CN117304278A - Recombinant RSV F protein and application thereof - Google Patents

Recombinant RSV F protein and application thereof Download PDF

Info

Publication number
CN117304278A
CN117304278A CN202311594753.5A CN202311594753A CN117304278A CN 117304278 A CN117304278 A CN 117304278A CN 202311594753 A CN202311594753 A CN 202311594753A CN 117304278 A CN117304278 A CN 117304278A
Authority
CN
China
Prior art keywords
protein
amino acid
recombinant rsv
rsv
wild
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
CN202311594753.5A
Other languages
Chinese (zh)
Other versions
CN117304278B (en
Inventor
程谱志
柏浩
陈吉
徐志伟
胡颖嵩
姚文荣
杨克俭
洪坤学
刘勇
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.)
Abzymo Biosciences Co ltd
Wuhan Ruike Biotechnology Co ltd
Jiangsu Ruike Biotechnology Co ltd
Original Assignee
Abzymo Biosciences Co ltd
Wuhan Ruike Biotechnology Co ltd
Jiangsu Ruike Biotechnology Co ltd
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 Abzymo Biosciences Co ltd, Wuhan Ruike Biotechnology Co ltd, Jiangsu Ruike Biotechnology Co ltd filed Critical Abzymo Biosciences Co ltd
Priority to CN202311594753.5A priority Critical patent/CN117304278B/en
Publication of CN117304278A publication Critical patent/CN117304278A/en
Application granted granted Critical
Publication of CN117304278B publication Critical patent/CN117304278B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Abstract

The invention belongs to the technical field of biological medicine, and particularly relates to a recombinant RSV F protein and application thereof in preparation of respiratory syncytial virus vaccine. The recombinant RSV F protein is provided in the form of a combination mutation without engineering the natural furin cleavage site and pep27 of the wild-type RSV F protein. The recombinant RSV F protein has high expression quantity and good stability, and can be used for preparing products such as vaccine, respiratory syncytial virus antibody, respiratory syncytial virus resisting serum, diagnostic antigen and the like.

