CN118340876A - Varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof - Google Patents
Varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof Download PDFInfo
- Publication number
- CN118340876A CN118340876A CN202410403081.3A CN202410403081A CN118340876A CN 118340876 A CN118340876 A CN 118340876A CN 202410403081 A CN202410403081 A CN 202410403081A CN 118340876 A CN118340876 A CN 118340876A
- Authority
- CN
- China
- Prior art keywords
- lipid
- seq
- mrna
- pharmaceutical composition
- protein
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 239000008194 pharmaceutical composition Substances 0.000 title claims abstract description 24
- 241000701085 Human alphaherpesvirus 3 Species 0.000 title abstract description 37
- 108700021021 mRNA Vaccine Proteins 0.000 title abstract description 12
- 229940126582 mRNA vaccine Drugs 0.000 title abstract description 12
- 238000002360 preparation method Methods 0.000 claims abstract description 22
- 108020004999 messenger RNA Proteins 0.000 claims description 100
- 150000002632 lipids Chemical class 0.000 claims description 91
- 108090000623 proteins and genes Proteins 0.000 claims description 66
- 102000004169 proteins and genes Human genes 0.000 claims description 59
- 239000002105 nanoparticle Substances 0.000 claims description 52
- 150000001413 amino acids Chemical class 0.000 claims description 35
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 claims description 34
- 238000008416 Ferritin Methods 0.000 claims description 30
- 229960005486 vaccine Drugs 0.000 claims description 30
- 108050000784 Ferritin Proteins 0.000 claims description 28
- 102000008857 Ferritin Human genes 0.000 claims description 28
- 210000004027 cell Anatomy 0.000 claims description 22
- 238000000034 method Methods 0.000 claims description 21
- 239000000203 mixture Substances 0.000 claims description 21
- 230000007935 neutral effect Effects 0.000 claims description 18
- -1 cationic lipid Chemical class 0.000 claims description 17
- 235000012000 cholesterol Nutrition 0.000 claims description 17
- 230000028993 immune response Effects 0.000 claims description 14
- 239000002773 nucleotide Substances 0.000 claims description 14
- 125000003729 nucleotide group Chemical group 0.000 claims description 14
- 230000035772 mutation Effects 0.000 claims description 11
- 229920001223 polyethylene glycol Polymers 0.000 claims description 11
- 239000013598 vector Substances 0.000 claims description 11
- 108020003589 5' Untranslated Regions Proteins 0.000 claims description 9
- 108010076504 Protein Sorting Signals Proteins 0.000 claims description 9
- 238000009472 formulation Methods 0.000 claims description 9
- 229920000642 polymer Polymers 0.000 claims description 9
- NRJAVPSFFCBXDT-HUESYALOSA-N 1,2-distearoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCCCC NRJAVPSFFCBXDT-HUESYALOSA-N 0.000 claims description 8
- 125000002091 cationic group Chemical group 0.000 claims description 8
- 108020001507 fusion proteins Proteins 0.000 claims description 8
- 102000037865 fusion proteins Human genes 0.000 claims description 8
- KILNVBDSWZSGLL-KXQOOQHDSA-N 1,2-dihexadecanoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCC KILNVBDSWZSGLL-KXQOOQHDSA-N 0.000 claims description 6
- SNKAWJBJQDLSFF-NVKMUCNASA-N 1,2-dioleoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC SNKAWJBJQDLSFF-NVKMUCNASA-N 0.000 claims description 6
- 239000003814 drug Substances 0.000 claims description 6
- 230000002163 immunogen Effects 0.000 claims description 6
- 239000007908 nanoemulsion Substances 0.000 claims description 6
- KSXTUUUQYQYKCR-LQDDAWAPSA-M 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium;chloride Chemical compound [Cl-].CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KSXTUUUQYQYKCR-LQDDAWAPSA-M 0.000 claims description 5
- 108091026898 Leader sequence (mRNA) Proteins 0.000 claims description 5
- 230000001939 inductive effect Effects 0.000 claims description 5
- 238000004519 manufacturing process Methods 0.000 claims description 5
- 230000002438 mitochondrial effect Effects 0.000 claims description 5
- 241000710929 Alphavirus Species 0.000 claims description 4
- 108091034057 RNA (poly(A)) Proteins 0.000 claims description 4
- 108091036066 Three prime untranslated region Proteins 0.000 claims description 4
- 201000010099 disease Diseases 0.000 claims description 4
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 4
- QFMZQPDHXULLKC-UHFFFAOYSA-N 1,2-bis(diphenylphosphino)ethane Chemical compound C=1C=CC=CC=1P(C=1C=CC=CC=1)CCP(C=1C=CC=CC=1)C1=CC=CC=C1 QFMZQPDHXULLKC-UHFFFAOYSA-N 0.000 claims description 3
- LZLVZIFMYXDKCN-QJWFYWCHSA-N 1,2-di-O-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC LZLVZIFMYXDKCN-QJWFYWCHSA-N 0.000 claims description 3
- CITHEXJVPOWHKC-UUWRZZSWSA-N 1,2-di-O-myristoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCC CITHEXJVPOWHKC-UUWRZZSWSA-N 0.000 claims description 3
- FVXDQWZBHIXIEJ-LNDKUQBDSA-N 1,2-di-[(9Z,12Z)-octadecadienoyl]-sn-glycero-3-phosphocholine Chemical compound CCCCC\C=C/C\C=C/CCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC FVXDQWZBHIXIEJ-LNDKUQBDSA-N 0.000 claims description 3
- SLKDGVPOSSLUAI-PGUFJCEWSA-N 1,2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine zwitterion Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OCCN)OC(=O)CCCCCCCCCCCCCCC SLKDGVPOSSLUAI-PGUFJCEWSA-N 0.000 claims description 3
- YKIOPDIXYAUOFN-UHFFFAOYSA-N 2,3-di(icosanoyloxy)propyl 2-(trimethylazaniumyl)ethyl phosphate Chemical compound CCCCCCCCCCCCCCCCCCCC(=O)OCC(COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCCCCCC YKIOPDIXYAUOFN-UHFFFAOYSA-N 0.000 claims description 3
- NEZDNQCXEZDCBI-UHFFFAOYSA-N 2-azaniumylethyl 2,3-di(tetradecanoyloxy)propyl phosphate Chemical compound CCCCCCCCCCCCCC(=O)OCC(COP(O)(=O)OCCN)OC(=O)CCCCCCCCCCCCC NEZDNQCXEZDCBI-UHFFFAOYSA-N 0.000 claims description 3
- GUJDUXMBBAOBHN-UHFFFAOYSA-N 6-(dimethylamino)-n-(2h-tetrazol-5-yl)pyrazine-2-carboxamide Chemical compound CN(C)C1=CN=CC(C(=O)NC2=NNN=N2)=N1 GUJDUXMBBAOBHN-UHFFFAOYSA-N 0.000 claims description 3
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 claims description 3
- 102000014914 Carrier Proteins Human genes 0.000 claims description 3
- 108010078791 Carrier Proteins Proteins 0.000 claims description 3
- GZDFHIJNHHMENY-UHFFFAOYSA-N Dimethyl dicarbonate Chemical compound COC(=O)OC(=O)OC GZDFHIJNHHMENY-UHFFFAOYSA-N 0.000 claims description 3
- 101710162577 Fms-related tyrosine kinase 3 ligand protein Proteins 0.000 claims description 3
- JZNWSCPGTDBMEW-UHFFFAOYSA-N Glycerophosphorylethanolamin Natural products NCCOP(O)(=O)OCC(O)CO JZNWSCPGTDBMEW-UHFFFAOYSA-N 0.000 claims description 3
- 101000823778 Homo sapiens Y-box-binding protein 2 Proteins 0.000 claims description 3
- 102000007327 Protamines Human genes 0.000 claims description 3
- 108010007568 Protamines Proteins 0.000 claims description 3
- NYDLOCKCVISJKK-WRBBJXAJSA-N [3-(dimethylamino)-2-[(z)-octadec-9-enoyl]oxypropyl] (z)-octadec-9-enoate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(CN(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC NYDLOCKCVISJKK-WRBBJXAJSA-N 0.000 claims description 3
- 229960003724 dimyristoylphosphatidylcholine Drugs 0.000 claims description 3
- MWRBNPKJOOWZPW-CLFAGFIQSA-N dioleoyl phosphatidylethanolamine Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(COP(O)(=O)OCCN)OC(=O)CCCCCCC\C=C/CCCCCCCC MWRBNPKJOOWZPW-CLFAGFIQSA-N 0.000 claims description 3
- 210000001808 exosome Anatomy 0.000 claims description 3
- GLGLUQVVDHRLQK-WRBBJXAJSA-N n,n-dimethyl-2,3-bis[(z)-octadec-9-enoxy]propan-1-amine Chemical compound CCCCCCCC\C=C/CCCCCCCCOCC(CN(C)C)OCCCCCCCC\C=C/CCCCCCCC GLGLUQVVDHRLQK-WRBBJXAJSA-N 0.000 claims description 3
- 150000008104 phosphatidylethanolamines Chemical class 0.000 claims description 3
- 229940048914 protamine Drugs 0.000 claims description 3
- 102000002572 Alpha-Globulins Human genes 0.000 claims description 2
- 108010068307 Alpha-Globulins Proteins 0.000 claims description 2
- 108010038061 Chymotrypsinogen Proteins 0.000 claims description 2
- 101710157275 Ferritin subunit Proteins 0.000 claims description 2
- 102100020715 Fms-related tyrosine kinase 3 ligand protein Human genes 0.000 claims description 2
- 241000963438 Gaussia <copepod> Species 0.000 claims description 2
- 108010076685 HLA-B46 antigen Proteins 0.000 claims description 2
- 101710154606 Hemagglutinin Proteins 0.000 claims description 2
- 101000746367 Homo sapiens Granulocyte colony-stimulating factor Proteins 0.000 claims description 2
- 101000976075 Homo sapiens Insulin Proteins 0.000 claims description 2
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 claims description 2
- 102000008100 Human Serum Albumin Human genes 0.000 claims description 2
- 108091006905 Human Serum Albumin Proteins 0.000 claims description 2
- 241001529936 Murinae Species 0.000 claims description 2
- 101710093908 Outer capsid protein VP4 Proteins 0.000 claims description 2
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 claims description 2
- 101710176177 Protein A56 Proteins 0.000 claims description 2
- 229960000074 biopharmaceutical Drugs 0.000 claims description 2
- 229950001616 erenumab Drugs 0.000 claims description 2
- 230000004927 fusion Effects 0.000 claims description 2
- 239000000185 hemagglutinin Substances 0.000 claims description 2
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 claims description 2
- 239000010445 mica Substances 0.000 claims description 2
- 229910052618 mica group Inorganic materials 0.000 claims description 2
- 229960002621 pembrolizumab Drugs 0.000 claims description 2
- 230000002265 prevention Effects 0.000 claims description 2
- 229960002633 ramucirumab Drugs 0.000 claims description 2
- 241000712461 unidentified influenza virus Species 0.000 claims description 2
- 125000003275 alpha amino acid group Chemical group 0.000 claims 3
- WALUVDCNGPQPOD-UHFFFAOYSA-M 2,3-di(tetradecoxy)propyl-(2-hydroxyethyl)-dimethylazanium;bromide Chemical compound [Br-].CCCCCCCCCCCCCCOCC(C[N+](C)(C)CCO)OCCCCCCCCCCCCCC WALUVDCNGPQPOD-UHFFFAOYSA-M 0.000 claims 1
- 239000002202 Polyethylene glycol Substances 0.000 claims 1
- PSLWZOIUBRXAQW-UHFFFAOYSA-M dimethyl(dioctadecyl)azanium;bromide Chemical compound [Br-].CCCCCCCCCCCCCCCCCC[N+](C)(C)CCCCCCCCCCCCCCCCCC PSLWZOIUBRXAQW-UHFFFAOYSA-M 0.000 claims 1
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims 1
- NFQBIAXADRDUGK-KWXKLSQISA-N n,n-dimethyl-2,3-bis[(9z,12z)-octadeca-9,12-dienoxy]propan-1-amine Chemical compound CCCCC\C=C/C\C=C/CCCCCCCCOCC(CN(C)C)OCCCCCCCC\C=C/C\C=C/CCCCC NFQBIAXADRDUGK-KWXKLSQISA-N 0.000 claims 1
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 claims 1
- 230000005847 immunogenicity Effects 0.000 abstract description 10
- 230000008901 benefit Effects 0.000 abstract description 3
- 238000011161 development Methods 0.000 abstract description 3
- 230000001900 immune effect Effects 0.000 abstract description 2
- 238000009776 industrial production Methods 0.000 abstract description 2
- 235000018102 proteins Nutrition 0.000 description 45
- 238000001514 detection method Methods 0.000 description 33
- 239000000427 antigen Substances 0.000 description 25
- 102000036639 antigens Human genes 0.000 description 25
- 108091007433 antigens Proteins 0.000 description 25
- 229920002477 rna polymer Polymers 0.000 description 25
- 238000006467 substitution reaction Methods 0.000 description 17
- 235000001014 amino acid Nutrition 0.000 description 16
- 241000699670 Mus sp. Species 0.000 description 15
- 229940024606 amino acid Drugs 0.000 description 14
- 108090000765 processed proteins & peptides Proteins 0.000 description 14
- 150000007523 nucleic acids Chemical group 0.000 description 13
- 239000000243 solution Substances 0.000 description 13
- 241000699666 Mus <mouse, genus> Species 0.000 description 12
- 102000039446 nucleic acids Human genes 0.000 description 12
- 108020004707 nucleic acids Proteins 0.000 description 12
- 102000004127 Cytokines Human genes 0.000 description 11
- 108090000695 Cytokines Proteins 0.000 description 11
- 210000001744 T-lymphocyte Anatomy 0.000 description 11
- 238000002965 ELISA Methods 0.000 description 10
- 108010074328 Interferon-gamma Proteins 0.000 description 10
- 230000014509 gene expression Effects 0.000 description 10
