WO2022052984A1 - Universal sars-cov-2 vaccine and preparation method thereof - Google Patents
Universal sars-cov-2 vaccine and preparation method thereof Download PDFInfo
- Publication number
- WO2022052984A1 WO2022052984A1 PCT/CN2021/117415 CN2021117415W WO2022052984A1 WO 2022052984 A1 WO2022052984 A1 WO 2022052984A1 CN 2021117415 W CN2021117415 W CN 2021117415W WO 2022052984 A1 WO2022052984 A1 WO 2022052984A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- peptide fragment
- fusion protein
- seq
- vaccine
- Prior art date
Links
- 229940022962 COVID-19 vaccine Drugs 0.000 title claims abstract description 24
- 238000002360 preparation method Methods 0.000 title abstract description 19
- 108020001507 fusion proteins Proteins 0.000 claims abstract description 63
- 102000037865 fusion proteins Human genes 0.000 claims abstract description 63
- 101710139375 Corneodesmosin Proteins 0.000 claims abstract description 43
- 102100031673 Corneodesmosin Human genes 0.000 claims abstract description 43
- 229960005486 vaccine Drugs 0.000 claims abstract description 33
- 210000000612 antigen-presenting cell Anatomy 0.000 claims abstract description 32
- 102000004127 Cytokines Human genes 0.000 claims abstract description 26
- 108090000695 Cytokines Proteins 0.000 claims abstract description 26
- 101710085938 Matrix protein Proteins 0.000 claims abstract description 22
- 101710127721 Membrane protein Proteins 0.000 claims abstract description 22
- 101710204837 Envelope small membrane protein Proteins 0.000 claims abstract description 21
- 101710145006 Lysis protein Proteins 0.000 claims abstract description 21
- 101710141454 Nucleoprotein Proteins 0.000 claims abstract description 20
- 239000004365 Protease Substances 0.000 claims abstract description 20
- 101150001779 ORF1a gene Proteins 0.000 claims abstract description 19
- 108010033276 Peptide Fragments Proteins 0.000 claims description 49
- 102000007079 Peptide Fragments Human genes 0.000 claims description 49
- 108090000623 proteins and genes Proteins 0.000 claims description 43
- 241001678559 COVID-19 virus Species 0.000 claims description 22
- 150000007523 nucleic acids Chemical class 0.000 claims description 22
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 20
- 102000004169 proteins and genes Human genes 0.000 claims description 20
- 108020003175 receptors Proteins 0.000 claims description 19
- 102000005962 receptors Human genes 0.000 claims description 19
- 230000036961 partial effect Effects 0.000 claims description 18
- 108010076504 Protein Sorting Signals Proteins 0.000 claims description 16
- 239000013598 vector Substances 0.000 claims description 15
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 13
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 13
- 108020004707 nucleic acids Proteins 0.000 claims description 13
- 102000039446 nucleic acids Human genes 0.000 claims description 13
- 108010002350 Interleukin-2 Proteins 0.000 claims description 12
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 claims description 12
- 102000034342 Calnexin Human genes 0.000 claims description 11
- 108010056891 Calnexin Proteins 0.000 claims description 11
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 claims description 11
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 claims description 11
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 11
- 241000713666 Lentivirus Species 0.000 claims description 10
- 238000000034 method Methods 0.000 claims description 10
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 8
- 239000013604 expression vector Substances 0.000 claims description 7
- 229960004857 mitomycin Drugs 0.000 claims description 6
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 5
- 230000005251 gamma ray Effects 0.000 claims description 5
- 230000002779 inactivation Effects 0.000 claims description 5
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims description 3
- 210000004962 mammalian cell Anatomy 0.000 claims description 2
- 238000004806 packaging method and process Methods 0.000 claims description 2
- 239000013612 plasmid Substances 0.000 claims description 2
- 230000001177 retroviral effect Effects 0.000 claims description 2
- 230000002463 transducing effect Effects 0.000 claims description 2
- 101800000504 3C-like protease Proteins 0.000 claims 1
- 241000702421 Dependoparvovirus Species 0.000 claims 1
- 108090000765 processed proteins & peptides Proteins 0.000 abstract description 13
- 101710172711 Structural protein Proteins 0.000 abstract description 12
- 102000004196 processed proteins & peptides Human genes 0.000 abstract description 11
- 230000006054 immunological memory Effects 0.000 abstract description 8
- 230000028993 immune response Effects 0.000 abstract description 7
- 239000012634 fragment Substances 0.000 abstract description 6
- 210000000987 immune system Anatomy 0.000 abstract description 4
- 230000003308 immunostimulating effect Effects 0.000 abstract description 4
- 238000009776 industrial production Methods 0.000 abstract description 4
- 229920001184 polypeptide Polymers 0.000 abstract description 4
- 108091007576 SARS-CoV-2 structural proteins Proteins 0.000 abstract 1
- 210000004027 cell Anatomy 0.000 description 22
- 210000001744 T-lymphocyte Anatomy 0.000 description 21
- 208000025721 COVID-19 Diseases 0.000 description 14
- 239000000427 antigen Substances 0.000 description 11
- 108091007433 antigens Proteins 0.000 description 11
- 102000036639 antigens Human genes 0.000 description 11
- 241000700605 Viruses Species 0.000 description 10
- 230000000638 stimulation Effects 0.000 description 9
- 229940096437 Protein S Drugs 0.000 description 7
- 238000004458 analytical method Methods 0.000 description 6
- 230000004044 response Effects 0.000 description 6
- 102100020715 Fms-related tyrosine kinase 3 ligand protein Human genes 0.000 description 5
- 101710162577 Fms-related tyrosine kinase 3 ligand protein Proteins 0.000 description 5
- 229940030156 cell vaccine Drugs 0.000 description 5
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 5
- 238000000684 flow cytometry Methods 0.000 description 5
- 239000000499 gel Substances 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 102100027207 CD27 antigen Human genes 0.000 description 4
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 4
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 4
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 4
- 239000012636 effector Substances 0.000 description 4
- 208000015181 infectious disease Diseases 0.000 description 4
- 241000315672 SARS coronavirus Species 0.000 description 3
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 description 3
- 101710198474 Spike protein Proteins 0.000 description 3
- 108010067390 Viral Proteins Proteins 0.000 description 3
- 230000000890 antigenic effect Effects 0.000 description 3
- 210000004443 dendritic cell Anatomy 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- -1 i.e. Proteins 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 239000000203 mixture Substances 0.000 description 3
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 3
- 230000002265 prevention Effects 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- 102100035765 Angiotensin-converting enzyme 2 Human genes 0.000 description 2
- 108090000975 Angiotensin-converting enzyme 2 Proteins 0.000 description 2
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 2
- 101710088172 HTH-type transcriptional regulator RipA Proteins 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 108010052285 Membrane Proteins Proteins 0.000 description 2
- 102000018697 Membrane Proteins Human genes 0.000 description 2
- 241000127282 Middle East respiratory syndrome-related coronavirus Species 0.000 description 2
- 108090001074 Nucleocapsid Proteins Proteins 0.000 description 2
- 102100034574 P protein Human genes 0.000 description 2
- 101710181008 P protein Proteins 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- 101710177166 Phosphoprotein Proteins 0.000 description 2
- 108091005774 SARS-CoV-2 proteins Proteins 0.000 description 2
- 239000011543 agarose gel Substances 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 230000009089 cytolysis Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 210000003162 effector t lymphocyte Anatomy 0.000 description 2
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 238000011990 functional testing Methods 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 210000003519 mature b lymphocyte Anatomy 0.000 description 2
- 210000003071 memory t lymphocyte Anatomy 0.000 description 2
- 230000003278 mimic effect Effects 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 230000002085 persistent effect Effects 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 208000019585 progressive encephalomyelitis with rigidity and myoclonus Diseases 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 1
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 1
- 241000711573 Coronaviridae Species 0.000 description 1
- 206010050685 Cytokine storm Diseases 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 108010041986 DNA Vaccines Proteins 0.000 description 1
- 229940021995 DNA vaccine Drugs 0.000 description 1
- 101710091045 Envelope protein Proteins 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 102000000588 Interleukin-2 Human genes 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- 102000012750 Membrane Glycoproteins Human genes 0.000 description 1
- 108010090054 Membrane Glycoproteins Proteins 0.000 description 1
- 229930191564 Monensin Natural products 0.000 description 1
- GAOZTHIDHYLHMS-UHFFFAOYSA-N Monensin A Natural products O1C(CC)(C2C(CC(O2)C2C(CC(C)C(O)(CO)O2)C)C)CCC1C(O1)(C)CCC21CC(O)C(C)C(C(C)C(OC)C(C)C(O)=O)O2 GAOZTHIDHYLHMS-UHFFFAOYSA-N 0.000 description 1
- 208000034486 Multi-organ failure Diseases 0.000 description 1
- 208000010718 Multiple Organ Failure Diseases 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 108010089430 Phosphoproteins Proteins 0.000 description 1
- 102000007982 Phosphoproteins Human genes 0.000 description 1
- 108010076039 Polyproteins Proteins 0.000 description 1
- 101710188315 Protein X Proteins 0.000 description 1
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 1
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 1
- 230000010398 acute inflammatory response Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 206010052015 cytokine release syndrome Diseases 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- XEYBRNLFEZDVAW-ARSRFYASSA-N dinoprostone Chemical compound CCCCC[C@H](O)\C=C\[C@H]1[C@H](O)CC(=O)[C@@H]1C\C=C/CCCC(O)=O XEYBRNLFEZDVAW-ARSRFYASSA-N 0.000 description 1
- 229960002986 dinoprostone Drugs 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 230000000694 effects Effects 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 239000012642 immune effector Substances 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 229940121354 immunomodulator Drugs 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 210000005007 innate immune system Anatomy 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 108700021021 mRNA Vaccine Proteins 0.000 description 1
- 229940126582 mRNA vaccine Drugs 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 230000015654 memory Effects 0.000 description 1
- 229960005358 monensin Drugs 0.000 description 1
- GAOZTHIDHYLHMS-KEOBGNEYSA-N monensin A Chemical compound C([C@@](O1)(C)[C@H]2CC[C@@](O2)(CC)[C@H]2[C@H](C[C@@H](O2)[C@@H]2[C@H](C[C@@H](C)[C@](O)(CO)O2)C)C)C[C@@]21C[C@H](O)[C@@H](C)[C@@H]([C@@H](C)[C@@H](OC)[C@H](C)C(O)=O)O2 GAOZTHIDHYLHMS-KEOBGNEYSA-N 0.000 description 1
- 208000029744 multiple organ dysfunction syndrome Diseases 0.000 description 1
- 208000025113 myeloid leukemia Diseases 0.000 description 1
- 210000005259 peripheral blood Anatomy 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- XEYBRNLFEZDVAW-UHFFFAOYSA-N prostaglandin E2 Natural products CCCCCC(O)C=CC1C(O)CC(=O)C1CC=CCCCC(O)=O XEYBRNLFEZDVAW-UHFFFAOYSA-N 0.000 description 1
- 239000012460 protein solution Substances 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 239000002994 raw material Substances 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 239000012488 sample solution Substances 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4615—Dendritic cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4622—Antigen presenting cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/463—Cellular immunotherapy characterised by recombinant expression
- A61K39/4634—Antigenic peptides; polypeptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/464838—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/525—Virus
- A61K2039/5252—Virus inactivated (killed)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55522—Cytokines; Lymphokines; Interferons
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/02—Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/40—Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/15011—Lentivirus, not HIV, e.g. FIV, SIV
- C12N2740/15021—Viruses as such, e.g. new isolates, mutants or their genomic sequences
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/15011—Lentivirus, not HIV, e.g. FIV, SIV
- C12N2740/15041—Use of virus, viral particle or viral elements as a vector
- C12N2740/15043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16041—Use of virus, viral particle or viral elements as a vector
- C12N2740/16043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/20011—Coronaviridae
- C12N2770/20022—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/20011—Coronaviridae
- C12N2770/20034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- the present disclosure belongs to the field of biological medicine, and relates to a universal SARS-CoV-2 vaccine and a preparation method thereof.