Description

Recombinant RSV F protein and application thereof
Technical Field
The invention belongs to the technical field of biological medicine, and particularly relates to a recombinant RSV F protein, a preparation method thereof and application thereof in preparation of respiratory syncytial virus vaccine.
Background
Human respiratory syncytial virus (Human Respiratory Syncytial Virus, hRSV) is one of the main causes of severe respiratory diseases in infants and elderly, and since hRSV was found in the 50 th century, scientific researchers at home and abroad have largely developed hRSV vaccines, which have been faced with great challenges of insufficient safety or poor immunogenicity, various hRSV vaccines entering the clinical trial stage have been declared "failed". In 2013, after the conformation of the F protein (prefusion F protein, pre-F) before fusion is successfully resolved, the hRSV vaccine based on Pre-F shows good application prospect. Despite extensive research on the conformational stability of RSV F protein prior to fusion, the safety and efficacy of its use as a vaccine remains a significant challenge.
Accordingly, it is an urgent need for a vaccine comprising the same to provide RSV Pre-F protein having high safety, high expression level, enhanced immunogenicity and improved stability.
Disclosure of Invention
In order to solve the technical problems, the invention discloses a recombinant RSV F protein, a preparation method thereof and application thereof in preparing respiratory syncytial virus vaccine. The recombinant RSV F protein incorporates a mutation in the amino acid sequence relative to the amino acid sequence of a corresponding wild-type RSV F protein, the mutation comprising an amino acid substitution, deletion or addition.
In one aspect, the invention provides a recombinant Respiratory Syncytial Virus (RSV) protein comprising a soluble F protein polypeptide comprising at least one modification selected from the group consisting of:
(a) At least one pair of amino acid residues is substituted with cysteine;
(b) At least one cavity filling abrupt change;
(c) Partial or complete deletion of fusion peptide;
(d) Proline mutations for preventing formation of long helices; and
(e) Amino acid sequences comprising a heterotrimeric domain are added.
In some embodiments, the at least one modification comprises substitution of at least one pair of amino acid residues in the F1 subunit and/or the F2 subunit of the F protein polypeptide with cysteine.
In some embodiments, the cysteine-substituted amino acid residues comprise at least one pair of positions 159+291, 176+190, 89+231, and 151+288 of the amino acid sequence of the wild-type RSV F protein.
In some embodiments, the at least one cavity filling mutation is an asparagine mutation at position 67 of the amino acid sequence of wild-type RSV F protein, said mutation being a substitution of a hydrophobic amino acid, utilizing steric filling of the multiple side chain hydrophobic amino acids to maintain the epitope Φ pre-fusion conformation.
In some embodiments, the asparagine at position 67 is mutated to phenylalanine or tryptophan.
In some embodiments, the fusion peptide portion of the F protein polypeptide is deleted.
In some aspects, several flexible amino acids between α4 and α5 of the RR1 region of RSV Pre-F protein form loops in the Pre-fusion structure, forming an α -helix structure after fusion. To prevent long helix formation, proline substitution mutations are introduced in the α4- α5 link loop region or other loop region to maintain the pre-fusion conformation of the F protein.
In some embodiments, the proline is mutated to at least one of a74P and D489P.
In some embodiments, the recombinant RSV F protein is mutated as compared to the wild-type respiratory syncytial virus F protein by:
(a) At least one pair of amino acid residues in the F1 subunit and/or F2 subunit of the recombinant RSV F protein compared to the wild-type respiratory syncytial virus F protein are substituted with cysteine;
(b) Asparagine at position 67 is mutated to phenylalanine or tryptophan;
(c) Partial or complete deletion of fusion peptide; and
(d) Proline mutations for preventing formation of long helices;
the amino acid sequence of the F protein of the wild respiratory syncytial virus is shown as SEQ ID NO. 1.
In some embodiments, the cysteine substitutions result in formation of a non-native disulfide bond linkage between the F1 subunit and the F2 subunit, or within the F1 subunit, of the recombinant RSV F protein, including disulfide bonds other than Cys69-Cys212 and Cys37-Cys439 formed between the F1 subunit and the F2 subunit.
In the pre-fusion conformation of the RSV F protein, the β2 and β4 chains are close to each other, facilitating the spatial folding of two discrete epitopes of the Φ epitope; the space position between the beta 2 chain and the beta 4 chain can be relatively stabilized by adding an unnatural disulfide bond, and the structure of the Pre-F protein can be well stabilized.
In some embodiments, the cysteine substitutions are selected from one or more of H159c+i291C, K c+s190C, A89 8c+l231C and g151 c+i288C.
In some embodiments, the cysteine substitutions are a89c+l231C, a89c+l231C such that an interchain disulfide bond is formed between positions 89 and 231 of the recombinant RSV F protein.
In some embodiments, the cysteine substitutions are K176c+s190C and a89c+l231C such that an intrachain disulfide bond is formed between positions 176 and 190 and an interchain disulfide bond is formed between positions 89 and 231 of the recombinant RSV F protein.
In some embodiments, the cysteine substitution is h159c+i291C such that an intrachain disulfide bond is formed between position 159 and 291 of the recombinant RSV F protein.
In some embodiments, to maintain the spatial conformation and stability of the two discrete epitope peptides aa62-69 and aa196-209 prior to epitope Φfusion, hydrophobic amino acid substitutions are made in the region between the β1 and β2, and/or β3 and β4 structures near the epitope, with steric filling of the multiple side chain hydrophobic amino acids, to maintain the pre-epitope Φfusion conformation.
In some embodiments, the recombinant RSV F protein has at least one cavity filling mutation compared to the wild-type respiratory syncytial virus F protein, the cavity filling mutation comprising at least an asparagine mutation at position 67 to phenylalanine (i.e., N67F) or tryptophan (i.e., N67W).
In some embodiments, the abrupt cavity filling change is preferably N67F.
In other embodiments, the abrupt cavity filling change is preferably N67W.
In some embodiments, the recombinant RSV F protein has a partial deletion of the fusion peptide compared to the wild-type respiratory syncytial virus F protein.
In some embodiments, the portion of the fusion peptide that is deleted comprises at least amino acid residues 137-144 of the wild-type RSV F protein.
In some embodiments, the portion of the fusion peptide that is deleted is amino acid residues 137-144 of the wild-type RSV F protein.
In some embodiments, the portion of the fusion peptide that is deleted is amino acid residues 137-146 of the wild-type RSV F protein.
In some embodiments, the portion of the fusion peptide that is deleted is amino acid residues 110-146 of the wild-type RSV F protein.
In some embodiments, the proline is mutated to at least one of a74P and D489P.
In some embodiments, the cysteine substitution is h159c+i291C, the asparagine at position 67 of the F protein amino acid sequence is mutated to tryptophan, the deleted portion of the fusion peptide comprises amino acid residues 137-144 of the wild-type respiratory syncytial virus F protein, and the proline is mutated to a74P.
In some embodiments, the cysteine substitution is h159c+i291C, the asparagine at position 67 of the F protein amino acid sequence is mutated to phenylalanine, the deleted portion of the fusion peptide comprises amino acid residues 137-144 of the wild-type respiratory syncytial virus F protein, and the proline is mutated to a74P.
In some embodiments, the cysteine substitution is h159c+i291C, the asparagine at position 67 of the F protein amino acid sequence is mutated to tryptophan, the deleted portion of the fusion peptide comprises amino acid residues 137-144 of the wild-type respiratory syncytial virus F protein, and the proline is mutated to D489P.
In some embodiments, the C-terminus of the recombinant RSV F protein comprises a trimerization domain selected from foldon or GCN 4.
In some embodiments, the trimerization domain comprised at the C-terminus of the recombinant RSV F protein is preferably foldon.
In some embodiments, the trimerization domain may be linked to the F1 subunit or a functional fragment thereof via a linker (such as an amino acid linker, e.g., sequence GG, GS, or SAIG). The linker may also be a longer linker, e.g. comprising the repeat sequence GG.