- 244000052769 pathogen Species 0.000 description 10
- 239000000523 sample Substances 0.000 description 10
- 208000007514 Herpes zoster Diseases 0.000 description 9
- 108010002350 Interleukin-2 Proteins 0.000 description 9
- 102000000588 Interleukin-2 Human genes 0.000 description 9
- 230000007969 cellular immunity Effects 0.000 description 9
- 230000004044 response Effects 0.000 description 9
- 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 8
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 8
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 8
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 8
- 239000002245 particle Substances 0.000 description 8
- 108020004414 DNA Proteins 0.000 description 7
- 102000053602 DNA Human genes 0.000 description 7
- 102100037850 Interferon gamma Human genes 0.000 description 7
- 230000033289 adaptive immune response Effects 0.000 description 7
- 239000000872 buffer Substances 0.000 description 7
- 238000005538 encapsulation Methods 0.000 description 7
- 230000015788 innate immune response Effects 0.000 description 7
- 239000013612 plasmid Substances 0.000 description 7
- 102000004196 processed proteins & peptides Human genes 0.000 description 7
- 241001465754 Metazoa Species 0.000 description 6
- 108700026244 Open Reading Frames Proteins 0.000 description 6
- 210000005006 adaptive immune system Anatomy 0.000 description 6
- 239000008346 aqueous phase Substances 0.000 description 6
- 238000000338 in vitro Methods 0.000 description 6
- 239000003112 inhibitor Substances 0.000 description 6
- 230000003834 intracellular effect Effects 0.000 description 6
- 230000001717 pathogenic effect Effects 0.000 description 6
- 229920001184 polypeptide Polymers 0.000 description 6
- 238000000746 purification Methods 0.000 description 6
- 230000032258 transport Effects 0.000 description 6
- 108010067902 Peptide Library Proteins 0.000 description 5
- 238000003776 cleavage reaction Methods 0.000 description 5
- 150000001875 compounds Chemical class 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 230000003053 immunization Effects 0.000 description 5
- 238000002649 immunization Methods 0.000 description 5
- 230000008569 process Effects 0.000 description 5
- 230000007017 scission Effects 0.000 description 5
- 210000004989 spleen cell Anatomy 0.000 description 5
- 238000013518 transcription Methods 0.000 description 5
- 108091026890 Coding region Proteins 0.000 description 4
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 4
- 206010036376 Postherpetic Neuralgia Diseases 0.000 description 4
- 230000024932 T cell mediated immunity Effects 0.000 description 4
- 230000004721 adaptive immunity Effects 0.000 description 4
- 238000013461 design Methods 0.000 description 4
- 239000000975 dye Substances 0.000 description 4
- 230000000694 effects Effects 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 230000004727 humoral immunity Effects 0.000 description 4
- 210000000987 immune system Anatomy 0.000 description 4
- 208000015181 infectious disease Diseases 0.000 description 4
- 102000040430 polynucleotide Human genes 0.000 description 4
- 108091033319 polynucleotide Proteins 0.000 description 4
- 239000002157 polynucleotide Substances 0.000 description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 4
- 238000007789 sealing Methods 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 230000035897 transcription Effects 0.000 description 4
- 108020005345 3' Untranslated Regions Proteins 0.000 description 3
- 238000011740 C57BL/6 mouse Methods 0.000 description 3
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 3
- 239000004472 Lysine Substances 0.000 description 3
- 102000006833 Multifunctional Enzymes Human genes 0.000 description 3
- 108010047290 Multifunctional Enzymes Proteins 0.000 description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 description 3
- 229920004890 Triton X-100 Polymers 0.000 description 3
- 239000013504 Triton X-100 Substances 0.000 description 3
- 241000700605 Viruses Species 0.000 description 3
- 150000003838 adenosines Chemical class 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 3
- 230000028996 humoral immune response Effects 0.000 description 3
- 210000005007 innate immune system Anatomy 0.000 description 3
- 235000018977 lysine Nutrition 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 238000002156 mixing Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 230000028327 secretion Effects 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 230000004936 stimulating effect Effects 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- QGWBEETXHOVFQS-UHFFFAOYSA-N 6-[6-(2-hexyldecanoyloxy)hexyl-(4-hydroxybutyl)amino]hexyl 2-hexyldecanoate Chemical compound CCCCCCCCC(CCCCCC)C(=O)OCCCCCCN(CCCCO)CCCCCCOC(=O)C(CCCCCC)CCCCCCCC QGWBEETXHOVFQS-UHFFFAOYSA-N 0.000 description 2
- 239000004475 Arginine Substances 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 108091093094 Glycol nucleic acid Proteins 0.000 description 2
- 241000700586 Herpesviridae Species 0.000 description 2
- 108091092195 Intron Proteins 0.000 description 2
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 2
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 2
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 2
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 2
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 2
- 108091027974 Mature messenger RNA Proteins 0.000 description 2
- 208000002193 Pain Diseases 0.000 description 2
- 108091093037 Peptide nucleic acid Proteins 0.000 description 2
- 241000710961 Semliki Forest virus Species 0.000 description 2
- 108091046915 Threose nucleic acid Proteins 0.000 description 2
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 2
- 241000710959 Venezuelan equine encephalitis virus Species 0.000 description 2
- 230000002378 acidificating effect Effects 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 239000012736 aqueous medium Substances 0.000 description 2
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 238000001976 enzyme digestion Methods 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 238000007306 functionalization reaction Methods 0.000 description 2
- 235000013922 glutamic acid Nutrition 0.000 description 2
- 239000004220 glutamic acid Substances 0.000 description 2
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 2
- 235000004554 glutamine Nutrition 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 229960000310 isoleucine Drugs 0.000 description 2
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 2
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 210000000822 natural killer cell Anatomy 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 239000012074 organic phase Substances 0.000 description 2
- 210000000278 spinal cord Anatomy 0.000 description 2
- 210000004988 splenocyte Anatomy 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 239000011550 stock solution Substances 0.000 description 2
- 229960000814 tetanus toxoid Drugs 0.000 description 2
- 239000004474 valine Substances 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 1
- 108010071134 CRM197 (non-toxic variant of diphtheria toxin) Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 102000004533 Endonucleases Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 239000005977 Ethylene Substances 0.000 description 1
- 108060002716 Exonuclease Proteins 0.000 description 1
- 101000597577 Gluconacetobacter diazotrophicus (strain ATCC 49037 / DSM 5601 / CCUG 37298 / CIP 103539 / LMG 7603 / PAl5) Outer membrane protein Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000893103 Homo sapiens Ferritin, mitochondrial Proteins 0.000 description 1
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 1
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 108700028075 Human Herpesvirus 3 gp 118 Proteins 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 241000588650 Neisseria meningitidis Species 0.000 description 1
- 239000012124 Opti-MEM Substances 0.000 description 1
- 101710176384 Peptide 1 Proteins 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 229940096437 Protein S Drugs 0.000 description 1
- 208000003251 Pruritus Diseases 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 101710198474 Spike protein Proteins 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 101710172711 Structural protein Proteins 0.000 description 1
- 108091027544 Subgenomic mRNA Proteins 0.000 description 1
- 108700009124 Transcription Initiation Site Proteins 0.000 description 1
- 108091023045 Untranslated Region Proteins 0.000 description 1
- 101900123149 Varicella-zoster virus Envelope glycoprotein E Proteins 0.000 description 1
- 241000701067 Varicellovirus Species 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 108010087302 Viral Structural Proteins Proteins 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000009824 affinity maturation Effects 0.000 description 1
- 238000000246 agarose gel electrophoresis Methods 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- HMFHBZSHGGEWLO-NEEWWZBLSA-N alpha-L-ribose Chemical compound OC[C@@H]1O[C@@H](O)[C@@H](O)[C@H]1O HMFHBZSHGGEWLO-NEEWWZBLSA-N 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- HMFHBZSHGGEWLO-TXICZTDVSA-N beta-D-ribose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-TXICZTDVSA-N 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 229940017687 beta-d-ribose Drugs 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 230000007910 cell fusion Effects 0.000 description 1
- 239000002458 cell surface marker Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000012501 chromatography medium Substances 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 230000035071 co-translational protein modification Effects 0.000 description 1
- 230000024203 complement activation Effects 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 239000012470 diluted sample Substances 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 229960003983 diphtheria toxoid Drugs 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 102000013165 exonuclease Human genes 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 230000012178 germinal center formation Effects 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 102000016932 human mitochondrial ferritin Human genes 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 230000006054 immunological memory Effects 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000004255 ion exchange chromatography Methods 0.000 description 1
- 230000007803 itching Effects 0.000 description 1
- 230000002045 lasting effect Effects 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000015654 memory Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 229910000402 monopotassium phosphate Inorganic materials 0.000 description 1
- 235000019796 monopotassium phosphate Nutrition 0.000 description 1
- 230000002276 neurotropic effect Effects 0.000 description 1
- 238000006386 neutralization reaction Methods 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 230000014207 opsonization Effects 0.000 description 1
- 102000007863 pattern recognition receptors Human genes 0.000 description 1
- 108010089193 pattern recognition receptors Proteins 0.000 description 1
- 230000008823 permeabilization Effects 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- PJNZPQUBCPKICU-UHFFFAOYSA-N phosphoric acid;potassium Chemical compound [K].OP(O)(O)=O PJNZPQUBCPKICU-UHFFFAOYSA-N 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 210000000413 sensory ganglia Anatomy 0.000 description 1
- 239000012089 stop solution Substances 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 229960001005 tuberculin Drugs 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 238000000108 ultra-filtration Methods 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
Landscapes
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The invention relates to a varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof. Specifically, compared with the prior art, the preparation method has the advantages of good immunogenicity, strong immune effect, simple preparation process, suitability for clinical development and industrial production, and higher industrialization value.