- Spike protein S protein
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the S protein plays a same role in SARS and MERS viruses. Therefore, the S protein determines the host range and specificity of the virus, and it is an important target for antibody binding in the host, and is also a key target for the development of vaccines, therapeutic antibodies, and diagnostic reagents.
- the virus causing COVID-19 is a newly emerged coronavirus variant.
- the spike protein of this virus binds to ACE2 receptor with a stronger force than SARS-CoV, which explains the strong infection ability of SARS-CoV-2.
- SARS-CoV-2 stimulates the innate immune system in an infected individual, causing the release of a large amount of cytokines in the body and causing a cytokine storm and an acute inflammatory response, thereby leading to acute respiratory distress syndrome (ARDS) and multiple organ failure.
- ARDS acute respiratory distress syndrome
- a great challenge in vaccine development is to find a suitable animal model.
- non-human primate infection models are mainly used to verify the safety and efficacy of the developed vaccines, but the cost is extremely high and it takes a long time.
- the whole virus antigen vaccine is based on the traditional inactivated virus or attenuated virus as antigen source, which has the problems of low safety and high cost;
- the viral specific antigen vaccine is based on synthetic S protein as the antigen, while artificially synthesized S protein is mainly expressed by bacteria, yeast, cells or viral vectors, which has the problems of a length process of manufacture, single valence, easy escape of the virus, short validity period, and high cost;
- DNA and mRNA vaccines need to enter cells and be converted into proteins to exert immune effects, and the low immunogenicity issue has been the bottleneck that the technology still cannot overcome at present time.
- the present disclosure provides a universal SARS-CoV-2 vaccine composition and a preparation method thereof.
- the vaccine uses immunologically modified antigen-presenting cells expressing key structural proteins and a non-structural protein of SARS-CoV-2 to induce the body to produce an immune response and form an immunological memory for the prevention and treatment of SARS-CoV-2 infection.
- the present disclosure provides a protein or a combination of proteins for a SARS-CoV-2 vaccine.
- the protein or combination of proteins includes any one or a combination of at least two of Spike protein (Sprotein) , Envelope protein (E protein) , Membrane protein (M protein) , Nucleocapsid protein (N protein) or ORF1a poly-protease (P protein) of SARS-CoV-2.
- the protein or the combination of proteins for a vaccine containing key structural proteins and a non-structural protein of SARS-CoV-2 can stimulate the body to produce an immune response and form an immunological memory, with a broad-spectrum immunostimulatory effect.
- the present disclosure provides a fusion protein for a SARS-CoV-2 vaccine.
- the fusion protein includes an S protein signal peptide and an S protein receptor binding domain of SARS-CoV-2.
- the S protein receptor binding domain includes the amino acid sequence as shown in SEQ ID NO: 1.
- the S protein signal peptide includes the amino acid sequence as shown in SEQ ID NO: 2.
- the fusion protein contains an S protein receptor binding domain (RBD) , which is the most important structure for SARS-CoV-2 to invade human cells.
- RBD S protein receptor binding domain
- the fusion protein helps stimulate the body to produce anti-SARS-CoV-2 antibodies having high specificity.
- the fusion protein contains only the S protein signal peptide and the S protein receptor binding domain, and a short peptide at the carboxyl terminal is modified.
- a designed fusion protein S includes the amino acid sequence as shown in SEQ ID NO: 3.
- the fusion protein of the present disclosure includes the S protein signal peptide and the S protein receptor binding domain, and further includes a partial peptide fragment of the E protein and a partial peptide fragment of the M protein.
- the partial peptide fragment of the E protein includes an amino terminal peptide fragment of the E protein.
- the partial peptide fragment of the M protein includes a carboxyl terminal peptide fragment of the M protein.
- the amino terminal peptide fragment of the E protein includes the amino acid sequence as shown in SEQ ID NO: 4.
- the carboxyl terminal peptide fragment of the M protein includes the amino acid sequence as shown in SEQ ID NO: 5.
- the fusion protein consists of an S protein signal peptide, an S protein receptor binding domain, an amino terminal peptide fragment of the E protein and a peptide fragment of the M protein.
- a designed fusion protein SEM comprises the amino acid sequence as shown in SEQ ID NO: 6.
- the fusion protein of the present disclosure includes the S protein signal peptide, the S protein receptor binding domain, the amino terminal peptide fragment of the E protein, and the peptide fragment of the M protein, and further includes a partial peptide fragment of the N protein and a partial peptide fragment of the ORF1a poly-protease
- the partial peptide fragment of the N protein includes a helix-turn domain of the N protein.
- the partial peptide fragment of the ORF1a poly-protease includes a functional domain peptide fragment of the ORF1a poly-protease.
- the helix-turn domain of the N protein includes the amino acid sequence as shown in SEQ ID NO: 7.
- the peptide fragment of the ORF1a poly-protease includes the amino acid sequence as shown in SEQ ID NO: 8.
- the fusion protein consists of an S protein signal peptide, an S protein receptor binding domain, an amino terminal peptide fragment of the E protein, a peptide fragment of the M protein, a helix-turn domain of the N protein and a functional domain peptide fragment of the ORF1a poly-protease.
- a designed fusion protein SEMNP comprises the amino acid sequence as shown in SEQ ID NO: 9.
- a fusion protein containing several key proteins of SARS-CoV-2 is constructed based on the nucleic acid sequence of SARS-CoV-2.
- the fusion protein produces polypeptide fragments after enzymolysis, which, after presented by antigen-presenting cells, stimulate B cells and T cells in the body to become mature B cells and T cells that are specifically directed against viral antigens, and stimulate further proliferation of activated immune cells, to achieve prevention and controlling of infections.
- the present disclosure provides a coding gene of the fusion protein in the second aspect.
- the coding gene comprises a nucleic acid molecule encoding an S protein signal peptide and a nucleic acid molecule encoding an S protein receptor binding domain.
- the coding gene of the fusion protein S comprises the nucleic acid sequence as shown in SEQ ID NO: 10.
- the coding gene further includes a nucleic acid molecule encoding the amino terminal peptide fragment of the E protein and a nucleic acid molecule encoding the peptide fragment of the M protein.
- the encoding gene of the fusion protein SEM includes the nucleic acid sequence as shown in SEQ ID NO: 11.
- the coding gene further includes a nucleic acid molecule encoding the helix-turn domain of the N protein and a nucleic acid molecule encoding the functional domain peptide fragment of the ORF1a poly-protease.
- the coding gene of the fusion protein SEMNP includes the nucleic acid sequence as shown in SEQ ID NO: 12.
- the present disclosure provides an expression vector, comprising the coding gene in the third aspect.
- the expression vector includes any one of a lentiviral vector, a retroviral vector an adeno-associated viral vector or an adenovirus vector, preferably a lentiviral vector.