The F1 subunit of the recombinant RSV F protein can have the same length as the full-length F1 polypeptide of the corresponding wild-type RSV F protein; however, it may also have deletions, such as 1, 2, 3, 5, 10, 20 amino acid residues, up to 60 amino acid residues from the C-terminus of the full-length F1 polypeptide. The full length F1 polypeptide of the recombinant RSV F protein corresponds to amino acid positions 137-574 of the natural RSV F0 precursor and includes (from N-terminus to C-terminus) the extracellular domain (residues 137-524), the transmembrane domain (residues 525-550) and the cytoplasmic domain (residues 551-574).
In some embodiments, the F1 subunit of the recombinant RSV F protein lacks the entire cytoplasmic domain. In other embodiments, the F1 subunit lacks a cytoplasmic domain and a portion or all of a transmembrane domain. In some embodiments, the recombinant RSV F protein comprises an F1 domain in which the amino acid residue from position 510, 511, 512, 513, 514, 515, 520, 525, or 530 to 574 is absent. Typically, amino acids 514 through 574 may not be present for recombinant RSV F protein linked to trimerization domain. Thus, in some embodiments, amino acid residues 514 through 574 are absent from the F1 subunit of the recombinant RSV F protein. In other specific embodiments, the F1 subunit of the recombinant RSV F protein comprises amino acid residues 137-513 of the native F0 polypeptide sequence.
The sequence of the recombinant RSV F protein of the invention includes a protease cleavage site sequence, such as the thrombin cleavage site (LVPRGS); protein tags, such as 6×His-tag (HHHHH) and strep II tag (WSHPQFEK); or linker sequences (such as GG, GS, SAIG) that are not necessary for the function of the RSV F protein, such as induction of an immune response. Those skilled in the art will recognize such sequences and understand that, where appropriate, these sequences are not included in the disclosed recombinant RSV F proteins.
The "foldon" or "foldon domain" according to the present invention generally refers to the residue at the C-terminal end of bacteriophage T4 fibrin. In this application, the foldon domain may be the 27 residues of the C-terminal end of phage T4 fibrin or a mutant thereof. An example is a sequence having GYIPEAPRDGQAYVRKDGEWVLLSTFL (SEQ ID NO: 6). In this application, the foldon domain may be a truncated or increased N-terminal or C-terminal 1, 2, 3, 4, 5, 6 or 10 amino acid derived truncations or increases of 27 residues of the C-terminal end of bacteriophage T4 fibrin. As used herein, a mutant generally refers to a sequence that differs from a reference sequence by the inclusion of one or more differences (mutations). The difference may be a substitution, deletion or insertion of one or more amino acids.
In some embodiments, the amino acid sequence of the recombinant RSV F protein is set forth in SEQ ID NO: 11-13.
In another aspect, the invention provides a recombinant nucleic acid comprising a polynucleotide sequence encoding the recombinant RSV F protein described above. In certain embodiments, the recombinant nucleic acid is codon optimized for expression in a selected prokaryotic or eukaryotic host cell. Host cells comprising nucleic acids encoding recombinant RSV F proteins are also a feature of the invention. Advantageous host cells include prokaryotic (i.e., bacterial) host cells, such as E.coli (E.coli), as well as many eukaryotic host cells, including fungal (e.g., yeast) cells, insect cells, and mammalian cells (such as CHO, VERO, HEK293 and Expi293F cells).
The invention also provides a method for preparing the recombinant RSV F protein, which comprises the following steps:
s1, synthesizing a DNA sequence corresponding to the recombinant RSV F protein, and cloning the DNA sequence into a plasmid vector;
s2, transfecting a host cell with a recombinant plasmid vector and expressing the host cell, wherein the host cell is a eukaryotic cell;
s3, purifying the culture product to obtain the recombinant RSV F protein.
In another aspect, the invention also provides the use of any of the recombinant RSV F proteins described above in the preparation of a vaccine for the prevention of RSV infection.
The invention further provides a vaccine comprising any of the recombinant RSV F proteins described above and a pharmaceutically acceptable carrier or excipient.
In some embodiments, the single dose of human vaccine comprises 60-120 μg of the recombinant RSV F protein. In some embodiments, the single dose of human vaccine contains preferably 60 μg of the recombinant RSV F protein. In some embodiments, the single dose of human vaccine comprises preferably 120 μg of the recombinant RSV F protein.
In some embodiments, the carrier or excipient comprises a buffer. Pharmaceutically acceptable carriers and excipients are well known and may be selected by those skilled in the art. Optionally, the pharmaceutically acceptable carrier or excipient further comprises at least one component that increases solubility and/or stability. Examples of solubilizing/stabilizing agents include detergents, such as laurel sarcosine and/or tween. Other examples of solubilizing agents/stabilizers include arginine, sucrose, trehalose, and the like. Thus, one skilled in the art can select suitable excipients and carriers to produce a formulation suitable for administration to a subject by a selected use. Suitable excipients include, but are not limited to, glycerol, polyethylene glycol, KCl, calcium ions, magnesium ions, manganese ions, zinc ions, and other divalent cation related salts, and the like.
The vaccine provides protection against infection by at least one of RSV subtypes a and B. The amino acid sequences of the F proteins of subtypes a and B are about 90% identical. Example sequences of the F0 precursor polypeptide of subtype A are provided in SEQ ID NO. 1 (strain A2; genBank: AAB 86664.1), and example sequences of the F0 precursor polypeptide of subtype B are provided in SEQ ID NO. 2 (strain 18537; swiss-Prot: P13843.1). Both are 574 amino acid sequences, of which the signal peptide sequence has also been reported as amino acids 1-25. In both sequences, the TM domain is approximately amino acids 530 to 550, but is alternatively reported as 525-548. Cytoplasmic tail starts at amino acid 548 or 550 and ends at amino acid 574, wherein the palmitoylated cysteine residue is at amino acid 550.
In some embodiments, the vaccine further comprises an adjuvant.
In some embodiments, the adjuvant comprises at least one of aluminum adjuvant, squalene, tocopherol, MPL, LPA, cpG, poly (I: C), and QS-21.
The adjuvant should generally be capable of enhancing a Th 1-biased immune response in a subject or population of subjects receiving vaccine administration and be safe and effective in the subject or population of subjects at the time of selection.
Optionally, the vaccine may further comprise at least one other antigen of a pathogenic organism different from RSV, for example, the pathogenic organism is a virus different from RSV, such as herpes zoster virus, human papilloma virus, hepatitis b virus, coronavirus or influenza virus. Alternatively the pathogenic organism may be a bacterium such as diphtheria, tetanus, pertussis, haemophilus influenzae or pneumococcus.
The invention also provides a method for preparing the vaccine for preventing RSV infection, which comprises the following steps:
s1, culturing a mammalian host cell to express the recombinant RSV F protein;
s2, purifying the expressed recombinant RSV F protein;
s3, packaging the purified recombinant RSV F protein and the adjuvant according to a proportion or fully mixing.
In some embodiments, the method further comprises additional steps of gene synthesis, construction of expression vectors, lyophilization of purified proteins, and the like. The expressed recombinant RSV F protein can be recovered and purified from recombinant cell culture by any one or several of a number of methods well known in the art, including ammonium sulfate or ethanol precipitation, acid extraction, filtration, ultrafiltration, centrifugation, anion/cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxyapatite chromatography, and lectin chromatography. Finally, high performance liquid chromatography HPLC can be employed in the final purification step.
Compared with the prior art, the invention has the following beneficial effects:
the invention obtains the pre-fusion conformation of the RSV F protein capable of exposing more neutralizing antibody epitopes by carrying out various amino acid substitutions, deletions and additions on the wild type RSV F protein, and ensures that the pre-fusion conformation is maintained stably and simultaneously can cause effective neutralizing antibody reaction and binding antibody reaction on the RSV subtypes A and B. More particularly, the recombinant RSV F protein is used as an immunogen combined adjuvant, so that stronger immunity induction effect is obtained, humoral immunity response is improved, th1 type immunity is stimulated effectively, and immunogenicity of RSV antigen is improved greatly. The mutation mode disclosed by the invention is applicable to other virus strains of human RSV; is suitable for various vaccine forms which take RSV F protein as antigen, such as protein vaccine, nucleic acid vaccine, virus-like particle vaccine and carrier vaccine.