Description
Technical Field
The invention relates to the technical field of biology, in particular to a preparation method and application of a self-replicating varicella-zoster virus (VZV) mRNA vaccine preparation.
Background
Varicella-zoster virus (VZV, also known as human herpesvirus type 3) is a double stranded DNA virus with neurotropic and lymphotropic properties. Most children before age 10 will come into contact with VZV, causing fever and varicella. VZV infection is followed by latency in the dorsal sensory ganglion of the spinal cord. When immunity declines, the reactivated VZV in the elderly migrates to the skin area associated with the affected spinal cord area, causing shingles. Typical areas of skin for acute shingles include clustered blisters, causing severe pain and itching. The incidence of shingles is about 0.4% in the 50-60 year old population, while it increases significantly to 1% in the over 80 year old population. Shingles is often associated with chronic, debilitating and pain lasting for several months, known as Post Herpetic Neuralgia (PHN), especially in the elderly. About 15% of cases of post-severe PHN postherpetic neuralgia persist for at least 3 months and are not effectively alleviated by antiviral drugs.
The advantages of efficient utilization of mRNA, rapid production and development, safe administration and the like are widely paid attention to. Self-replicating mRNA (saRNA) vaccines are genetically engineered replicons derived from self-replicating single-stranded RNA viruses, and the sense alphavirus genomes commonly used in saRNA vaccine design include venezuelan equine encephalitis Virus (VEE), SINV, and Semliki Forest Virus (SFV). In the case of the alphavirus-based self-amplifying mRNA, the additional RNA contains four non-structural protein coding regions (nsP 1-4), a subgenomic promoter and a large open reading frame, and the gene encoding the viral structural protein in the viral genome is replaced by the gene of interest, so that the mRNA is not capable of producing infectious virus. The expression of conventional mRNA vaccine antigens is often proportional to the copy number of mRNA delivered into a body, so that large dosage and multiple immunity are needed, side effects are large, the problem is solved by self-replication of the saRNA, and compared with the conventional mRNA vaccine, the saRNA vaccine antigens can be delivered at a lower dosage, and the same immune protection is realized. Combining the VZV immunoprotection antigen with the self-replicating RNA is expected to be a powerful tool for preventing the VZV at low cost, quickly and efficiently.
Therefore, there is a need to develop self-replicating mRNA vaccines against VZV that are simple in process and more immunogenic.
Disclosure of Invention
It is a first object of the present invention to provide a Varicella Zoster Virus (VZV) vaccine. More specifically, an mRNA vaccine of varicella-zoster virus (VZV) is provided that can elicit an immune response against VZV and also can produce antibodies against VZV.
A second object of the present invention is to provide mRNA constructs comprised in the above vaccine, and corresponding vectors, lipid Nanoparticles (LNP).
The third object of the present invention is to provide the mRNA construct, and the corresponding vector, pharmaceutical use.
A fourth object of the present invention is to provide a method of inducing an immune response in a subject in need thereof or a method of preventing varicella-zoster in a subject in need thereof based on the vaccine described above.
In order to solve the technical problems, the invention comprises the following technical scheme:
In one aspect, the invention provides a vaccine that is a VZV mRNA vaccine comprising a VZV mRNA construct as an immunogenic component. Wherein the mRNA construct comprises mRNA encoding the gE and/or gH, gL regions of VZV.
In some specific embodiments, the mRNA encoding the gE and/or gH, gL regions of VZV is expressed alone or in fusion within the host cell.
In some specific embodiments, the mRNA encodes a protein that further comprises a mutation.
In some specific embodiments, the mRNA encodes a protein that further comprises Flt3L.
In some specific embodiments, the mRNA encodes a protein that further comprises a signal peptide sequence.
Optionally, the signal peptide is located at the N-terminus of the protein.
Preferably, the signal peptide is derived from murine H-2Kb, human IgE, HLA-B46, MICA 008, OSM, VSV-G, mouse Ig Kappa, mouse heavy chain, BM40, human chymotrypsinogen, human prothrombin-2, human IL-2, human G-CSF, human hemagglutinin IX, human albumin, gaussia luc, HAS, influenza virus, human insulin, silk LC, erenumab antibody light chain, pembrolizumab light chain, ramucirumab light chain, E SIGNAL PEPTIDE, SP1 (LZJ human IgG1, SP2, SP3 (ZLQ).
In some specific embodiments, the gE extracellular region (gE Ecto) protein binds to ferritin (Ferritin) to form a fusion protein.
The gE Ecto-ferritin fusion proteins of the invention comprise the unit subunits of ferritin which can be assembled extracellularly into nanoparticles, with 24 monomers constituting 20 panels to fully display the immunogenic portion of gE Ecto.
The ferritin subunit of the invention is a full length or any portion of ferritin, wild type or partial amino acid mutation. In a specific embodiment, the monomeric subunit is from mitochondrial ferritin or heavy chain ferritin.
In some specific embodiments, the mRNA encodes a protein comprising an amino acid sequence that is at least 90% identical to the amino acid sequence set forth in SEQ ID NO. 3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID NO. 13.
In some specific embodiments, the mRNA encodes a protein comprising an amino acid sequence that is at least 95% identical to the amino acid sequence set forth in SEQ ID NO. 3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID NO. 13.
In some specific embodiments, the mRNA encodes a protein comprising an amino acid sequence that is at least 99% identical to the amino acid sequence set forth in SEQ ID NO. 3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID NO. 13.
In a preferred embodiment, the mRNA encodes a protein comprising the amino acid sequence set forth in SEQ ID NO. 3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID NO. 13.
In a preferred embodiment, the mRNA comprises the coding nucleotide corresponding to the amino acid sequence shown as SEQ ID NO.3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID NO. 13.
In a more preferred embodiment, the mRNA comprises the nucleotide sequence set forth in SEQ ID NO. 2,SEQ ID NO:4,SEQ ID NO:6,SEQ ID NO:8,SEQ ID NO:10 or SEQ ID NO. 12.
In some specific embodiments, the mRNA construct further comprises a 5' utr sequence.
Optionally, the 5' UTR sequence is a human alpha globulin 5' UTR or a non-native 5' UTR sequence.
In some specific embodiments, the mRNA construct further comprises a 3' utr sequence.
In some specific embodiments, the mRNA construct further comprises a Poly (a) sequence.
In some specific embodiments, the mRNA construct further comprises an alphavirus nonstructural protein (nsp) gene.
Optionally, the nsp gene is shown as SEQ ID NO. 1.
In a specific embodiment, the vaccine further comprises a delivery formulation.
Preferably, the delivery formulation is a nanoparticle.
Preferably, the delivery formulation comprises lipid nanoparticles (Lipid nanoparticle, LNP), lipid multipolymers (lipopolyplex, LPP), polymer nanoparticles (Polymer nanoparticles, PNP), inorganic nanoparticles (Inorganic nanoparticles, INP), cationic nanoemulsions (Cationic nanoemulsion, CNE), exosomes, biologicals, and protamine, etc.
More preferably, the nanoparticle is a lipid nanoparticle (Lipid nanoparticle, LNP).
In some specific embodiments, the lipid comprises one or more of a cationic lipid, a neutral helper lipid, cholesterol, and a PEG-modified lipid; preferably, two or more are included.
In some specific embodiments, the lipid comprises:
a) Cationic lipids;
b) Neutral helper lipids;
c) Cholesterol; and
D) PEG modified lipids.
In some embodiments, the lipid nanoparticle has a molar ratio of each lipid component, based on 100% total molar amount of lipid, of:
a) 45% -50% of cationic lipid;
b) Neutral auxiliary lipid 5-10%;
c) 38% -48% of cholesterol;
and d) PEG modified lipid 0-3%.
In some specific embodiments, the cationic lipid is selected from the group consisting of N, N-dimethyl-2, 3-dioleoyloxypropylamine (DODMA), 1, 2-dimyristoxypropyl-3-dimethyl-hydroxyethylammonium bromide (dmrii), N-dioleoyln, N-dimethylammonium chloride (DODAC), 1, 2-dioleoyl-3-dimethylammonium-propane (DODAP), N-distearyl-N, N-dimethylammonium bromide (DDAB), N- (l- (2, 3-dioleoyloxy) propyl) -N, N-trimethylammonium chloride (DOTAP), N- (1- (2, 3-dioleoyloxy) propyl) -N, N-trimethylammonium chloride (DOTMA), 1, 2-dimethanoyloxy-N, N-dimethylaminopropane (DLin), 2-dioleylene-4- (2-dimethylaminoethyl) - [1,3] -dioxolane (DLin), N- (2, 3-dioleyloxy) propyl) -N, N-trimethylammonium chloride (dlma), 1- (2, 3-dioleyloxy) 2-dlm-4-dlm, dlm-2-di-N-butanoic acid (dlm), and any of more than one of these.
