WO2023160654A1 - 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 - Google Patents

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 Download PDF

Info

Publication number
WO2023160654A1
WO2023160654A1 PCT/CN2023/078135 CN2023078135W WO2023160654A1 WO 2023160654 A1 WO2023160654 A1 WO 2023160654A1 CN 2023078135 W CN2023078135 W CN 2023078135W WO 2023160654 A1 WO2023160654 A1 WO 2023160654A1
Authority
WO
WIPO (PCT)
Prior art keywords
strain
mutant
adjuvant
seq
immunogenic
Prior art date
Application number
PCT/CN2023/078135
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
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 CN202380013961.3A priority Critical patent/CN118076646A/zh
Publication of WO2023160654A1 publication Critical patent/WO2023160654A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • 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
    • C07K14/08RNA viruses
    • C07K14/165Coronaviridae, e.g. avian infectious bronchitis virus
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof

Definitions

  • the invention relates to the field of molecular vaccinology, and relates to the preparation and application of a recombinant multi-component novel coronavirus trimeric protein vaccine capable of inducing broad-spectrum neutralization activity.
  • 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. Since the SARS-CoV-2 pandemic, more than 200 new crown vaccines have been developed by various countries. As of December 2, 2022, 50 vaccines have been approved for use or conditional use worldwide, and another 242 vaccines have entered clinical research (https://covid19.trackvaccines.org/vaccines/).
  • Angiotensin-converting enzyme 2 is a common host cell receptor protein of SARS-CoV-2 and SARS-CoV [1] .
  • the trimeric spike protein (Spike) of the virus binds to the ACE2 receptor and 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 [2 ] .
  • the S protein is the main component of the viral envelope and plays an important role in receptor binding, fusion, virus entry and host immune defense.
  • 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, prone to deletion mutations, and such mutations mostly occur in the recurrent deletion regions (RDRs) of the S protein. Deletion or mutation may change the conformation of the S protein, reducing the binding and neutralization of the mutant S protein by antibodies induced by previous vaccine immunization, resulting in a decline in the immune effect of the vaccine and 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 [3,4] .
  • Alpha spreads rapidly and can increase the risk of death by 61% [6] .
  • the results of the neutralization effect study showed that the neutralization ability of the plasma of convalescents or the serum of vaccine immunized persons remained basically unchanged to Alpha, but the neutralization ability of Beta decreased significantly [7-12] .
  • Clinical results also show that Alpha has little effect on the protective effect of the vaccine, while Beta will greatly reduce the protective effect on mild disease [13-16] .
  • the Delta mutation has stronger transmission power, shorter incubation period, faster disease progression, and can reduce the protective effect of the vaccine. Omicron is the most severely mutated strain so far, and its S protein contains about 30 amino acid mutations.
  • the first aspect of the present invention provides a method for improving the ECD antigen immunogenicity/antigen trimer stability of SARS-CoV-2 mutant strains
  • the mutant strain contains A67V, H69del, V70del, T95l, G142D, V143del, Y144del, Y145del, N211del, L212l, ins214EPE, G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K , E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, and L981F are high-risk mutant strains with at least one mutation;
  • the mutant strain is BA.1.
  • the ECD antigen and an adjuvant are co-administered to the subject, and the adjuvant is selected from the group consisting of aluminum adjuvants, oil-emulsion adjuvants, Toll-like receptor (TLR) agonists, combinations of immunopotentiators, microbial adjuvants one or more of propolis adjuvant, levamisole adjuvant, liposome adjuvant, traditional Chinese medicine adjuvant and small peptide adjuvant;
  • TLR Toll-like receptor
  • the oil-emulsion adjuvant contains squalene
  • TLR Toll-like receptor agonists comprising CpG or monophosphoryl lipid A (MPL) adsorbed on aluminum salts; and
  • Combinations of immune boosters include QS-21 and/or MPL.
  • the second aspect of the present invention provides a method for improving the immunogenicity of the SARS-CoV-2 mutant strain ECD antigen immunogenicity/antigen trimer stability, the method comprises the amino acid sequence shown in SEQ ID No: 8 by constructing the code, or Polynucleotides of immunogenic fragments and/or immunogenic variants thereof, thereby expressing the trimeric form of the ECD in a stable prefusion conformation.
  • the mutant strain contains A67V, H69del, V70del, T95l, G142D, V143del, Y144del, Y145del, N211del, L212l, ins214EPE, G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K , E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, and L981F are high-risk mutant strains with at least one mutation;
  • the mutant strain is BA.1.
  • the method comprises constructing a polynucleotide comprising the nucleotide sequence shown in SEQ ID No: 7 or a fragment thereof.
  • the third aspect of the present invention provides a SARS-CoV-2 mutant strain ECD immunogenic protein/peptide with improved immunogenicity/antigen trimer stability, the immunogenic protein/peptide comprising SEQ ID No: 8
  • the amino acid sequence shown, or immunogenic fragments and/or immunogenic variants thereof, the ECD immunogenic protein/peptide is in the form of a trimer in a stable pre-fusion conformation.
  • the mutant strain contains A67V, H69del, V70del, T95l, G142D, V143del, Y144del, Y145del, N211del, L212l, ins214EPE, G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K , E484A, High-risk mutant strains with at least one mutation among Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, and L981F;
  • the mutant strain is BA.1.
  • the fourth aspect of the present invention provides a polynucleotide encoding the above-mentioned immunogenic protein/peptide.
  • the polynucleotide comprises the nucleotide sequence shown in SEQ ID No: 7.
  • the fifth aspect of the present invention provides an immunogenic composition comprising:
  • an adjuvant is included.
  • the immunogenic composition further comprises amino acid sequences shown in SEQ ID No: 16, SEQ ID No: 20, SEQ ID No: 28, or immunogenic fragments thereof and/or immunogenic Variants.
  • the immunogenic composition further comprises amino acid sequences encoding SEQ ID No: 16, SEQ ID No: 20, SEQ ID No: 28, or immunogenic fragments and/or immunogens thereof Nucleotide sequence of the sex variant.
  • nucleotide sequences encoding the amino acid sequences shown in SEQ ID No: 16, SEQ ID No: 20, and SEQ ID No: 28 are SEQ ID No: 15, SEQ ID No: 19, SEQ ID No: 19, and SEQ ID No: 28, respectively. ID No: the nucleotide sequence shown in 27.
  • the adjuvant is selected from:
  • Aluminum adjuvant oil emulsion adjuvant, Toll-like receptor (TLR) agonist, combination of immune enhancer, microbial adjuvant, propolis adjuvant, levamisole adjuvant, liposome adjuvant, traditional Chinese medicine adjuvant and small
  • TLR Toll-like receptor
  • the oil-emulsion adjuvant contains squalene
  • TLR Toll-like receptor agonists comprising CpG or monophosphoryl lipid A (MPL) adsorbed on aluminum salts; and
  • Combinations of immune boosters include QS-21 and/or MPL.
  • the sixth aspect of the present invention provides the use of the above-mentioned immunogenic protein/peptide, polynucleotide and immunogenic composition for preventing or treating diseases caused by mutant SARS-CoV-2 strains.
  • the mutant strain is a high-risk mutant strain
  • mutant strains are containing L18F, T19L, T19R, L24DEL, P25DEL, P26DEL, A27S, A67V, H68DEL, H69DEL, V70DEL, D80A, T95L, G143DEL, Y145DEL, Y145DEL, E156G, F15, F15, F15, F15, F15, F15, F15, F15, F15, F15, F15, F15 7DEL, R158DEL, N211DEL, L212l, V213G, ins214EPE, D215G, L242del, A243del, L244del, R246l, G339D, R346K, S371F, S371L, S373P, S375F, T376A, D405N, R408S, K417N, N440K, G446S, L 452R, S477N, T478K
  • the strain is selected from D614G mutant strain, Beta strain, Alpha strain, Delta strain, Gamma strain, Epsilon strain, BA.1 strain, BA.1.1 strain, BA.2 At least one of strains, BA.2.12.1 strains, BA.3 strains and/or BA.4/5 strains;
  • the strain comprises Alpha strain, Beta strain, Delta strain, BA.1 strain, BA.1.1 strain, BA.2 strain, BA.2.12.1 strain, BA.3 strain and/or at least one of the BA.4/5 strains.
  • the seventh aspect of the present invention provides the use of the above-mentioned immunogenic protein/peptide, polynucleotide and immunogenic composition in the preparation of vaccines or medicines for preventing or treating diseases caused by mutant SARS-CoV-2 strains.
  • the mutant strain is a high-risk mutant strain
  • mutant strains are containing L18F, T19L, T19R, L24DEL, P25DEL, P26DEL, A27S, A67V, H68DEL, H69DEL, V70DEL, D80A, T95L, G143DEL, Y145DEL, Y145DEL, E156G, F15, F15, F15, F15, F15, F15, F15, F15, F15, F15, F15, F15 7DEL, R158DEL, N211DEL, L212l, V213G, ins214EPE, D215G, L242del, A243del, L244del, R246l, G339D, R346K, S371F, S371L, S373P, S375F, T376A, D405N, R408S, K417N, N440K, G446S, L 452R, S477N, T478K
  • the strain is selected from D614G mutant strain, Alpha strain, Beta strain, Delta strain, Gamma strain, Epsilon strain, BA.1 strain, BA.1.1 strain, BA.2 At least one of strains, BA.2.12.1 strains, BA.3 strains and/or BA.4/5 strains;
  • the strain is selected from Alpha strain, Beta strain, Delta strain, BA.1 strain, BA.1.1 strain, BA.2 strain, BA.2.12.1 strain, BA.3 strain and/or at least one of the BA.4/5 strains.
  • Figure 1 is a schematic diagram of the primary structure (A) and high-order structure (B, reference PDB: 6XLR) of the modified S-ECD.
  • Figure 2 is the analysis of the purity of TM41 protein, wherein (A) is a representative spectrum of non-reducing SDS-PAGE; (B) is a representative spectrum of SEC-HPLC.
  • Figure 3 shows the detection results of serum antibody titers (GeoMean ⁇ SD) after immunizing C57BL/6 mice with TM41 single-component and multi-component vaccines.
  • Figure 4 shows the neutralization titer detection results (GeoMean ⁇ SD) of different pseudoviruses neutralized in serum after immunization of C57BL/6 mice with TM41 single-component and multi-component vaccines for 2 days and 7 days.
  • Fig. 5 shows different doses of TM22+TM23+TM28+TM41 four-component vaccine to immunize C57BL/6 mice for 2 days and 7 days after serum neutralizes the titer detection (GeoMean ⁇ SD) of different mutant strains of pseudovirus.
  • Fig. 6 shows TM22+TM23 two-component vaccine and TM22+TM23+TM28+TM41 four-component vaccine immunization C57BL/6 mouse 2 immunizations after 7 days serum neutralizes different variant strain pseudovirus titer detection results (GeoMean ⁇ SD ).
  • Fig. 7 shows C57BL/6 mouse 2 immunization 7 days and 3 immunization 7 days serum and Omicron (BA.1) pseudovirus titer detection result (GeoMean ⁇ SD); 2M7D represents 2 immunizations 7 days; 3 7 days after exemption.
  • Figure 8 shows the titer detection results of TM22+TM23+TM28+TM41 four-component vaccine and single-component/two-component vaccine immunization of C57BL/6 mice for 2 days and 14 days after serum neutralization of different mutant pseudoviruses.
  • Figure 9 shows the detection results of cellular immune responses induced by different vaccine antigens, wherein (A) is the result of the number of IFN-positive cells; (B) is the result of the number of IL-4 positive cells; (C) is the result of the number of CD137+CD134+ double positives The results of the proportion of CD4 T lymphocytes; (D) is the result of the proportion of CD137+CD69+ double positive CD8 T lymphocytes.
  • Figure 10 shows the comparison of the neutralization titers of the serum to each subtype of Omicron (BA.1, BA.2, BA.3, BA.4/5) pseudoviruses after booster immunization.
  • BA.1, BA.2, BA.3, BA.4/5 pseudoviruses after booster immunization.
  • a indicates the fold change compared with TM8 two-shot immunization (2 immunizations for 7 days);
  • b indicates the fold change compared with TM8 booster immunization].
  • Figure 11 shows the comparison of neutralizing titers of sera to Alpha, Beta, and Delta pseudoviruses after 1 shot of booster immunization respectively. [a indicates the fold change compared with TM8 two-shot immunization (2 immunizations for 7 days); b indicates the fold change compared with TM8 booster immunization].
  • Figure 12 (A-D) respectively shows the comparison of the neutralization titer of serum to each subtype of Omicron (BA.1, BA.2, BA.3, BA.4/5) pseudoviruses after 2 injections of booster immunization.
  • BA.1, BA.2, BA.3, BA.4/5 pseudoviruses after 2 injections of booster immunization.
  • a indicates the fold change compared with TM8 two-shot immunization (2 immunizations for 7 days);
  • b indicates the fold change compared with TM8 booster immunization].
  • 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”.
  • a "hapten” refers to 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 "cell-mediated immune response” is an immune response mediated by T cells 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”.
  • immunogenic fragments and “immunogenic variants” of the present invention are 99%, 98%, 97%, 95%, 90% identical to the corresponding (the former derived from the latter) "immunogenic protein/peptide" sequence identity.
  • the SCTV01E 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 natural spike protein of SARS-CoV-2 is a trimeric structure. During its production and infection function, it is easily cleaved by proteases in the Golgi apparatus and on the cell surface due to the RRAR site between S1 and S2 After opening, S1 falls off, and the S2 structure changes from prefusion conformation to postfusion conformation, thus completing the membrane fusion process[20].
  • the present invention carried out the following three-part transformation on the basis of the S protein of different strain variants (Table 1 and Fig. 1):
  • the Furin site is modified and removed in the SCTV01E recombinant protein vaccine, that is, the amino acid sequence from 679 to 688 is fixed as NSPGSASSVA, so as to reduce the possibility of S1 breaking and falling off.
  • the present invention introduces the SCTV01E recombinant protein vaccine HexaPro mutations that can effectively improve 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 to proline) [26] .
  • 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. T4 foldon has been used in RSV vaccine candidates, and proved to be safe in Phase I clinical studies [27] .
  • the corresponding trimer was prepared Protein, that is, in the previous invention (recorded in PCT/CN2022/095609 and PCT/CN2022/107213) D614G mutant strain S-Trimer-TM8 protein (hereinafter referred to as TM8), Alpha mutant strain S-Trimer-TM22 protein (hereinafter referred to as TM22) , Beta mutant strain S-Trimer-TM23 protein (hereinafter referred to as TM23), Delta mutant strain S-Trimer-TM28 protein (hereinafter referred to as TM28) and BA.1 mutant strain S-Trimer-TM41 protein of the present invention (hereinafter referred to as TM41, Table 1 outlines its molecular modification scheme).
  • TM8 D614G mutant strain S-Trimer-TM8 protein
  • TM22 Alpha mutant strain S-Trimer-TM22 protein
  • Beta mutant strain S-Trimer-TM23 protein hereinafter referred to as TM23
  • Delta mutant strain S-Trimer-TM28 protein hereinafter
  • the prepared D614G variant strain TM8 protein vaccine was used to immunize mice for immunological determination, the immunological determination of Beta variant strain TM23 protein vaccine in cynomolgus monkeys and the Alpha variant strain TM22 protein vaccine in mice Immunological assays all show that these three vaccines prepared by the present invention can produce antibody immune responses of sufficient titer in experimental animals; It is also suggested in the immunological evaluation that the two-component vaccine of the present invention has higher and similar neutralizing titers to different strains, so it has better broad-spectrum neutralization ability than the single-component vaccine. The neutralizing titer for different mutant strains is much higher than that of the convalescent serum for the early epidemic strain (its genome sequence: GenBank Accession No.NC_045512).
  • the four-component vaccine (TM22+TM23+TM28+TM41) of the present invention has more broad-spectrum neutralizing activity to mutant strains such as Alpha, Beta, Delta and Omicron, but keeps the same single-component vaccine and two-component vaccine.
  • a similar high level of T cell immune response is expected to produce cross-protection against a variety of mutant strains (Table 2 records the S protein mutations of related SARS-CoV-2 mutant strains) and improve the protection rate against mutant strain infection .
  • 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.
  • TM41 contains a 3699bp gene fragment, and the TM41 gene fragment was obtained by overlapping PCR from the template pCMV3-CoV2-B.1.1.529 and pD2535nt-CoV2-S-ECDTM8-T4F-trimer. Constructed into the pD2535nt-HDP stable strain expression vector digested with Xba I+Asc I by the In-fusion method to obtain the pD2535nt-CoV2-S-ECDTM41-T4F-trimer expression vector.
  • 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) Samples were boiled at 100°C for 2 minutes, centrifuged and loaded with 8 samples-; (3) Coomassie brilliant blue staining After bleaching.
  • 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 NaH2P04, 100mM Arginine, pH 6.5, 0.01% isopropanol (IPA); (3) The sample volume is 80 ⁇ g; (3) The detection wavelength is 280nM, the analysis time is 35min, and the flow rate is 0.15mL/min.
  • DLS Dynamic Light Scattering Instrument (Wyatt Technology Company, model: DynaPro NanoStar); (2) The sample volume is 50 ⁇ L; (3) After collecting the data, use Dynamics 7.1.8 software to analyze the data.
  • the recombinant TM41 protein has a homotrimeric structure due to its non-covalent hydrophobic interaction. After non-reducing SDS-PAGE treatment, it becomes a monomer molecule with a molecular weight of about 148KDa ( Figure 2), and the purity is 99.0%. %, the average molecular weight of its main peak is 512KDa; the dynamic light scattering results show that the average molecular radius of TM41 trimeric protein is 8.8nm (Table 3).
  • Recombinant TM41 trimer protein was stored at 37°C for 2 weeks (37T2W), stored at -80°C for 8 hours, and then transferred to 25°C for 0.5h (F/T), and repeated freezing and thawing was carried out 4 times.
  • Embodiment 3 TM41 single-component vaccine and multi-component vaccine in Immunological evaluation of mice
  • the purified TM22, TM23, TM28 and TM41 trimeric proteins were pre-diluted with PBS and mixed with MF59 (8 ⁇ , source: Shenzhou Cell Engineering Co., Ltd., the same below) in equal volumes to prepare Single or multi-component vaccine samples.
  • mice immunized with one-component vaccine was coated with 5 ⁇ g/mL of TM41 protein
  • the immune serum of mice immunized with two-component vaccine was coated with 5 ⁇ g/mL of TM22 and TM23 proteins (1:1)
  • the immune serum of mice immunized with two-component vaccine was coated with 5 ⁇ g/mL of TM22 and TM23 proteins (1:1)
  • TM22, TM23, TM28 and TM41 proteins (1:1:1:1 were coated with 5 ⁇ g/mL of mouse immune serum, 100 ⁇ L/well was coated on a 96-well plate, and coated overnight at 2-8°C.
  • the plate was washed 3 times, 100 ⁇ L/well of 80 ng/mL rabbit anti-mouse IgG F(ab)2/HRP detection secondary antibody (source: Jackson ImmunoResearch, the same below) was added, and incubated at room temperature for 1 h. Wash the plate 5 times, add the substrate chromogenic solution to develop the color for 10-15 minutes, read the OD 450 with a microplate reader after the termination of 2M H 2 SO 4 , and calculate the immune antibody titer.
  • Antibody titer negative serum OD 450 ⁇ maximum dilution factor of 2.1.
  • the total IgG antibody titer induced by the TM22+TM23 two-component vaccine antigen was 960000, while the TM22+TM23+TM28+TM41 four-component vaccine antigen immunization group (0.5+0.5+0.5+1.5 ⁇ g) induced the highest total IgG antibody titer.
  • the total IgG antibody titers induced by different doses of TM41 single-component vaccine antigen (0.25 ⁇ g/dose, 0.5 ⁇ g/dose and 1 ⁇ g/dose) showed a dose-effect relationship, and the antibody titers were 256,000, 512,000, and 576,000, respectively.
  • the total IgG antibody titers induced by different doses of TM22+TM23+TM28+TM41 four-component vaccine antigen immunization group were higher than that of TM41 single-component vaccine.
  • pseudoviruses are replication-deficient vesicular stomatitis viruses that replace the VSV-G protein gene in the viral genome with the luciferase reporter gene (i.e.
  • VSV ⁇ G-Luc-G as a carrier, amplified and prepared in a cell line expressing Spike and its mutant proteins, prepared by Shenzhou Cell Engineering Co., Ltd., the same below), mixed and placed at 37 ° C, 5% Incubate for 1 h in a CO 2 incubator. Serum-free cell wells containing pseudovirus were used as positive controls, and cell wells without serum and pseudoviruses were used as negative controls. After the incubation, 100 ⁇ L/well was inoculated with 2 ⁇ 10 4 Huh-7 cells, mixed evenly, and placed in a 37° C., 5% CO 2 incubator for static culture for about 20 h.
  • TM41 single-component and four-component vaccines TM22+TM23+TM28+TM41 immunized C57BL/6 mice for 2 days and 7 days after serum neutralization of different mutant strains Alpha(B.1.1.7), Beta(B.1.351), Delta (B.1.617.2) and BA.1 (B.1.1.529.1) pseudovirus neutralization potency.
  • the TM41 single-component vaccine at different doses can induce the specific neutralizing antibodies against the BA.1 variant strain, and 1 ⁇ g of the single-component vaccine TM41 can induce the highest anti-BA.1 variant strain in C57BL/6J mice Neutralizing activity, the neutralizing antibody titer was 2730, which reached the saturation dose in this experiment.
  • the TM41 single-component vaccine has no neutralizing activity against the Alpha, Beta and Delta mutant strains, and the neutralizing antibody titer detection values are all 60, which is lower than the detection limit.
  • the TM22+TM23+TM28+TM41 four-component vaccines in different dosage groups had strong neutralizing activity against BA. 1
  • the detection value of the neutralizing antibody titer of the variant strain was 618-2730.
  • the neutralizing activity of the TM22+TM23+TM28+TM41 four-component vaccine against the BA.1 mutant strain is comparable to that of the TM41 single-component vaccine, and it also has higher neutralizing activity against the Alpha, Beta, Delta and other mutant strains, indicating that Compared with the TM41 single-component vaccine, the TM22+TM23+TM28+TM41 four-component vaccine has a broader spectrum of neutralizing activity against different variants of SARS-CoV-2 ( Figure 4).
  • TM22+TM23+TM28+TM41 four-component vaccine to immunize C57BL/6 mice for 2 days and 7 days to neutralize different mutant strains Alpha(B.1.1.7), Beta(B.1.351), Delta(B. 1.617.2) and BA.1 (B.1.1.529.1) pseudovirus neutralization titers.
  • Different doses of TM22+TM23+TM28+TM41 four-component vaccines can induce higher levels of neutralizing antibodies against Alpha, Beta and Delta variants. Compared with the above three neutralizing antibodies, the four-component vaccines induced BA .1 The overall level of neutralizing antibodies is low. As the dose of TM41 increased, the neutralizing activity against BA.1 mutant strains tended to increase, showing a dose-effect relationship (Fig. 5).
  • TM22+TM23 two-component vaccine and different doses of TM22+TM23+TM28+TM41 four-component vaccine immunized C57BL/6 mice for 2 days and 7 days after serum neutralization of different mutant strains Alpha(B.1.1.7), Beta( B.1.351), Delta (B.1.617.2) and BA.1 (B.1.1.529.1) pseudovirus titers.
  • the specific neutralizing antibody titers of the TM22+TM23 two-component vaccine group against the Alpha, Beta, Delta and BA.1 mutant strains were 4598, 4972, 1384 and 247, respectively. Higher specific neutralizing antibody titers against Alpha, Beta, and Delta were generated, but the protective effect on BA.1 mutant strains was relatively weak.
  • the neutralizing antibody titers produced by the TM22+TM23+TM28+TM41 four-component vaccine at different doses against the Alpha and Beta mutant strains were comparable to those of the two-component vaccines, while the neutralizing antibody titers against the Delta and BA.1 mutant strains than two-component vaccines.
  • the low-dose group of the four-component vaccine induced 5.4 times and 1.9 times the neutralizing antibodies against the Delta and BA.1 variants than the two-component vaccine
  • the medium dose of the four-component vaccine induced neutralizing antibodies against the Delta and BA.1 variants.
  • TM22+TM23+TM28+TM41 four-component vaccine and single-component/two-component vaccine immunization of C57BL/6 mice for 14 days after 2 immunizations to neutralize different mutant strains (D614G strain, Alpha strain, Beta strain, Delta strain, BA.1 strain, BA.1.1 strain, BA.2 strain, BA.2.12.1 strain, BA.3 strain and BA.4/5 strain) pseudovirus titer. Comparing the neutralizing activity of the four-component vaccine TM22+TM23+TM28+TM41 with the TM8, TM41 single-component vaccine and the two-component vaccine TM22+TM23 against different new coronavirus mutant strains pseudoviruses, the test results are shown in Figure 8A.
  • the four-component vaccine TM22+TM23+TM28+TM41 can significantly improve the response to Beta, Delta, Omicron BA.1, BA.1.1, BA.2, BA.3 and BA.4/5 variation Strain neutralizing activity.
  • the four-component vaccine TM22+TM23+TM28+TM41 can significantly improve the neutralizing activity against D614G, Alpha, Beta and Delta mutant strains.
  • the four-component vaccine TM22+TM23+TM28+TM41 can significantly improve the variation of Delta, Omicron BA.1, BA.1.1, BA.2, and BA.3 Strain neutralizing activity.
  • the four-component vaccine TM22+TM23+TM28+TM41 can significantly improve the effect on Omicron BA.2.12.1 and BA.4/ 5 Neutralizing activity of mutant strains.
  • ELISpot method for detection of T cell immunity isolate mouse splenocytes, inoculate 100 ⁇ L/well of mouse splenocytes on pre-treated ELISpot well plates (source: Mabtech, the same below), and the cell inoculation density is 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 GraphPad Prism software was used for data statistics.
  • Detect activated T cell subsets by flow cytometry Grind the spleen into a single cell suspension, use wild-type (original strain: genome sequence: GenBank Accession No.NC_045512) polypeptide library and Omicron (BA.1) polypeptide library Stimulate splenocytes immunized with different vaccine antigens for 20 hours in a 37°C 5% CO 2 incubator. After the stimulation, wash the cells with PBS, centrifuge at 1000rpm for 5min and discard the supernatant.
  • BV510 anti- mouse CD3e, CD4 Antibody (FITC), Rabbit Mab, CD8a Antibody (APC), Rabbit Mab, BV650 Hamster Anti-Mouse CD69, PE Rat Anti-Mouse CD137, Brilliant Violet 421TM anti-mouse CD134 (OX-40) corresponding antibody Spleen cells were stained at 4°C in the dark for 20 minutes, and detected by flow cytometry after staining.
  • the number of IL-4 positive cells induced by TM41 single-component, TM22+TM23 two-component and TM22+TM23+TM28+TM41 four-component vaccine antigens was comparable, and the vaccine-induced Th2 cells The immune response was not significantly different between the groups (Fig. 9B).
  • the spleen cells of the mice were stimulated with wild-type and BA.1 antigen peptides respectively, and the activated CD4 + and CD8 + T cells were induced higher than the levels of the blank and adjuvant control groups, and There were no significant differences between the three live vaccines.
  • the wild-type and BA.1 antigen peptides have similar T cell activation stimulation levels, indicating that there are conserved T cell epitopes among different strains (Fig. 9C-D).
  • Example 3.1 To prepare TM8 single-component vaccine, TM22+TM23 two-component vaccine and TM22+TM23+TM28+TM41 four-component vaccine.
  • mice About 6 weeks old, C57BL/6J female mice (source: Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.) were immunized with 100 ⁇ L TM8 single-component vaccine antigen containing MF59 adjuvant by intramuscular injection on day 0 and day 14 respectively ( 1 ⁇ g/dose), and the serum immunoassay was carried out in the orbit on the 2nd and 7th day.
  • Example 3.4 detect the neutralizing titer of mouse immune serum to Alpha, Beta, Delta, each subtype of Omicron (BA.1, BA.2, BA.3, BA.4/5) pseudovirus.
  • the values were 1293, 1178, 803 and 722, respectively, which were 15.0 times, 9.1 times, 10.7 times and 10.6 times of the neutralization titer of the TM8 single-component vaccine 7 days after the second immunization, and 2.9 times of the TM8 single-component vaccine booster immunization group.
  • TM22+TM23 two-component vaccine booster group (Fig. 10A-D); the neutralization titer to the Delta strain was 22298 , which is 5.2 times the neutralization titer of TM8 single-component vaccine 7 days after the second immunization, 1.6 times that of the single-component vaccine booster immunization group, and 1.6 times that of the two-component vaccine booster immunization group; in addition, TM22+TM23+TM28 +TM41 four-component vaccine booster immunization can induce high levels of neutralizing activities of the Alpha and Beta variants, which are 21257 and 12898, respectively (Fig.
  • the four-component vaccine has a broader neutralizing activity against SARS-CoV-2 mutants Alpha, Beta, Delta and Omicron (BA.1, BA.2, BA.3, BA.4/5), and is superior to Booster immunization with TM8 single-component vaccine or TM22+TM23 two-component vaccine.
  • the geometric mean neutralization titers of Omicron BA.1, BA.2, BA.3 and BA.4/5 induced by two injections of TM22+TM23+TM28+TM41 four-component vaccine after booster immunization (4 days and 7 days) were respectively 14872, 6897, 8768 and 1136, which were 172.9 times, 53.5 times, 116.9 and 16.7 times the neutralization titers of the TM8 single-component vaccine 7 days after the second immunization, and 22.6 times and 17.4 times the TM8 single-component vaccine booster immunization group.
  • the four-component vaccine has a broad-spectrum neutralization ability against different mutant strains, but maintains a similar high
  • the level of T cell immune response is expected to produce cross-protection ability against multiple mutant strains and improve the protection rate against mutant strain infection.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Communicable Diseases (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention se rapporte au domaine de la vaccinologie moléculaire. La présente invention concerne 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. Les ingrédients protéiques recombinants incluent, entre autres, des protéines homotrimériques formées au moyen de l'introduction de sites de mutation et de structures auxiliaires trimériques dans les domaines extracellulaires (ECD) de protéines de spicule (S protéines) de Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2) et BA.1 (B.1.1.529.1). Le vaccin à composants multiples contient une protéine trimérique ECD des variants ci-dessus, seuls ou en combinaison, et un adjuvant pharmaceutiquement acceptable. La combinaison de vaccins présente une excellente immunogénicité chez les souris, tout en maintenant également une immunité humorale à long terme et des réponses immunitaires cellulaires à long terme. Le vaccin à composants multiples à base de protéine trimérique de SARS-CoV-2 peut être utilisé pour prévenir des maladies liées à une infection provoquées par des infections par le SARS-CoV-2 et des variants de celui-ci.
PCT/CN2023/078135 2022-02-25 2023-02-24 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 WO2023160654A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202380013961.3A CN118076646A (zh) 2022-02-25 2023-02-24 一种可诱导广谱中和活性重组多组分新冠病毒三聚体蛋白疫苗的制备及应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210184528.3 2022-02-25
CN202210184528 2022-02-25

Publications (1)

Publication Number Publication Date
WO2023160654A1 true WO2023160654A1 (fr) 2023-08-31

Family

ID=87764840

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/078135 WO2023160654A1 (fr) 2022-02-25 2023-02-24 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

Country Status (2)

Country Link
CN (1) CN118076646A (fr)
WO (1) WO2023160654A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117070535A (zh) * 2023-10-17 2023-11-17 中国医学科学院医学生物学研究所 具有体液免疫和细胞免疫功能的新冠病毒突变体广谱疫苗

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112552380A (zh) * 2020-12-10 2021-03-26 武汉博沃生物科技有限公司 一种SARS-CoV-2病毒的免疫原及其应用
CN113388041A (zh) * 2020-03-12 2021-09-14 厦门大学 具有融合前早期构象的SARS-CoV-2 S三聚体蛋白及其应用
WO2021204825A2 (fr) * 2020-04-06 2021-10-14 Valneva Austria Gmbh Vaccin à virus sars-cov-2 inactivé
US11225508B1 (en) * 2020-09-23 2022-01-18 The University Of North Carolina At Chapel Hill Mouse-adapted SARS-CoV-2 viruses and methods of use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113388041A (zh) * 2020-03-12 2021-09-14 厦门大学 具有融合前早期构象的SARS-CoV-2 S三聚体蛋白及其应用
WO2021204825A2 (fr) * 2020-04-06 2021-10-14 Valneva Austria Gmbh Vaccin à virus sars-cov-2 inactivé
US11225508B1 (en) * 2020-09-23 2022-01-18 The University Of North Carolina At Chapel Hill Mouse-adapted SARS-CoV-2 viruses and methods of use thereof
CN112552380A (zh) * 2020-12-10 2021-03-26 武汉博沃生物科技有限公司 一种SARS-CoV-2病毒的免疫原及其应用

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MALIK, J.A.: "Targets and Strategies for Vaccine Development against SARS-CoV-2", BIOMEDICINE & PHARMACOTHERAPY, vol. 137, 31 May 2021 (2021-05-31) *
PINGPING JIANG, WANG TING: "SARS-CoV-2 immunity and vaccine progress ", CHINESE JOURNAL OF PHARMACEUTICS(ON LINE), vol. 20, no. 1, 15 January 2022 (2022-01-15), pages 40 - 46, XP093088303, DOI: 10.14146/j.cnki.cjp.2022.01.004 *
XUN WANG, WANG PENGFEI: "Advances in Vaccine Research against Novel Coronavirus Mutant Strains", ZHANGJIANG TECHNOLOGY REVIEW, vol. 6, 28 December 2021 (2021-12-28), pages 35 - 37, XP093088284 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117070535A (zh) * 2023-10-17 2023-11-17 中国医学科学院医学生物学研究所 具有体液免疫和细胞免疫功能的新冠病毒突变体广谱疫苗
CN117070535B (zh) * 2023-10-17 2024-01-26 中国医学科学院医学生物学研究所 具有体液免疫和细胞免疫功能的新冠病毒突变体广谱疫苗

Also Published As

Publication number Publication date
CN118076646A (zh) 2024-05-24

Similar Documents

Publication Publication Date Title
CN110951756B (zh) 表达SARS-CoV-2病毒抗原肽的核酸序列及其应用
EP3054971B1 (fr) Vaccins contre le virus d'epstein-barr
US9731006B2 (en) Attenuated recombinant vesicular stomatitis virus vaccine vectors comprising modified matrix proteins
JP2023524054A (ja) ベータコロナウイルスの予防と治療
US20220054625A1 (en) Immunogenic composition
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
TW202328210A (zh) 用於預防感染與治療遠程新冠肺炎之針對SARS-CoV-2變體的疫苗組合物
Sun et al. Vaccine: X
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
US20190231866A1 (en) Methods for safe induction of cross-clade immunity against hiv infection in humans
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
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
CN118119646A (zh) 一种可诱导广谱中和活性重组五组分新冠病毒三聚体蛋白疫苗的制备及应用
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
US12005115B2 (en) Epstein-barr virus vaccines
US20240092840A1 (en) Vaccine formulation comprising recombinant overlapping peptides and native proteins
Lee et al. T-cell immunity induced by a nonadjuvanted HLA-restricted peptide COVID-19 vaccine
Lee et al. T cell immunity of the nonadjuvanted HLA-restricted peptide COVID-19 vaccine
WO2023187366A1 (fr) Compositions immunogènes pour la prévention de la grippe a
Lucy Kuo et al. A Novel SARS-CoV-2 Multitope Protein/Peptide Vaccine Candidate is Highly
OA17659A (en) Modified matrix proteins of vesicular stomatitis virus.

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: 23759276

Country of ref document: EP

Kind code of ref document: A1