Detailed Description
The invention will be further illustrated by the following non-limiting examples, which are well known to those skilled in the art, that many modifications can be made to the invention without departing from the spirit thereof, and such modifications also fall within the scope of the invention. The following examples are merely illustrative of the present invention and should not be construed as limiting the scope of the invention as embodiments are necessarily varied. The terminology used in the description is for the purpose of describing particular embodiments only and is not intended to be limiting, the scope of the present invention being defined in 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. Preferred methods and materials of the invention are described below, but any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention. The following experimental methods are all methods described in conventional methods or product specifications unless otherwise specified, and the experimental materials used are readily available from commercial companies unless otherwise specified.
Definition of terms
Reference throughout this application to "one embodiment" means that a particular parameter, step, etc. described in that embodiment is at least included in one embodiment according to the present invention. Thus, references to "one embodiment according to the present invention," "in an embodiment," and the like, are not intended to be interpreted as referring to the same embodiment, nor are references to features intended to be included in a particular embodiment, unless references to "in another embodiment," "in a different embodiment according to the present invention," and the like are used in this application. It will be appreciated by those of skill in the art that the specific parameters, steps, etc. disclosed in one or more of the embodiments of the invention can be combined in any suitable manner.
In this application, the term "cavity filling mutation" refers to the substitution of amino acid residues in the wild-type RSV F protein by amino acids that are expected to fill the internal cavity of the mature RSV F protein. In one application, such cavity filling mutations help stabilize the pre-fusion configuration of the F protein. The cavities in the pre-fusion configuration of the RSV F protein can be identified by methods known in the art, such as by visual inspection of the crystal structure of RSV F in the pre-fusion configuration, or by using computational protein design software.
In the present application, the term "DS-Cav1" refers to a form of the RSV F protein having the amino acid sequence described in McLellan et al, science, 342 (6158), 592-598, 2013.
In the present application, the term "pXCS847" refers to a form of the RSV F protein having the amino acid sequence shown in SEQ ID NO. 19 described in the J.sub.CN 201680075615.8.
In the present application, the term "D25" refers to an antibody described in CN200880023301.9, which has the amino acid sequences of the heavy and light chain CDRs shown in fig. 11D of the specification of the application, respectively.
In the present application, the term "101F" refers to an antibody described in US11261356, which has the amino acid sequences of the heavy and light chain CDRs of anti-RSV mAb 101F shown in fig. 2a and 2b, respectively, of the specification of this application.
In the present application, the term "Mota" or "motavizumab" refers to an antibody described in WO2007002543A2, which has the amino acid sequences of the heavy and light chain CDRs of "antibody A4B4L1FR-S28R (aka motavizumab)" shown in table 2 of the specification, respectively.
In this application, the terms "comprises," "comprising," and "includes" are used in their plain, inclusive, and open-ended meaning. In some cases, the meaning of "as", "consisting of … …" is also indicated.
EXAMPLE 1 Single mutant design and Gene Synthesis
The recombinant RSV F protein of the present invention is designed and prepared based on the amino acid sequence of Wild Type (WT) RSV F protein described in SEQ ID NO. 1. This example illustrates the design of various recombinant RSV F proteins, including a foldon domain and introduced amino acid mutations.
The template amino acid sequence before introducing the amino acid mutation is:
MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLSALRTGWYTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQSTPATNNRARRELPRFMNYTLNNAKKTNVTLSKKRKRRFLGFLLGVGSAIASGVAVSKVLHLEGEVNKIKSALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDKQLLPIVNKQSCSISNIATVIEFQQKNNRLLEITREFSVNAGVTTPVSTYMLTNSELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSIIKEEVLAYVVQLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGSVSFFPQAETCKVQSNRVFCDTMNSLTLPSEVNLCNVDIFNPKYDCKIMTSKTDVSSSVITSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKQEGKSLYVKGEPIINFYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELLSAIGGYIPEAPRDGQAYVRKDGEWVLLSTFLGGLVPRGSHHHHHHSAWSHPQFEK(SEQ ID NO: 7)。
the amino acid sequences designed as follows were subjected to total gene synthesis according to codon optimisation of host Expi293F cells to determine the nucleic acid sequences.
TABLE 1 design of cysteine substituted mutants
Mutant ID Mutation
CM-1 G151C+S287C
CM-2 G151C+I288C
CM-3 Y53C+G151C
CM-4 Y53C+L303C
CM-5 V56C+T189C
CM-6 T58C+T189C
CM-8 W52C+S150C
CM-11 T54C+V301C
CM-12 T54C+Q302C
CM-13 T54C+L303C
CM-14 T54C+G151C
CM-15 A177C+T189C
CM-16 A177C+S190C
CM-17 A177C+L188C
CM-18 H159C+I291C
CM-19 A89C+L231C
CM-20 A177C+T189C,T174P
CM-21 A177C+S190C,T174P
CM-22 A177C+L188C,T174P
CM-23 I57C+S190C
CM-24 T58C+V192C
CM-25 I59C+V192C
CM-26 I59C+L193C
CM-27 G151C+Q302C
CM-28 K176C+S190C
CM-29 N175C+K191C
CM-30 A177C+T189C
CM-31 A89C+234C
CM-32 F505C+K508C
CM-33 G143C+S405C
CM-34 A102C+I148C
CM-35 L142C+N371C
CM-36 S146C+Y457C
TABLE 2 mutant design of cavity filling mutations
Mutant ID Mutation
VM-1 S46L
VM-2 V56F
VM-3 I57F
VM-4 N67F
VM-5 N67W
VM-6 K68H
VM-7 G184I
VM-8 V185E
VM-9 V187F
VM-10 T189F
VM-11 A177K
VM-12 L160K
VM-13 G162I
VM-14 V178K
VM-15 F137W,F140W
VM-16 V56F,N67F
TABLE 3 mutant design of fusion peptide deletions
Mutant ID Mutation
△FP Delta (110-146) deletions
TABLE 4 design of proline substituted mutants
Mutant ID Mutation
PM-1 S213P,I214G
PM-2 I214P,S215G
PM-3 N216P,I217G
PM-4 I217A,E218P
PM-5 T219P
PM-6 S213P,I214P
PM-7 I214P,N216P
PM-8 E218P,T219P
PM-9 I217A,E218P,T219P
PM-10 I217A,E218P,T219G
PM-11 M 213 SGKPAPGSEM 218
PM-12 M 213 SGAPEPAPGEM 218
PM-13 D73P,I214P,S215G
PM-14 D73P, M 213 SGKPAPGSEM 218
PM-15 A74P,I214P,S215G
PM-16 A74P, M 213 SGKPAPGSEM 218
PM-17 T58P,I214P,S215G
PM-18 T58P, M 213 SGKPAPGSEM 218
PM-19 E60P,I214P,S215G
PM-20 E60P, M 213 SGKPAPGSEM 218
PM-21 L195P,I214P,S215G
PM-22 L195P, M 213 SGKPAPGSEM 218
PM-28 D489P
PM-29 A74P
Note that: m is M 213 And M 218 Etc. represent the mutation sites of amino acid residues in the F protein, e.g.M 213 SGKPAPGSEM 218 The amino acid residues 213-218 in the F protein are replaced by SGKPAPGSE.
Table 5 control design
Control group SEQ ID NO:
WT 1
DS-Cav1 3
pXCS847 4
Post-F 5
EXAMPLE 2 construction, expression and Primary screening of recombinant plasmids
The synthesized target gene is inserted into eukaryotic plasmid PTT5 vector for transfection of the Expi293F cell for expression. The epi 293F cells in logarithmic growth phase are passaged one day before transfection and inoculated into 96-well deep-well plates according to a certain cell density to enable the cell density to reach 2.5X10 next day 6 /ml. Mu.g of antigen expression plasmid was diluted in 40. Mu.l Opti-MEMI and 2.4. Mu.l of transfection reagent was diluted in 40. Mu.l Opti-MEM. The DNA and transfection reagent diluted separately were mixed and incubated for 20min at room temperature. Adding DNA-liposome complex into cells, and standing at 37deg.C with 5% CO 2 On a 3mm orbital shaker, 1200rpm culture. Cell supernatants were collected 3 days after expression, and a double antibody sandwich ELISA method was used, in which 101F (Site IV specific antibody binding to both pre-and post-fusion F) was used as a coating antibody, D25 (monoclonal antibody specifically recognizing the epitope of pre-fusion F protein phi) and motavizumab (Site II specific antibody simultaneously recognizing both pre-and post-fusion F, and subsequently referred to simply as Mota) were used as marker antibody detection pairs, and expression of Site phi and Site II epitopes by each mutant was detected as OD values.
The double antibody sandwich ELISA method comprises the following specific steps:
1) Solution preparation
Coating liquid: sodium bicarbonate is 1.59g/L, sodium bicarbonate is 2.94mg/L;
20mM PB solution: disodium hydrogen phosphate; sodium dihydrogen phosphate;
sealing liquid: PB 0.02M, 1.5% BSA+5% sucrose, 0.1% Proclin300;