In some specific embodiments, the molar ratio of the cationic lipid is 45% to 48% based on 100% total molar amount of lipid.
In some specific embodiments, the neutral helper lipid is selected from any one or more of distearoylphosphatidylcholine(DSPC)、dioleoylphosphatidylcholine(DOPC)、dimyristoylphosphatidylcholine(DMPC)、dipalmitoylphosphatidylcholine(DPPC)、diarachidoylphosphatidylcholine(DAPC)、dibehenoylphosphatidylcholine(DBPC)、ditricosanoylphosphatidylcholine(DTPC)、dilignoceroylphatidylcholine(DLPC)、dioleoylphosphatidylethanolamine(DOPE)、dipalmitoyl-phosphatidylethanolamine(DPPE)、dimyristoyl-phosphatidylethanolamine(DMPE) or phosphatidylethanolamine (DLPE).
In some embodiments, the neutral helper lipid is present in a molar ratio of 6% to 10% based on 100% total molar amount of lipid.
In some embodiments, the neutral helper lipid is present in a molar ratio of 8% to 10% based on 100% total molar amount of lipid.
In some embodiments, the PEG-modified lipid is selected from any one or more of methoxypolyethylene glycol bitetradecylamide (ALC-0159), DMG-PEG2000, DMG-PEG5000, PEG 2000.
In some embodiments, the PEG-modified lipids are present in a molar ratio of 1% to 3% based on 100% total molar amount of lipids.
In some embodiments, the PEG-modified lipids are present in a molar ratio of 1% to 2% based on 100% total molar amount of lipids.
In some embodiments, the cholesterol is in a molar ratio of 40% to 46% based on 100% total molar amount of lipid.
In some embodiments, the cholesterol is in a molar ratio of 42% to 45% based on 100% total molar amount of lipid.
In another aspect, the invention provides an mRNA construct that is any one of the mRNA constructs described herein as an immunogenic component in the vaccine, or a combination thereof.
In another aspect, the invention provides a vector comprising any one of the mRNA constructs described herein, or a combination thereof.
In another aspect, the invention provides a cell comprising any one of the mRNA constructs described herein, or a combination thereof, or a vector described herein.
In another aspect, the invention provides a nanoparticle comprising any one of the mRNA constructs described herein, or a combination thereof.
Optionally, the nanoparticles include lipid nanoparticles (Lipid nanoparticle, LNP), lipid multipolymers (lipopolyplex, LPP), polymer nanoparticles (Polymer nanoparticles, PNP), inorganic nanoparticles (Inorganic nanoparticles, INP), cationic nanoemulsions (Cationic nanoemulsion, CNE), exosomes, biological microvesicles, protamine, and the like.
Preferably, the nanoparticle is a lipid nanoparticle (Lipid nanoparticle, LNP).
In some specific embodiments, the lipid comprises one or more of a cationic lipid, a neutral helper lipid, cholesterol, and a PEG-modified lipid; preferably, two or more are included.
In some specific embodiments, the lipid comprises:
a) Cationic lipids;
b) Neutral helper lipids;
c) Cholesterol; and
D) PEG modified lipids.
In some embodiments, the lipid nanoparticle has a molar ratio of each lipid component, based on 100% total molar amount of lipid, of:
a) 45% -50% of cationic lipid;
b) Neutral auxiliary lipid 5-10%;
c) 38% -48% of cholesterol;
and d) PEG modified lipid 0-3%.
In some specific embodiments, the cationic lipid is selected from the group consisting of N, N-dimethyl-2, 3-dioleoyloxypropylamine (DODMA), 1, 2-dimyristoxypropyl-3-dimethyl-hydroxyethylammonium bromide (dmrii), N-dioleoyln, N-dimethylammonium chloride (DODAC), 1, 2-dioleoyl-3-dimethylammonium-propane (DODAP), N-distearyl-N, N-dimethylammonium bromide (DDAB), N- (l- (2, 3-dioleoyloxy) propyl) -N, N-trimethylammonium chloride (DOTAP), N- (1- (2, 3-dioleoyloxy) propyl) -N, N-trimethylammonium chloride (DOTMA), 1, 2-dimethanoyloxy-N, N-dimethylaminopropane (DLin), 2-dioleylene-4- (2-dimethylaminoethyl) - [1,3] -dioxolane (DLin), N- (2, 3-dioleyloxy) propyl) -N, N-trimethylammonium chloride (dlma), 1- (2, 3-dioleyloxy) 2-dlm-4-dlm, dlm-2-di-N-butanoic acid (dlm), and any of more than one of these.
In some specific embodiments, the molar ratio of the cationic lipid is 45% to 48% based on 100% total molar amount of lipid.
In some specific embodiments, the neutral helper lipid is selected from any one or more of distearoylphosphatidylcholine(DSPC)、dioleoylphosphatidylcholine(DOPC)、dimyristoylphosphatidylcholine(DMPC)、dipalmitoylphosphatidylcholine(DPPC)、diarachidoylphosphatidylcholine(DAPC)、dibehenoylphosphatidylcholine(DBPC)、ditricosanoylphosphatidylcholine(DTPC)、dilignoceroylphatidylcholine(DLPC)、dioleoylphosphatidylethanolamine(DOPE)、dipalmitoyl-phosphatidylethanolamine(DPPE)、dimyristoyl-phosphatidylethanolamine(DMPE) or phosphatidylethanolamine (DLPE).
In some embodiments, the neutral helper lipid is present in a molar ratio of 6% to 10% based on 100% total molar amount of lipid.
In some embodiments, the neutral helper lipid is present in a molar ratio of 8% to 10% based on 100% total molar amount of lipid.
In some embodiments, the PEG-modified lipid is selected from any one or more of methoxypolyethylene glycol bitetradecylamide (ALC-0159), DMG-PEG2000, DMG-PEG5000, PEG 2000.
In some embodiments, the PEG-modified lipids are present in a molar ratio of 1% to 3% based on 100% total molar amount of lipids.
In some embodiments, the PEG-modified lipids are present in a molar ratio of 1% to 2% based on 100% total molar amount of lipids.
In some embodiments, the cholesterol is in a molar ratio of 40% to 46% based on 100% total molar amount of lipid.
In some embodiments, the cholesterol is in a molar ratio of 42% to 45% based on 100% total molar amount of lipid.
In another aspect, the invention provides a fusion protein comprising a protein encoded by any one of the mRNAs described herein and a second protein fused thereto, wherein said protein encoded by the mRNAs is an unfused protein.
In some specific embodiments, the second protein is a carrier protein.
In some specific embodiments, the second protein is ferritin (Ferritin).
Preferably, the ferritin is mitochondrial ferritin or heavy chain ferritin.
In some specific embodiments, the ferritin comprises a domain that allows the fusion protein to self-assemble into a nanoparticle.
In another aspect, the invention provides the use of any of the vaccines, mRNA constructs, vectors, or nanoparticles described herein in the manufacture of a medicament for inducing an immune response in a subject in need thereof.
In another aspect, the invention provides the use of any of the vaccines, mRNA constructs, vectors, or nanoparticles described herein in the manufacture of a medicament for treating or preventing varicella-zoster in a subject in need thereof.
In another aspect, the invention provides a method of inducing an immune response in a subject in need thereof, comprising administering to the subject any of the vaccines described herein;
preferably, the method induces an immune response against VZV by using self-replicating mRNA.
In another aspect, the invention provides a method of treating or preventing varicella-zoster in a subject in need thereof comprising administering to the subject any of the vaccines described herein;
preferably, the method induces an immune response against VZV by using self-replicating mRNA.
In some specific embodiments, the vaccine composition is administered as part of a therapeutic regimen.
Advantageous effects
Compared with the prior art, the technical scheme of the invention has at least one of the following beneficial effects:
1. The VZV mRNA vaccine has better immunogenicity, can effectively stimulate organisms to generate high-level immune response, and can induce high-level specific humoral immune response and cell immunity.
2. The invention provides a self-replicating VZV mRNA vaccine, which can express gH and gL proteins besides gE protein, and comprehensively verify the effect from the angles of cells and animal models, and the result proves that the VZV vaccine has stronger immune effect.
3. The mRNA vaccine provided by the invention has a simple preparation process, is more suitable for clinical development and industrial production, and has higher industrialization value.
4. The LNP delivery formulation formulations provided in the present invention were tested to exhibit excellent performance in a variety of ways.
Definition and description of terms
Unless defined otherwise herein, scientific and technical terms used in connection with the present invention shall have the meaning as understood by one of ordinary skill in the art.
Furthermore, unless otherwise indicated herein, terms in the singular herein shall include the plural and terms in the plural shall include the singular. More specifically, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise.
The terms "comprising," "including," and "having" are used interchangeably herein to mean that the elements are included in an arrangement, meaning that the arrangement may exist in addition to the elements listed. It should also be understood that the use of "including," "comprising," and "having" descriptions herein also provides a "consisting of … …" scheme.
The term "and/or" as used herein includes the meaning of "and", "or" and "all or any other combination of the elements linked by the term of interest".
The term "and/or" as used herein includes the meaning of "and", "or" and "all or any other combination of the elements linked by the term of interest".
The term "gene" as used herein refers to a nucleic acid fragment encoding a protein or RNA alone (also referred to as a "coding sequence" or "coding region") and associated regulatory regions such as promoters, operators, terminators, etc., which may be located upstream or downstream of the coding sequence.
The term "nucleic acid" herein in its broadest sense includes any compound and/or substance comprising a polymer of nucleotides. These polymers are referred to as polynucleotides.
The nucleic acid (also referred to as a polynucleotide) may be or may include, for example, ribonucleic acid (RNA), deoxyribonucleic acid (DNA), threose Nucleic Acid (TNA), glycol Nucleic Acid (GNA), peptide Nucleic Acid (PNA), locked nucleic acid (LNA, including LNA having a βd ribose configuration, αlna having an αl ribose configuration (a diastereomer of LNA), 2 'amino LNA having 2' amino functionalization, and 2 'amino αlna having 2' amino functionalization), ethylene Nucleic Acid (ENA), cyclohexenyl nucleic acid (CeNA), or a chimeric or combination thereof.
The term "mRNA" herein is messenger RNA, meaning any polynucleotide that encodes (at least one) polypeptide (naturally occurring, non-naturally occurring or modified amino acid polymer) and can be translated in vitro, in vivo, in situ, or ex vivo to produce the encoded polypeptide.
The basic components of an mRNA molecule typically include at least one coding region, a 5' untranslated region (UTR), a 3' UTR, a 5' cap, and a poly a tail. Polynucleotides of the present disclosure may serve as mrnas, but may differ from wild-type mrnas in their functional and/or structural design features that are used to overcome the existing problems of efficient polypeptide expression using nucleic acid-based therapeutics.
The mRNA as used herein refers to mRNA comprising a nucleic acid sequence encoding a VZV antigen, which may be 1) mRNA encoding and translating only a certain VZV antigen, or 2) a mixture of mRNA encoding and translating a plurality of VZV antigens, or 3) a mixture of 1) 2) mRNA encoding other non-VZV antigens.
The term 5' UTR (5 ' -untranslated region) herein refers to a specific portion of a messenger RNA (mRNA) that is located 5' of the open reading frame of the mRNA. Typically, the 5' UTR begins at the transcription initiation site and ends one nucleotide before the initiation codon of the open reading frame. The 5' utr may include elements for controlling gene expression, also known as regulatory elements. The regulatory element may be, for example, a ribosome binding site or a 5' -terminal oligopyrimidine sequence. The 5'UTR may be post-transcriptionally modified, for example by the addition of 5' CAP.
The term 3'UTR (3' -untranslated region) herein is the portion of an mRNA that is located between the protein coding region (i.e., the open reading frame) and the poly (A) sequence of the mRNA. The 3' UTR of mRNA is not translated into amino acid sequences. The 3' utr sequence is typically encoded by a gene that: the gene is transcribed into the corresponding mRNA during gene expression. The genomic sequence is first transcribed into mature pre-mRNA, which includes optional introns. The pre-mature mRNA is then further processed into mature mRNA during the maturation process. The maturation process comprises the following steps: 5' capping, splicing of the mature pre-mRNA to cleave off optional introns, and modification of the 3' end, such as polyadenylation of the 3' end of the mature pre-mRNA and optional endo-or exonuclease cleavage, etc.
The term "Poly (a)" herein refers to a stretch of (long) adenosine nucleotides added at the 3' end of an RNA of up to about 400 adenosine nucleotides, for example, about 25 to about 400, preferably about 50 to about 400, more preferably about 50 to about 300, even more preferably about 50 to about 250, most preferably about 60 to about 250 adenosine nucleotides.
The term "mutation" herein includes genetic mutations and amino acid mutations, wherein a genetic mutation refers to a deletion, insertion, inversion or substitution of a heterologous nucleic acid, which may result in an alteration of the amino acid sequence in the corresponding protein product; amino acid mutations are also known as nonsensical single nucleotide mutations, because of the change in the amino acid sequence in the protein product due to some single base changes. Amino acid changes affect protein stability, interactions and enzyme activity, resulting in disease.
The terms "protein," "polypeptide," and "peptide" are used interchangeably herein and refer to a peptide-bond-linked chain of any amino acid, whether length or co-translational or post-translational modification. Such definition of a protein polypeptide or protein not encoded on a nucleic acid construct, wherein in particular and additionally such chains are included: the chain comprises one or more unnatural amino acid or amino acid-like structural units.
The term "amino acid substitution" herein refers to those in which at least one amino acid residue in the natural or starting sequence is removed and a different amino acid is inserted into its place at the same position. Substitutions may be single, wherein only one amino acid in the molecule has been substituted, or they may be multiple, wherein two or more amino acids in the same molecule have been substituted.
The term "conservative amino acid substitution" herein refers to the substitution of an amino acid that is normally present in a sequence with a different amino acid of similar size, charge, or polarity. Examples of conservative substitutions include the substitution of a nonpolar (hydrophobic) residue such as isoleucine, valine and leucine for another nonpolar residue. Likewise, examples of conservative substitutions include the substitution of one polar (hydrophilic) residue for another, such as between arginine and lysine, between glutamine and asparagine, and between glycine and serine. In addition, substitution of a basic residue such as lysine, arginine, or histidine for another residue, or substitution of one acidic residue such as aspartic acid or glutamic acid for another acidic residue, is an additional example of a conservative substitution. Examples of non-conservative substitutions include the substitution of a non-polar (hydrophobic) amino acid residue such as isoleucine, valine, leucine, alanine, methionine for a polar (hydrophilic) residue such as cysteine, glutamine, glutamic acid or lysine, and/or the substitution of a polar residue for a non-polar residue.
The term "mutant" herein refers to a "variant" of the protein or peptide that may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% amino acid identity to the amino acid sequence of the protein or peptide.
The term "vector" as used herein refers to a DNA molecule comprising single-stranded, double-stranded, circular or supercoiled DNA. Suitable vectors include retroviruses, adenoviruses, adeno-associated viruses, poxviruses and bacterial plasmids.
The term "antigen" herein refers to a substance that can be recognized by the immune system and is capable of triggering an antigen-specific immune response. Such a response may include the formation of antibodies specific for the antigen, or the activation of T cells specific for the antigen, as part of an adaptive immune response. Typically, the antigen may be or may comprise a peptide or protein that may be presented to T cells by MHC. An antigen in the sense of the present invention may be a translation product of a provided nucleic acid molecule, preferably an mRNA as defined herein. Fragments, variants and derivatives of peptides and proteins comprising at least one epitope are also understood in this context as antigens. The term "carrier protein" as used herein refers to proteins that are non-toxic to humans, do not cause allergic reactions and enhance vaccine immune efficacy, including ferritin (Ferritin), diphtheria toxoid (DT, DT CRM 197) and Tetanus Toxoid (TT), keyhole Limpet Hemocyanin (KLH), OMPC from Neisseria meningitidis (N.menningitidis), pure protein derivatives of tuberculin (PPD), BSA and/or OVA, and the like.
The term "vaccine" herein refers to a prophylactic or therapeutic substance that provides at least one antigen or antigen function, which is capable of stimulating an immune response in the body without causing a disease. The antigen or antigen function may stimulate the adaptive immune system of the body to provide an adaptive immune response.
The terms "nanoparticle" and "nanoparticle" are used interchangeably herein.
The term "immune system" herein may protect an organism from infection. If a pathogen breaks through the physical barrier of an organism and enters the organism, the innate immune system provides an immediate but non-specific response. If the pathogen bypasses the innate response, the vertebrate has a second layer of protection, the adaptive immune system. Here, the immune system alters its response during infection to improve its recognition of pathogens. This improved response is then retained in the form of an immunological memory after the pathogen is eliminated, and allows the adaptive immune system to establish a faster and stronger attack each time the pathogen is encountered. Thus, it can be seen that the immune system comprises two main forms: innate immunity and adaptive immunity. Innate immunity recognizes pathogens through non-specific pattern recognition receptors, involving a variety of humoral and cytokines. Adaptive immunity, in turn, distinguishes pathogens through specific antigen recognition, relying on humoral immunity and cell-mediated immune responses.
The term "immune response" herein may typically be a specific response of the adaptive immune system against a specific antigen (so-called specific or adaptive immune response) or a non-specific response of the innate immune system (so-called non-specific or innate immune response). One basis of the present invention relates to the specific response of the adaptive immune system (adaptive immune response); in particular an adaptive immune response following exposure to an antigen (such as an immunogenic polypeptide). However, this specific response may be supported by an additional non-specific response (innate immune response). Thus, one basis of the present invention also relates to compounds for stimulating both the innate and adaptive immune systems simultaneously to elicit an effective adaptive immune response. In the context of the present invention, "antigen composition" refers to a compound or mixture of compounds (such as in a solution or pharmaceutical formulation): which is capable of, is used for, is capable of, or in practice can elicit, augment, generate, or elicit an immune response (preferably, an effective adaptive immune response) upon administration to or otherwise exposure to a subject.
The term "cellular immunity/cellular immune response" herein refers to an immune response that involves primarily macrophages, natural killer cells, antigen-specific T cells, and various immune cytokines. Cellular immunity does not utilize antibodies, but rather works by activating immune cells. For example, it can activate antigen-specific cytotoxic T lymphocytes, which are capable of recognizing and killing cells infected with viruses or bacteria, as well as tumor cells in vivo; it can also activate macrophages and natural killer cells to clear and kill pathogens; in addition, cellular immunity can also stimulate immune cells to secrete a variety of cytokines that can regulate the function of adaptive immunity, innate immunity, and other cells.
The term "humoral immunity/humoral immune response" herein typically refers to antibody production and possibly the ancillary processes that accompany it. For example, humoral immune responses may typically be characterized by Th2 activation and cytokine production, germinal center formation and allotypic switching, affinity maturation and memory cell production. Humoral immunity also typically may refer to effector functions of antibodies, including pathogen and toxin neutralization, classical complement activation and phagocytosis, and opsonization of pathogen elimination.
The term VZV herein, i.e., varicella-zoster virus, varicella-zoster virus, belongs to the family Herpesviridae (Herpesviridae), the subfamily alpha-Herpesviridae (alpha-hermesviridae), the genus varicella (Varicellovirus).
The term "internal aqueous phase" as used herein means that during the preparation of the lipid nanoparticle, the nucleic acid is dissolved in an aqueous medium, i.e. the internal aqueous phase, prior to mixing the nucleic acid with the lipid component to form the nanoparticle.
The term "external aqueous phase" as used herein means that, during the preparation of the lipid nanoparticle, after mixing the nucleic acid with the lipid component to form the nanoparticle, the nanoparticle is in an aqueous medium (such as a buffer), which is the external aqueous phase.
The term ALC-0315 as used herein is a structural compound with CAS number: 2036272-55-4, the structural formula is as follows:
Drawings
FIG. 1 schematic design of self-replicating repRNA-VZV constructs.
FIG. 2 is a flow chart of detection of LNP-XR003-1, LNP-XR003-2, and LNP-XR003-4 expression.
FIG. 3 shows the expression of LNP-XR003-5, LNP-XR003-6 and LNP-XR003-7 before and after the addition of protein transport inhibitor.
FIG. 4 shows the mean fluorescence intensity of LNP-XR003-5, LNP-XR003-6 and LNP-XR003-7 before and after the addition of the protein transport inhibitor.
FIG. 5 is an ELISA for detection of XR003-1 and XR003-2 group mice gE antibody levels.
FIG. 6 shows ELISPOT detection of IFN-gamma levels in mice of groups XR003-1 and XR 003-2.
FIG. 7 is a flow chart of INF-gamma, IL-2 and TNF-alpha levels released by CD4+ T cells from mice of groups XR003-1 and XR 003-2.
FIG. 8 is an ELISA for detection of XR003-5 to XR003-7 group mice gE antibody levels.
FIG. 9 shows ELISPOT detection of IFN-gamma levels in mice groups XR003-5 through XR 003-7.
FIG. 10 is a flow chart of INF-gamma, IL-2 and TNF-alpha levels released by CD4+ T cells from mice of groups XR003-5 through XR 003-7.
FIG. 11 shows ELISA detection of XR003-4 group mice gH antibody levels.
FIG. 12 shows ELISPOT detection of IFN-gamma levels in mice of group XR 003-4.
FIG. 13 is a flow chart of INF-gamma, IL-2 and TNF-alpha levels released by CD4+ T cells from mice of group XR 003-4.
FIG. 14 is a flow chart of INF-gamma, IL-2 and TNF-alpha levels released by CD8+ T cells from mice of group XR 003-4.
Detailed Description
The invention will be further described with reference to specific embodiments, and advantages and features of the invention will become apparent from the description. The specific conditions are not noted in the examples and are carried out according to conventional conditions or conditions recommended by the manufacturer. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention.
The present embodiments are merely examples and do not limit the scope of the present invention in any way. It will be understood by those skilled in the art that various changes and substitutions of details and forms of the technical solution of the present invention may be made without departing from the spirit and scope of the present invention, but these changes and substitutions fall within the scope of the present invention.
Example 1XR003-1 to XR003-7 construct designs
6 MRNA molecules were designed using the RNA self-replication platform (NSP 1-4 nucleic acid sequence shown in SEQ ID NO: 1) as shown in FIG. 1, wherein:
1.XR003-1
The VZV-OKA strain glycoprotein gE is selected as an antigen, the C terminal 50 amino acids are deleted, and mutation Y569A is introduced, wherein the nucleotide and amino acid sequences are shown as SEQ ID NO. 2-3.
2.XR003-2
XR003-2 is connected with Flt3L gene through P2A at N end of XR003-1, and nucleotide and amino acid sequences are shown in SEQ ID NO. 4-5.
3.XR003-4
XR003-4 lacks 8AA at the C end of gH protein, the N end is connected through the C end of P2A gL, his tag is added at gLC end for protein expression detection, and the nucleotide and amino acid sequences are shown in SEQ ID NO. 6-7.
4.XR003-5
XR003-5 inserts signal peptide into N end of gE protein, and lacks gE transmembrane domain and intracellular region, and its nucleotide and amino acid sequences are shown in SEQ ID NO. 8-9.
5.XR003-6
XR003-6 is connected with human mitochondrial ferritin through (G4S) 4 at the C end of XR003-5 protein sequence, and the nucleotide and amino acid sequences are shown in SEQ ID NO 10-11.
6.XR003-7
XR003-7 is connected with human heavy chain ferritin through (G4S) 4 at the C end of XR003-5 protein sequence, and the nucleotide and amino acid sequences are shown in SEQ ID NO. 12-13.
TABLE 1 sequence information Table
Note that: the nucleotide sequences in the tables are indicated by ATGC. Those skilled in the art know that ATGC when the nucleotide is DNA; AUGC when transcribed into mRNA.
EXAMPLE 2 preparation of XR003-1 to XR003-7 stock solutions
The 6 construct plasmids designed in example 1 were subjected to enzymatic tangentially and mRNA was prepared by In Vitro Transcription (IVT), capping and purification. The method comprises the following specific steps:
2.1 plasmid linearization cleavage
The six XR003 plasmids were subjected to enzymatic cleavage and purification according to conventional plasmid linearization cleavage and purification methods in the art, and subjected to capping and purification by in vitro co-transcription to prepare mRNA.
Plasmid linearized cleavage was performed using NotI (NEB, R3189L) or BspQ I (Nanjinovazan, DD 4302-03) under conventional systems. After the enzyme digestion, 1% agarose gel electrophoresis and a gel imager are carried out to observe whether the enzyme digestion of the sample is complete.
2.2 In vitro transcription and purification
An in vitro transcription system was prepared using linearized plasmid templates according to methods conventional in the art, incubated at 37℃for 2h, subjected to in vitro transcription, then digested at 37℃for 15min with DNase I, and the DNA template removed, followed by purification of mRNA by ion exchange chromatography media. Gel electrophoresis was performed on the purified RNA, resulting in: the size of all RNA molecules after electrophoresis was consistent with that expected, confirming that RNA was successfully prepared.
2.3MRNA integrity detection
2.3.1 Sample treatment: mRNA dilution with 1 XTE buffer
2.3.2 Adding the RNA diluent standard into a 96-well sample plate, adding diluted sample solution and RNA LADDER solution, sealing by using a sealing plate film, and centrifuging for 2 minutes at 3000 rpm/min. Mix well for 2 minutes with shaking at 2000 rpm. RNA integrity was obtained using a 5200Fragment Analyzer assay.
2.4 Detection of RNA concentration
Ribogreen dyes (ThermoFisher, R11490) were mixed with 1×te buffer for use. RNA standards were diluted with 1 XTE. 100. Mu.L of mRNA sample to be detected was added to a 96-well plate, followed by addition of 100. Mu. L Ribogreen dye solution and placed on a plate shaker for 5 minutes. And detecting by using a SpectraMax iD3 multifunctional enzyme-labeled instrument. Total RNA concentration was calculated using a standard curve.
Concentration and integrity measurements were performed on the transcribed and purified XR003 mRNA. The integrality of six mRNA in XR003 exceeds 85%, the concentration is between 2.4mg/ml and 2.5mg/ml, and the subsequent preparation requirements of LNP can be met.
EXAMPLE 3 preparation of XR003-1 to 7 constructs LNP
TABLE 2 materials and reagents
Sample name | Sample name |
ALC0315 | Quant-itTMRiboGreen RNA Assay Kit |
ALC0159 | 1 XTE buffer |
Cholesterol | Triton X-100 |
DSPC | Spike protein liquid |
3.1LNP/mRNA preparation
The lipid nanoparticle is prepared by a microfluidic method.
Preparing an aqueous phase: the RNA stock solutions (XR 003-1, XR003-2, XR003-3, XR003-4, XR003-5, XR003-6, XR 003-7) were treated with 50mM potassium dihydrogen phosphate solution pH4.5;
Organic phase preparation: the lipid was diluted with absolute ethanol (ALC 0315: ALC0159: DSPC: cholesterol, molar ratio 47.4%:1.8%:9.9%: 40.8%).
The aqueous phase and the organic phase are subjected to micro-fluidic, the prepared LNP is diluted by PBS diluent, an ultrafiltration centrifuge tube is used for concentration, liquid exchange, constant volume, split charging by a microporous filter membrane with the size of 0.22 mu m in a biosafety cabinet, and after frozen for 24 hours at the temperature of minus 60 ℃, particle size, polydispersity index, zeta potential, total concentration of mRNA, encapsulation rate and mRNA integrity detection are carried out.
3.2LNP/mRNA preparation particle size, PDI and Zeta potential detection
LNP/mRNA preparation particle size and potential were measured using Malvern Zetasizer ultra. mu.L of the solution containing LNP particles was diluted to 1mL with water for injection, and placed in a detection cell to detect the particle size, PDI and Zeta potential of LNP/mRNA preparation particles.
3.3 Detection of Total RNA concentration and encapsulation efficiency in LNP/mRNA preparation
Total RNA concentration and encapsulation efficiency in LNP/mRNA preparations were measured by the RiboGreen method. The LNP/mRNA preparation solution was demulsified with triton X-100, 100. Mu.L of the demulsified solution was added to a 96-well plate, and then 100. Mu.L of the riboGreen dye solution was added, and the solution was placed on a plate shaker and shaken for 5 minutes at 600rpm. And detecting by using a SpectraMax iD3 multifunctional enzyme-labeled instrument. Total RNA concentration was calculated using a standard curve.
10. Mu.l of LNP/mRNA preparation solution was taken, 990. Mu.l of buffer was added, mixed well, 100. Mu.l of riboGreen dye solution was added, and placed on a plate shaker, and the shaking was carried out for 5 minutes at 600rpm. And detecting by using a SpectraMax iD3 multifunctional enzyme-labeled instrument. The free RNA concentration was calculated using a standard curve. The encapsulation efficiency was then calculated according to the following formula.
Encapsulation (%) = 100% - (free mRNA concentration/total RNA concentration)%
3.4 Detection of mRNA integrity in LNP/mRNA preparation
MRNA integrity detection: 10% of triton X-100 is added into LNP/mRNA sample to carry out demulsification, thus obtaining broken emulsion. Diluting to a concentration of about 8 ng/. Mu.l by using a1 XTE buffer solution, uniformly mixing, denaturing at 70 ℃ for 2min, rapidly transferring to ice for preservation, then adding a 96-well sample plate, sealing by using a sealing plate film, and centrifuging to remove bubbles. And detecting by using an Agilent 5200 fragment analyzer to obtain the mRNA integrity.
3.5 Detection results
The mass measurements of the seven LNP formulations LNP/XR003-1, LNP/XR003-2, LNP/XR003-3, LNP/XR003-4, LNP/XR003-5, LNP/XR003-6, LNP/XR003-7 are shown in the following table. The indexes of LNP/XR003-1, LNP/XR003-2, LNP/XR003-3, LNP/XR003-4, LNP/XR003-5, LNP/XR003-6 and LNP/XR003-7 prepared by the representative process meet the standard specification. The particle size of the 7 preparations is 64-69 nm, PDI is 0.084-0.153, electric potential is-5 mV-7 mV, total RNA concentration is 0.32-0.45 mg/ml, encapsulation rate is above 92%, and mRNA integrity is 65% -70%.
TABLE 3 detection results
Name of the name | Particle size | PDI | Potential of | Total RNA concentration | Encapsulation efficiency | MRNA integrity |
LNP/XR003-1 | 66nm | 0.084 | -5mV | 0.45mg/ml | 93% | 68% |
LNP/XR003-2 | 69nm | 0.153 | 7mV | 0.42mg/ml | 92% | 65% |
LNP/XR003-4 | 64nm | 0.098 | 7mV | 0.32mg/ml | 92% | 67% |
LNP/XR003-5 | 64nm | 0.111 | 5mV | 0.41mg/ml | 92% | 70% |
LNP/XR003-6 | 66nm | 0.123 | 6mV | 0.38mg/ml | 92% | 68% |
LNP/XR003-7 | 65nm | 0.101 | 6mV | 0.37mg/ml | 92% | 66% |
Example 4 detection of expression of LNP-XR003-1 to 7 in 293T cells
293T cells were aliquoted into 24 well plates, cultured overnight, 100. Mu.l/well was aspirated when the cell fusion should be greater than or equal to 80%, opti-MEM medium (gibco, cat# 31985-062) was placed into sterile centrifuge tubes, 1. Mu. gLNP-XR003-1, 2, 4, 5, 6, 7 molecules were added to each well, mixed well, 293T cells were added, XR003-5 to XR003-7 transfected 2 parts, 1. Mu.l of the eBioscience TM protein transport inhibitor mixture (Thermo fisher, cat# 00-4980-03) was added in one part, and sample treatment and expression measurement were performed after 48 h.