- the present disclosure provides a recombinant lentivirus, which is obtained by packaging of the lentiviral vector in the fourth aspect and a helper plasmid in mammalian cells.
- the present disclosure provides an antigen-presenting cell that expresses the fusion protein for a SARS-CoV-2 vaccine as described above, wherein the antigen-presenting cell further expresses a cytokine or cytokines; wherein the cytokine or cytokines include any one or a combination of at least two of Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12.
- CNX Calnexin
- artificial antigen presenting cells are constructed by introducing cytokines such as Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12 into myeloid-derived antigen-presenting cells using lentiviral technology.
- cytokines such as Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12
- the present disclosure provides a universal SARS-CoV-2 vaccine, which is an antigen-presenting cell expressing the fusion protein in the second aspect.
- the present disclosure adopts antigen-presenting cells expressing key proteins of SARS-CoV-2 to construct cellular vaccines, which is conducive to solving the safety and high cost problems of individualized cellular infusion, and can accelerate the preparation of cellular vaccines and rapidly realize large-scale industrial production. Moreover, the present disclosure adopts mitomycin-C and gamma-ray irradiation treatment to further strengthen the safety factor, which solves the risks in preparation and safety control of vaccine cells. In addition, the present disclosure strictly regulates and controls the preparation process, and reduces costs.
- the antigen-presenting cell expresses a cytokine or cytokines.
- the antigen-presenting cell expresses any one or a combination of at least two of Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12.
- CNX Calnexin
- the prophylactic and therapeutic process of the vaccine of the present disclosure is designed to mimic the natural immune system of the body.
- the structural and non-structural proteins of SARS-CoV-2 are exposed to stimulate the body to produce an immune response and form an immunological memory, and to produce specific antibodies against the SARS-CoV-2 proteins.
- a stronger immunological memory may be rapidly induced when re-infected with the virus.
- the coding gene in the third aspect is integrated into the genome of the antigen-presenting cell.
- the antigen-presenting cell includes the expression vector in the fourth aspect and/or the recombinant lentivirus in the fifth aspect.
- the present disclosure provides a method for preparing the vaccine in the seventh aspect, comprising:
- a cytokine or cytokines including any one or a combination of at least two of Calnexin, GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12
- a minigene encoding a fusion protein composed of several structural proteins and a non-structural protein of SARS-CoV-2 is synthesized, and transduced into immunologically modified artificial antigen-presenting cells (aAPC) through a lentiviral system.
- aAPC immunologically modified artificial antigen-presenting cells
- the constructed cellular vaccine has a broad-spectrum immunostimulatory effect, effectively stimulates cellular immune responses and production of corresponding antibodies in the body, and can be produced on a large scale.
- the inactivation treatment includes mitomycin-C treatment and/or gamma-ray irradiation.
- a concentrated minigene-containing lentiviral vector LV-SEMNP, LV-SEM or LV-S is used to transduce aAPC to establish a persistent artificially synthesized antigen-presenting cell line (COVID-19-: aAPC-SEMNP, aAPC-SEM or aAPC-S) , which is inactivated by mitomycin-C and/or gamma-ray irradiation to further enhance the vaccine safety factor, to prepare a universal COVID-19/aAPC vaccine.
- COVID-19- a persistent artificially synthesized antigen-presenting cell line
- the present disclosure has the following beneficial effects.
- a fusion protein containing structural proteins S protein, E protein, M protein, and N protein, and a non-structural protein ORF1a poly-protease of SARS-CoV-2, and a coding minigene thereof are constructed.
- the minigene is transduced into immunologically modified artificial antigen-presenting cells using a lentiviral system.
- the fusion protein is enzymatically digested in the body into several polypeptide fragments, which, after presented by antigen-presenting cells, can stimulate B cells and T cells in the body to become mature B cells and T cells that are specifically directed against viral antigens, and stimulate further proliferation of activated immune cells, to achieve prevention and controlling of infections.
- the prophylactic and therapeutic process of the vaccine of the present disclosure is designed to mimic the natural immune system of the body.
- the structural and non-structural proteins of SARS-CoV-2 are exposed to stimulate the body to produce an immune response and form an immunological memory, and to produce specific antibodies against the SARS-CoV-2 proteins.
- a stronger immunological memory may be rapidly induced when re-infected with the virus, with a broad-spectrum immunostimulatory effect.
- the present disclosure adopts a preferred antigen-presenting cell strain expressing key proteins of SARS-CoV-2 to construct cellular vaccines. It is conducive to solving the safety and high cost problems of individualized cellular infusion, accelerating the preparation time of cellular vaccines, rapidly realizing large-scale industrial production.
- mitomycin C and/or gamma-radiation are used for inactivation treatment, which further improve (s) the safety factor of the vaccine, solve (s) the risk of vaccine cell preparation and safety control, and strictly regulate (s) and control (s) the preparation process, and the costs are reduced.
- Figure 1 shows a schematic diagram of the preparation process of a universal SARS-CoV-2 vaccine.
- Figure 2 shows a lentiviral vector system containing a minigene.
- Figure 3 is a diagram showing the phenotype analysis of universal artificial vaccine cells (aAPCs) modified by lentivirus; wherein, Figure 3A shows a flow cytometric analysis comparing the expression of HLA antigens (-ABC, and -DP, DR, DQ) on cell surface between dendritic cells and aAPCs; and Figure 3B shows a flow cytometric analysis comparing the expression of CD80/CD86 and FL on cell surface and intracellular GM-CSF and IL-2 between unmodified aAPCs and lentivirus-modified aAPCs.
- aAPCs universal artificial vaccine cells
- Figure 4 shows the expression of viral proteins in aAPCs modified by lentiviruses containing different minigenes (aAPC-S, -SEM, -SEMNP) .
- Figure 5A shows the amplification of T cells from Donor 1 after aAPC-SEMNP stimulation
- Figure 5B shows the amplification of T cells from Donor 2 after aAPC-SEMNP stimulation
- Figure 5C shows the antigenic phenotypes of CD4+ T cells after stimulation of T cells from different donors with aAPC-SEMNP
- Figure 5D shows the antigenic phenotypes of CD8+ T cells after stimulation of T cells from different donors with aAPC-SEMNP.
- Figure 6A shows results of an effector response assay where the universal SARS-CoV-2 vaccine aAPC-SEMNP was used to stimulate T cells from Donor 1;
- Figure 6B shows results of an effector response test where the universal SARS-CoV-2 vaccine aAPC-SEMNP is used to stimulate T cells from Donor 2;
- Figure 6C shows results of an effector response test where the universal SARS-CoV-2 vaccine aAPC-SEMNP is used to stimulate T cells from Donor 3; and
- Figure 6D shows results of an effector response test where the universal SARS-CoV-2 vaccine aAPC-SEMNP is used to stimulate T cells from Donor 4.
- SARS-CoV-2 mainly expresses four structural proteins, i.e., surface glycoprotein (Sprotein) , envelop protein (E protein) , membrane protein (M protein) , and nucleocapsid phosphoprotein (N protein) , and a functional protein, polyprotein cleavage protease (ORF1a poly-protease) .
- Sprotein surface glycoprotein
- E protein envelop protein
- M protein membrane protein
- N protein nucleocapsid phosphoprotein
- ORF1a poly-protease polyprotein cleavage protease
- the S protein signal peptide (SEQ ID NO: 2) and S protein receptor binding domain (SEQ ID NO: 1) were selected as the first part of the fusion protein; the amino-terminal 29 aa peptide fragment (SEQ ID NO: 4) of the E protein was selected as the second part of the fusion protein; the carboxyl-terminal 58 aa peptide fragment (SEQ ID NO: 5) of the M protein was selected as the third part of the fusion protein; the intermediate 162 aa helix-turn domain (SEQ ID NO: 7) of the N protein was selected as the fourth part of the fusion protein; and the carboxyl-terminal 123 aa peptide (SEQ ID NO: 8) of the ORF1a poly-protease was selected as the fifth part of the fusion protein.
- the fusion protein SEMNP was truncated. Most peptides of the helix-turn domain of the N protein and all peptides of the carboxyl-terminal of the ORF1a poly-protease were deleted. A fusion protein SEM (SEQ ID NO: 6) with a length of 315 aa was obtained.
- the fusion protein SEMNP was further truncated. Only the S protein signal peptide and the S protein receptor binding domain were retained, and a short peptide fragment at the carboxyl-terminal was modified. A fusion protein S (SEQ ID NO: 3) with a length of 228 aa was obtained.
- the amino acid sequence of the fusion protein SEMNP was converted to a corresponding nucleic acid sequence, which was optimized to obtain the coding gene of the fusion protein SEMNP as shown in SEQ ID NO: 12.
- Partial NP sequence at the 3' end of the coding gene of SEMNP was deleted (nt. 4990-5840, NheI-SpeI deletion) , to obtain the coding gene of fusion protein SEM as shown in SEQ ID NO: 11.
- Partial EMNP sequence at the 3' end of the coding gene of SEMNP was deleted (nt. 4680-5850, BstBI-BstBI deletion) , to obtain the coding gene of fusion protein S as shown in SEQ ID NO: 10.
- the coding genes of the fusion proteins S, SEM and SEMNP as shown in SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12, respectively, were artificially synthesized.
- the above nucleic acid molecules were double digested, incubated in a 37 °C water bath for 30 min.
- the digested products were subjected to DNA electrophoresis on a 1.5%agarose gel.
- the digested fragments were purified and recovered using an agarose gel kit.