dilution liquid: PB 0.02M, 1.5% BSA, 0.1% Proclin300, aminopyrine.
2) Antibody coating: the 101F antibody is diluted to 4 mug/ml by coating liquid, 100 mug/well is added into the enzyme-labeled plate hole, and the mixture is coated for 16 hours at the temperature of 2-8 ℃.
3) Closing: plate washer 300. Mu.l/well, 2 wash beats, add sealing solution 150. Mu.l/well, 37℃and incubate for 2 hours.
4) Sample adding: the sample to be tested and the enzyme-labeled antibody (2000-fold dilution) were added, incubated at 37℃for 30 minutes at 100. Mu.l/well, and washed 5 times with a plate washer.
5) Color development: the reaction mixture (A+B) was added to 100. Mu.l/well, and the mixture was reacted at room temperature for 15 minutes in the absence of light.
6) Reading: the microplate reader is opened in advance, the color development is complete, 50 μl/hole of stop solution is added into the reaction hole of the microplate, and the microplate reader is placed for reading.
The primary screen gave a design with high expression for the next screening or evaluation, the results are shown in the following table.
TABLE 6 high throughput transfection of cysteine-substituted mutant 96 well plates for expression primary screening
The results show that mutants CM-2, CM-5, CM-6, CM-8, CM-14, CM-15, CM-18, CM-19, CM-23, CM-26, CM-27, CM-28, CM-32, CM-33, CM-34 and CM-35 can stably maintain neutralizing antibody epitopes Site phi and Site II, and can be used for the next screening and evaluation.
TABLE 7 mutant 96 well plate high throughput transfection expression primary screen for cavity filling mutations
The results show that the mutants VM-3, VM-4, VM-5, VM-6, VM-8, VM-10, VM-11, VM-12, VM-14 and VM-15 can stably maintain neutralizing antibody epitopes Site phi and Site II and can be used for the next screening and evaluation.
Table 8 fusion peptide deleted mutant 96 well plate high throughput transfection expression preliminary screening
The result shows that the mutant delta FP can stably maintain neutralizing antibody epitopes Site phi and Site II, and can be used for the next screening and evaluation.
TABLE 9 high throughput transfection expression preliminary screening of proline substituted mutant 96 well plates
The results show that the mutants PM-2, PM-6, PM-7, PM-8, PM-11, PM-12, PM-13 and PM-21 can stably maintain neutralizing antibody epitopes Site phi and Site II, and can be used for the next screening and evaluation.
EXAMPLE 3 evaluation of Single mutant expression level and stability
Expi293F cells in logarithmic growth phase were passaged 1.2X10 day before transfection 6 Cell density/ml was seeded in 96-well deep well plates until the next day cell density reached 2.5X10 6 Per ml. Diluting the expression plasmid and the transfection reagent by using Opti-MEMI respectively, standing at room temperature for 5min, slightly dripping the transfection reagent PEI into diluted DNA, mixing, and incubating at room temperature for 20min. Adding the DNA-PEI complex into cells, and placing at 37deg.C and 5% CO 2 On a 3mm orbital shaker, 1200rpm culture. After the single mutant is expressed for three days, centrifugally collecting the expressed cell supernatant at 10000rpm, taking the expressed supernatant, respectively storing for 7 days and 14 days at 4 ℃, then adopting a double antibody sandwich ELISA method, taking 101F as a coating antibody, taking D25 and movizumab (later referred to as Mota) as labeled antibody detection pairs, detecting the expression quantity of each mutant expressed by the recombinant plasmid, and standing for different time at 4 ℃, wherein the expression quantity is expressed as the concentration of the epitope specific neutralizing antibody detection activity, and the conformational stability is expressed as the active concentration ratio of day14/day0, and the results are shown in the following table.
TABLE 10 selection of expression levels and conformational stability of cysteine-substituted mutants
Note that: "/" indicates no reactivity detected or no assay
TABLE 11 conformational stability of cysteine-substituted mutants
Note that: "/" indicates no reactivity detected or no assay
The cysteine-substituted mutants CM-2, CM-18, CM-19, CM-28, CM-35, CM-33 and CM-14 perform better by comprehensively considering the expression level and the thermostability. The decrease of the D25 detection activity concentration is not obvious or occurs in comparison with the wild type after 14 days of storage at 4 ℃, and the retention rate of the conformation before fusion is obviously higher than that of the wild type F protein. And has stability comparable to or higher than DS-Cav 1.
TABLE 12 mutant expression level and conformational stability screening of cavity filling mutations
Note that: "/" indicates no reactivity detected or no assay
TABLE 13 mutant conformational stability of cavity filling mutations
The expression quantity and the thermal stability are comprehensively considered, and the mutants VM-4 and VM-5 have better performance. The decrease of the D25 detection activity concentration is not obvious or occurs in comparison with the wild type after 14 days of storage at 4 ℃, and the retention rate of the conformation before fusion is obviously higher than that of the wild type F protein. And has stability comparable to DS-Cav 1.
TABLE 14 screening for expression levels and conformational stability of fusion peptide deleted mutants
TABLE 15 conformational stability of fusion peptide deleted mutants
Compared with the wild type, the D25 detection activity concentration of the fusion peptide-deleted mutant is not obviously reduced after the mutant is stored for 14 days at the temperature of 4 ℃, and the conformational retention rate before fusion is obviously higher than that of the wild type F protein. And has stability comparable to DS-Cav 1. In further studies, it was found that when combined mutants were formed with other mutations, only the 137 th to 144 th amino acid residues were deleted and the expression level was higher, and therefore, mutants lacking the 137 th to 144 th amino acid residues were mainly selected for the next screening and evaluation.
TABLE 16 selection of proline substituted mutant expression levels and conformational stability
Note that: "/" indicates no reactivity detected or no assay
TABLE 17 conformational stability of proline substituted mutants
Note that: "/" indicates no reactivity detected or no assay
The mutants PM-15 and PM-29 show better performance by comprehensively considering the expression quantity and the thermal stability. The D25 detection activity concentration is not obviously reduced or is not reduced after being stored for 14 days at the temperature of 4 ℃ compared with the wild type, and the conformational residue rate before fusion is obviously higher than that of the wild type F protein. And has stability comparable to or higher than DS-Cav 1. In further studies, it was found that proline substitution D489P and a74P had higher expression levels when combined mutants were formed with other mutations, and therefore mutants in which proline substitution D489P and a74P were mainly selected for the next screening and evaluation.
Example 4 evaluation of combinatorial mutant design, expression level and stability
The signal peptide of the amino acid sequence of the template was replaced with CD33 signal peptide (SEQ ID NO: 8), the junction between the foldon domain and F1 subunit was replaced with GGSGGSG, the amino acid sequence was designed based on the above-selected single mutant with superior expression and thermal stability, and the nucleic acid sequence was determined by codon optimization according to the host Expi293F cell to perform total gene synthesis, and recombinant plasmids were constructed and expressed according to the method of example 2.
Table 18 combinatorial mutant design
Mutant ID Mutation SEQ ID NO:
RC169 K176C+S190C,A89C+L231C 9
RC171 K176C+S190C, A89C+L231C, deltaV (137-144) deletion 10
RC203 H159C, I291C, N67F, A74P, delta (137-144) deletions 11
RC204 H159C, I291C, N67W, A74P, delta (137-144) deletion 12
RC206 H159C, I291C, N67W, D489P, delta (137-144) deletion 13
RC210 G151C, I288C, N67F, delta (137-144) deletion 14
RC217 H159C, I291C, A89C, L231C, N67F, delta (137-144) deletion 15
RC220 G151C, I288C, A89C, L231C, N67W, delta (137-144) deletion 16
RC221 G151C, I288C,176C,190C, N67W, delta (137-144) deletion 17
RC222 H159C, I291C,176C,190C, N67W, delta (137-144) deletion 18
RC231 H159C,I291C,N67W,D489P 19
After three days of combined mutant expression, centrifugally collecting the expressed cell supernatant at 10000rpm, taking the expression supernatant, respectively incubating for 1 hour at 50 ℃ and 60 ℃, then adopting a double-antibody sandwich ELISA method, taking 101F as a coating antibody and D25 as a labeled antibody detection pair, and detecting the expression quantity and conformational stability of Site phi epitopes after each mutant expressed by the recombinant plasmid is incubated at 4 ℃ for 1h and at 50 ℃ for 1h and at 60 ℃ for 1h, wherein the expression quantity is expressed as the epitope specific neutralizing antibody detection active concentration, and the conformational stability is expressed as the active concentration ratio of 50 ℃/4 ℃ and 60 ℃/4 ℃, and the results are shown in the following table.
TABLE 19 expression level and conformational stability of combination mutants
Mutants RC203, RC204 and RC206 exhibited higher expression levels than the 4 control groups, and had conformational stability comparable to or better than that of the pXCS847 and DS-Cav1 control groups even at 50℃and 60℃and significantly higher than that of the Post-F and WT control groups, and as vaccine immunogen components, the temporary storage at extreme temperatures of 50-60℃for 1 hour was ensured.
Example 5 preparation of proteins
5.1 Large Scale expression of proteins
Recombinant proteins were expressed by transient transfection in Expi293F cells using PEI MAX. The day prior to transfection, the logarithmic growth phase of the Expi293F cells was passaged at 2.5X10 6 Cell density/ml was seeded in 500ml/1000ml shake flasks. Cell density was diluted to 2.5X10 on the day of transfection 6 And (3) diluting the expression plasmid DNA and the transfection reagent PEIMAX by using Opti-MEM respectively, standing at room temperature for 5min, slightly dripping the diluted transfection reagent PEIMAX into the diluted expression plasmid DNA, uniformly mixing, and incubating at room temperature for 10min. Adding the DNA-PEI complex into cells, and placing at 37deg.C and 8% CO 2 Culturing in a carbon dioxide shaker at 120 rpm. Culture supernatants were collected on day 4 post-transfection and cell debris was removed by centrifugation twice at 4000rpm and 10000 rpm. The culture supernatant was sterile filtered and the protein purified by two-step affinity chromatography with HiS Trap HP and strep ii.
5.2 protein purification
Starting an SCG chromatographic system and connecting with an instrument, regulating the system parameter pressure to be not more than 0.3Mpa, alarming to be high-sensitivity, centrifuging the cell supernatant to be purified at 10000rpm for 8min, and filtering for later use. The expressed immunogenic proteins were purified using a two-step chromatography.
(1) HiS Trap HP (5 ml) chromatography: the filtered supernatant was first subjected to pretreatment at a flow rate of 2ml/min after completion of equilibration using 50ml Buffer A (20 mM PB, 500 mM NaCl, pH 7.4) at a flow rate of 5ml/min for 10 minutes. After loading, the column was washed with 50ml Buffer A (20 mM PB, 500 mM NaCl, pH 7.4) at a flow rate of 5ml/min for 10 minutes until the UV was brought to the baseline position, and then the immunogenic protein was eluted with Buffer B (20 mM PB, 500 mM NaCl, 500 mM Imidazole, pH 7.4) at a flow rate of 5 ml/min.
(2) Strep ii (1 ml) chromatography: the column (strep II purification column) was equilibrated to UV towards baseline position using 30ml Buffer C (100 mM Tris-HCl, 150 mM NaCl, 1mM EDTA, pH 8.0) at a flow rate of 1 ml/min; loading HiS Trap HP (5 ml) chromatographic eluent at a flow rate of 1ml/min, balancing the chromatographic column to ultraviolet UV (ultraviolet) trend baseline at a flow rate of 1ml/min by Buffer C; finally, buffer D (50 mM biotin in Binding Buffer) was used to elute the immunogenic protein at a flow rate of 1 ml/min. After washing with pure water, the NaOH (50 mM) caustic wash column was regenerated.
Quantification: and uniformly mixing the eluted proteins, quantifying the proteins at UV280nm, taking out a small amount of proteins for electrophoresis detection, and after the rest of proteins are packaged and frozen at-80 ℃.
EXAMPLE 6 evaluation of conformational stability of immunogens
The stability of the purified protein immunogen conformation was identified using Pre-F Pre-fusion conformation specific mab D25. The stability of the candidate immunogen in conformation under different conditions of temperature, osmotic pressure, repeated freezing and thawing and the like is examined, and the affinity of the candidate immunogen with the D25 monoclonal antibody is tested.
Table 20 Condition of investigation
The results indicate that although each recombinant RSV F protein contains a furin cleavage site and pep27 that are native to the wild-type RSV F protein, the mutations disclosed by the invention are sufficient to stabilize it in the pre-fusion conformation and the ratio of the concentration of neutralizing antibody detection activity to D25 before treatment is close to 1 with 1h at 50 ℃, high/low osmotic pressure, and repeated freeze thawing, and particularly, the levels of stability of RC203, RC204, and RC206 are comparable to the DS-Cav1 and pXCS847 levels of the control group, all significantly higher than wild-type WTs after 1 hour of incubation at 60 ℃.
Example 7 animal test
Female BALB/C mice of 6-8 weeks of age are selected for immunization, 6 mice in each group are randomly grouped, and vaccine double-dot immunization is carried out on the mice by intramuscular injection on days 0 and 21, and 50 mu L of each of left and right legs comprises recombinant RSV F protein candidate antigens RC203, RC204 and RC206; WT, DS-Cav1, pXCS847 control antigen group; and a PBS control group. The antigen dose was 12 μg/mouse. Serum was collected on day 20 and day 35, respectively, and the collected serum was stored at-20 ℃ for subsequent detection of neutralizing antibodies and bound antibodies.
7.1 neutralizing antibody detection:
the university of martial arts virology focus laboratory was commissioned to detect neutralizing antibodies using the stored RSV A2 strain. The method comprises the following specific steps:
(1) After immunization, the serum is incubated in a water bath at 56 ℃ for 30 minutes;
(2) The inactivated serum samples were diluted 2-fold in 96-well plates at a starting dilution of 1:8 using DMEM medium containing 2% fbs;
(3) 50-100 PFU of RSV A2 strain is added into the hole, and after being uniformly mixed, the mixture is subjected to 37 ℃ and 5% CO 2 Incubating for 1 hour under the condition;
(4) Transferring the virus and serum mixture into a 96-well plate pre-inoculated with monolayer Vero cells at 37deg.C with 5% CO 2 Culturing for 7 days under the condition;
(5) Cytopathy was observed, and neutralizing antibody titer, defined as the highest serum dilution with more than 50% of intact Vero cells, was calculated using Reed-Muench method.
The results showed that the candidate antigens RC203, RC204 and RC206 induced neutralizing antibodies at a geometric mean concentration comparable to DS-Cav1 levels, higher than pXCS847, but without significant differences, all of which induced neutralizing antibodies at a geometric mean concentration significantly higher than the WT and post-F proteins.
7.2 detection of bound antibodies:
(1) Purified pre-F and post-F proteins were diluted to 3. Mu.g/mL coated 96-well plates, 100. Mu.L/well, incubated overnight at 4℃blocked with 1.5% BSA-PBS for 2 hours at 37 ℃, washed, and dried;
(2) The serum sample is subjected to 2-time serial dilution by using a sample diluent, 100 mu L of the sample is added to each hole by taking 1:200 as a detection initial point, and the sample is incubated for 30 minutes at 37 ℃;
(3) Horseradish peroxidase-conjugated goat anti-mouse IgG secondary antibody (ablonal, #as 003) was diluted 1:4000, 100 μl per well was added and incubated for 30 min at 37 ℃;
(4) Color development was performed using 3,3', 5' -Tetramethylbenzidine (TMB), 100. Mu.L was added to each well, and incubated at room temperature for 15 minutes in the absence of light;
(5) 50 mu L of 2M H are added to each well 2 SO 4 The reaction was stopped and the OD values at 450nm and 630nm were measured for each well using a microplate reader (MD, spectromax I3X);
(6) The absorbance value of the negative control is 2.1 times of the Cut-off value, and the maximum dilution multiple of the absorbance value of the sample which is more than or equal to the Cut-off value is the titer of the sample.
The results show that the geometric mean concentration of Pre-F specific antibodies induced by candidate antigens RC203, RC204 and RC206 is about 2.5 times that of WT, comparable to pXCS847 and DS-Cav1 levels.
In conclusion, the recombinant RSV F protein provided by the invention has higher stability and expression level, and the vaccine composition containing the recombinant RSV F protein can induce the generation of neutralizing antibodies with high titer against RSV, which indicates that the vaccine provides better protective effect during virus invasion and has important significance for the research and development of RSV vaccine candidates.
The foregoing description of the embodiments has been provided for the purpose of illustrating the general principles of the invention, and is not meant to limit the invention thereto, but to limit the invention thereto, and any modifications, equivalents, improvements and equivalents thereof may be made without departing from the spirit and principles of the invention.

Claims (19)

1. A recombinant Respiratory Syncytial Virus (RSV) protein comprising a soluble F protein polypeptide comprising at least one modification selected from the group consisting of:
(a) At least one pair of amino acid residues is substituted with cysteine;
(b) At least one cavity filling abrupt change;
(c) Partial or complete deletion of fusion peptide;
(d) Proline mutations for preventing formation of long helices; and
(e) Amino acid sequences comprising a heterotrimeric domain are added.
2. The recombinant RSV protein according to claim 1, wherein said at least one modification comprises substitution of at least one pair of amino acid residues in the F1 subunit and/or F2 subunit of the F protein polypeptide with cysteine.
3. The recombinant RSV protein of claim 1 or 2, wherein the cysteine-substituted amino acid residue comprises at least one pair of amino acid sequences 159+291, 176+190, 89+231, and 151+288 of the wild-type RSV F protein amino acid sequence.
4. The recombinant RSV protein according to claim 1, wherein said at least one cavity filling mutation is an asparagine mutation at amino acid sequence position 67 of wild-type RSV F protein.
5. The recombinant RSV protein of claim 4, wherein the asparagine at position 67 is mutated to phenylalanine or tryptophan.
6. The recombinant RSV protein of claim 1, wherein the fusion peptide portion of the F protein polypeptide is deleted.
7. The recombinant RSV protein of claim 1, wherein the proline mutation is at least one of a74P and D489P.
8. The recombinant RSV protein of claim 1, wherein the recombinant RSV F protein is mutated as compared to a wild-type respiratory syncytial virus F protein by:
(a) At least one pair of amino acid residues in the F1 subunit and/or F2 subunit of the recombinant RSV F protein compared to the wild-type respiratory syncytial virus F protein are substituted with cysteine;
(b) Asparagine at position 67 is mutated to phenylalanine or tryptophan;
(c) Partial or complete deletion of fusion peptide; and
(d) Proline mutations for preventing formation of long helices;
wherein the amino acid sequence of the F protein of the wild type respiratory syncytial virus is shown as SEQ ID NO. 1.
9. The recombinant RSV F protein according to claim 8, wherein said cysteine substitution is selected from one or more of H159c+i291C, K c+s190C, A c+l231C and g151 c+i288C.
10. The recombinant RSV F protein according to claim 8, wherein said fusion peptide deleted portion comprises at least amino acid residues 137-144 of a wild-type respiratory syncytial virus F protein.
11. The recombinant RSV F protein according to claim 8, wherein said proline mutation is at least one of a74P and D489P.
12. The recombinant RSV F protein according to any of claims 8-11, wherein said cysteine substitution is h159c+i291C, asparagine at amino acid sequence 67 of F protein is mutated to tryptophan, said deleted portion of fusion peptide comprises amino acid residues 137-144 of wild-type respiratory syncytial virus F protein, and said proline is mutated to a74P.
13. The recombinant RSV F protein according to any of claims 8-11, wherein said cysteine substitution is h159c+i291C, wherein the asparagine at amino acid sequence 67 of the F protein is mutated to phenylalanine, wherein said deleted portion of the fusion peptide comprises amino acid residues 137-144 of the wild-type respiratory syncytial virus F protein, and wherein said proline is mutated to a74P.
14. The recombinant RSV F protein according to any of claims 8-11, wherein said cysteine substitution is h159c+i291C, asparagine at amino acid sequence 67 of F protein is mutated to tryptophan, said deleted portion of fusion peptide comprises amino acid residues 137-144 of wild-type respiratory syncytial virus F protein, and said proline is mutated to D489P.
15. The recombinant RSV F protein according to any of claims 8-11, wherein said recombinant RSV F protein comprises a trimerization domain selected from the group consisting of foldon and GCN4 at the C-terminus.
16. The recombinant RSV F protein according to claim 15, wherein said recombinant RSV F protein has an amino acid sequence as set forth in any one of SEQ ID NOs 11-13.
17. Use of the recombinant RSV F protein of any one of claims 1-16 for the preparation of a vaccine for preventing RSV infection.
18. A vaccine comprising the recombinant RSV F protein of any one of claims 1-16 and a pharmaceutically acceptable carrier or excipient.
19. The vaccine of claim 18, further comprising an adjuvant.
CN202311594753.5A 2023-11-28 2023-11-28 Recombinant RSV F protein and application thereof Active CN117304278B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311594753.5A CN117304278B (en) 2023-11-28 2023-11-28 Recombinant RSV F protein and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311594753.5A CN117304278B (en) 2023-11-28 2023-11-28 Recombinant RSV F protein and application thereof

Publications (2)

Publication Number Publication Date
CN117304278A true CN117304278A (en) 2023-12-29
CN117304278B CN117304278B (en) 2024-04-16

Family

ID=89255531

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311594753.5A Active CN117304278B (en) 2023-11-28 2023-11-28 Recombinant RSV F protein and application thereof

Country Status (1)

Country Link
CN (1) CN117304278B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117586357A (en) * 2024-01-19 2024-02-23 北京安百胜生物科技有限公司 Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity
CN117586359A (en) * 2024-01-19 2024-02-23 北京安百胜生物科技有限公司 Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102639147A (en) * 2009-07-15 2012-08-15 诺华有限公司 RSV F protein compositions and methods for making same
CN105473604A (en) * 2013-03-13 2016-04-06 美国政府(由卫生和人类服务部的部长所代表) Prefusion RSV F proteins and their use
CN108738312A (en) * 2015-12-23 2018-11-02 辉瑞公司 Rsv f protein mutant
CN109851678A (en) * 2019-03-07 2019-06-07 苏州宇之波生物科技有限公司 A kind of inferior stable state bovine respiratory syncytial virus of improvement merges DNA molecular and its application of precursor F protein matter and coding
CN115850396A (en) * 2022-12-22 2023-03-28 北京吉诺卫生物科技有限公司 RSV (respiratory syncytial virus) nanoparticle vaccine as well as preparation method and application thereof
CN116327910A (en) * 2023-03-31 2023-06-27 北京吉诺卫生物科技有限公司 Combination vaccine of novel coronavirus, influenza virus and/or RSV, preparation method and application thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102639147A (en) * 2009-07-15 2012-08-15 诺华有限公司 RSV F protein compositions and methods for making same
CN105473604A (en) * 2013-03-13 2016-04-06 美国政府(由卫生和人类服务部的部长所代表) Prefusion RSV F proteins and their use
CN108738312A (en) * 2015-12-23 2018-11-02 辉瑞公司 Rsv f protein mutant
CN109851678A (en) * 2019-03-07 2019-06-07 苏州宇之波生物科技有限公司 A kind of inferior stable state bovine respiratory syncytial virus of improvement merges DNA molecular and its application of precursor F protein matter and coding
CN115850396A (en) * 2022-12-22 2023-03-28 北京吉诺卫生物科技有限公司 RSV (respiratory syncytial virus) nanoparticle vaccine as well as preparation method and application thereof
CN116327910A (en) * 2023-03-31 2023-06-27 北京吉诺卫生物科技有限公司 Combination vaccine of novel coronavirus, influenza virus and/or RSV, preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MARYAMMUKHAMEDOVA等: ""Vaccination with prefusion-stabilized respiratory syncytial virus fusion protein induces genetically and antigenically diverse antibody responses"", 《IMMUNITY》, vol. 54, no. 4, pages 769 - 780 *
易高;潘嘉宇;林纯意;刘兆宇;: "呼吸道合胞体病毒F蛋白二级结构分析及其B细胞表位预测", 山东医药, no. 27, pages 25 - 28 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117586357A (en) * 2024-01-19 2024-02-23 北京安百胜生物科技有限公司 Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity
CN117586359A (en) * 2024-01-19 2024-02-23 北京安百胜生物科技有限公司 Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity

Also Published As

Publication number Publication date
CN117304278B (en) 2024-04-16

Similar Documents

Publication Publication Date Title
CN117304278B (en) Recombinant RSV F protein and application thereof
US20230265127A1 (en) Stabilized soluble pre-fusion rsv f polypeptides
CN117304281B (en) Recombinant RSV F protein and application thereof
CN117304280B (en) Recombinant RSV F protein and application thereof
JP6679475B2 (en) Stabilized soluble pre-fusion RSV F polypeptide
CN115515627A (en) SARS-CoV-2 vaccine
CN117304279B (en) Recombinant RSV F protein and application thereof
JP2009022186A (en) Antigen peptide and use thereof
CN117586358A (en) Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity
CN117586357A (en) Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity
WO2024017682A1 (en) Rsv immunogens
CN117586359A (en) Respiratory Syncytial Virus (RSV) polypeptides having immunogenicity
CN117586425A (en) Recombinant respiratory syncytial virus particle antigen, preparation method and application thereof
OA19273A (en) Stabilized pre-fusion RSV F proteins.
OA17598A (en) Stabilized soluble pre-fusion RSV F polypeptides
OA17539A (en) Stabilized soluble prefusion RSV F polypeptides.
NZ714594B2 (en) Stabilized soluble pre-fusion rsv f polypeptides
NZ752808A (en) Stabilized soluble prefusion rsv f polypeptides

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