As shown in FIG. 2, the gE expression can be detected by all the XR003-1 to XR003-3 molecules, the XR003-4 gL protein can be expressed on a membrane, and the XR003-5 to XR003-7 molecules can be secreted outside the cell after the transmembrane region sequence is removed; as shown in fig. 3 and 4, the protein transport inhibitor mixture prevents transport of proteins outside the cell, and is effective to increase intracellular gE protein accumulation upon addition of the inhibitor.
Example 5XR003-1 and 2 molecular mouse immunogenicity detection
The animals groups referred to in example 5
SPF-class female 6-8 week-old C57BL/6 mice were randomly divided into 3 groups of 6 animals each, grouped as follows:
a first group: LNP-XR003-1 1. Mu.g/dose
Second group: LNP-XR003-2 1. Mu.g/dose
Third group: 50 μl PBS
Each group was vaccinated with 1 μg (50 μl) of vaccine, and 2 immunizations were performed at 4 weeks intervals, and ELISA detection of gE antibody levels was performed by taking blood on days D14, D28, and D47, respectively, and spleen of mice was collected on day D47 for IFN-. Gamma.ELISPOT detection and cytokine detection.
5.1ELISA method for detecting gE antibody level in mouse serum
96-Well ELISA plates were coated with VZV-gE (0.5. Mu.g/ml) overnight at 4 ℃. Plates were washed 5 times with 260 μl/well PBST and blocked with 3% BSA in PBST for 2 hours at 37deg.C. Immunized mouse serum was serially diluted twice and added to each well, incubated at 37 ℃ for 1 hour, plated 5 times with PBST, and dried by pipetting with HRP-labeled goat anti-mouse IgG. TMB was added to develop the color for 10min, ELISA stop solution was added to stop the reaction. The absorbance at 450nm of each well is read by an enzyme-labeled instrument, and the sample OD450 is equal to or greater than the maximum dilution corresponding to the average value of all negative control samples multiplied by 2.1.
As shown in fig. 5, the differences in the levels of gE antibodies were not significant in the XR003-1 and XR003-2 groups D14, D28, and the levels of gE antibodies to D47 were significantly increased after D28 days of booster immunization; d47 Both XR003-1 and XR003-2 groups had higher antibody levels, with the XR003-2 group being slightly higher than the XR003-1 group, but the difference was not significant.
5.2ELISPot detection of IFN-gamma levels secreted by mouse spleen cells
Incubation of spleen cell suspensions (2X 10 5 cells) with PRIM 1640 medium (negative control), gE peptide pool (6.67. Mu.g/ml), or ConA (6.67. Mu.g/ml, positive control) for 16-20h was performed using the mouse IFN-. Gamma.ELISPOT kit (Abcam ab 64029) according to the manufacturer's instructions. The number of spots was determined using an enzyme-linked immunosorbent assay (Cellular Technology Limited, S6 Entry). The prevention of shingles requires intense VZV-specific cellular immunity, which is characterized by IFN- γ secretion. T cells are responsible for producing IFN- γ in adaptive immunity, which means that the level of IFN- γ produced by a herpes zoster vaccine may reflect its ability to induce specific cellular immunity. To study VZV-specific cellular immunity, the enzyme-linked immunosorbent spot assay stimulated the number of IFN- γ secreting splenocytes with the gE protein peptide pool on day 47.
As shown in FIG. 6, both XR003-1 and XR003-2 had higher levels of IFN-gamma secretion, and in addition XR003-2 induced higher specific cellular immunity when splenocytes were stimulated with the gE peptide pool than XR003-1, but the difference was not significant.
5.3 Intracellular cytokine detection
1.5X10 6 spleen cells were incubated with 0.2. Mu.g of gE protein peptide library or 1 XeBioscience TM cell stimulating mixture (Thermo, 00-4970-03) at 37℃for 1h followed by addition of 1 XeBioscience TM protein transport inhibitor mixture (Thermo, 00-4980-93) and incubation in a 5% carbon dioxide incubator for 5h to block cytokine release.
Cell surface marker staining: cells were blocked with 50. Mu.l of 4% TruStain FcX TM anti-mouse CD16/32 (Biolegend, 101320) in PBS and incubated at 4℃for 8-10min, 50. Mu.l of 1:500LIVE/DEAD TM A mixture of pale green DEAD cell stain/L34957 (Thermo, L34957) with 2% CD45 (bioleged, 103116), CD8 (bioleged, 100734), CD4 (bioleged, 100422) direct fluorescent antibody in PBS was incubated at 4℃for 20min.
Cytokine staining: the mixture was fixed at IC Fixation Buffer (thermo Fisher, 00-8222-49) for 20min at 4 ℃. Membrane-through staining was performed with 100. Mu.l (containing 1% IL-2 (Biolegend, 503808), IFN-. Gamma. (Biolegend, 505830), TNF-. Alpha. (Biolegend, 506304) direct-labeled fluorescent antibody) 1X Permeabilization Buffer (ThermoFisher, 00-8333-56) and incubated at 4℃for 20min in the absence of light. After staining, the cells were subjected to loop gating (forward and side scatter, FSC/SSC) and samples were analyzed using Attune NxT acoustic focusing flow cytometry (Thermo, 2AFC 236901121).
As a result, as shown in FIG. 7, CD4 + cells, after stimulation with gE peptide library, can specifically secrete Th1 type cytokines IFN-gamma, IL-2 and TNF-alpha, and XR003-2 is superior to XR003-1.
Based on the experimental data, XR003-1 and XR003-2 can both induce corresponding antibodies to generate better, and have better immunogenicity, and the addition of FIT3L ligand has a certain degree of improvement on the immunogenicity.
EXAMPLE 6XR003-5 to 7 molecular mouse immunogenicity
The animals group referred to in example 6
SPF-class female 6-8 week-old C57BL/6 mice were randomly divided into 4 groups of 6 animals each, each group being as follows:
A first group: LNP-XR003-5 1. Mu.g/dose
Second group: LNP-XR003-6 1. Mu.g/dose
Third group: LNP-XR003-7 1. Mu.g/dose
Fourth group: 50 μl PBS
Each group was vaccinated with 1 μg (50 μl) of vaccine, and 2 immunizations were performed at 4 weeks intervals, and ELISA detection of gE antibody levels was performed by taking blood on days D14, D28, and D47, respectively, and spleen of mice was collected on day D47 for IFN-. Gamma.ELISPOT detection and cytokine detection.
6.1ELISA method for detecting gE antibody level in mouse serum the experimental method is the same as that of 5.1, and the results are shown in FIG. 8, D14 and D28 immune groups show that gE antibody level is not obviously different, gE antibody level to D47 is obviously increased after D28 days of booster immunization, and XR003-5 to XR003-7 groups show that the XR003-5 group has the highest antibody level.
6.2ELISPot the same experimental procedure as 5.2 for detecting INF-gamma secretion from spleen cells of mice, and the results are shown in FIG. 9, XR003-5, XR003-6, XR003-7, in which gE peptide library-specific cellular immunity was generated and the number of IFN-gamma spots in XR003-6 group was the greatest.
6.3 Intracellular cytokine detection experiments were performed in the same manner as 5.3, and as shown in FIG. 10, after stimulation with gE peptide library, the vaccine group and the control group, CD4+ T cells can specifically secrete Th1 type cytokines IFN-gamma, IL-2 and TNF-alpha, wherein the cell proportion of IFN-gamma, IL-2 and TNF-alpha secreted by the XR003-6 group CD4 + T cells is the highest.
Based on the experimental data, XR003-5 to XR003-7 can induce corresponding antibodies to generate better, have better immunogenicity, and the added mitochondrial ferritin has a certain degree of improvement on the immunogenicity.
EXAMPLE 7XR003-4 molecular mouse immunogenicity
Animal grouping in example 7
SPF-class female 6-8 week-old C57BL/6 mice were randomly divided into 2 groups of 6 animals each, grouped as follows:
a first group: LNP-XR003-4 1. Mu.g/dose
Second group: 50 μl PBS
The first group was vaccinated with 1 μg (50 μl) of vaccine, and immunized 2 times at 4 weeks intervals, ELISA was performed to detect the gH antibody level by taking blood on days D14, D28, and D47, and spleen of the mice was collected on day D47 for IFN-. Gamma.ELISPOT detection and intracellular cytokine detection, respectively.
7.1ELISA method for detecting gH antibody level in mouse serum
96-Well ELISA plates were coated overnight with VZV-gH protein (4. Mu.g/ml) at 4℃and tested in the same manner as 5.1. As a result, as shown in FIG. 11, XR003-4 group produced gH-specific antibodies, which had slightly higher antibody titer at D28 than at D14, and significantly increased gH antibody levels in XR003-4 group after booster immunization;
7.2ELISPot detection of INF-gamma levels secreted by mouse spleen cells
Experimental methods were similar to 5.2, and as shown in FIG. 12, XR003-4 immunized mice produced a high level of gH-specific Th1 type cellular immune response;
7.3 intracellular cytokine detection
The experimental method is the same as 5.3, and the results are shown in FIG. 13 and FIG. 14, and CD4 + and CD8 + T cells (cell surface CD8 antibody-derived Biolegend, 100734) can specifically secrete Th1 type cytokines IFN-gamma, IL-2 and TNF-alpha after being stimulated by the gH peptide library;
Based on the experimental data, XR003-4 can induce humoral immunity and cellular immune response well, and the induced cellular immune response comprises two T cell types of CD4 + and CD8 +, so that the cell type has good immunogenicity.
Claims (23)
1. A pharmaceutical composition comprising an mRNA molecule construct and a delivery formulation; preferably, the mRNA construct comprises mRNA encoding the gE and/or gH, gL regions of VZV; more preferably, the mRNAs encoding the gE and/or gH, gL regions of VZV are expressed alone or in fusion in a host cell.
2. The pharmaceutical composition of claim 1, wherein the mRNA encodes a protein further comprising a mutation; optionally, the mRNA encodes a protein that further comprises Flt3L; optionally, the mRNA encodes a protein further comprising a signal peptide sequence; optionally, the signal peptide is located at the N-terminus of the protein; preferably, the signal peptide is derived from murine H-2Kb, human IgE, HLA-B46, MICA 008, OSM, VSV-G, mouse Ig Kappa, mouse heavy chain, BM40, human chymotrypsinogen, human prothrombin-2, human IL-2, human G-CSF, human hemagglutinin IX, human albumin, gaussia luc, HAS, influenza virus, human insulin, silk LC, erenumab antibody light chain, pembrolizumab light chain, ramucirumab light chain, E SIGNAL PEPTIDE, SP1 (LZJ human IgG1, SP2, SP3 (ZLQ).
3. The pharmaceutical composition of claim 1 or 2, wherein the gE extracellular region (gE Ecto) protein binds to ferritin (Ferritin) to form a fusion protein; preferably, gE Ecto-ferritin fusion proteins comprise the unit subunit of ferritin; alternatively, the ferritin subunit is a full length or any portion of ferritin, wild type or partial amino acid mutation; alternatively, the monomeric subunit is from mitochondrial ferritin or heavy chain ferritin.
4. A pharmaceutical composition according to any one of claims 1 to 3, wherein the protein encoded by the mRNA comprises an amino acid sequence having at least 90%, 95%, 99%, 100% identity to the amino acid sequence as set forth in SEQ ID No. 3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID No. 13.
5. The pharmaceutical composition of any one of claims 1-4, wherein the mRNA comprises a coding nucleotide corresponding to the amino acid sequence set forth in SEQ ID No. 3,SEQ ID NO:5,SEQ ID NO:7,SEQ ID NO:9,SEQ ID NO:11 or SEQ ID No. 13.
6. The pharmaceutical composition of any one of claims 1-5, wherein the mRNA comprises the nucleotide sequence set forth in SEQ ID No. 2,SEQ ID NO:4,SEQ ID NO:6,SEQ ID NO:8,SEQ ID NO:10 or SEQ ID No. 12.
7. The pharmaceutical composition of any one of claims 1-6, wherein the mRNA construct further comprises a 5'utr and/or a 3' utr sequence; optionally, the 5' UTR sequence is a human alpha globulin 5' UTR or a non-native 5' UTR sequence; optionally, the mRNA construct further comprises a Poly (a) sequence.
8. The pharmaceutical composition of any one of claims 1-7, wherein the mRNA construct further comprises an alphavirus nonstructural protein (nsp) gene; optionally, the nsp gene is shown as SEQ ID NO. 1.
9. The pharmaceutical composition of any one of claims 1-8, wherein the delivery formulation is a nanoparticle; preferably, the delivery formulation comprises lipid nanoparticles (Lipid nanoparticle, LNP), lipid multipolymer (lipopolyplex, LPP), polymer nanoparticles (Polymer nanoparticles, PNP), inorganic nanoparticles (Inorganic nanoparticles, INP) cationic nanoemulsions (Cationic nanoemulsion, CNE), exosomes, biologicals microvesicles, protamine, etc.; more preferably, the nanoparticle is a lipid nanoparticle (Lipid nanoparticle, LNP); more preferably, the lipid comprises one or more of a cationic lipid, a neutral helper lipid, cholesterol, and a PEG-modified lipid; more preferably, two or more are included.
10. The pharmaceutical composition of any one of claims 1-9, wherein the lipid nanoparticle comprises: a) a cationic lipid, b) a neutral helper lipid, c) cholesterol, and d) a PEG-modified lipid; preferably, in the lipid nanoparticle, the molar ratio of each lipid component is as follows, based on 100% of the total molar amount of lipid: 45% -50% of a cationic lipid, 5% -10% of b) a neutral auxiliary lipid, 38% -48% of c) cholesterol and 0% -3% of d) PEG modified lipid.
11. The pharmaceutical composition according to any one of claims 1 to 10, wherein the molar ratio of the cationic lipid is 45% to 48% based on 100% of the total molar amount of lipid; preferably, the cationic lipid is selected from the group consisting of N, N-dimethyl-2, 3-dioleoyloxypropylamine (DODMA), 1, 2-dimyristoxypropyl-3-dimethyl-hydroxyethylammonium bromide (DMRIE), N, N-dioleoyln, N-dimethylammonium chloride (DODAC), 1, 2-dioleoyl-3-dimethylammonium-propane (DODAP), N, N-distearyl-N, N-dimethylammonium bromide (DDAB), N- (l- (2, 3-dioleoyloxy) propyl) -N, N, N-trimethylammonium chloride (DOTAP), N- (1- (2, 3-dioleoyloxy) propyl) -N, N, N-trimethylammonium chloride (DOTMA), 1, 2-dimethanooxy-N, N-dimethylaminopropane (DLinDMA), 2-dioleylene-4- (2-dimethylaminoethyl) - [1,3] -dioxolane (DLIn), N- (2, N-dimethyl-MC-4- (DLMC-2-DMA), or any one of the methyl-2- (3-dioleyloxy) propyl) -N, N, N-trimethylammonium chloride (DOTAP), N- (1- (2, 3-dioleyloxy) propyl) -N, N-trimethyl ammonium chloride (DLMA).
12. The pharmaceutical composition according to any one of claims 1 to 11, wherein the molar ratio of neutral helper lipids is 6% to 10% based on 100% total molar amount of lipids; preferably, the molar ratio of the neutral auxiliary lipid is 8% -10%; optionally, the neutral helper lipid is selected from any one or more of distearoylphosphatidylcholine(DSPC)、dioleoylphosphatidylcholine(DOPC)、dimyristoylphosphatidylcholine(DMPC)、dipalmitoylphosphatidylcholine(DPPC)、diarachidoylphosphatidylcholine(DAPC)、dibehenoylphosphatidylcholine(DBPC)、ditricosanoylphosphatidylcholine(DTPC)、dilignoceroylphatidylcholine(DLPC)、dioleoylphosphatidylethanolamine(DOPE)、dipalmitoyl-phosphatidylethanolamine(DPPE)、dimyristoyl-phosphatidylethanolamine(DMPE) or phosphatidylethanolamine (DLPE).
13. The pharmaceutical composition according to any one of claims 1 to 12, wherein the PEG-modified lipid is present in a molar ratio of 1% to 3% based on 100% total molar amount of lipid; preferably, the molar ratio of the PEG modified lipid is 1% -2%; optionally, the PEG modified lipid is selected from any one or more of methoxy polyethylene glycol bitetradecylacetamide (ALC-0159), MG-PEG2000, DMG-PEG5000 and PEG 2000.
14. The pharmaceutical composition according to any one of claims 1 to 13, wherein the molar ratio of cholesterol is 40% to 46% based on 100% of the total molar amount of lipid; preferably, the molar ratio of the cholesterol is 42% -45%.
15. A vaccine comprising the pharmaceutical composition of any one of claims 1-14; preferably, the vaccine is a VZV vaccine.
16. An mRNA construct as an immunogenic component in the medicament of any one of claims 1-14 or the vaccine of claim 15, or a combination thereof.
17. A vector comprising the mRNA construct of claim 16.
18. A nanoparticle in a pharmaceutical composition as claimed in any one of claims 9 to 14.
19. A fusion protein comprising a protein encoded by the mRNA construct of claim 16 and a second protein fused thereto; optionally, the protein encoded by the mRNA is an unfused protein; preferably, the second protein is a carrier protein; more preferably, the second protein is ferritin (Ferritin); more preferably, the ferritin is mitochondrial ferritin or heavy chain ferritin; more preferably, the ferritin comprises a domain that allows the fusion protein to self-assemble into a nanoparticle.
20. Use of the pharmaceutical composition of any one of claims 1-14, the vaccine of claim 15, the mRNA construct of claim 16, the vector of claim 17, the nanoparticle of claim 18 in the preparation of a medicament for inducing an immune response in a subject in need thereof.
21. Use of the pharmaceutical composition of any one of claims 1-14, the vaccine of claim 15, the mRNA construct of claim 16, the vector of claim 17, the nanoparticle of claim 18 in the manufacture of a medicament for the treatment or prevention of a VZV-related disease in a subject in need thereof.
22. A method of inducing an immune response in a subject in need thereof, comprising administering to the subject the pharmaceutical composition of any one of claims 1-14 or the vaccine of claim 15.
23. A method of treating or preventing a VZV-related disease in a subject in need thereof, wherein the pharmaceutical composition of any one of claims 1-14 or the vaccine of claim 15 is administered to the subject.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202410403081.3A CN118340876A (en) | 2024-04-03 | 2024-04-03 | Varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202410403081.3A CN118340876A (en) | 2024-04-03 | 2024-04-03 | Varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN118340876A true CN118340876A (en) | 2024-07-16 |
Family
ID=91814968
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202410403081.3A Pending CN118340876A (en) | 2024-04-03 | 2024-04-03 | Varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN118340876A (en) |
-
2024
- 2024-04-03 CN CN202410403081.3A patent/CN118340876A/en active Pending
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN114081943B (en) | Varicella-zoster mRNA vaccine composition and preparation method and application thereof | |
CN105473158B (en) | Respiratory Syncytial Virus (RSV) vaccine | |
US20240207392A1 (en) | Epstein-barr virus mrna vaccines | |
CN113151196A (en) | Recombinant vaccinia virus, vaccinia virus vector vaccine, application and preparation method thereof | |
WO2023036191A1 (en) | Vaccine for novel coronavirus sars-cov-2 delta variant and application | |
EP4034548A1 (en) | Coronavirus vaccines and uses thereof | |
ES2902787T3 (en) | DNAi vaccines and procedures for using the same | |
CN112662695B (en) | Construction method and application of bacterial biofilm vesicle BBV as vaccine vector | |
JP2010511406A (en) | Linear expression cassette vaccine | |
KR20240099279A (en) | RNA molecule | |
CN116426543B (en) | Novel coronavirus vaccine and preparation method thereof | |
CN118340876A (en) | Varicella-zoster virus self-replicating mRNA vaccine pharmaceutical composition and application thereof | |
CN116200403A (en) | Novel coronavirus mRNA vaccine for preventing mutant strain | |
Koppu et al. | Current perspectives and future prospects of mRNA vaccines against viral diseases: A brief review | |
WO2022109093A1 (en) | Compositions and methods for treating and suppressing allergic responses | |
CN118340875A (en) | MRNA vaccine of varicella-zoster virus and application thereof | |
CN117205309B (en) | Influenza immunogen composition, preparation method and application thereof | |
JP2002520299A (en) | Polynucleotide vaccine formulation | |
WO2024055273A1 (en) | Rabies mrna vaccine and preparation and use thereof | |
CN116904489B (en) | Duck tembusu virus nucleic acid vaccine and application thereof | |
WO2023202711A1 (en) | Mrna vaccine based on novel coronavirus | |
WO2024120490A1 (en) | Self-replicating rna vaccines and methods of use | |
US20170224808A1 (en) | Therapeutic compositiojns and methods for inducing an immune response to herpes simplex virus type 2 (hsv-2) | |
CN117683790A (en) | mRNA vaccine of Legionella pneumophila related antigen, preparation method and application thereof | |
WO2023236041A1 (en) | Mrna vaccine encoding pcrv and/or oprf-i protein |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |