WO2022253134A1 - Procédé pour améliorer l'immunogénicité/la stabilité de trimère antigénique d'un antigène ecd de souche mutante de sars-cov-2 - Google Patents

Procédé pour améliorer l'immunogénicité/la stabilité de trimère antigénique d'un antigène ecd de souche mutante de sars-cov-2 Download PDF

Info

Publication number
WO2022253134A1
WO2022253134A1 PCT/CN2022/095609 CN2022095609W WO2022253134A1 WO 2022253134 A1 WO2022253134 A1 WO 2022253134A1 CN 2022095609 W CN2022095609 W CN 2022095609W WO 2022253134 A1 WO2022253134 A1 WO 2022253134A1
Authority
WO
WIPO (PCT)
Prior art keywords
strain
immunogenic
cov
seq
sars
Prior art date
Application number
PCT/CN2022/095609
Other languages
English (en)
Chinese (zh)
Inventor
谢良志
孙春昀
张延静
Original Assignee
神州细胞工程有限公司
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
Priority claimed from CN202110606512.2A external-priority patent/CN115477703A/zh
Application filed by 神州细胞工程有限公司 filed Critical 神州细胞工程有限公司
Priority to CN202280034785.7A priority Critical patent/CN117295771A/zh
Publication of WO2022253134A1 publication Critical patent/WO2022253134A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the invention relates to the field of molecular vaccinology, and relates to a method for improving the ECD antigen immunogenicity/antigen trimer stability of SARS-CoV-2 mutant strains and a SARS-CoV-2 improved immunogenicity/antigen trimer stability - CoV-2 mutant strain ECD immunogenic proteins/peptides.
  • the new coronavirus (SARS-CoV-2) has a strong ability to spread, and a safe and effective vaccine is the most powerful technical means to control the epidemic.
  • vaccines can be divided into the following categories: inactivated vaccines, recombinant protein vaccines, viral vector vaccines, RNA vaccines, live attenuated vaccines, and virus-like particle vaccines.
  • the trimeric Spike protein (Spike, S protein) of SARS-CoV-2 is the main component of the virus envelope and plays an important role in receptor binding, fusion, virus entry and host immune defense.
  • SARS-CoV-2 and SARS-CoV share a common host cell receptor protein, angiotensin-converting enzyme 2 (ACE2) (Zhou,P.,et al.,Discovery of a novel coronavirus associated with the recent pneumonia outbreak in humans and its potential bat origin. BioRxiv, 2020.).
  • ACE2 angiotensin-converting enzyme 2
  • SARS-CoV-2 After the S protein of SARS-CoV-2 binds to the ACE2 receptor, it is cleaved by the host protease into the S1 polypeptide containing the receptor binding domain (RBD) and the S2 polypeptide responsible for mediating the fusion of the virus with the cell membrane (Hoffmann, M. , et al., SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell, 2020.).
  • RBD receptor binding domain
  • the RBD region of the S protein contains major neutralizing antibody epitopes, which can stimulate B cells to produce high-titer neutralizing antibodies against RBD.
  • the S protein also contains abundant T cell epitopes, which can induce specific CTL responses in T cells and clear virus-infected cells. Therefore, the S protein is the most critical antigen for the design of the new crown vaccine.
  • the vast majority of vaccines currently designed have selected S protein or RBD domain protein as the core immunogen.
  • SARS-CoV-2 is an RNA single-stranded virus, which is prone to deletion mutations, and such mutations mostly occur in the repeat deletion regions (Recurrent deletion regions, RDRs) of the S protein. Deletion or mutation may change the conformation of the S protein, so that the antibodies induced by the previous vaccine immunization reduce the binding and neutralization of the mutant S protein, resulting in the decline of the immune effect of the vaccine and the immune escape of the virus.
  • the early D614G mutation (B.1) enhanced the affinity of the S protein to the ACE2 receptor and quickly became a popular strain, but the mutation did not reduce the sensitivity to neutralizing antibodies (Korber, B., et al., Spike mutation pipeline reveals the emergence of a more transmissible form of SARS-CoV-2.bioRxiv,2020:p.2020.04.29.069054.; Korber,B.,et al.,Tracking Changes in SARS-CoV-2 Spike:Evidence that D614G Increases Infectivity of the COVID-19 Virus. Cell, 2020.182(4): p.812-827.e19.).
  • Omicron variant of SARS-CoV-2 Genomics, transmissibility, and responses to current COVID-19 vaccines. 2022.
  • Omicron has spread to at least 49 countries around the world, and has replaced Delta as the main epidemic strain in the world.
  • the current vaccines are all designed based on the sequence of the early epidemic strain (its genome sequence: GenBank Accession No.NC_045512).
  • GenBank Accession No.NC_045512 the genome sequence: GenBank Accession No.NC_045512.
  • the present invention provides a method for improving the ECD antigen immunogenicity/antigen trimer stability of SARS-CoV-2 mutant strains , the method is by constructing an ECD antigen comprising at least any amino acid sequence shown in SEQ ID No:8, SEQ ID No:12 or SEQ ID No:16, or an immunogenic fragment and/or an immunogenic variant thereof,
  • ECD is a trimeric form in a stable prefusion conformation.
  • the mutant strain contains T19I, L24S, ⁇ 25/27, H49Y, A67V, ⁇ 69/70, T95I, G142D, ⁇ 143/145, ⁇ 145-146, N211I, ⁇ 212/ 212 ⁇ V213G ⁇ G339D ⁇ R346K ⁇ R346S ⁇ S371L ⁇ S373P ⁇ S375F ⁇ T376A ⁇ D405N ⁇ R408S ⁇ K417N ⁇ N440K ⁇ L452Q ⁇ L452R ⁇ S477N ⁇ T478K ⁇ E484A ⁇ E484K ⁇ E484Q ⁇ F490S ⁇ Q493R ⁇ G496S ⁇ Q498R ⁇ N501Y ⁇ High-risk mutant strains of at least any one of Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, and L98
  • the strains include B.1 strain, B.1.351 strain, B.1.1.7 strain, P.1 strain, B.1.427 strain, B.1.429 strain, B. .1.526 strain, C.37 strain, B.1.621 strain, B.1.618 strain, C.36.3 strain, 20I/484Q strain, BA.1 strain, BA.1.1 strain and BA.2 at least one of the strains.
  • the ECD antigen is co-administered to the subject with one or more adjuvants selected from:
  • TLR Toll-like receptor
  • the oil-emulsion adjuvant contains squalene
  • TLR Toll-like receptor
  • MPL monophosphoryl lipid A
  • the present invention also provides a method for improving the immunogenicity of the SARS-CoV-2 mutant strain ECD antigen immunogenicity/antigen trimer stability, the method comprises SEQ ID No: 8, SEQ ID No: 12 or SEQ ID by constructing a code A polynucleotide of at least any amino acid sequence shown in No: 16, or an immunogenic fragment and/or an immunogenic variant thereof, so as to express a trimer form of ECD in a stable prefusion conformation.
  • the mutant strain contains T19I, L24S, ⁇ 25/27, H49Y, A67V, ⁇ 69/70, T95I, G142D, ⁇ 143/145, ⁇ 145-146, N211I, ⁇ 212 /212, V213G, G339D, R346K, R346S, S371L, S373P, S375F, T376A, D405N, R408S, K417N, N440K, L452Q, L452R, S477N, T478K, E484A, E484K, E484Q, N991R, G409, Q5 , Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, L981F at least any high-risk mutant strain.
  • the strains include B.1 strain, B.1.351 strain, B.1.1.7 strain, P.1 strain, B.1.427 strain, B.1.429 strain, B. .1.526 strain, C.37 strain, B.1.621 strain, B.1.618 strain, C.36.3 strain, 20I/484Q strain, BA.1 strain, BA.1.1 strain and BA.2 at least one of the strains.