- the digested fragments were inserted into the linearized lentiviral vector TYF to construct lentiviral vectors LV-S, LV-SEM and LV-SEMNP containing minigenes.
- aAPCs were prepared using human-derived myeloid-associated cells: myelogenous leukemia AML, KG-1 or K562, for examples.
- Cells were modified with lentivirus to express CNX, GM-CSF, CD80, CD86, Flt-L, IL-2 and IL-12 in order to enhance the antigen presentation function of aAPCs.
- Cell strains expressing high levels of cytokines were screened as vaccine strains.
- Peripheral blood mononuclear cells were plated in culture dishes and cultured for 2 h in AIM-V medium (Gibco-BRL, CA, USA) ;
- Nonadherent cells were gently removed, and the culture was continued for 24 h using AIM-V medium supplemented with 50 ng/mL GM-CSF (Biosource, CA, USA) and 25 ng/mL IL-4 (Biosource, CA, USA) .
- AIM-V medium containing 20 ng/mL IFN- ⁇ (Gentaur) , 50 ng/mL TNF- ⁇ (R&D systems, MN, USA) , 10 ng/mL IL-1 ⁇ (R&D systems, MN, USA) , 10 ng/mL IL-6 (R&D systems) and 1 ⁇ M PGE2 (Sigma-Aldrich, MO, USA) , and cultured for 24 h to generate mature dendritic cells.
- the screened aAPC cell strains had been modified with lentivirus to express cytokines. Phenotypic analysis was performed by flow cytometry as follows.
- aAPCs were stained with antibodies PE-anti-HLA-ABC, FITC-anti-HLA-DR, DP, DQ, FITC-anti-CD80, and FITC-anti-CD86, kept at 4 °C for 30 min, and washed twice with PBS containing 1%FBS.
- Intracellular staining analysis was performed according to the literature. In brief, cells were treated with Monensin (Sigma) for 5 h, fixed and permeablized, followed by the addition of antibodies (BD Bioscience) against GM-CSF, IL-2, and Flt3-ligand (FL) , stained with FIX/PERM and PERM/Wash solution (BD) , and then loaded onto flow cytometry. The percentage of different antigen presenting cell subpopulations was analyzed with Flowjo software. The results are shown in Figure 3.
- aAPCs did not express HLA-ABC and -DP, -DR, -DQ, thus the immune rejection response was reduced.
- unmodified APCs expressed low levels of CD80 and CD86, and did not express GM-CSF and IL-2; modified aAPCs expressed high levels of CD80, CD86, GM-CSF and IL-2; unmodified APCs did not express Flt-L (FL) ; and modified aAPCs expressed a high level of FL.
- aAPCs were transduced with concentrated minigene-containing lentiviral vectors LV-SEMNP, LV-SEM or LV-S to establish persistent artificially synthesized antigen-presenting cell strains (COVID-19-: aAPC-SEMNP, aAPC-SEM and aAPC-S) , which were then inactivated through mitomycin-C and/or gamma-ray irradiation, to produce universal SARS-CoV-2 vaccines COVID-19/aAPC-SEMNP, COVID-19/aAPC-SEM and COVID-19/aAPC-S.
- Cell samples were lysed for 1 h with RIPA lysis solution.
- the protein concentration was determined by a BCA method.
- the protein concentration and the sample loading amount of the sample solution were calculated.
- the protein solution, 6 ⁇ SDS buffer and RIPA lysis solution were proportionally prepared into a total volume of 25 ⁇ L protein loading solution and incubated at 100 °C for 5 min.
- 10%upper and lower gels were prepared according to the Gel Fast Preparation Kit.
- the stacking gel was run at a voltage of 70 V, and the separating gel was run at a voltage of 120 V.
- a PVDF membrane was cut out and soaked in absolute ethanol. Filter paper used for transfer was soaked in the transfer buffer. A semi-dry transfer device was used to perform transfer.
- the membrane was blocked in 5%BSA for 1 h at room temperature.
- Primary antibodies of S protein, N protein, P protein and M protein were added after blocking and then incubated overnight in a refrigerator at 4°C.
- aAPC-S cells expressed a high level of S protein (26 kD)
- aAPC-SEMNP cell strains expressed a high level of SMENP fusion protein (66 kD)
- aAPC-SEM cells expressed SEM fusion protein (35 kD) .
- cytotoxic T cells were analyzed based on CD27, CD28, CD45RA, CD4 and CD8 surface staining by flow cytometry. Results are shown in Figures 5A, 5B, 5C and 5D, wherein Tcm refers to central memory T cells (CD27 + , CD28 + , CD45RA - ) , Tem refers to effector-memory T cells (CD27 +/- , CD28 +/- , CD45RA - ) , and Teff refers to terminal effector T cells (CD27 - , CD28 - , CD45RA - ) .
- Tcm refers to central memory T cells (CD27 + , CD28 + , CD45RA - )
- Tem refers to effector-memory T cells (CD27 +/- , CD28 +/- , CD45RA - )
- Teff refers to terminal effector T cells (CD27 - , CD28 - , CD45RA
- T cells from two different donors expanded after 2-3 weeks of stimulation.
- the expanded T cells were analyzed through surface antigen staining, and it showed the expression of antigen phenotypes of memory and effector T cells.
- Engineered immune effector cytotoxic T cells (EIE CTLs) generated after stimulation with COVID-19/aAPC vaccine were added with relevant antigenic peptides (a SEMNP mixture, S, E, M, N, P, and HIV peptides) , or artificial vaccine cells aAP-SEMNP, aAPC-SEM, or aAPC-S, and stimulated overnight. The strength of the antigen-specific response was detected using ELISPOT immunoassay.
- EIE EIE
- PBMCs peripheral blood mononuclear cells
- the universal SARS-CoV-2 vaccine of the present disclosure mimics the natural immune system of the body.
- the several polypeptide fragments formed from the fusion protein are delivered by antigen-presenting cells and stimulate the body to generate an immune response and form an immunological memory.
Landscapes
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Cell Biology (AREA)
- Microbiology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- Virology (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Biotechnology (AREA)
- Biophysics (AREA)
- General Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Physics & Mathematics (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Plant Pathology (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
A universal SARS-CoV-2 vaccine and a preparation method thereof. The vaccine is an artificial antigen-presenting cell expressing a fusion protein of SARS-CoV-2 structural proteins S protein, E protein, M protein, N protein and non-structural protein ORF1a poly-protease P. The vaccine mimics the natural immune system of the body, and in the presence of cytokines, multiple polypeptide fragments formed by the fusion protein are presented by the antigen-presenting cells, which can stimulate the body to generate immune responses and form immunological memory, with a broad-spectrum immunostimulatory effect; the vaccine can realize rapid large-scale industrial production, with the advantages of high safety and low cost.
Description
The present disclosure belongs to the field of biological medicine, and relates to a universal SARS-CoV-2 vaccine and a preparation method thereof.
Binding of Spike protein (Sprotein) to ACE2 receptor on the cell surface is key for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to infect human cells. The S protein plays a same role in SARS and MERS viruses. Therefore, the S protein determines the host range and specificity of the virus, and it is an important target for antibody binding in the host, and is also a key target for the development of vaccines, therapeutic antibodies, and diagnostic reagents.
The virus causing COVID-19 is a newly emerged coronavirus variant. The spike protein of this virus binds to ACE2 receptor with a stronger force than SARS-CoV, which explains the strong infection ability of SARS-CoV-2. SARS-CoV-2 stimulates the innate immune system in an infected individual, causing the release of a large amount of cytokines in the body and causing a cytokine storm and an acute inflammatory response, thereby leading to acute respiratory distress syndrome (ARDS) and multiple organ failure. At the present stage, the preventive effects of available vaccines or drugs to COVID-19 are limited, and vaccine development is challenging due to the limited knowledge about SARS-CoV-2. A great challenge in vaccine development is to find a suitable animal model. Currently, non-human primate infection models are mainly used to verify the safety and efficacy of the developed vaccines, but the cost is extremely high and it takes a long time.
In the existing virus vaccine technology, the whole virus antigen vaccine is based on the traditional inactivated virus or attenuated virus as antigen source, which has the problems of low safety and high cost; the viral specific antigen vaccine is based on synthetic S protein as the antigen, while artificially synthesized S protein is mainly expressed by bacteria, yeast, cells or viral vectors, which has the problems of a length process of manufacture, single valence, easy escape of the virus, short validity period, and high cost; DNA and mRNA vaccines need to enter cells and be converted into proteins to exert immune effects, and the low immunogenicity issue has been the bottleneck that the technology still cannot overcome at present time.
SUMMARY
The present disclosure provides a universal SARS-CoV-2 vaccine composition and a preparation method thereof. The vaccine uses immunologically modified antigen-presenting cells expressing key structural proteins and a non-structural protein of SARS-CoV-2 to induce the body to produce an immune response and form an immunological memory for the prevention and treatment of SARS-CoV-2 infection.
To achieve this, the following technical solutions are used in the present disclosure.
In a first aspect, the present disclosure provides a protein or a combination of proteins for a SARS-CoV-2 vaccine. The protein or combination of proteins includes any one or a combination of at least two of Spike protein (Sprotein) , Envelope protein (E protein) , Membrane protein (M protein) , Nucleocapsid protein (N protein) or ORF1a poly-protease (P protein) of SARS-CoV-2.
In the present disclosure, the protein or the combination of proteins for a vaccine containing key structural proteins and a non-structural protein of SARS-CoV-2 can stimulate the body to produce an immune response and form an immunological memory, with a broad-spectrum immunostimulatory effect.
In a second aspect, the present disclosure provides a fusion protein for a SARS-CoV-2 vaccine. The fusion protein includes an S protein signal peptide and an S protein receptor binding domain of SARS-CoV-2.
In a specific embodiment, the S protein receptor binding domain includes the amino acid sequence as shown in SEQ ID NO: 1.
SEQ ID NO: 1:
In a specific embodiment, the S protein signal peptide includes the amino acid sequence as shown in SEQ ID NO: 2.
SEQ ID NO: 2:
MFVFLVLLPLVSSQCVNLTTRTQLP.
In the present disclosure, the fusion protein contains an S protein receptor binding domain (RBD) , which is the most important structure for SARS-CoV-2 to invade human cells. The fusion protein helps stimulate the body to produce anti-SARS-CoV-2 antibodies having high specificity.
Preferably, the fusion protein contains only the S protein signal peptide and the S protein receptor binding domain, and a short peptide at the carboxyl terminal is modified. A designed fusion protein S includes the amino acid sequence as shown in SEQ ID NO: 3.
SEQ ID NO: 3:
The fusion protein of the present disclosure includes the S protein signal peptide and the S protein receptor binding domain, and further includes a partial peptide fragment of the E protein and a partial peptide fragment of the M protein.
Preferably, the partial peptide fragment of the E protein includes an amino terminal peptide fragment of the E protein.
Preferably, the partial peptide fragment of the M protein includes a carboxyl terminal peptide fragment of the M protein.
Preferably, the amino terminal peptide fragment of the E protein includes the amino acid sequence as shown in SEQ ID NO: 4.
SEQ ID NO: 4:
MYSFVSEETGTLIVNSVLLFLAFVVFLLV.
Preferably, the carboxyl terminal peptide fragment of the M protein includes the amino acid sequence as shown in SEQ ID NO: 5.
SEQ ID NO: 5:
PKEITVATSRTLSYYKLGASQRVAGDSGFAAYSRYRIGNYKLNTDHSSSSDNIALLVQ.
Preferably, the fusion protein consists of an S protein signal peptide, an S protein receptor binding domain, an amino terminal peptide fragment of the E protein and a peptide fragment of the M protein. A designed fusion protein SEM comprises the amino acid sequence as shown in SEQ ID NO: 6.
SEQ ID NO: 6:
The fusion protein of the present disclosure includes the S protein signal peptide, the S protein receptor binding domain, the amino terminal peptide fragment of the E protein, and the peptide fragment of the M protein, and further includes a partial peptide fragment of the N protein and a partial peptide fragment of the ORF1a poly-protease
Preferably, the partial peptide fragment of the N protein includes a helix-turn domain of the N protein.
Preferably, the partial peptide fragment of the ORF1a poly-protease includes a functional domain peptide fragment of the ORF1a poly-protease.
Preferably, the helix-turn domain of the N protein includes the amino acid sequence as shown in SEQ ID NO: 7.
SEQ ID NO: 7:
Preferably, the peptide fragment of the ORF1a poly-protease includes the amino acid sequence as shown in SEQ ID NO: 8.
SEQ ID NO: 8:
Preferably, the fusion protein consists of an S protein signal peptide, an S protein receptor binding domain, an amino terminal peptide fragment of the E protein, a peptide fragment of the M protein, a helix-turn domain of the N protein and a functional domain peptide fragment of the ORF1a poly-protease. A designed fusion protein SEMNP comprises the amino acid sequence as shown in SEQ ID NO: 9.
SEQ ID NO: 9:
In the present disclosure, a fusion protein containing several key proteins of SARS-CoV-2 is constructed based on the nucleic acid sequence of SARS-CoV-2. The fusion protein produces polypeptide fragments after enzymolysis, which, after presented by antigen-presenting cells, stimulate B cells and T cells in the body to become mature B cells and T cells that are specifically directed against viral antigens, and stimulate further proliferation of activated immune cells, to achieve prevention and controlling of infections.
In a third aspect, the present disclosure provides a coding gene of the fusion protein in the second aspect. The coding gene comprises a nucleic acid molecule encoding an S protein signal peptide and a nucleic acid molecule encoding an S protein receptor binding domain. The coding gene of the fusion protein S comprises the nucleic acid sequence as shown in SEQ ID NO: 10.
SEQ ID NO: 10:
Preferably, the coding gene further includes a nucleic acid molecule encoding the amino terminal peptide fragment of the E protein and a nucleic acid molecule encoding the peptide fragment of the M protein. The encoding gene of the fusion protein SEM includes the nucleic acid sequence as shown in SEQ ID NO: 11.
SEQ ID NO: 11:
Preferably, the coding gene further includes a nucleic acid molecule encoding the helix-turn domain of the N protein and a nucleic acid molecule encoding the functional domain peptide fragment of the ORF1a poly-protease. The coding gene of the fusion protein SEMNP includes the nucleic acid sequence as shown in SEQ ID NO: 12.
SEQ ID NO: 12:
In a fourth aspect, the present disclosure provides an expression vector, comprising the coding gene in the third aspect.
Preferably, the expression vector includes any one of a lentiviral vector, a retroviral vector an adeno-associated viral vector or an adenovirus vector, preferably a lentiviral vector.
In a fifth aspect, the present disclosure provides a recombinant lentivirus, which is obtained by packaging of the lentiviral vector in the fourth aspect and a helper plasmid in mammalian cells.
In a sixth aspect, the present disclosure provides an antigen-presenting cell that expresses the fusion protein for a SARS-CoV-2 vaccine as described above, wherein the antigen-presenting cell further expresses a cytokine or cytokines; wherein the cytokine or cytokines include any one or a combination of at least two of Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12.
In the present disclosure, artificial antigen presenting cells (aAPCs) are constructed by introducing cytokines such as Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12 into myeloid-derived antigen-presenting cells using lentiviral technology.
In a seventh aspect, the present disclosure provides a universal SARS-CoV-2 vaccine, which is an antigen-presenting cell expressing the fusion protein in the second aspect.
The present disclosure adopts antigen-presenting cells expressing key proteins of SARS-CoV-2 to construct cellular vaccines, which is conducive to solving the safety and high cost problems of individualized cellular infusion, and can accelerate the preparation of cellular vaccines and rapidly realize large-scale industrial production. Moreover, the present disclosure adopts mitomycin-C and gamma-ray irradiation treatment to further strengthen the safety factor, which solves the risks in preparation and safety control of vaccine cells. In addition, the present disclosure strictly regulates and controls the preparation process, and reduces costs.
Preferably, the antigen-presenting cell expresses a cytokine or cytokines.
Preferably, the antigen-presenting cell expresses any one or a combination of at least two of Calnexin (CNX) , GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12.
The prophylactic and therapeutic process of the vaccine of the present disclosure is designed to mimic the natural immune system of the body. In the presence of cytokines, the structural and non-structural proteins of SARS-CoV-2 are exposed to stimulate the body to produce an immune response and form an immunological memory, and to produce specific antibodies against the SARS-CoV-2 proteins. Thereby, a stronger immunological memory may be rapidly induced when re-infected with the virus.
Preferably, the coding gene in the third aspect is integrated into the genome of the antigen-presenting cell.
Preferably, the antigen-presenting cell includes the expression vector in the fourth aspect and/or the recombinant lentivirus in the fifth aspect.
In an eighth aspect, the present disclosure provides a method for preparing the vaccine in the seventh aspect, comprising:
transducing an antigen-presenting cell that expresses a cytokine or cytokines including any one or a combination of at least two of Calnexin, GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12 with the recombinant lentivirus in the fifth aspect, and carrying out inactivation treatment, to obtain the vaccine.
In the present disclosure, a minigene encoding a fusion protein composed of several structural proteins and a non-structural protein of SARS-CoV-2 is synthesized, and transduced into immunologically modified artificial antigen-presenting cells (aAPC) through a lentiviral system. The constructed cellular vaccine has a broad-spectrum immunostimulatory effect, effectively stimulates cellular immune responses and production of corresponding antibodies in the body, and can be produced on a large scale.
Preferably, the inactivation treatment includes mitomycin-C treatment and/or gamma-ray irradiation.
In the present disclosure, a concentrated minigene-containing lentiviral vector LV-SEMNP, LV-SEM or LV-S is used to transduce aAPC to establish a persistent artificially synthesized antigen-presenting cell line (COVID-19-: aAPC-SEMNP, aAPC-SEM or aAPC-S) , which is inactivated by mitomycin-C and/or gamma-ray irradiation to further enhance the vaccine safety factor, to prepare a universal COVID-19/aAPC vaccine.
Compared with the prior art, the present disclosure has the following beneficial effects.
(1) In the present disclosure, a fusion protein containing structural proteins S protein, E protein, M protein, and N protein, and a non-structural protein ORF1a poly-protease of SARS-CoV-2, and a coding minigene thereof are constructed. The minigene is transduced into immunologically modified artificial antigen-presenting cells using a lentiviral system. The fusion protein is enzymatically digested in the body into several polypeptide fragments, which, after presented by antigen-presenting cells, can stimulate B cells and T cells in the body to become mature B cells and T cells that are specifically directed against viral antigens, and stimulate further proliferation of activated immune cells, to achieve prevention and controlling of infections.
(2) The prophylactic and therapeutic process of the vaccine of the present disclosure is designed to mimic the natural immune system of the body. In the presence of cytokines, the structural and non-structural proteins of SARS-CoV-2 are exposed to stimulate the body to produce an immune response and form an immunological memory, and to produce specific antibodies against the SARS-CoV-2 proteins. Thereby, a stronger immunological memory may be rapidly induced when re-infected with the virus, with a broad-spectrum immunostimulatory effect.
(3) In order to realize rapid large-scale industrial production of vaccines, the present disclosure adopts a preferred antigen-presenting cell strain expressing key proteins of SARS-CoV-2 to construct cellular vaccines. It is conducive to solving the safety and high cost problems of individualized cellular infusion, accelerating the preparation time of cellular vaccines, rapidly realizing large-scale industrial production. In the vaccine preparation process, mitomycin C and/or gamma-radiation (are) is used for inactivation treatment, which further improve (s) the safety factor of the vaccine, solve (s) the risk of vaccine cell preparation and safety control, and strictly regulate (s) and control (s) the preparation process, and the costs are reduced.
BRIEF DESCRIPTION OF DRAWINGS
Figure 1 shows a schematic diagram of the preparation process of a universal SARS-CoV-2 vaccine.
Figure 2 shows a lentiviral vector system containing a minigene.
Figure 3 is a diagram showing the phenotype analysis of universal artificial vaccine cells (aAPCs) modified by lentivirus; wherein, Figure 3A shows a flow cytometric analysis comparing the expression of HLA antigens (-ABC, and -DP, DR, DQ) on cell surface between dendritic cells and aAPCs; and Figure 3B shows a flow cytometric analysis comparing the expression of CD80/CD86 and FL on cell surface and intracellular GM-CSF and IL-2 between unmodified aAPCs and lentivirus-modified aAPCs.
Figure 4 shows the expression of viral proteins in aAPCs modified by lentiviruses containing different minigenes (aAPC-S, -SEM, -SEMNP) .
Figure 5A shows the amplification of T cells from Donor 1 after aAPC-SEMNP stimulation; Figure 5B shows the amplification of T cells from Donor 2 after aAPC-SEMNP stimulation; Figure 5C shows the antigenic phenotypes of CD4+ T cells after stimulation of T cells from different donors with aAPC-SEMNP; and Figure 5D shows the antigenic phenotypes of CD8+ T cells after stimulation of T cells from different donors with aAPC-SEMNP.
Figure 6A shows results of an effector response assay where the universal SARS-CoV-2 vaccine aAPC-SEMNP was used to stimulate T cells from Donor 1; Figure 6B shows results of an effector response test where the universal SARS-CoV-2 vaccine aAPC-SEMNP is used to stimulate T cells from Donor 2; Figure 6C shows results of an effector response test where the universal SARS-CoV-2 vaccine aAPC-SEMNP is used to stimulate T cells from Donor 3; and Figure 6D shows results of an effector response test where the universal SARS-CoV-2 vaccine aAPC-SEMNP is used to stimulate T cells from Donor 4.
In order to further illustrate the technical means adopted by the present disclosure and effects thereof, the present disclosure will be further described below in conjunction with examples and drawings. It should be understood that the specific embodiments described herein are only used to explain the present disclosure, but not to limit the present disclosure.
Experiments without specific techniques or conditions noted in the examples were conducted according to techniques or conditions described in the literature in the art or a product instruction. The reagents or instruments used herein without manufacturers specified were conventional products commercially available from proper channels.
Example 1 Design of fusion protein
Comparison of the nucleic acid sequences of SARS-CoV-2 with SARS and MERS viruses reveals that SARS-CoV-2 mainly expresses four structural proteins, i.e., surface glycoprotein (Sprotein) , envelop protein (E protein) , membrane protein (M protein) , and nucleocapsid phosphoprotein (N protein) , and a functional protein, polyprotein cleavage protease (ORF1a poly-protease) .
In this example, based on the published full-length 1273 aa amino acid sequence of S protein (MN908947.3) , the S protein signal peptide (SEQ ID NO: 2) and S protein receptor binding domain (SEQ ID NO: 1) were selected as the first part of the fusion protein; the amino-terminal 29 aa peptide fragment (SEQ ID NO: 4) of the E protein was selected as the second part of the fusion protein; the carboxyl-terminal 58 aa peptide fragment (SEQ ID NO: 5) of the M protein was selected as the third part of the fusion protein; the intermediate 162 aa helix-turn domain (SEQ ID NO: 7) of the N protein was selected as the fourth part of the fusion protein; and the carboxyl-terminal 123 aa peptide (SEQ ID NO: 8) of the ORF1a poly-protease was selected as the fifth part of the fusion protein. A fusion protein SEMNP (SEQ ID NO: 9) with a length of 595 aa was obtained. Further, the length of each viral protein in the fusion protein can be increased or decreased, for example as follows.
In this example, the fusion protein SEMNP was truncated. Most peptides of the helix-turn domain of the N protein and all peptides of the carboxyl-terminal of the ORF1a poly-protease were deleted. A fusion protein SEM (SEQ ID NO: 6) with a length of 315 aa was obtained.
In this example, the fusion protein SEMNP was further truncated. Only the S protein signal peptide and the S protein receptor binding domain were retained, and a short peptide fragment at the carboxyl-terminal was modified. A fusion protein S (SEQ ID NO: 3) with a length of 228 aa was obtained.
Example 2 Design of minigene
The amino acid sequence of the fusion protein SEMNP was converted to a corresponding nucleic acid sequence, which was optimized to obtain the coding gene of the fusion protein SEMNP as shown in SEQ ID NO: 12.
Partial NP sequence at the 3' end of the coding gene of SEMNP was deleted (nt. 4990-5840, NheI-SpeI deletion) , to obtain the coding gene of fusion protein SEM as shown in SEQ ID NO: 11.
Partial EMNP sequence at the 3' end of the coding gene of SEMNP was deleted (nt. 4680-5850, BstBI-BstBI deletion) , to obtain the coding gene of fusion protein S as shown in SEQ ID NO: 10.
Example 3 Construction of lentiviral vector
The coding genes of the fusion proteins S, SEM and SEMNP as shown in SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12, respectively, were artificially synthesized. The above nucleic acid molecules were double digested, incubated in a 37 ℃ water bath for 30 min. The digested products were subjected to DNA electrophoresis on a 1.5%agarose gel. The digested fragments were purified and recovered using an agarose gel kit.
As shown in Figure 2, the digested fragments were inserted into the linearized lentiviral vector TYF to construct lentiviral vectors LV-S, LV-SEM and LV-SEMNP containing minigenes.
Example 4 Preparation of artificial antigen-presenting cells (aAPCs) and dendritic cells (DCs)
(1) Preparation of aAPCs
aAPCs were prepared using human-derived myeloid-associated cells: myelogenous leukemia AML, KG-1 or K562, for examples. Cells were modified with lentivirus to express CNX, GM-CSF, CD80, CD86, Flt-L, IL-2 and IL-12 in order to enhance the antigen presentation function of aAPCs. Cell strains expressing high levels of cytokines were screened as vaccine strains.
(2) Preparation of DCs
Peripheral blood mononuclear cells were plated in culture dishes and cultured for 2 h in AIM-V medium (Gibco-BRL, CA, USA) ;
Nonadherent cells were gently removed, and the culture was continued for 24 h using AIM-V medium supplemented with 50 ng/mL GM-CSF (Biosource, CA, USA) and 25 ng/mL IL-4 (Biosource, CA, USA) .
Cells were washed with PBS, replaced with fresh AIM-V medium containing 20 ng/mL IFN-γ (Gentaur) , 50 ng/mL TNF-α (R&D systems, MN, USA) , 10 ng/mL IL-1β (R&D systems, MN, USA) , 10 ng/mL IL-6 (R&D systems) and 1 μM PGE2 (Sigma-Aldrich, MO, USA) , and cultured for 24 h to generate mature dendritic cells.
Example 5 Phenotypic analysis of artificial antigen-presenting cells (aAPCs)
The screened aAPC cell strains had been modified with lentivirus to express cytokines. Phenotypic analysis was performed by flow cytometry as follows.
aAPCs were stained with antibodies PE-anti-HLA-ABC, FITC-anti-HLA-DR, DP, DQ, FITC-anti-CD80, and FITC-anti-CD86, kept at 4 ℃ for 30 min, and washed twice with PBS containing 1%FBS.
Intracellular staining analysis was performed according to the literature. In brief, cells were treated with Monensin (Sigma) for 5 h, fixed and permeablized, followed by the addition of antibodies (BD Bioscience) against GM-CSF, IL-2, and Flt3-ligand (FL) , stained with FIX/PERM and PERM/Wash solution (BD) , and then loaded onto flow cytometry. The percentage of different antigen presenting cell subpopulations was analyzed with Flowjo software. The results are shown in Figure 3.
As shown in Figure 3A, compared with normal immature DC cells, aAPCs did not express HLA-ABC and -DP, -DR, -DQ, thus the immune rejection response was reduced. As shown in Figure 3B, unmodified APCs expressed low levels of CD80 and CD86, and did not express GM-CSF and IL-2; modified aAPCs expressed high levels of CD80, CD86, GM-CSF and IL-2; unmodified APCs did not express Flt-L (FL) ; and modified aAPCs expressed a high level of FL.
Example 6 Preparation of a universal SARS-CoV-2 vaccine
The preparation process of the universal SARS-CoV-2 vaccine is shown in Figure 1. First, aAPCs were transduced with concentrated minigene-containing lentiviral vectors LV-SEMNP, LV-SEM or LV-S to establish persistent artificially synthesized antigen-presenting cell strains (COVID-19-: aAPC-SEMNP, aAPC-SEM and aAPC-S) , which were then inactivated through mitomycin-C and/or gamma-ray irradiation, to produce universal SARS-CoV-2 vaccines COVID-19/aAPC-SEMNP, COVID-19/aAPC-SEM and COVID-19/aAPC-S.
Example 7 Analysis of viral protein expression by universal SARS-CoV-2 vaccine (aAPC-SEMNP)
Cell samples were lysed for 1 h with RIPA lysis solution. The protein concentration was determined by a BCA method. The protein concentration and the sample loading amount of the sample solution were calculated.
According to the protein quantification results, the protein solution, 6×SDS buffer and RIPA lysis solution were proportionally prepared into a total volume of 25 μL protein loading solution and incubated at 100 ℃ for 5 min.
10%upper and lower gels were prepared according to the Gel Fast Preparation Kit. The stacking gel was run at a voltage of 70 V, and the separating gel was run at a voltage of 120 V.
According to the position of the protein molecular weight (Marker) and the size of the gel, a PVDF membrane was cut out and soaked in absolute ethanol. Filter paper used for transfer was soaked in the transfer buffer. A semi-dry transfer device was used to perform transfer.
After the transfer, the membrane was blocked in 5%BSA for 1 h at room temperature. Primary antibodies of S protein, N protein, P protein and M protein were added after blocking and then incubated overnight in a refrigerator at 4℃.
The next day, secondary antibodies were added and incubated at room temperature for 1 h. Finally, the PVDF membrane was treated with a HRP Substrate Kit and exposed on an Imager.
The results are shown in Figure 4. aAPC-S cells expressed a high level of S protein (26 kD) , aAPC-SEMNP cell strains expressed a high level of SMENP fusion protein (66 kD) , and aAPC-SEM cells expressed SEM fusion protein (35 kD) .
Example 8 Analysis of function of T cells stimulated by universal SARS-CoV-2 vaccine aAPC-SEMNP
(1) Functional test 1 of Covid-19/aAPC-stimulated T cells:
After stimulation of peripheral blood T cells by COVID-19/aAPC vaccine, cytotoxic T cells (CTLs) were analyzed based on CD27, CD28, CD45RA, CD4 and CD8 surface staining by flow cytometry. Results are shown in Figures 5A, 5B, 5C and 5D, wherein Tcm refers to central memory T cells (CD27
+, CD28
+, CD45RA
-) , Tem refers to effector-memory T cells (CD27
+/-, CD28
+/-, CD45RA
-) , and Teff refers to terminal effector T cells (CD27
-, CD28
-, CD45RA
-) .
As shown in Figure 5A and Figure 5B, T cells from two different donors expanded after 2-3 weeks of stimulation. As shown in Figure 5C and Figure 5D, the expanded T cells were analyzed through surface antigen staining, and it showed the expression of antigen phenotypes of memory and effector T cells.
(2) Functional test 2 of Covid-19/aAPC stimulated T cells.
Engineered immune effector cytotoxic T cells (EIE CTLs) generated after stimulation with COVID-19/aAPC vaccine were added with relevant antigenic peptides (a SEMNP mixture, S, E, M, N, P, and HIV peptides) , or artificial vaccine cells aAP-SEMNP, aAPC-SEM, or aAPC-S, and stimulated overnight. The strength of the antigen-specific response was detected using ELISPOT immunoassay.
CTLs (EIE) generated after stimulation of peripheral blood mononuclear cells (PBMCs) from 4 healthy donors with SEMNP antigen were stimulated again with Covid-19 peptides S, M, E, N, P, and SEMNP (a mixture of 5 peptides) , and different aAPC vaccine cells for overnight, and ELISpot analysis was performed.
The results are shown in Figure 6A, Figure 6B, Figure 6C and Figure 6D. Each donor expressed and produced IFN-γ after stimulation with different protein peptides, indicating that the aAPC vaccines (-S, -SEM, or -SEMNP) induced T cell-specific immune responses in large scale.
In summary, the universal SARS-CoV-2 vaccine of the present disclosure mimics the natural immune system of the body. In the presence of cytokines, the several polypeptide fragments formed from the fusion protein are delivered by antigen-presenting cells and stimulate the body to generate an immune response and form an immunological memory.
The applicant declares that the present disclosure uses the above-mentioned examples to illustrate the detailed methods of the present disclosure, but the present disclosure is not limited to the above-mentioned detailed methods, which does not mean that the present disclosure must rely on the above-mentioned detailed methods to be implemented. Those skilled in the art should understand that any improvement of the present disclosure, the equivalent replacement of each raw material of the product of the present disclosure, the addition of auxiliary components, the selection of specific methods, etc. fall within the scope of protection and disclosure of the present disclosure.
Claims (14)
- A protein or a combination of proteins for a SARS-CoV-2 vaccine, comprising any one or a combination of at least two of S protein, E protein, M protein, N protein or ORF1a poly-protease P of SARS-CoV-2.
- A fusion protein for a SARS-CoV-2 vaccine, comprising an S protein signal peptide and an S protein receptor binding domain of SARS-CoV-2.
- The fusion protein according to claim 2, wherein the S protein receptor binding domain includes the amino acid sequence as shown in SEQ ID NO: 1; and/orthe S protein signal peptide includes the amino acid sequence as shown in SEQ ID NO: 2.
- The fusion protein according to claim 2 or 3, wherein the fusion protein consists of the S protein signal peptide and the S protein receptor binding domain;preferably, the fusion protein includes the amino acid sequence as shown in SEQ ID NO: 3.
- The fusion protein according to claim 2 or 3, wherein the fusion protein further includes a partial peptide fragment of E protein and a partial peptide fragment of M protein;preferably, the partial peptide fragment of the E protein includes an amino terminal peptide fragment of the E protein;preferably, the partial peptide fragment of the M protein includes a carboxyl terminal peptide fragment of the M protein;preferably, the amino terminal peptide fragment of the E protein includes the amino acid sequence as shown in SEQ ID NO: 4;preferably, the carboxyl terminal peptide fragment of the M protein includes the amino acid sequence as shown in SEQ ID NO: 5.
- The fusion protein according to claim 5, wherein the fusion protein consists of the S protein signal peptide, the S protein receptor binding domain, the amino terminal peptide fragment of the E protein, and the peptide fragment of the M protein;preferably, the fusion protein includes the amino acid sequence as shown in SEQ ID NO: 6.
- The fusion protein according to any one of claims 2-3 and 5-6, wherein the fusion protein further includes a partial peptide fragment of N protein and a partial peptide fragment of ORF1a poly-protease;preferably, the partial peptide fragment of the N protein includes a helix-turn domain of the N protein;preferably, the partial peptide fragment of the ORF1a poly-protease includes a functional domain peptide fragment (3CLpro) of the ORF1a poly-protease;preferably, the helix-turn domain of the N protein includes the amino acid sequence as shown in SEQ ID NO: 7;preferably, the peptide fragment of the ORF1a poly-protease includes the amino acid sequence as shown in SEQ ID NO: 8.
- The fusion protein according to claim 7, wherein the fusion protein consists of the S protein signal peptide, the S protein receptor binding domain, the amino terminal peptide fragment of the E protein, the peptide fragment of the M protein, the helix-turn domain of the N protein and the functional domain peptide fragment of the ORF1a poly-protease;preferably, the fusion protein includes the amino acid sequence as shown in SEQ ID NO: 9.
- A coding gene of the fusion protein of any one of claims 2-8, including a nucleic acid molecule encoding the S protein signal peptide and a nucleic acid molecule encoding the S protein receptor binding domain;preferably, the coding gene includes the nucleic acid sequence as shown in SEQ ID NO: 10;preferably, the coding gene further includes a nucleic acid molecule encoding the amino terminal peptide fragment of the E protein and a nucleic acid molecule encoding the peptide fragment of the M protein;preferably, the coding gene includes the nucleic acid sequence as shown in SEQ ID NO: 11;preferably, the coding gene further includes a nucleic acid molecule encoding the helix-turn domain of the N protein and a nucleic acid molecule encoding the functional domain peptide fragment of the ORF1a poly-protease;preferably, the coding gene includes the nucleic acid sequence as shown in SEQ ID NO: 12.
- An expression vector, including the coding gene of claim 9;preferably, the expression vector includes any one of a lentiviral vector, a retroviral vector or an adeno-associated virus vector, preferably a lentiviral vector.
- A recombinant lentivirus, which is obtained by packaging of the expression vector of claim 10 and a helper plasmid in mammalian cells.
- An antigen-presenting cell that expresses the fusion protein for a SARS-CoV-2 vaccine of any one of claims 2-8, wherein the antigen-presenting cell further expresses a cytokine or cytokines;wherein the cytokine or cytokines include any one or a combination of at least two of Calnexin, GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12.
- A universal SARS-CoV-2 vaccine, which is an antigen-presenting cell expressing the fusion protein of any one of claims 2-8;preferably, the antigen-presenting cell further expresses a cytokine or cytokines;preferably, the cytokine or cytokines include any one or a combination of at least two of Calnexin, GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12;preferably, the coding gene of claim 9 is integrated into the genome of the antigen-presenting cell;preferably, the antigen-presenting cell includes the expression vector of claim 10 and/or the recombinant lentivirus of claim 11.
- A method for preparing the vaccine of claim 13, comprising:transducing an antigen-presenting cell that expresses a cytokine or cytokines including any one or a combination of at least two of Calnexin, GM-CSF, CD80, CD86, Flt3-L, IL-2 or IL-12 with the recombinant lentivirus of claim 11, and carrying out inactivation treatment, to obtain the vaccine;preferably, the inactivation treatment includes mitomycin-C treatment and/or gamma-ray irradiation.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202010955986.3 | 2020-09-11 | ||
CN202010955986.3A CN112048007B (en) | 2020-09-11 | 2020-09-11 | Universal novel coronavirus vaccine and preparation method thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022052984A1 true WO2022052984A1 (en) | 2022-03-17 |
Family
ID=73610244
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2021/117415 WO2022052984A1 (en) | 2020-09-11 | 2021-09-09 | Universal sars-cov-2 vaccine and preparation method thereof |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN112048007B (en) |
WO (1) | WO2022052984A1 (en) |
Families Citing this family (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN112048007B (en) * | 2020-09-11 | 2022-07-12 | 北京美康基免生物科技有限公司 | Universal novel coronavirus vaccine and preparation method thereof |
CN114517205A (en) * | 2020-11-20 | 2022-05-20 | 北京震旦鼎泰生物科技有限公司 | Fusion gene, recombinant novel coronavirus efficient immune tripod molecular DNA vaccine, and construction method and application thereof |
WO2022122036A1 (en) * | 2020-12-10 | 2022-06-16 | 武汉博沃生物科技有限公司 | Immunogen and pharmaceutical composition for sars-cov-2 virus, and use thereof |
CN114751964B (en) * | 2020-12-29 | 2024-03-08 | 苏州方舟生物科技有限公司 | Universal vaccine protein fragment of beta coronavirus, screening method and application thereof |
CN114656529B (en) * | 2021-02-08 | 2024-05-31 | 暨南大学 | Novel antigen epitope peptide of coronavirus T cell and application thereof |
CN113018427B (en) * | 2021-03-10 | 2023-09-19 | 江苏健安生物科技有限公司 | Multivalent fusion protein vaccine based on neutralizing epitope of novel coronavirus |
CN114907452B (en) * | 2022-04-08 | 2023-07-25 | 国科宁波生命与健康产业研究院 | M protein polypeptide for treating SARS-CoV-2 virus infection |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111218458A (en) * | 2020-02-27 | 2020-06-02 | 珠海丽凡达生物技术有限公司 | mRNAs encoding SARS-CoV-2 virus antigen and vaccine and preparation method of vaccine |
CN111607003A (en) * | 2020-05-21 | 2020-09-01 | 泰州市百英生物科技有限公司 | SARS-CoV-2N/S1(RBD) recombinant protein and its preparation method and application |
CN111732638A (en) * | 2020-07-02 | 2020-10-02 | 重庆博唯佰泰生物制药有限公司 | Vaccine against SARS-CoV-2 |
CN112048007A (en) * | 2020-09-11 | 2020-12-08 | 北京美康基免生物科技有限公司 | Universal novel coronavirus vaccine and preparation method thereof |
CN113336832A (en) * | 2020-03-02 | 2021-09-03 | 成都威斯克生物医药有限公司 | Protein for resisting SARS-CoV-2 infection and vaccine containing the protein |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
SE0301109D0 (en) * | 2003-04-14 | 2003-04-14 | Mallen Huang | Nucleotide vaccine composition |
CN111607002B (en) * | 2020-02-24 | 2021-07-09 | 中山大学 | Novel coronavirus S protein double-region subunit nano vaccine based on helicobacter pylori ferritin |
-
2020
- 2020-09-11 CN CN202010955986.3A patent/CN112048007B/en active Active
-
2021
- 2021-09-09 WO PCT/CN2021/117415 patent/WO2022052984A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111218458A (en) * | 2020-02-27 | 2020-06-02 | 珠海丽凡达生物技术有限公司 | mRNAs encoding SARS-CoV-2 virus antigen and vaccine and preparation method of vaccine |
CN113336832A (en) * | 2020-03-02 | 2021-09-03 | 成都威斯克生物医药有限公司 | Protein for resisting SARS-CoV-2 infection and vaccine containing the protein |
CN111607003A (en) * | 2020-05-21 | 2020-09-01 | 泰州市百英生物科技有限公司 | SARS-CoV-2N/S1(RBD) recombinant protein and its preparation method and application |
CN111732638A (en) * | 2020-07-02 | 2020-10-02 | 重庆博唯佰泰生物制药有限公司 | Vaccine against SARS-CoV-2 |
CN112048007A (en) * | 2020-09-11 | 2020-12-08 | 北京美康基免生物科技有限公司 | Universal novel coronavirus vaccine and preparation method thereof |
Non-Patent Citations (5)
Title |
---|
CHANG CHENG-WEI, LIU YUCHEN, JIAO CHENG, LIU HONGWEI, GONG JIE, CHEN XIAOCHUAN, CHANG LUNG-JI: "A Glimpse into the Diverse Cellular Immunity against SARS-CoV-2", VACCINES, vol. 9, no. 8, pages 1 - 13, XP055911856, DOI: 10.3390/vaccines9080827 * |
DATABASE PROTEIN 27 April 2020 (2020-04-27), ANONYMOUS: "SARS_CoV_2RBD_his [synthetic construct]", XP055862868, retrieved from GENBANK Database accession no. QJE37811 * |
GONG, GUO LI; WANG, LIANG; WU, ZHOU: "Research Progress of Novel Coronavirus SARS-CoV-2", JOURNAL OF CHONGQING UNIVERSITY OF TECHNOLOGY(NATURAL SCIENCE), vol. 34, no. 4, 30 April 2020 (2020-04-30), pages 42 - 50, XP009535560, ISSN: 1674-8425 * |
XU, ZHI-WIANG; LU, CHEN; HU, XIAO-JUN; GUO, YOU; HU, QIAO-LI: "Epitope-based peptide vaccine design and immunogenicity analysis of the spike protein from SARS-CoV-2", JOURNAL OF GANNAN MEDICAL UNIVERSITY, vol. 40, no. 3, 31 March 2020 (2020-03-31), CN , pages 217 - 224, XP009535561, ISSN: 1001-5779 * |
ZHANG,N.R.ET AL.: "Current development of COVID-19 diagnostics, vaccines and therapeutics", MICROBES AND INFECTION, vol. 22, 6 May 2020 (2020-05-06), XP086233891, DOI: 10.1016/j.micinf.2020.05.001 * |
Also Published As
Publication number | Publication date |
---|---|
CN112048007A (en) | 2020-12-08 |
CN112048007B (en) | 2022-07-12 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2022052984A1 (en) | Universal sars-cov-2 vaccine and preparation method thereof | |
US20240091339A1 (en) | Pre-immunization and immunotherapy | |
Gardner et al. | Infection of human dendritic cells by a sindbis virus replicon vector is determined by a single amino acid substitution in the E2 glycoprotein | |
CN111450244B (en) | Cell combination for preventing and treating coronavirus infection and application thereof | |
US9080152B2 (en) | Methods for proliferation of antigen-specific T cells | |
US20110136208A1 (en) | Antigen-specific regulatory t-cell induction | |
JP7268055B2 (en) | Human DC cell amplification method and human DC cell resource bank | |
EA010434B1 (en) | Process for producing cytotoxic lymphocyte | |
Chen et al. | Enhancement of CTLs induced by DCs loaded with ubiquitinated hepatitis B virus core antigen | |
JP2023516685A (en) | On-demand expression of exogenous factors in lymphocytes to treat HIV | |
Thompson et al. | Alphavirus replicon particles acting as adjuvants promote CD8+ T cell responses to co-delivered antigen | |
AU769538B2 (en) | Efficient ex vivo expansion of CD4+ and CD8+ T-cells from HIV infected subjects | |
Anticoli et al. | Engineered exosomes boost the HCV NS3-specific CD8+ T lymphocyte immunity in humans | |
Nazarkina et al. | Maturation and antigen loading protocols influence activity of anticancer dendritic cells | |
CN102234659B (en) | Plasmid for silencing cytokine signal inhibitory factor 1 and expressing high mobility group B1 protein and tumor associated antigen and preparation method thereof | |
CN115820742A (en) | Method for improving transduction efficiency of NK cell lentivirus | |
Shen et al. | Modulation of the immune system by Listeria monocytogenes‐mediated gene transfer into mammalian cells | |
Kos et al. | Specific epitope‐induced conversion of CD8+ memory cells into effector cytotoxic T lymphocytes in vitro: presentation of peptide antigen by CD8+ T cells | |
KR20210110940A (en) | Vaccine composition comprising enhanced avian influenza virus-like particles and method thereof | |
WO2018032619A1 (en) | Applications of soluble protein baff in b cell in-vitro culture and proliferation | |
CN117777314B (en) | Lentiviral vector and application thereof | |
CN111620955B (en) | Multi-target-site composite antigen and application thereof | |
EP2926831A1 (en) | CD8+ T-cell epitopes from polyomavirus capsid protein VP1 for vaccination | |
CN112779224A (en) | NK trophoblast cell expressing cytokine composition and preparation method and application thereof | |
Wade-Evans et al. | Specific proliferative T cell responses and antibodies elicited by vaccination with simian immunodeficiency virus Nef do not confer protection against virus challenge |
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: 21866036 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21866036 Country of ref document: EP Kind code of ref document: A1 |