val Glu Trp Thr Ser Asn Gly His Thr Glu Glu
405 410 415
Asn Tyr Lys Asp Thr Ala Pro Val Leu Asp Ser Asp Gly Ser Tyr Phe
420 425 430
Ile Tyr Ser Lys Leu Asn Met Lys Thr Ser Lys Trp Glu Lys Thr Asp
435 440 445
Ser Phe Ser Cys Asn Val Arg His Glu Gly Leu Lys Asn Tyr TyrLeu
450 455 460
Lys Lys Thr Ile Ser Arg Ser Pro Gly Lys
465 470
<210>36
<211>119
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>36
Gln Val Gln Leu Gln Gln Pro Gly Thr Glu Leu Val Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Met Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Met Ile His Pro Ser Asp Ser Glu Thr Arg Leu Asn Gln Arg Phe
50 55 60
Lys Asp Lys Ala Thr Leu Thr Val Asp Lys Ser Ser Asn Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Ser Pro Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Glu Gly Tyr Asp Tyr Asp Gly Phe Ala Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr ValSer Ala
115
<210>37
<211>708
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>37
atggattttc aagtgcagat tttcagcttc ctgctaatca gtgcctcagt catactgtcc 60
agaggacaaa ttgttctcac ccagtctcca gcagccatgt ctgcatctcc aggggagaag 120
gtcaccatga cctgcagtgc cagctcaagt gtgatttaca tgtactggta ccagcagaag 180
ccaggatcct cccccagact cctgatttat gacacatcca acctggcttc tggagtccct 240
gttcgcttca gtggcagtgg gtctgggacc tcttactctc tcacaatcag ccgaatggag 300
gctgaagatg ctgccactta ttactgccag cagtggagta gtttcccgta cacgttcgga 360
ggggggacca agctggaaat aaaacgggct gatgctgcac caactgtatc catcttccca 420
ccatccagtg agcagttaac atctggaggt gcctcagtcg tgtgcttctt gaacaacttc 480
taccccaaag acatcaatgt caagtggaag attgatggca gtgaacgaca aaatggcgtc 540
ctgaacagtt ggactgatca ggacagcaaa gacagcacct acagcatgag cagcaccctc 600
acgttgacca aggacgagta tgaacgacat aacagctata cctgtgaggc cactcacaag 660
acatcaactt cacccattgt caagagcttc aacaggaatg agtgttag 708
<210>38
<211>318
<212>DNA
<213> Artificial sequence (Artificial sequence)
<400>38
caaattgttc tcacccagtc tccagcagcc atgtctgcat ctccagggga gaaggtcacc 60
atgacctgca gtgccagctc aagtgtgatt tacatgtact ggtaccagca gaagccagga 120
tcctccccca gactcctgat ttatgacaca tccaacctgg cttctggagt ccctgttcgc 180
ttcagtggca gtgggtctgg gacctcttac tctctcacaa tcagccgaat ggaggctgaa 240
gatgctgcca cttattactg ccagcagtgg agtagtttcc cgtacacgtt cggagggggg 300
accaagctgg aaataaaa 318
<210>39
<211>235
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>39
Met Asp Phe Gln Val Gln Ile Phe Ser Phe Leu Leu Ile Ser Ala Ser
1 5 10 15
Val Ile Leu Ser Arg Gly Gln Ile Val Leu Thr Gln Ser Pro Ala Ala
20 25 30
Met Ser Ala Ser Pro Gly Glu Lys Val Thr Met Thr Cys Ser Ala Ser
35 40 45
Ser Ser Val Ile Tyr Met Tyr Trp Tyr Gln Gln Lys Pro Gly Ser Ser
50 55 60
Pro Arg Leu Leu Ile Tyr Asp Thr Ser Asn Leu Ala Ser Gly Val Pro
65 70 75 80
Val Arg Phe Ser Gly Ser Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile
85 90 95
Ser Arg Met Glu Ala Glu Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Trp
100 105 110
Ser Ser Phe Pro Tyr Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
115 120 125
Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu
130 135 140
Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe
145 150 155 160
Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg
165 170 175
Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser
180 185 190
Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu
195 200 205
Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser
210 215 220
Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys
225 230 235
<210>40
<211>106
<212>PRT
<213> Artificial sequence (Artificial sequence)
<400>40
Gln Ile Val Leu Thr Gln Ser Pro Ala Ala Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Met Thr Cys Ser Ala Ser Ser Ser Val Ile Tyr Met
20 25 30
Tyr Trp Tyr Gln Gln Lys Pro Gly Ser Ser Pro Arg Leu Leu Ile Tyr
35 40 45
Asp Thr Ser Asn Leu Ala Ser Gly Val Pro Val Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Trp Ser Ser Phe Pro Tyr Thr
85 90 95
Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105

Claims (13)

1. An antibody heavy chain variable region having Complementarity Determining Regions (CDRs) selected from the group consisting of:
VH-CDR1 shown in SEQ ID NO. 2,
VH-CDR2 shown in SEQ ID NO. 4,
VH-CDR3 shown in SEQ ID NO 6,
VH-CDR1 shown in SEQ ID NO:22,
VH-CDR1 shown in SEQ ID NO:24, and
VH-CDR1 shown in SEQ ID NO. 26;
preferably, the heavy chain variable region has the amino acid sequence shown in SEQ ID NO 16 and/or SEQ ID NO 36.
2. An antibody heavy chain having the heavy chain variable region and the heavy chain constant region of claim 3;
preferably, the heavy chain has the amino acid sequence shown in SEQ ID NO. 15 and/or SEQ ID NO. 35.
3. An antibody light chain variable region having Complementarity Determining Regions (CDRs) selected from the group consisting of:
VL-CDR1 shown in SEQ ID NO. 8,
VL-CDR2 shown in SEQ ID NO. 10,
VL-CDR3 shown in SEQ ID NO. 12;
VL-CDR1 shown in SEQ ID NO 28,
VL-CDR2 shown in SEQ ID NO. 30, and
VL-CDR3 shown in SEQ ID NO. 32;
preferably, the light chain variable region has the amino acid sequence shown in SEQ ID NO. 20 and/or SEQ ID NO. 40.
4. An antibody light chain having the light chain variable region and the light chain constant region of claim 1;
preferably, the light chain has the amino acid sequence shown as SEQ ID NO 19 and/or SEQ ID NO 39.
5. An antibody, wherein said antibody has:
(1) the heavy chain variable region of claim 1; and/or
(2) The light chain variable region of claim 3;
alternatively, the antibody has: the heavy chain of claim 2; and/or the light chain of claim 4.
6. A recombinant protein, said recombinant protein having:
(i) the sequence of the heavy chain variable region of claim 1, the sequence of the heavy chain of claim 2, the sequence of the light chain variable region of claim 3, the sequence of the light chain of claim 4, or the sequence of the antibody of claim 5; and
(ii) optionally a tag sequence to facilitate expression and/or purification.
7. A polynucleotide encoding a polypeptide selected from the group consisting of:
(1) the heavy chain variable region of claim 1, the heavy chain of claim 2, the light chain variable region of claim 3, the light chain of claim 4, or the antibody of claim 5; or
(2) The recombinant protein of claim 6.
8. A vector comprising the polynucleotide of claim 7.
9. A genetically engineered host cell comprising the vector or genome of claim 8 having the polynucleotide of claim 7 integrated therein.
10. An immunoconjugate, comprising:
(a) the heavy chain variable region of claim 1, the heavy chain of claim 2, the light chain variable region of claim 3, the light chain of claim 4, or the antibody of claim 5, or the recombinant protein of claim 6; and
(b) a coupling moiety selected from the group consisting of: a detectable label, a drug, a toxin, a cytokine, a radionuclide, or an enzyme.
11. A pharmaceutical composition comprising:
(i) the heavy chain variable region of claim 1, the heavy chain of claim 2, the light chain variable region of claim 3, the light chain of claim 4, or the antibody of claim 5, or the recombinant protein of claim 6, or the immunoconjugate of claim 10; and
(ii) a pharmaceutically acceptable carrier.
12. Use of the heavy chain variable region of claim 1, the heavy chain of claim 2, the light chain variable region of claim 3, the light chain of claim 4, the antibody of claim 5, the recombinant protein of claim 6, or the immunoconjugate of claim 10, for the preparation of a medicament, a reagent, a detection panel, or a kit.
13. A method for detecting enterovirus D68 in a sample, the method comprising the steps of:
(1) contacting a sample with the antibody of claim 5;
(2) detecting the formation of an antigen-antibody complex, wherein the formation of the complex is indicative of the presence of enterovirus type D68 in the sample.
CN201811326695.7A 2018-11-08 2018-11-08 Broad-spectrum neutralizing monoclonal antibody against enterovirus D68 Active CN111153988B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811326695.7A CN111153988B (en) 2018-11-08 2018-11-08 Broad-spectrum neutralizing monoclonal antibody against enterovirus D68

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811326695.7A CN111153988B (en) 2018-11-08 2018-11-08 Broad-spectrum neutralizing monoclonal antibody against enterovirus D68

Publications (2)

Publication Number Publication Date
CN111153988A true CN111153988A (en) 2020-05-15
CN111153988B CN111153988B (en) 2022-11-04

Family

ID=70554940

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811326695.7A Active CN111153988B (en) 2018-11-08 2018-11-08 Broad-spectrum neutralizing monoclonal antibody against enterovirus D68

Country Status (1)

Country Link
CN (1) CN111153988B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113150131A (en) * 2021-02-26 2021-07-23 上海市公共卫生临床中心 Monoclonal antibody for broad-spectrum recognition of group A enterovirus 2C protein and application thereof
WO2022072495A3 (en) * 2020-10-01 2022-08-18 Academia Sinica Potent neutralizing antibodies for prevention and treatment of covid-19
CN116789836A (en) * 2023-08-14 2023-09-22 浙江时迈药业有限公司 Antibodies against DLL3 and uses thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015189715A (en) * 2014-03-28 2015-11-02 公益財団法人神奈川科学技術アカデミー Monoclonal antibodies against pb2 subunit of rna-dependent rna polymerase of influenza virus
WO2017180913A2 (en) * 2016-04-13 2017-10-19 Sanofi Trispecific and/or trivalent binding proteins
JP2018102211A (en) * 2016-12-27 2018-07-05 国立大学法人東北大学 Anti-phenyl sulphate derivative antibody
CN108727488A (en) * 2017-04-13 2018-11-02 中国科学院上海巴斯德研究所 The preparation and application of anti-norovirus GII.17 monoclonal antibodies

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015189715A (en) * 2014-03-28 2015-11-02 公益財団法人神奈川科学技術アカデミー Monoclonal antibodies against pb2 subunit of rna-dependent rna polymerase of influenza virus
WO2017180913A2 (en) * 2016-04-13 2017-10-19 Sanofi Trispecific and/or trivalent binding proteins
JP2018102211A (en) * 2016-12-27 2018-07-05 国立大学法人東北大学 Anti-phenyl sulphate derivative antibody
CN108727488A (en) * 2017-04-13 2018-11-02 中国科学院上海巴斯德研究所 The preparation and application of anti-norovirus GII.17 monoclonal antibodies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GENBANK: "immunoglobulin kappa light chain,partial[Mus musculus]", 《NCBI》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022072495A3 (en) * 2020-10-01 2022-08-18 Academia Sinica Potent neutralizing antibodies for prevention and treatment of covid-19
CN113150131A (en) * 2021-02-26 2021-07-23 上海市公共卫生临床中心 Monoclonal antibody for broad-spectrum recognition of group A enterovirus 2C protein and application thereof
CN116789836A (en) * 2023-08-14 2023-09-22 浙江时迈药业有限公司 Antibodies against DLL3 and uses thereof
CN116789836B (en) * 2023-08-14 2024-01-05 浙江时迈药业有限公司 Antibodies against DLL3 and uses thereof

Also Published As

Publication number Publication date
CN111153988B (en) 2022-11-04

Similar Documents

Publication Publication Date Title
EP3689909A1 (en) Tigit antibody, antigen-binding fragment thereof, and medical use thereof
RU2553325C2 (en) Neutralising anti-influenza a virus antibodies and using them
AU2015218905A1 (en) Ebola monoclonal antibodies
CN113423733A (en) Antibodies that bind TSLP and uses thereof
CN108026166A (en) Polyomavirus neutralizing antibody
CN111153988B (en) Broad-spectrum neutralizing monoclonal antibody against enterovirus D68
CN111606993B (en) Fully human broad-spectrum neutralizing antibody 4F1 for resisting respiratory syncytial virus and application thereof
CN111732654B (en) Monoclonal antibody 1E10 for resisting SARS-CoV-2
EP3220947B1 (en) Antibodies that potently neutralize rabies virus and other lyssaviruses and uses thereof
US20170158753A1 (en) Monoclonal antibodies directed against envelope glycoproteins from multiple filovirus species
CN114644708B (en) Respiratory syncytial virus specific binding molecules
CA3001231C (en) Antibody against hepatitis b surface antigen and use thereof
CN107840889A (en) The anti-CD123 antibody of high-affinity and its application
WO2016173559A1 (en) Preparation and use of murine monoclonal antibody against gi.1 norovirus
WO2015010792A1 (en) Antibodies that potently neutralize rsv and uses thereof
CN114174331A (en) Antibodies that bind human metapneumovirus fusion proteins and uses thereof
CN108727488B (en) Preparation and application of anti-norovirus GII.17 monoclonal antibody
EP4353744A1 (en) Antibody against respiratory syncytial virus and use thereof
CN114805579A (en) Anti-human ACE2 protein monoclonal antibody, nucleic acid molecule and application
CN114685670A (en) CLDN18.2 antibodies and uses thereof
CN114773461B (en) Japanese encephalitis virus antibody 1D11 and application thereof
CN109957014B (en) Preparation and application of anti-norovirus GII.3 murine monoclonal antibody
CN113355344B (en) Expression plasmid, recombinant protein and specific monoclonal antibody of influenza virus NS1 protein and application
RU2769223C1 (en) Means and method for therapy and emergency prevention of diseases caused by the sars-cov-2 virus based on a recombinant antibody and a humanized monoclonal antibody
CN111423507B (en) Fully human antibodies that neutralize influenza viruses in a broad spectrum

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
CP03 Change of name, title or address

Address after: Building 2, No. 225, Chongqing South Road, Huangpu District, Shanghai, 200025

Patentee after: Shanghai Institute of Immunology and Infection, Chinese Academy of Sciences

Address before: No. 411, Hefei Road, Huangpu District, Shanghai 200025

Patentee before: INSTITUT PASTEUR OF SHANGHAI, CHINESE ACADEMY OF SCIENCES

CP03 Change of name, title or address