  • the present invention provides a SARS-CoV-2 mutant strain ECD immunogenic protein/peptide with improved immunogenicity/antigen trimer stability, said immunogenic protein/peptide comprising SEQ ID No: 8, SEQ ID No:12 or SEQ ID No:16 at least one of the amino acid sequences shown in, or immunogenic fragments and / or immunogenic variants, the ECD immunogenic protein / peptide is a stable prefusion conformation of three aggregate form.
  • the mutant strain contains T19I, L24S, ⁇ 25/27, H49Y, A67V, ⁇ 69/70, T95I, G142D, ⁇ 143/145, ⁇ 145-146, N211I, ⁇ 212/ 212 ⁇ V213G ⁇ G339D ⁇ R346K ⁇ R346S ⁇ S371L ⁇ S373P ⁇ S375F ⁇ T376A ⁇ D405N ⁇ R408S ⁇ K417N ⁇ N440K ⁇ L452Q ⁇ L452R ⁇ S477N ⁇ T478K ⁇ E484A ⁇ E484K ⁇ E484Q ⁇ F490S ⁇ Q493R ⁇ G496S ⁇ Q498R ⁇ N501Y ⁇ High-risk mutant strains of at least any one of Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, and L98
  • the strains include B.1 strain, B.1.351 strain, B.1.1.7 strain, P.1 strain, B.1.427 strain, B.1.429 strain, B. .1.526 strain, C.37 strain, B.1.621 strain, B.1.618 strain, C.36.3 strain, 20I/484Q strain, BA.1 strain, BA.1.1 strain and BA.2 at least one of the strains.
  • the present invention also provides a polynucleotide encoding the ECD immunogenic protein/peptide of the SARS-CoV-2 mutant strain with improved immunogenicity/antigen trimer stability as described above; preferably, the polynucleotide comprises A nucleotide sequence of at least one of SEQ ID No: 7, SEQ ID No: 11 or SEQ ID No: 15.
  • the present invention provides an immunogenic composition
  • an immunogenic composition comprising at least one immunogenic protein/peptide as described above, or at least one immunogenic protein/peptide encoding an immunogenic/antigenic trimer as described above
  • Polynucleotides of ECD immunogenic proteins/peptides of mutant strains of SARS-CoV-2 with increased stability and
  • the immunogenic composition also includes an adjuvant.
  • the present invention provides an immunogenic composition comprising the amino acid sequences shown in SEQ ID No: 12 and SEQ ID No: 16, or immunogenic fragments and/or immunogens thereof sexual variants, or the amino acid sequences shown in SEQ ID No: 8 and SEQ ID No: 16, or immunogenic fragments and/or immunogenic variants thereof.
  • the present invention provides an immunogenic composition
  • the adjuvant of the immunogenic composition is selected from one or more of the following: aluminum adjuvant, oil emulsion adjuvant, Toll-like receptor (TLR ) agonists, combinations of immunopotentiators, microbial adjuvants, propolis adjuvants, levamisole adjuvants, liposome adjuvants, traditional Chinese medicine adjuvants and small peptide adjuvants; preferably, the oil emulsion adjuvant contains squal Alkenes;
  • TLR Toll-like receptor
  • MPL monophosphoryl lipid A
  • the present invention also provides the immunogenic protein/peptide as described above, polynucleotides encoding the immunogenic protein/peptide as described above and/or comprising the immunogenic protein/peptide as described above or encoding the immunogenic protein/peptide as described above
  • the immunogenic composition of the polynucleotide of the original protein/peptide is applied to the purposes of preventing or treating the diseases caused by the mutant strain of SARS-CoV-2; meanwhile, the present invention also provides the above-mentioned immunogenic protein /peptides, polynucleotides encoding immunogenic proteins/peptides as described above and/or immunogenic combinations comprising immunogenic proteins/peptides as described above or polynucleotides encoding said immunogenic proteins/peptides
  • the medicine is applied to the purposes of preparing vaccines or medicines for preventing or treating diseases caused by SARS-CoV-2 mutant strains.
  • Figure 1 is a schematic diagram of the primary structure (A) and high-order structure (B, refer to PDB:6XLR) of the modified S-ECD.
  • FIG. 2 shows the analysis results of the purity of the recombinant spike protein extracellular domain (S-ECD) trimer protein, wherein (A) is the non-reducing SDS-PAGE profile, and (B) is the SEC-HPLC profile.
  • S-ECD spike protein extracellular domain
  • Fig. 3 shows the negative staining electron microscope results of the recombinant spike protein extracellular domain (S-ECD) trimer protein.
  • Figure 4 shows SCTV01C-TM8 vaccine immunization after 6-8 weeks of Balb/c mice (A), 6-8 weeks of C57BL/6 mice (B) and 7-8 months old Balb/c mice (C) Test results of serum antibody titers.
  • FIG. 5 shows that SCTV01C-TM8 vaccine immunizes 6-8 weeks of Balb/c mice (A), 6-8 weeks of C57BL/6 mice (B) and 7-8 months old Balb/c mice (C) Detection results of serum pseudovirus neutralization titer.
  • Figure 6 shows the statistical results of ELISpot detection of the number of T lymphocytes secreting IFN- ⁇ (A) and IL-4 (B) in splenocytes under different peptide library stimulation conditions after SCTV01C-TM8 vaccine immunization of three mouse models.
  • Figure 7 shows the results of detection of SCTV01C-TM23 vaccine cynomolgus monkey immune serum antibody titer (A) and pseudovirus neutralization titer (B).
  • Figure 8 shows the statistical results of ELISpot detection of the number of T lymphocytes secreting IFN- ⁇ (A) and IL-4 (B) in PBMCs under different peptide library stimulation conditions after SCTV01C-TM23 vaccine immunization in cynomolgus monkeys.
  • FIG. 9 shows the results of titer detection of partial antibody against Foldon in cynomolgus monkey immune serum of SCTV01C-TM23 vaccine.
  • Fig. 10 shows the statistical results of ELISpot detection of the number of T lymphocytes secreting IFN- ⁇ (A) and IL-4 (B) in PBMCs under stimulation conditions of Foldon protein or 6P+Furin mutation modified peptide library.
  • Figure 11 shows the results of detection of SCTV01C-TM22 vaccine mouse immune serum antibody titer (A) and pseudovirus neutralization titer (B).
  • Fig. 12 shows the results of detection of pseudovirus broad-spectrum neutralization titer in immune sera of SCTV01C-TM22 vaccine mice.
  • Figure 13 shows the results of the detection of pseudovirus neutralization titer in immune sera of mice with TM8+TM23 bivalent vaccine.
  • Figure 14 shows the statistical results of ELISpot detection of the number of T lymphocytes secreting IFN- ⁇ (A) and IL-4 (B) in splenocytes under different peptide library stimulation conditions after immunizing mice with TM8+TM23 bivalent vaccine.
  • Figure 15 shows the detection results of the neutralization titer of TM22+TM23 bivalent vaccine mouse immune sera to B.1 strain, B.1.351 strain and B.1.1.7 strain pseudovirus.
  • Figure 16 shows that TM22+TM23 bivalent vaccine mouse immune serum is to B.1.526 virus strain, C.37 virus strain, B.1.621 virus strain, B.1.618 virus strain, C.36.3 virus strain and 20I/484Q virus Strain pseudovirus neutralization titer test results.
  • Fig. 17 shows the detection results of the neutralization titer of TM22+TM23 bivalent vaccine mouse immune serum to BA.1 strain, BA.1.1 strain and BA.2 strain pseudovirus.
  • compositions refers to the inclusion of specific components without excluding any other components.
  • Terms such as “consisting essentially of” allow for the inclusion of other ingredients or steps that do not impair the novel or essential characteristics of the invention, ie they exclude other unrecited ingredients or steps that impair the novel or essential characteristics of the invention.
  • Consisting of means the inclusion of a specific ingredient or group of ingredients and the exclusion of all other ingredients.
  • antigen refers to a foreign substance that is recognized (specifically bound) by antibodies or T cell receptors, but which does not definitively induce an immune response. Exogenous substances that induce specific immunity are called “immunizing antigens” or “immunogens”. By “hapten” is meant an antigen that by itself does not elicit an immune response (although a combination of several molecules of the hapten, or a combination of a hapten and a macromolecular carrier may elicit an immune response).
  • a “humoral immune response” is an antibody-mediated immune response and involves the introduction and production of antibodies that recognize and bind with affinity to the antigen in the immunogenic composition of the invention
  • a “cell-mediated immune response” is composed of T cells and and/or other white blood cell-mediated immune responses.
  • a “cell-mediated immune response” is induced by presenting an antigenic epitope associated with a major histocompatibility complex (MHC) class I or class II molecule, CD1 or other atypical MHC-like molecule.
  • MHC major histocompatibility complex
  • immunogenic composition refers to any pharmaceutical composition containing an antigen, such as a microorganism or a component thereof, which is useful for eliciting an immune response in an individual.
  • immunogenicity means that an antigen (or an epitope of an antigen) such as the coronavirus spike protein receptor binding region or an immunogenic composition induces humoral or cell-mediated immune response, or both.
  • a “protective" immune response refers to the ability of an immunogenic composition to elicit a humoral or cell-mediated immune response, or both, to protect an individual from infection.
  • the protection conferred need not be absolute, i.e., the infection need not be completely prevented or eradicated, so long as there is a statistically significant improvement relative to a control population of individuals (e.g., infected animals not administered the vaccine or immunogenic composition) . Protection may be limited to moderation of severity or rapidity of onset of symptoms of infection.
  • immunogenic amount and “immunologically effective amount” are used interchangeably herein to refer to an antigen or immunogenic composition sufficient to elicit an immune response (cellular (T cell) or humoral (B cell or antibody) response or both. or, as measured by standard assays known to those skilled in the art).
  • the effectiveness of an antigen as an immunogen can be measured, for example, by a proliferation assay, by a cell lysis assay, or by measuring the level of B cell activity.
  • polypeptide and “protein” are used interchangeably herein to refer to a polymer of contiguous amino acid residues.
  • nucleic acid refers to RNA, DNA, cDNA or cRNA and derivatives thereof, such as those containing modified backbones. It is to be understood that the invention provides polynucleotides comprising sequences that are complementary to the sequences described herein.
  • a “polynucleotide” contemplated in the present invention includes the forward strand (5' to 3') and the reverse complementary strand (3' to 5').
  • the polynucleotides according to the invention can be prepared in different ways (e.g. by chemical synthesis, by gene cloning, etc.) and can take various forms (e.g. linear or branched, single or double stranded, or hybrids thereof , primers, probes, etc.).
  • immunogenic protein/peptide includes a polypeptide that is immunologically active in the sense that it is capable of eliciting a humoral and/or cell-type immune response against the protein once administered to a host.
  • a protein fragment according to the invention comprises or consists essentially of or consists of at least one epitope or antigenic determinant.
  • an "immunogenic" protein or polypeptide includes the full-length sequence of the protein, an analog thereof, or an immunogenic fragment thereof.
  • immunogenic fragment refers to a fragment of a protein that contains one or more epitopes that elicit an immune response as described above.
  • immunogenic protein/peptide also covers deletions, additions and substitutions to sequences so long as the polypeptide functions to generate an immune response as defined herein, ie “immunogenic variants”.
  • the SCTV01C recombinant protein vaccine provided by the present invention is transformed based on the extracellular domain (ECD, including S1 and S2 parts) of the SARS-CoV-2 spike protein.
  • ECD extracellular domain
  • the known natural spike protein of SARS-CoV-2 has a trimeric structure. During its production and infection function, the completion of the membrane fusion process is easily detected by the RRAR site between S1 and S2. Proteases in the Golgi apparatus and on the cell surface cut open, and then S1 falls off, and the S2 structure changes from a prefusion conformation to a postfusion conformation, thereby completing membrane fusion (Cai, Y., J. Zhang, and T. Xiao, Distinct conformational states of SARS-CoV-2 spike protein. 2020.369(6511): p.1586-1592.).
  • the present invention carried out the following three-part modification on the basis of the S protein of different strain variants:
  • the Furin site is modified and removed in the SCTV01C recombinant protein vaccine, that is, the amino acid sequence at positions 679 to 688 is fixed as NSPGSASSVA, so as to reduce the possibility of S1 breaking and falling off.
  • the S-2P i.e. mutating amino acids at positions 986 and 987 to proline
  • S-2P i.e. mutating amino acids at positions 986 and 987 to proline
  • Mercado,N.B.,et al.,Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques.2020.586(7830):p.583-588 .
  • the present invention also introduces a HexaPro mutation that can effectively improve the stability without affecting its three-dimensional structure (that is, in addition to the S-2P mutation, the amino acids at positions 817, 892, 899 and 942 are mutated into proline) (Hsieh, C.L., et al., Structure-based Design of Prefusion-stabilized SARS-CoV-2 Spikes. bioRxiv, 2020.). These mutation sites are all located at the N-terminus or Loop region of the ⁇ -helix in S2. After mutation to the proline (P) type with this secondary structure tendency, it can effectively reduce the allosteric tendency of S2 and stabilize the prefusion of S2 Conformation.
  • P proline
  • trimerization module T4foldon to the C-terminus of the vaccine molecule.
  • This module is derived from the C-terminal domain of fibrin of T4 phage and has 27 amino acids. T4foldon has been used in RSV candidate vaccines, and has been shown to be safe in Phase I clinical studies (Crank, M.C., A proof of concept for structure-based vaccine design targeting RSV in humans. 2019.365(6452): p. 505-509.).
  • B.1 strain, B.1.351 and B.1.1.7 strain respectively through the expression vector of the S-ECD trimer protein antigen of above-mentioned transformation, and routinely carry out the expression recombinant S-ECD trimer protein Purity and stability analysis, preparation of corresponding vaccines, namely B.1 strain SCTV01C-TM8 vaccine, B.1.351 strain SCTV01C-TM23 vaccine and B.1.1.7 strain SCTV01C-TM22 vaccine.
  • the ECD trimer immunogenic protein/peptide of the present invention shows excellent immunogenicity in mice and cynomolgus monkeys, and can maintain long-term humoral and cellular immune responses.
  • mice After immunizing mice with the prepared B.1 strain SCTV01C-TM8 vaccine, the immunological assay, the immunological assay of the B.1.351 strain SCTV01C-TM23 vaccine in cynomolgus monkeys, and the B.1.1.7 strain SCTV01C-
  • the immunological determination of TM22 vaccine in mice all shows that these three vaccines prepared by the present invention can produce sufficient titer antibody immune response in experimental animals; SCTV01C-TM8+SCTV01C-TM23 bivalent vaccine and SCTV01C-TM22+SCTV01C -
  • the mouse immunological evaluation of the TM23 bivalent vaccine also suggests that the bivalent vaccine of the present invention has higher and similar neutralizing titers to different strains, so it has better broad-spectrum neutralization ability than the monovalent vaccine,
  • the neutralizing titer of the bivalent vaccine against different mutant strains is much higher than that of the recovered serum against the SARS-CoV-2 strain whose genome sequence number is GenBank Accession No. MN90
  • SCTV01C-TM8 contains a 3708bp gene fragment, and the target gene fragment was obtained from the template pSE-CoV2-S-ECDTM2-T4F-trimer by PCR splicing.
  • the expression vector pGS3-2-CoV2-S-ECDTM22-T4F-trimer was constructed by the In-fusion method into the pGS3-2-SCT-1 expression vector digested with Kpn I+Not I enzymes.
  • the SCTV01C-TM8 gene fragment was obtained by PCR amplification, and the expression vector of pD2535nt-HDP stable strain digested with Xba I+Asc I was constructed by In-fusion method , the expression vector pD2535nt-CoV2-S-ECDTM8-T4F-trimer of SCTV01C-TM8 was obtained.
  • SCTV01C-TM22 contains a 3699bp gene fragment, and the target gene fragment was obtained from the template pD2535nt-CoV2-S-ECDTM8-T4F-trimer by PCR splicing.
  • the expression vector pGS5-CoV2-S-ECDTM22-T4F-trimer was constructed by the In-fusion method into the pGS5-SCT-1 expression vector digested with Kpn I+Not I enzymes.
  • the SCTV01C-TM22 gene fragment was obtained by PCR amplification, and constructed into the pD2535nt-HDP stable strain expression vector digested with Xba I+Asc I by the In-fusion method, The expression vector pD2535nt-CoV2-S-ECDTM22-T4F-trimer of SCTV01C-TM22 was obtained.
  • SCTV01C-TM23 contains a 3699bp gene fragment.
  • the target gene fragment was obtained from the template pD2535nt-CoV2-S-ECDTM8-T4F-trimer by PCR splicing, and constructed into pD2535nt-HDP digested with Xha I+Asc I by the In-fusion method Among the stable strain expression vectors, the expression vector pD2535nt-CoV2-S-ECDTM23-T4F-trimer of SCTV01C-TM23 was obtained.
  • the target gene constructed above was chemically transferred into HD-BIOP3(GS-) cells (Horizon), cultured in a self-developed serum-free medium, and a cell line with stable expression was obtained through MSX pressurized screening, and cultured for 14 hours. Days later, the culture supernatant was obtained by centrifugation and filtration.
  • the culture supernatant was first captured by cation exchange chromatography (POROS XS, Thermo) and eluted with high-salt buffer; then anion chromatography (NanoGel-50Q, NanoMicro) combined mode and mixed anion chromatography (DiamondMIX-A , Borglon) flow-through mode for further purification to remove product and process-related impurities; secondly, use low pH incubation and virus removal filtration (Planova) to inactivate and remove viruses, and finally use ultrafiltration membrane packs (Millipore ) for ultrafiltration to citrate buffer. S-ECD trimer expression level >500mg/L.
  • Example 2 Analysis of the purity and stability of the trimer protein of the new coronavirus recombinant spike protein extracellular domain (S-ECD)
  • SDS-PAGE specific operation steps (1) SDS-PAGE gel preparation: 3.9% stacking gel, 7.5% separating gel; (2) Boil the sample at 100°C for 2 minutes, and load 8 ⁇ g of sample after centrifugation; (3) Decolorize after Coomassie brilliant blue staining .
  • SEC-HPLC operation step is: (1) instrument: liquid chromatography system (Agilent company, model: Agilent1260), water-soluble size exclusion chromatographic column (Sepax company, model: SRT-C SEC-500 chromatographic column); (2 ) Mobile phase: 200mM NaH 2 PO 4 , 100mM Arginine, pH 6.5, 0.01% isopropanol (IPA); (3) Loading amount is 80 ⁇ g; (3) Detection wavelength is 280nM, analysis time is 35min, flow rate is 0.15mL /min.
  • SCTV01C-TM8, SCTV01C-TM22, and SCTV01C-TM23 proteins are homotrimeric structures due to their non-covalent hydrophobic interactions.
  • monomeric molecules with a molecular weight of about 148 KDa were obtained (Fig. 2), and the purities were 98.0%, 98.8%, and 97.7%, respectively.
  • SEC-HPLC shows that the main peaks have a purity of 96.6%, 95.5%, and 96.6%, respectively, and the proportions of aggregates and fragments are relatively small, and the average molecular weight of the main peak is 530KDa.
  • Figure 2 is a representative test result of SCTV01C-TM8.
  • Recombinant S-ECD trimer protein was stored at -80°C for 8h, then transferred to 25°C for 0.5h (F/T-4C), and then frozen and thawed 4 times, and its trimer was analyzed by SEC-HPLC content changes.
  • the purified SCTV01C-TM8 protein was pre-diluted with PBS to 20 ⁇ g/mL or 60 ⁇ g/mL, and the diluted antigen was mixed with MF59 (source: China Cell Engineering Co., Ltd., the same below) in equal volumes to obtain a final antigen concentration of 10 ⁇ g/mL. mL or 30 ⁇ g/mL of finished vaccine containing MF59.
  • MF59 source: China Cell Engineering Co., Ltd., the same below
  • mice 6-8 weeks of Balb/c mice, 6-8 weeks of C57BL/6 mice, 7-8 months old Balb/c mice (source: Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.), intramuscular injection of 0.1mL containing The finished vaccine with MF59 adjuvant.
  • Three types of mice were immunized with 1 ⁇ g, 3 ⁇ g and 3 ⁇ g of antigens respectively. A total of 3 immunizations were carried out, and the immunization interval was 3 weeks. Two weeks after the first immunization, orbital blood was collected every other week, and serum was collected by centrifugation at 4500rpm for 15 minutes for subsequent serological immune analysis.
  • VSV ⁇ G-Luc-G The replication-defective vesicular stomatitis virus (VSV ⁇ G-Luc-G) in which the VSV-G protein gene in the viral genome was replaced by a luciferase reporter gene was used as a vector, and carried out in a cell line expressing Spike and its mutant proteins Amplification preparation, prepared by Shenzhou Cell Engineering Co., Ltd., the same below), mixed evenly and placed in a 37°C, 5% CO 2 incubator for 1h incubation.
  • VSV ⁇ G-Luc-G The replication-defective vesicular stomatitis virus (VSV ⁇ G-Luc-G) in which the VSV-G protein gene in the viral genome was replaced by a luciferase reporter gene was used as a vector, and carried out in a cell line expressing Spike and its mutant proteins Amplification preparation, prepared by Shenzhou Cell Engineering Co., Ltd., the same below), mixed evenly and placed in a 37°C, 5% CO 2 incubator for
  • Serum-free cell wells containing pseudovirus were used as positive controls, and cell wells without serum and pseudoviruses were used as negative controls.
  • 3 ⁇ 10 4 293FT-ACE2 cells were inoculated at 100 ⁇ L/well, mixed well, and placed in a 37° C., 5% CO 2 incubator for static culture for about 20 hours. After the culture, remove the culture supernatant, add 50 ⁇ L/well of 1 ⁇ Passive lysis buffer, and mix well to lyse the cells.
  • the antibody titer results after immunization are shown in Figure 4.
  • SCTV01C-TM8 was tested in 6-8 week Balb/c mice, 6-8 week C57BL/6 mice and 7-8 month old Balb/c mice. All models can induce high-titer antibody immune responses.
  • the pseudovirus neutralization titer results of the SARS-CoV-2 strain whose genome sequence number is GenBank Accession No. MN908947.3 are shown in Figure 5.
  • SCTV01C-TM8 can induce high-titer pseudoviruses in all three mouse models Neutralizing antibodies. After immunization, the neutralizing titer of the pseudovirus is significantly higher than that of the SARS-CoV-2 strain whose genome sequence number is GenBank Accession No. MN908947.3 by the serum (HCS) of recovered patients with new crown infection, 3 free for 7 days
  • the neutralizing titers of HCS were 33.3 times, 13.9 times and 3.9 times respectively.
  • Mouse splenocytes were isolated, and 100 ⁇ L/well of mouse splenocytes were inoculated on pre-treated ELISpot well plates (source: Mabtech, the same below), at a cell inoculation density of 2 ⁇ 10 5 cells/well. Then 100 ⁇ L/well was added to RBD, S1, S2 or S protein peptide library with a final concentration of 2 ⁇ g/mL (15 amino acids/peptide, overlapping 11 amino acids, source: Beijing Zhongke Yaguang Biotechnology Co., Ltd. Synthesis, the same below ), and incubated in a 37°C, 5% CO 2 incubator for about 20h.
  • the cell supernatant of the ELISpot well plate was removed, the plate was washed 5 times with PBS, and then 100 ⁇ L/well of the diluted detection antibody was added. After incubation for 2 hours, the plate was washed 5 times with PBS, and diluted Streptavidin-ALP (1:1000) was added to 100 ⁇ L/well. After incubation at room temperature for 1 h, the plate was washed 5 times with PBS, and then 100 ⁇ L/well of BCIP/NBT-plus substrate filtered with a 0.45 ⁇ m filter membrane was added. Keep away from light at room temperature for 10-30 minutes to develop color until clear spots appear, and stop with deionized water.
  • the ELISpot well plate Place the ELISpot well plate in a cool place at room temperature, wait for it to dry naturally, and analyze the results with an enzyme-linked spot analyzer.
  • the number of antigen-specific IFN- ⁇ or IL-4 secreting positive T cells was represented by SFC (Spot-forming cells) per 10 6 mouse splenocytes, and the GrapPad Prism software was used for data statistics.
  • SCTV01C-TM8 can induce higher Th1 (IFN- ⁇ ) and Th2 (IL- 4) Cellular response.
  • Example 4 Immunological evaluation of B.1.351 strain SCTV01C-TM23 vaccine in cynomolgus monkeys
  • the purified SCTV01C-TM23 protein was pre-diluted with PBS to 120 ⁇ g/mL, and the diluted antigen was mixed with MF59 in equal volumes to obtain a finished vaccine containing MF59 with a final antigen concentration of 60 ⁇ g/mL.
  • Cynomolgus monkeys (source: Guangxi Xiongsen Primate Experimental Animal Breeding and Development Co., Ltd.), intramuscularly injected 0.5 mL of finished vaccine containing MF59 adjuvant, with an antigenic amount of 30 ⁇ g.
  • a total of 2 immunizations were performed with an interval of 3 weeks.
  • Two weeks after the first immunization venous blood was collected every other week, and non-anticoagulant centrifuge tubes were pre-incubated in ice water before use. After the blood sample was collected, it was transferred to a centrifuge tube without anticoagulant for temporary storage, and then centrifuged at 3000 ⁇ g for 10 min at 2-8°C. Separated serum samples were subjected to subsequent serological immunoassays.
  • Blood samples collected using anticoagulant centrifuge tubes were routinely isolated from peripheral blood lymphocytes (PBMCs) for cellular immunoassays.
  • PBMCs peripheral blood lymphocytes
  • Anti-SCTV01C-TM23 or Foldon part-specific IgG antibodies in serum of cynomolgus monkeys were detected by ELISA. Dilute SCTV01C-TM23 with coating solution to 2 ⁇ g/mL, add 100 ⁇ L/well into the microplate, and incubate overnight at 2-8°C. Wash the plate 3 times and pat dry, add 300 ⁇ L/well of 2% casein-PBST blocking solution, and block at room temperature for at least 1 h.
  • MN908947.3 is still used for T cell immune detection.
  • Monkey PBMCs were isolated by density gradient centrifugation, and 100 ⁇ L/well of PBMCs cells were inoculated on the pre-treated ELISpot well plate at a cell seeding density of 2.5 ⁇ 10 5 cells/well, and then RBD with a final concentration of 2 ⁇ g/mL was added to 100 ⁇ L/well , S1 or S protein peptide library, and incubated in a 37°C, 5% CO 2 incubator for about 20 hours.
  • the cell supernatant of the ELISpot well plate was removed, the plate was washed 5 times with PBS, and then 100 ⁇ L/well of the diluted detection antibody with a final concentration of 1 ⁇ g/mL was added. After incubation for 2 hours, the plate was washed 5 times with PBS, 100 ⁇ L/well of diluted Streptavidin-ALP (1:1000) was added, and the plate was washed 5 times with PBS after incubation at room temperature for 1 hour. Then 100 ⁇ L/well of BCIP/NBT-plus substrate filtered with a 0.45 ⁇ m filter membrane was added, and the color was developed for 10-30 minutes at room temperature in the dark until clear spots appeared and terminated with deionized water.
  • SCTV01C-TM23 can induce Th1 (IFN- ⁇ ) and Th2 (IL-4) cell responses against RBD, S1, and S polypeptide libraries in cynomolgus monkeys after 2 immunizations and 7 days.
  • the Foldon-containing RSV F recombinant protein (RSV-F-Foldon) was used as the coating antigen to detect the antibody titer against Foldon after SCTV01C-TM23 was immunized with cynomolgus monkeys with reference to Example 4.3.
  • the results are shown in Figure 9, the immunogenicity induced by Foldon in the S-ECD trimer molecule is very weak, and the immune titer of SCTV01C-TM23 in cynomolgus monkeys at different time points is 54-76 times that of Foldon.
  • the RSV-F-Foldon protein or the peptide library containing the "6P" mutation and the Furin site mutation were used to detect the effect of the introduction of Foldon and the mutation modification on the immune response of T cells with reference to Example 4. The results are shown in Figure 10. After 2 immunizations and 7 days, Foldon had a very low T cell immune response in cynomolgus monkeys, and the "6P" and Furin site mutations had no effect on the cellular immune response.
  • the purified SCTV01C-TM22 protein was pre-diluted to 20 ⁇ g/mL with PBS, and the diluted antigen was mixed with MF59 in equal volumes to obtain a finished vaccine containing MF59 with a final antigen concentration of 10 ⁇ g/mL.
  • C57BL/6 mice (source: Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.) at 6-8 weeks were intramuscularly injected with 0.1 mL of the finished vaccine containing MF59 adjuvant, and the amount of antigen was 1 ⁇ g.
  • a total of 3 immunizations were performed with an interval of 2 weeks.
  • SCTV01C-TM22 as the coating antigen, refer to Example 3.3 to detect the antibody titer after SCTV01C-TM22 immunized C57BL/6 mice.
  • B.1.1.7 Strain pseudovirus neutralizing titer results As shown in Figure 11, SCTV01C-TM22 can induce high titers of pseudovirus neutralizing antibodies in C57BL/6 mice.
  • the neutralizing titer after immunization is higher than the neutralizing titer of the SARS-CoV-2 strain whose genome sequence number is GenBank Accession No. MN908947.3 by the serum (HCS) of recovered patients with new crown infection.
  • the neutralizing titer is 56.5 times that of HCS.
  • the neutralizing activity test results of SCTV01C-TM22 vaccine showed that the monovalent vaccine could not produce high-titer neutralizing antibodies against mutant strains, and had poor broad-spectrum neutralizing activity.
  • the following examples prepared bivalent vaccines containing S-ECD proteins of different mutant strains, and tested the broad-spectrum of the bivalent vaccine. Immunological evaluation was carried out with the ability.
  • the purified SCTV01C-TM8 and SCTV01C-TM23 proteins were pre-diluted to 20 ⁇ g/mL with PBS, and the diluted antigen was mixed with MF59 in equal volumes to obtain a finished monovalent vaccine containing MF59 with a final antigen concentration of 10 ⁇ g/mL.
  • the SCTV01C-TM8 and SCTV01C-TM23 proteins were pre-diluted with PBS to 40 ⁇ g/mL, and the diluted antigens were mixed in equal volumes to obtain a mixed antigen sample with a final antigen concentration of 20 ⁇ g/mL.
  • the mixed antigen sample was mixed with MF59 in equal volumes to obtain the finished bivalent vaccine TM8+TM23 containing MF59 with the final antigen concentration of SCTV01C-TM8 and SCTV01C-TM23 both being 10 ⁇ g/mL.
  • Example 5.3 detect the immune serum of the bivalent vaccine for 2 immunizations for 7 days to different mutant strains (B.1 strain, B.1.351 strain, B.1.1.7 strain, P.1 strain, and B.1.429 virus strain and/or B.1.427 strain) pseudovirus neutralization titer.
  • B.1 strain, B.1.351 strain, B.1.1.7 strain, P.1 strain, and B.1.429 virus strain and/or B.1.427 strain pseudovirus neutralization titer.
  • the results are shown in Figure 13, the SCTV01C-TM8 monovalent vaccine has a higher neutralizing titer to the B.1 strain and the B.1.1.7 strain pseudovirus, but to the B.1.351 strain and the P.1 strain pseudovirus
  • the neutralizing titer is low, and the reduction factor is about 5-11 times of the neutralizing titer of the B.1 strain.
  • SCTV01C-TM23 monovalent vaccine has higher neutralizing titer to B.1.351 strain and P.1 strain pseudovirus, but to B.1 strain, B.1.1.7 strain, and B.1.429 strain and/or Or the neutralizing titer of the B.1.427 strain pseudovirus is low, and the reduction factor is about 4 to 12 times of the neutralizing titer of the B.1.351 strain.
  • the TM8+TM23 bivalent vaccine has high and similar neutralizing titers against different strains, so it has better broad-spectrum neutralizing ability than the monovalent vaccine.
  • the neutralizing titer of the bivalent vaccine against different mutant strains is much higher than that of the recovered serum against the SARS-CoV-2 strain whose genome sequence number is GenBank Accession No. MN908947.3.
  • MN908947.3 has a higher T cell response after stimulating the splenocytes of mice immunized with monovalent vaccine and bivalent vaccine. And T cell immune response is similar.
  • the S-peptide library and the S+TM23-mix mixed peptide library stimulated similar T cell responses, indicating that the differential polypeptides of the B.1.351 strain had no T-cell immune response, and further proved that there were conserved T-cell epitopes among mutant strains.
  • the purified SCTV01C-TM22 and SCTV01C-TM23 proteins were pre-diluted with PBS to 20 ⁇ g/mL, and the diluted antigen was mixed with MF59 in equal volumes to obtain a finished monovalent vaccine containing MF59 with a final antigen concentration of 10 ⁇ g/mL.
  • the SCTV01C-TM22 and SCTV01C-TM23 proteins were pre-diluted with PBS to 40 ⁇ g/mL, and the diluted antigens were mixed in equal volumes to obtain a mixed antigen sample with a final antigen concentration of 20 ⁇ g/mL.
  • the mixed antigen sample was mixed with MF59 in equal volumes to obtain the finished bivalent vaccine TM22+TM23 containing MF59 with the final antigen concentration of SCTV01C-TM22 and SCTV01C-TM23 both being 10 ⁇ g/mL.
  • Example 5.3 to detect the pseudovirus neutralization titer of the bivalent vaccine 2-immune 7-day immune serum to different mutant strains (B.1 strain, B.1.351 strain and B.1.1.7 strain).
  • the results are shown in Figure 15.
  • SCTV01C-TM22 monovalent vaccine has a higher neutralizing titer to B.1.1.7 strains and B.1 strain pseudoviruses, but the neutralizing ability to B.1.351 strains decreased by about It is 8.8 times of the neutralizing titer of B.1.1.7 strain.
  • SCTV01C-TM23 monovalent vaccine has higher neutralizing titer to B.1.351 strain, but lower neutralizing potency to B.1.1.7 strain and B.1 strain, and the reduction times are B.1.351 strain neutralization The potency is 6.4 times and 5.1 times.
  • the TM22+TM23 bivalent vaccine has high and similar neutralizing titers against different strains, so it has better broad-spectrum neutralizing ability than the monovalent vaccine.
  • the neutralizing titer of the bivalent vaccine against different mutant strains is much higher than that of the recovered serum against the SARS-CoV-2 strain whose genome sequence number is GenBank Accession No. MN908947.3.
  • the immune serum of the bivalent vaccine was tested for 2 immunizations for 7 days to different mutant strains of SARS-CoV-2 (B.1.526 strain, C.37 strain, B.1.621 strain, B.1.618 strain, C. 36.3 strain and 20I/484Q strain) pseudovirus neutralization titer.
  • SCTV01C-TM22+TM23 bivalent vaccine has Higher and similar neutralizing titer, so it has better broad-spectrum neutralizing ability than monovalent vaccine.
  • Example 5.3 detect the pseudovirus neutralizing titer of the bivalent vaccine for 2 immune sera in 21 days to different mutant strains (BA.1 strain, BA.1.1 strain, BA.2 strain) of SARS-CoV-2Omicron. The results are shown in Figure 17.
  • the GMT value has a certain degree of increase (increasing multiples are 18.3 times, 25.3 times, 10.4 times respectively), indicating that the SCTV01C-TM22+TM23 bivalent vaccine has different effects on SARS-CoV-2Omicron variation. All strains have higher neutralizing titers, so they have better broad-spectrum neutralizing ability than monovalent vaccines.
  • bivalent vaccines have broad-spectrum neutralization capabilities against different mutant strains, and are expected to produce cross-protection capabilities against multiple mutant strains and improve the protection rate against mutant strain infection.

Abstract

La présente invention relève du domaine de la vaccinologie moléculaire, et concerne un procédé permettant d'améliorer l'immunogénicité/la stabilité de trimère antigénique d'un antigène à domaine extracellulaire (ECD) d'une souche mutante de SARS-CoV-2, et une protéine/peptide immunogène ECD, ayant une immunogénicité/stabilité de trimère antigénique améliorée, de la souche mutante de SARS-CoV-2. La présente invention comprend, mais sans caractère limitatif, un ECD d'une protéine de spicule (protéine S) d'une souche de SARS-CoV-2, d'une souche B.1, d'une souche B.1.1.7 ou d'une souche B.1.351 ayant un numéro de séquence génomique du numéro d'accès GenBank MN908947.3; par l'introduction d'un homotrimère formé par un site de mutation et une structure assistée par trimérisation, l'immunogénicité/la stabilité de trimère antigénique de l'antigène ECD est améliorée. Un vaccin comprend en outre un adjuvant pharmaceutiquement acceptable. Une composition de vaccin a une excellente immunogénicité chez les souris et le Macaca fascicularis, et peut maintenir des réponses immunitaires tumorales et cellulaires à long terme. Un vaccin à base de protéine trimère recombinante peut être utilisé pour prévenir des maladies associées aux infections par le SARS-CoV-2.
PCT/CN2022/095609 2021-05-31 2022-05-27 Procédé pour améliorer l'immunogénicité/la stabilité de trimère antigénique d'un antigène ecd de souche mutante de sars-cov-2 WO2022253134A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280034785.7A CN117295771A (zh) 2021-05-31 2022-05-27 一种提高SARS-CoV-2突变毒株ECD抗原免疫原性/抗原三聚体稳定性的方法

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202110606512.2 2021-05-31
CN202110606512.2A CN115477703A (zh) 2021-05-31 2021-05-31 一种提高SARS-CoV-2突变毒株ECD抗原免疫原性/抗原三聚体稳定性的方法
CN202111237604 2021-10-22
CN202111237604.4 2021-10-22

Publications (1)

Publication Number Publication Date
WO2022253134A1 true WO2022253134A1 (fr) 2022-12-08

Family

ID=84323876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/095609 WO2022253134A1 (fr) 2021-05-31 2022-05-27 Procédé pour améliorer l'immunogénicité/la stabilité de trimère antigénique d'un antigène ecd de souche mutante de sars-cov-2

Country Status (2)

Country Link
CN (1) CN117295771A (fr)
WO (1) WO2022253134A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111603557A (zh) * 2020-06-15 2020-09-01 苏州奥特铭医药科技有限公司 一种包膜替换型病毒载体疫苗及其构建方法
CN112076315A (zh) * 2020-08-25 2020-12-15 中国农业科学院生物技术研究所 新冠病毒s蛋白和铁蛋白亚基融合的纳米抗原颗粒、新冠疫苗及其制备方法和应用
CN112358533A (zh) * 2020-10-30 2021-02-12 上海泽润生物科技有限公司 重组刺突蛋白及其制备方法和用途
CN112480217A (zh) * 2020-11-30 2021-03-12 广州市锐博生物科技有限公司 基于SARS-CoV-2的S抗原蛋白的疫苗和组合物
CN112661865A (zh) * 2020-12-31 2021-04-16 厚朴生物科技(苏州)有限公司 针对新型冠状病毒的疫苗以及其应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111603557A (zh) * 2020-06-15 2020-09-01 苏州奥特铭医药科技有限公司 一种包膜替换型病毒载体疫苗及其构建方法
CN112076315A (zh) * 2020-08-25 2020-12-15 中国农业科学院生物技术研究所 新冠病毒s蛋白和铁蛋白亚基融合的纳米抗原颗粒、新冠疫苗及其制备方法和应用
CN112358533A (zh) * 2020-10-30 2021-02-12 上海泽润生物科技有限公司 重组刺突蛋白及其制备方法和用途
CN112480217A (zh) * 2020-11-30 2021-03-12 广州市锐博生物科技有限公司 基于SARS-CoV-2的S抗原蛋白的疫苗和组合物
CN112661865A (zh) * 2020-12-31 2021-04-16 厚朴生物科技(苏州)有限公司 针对新型冠状病毒的疫苗以及其应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GUO LIANG, BI WENWEN, WANG XINLING, XU WEI, YAN RENHONG, ZHANG YUANYUAN, ZHAO KAI, LI YANING, ZHANG MINGFENG, BAO XINGYUE, CAI XIA: "Engineered Trimeric ACE2 Binds and Locks "Three-up" Spike Protein to Potently Inhibit SARS-CoVs and Mutants", BIORXIV, 1 September 2020 (2020-09-01), pages 1 - 35, XP055938126, DOI: 10.1101/2020.08.31.274704 *
XU ZHIJUE, KU XIN, TIAN JIAQI, ZHANG HAN, HOU JINGLI, ZHANG CAN, SHI JINGJING, LI YANG, KAJI HIROYUKI, TAO SHENG-CE, KUNO ATSUSHI,: "Altered O-glycosylation Level of SARS-CoV-2 Spike Protein by Host O-glycosyltransferase Strengthens Its Trimeric Structure", BIORXIV, 6 April 2021 (2021-04-06), pages 1 - 30, XP093009388, DOI: 10.1101/2021.04.06.438614 *

Also Published As

Publication number Publication date
CN117295771A (zh) 2023-12-26

Similar Documents

Publication Publication Date Title
US9731006B2 (en) Attenuated recombinant vesicular stomatitis virus vaccine vectors comprising modified matrix proteins
WO2021076010A1 (fr) Agent pharmaceutique pour induire une immunité spécifique contre le sras-cov-2
Li et al. Self-assembling nanoparticle vaccines displaying the receptor binding domain of SARS-CoV-2 elicit robust protective immune responses in rhesus monkeys
CA2916789C (fr) Proteines de matrice modifiees du virus de la stomatite vesiculaire
WO2023160654A1 (fr) Préparation et utilisation d'un vaccin à composants multiples à base de protéine trimérique de sars-cov-2 recombinante capable d'induire une activité de neutralisation à large spectre
WO2023001259A1 (fr) Préparation et utilisation d'un vaccin de protéine trimère du nouveau coronavirus multivalent recombinant capable d'induire une activité à large spectre et de neutralisation
WO2022253134A1 (fr) Procédé pour améliorer l'immunogénicité/la stabilité de trimère antigénique d'un antigène ecd de souche mutante de sars-cov-2
CN115477703A (zh) 一种提高SARS-CoV-2突变毒株ECD抗原免疫原性/抗原三聚体稳定性的方法
US20190231866A1 (en) Methods for safe induction of cross-clade immunity against hiv infection in humans
WO2024032468A1 (fr) Préparation et utilisation d'un vaccin à protéine trimère du sars-cov-2 à cinq composants recombinant pouvant induire une activité de neutralisation à large spectre
US20210340188A1 (en) Recombinant gp120 protein with v1-loop deletion
JP7360544B2 (ja) Sars-cov-2に対する特異的免疫を誘導するための医薬品
McKechnie et al. Virus-like particle displaying SARS-CoV-2 receptor binding domain elicits neutralizing antibodies and is protective in a challenge model
Liao et al. Co‐delivery of a trimeric spike DNA and protein vaccine with aluminum hydroxide enhanced Th1‐dominant humoral and cellular immunity against SARS‐CoV‐2
US20240002446A1 (en) Proteoliposomes comprising a sars-cov-2 s glycoprotein ectodomain and their use as a vaccine
WO2022053016A1 (fr) Procédé d'amélioration de l'immunogénicité à l'aide d'un conjugué d'antigène de rbd de glyco-coronavirus
Lee et al. T cell immunity of the nonadjuvanted HLA-restricted peptide COVID-19 vaccine
JP2023182231A (ja) Sars-cov-2 s糖タンパク質エクトドメインを含むプロテオリポソームおよびワクチンとしてのそれらの使用
CA3162727A1 (fr) Proteoliposomes comprenant un ectodomaine de glycoproteine du coronavirus 2 du syndrome respiratoire aigu severe (sars-cov-2) et leur utilisation comme vaccin
WO2022197209A1 (fr) Induction d'une immunité contre le sras-cov-2 chez les enfants
EP4010015A1 (fr) Formulations de vaccin contre le chikungunya
CN112521453A (zh) 寨卡病毒优势t细胞表位肽及其在疫苗和诊断中的应用
CN115925824A (zh) SARS-CoV-2编码蛋白来源的T细胞表位多肽NVFAFPFTI及其应用
Wang et al. Protection against lethal subcutaneous challenge of virulent Y. pestis strain 141 using an F1-V subunit vaccine
US20150328304A1 (en) Dengue virus vaccine composition

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22815180

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE