WO2021206638A1 - Vaccine and/or antibody for viral infection - Google Patents
Vaccine and/or antibody for viral infection Download PDFInfo
- Publication number
- WO2021206638A1 WO2021206638A1 PCT/SG2021/050197 SG2021050197W WO2021206638A1 WO 2021206638 A1 WO2021206638 A1 WO 2021206638A1 SG 2021050197 W SG2021050197 W SG 2021050197W WO 2021206638 A1 WO2021206638 A1 WO 2021206638A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cov
- coronavirus
- protein
- sars
- antibody
- Prior art date
Links
- 229960005486 vaccine Drugs 0.000 title claims abstract description 41
- 230000009385 viral infection Effects 0.000 title claims description 23
- 208000036142 Viral infection Diseases 0.000 title claims description 22
- 101710141454 Nucleoprotein Proteins 0.000 claims abstract description 98
- 230000002163 immunogen Effects 0.000 claims abstract description 74
- 241001678559 COVID-19 virus Species 0.000 claims abstract description 73
- 239000012634 fragment Substances 0.000 claims abstract description 67
- 108091006197 SARS-CoV-2 Nucleocapsid Protein Proteins 0.000 claims abstract description 47
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 38
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 38
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 38
- 230000027455 binding Effects 0.000 claims abstract description 33
- 239000000427 antigen Substances 0.000 claims abstract description 16
- 102000036639 antigens Human genes 0.000 claims abstract description 16
- 108091007433 antigens Proteins 0.000 claims abstract description 16
- 208000025721 COVID-19 Diseases 0.000 claims description 37
- 241000315672 SARS coronavirus Species 0.000 claims description 29
- 208000015181 infectious disease Diseases 0.000 claims description 26
- 208000001528 Coronaviridae Infections Diseases 0.000 claims description 22
- 230000003053 immunization Effects 0.000 claims description 18
- 238000000034 method Methods 0.000 claims description 15
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 claims description 14
- 239000002671 adjuvant Substances 0.000 claims description 13
- 108090001074 Nucleocapsid Proteins Proteins 0.000 claims description 11
- 241000127282 Middle East respiratory syndrome-related coronavirus Species 0.000 claims description 9
- 239000013598 vector Substances 0.000 claims description 9
- 239000000203 mixture Substances 0.000 claims description 8
- 239000008194 pharmaceutical composition Substances 0.000 claims description 8
- 238000002360 preparation method Methods 0.000 claims description 8
- 230000000890 antigenic effect Effects 0.000 claims description 5
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 5
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims description 4
- 239000003814 drug Substances 0.000 claims description 4
- 101001024637 Severe acute respiratory syndrome coronavirus 2 Nucleoprotein Proteins 0.000 claims 1
- 238000005516 engineering process Methods 0.000 abstract description 5
- 108020004511 Recombinant DNA Proteins 0.000 abstract description 4
- 108090000765 processed proteins & peptides Proteins 0.000 description 65
- 238000002255 vaccination Methods 0.000 description 42
- 241000699670 Mus sp. Species 0.000 description 39
- 108090000623 proteins and genes Proteins 0.000 description 35
- 102000004169 proteins and genes Human genes 0.000 description 34
- 210000002966 serum Anatomy 0.000 description 27
- 241000699666 Mus <mouse, genus> Species 0.000 description 26
- 102000004196 processed proteins & peptides Human genes 0.000 description 23
- 108090000695 Cytokines Proteins 0.000 description 19
- 102000004127 Cytokines Human genes 0.000 description 19
- 210000001744 T-lymphocyte Anatomy 0.000 description 19
- 238000002649 immunization Methods 0.000 description 15
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 14
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 14
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 14
- 239000002953 phosphate buffered saline Substances 0.000 description 14
- 238000011725 BALB/c mouse Methods 0.000 description 13
- 101100454807 Caenorhabditis elegans lgg-1 gene Proteins 0.000 description 13
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 12
- 210000003071 memory t lymphocyte Anatomy 0.000 description 12
- 102100032912 CD44 antigen Human genes 0.000 description 11
- 241000711573 Coronaviridae Species 0.000 description 11
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 11
- 210000004027 cell Anatomy 0.000 description 11
- 238000010790 dilution Methods 0.000 description 10
- 239000012895 dilution Substances 0.000 description 10
- 241000283707 Capra Species 0.000 description 9
- 238000002965 ELISA Methods 0.000 description 9
- 210000004369 blood Anatomy 0.000 description 9
- 239000008280 blood Substances 0.000 description 9
- 230000008859 change Effects 0.000 description 9
- 230000036039 immunity Effects 0.000 description 9
- 230000003472 neutralizing effect Effects 0.000 description 9
- 230000003287 optical effect Effects 0.000 description 9
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 8
- 102100033467 L-selectin Human genes 0.000 description 8
- 230000016784 immunoglobulin production Effects 0.000 description 8
- 238000003491 array Methods 0.000 description 7
- 238000011161 development Methods 0.000 description 7
- 230000028993 immune response Effects 0.000 description 7
- 230000015654 memory Effects 0.000 description 7
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 6
- 238000012286 ELISA Assay Methods 0.000 description 6
- 150000001413 amino acids Chemical class 0.000 description 6
- 230000005875 antibody response Effects 0.000 description 6
- 229940098773 bovine serum albumin Drugs 0.000 description 6
- 239000000872 buffer Substances 0.000 description 6
- 238000013207 serial dilution Methods 0.000 description 6
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 5
- 238000011771 FVB mouse Methods 0.000 description 5
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 5
- 241000700605 Viruses Species 0.000 description 5
- 230000002788 anti-peptide Effects 0.000 description 5
- 210000003719 b-lymphocyte Anatomy 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 210000004408 hybridoma Anatomy 0.000 description 5
- 230000007774 longterm Effects 0.000 description 5
- 230000003612 virological effect Effects 0.000 description 5
- 102100031673 Corneodesmosin Human genes 0.000 description 4
- 101710139375 Corneodesmosin Proteins 0.000 description 4
- 210000003567 ascitic fluid Anatomy 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 210000000987 immune system Anatomy 0.000 description 4
- 238000009169 immunotherapy Methods 0.000 description 4
- 229920001184 polypeptide Polymers 0.000 description 4
- 230000000770 proinflammatory effect Effects 0.000 description 4
- 230000001681 protective effect Effects 0.000 description 4
- 229940023143 protein vaccine Drugs 0.000 description 4
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 3
- 101000797762 Homo sapiens C-C motif chemokine 5 Proteins 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 3
- 101000629318 Severe acute respiratory syndrome coronavirus 2 Spike glycoprotein Proteins 0.000 description 3
- 101001024647 Severe acute respiratory syndrome coronavirus Nucleoprotein Proteins 0.000 description 3
- 238000009825 accumulation Methods 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 210000002825 class switched memory b cell Anatomy 0.000 description 3
- 239000011248 coating agent Substances 0.000 description 3
- 238000000576 coating method Methods 0.000 description 3
- 230000000295 complement effect Effects 0.000 description 3
- 239000012530 fluid Substances 0.000 description 3
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 3
- 210000002865 immune cell Anatomy 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 230000035772 mutation Effects 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- 238000005406 washing Methods 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- YRNWIFYIFSBPAU-UHFFFAOYSA-N 4-[4-(dimethylamino)phenyl]-n,n-dimethylaniline Chemical compound C1=CC(N(C)C)=CC=C1C1=CC=C(N(C)C)C=C1 YRNWIFYIFSBPAU-UHFFFAOYSA-N 0.000 description 2
- 102100021943 C-C motif chemokine 2 Human genes 0.000 description 2
- OBMZMSLWNNWEJA-XNCRXQDQSA-N C1=CC=2C(C[C@@H]3NC(=O)[C@@H](NC(=O)[C@H](NC(=O)N(CC#CCN(CCCC[C@H](NC(=O)[C@@H](CC4=CC=CC=C4)NC3=O)C(=O)N)CC=C)NC(=O)[C@@H](N)C)CC3=CNC4=C3C=CC=C4)C)=CNC=2C=C1 Chemical compound C1=CC=2C(C[C@@H]3NC(=O)[C@@H](NC(=O)[C@H](NC(=O)N(CC#CCN(CCCC[C@H](NC(=O)[C@@H](CC4=CC=CC=C4)NC3=O)C(=O)N)CC=C)NC(=O)[C@@H](N)C)CC3=CNC4=C3C=CC=C4)C)=CNC=2C=C1 OBMZMSLWNNWEJA-XNCRXQDQSA-N 0.000 description 2
- 241000494545 Cordyline virus 2 Species 0.000 description 2
- 108010024636 Glutathione Proteins 0.000 description 2
- 102100039619 Granulocyte colony-stimulating factor Human genes 0.000 description 2
- 101000897480 Homo sapiens C-C motif chemokine 2 Proteins 0.000 description 2
- 101000746367 Homo sapiens Granulocyte colony-stimulating factor Proteins 0.000 description 2
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 2
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 2
- 241000712431 Influenza A virus Species 0.000 description 2
- 102000003814 Interleukin-10 Human genes 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- 208000025370 Middle East respiratory syndrome Diseases 0.000 description 2
- 101710176384 Peptide 1 Proteins 0.000 description 2
- 229940026233 Pfizer-BioNTech COVID-19 vaccine Drugs 0.000 description 2
- 230000005867 T cell response Effects 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 2
- 102100033732 Tumor necrosis factor receptor superfamily member 1A Human genes 0.000 description 2
- 101710187743 Tumor necrosis factor receptor superfamily member 1A Proteins 0.000 description 2
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 2
- 229960000723 ampicillin Drugs 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 238000002038 chemiluminescence detection Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 231100000517 death Toxicity 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 210000004379 membrane Anatomy 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 210000001806 memory b lymphocyte Anatomy 0.000 description 2
- 229940126619 mouse monoclonal antibody Drugs 0.000 description 2
- 210000003200 peritoneal cavity Anatomy 0.000 description 2
- 102000013415 peroxidase activity proteins Human genes 0.000 description 2
- 108040007629 peroxidase activity proteins Proteins 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 230000002516 postimmunization Effects 0.000 description 2
- 238000011321 prophylaxis Methods 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 208000026425 severe pneumonia Diseases 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000002459 sustained effect Effects 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 238000011282 treatment Methods 0.000 description 2
- 229940125575 vaccine candidate Drugs 0.000 description 2
- 239000011534 wash buffer Substances 0.000 description 2
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 1
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 1
- 108700022172 2019-nCoV Vaccine mRNA-1273 Proteins 0.000 description 1
- 102100035765 Angiotensin-converting enzyme 2 Human genes 0.000 description 1
- 108090000975 Angiotensin-converting enzyme 2 Proteins 0.000 description 1
- 230000003844 B-cell-activation Effects 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 1
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- -1 IFN-y Proteins 0.000 description 1
- 241000711450 Infectious bronchitis virus Species 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 102100022338 Integrin alpha-M Human genes 0.000 description 1
- 102100037850 Interferon gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 102000004388 Interleukin-4 Human genes 0.000 description 1
- 239000006137 Luria-Bertani broth Substances 0.000 description 1
- 229940026207 Moderna COVID-19 vaccine Drugs 0.000 description 1
- 208000034486 Multi-organ failure Diseases 0.000 description 1
- 102000016943 Muramidase Human genes 0.000 description 1
- 108010014251 Muramidase Proteins 0.000 description 1
- 241000711466 Murine hepatitis virus Species 0.000 description 1
- 108010062010 N-Acetylmuramoyl-L-alanine Amidase Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 229940096437 Protein S Drugs 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 230000010799 Receptor Interactions Effects 0.000 description 1
- 108091005634 SARS-CoV-2 receptor-binding domains Proteins 0.000 description 1
- 101100203795 Severe acute respiratory syndrome coronavirus 2 S gene Proteins 0.000 description 1
- 101000629313 Severe acute respiratory syndrome coronavirus Spike glycoprotein Proteins 0.000 description 1
- 101710198474 Spike protein Proteins 0.000 description 1
- 101710172711 Structural protein Proteins 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 241000710959 Venezuelan equine encephalitis virus Species 0.000 description 1
- 108010067390 Viral Proteins Proteins 0.000 description 1
- 210000001015 abdomen Anatomy 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 206010069351 acute lung injury Diseases 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 229940124691 antibody therapeutics Drugs 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- 239000013599 cloning vector Substances 0.000 description 1
- 230000007402 cytotoxic response Effects 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 239000012149 elution buffer Substances 0.000 description 1
- 230000002327 eosinophilic effect Effects 0.000 description 1
- 230000008029 eradication Effects 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 239000011536 extraction buffer Substances 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 230000005745 host immune response Effects 0.000 description 1
- 230000028996 humoral immune response Effects 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000006054 immunological memory Effects 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 229960003971 influenza vaccine Drugs 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 230000002045 lasting effect Effects 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 229960000274 lysozyme Drugs 0.000 description 1
- 239000004325 lysozyme Substances 0.000 description 1
- 235000010335 lysozyme Nutrition 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000001823 molecular biology technique Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 208000029744 multiple organ dysfunction syndrome Diseases 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 201000009240 nasopharyngitis Diseases 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229940023041 peptide vaccine Drugs 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000001932 seasonal effect Effects 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 230000007502 viral entry Effects 0.000 description 1
- 230000008478 viral entry into host cell Effects 0.000 description 1
- 229960004854 viral vaccine Drugs 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/42—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum viral
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/08—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
- C07K16/10—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
- C07K16/1002—Coronaviridae
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/08—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
- C07K16/10—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
- C07K16/1002—Coronaviridae
- C07K16/1003—Severe acute respiratory syndrome coronavirus 2 [SARS‐CoV‐2 or Covid-19]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55566—Emulsions, e.g. Freund's adjuvant, MF59
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/34—Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/20011—Coronaviridae
- C12N2770/20022—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/20011—Coronaviridae
- C12N2770/20034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- the present invention relates to prophylaxis and treatment of viral infection.
- the invention relates to immune therapies such as novel vaccines for prophylaxis and antibodies for treatment of viral infection, for example Coronavirus infection.
- Coronaviruses are enveloped, positive-sense, single-stranded RNA viruses of the family Coronaviridae. While most viruses cause mild illnesses such as the common cold, a few viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV) resulted in the severe acute respiratory syndrome (SARS) public health crises in 2003, Middle East respiratory syndrome coronavirus (MERS-CoV) caused Middle East respiratory syndrome (MERS) in 2009. In addition, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused Coronavirus disease 2019 (COVID-19) from late 2019. The outbreaks for SARS-CoV and MERS-CoV were regional, while that of SARS-CoV-2 is global.
- SARS-CoV severe acute respiratory syndrome coronavirus
- COVID-19 like flu, could become seasonal and may recur in the future even after recovery.
- the global pandemic of COVID-19 has prompted the current interest in the pursuit of immune therapies against SARS-CoV-2. It is desirable to develop novel and effective immune therapies such as vaccines and antibody therapeutics for coronavirus infections.
- the present invention relates to an isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof.
- SARS-CoV-2 nucleocapsid protein comprises or consists of:
- Another aspect of the present invention includes an isolated nucleic acid molecule encoding the SARS-CoV-2 nucleocapsid protein (N-protein) and/or an immunogenic fragment thereof.
- the SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may be for use as a medicament.
- the SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may be for use as a vaccine.
- the invention includes an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof.
- the immunogenic fragment thereof is also capable of binding to SARS-CoV-2 N- protein.
- the antibody binds to SARS-CoV-2 N-protein.
- the antigenic-binding fragment also binds to the SARS-CoV-2 N-protein.
- the antibody may be for use as a medicament.
- Figure 1 shows the antibody responses to vaccination of nucleocapsid protein in four Balb C mice (A, B, C, D). The whole N protein vaccination was repeated 4 times (2-week interval). Red arrow indicates the time points for vaccination. Blood samples were taken before vaccination followed by every 2 weeks until 22 nd week. Serum antibodies were detected by using anti-lgM, -lgG1 , -lgG2 and anti IgG Fc horseradish peroxidase (HRP) conjugated antibodies. Antibody responses can be detected after 2 nd vaccination and sustained till last sample collection in mouse#1 (A), #2 (B), #3 (C) and #4 (D).
- HRP horseradish peroxidase
- Figure 2 shows the sequences of peptides which were selected based on N-protein sequence.
- B anti-N polyclonal Abs (at 1: 1000 & 1 :2000 dilutions) were tested by Elisa for the binding affinity to three individual N-peptides, whole N protein as controls, which were coated respectively with 5ng & 20ng/well, detected by anti-mouse IgG Fc (HRP). The Optical Density (OD) was measured.
- anti-N polyclonal Abs bind not only whole N protein, but also enriched binding to Peptide#3, the highest OD compared to Peptide#1 and #2.
- C Peptide#3 was used to vaccine BALB/c mice in 2-week interval, 3 repeats.
- CD45*CD3*CD335 ' CD4*CD8 ⁇ T cells from the blood of Balb/c mice. Mice were either unvaccinated (WT) or vaccinated with Freund’s adjuvant and N protein (vaccinated mice).
- B Change in the percentage of live CD44 + CD62L ' memory T cells as a proportion of total live CD45*CD3*CD335 ‘ CD4*CD8 ⁇ T cells in unvaccinated and vaccinated mice.
- C Change in the percentage of live CD44 " CD62L* naive T cells as a proportion of total live CD45*CD3*CD335 CD4*CD8 ⁇ T cells in unvaccinated and vaccinated mice.
- the orange box indicates the cytokines which increased more than 2 folds than pre- immunization sample.
- B Map of cytokine array.
- C The table indicating fold increases in cytokine level based on pre-immunization sample.
- D Bar graph of cytokines with more than 2 folds increase compared to pre-immunization sample. Cytokine array performed for wild type(pre), 4 weeks treated (N4), and 12 weeks treated (N12) mice sera.
- FIG. 5 Clone 6H3 mouse monoclonal antibody binds to SARS-CoV-2 N- protein with good affinity.
- ELISA was done to analyzed the binding affinity of peptides & N-protein (SARS-CoV2) to in house produced mouse SARS-CoVAb (clone 6H3).
- ELISA plate was coated with 5ng & 20ng/ well of different peptides & N-protein (SARS-CoV2).
- mouse 6H3 antibodies were diluted at 1 :1000 & 1 :5000 dilution. The binding of antibody was detected by anti-mouse IgG (HRP). The Optical Density (OD) was measured.
- adjuvant refers to any substance or combination of substances which non-specifically enhances the immune response to an antigen.
- the term “comprising” or “including” is to be interpreted as specifying the presence of the stated features, integers, steps or components as referred to, but does not preclude the presence or addition of one or more features, integers, steps or components, or groups thereof.
- the term “comprising” or “including” also includes “consisting of.
- the variations of the word “comprising”, such as “comprise” and “comprises”, and “including”, such as “include” and “includes”, have correspondingly varied meanings.
- An immunogenic fragment is defined as a part of an antigen which is capable of inducing/eliciting an immune response in a host.
- An immunogenic fragment of a protein/polypeptide preferably comprises one or more epitopes of said protein/polypeptide.
- An epitope of a protein/polypeptide is defined as a fragment of said protein/polypeptide of at least about 4 or 5 amino acids in length, capable of eliciting a specific antibody and/or an immune cell (e.g., a T cell or B cell) bearing a receptor capable of specifically binding said epitope.
- a linear epitope comprises a stretch of consecutive amino acids.
- a conformational epitope is typically formed by several stretches of consecutive amino acids that are folded in position and together form an epitope in a properly folded protein.
- An immunogenic fragment as used herein refers to either one, or both, of said types of epitopes.
- the term “vaccine” refers to a composition comprising an antigen capable of stimulating an immune response specifically against the antigen and preferably to engender immunological memory that leads to mounting of a protective immune response should the subject encounter that antigen at some future time.
- the present invention relates to an isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof.
- SARS-CoV-2 N-protein comprises or consists of:
- Another aspect of the present invention includes an isolated nucleic acid molecule encoding the SARS-CoV2 N-protein and/or an immunogenic fragment thereof.
- the SARS-CoV-2 N-protein and/or immunogenic fragment thereof may be prepared by recombinant DNA technology or chemically synthesised.
- the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may also be prepared by recombinant DNA technology or chemically synthesised.
- a further aspect of the invention includes a vector comprising a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof.
- the invention further includes a host cell comprising a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof.
- the invention also includes a host cell comprising a vector comprising a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof.
- the SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may be for use as a vaccine.
- the immunogenic fragment of the SARS-CoV-2 N-protein comprises or consists of a sequence selected from the group consisting of:
- the immunogenic fragment of the SARS-CoV-2 N-protein comprises SEQ ID NO: 4. More in particular, the immunogenic fragment of the SARS-CoV-2 N-protein consists of SEQ ID NO: 4.
- the invention includes an immunogenic combination and/or immunogenic composition comprising two or more components as described herein according to any aspect of the invention. It will be appreciated that the components of an immunogenic combination are administered in combination, for example, they may be combined together before administration or may be administered simultaneously or sequentially.
- the immunogenic combination and/or immunogenic composition may comprise any two or more components selected from the group consisting of:
- N-protein an isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); (ii) the immunogenic fragment(s) as described herein.
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the immumogenic combination and/or immunogenic composition may comprise two or more immunogenic fragments as described herein.
- the invention includes the use of SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof in the preparation of a vaccine.
- the invention includes a pharmaceutical composition comprising a SARS-CoV- 2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof.
- the pharmaceutical composition may comprise at least one pharmaceutically acceptable excipient.
- the invention further includes a vaccine comprising a SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof.
- the vaccine may also comprise at least one pharmaceutically acceptable excipient.
- the vaccine may further comprise at least one adjuvant.
- the vaccine may be for immunizing a subject against a viral infection.
- the invention includes a method for immunizing a subject against a viral infection, comprising administering to the subject the isolated SARS-CoV-2 N- protein and/or immunogenic fragment thereof, the immunogenic combination and/or the immunogenic composition and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment and/or the vector; as described herein.
- the viral infection may be a Coronavirus infection.
- the vaccine may be for immunising against severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS- CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
- the vaccine is for immunising against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
- the invention includes an antibody capable of binding to the SARS-CoV-2 N-protein and/or an immunogenic fragment thereof or an antigen-binding fragment of the antibody.
- the immunogenic fragment thereof is also capable of binding to SARS-CoV-2 N-protein.
- the antibody binds to SARS-CoV-2 N-protein.
- the antigenic-binding fragment also binds to the SARS-CoV-2 N-protein.
- the antibody capable of binding to the to the SARS-CoV-2 N-protein may be a monoclonal antibody.
- the monoclonal antibody may be a chimeric or humanised antibody. It will be appreciated that the monoclonal antibody may be produced by any method, for example hybridoma technology or recombinant DNA technology.
- the antibody comprises a heavy chain comprising SEQ ID NO: 5:
- the regions of the heavy chain are arranged in the following order Signal oeotide-FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 ⁇ with the framework regions FR1 , FR2 and FR3 in bold and the complementary regions CDR1 , CDR2 and CDR3 underlined.
- the antibody comprises a heavy chain comprising a variable region comprising SEQ ID NO: 6:
- the antibody comprises a heavy chain comprising a CDR region 1 (CDR1) comprising NYGMN (SEQ ID NO: 7), a CDR region 2 (CDR2) comprising WINTYTGEPTYADDFKG (SEQ ID NO: 8), and a CDR region 3 (CDR3) comprising PLYYDYDGHAMDY (SEQ ID NO: 9).
- CDR1 comprising NYGMN (SEQ ID NO: 7)
- CDR2 comprising WINTYTGEPTYADDFKG
- CDR3 CDR region 3
- PLYYDYDGHAMDY SEQ ID NO: 9
- the invention includes an isolated nucleic acid molecule encoding the heavy chain of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- the isolated nucleic acid molecule encoding the heavy chain of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) comprises SEQ ID NO: 10:
- nucleotide regions encoding the regions of the heavy chain are shown in the following order, Signal sequence-FR1-CDR1- FR2-CDR2-FR3-CDR3-FR4 with framework regions FR1 , FR2 and FR3 in bold and the complementary regions CDR1, CDR2 and CDR3 underlined.
- the invention also includes an isolated nucleic acid molecule encoding the heavy chain variable region of an antibody capable of binding to the isolated N- protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the invention includes an isolated nucleic acid molecule encoding a heavy chain antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) comprising a variable region comprising SEQ ID NO: 6.
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the invention includes an isolated nucleic acid molecule encoding a heavy chain antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) comprising a CDR region 1 (CDR1) comprising NYGMN (SEQ ID NO: 7), a CDR region 2 (CDR2) comprising WINTYTGEPTYADDFKG (SEQ ID NO: 8), and a CDR region 3 (CDR3) comprising PLYYDYDGHAMDY (SEQ ID NO: 9).
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the invention includes an isolated nucleic acid encoding the light chain of an antibody as described herein.
- the invention includes an isolated nucleic acid molecule encoding the light chain of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the invention also includes an isolated nucleic acid molecule encoding the light chain variable region of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- the antibody and/or antigenic fragment thereof may be for use as a medicament.
- the antibody may be formulated into a pharmaceutical composition.
- the pharmaceutical composition may comprise at least one pharmaceutically acceptable excipient.
- the invention includes the antibody and/or antigen-binding fragment thereof for use in treating a viral infection.
- the invention also includes the use of the antibody and/or antigen-binding fragment thereof as described herein in the preparation of a pharmaceutical composition for treating a viral infection.
- the invention further includes a method for treating a viral infection in a subject comprising administering the antibody and/or antigenic fragment as described herein thereof to the subject.
- the viral infection may be a Coronavirus infection.
- the viral infection may be severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
- SARS-CoV severe acute respiratory syndrome coronavirus
- MERS-CoV Middle East respiratory syndrome coronavirus
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- Example 1 Vaccination of mice with SARS-CoV-2 nucleocapsid protein (N- protein) or immunogenic peptides derived from SARS-CoV-2 nucleocapsid protein (N-protein) Materials and methods
- SARS-CoV-2 GST-N protein bacterial clone was kindly supplied by Dr Yee Joo TAN (Monoclonal Antibody Unit, IMCB, A*STAR, Singapore).
- Bacterial clone was inoculated in 5 mL of Luria-Bertani (LB media) with 100 ⁇ g/mL ampicillin and cultured overnight, added to 500 mL of Luria-Bertani/ampicillin and grown until its OD reached 0.6 to 0.8 at A600 nm.
- Isopropyl-L-thio-h-D- galactopyranoside was added to the culture at 0.5 mmol/L/mL and the culture was shaken overnight at room temperature.
- the culture was then centrifuged at 5,000 rpm for 10 minutes.
- the pellet was in 25 mL GST extraction buffer [1 mg/mL lysozyme, 5 mmol/L DTT and 0.5 mmol/L phenylmethylsulfonyl fluoride in GST buffer PBS, 50 mmol/L Tris (pH 8) and 0.5 mmol/L MgCI2],
- the lysate was incubated on ice for 15 minutes and sonicated for 5 minutes followed by centrifugation at 15,000 rpm for 30 minutes at 4°C.
- the supernatant was passed through a 0.45- ⁇ filter.
- GST-Nucleocapsid protein (GST-N- protein) was purified from GST-N protein bacterial clone. Nucleocapsid peptide with or without KLH tagged, Peptide#1 , Peptide#2, Peptide#3 were synthesized by Genemed Synthesis, Inc (USA). GST-N-protein (75ug), KLH-Peptide#3 (50ug) and Peptide#3 vaccine (20ug) in 100 ul of PBS were mixed thoroughly with 100 ⁇ L of Freund’s adjuvant (complete adjuvant for 1 st immunization and incomplete adjuvant for subsequent immunization, Pierce).
- Freund’s adjuvant complete adjuvant for 1 st immunization and incomplete adjuvant for subsequent immunization, Pierce).
- mice were immunized by intraperitoneal injection of each vaccine in 2 week intervals for 3 or 4 times.
- the vaccine without any protein or peptide (Adjuvant only) was injected in a group of mice as control.
- Blood samples (20 ⁇ L each time) were taken by tail bleed in Eppendorf tube, and serum was prepared. The antibody titer was measured by ELISA.
- the peptides are:
- Peptide#1 (P1): CIRQGTDYKHWPQIAQFAPSASAFFGMSRIG (SEQ ID NO: 2)
- Peptide#2 (P2): CIAQFAPSASAFFGMSRIGMEVTPSGTWLTY (SEQ ID NO: 3)
- Peptide#3 (P3): CVILLNKHIDAYKTFPPTEPKKDKKKKADET (SEQ ID NO: 4) Preparation of serum samples for Elisa:
- Anti-N Protein IgM antibody was detected by goat anti-mouse IgM-HRP Antibody (Invitrogen 626820). Subtype of anti N Protein IgG, lgG2a & lgG1, were detected by goat anti-mouse lgG2a-HRP (Invitrogen A10685) & goat antimouse lgG1-HRP (Invitrogen A10551) secondary antibodies. Anti N-protein IgG (whole IgG) were detected by goat anti-mouse IgG-HRP (H+L) secondary antibody (Invitrogen 31430).
- TMB tetramethylbenzidine
- Pierce peroxidase substrate
- OD Optical Density
- Anti-N Protein IgM antibody was detected by goat anti-mouse IgM-HRP Antibody (Invitrogen 626820). Subtype of anti N Protein IgG, lgG2a & lgG1 , were detected by goat anti-mouse lgG2a-HRP (Invitrogen A10685) & goat antimouse lgG1-HRP (Invitrogen A10551) secondary antibodies. Anti N-protein IgG (whole IgG) were detected by goat anti-mouse IgG-HRP (H+L) secondary antibody (Invitrogen 31430).
- TMB tetramethylbenzidine
- Pierce peroxidase substrate
- OD Optical Density
- mice Whole blood from mice was lysed with ACK Lysing Buffer (Gibco, A1049201) for 10 min at RT to remove RBCs. The remaining single-cell suspensions were then stained with Zombie UV Fixable Viability dye (BioLegend) for 30 min at 4 °C, approximately 300,000 - 500,000 cells were used per stain.
- ACK Lysing Buffer Gibco, A1049201
- Zombie UV Fixable Viability dye BioLegend
- Non-specific labelling was blocked with anti-CD16/32 (clone 2.4G2; BD Biosciences) for 30 min at 4 °C before multiplex labelling for 30 min at 4 °C with the following antibodies from BioLegend: Brilliant Violet 711 anti-mouse CD3e (clone 145- 2C11), PE-Cy7 anti-mouse CD4 (clone RM4-5), Brilliant Violet 786 anti-mouse CD8a (clone 53-6.7), PE/Dazzle 594 anti-mouse CD11b (clone M1/70), APC- Cy7 anti-mouse CD19 (clone 6D5), Brilliant Violet 510TM anti-mouse CD25 (clone PC61), AF488 anti-mouse CD45 (clone 30-F11), APC anti-mouse CD69 (clone H1.2F3) PE/Dazzle 594 anti-mouse CD127 (clone A7R34), Brilliant Violet 421 anti-mouse CD
- BV711 Anti-Mouse CD3e (clone 145-2C11), APC-Cy7 Rat Anti-Mouse CD19 (clone 1 D3), PE Anti-Mouse CD44 (clone IM7), FITC Anti-Mouse CD45 (clone 30- F11), BV650 Anti-Mouse CD62L (clone MEL-14), PE-CF594 Anti-Mouse CD80 (clone 16-10A1), BV786 Anti-Mouse CD138 (clone 281-2), BV510 Anti-Mouse CD273 (clone TY25), APC Anti-Mouse IgD (clone 11-26c.2a). All samples were run on a BD LSR II flow cytometer and analysed using the FlowJo software 10.5.3 (FlowJo).
- N-Protein vaccination can produce high and sustainable anti N-protein different antibody subtypes
- Immunization of N-protein vaccine was done in BALB/c mice and the antibody response at different time intervals was analyzed. Serum IgM, IgG and the subclasses lgG1 and lgG2a were measured using ELISA to evaluate the profile of the immune response. lgG1 indicates a humoral immune response, whereas lgG2a indicates a cellular immune response.
- Antibody production was not detected after 1 st dose of vaccination.
- IgM antibody was detected after 2 nd vaccination, but it stays at the plateau phase, and at a sustainable level throughout the time point.
- IgG and its subclasses lgG1 & lgG2a antibodies can also be detected after the 2 nd dose of vaccination. These antibodies gradually increase upon subsequent vaccination and remain in the plateau phase throughout the time points. Peak antibody responses were detected after the 4 th dose of vaccination in each BALB/c mouse ( Figure 1A, B, C, D).
- the second dose of vaccine significantly boosted the concentrations of all antibody subtypes, IgM (p-value ⁇ 0.001), lgG2a (p-value ⁇ 0.001), lgG1 (p-value ⁇ 0.0001) and IgG (p-value ⁇ 0.0001) comparison between 2 weeks (2weeks after 1 st vaccination) and 4 weeks (2 weeks after 2 nd vaccination) time point (Figure 1 E).
- mice vaccinated with adjuvant only All types of antibodies were not detected in the mice vaccinated with adjuvant only.
- the second dose of vaccine elicited a greater increase in the lgG1 antibody concentration than in the lgG2a antibody concentration, which resulted in a lower lgG2a/lgG1 ratio compared to the ratio observed after the first vaccination.
- mice were immunized in another mouse species, FVB mice.
- the same trend of antibody production was detected in FVB mice also ( Figure 1 F).
- mice vaccinated with whole N protein produced high titer of specific anti-N antibody subtypes: IgM (p-value ⁇ 0.045), IgG (p-value ⁇ 0.0037), lgG1(p- value ⁇ 0.045), lgG2a (p-value ⁇ 0.029), suggesting that N-protein is an excellent antigen with high immunogenicity to evoke a protective immunity and produce anti-N specific antibodies at high titers.
- Peptides selected on N-protein sequence could bind specifically to anti-N protein antibody
- N protein vaccination resulted in a high and sustained production of different IgG subtypes in mice, indicating that N-protein is a good vaccine candidate.
- N-protein sequences (419 amino acid sequence)
- We selected 3 different peptides: peptide 1 , peptide 2, and peptide 3 that are derived from N-protein, and 3 N-protein peptides were synthesized, 30 amino acids in the length of each peptide (Figure 2A) with or without KLH carrier.
- the OD of Peptide #3 (with or without KLH carrier) were nearly 2/3 of N-protein OD at dilution step 10.
- the OD of other peptides, peptide #1 and Peptide #2 were nearly 10 times lower than peptide #3, indicating that Peptide 3 is the best candidate to represent N protein epitope to produce antibody which is specific to N-protein, and the peptide#3 could potentially be combined with the cocktail of traditional Influenza vaccine to be a general safe vaccine.
- Peptide vaccination could induce high and sustainable antibody production similar to N protein vaccine.
- 3 peptides synthesized most polyclonal anti-N antibodies react highly with peptide#3, but not peptide 1 and peptide 2, suggesting that peptide 3 alone could be a good immunogen for vaccination to evoke host immune system to produce antibody specific to N protein.
- Freund adjuvant By mixing with Freund adjuvant, the immunization was done on BALB/c mice for 3 times in a 2-week interval. The blood collection (20 ⁇ L each) was done before each immunization and every 2 weeks after immunization.
- mice 8-week-old BALB/c mice were vaccinated once weekly with a combination of Freund’s adjuvant and N protein (Vaccinated mice) for four weeks. Vaccinated mice were then bled and sacrificed eight weeks after the last vaccination to determine if they had elevated levels of memory cells compared to unvaccinated mice.
- T cells may be more important for mediating long term immunity against SARS-CoV-2 and may explain why some former SARS-CoV-2 patients experience a decline in antibodies several months after recovery [Marot et al., (2021); Self et al., 2020], Example 2: Cvtokine production in vaccinated mice
- Mouse serum cytokines from unvaccinated and vaccinated mice were analysed with the RayBiotech mouse cytokine array C1 (Cat: #AMM-CYT-1-8) using the provided experimental protocol unless otherwise indicated.
- blots were blocked with 2m L of provided blocking buffer and incubated for 30m ins at room temperature. 6pL of each serum sample was diluted to a total volume of 500 ⁇ L with blocking buffer and arrays were incubated overnight at 4°C with dilute serum samples. Arrays were then washed with provided washing buffers according to the standard protocol. Next arrays were incubated with 500 ⁇ L of pre-diluted biotinylated antibody cocktail for 4hrs at room temperature.
- Arrays were then washed with provided washing buffers according to the standard protocol. Arrays were then incubated with 500 ⁇ _ of x1 HRP-Streptavidin for 2hrs at room temperature. Arrays were washed and incubated with detection buffers for chemiluminescence detection.
- Vaccination N protein with complete Freund’s adjuvant can induce the secretion of pro-inflammatory memory cell and TH1 associated cytokines.
- N-protein hybridoma clone generated by fusion of splenocytes from N -protein immunized BALB/c mice and BALB/c parental myeloma SP2/0 cells was kindly provided by Dr Yee Joo TAN (Monoclonal Antibody Unit, IMCB, A*STAR, Singapore).
- Hybridoma cells (5 x 10 5 ) were suspended in 200 ⁇ L of Phosphate Buffered Saline (PBS) and injected into the peritoneal cavity and wait until the mouse developed a large quantity of ascitic fluid. The mouse was sacrificed and ascitic fluid was collected, centrifuged and frozen at -70°C until further use.
- PBS Phosphate Buffered Saline
- Hybridoma cells (5 x 10 6 ) were suspended in 200 L of serum-free DMEM medium and injected with a 26-gauge needle into the peritoneal cavity to BALB/c mice. After 10 days, the mouse developed a large quantity of ascitic fluid, and the abdomen was greatly distended. The mouse was sacrificed and a small shallow was cut to open the abdominal cavity. The ascitic fluid was drawn with a 10-mL syringe fitted with an 18-gauge needle. The fluid was centrifuged at 200 x g for 10 minutes at 4°C. The supernatant fluid was collected and frozen at -70°C until further use.
- Mouse SAR-CoV-N-protein antibody could not bind well to Peptide #1 , Peptide #2, and Peptide #3 but bind strongly to SARS-CoV2 N-protein in a concentration dependent manner ( Figure 5), suggesting clone 6H3 epitope presents in the whole N protein but not in all 3 peptides.
- the mouse antibody (6H3) can be developed for the First in Class humanized antibody to treat patients infected with coronavirus.
- Severe Acute Respiratory Syndrome Coronavirus 2 (SAR-CoV-2) caused the global pandemic of the Coronavirus disease in late 2019 (COVID-19).
- Vaccine development efforts have predominantly been aimed at ‘Extra-viral’ Spike (S) mRNA as vaccine vehicles but there are concerns regarding ‘viral immune escape’ since multiple mutations may enable the mutated virus strains to escape from immunity against S protein.
- the ‘Intra-viral’ Nucleocapsid (N- protein) is relatively conserved among mutant strains of coronaviruses during spread and evolution.
- N- protein is relatively conserved among mutant strains of coronaviruses during spread and evolution.
- mice vaccinated with N protein/or N protein fragment/peptides had sustainably high titers of anti-N antibodies (IgG, lgG1 , lgG2a, IgM).
- vaccination with peptide #3 gave similar results as that of the whole N protein, suggesting that peptide #3 is not only the major epitope in the N-protein but also sufficient to elicit protective immunity in the host.
- mice immunized with S protein exhibited reduced viral titers after vaccination, in contrast to vaccination with other SARS-CoV structural proteins, such as the N, membrane (M), and envelop (E) proteins [Yasui etal., (2008)].
- SARS-CoV structural proteins such as the N, membrane (M), and envelop (E) proteins
- SARS-CoV-2 Similar to the SARS coronavirus (SARS-CoV), SARS-CoV-2 infects target cells via spike protein receptor binding domain (RBD) and ACE2 receptor interactions [Hoffmann et al., (2020); Zhou etal., (2020)]. To generate effective neutralizing antibodies to block SARS-CoV-2 viral entry, the SARS-CoV-2 spike protein and its RBD were selected as the leading target antigens in vaccine development [Chen et al., (2020); Pang et al., (2020)].
- RBD spike protein receptor binding domain
- ACE2 receptor interactions Hoffmann et al., (2020); Zhou etal., (2020)
- SARS-CoV-2 spike protein and its RBD were selected as the leading target antigens in vaccine development [Chen et al., (2020); Pang et al., (2020)].
- the SARS-CoV-2 S gene also has a relatively lower amino acid similarity (76%) compared to the SARS-CoV S gene with a higher rate of mutation compared to the more conserved (90)% N gene [Dutta et al., (2020); Grifoni et al., (2020a); Marra et al., (2003); Drosten et al., (2003); Zhu et al., (2005)].
- This data suggests that while anti-S protein antibodies may be key for controlling viral litres during an ongoing infection, other immune mediators may be responsible for conferring long-term immunity to SARS-CoV-2.
- N protein is highly immunogenic and is the most abundant viral protein during coronavirus infections [Dai et al., (2021); Long et al., (2020)]. It is also a major target for antibody and T cell responses [Sariol and Perlman (2020)]. Importantly, non-neutralizing antibodies against N protein can protect mice against some other viruses, such as the mouse hepatitis virus [Nakanaga et al., (1986); Lecomte et al., (1987)] and influenza A virus [Fujimoto et al., (2016)]. N protein is also commonly externalized on the cell surface membrane of infected cells, and can act as a potential target for both antibody and T cell responses [Fujimoto et al., (2016)].
- memory T cells may play a critical role in conferring long-term immunity to SARS-CoV-2.
- Grifoni et al (2020b) reports the induction of robust CD4* and CD8* T cells in convalescent SARS-CoV-2 patients. Surprisingly even some non-exposed individuals demonstrate T cell reactivity against SARS-CoV-2 epitopes, suggesting that prior infections in these individuals could also enhance immunity against SARS-CoV-2 [Grifoni et al (2020b)].
- Le Bert et al also demonstrates that former SARS-CoV patients possess long lasting memory T cells which are reactive to N protein over 17 years after the SARS epidemic in 2003 [Le Bert et al, (2020)].
- N protein vaccination provides sustainably long protective immunity.
- Much emphasis has been placed on the extra-viral spike (S) protein in vaccine development. This is due to its importance in the detection by host immune system and viral entry into host cells.
- S protein extra-viral spike
- Intra-viral nucleocapsid (N) protein is more conserved [Surjit et al., (2008)] in sequence.
- N protein can be detected by the host immune system as there is a presence of anti-N protein antibodies in the sera of SARS-CoV-2 infected patients [Burbelo et al., (2020b)].
- anti-N protein antibodies On the potential of anti-N protein antibodies in the prevention of infection, dominant helper T-cell epitopes in the N protein of SARS-CoV have been identified to assist in antiviral neutralizing antibody production [Zhao et al., (2007).
- the anti-N protein antibodies have been previously shown to confer protection against several types of lethal influenza A viruses [La Mere et al., (2011); Fujimoto et al., (2016); Carragher et al., (2008)].
- a combination of neutralizing antibodies targeting S protein and its RBD, anti-N protein antibodies, and memory T cells against N protein epitopes may be essential to confer long-term protection against SARS-CoV-2.
- SARS-CoV nucleocapsid protein a protein with multifarious activities. Infection, genetics and evolution : journal of molecular epidemiology and evolutionary genetics in infectious diseases, 2008. 8(4): p. 397-405.
- SARS-CoV severe acute respiratory syndrome-associated coronavirus
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Virology (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biochemistry (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- Pulmonology (AREA)
- Gastroenterology & Hepatology (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Peptides Or Proteins (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
The present invention relates to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) N-protein and/or an immunogenic fragment thereof and uses thereof. The invention also includes a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or an immunogenic fragment thereof; and uses thereof. The SARS-CoV-2 N protein and/or immunogenic fragment thereof may be produced by recombinant DNA technology or may be chemically synthesised. In particular, the SARS-CoV-2 N-protein and/or an immunogenic fragment thereof and/or nucleic acid molecule encoding the SARS-CoV-2 N-protein and immunogenic may be for use as a vaccine. The invention further includes an antibody capable of binding to the SARS-CoV-2 N-protein or antigen-binding fragment thereof and uses thereof.
Description
Vaccine and/or antibody for viral infection
Field of the invention
The present invention relates to prophylaxis and treatment of viral infection. In particular, the invention relates to immune therapies such as novel vaccines for prophylaxis and antibodies for treatment of viral infection, for example Coronavirus infection.
Background of the invention
Coronaviruses (CoVs) are enveloped, positive-sense, single-stranded RNA viruses of the family Coronaviridae. While most viruses cause mild illnesses such as the common cold, a few viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV) resulted in the severe acute respiratory syndrome (SARS) public health crises in 2003, Middle East respiratory syndrome coronavirus (MERS-CoV) caused Middle East respiratory syndrome (MERS) in 2009. In addition, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused Coronavirus disease 2019 (COVID-19) from late 2019. The outbreaks for SARS-CoV and MERS-CoV were regional, while that of SARS-CoV-2 is global. The World Health Organisation (WHO) declared COVID-19 as a pandemic on 11th March 2020 and SARS-CoV-2 has infected almost 128 million people and caused over 2.8 million deaths worldwide as of 3rd April 2021 , with severe outbreaks occurring in first in China, then Europe and in the USA (WHO Coronavirus (COVID-19) Dashboard). While infections are generally self-resolving in healthy subjects, it can also lead to severe pneumonia, multi-organ failure, and death in significant portions of infected patients, especially those with pre-existing comorbidities. Along with drastic social distancing measures in an attempt to slow the spread of the virus, the current COVID-19 pandemic has caused widespread medical, social, political, and financial repercussions. There are predictions that COVID-19, like flu, could
become seasonal and may recur in the future even after recovery. The global pandemic of COVID-19 has prompted the current interest in the pursuit of immune therapies against SARS-CoV-2. It is desirable to develop novel and effective immune therapies such as vaccines and antibody therapeutics for coronavirus infections.
Summarv of the invention
According to a first aspect, the present invention relates to an isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof. The sequence of the SARS-CoV-2 nucleocapsid protein (N-protein) comprises or consists of:
Another aspect of the present invention includes an isolated nucleic acid molecule encoding the SARS-CoV-2 nucleocapsid protein (N-protein) and/or an immunogenic fragment thereof.
The SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may be for use as a medicament.
The SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may be for use as a vaccine.
According to a further aspect, the invention includes an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof. The immunogenic fragment thereof is also capable of binding to SARS-CoV-2 N- protein. In particular, the antibody binds to SARS-CoV-2 N-protein. The antigenic-binding fragment also binds to the SARS-CoV-2 N-protein.
The antibody may be for use as a medicament.
Brief description of the figures
Figure 1 shows the antibody responses to vaccination of nucleocapsid protein in four Balb C mice (A, B, C, D). The whole N protein vaccination was repeated 4 times (2-week interval). Red arrow indicates the time points for vaccination. Blood samples were taken before vaccination followed by every 2 weeks until 22nd week. Serum antibodies were detected by using anti-lgM, -lgG1 , -lgG2 and anti IgG Fc horseradish peroxidase (HRP) conjugated antibodies. Antibody responses can be detected after 2nd vaccination and sustained till last sample collection in mouse#1 (A), #2 (B), #3 (C) and #4 (D). (E) Mean data of antibody productions in the BALB/c mice (n=4) Mean antibody production of N protein vaccination in FVB mice (n=3). Data represent Mean ±S.D. (F) shows the same trend in FVB mice vaccinated with N protein. Figure 1 shows that N protein is an excellent immunogen for vaccination.
Figure 2: (A) shows the sequences of peptides which were selected based on N-protein sequence. (B) anti-N polyclonal Abs (at 1: 1000 & 1 :2000 dilutions) were tested by Elisa for the binding affinity to three individual N-peptides, whole N protein as controls, which were coated respectively with 5ng & 20ng/well, detected by anti-mouse IgG Fc (HRP). The Optical Density (OD) was measured. anti-N polyclonal Abs bind not only whole N protein, but also enriched binding to Peptide#3, the highest OD compared to Peptide#1 and #2. (C) Peptide#3 was used to vaccine BALB/c mice in 2-week interval, 3 repeats.
Red arrow indicates each vaccine time point. (D) Anti-Peptide#3 Ab serum were classified by using anti-lgM, -lgG1 , -lgG2a and -IgG Fc horseradish peroxidase (HRP) conjugated antibodies. Data represent Mean ±S.D, n=3. (E) Anti- Peptide#3 Ab serum were tested by Elisa for the binding capacity. Anti- Peptide#3 Ab serum binds to Peptide#3 and whole N protein, detected by antimouse IgG Fc (HRP). The Optical Density (OD) was measured. Figure 2 shows that Peptide#3 is a good immunogen.
Figure 3. Vaccination results in an increased frequency of CD4* & CD8* memory T cells and a decreased frequency of memory T cells. (A) Representative CD62L and CD44 staining on live
CD45*CD3*CD335'CD4*CD8· T cells from the blood of Balb/c mice. Mice were either unvaccinated (WT) or vaccinated with Freund’s adjuvant and N protein (vaccinated mice). (B) Change in the percentage of live CD44+CD62L' memory T cells as a proportion of total live CD45*CD3*CD335‘CD4*CD8· T cells in unvaccinated and vaccinated mice. (C) Change in the percentage of live CD44" CD62L* naive T cells as a proportion of total live CD45*CD3*CD335 CD4*CD8· T cells in unvaccinated and vaccinated mice. (D) Representative CD62L and CD44 staining on live C D45*C D3*C D335 C D4 C D8* T cells from the blood of Balb/c mice. (E) Change in the percentage of live CD44*CD62I_· memory T cells as a proportion of total live CD45*CD3*CD335'CD4‘CD8* T cells in unvaccinated and vaccinated mice. (F) Change in the percentage of live CD44" CD62L* naive T cells as a proportion of total live CD45*CD3*CD335 CD4 CD8* T cells in unvaccinated and vaccinated mice. (G) Representative IgD and IgG staining on live CD45*CD19*CD138' B cells from the blood of unvaccinated and vaccinated Balb/c mice. Change in the percentage of (H) naive IgD* B cells and (I) IgG* class-switched memory B cells. Data representing meant SEM. n=4 in BALB/c mice & n=3 in FVB mice.
Figure 4. Vaccination with whole N protein in complete Freund’s adjuvant (CFA) can induce the secretion of pro-inflammatory memory cell and TH1 associated cytokines. (A) Cytokine array blot of pre- & post-immunization mouse sera. The orange box indicates the cytokines which increased more than 2 folds than pre- immunization sample. (B) Map of cytokine array. (C) The table indicating fold increases in cytokine level based on pre-immunization sample. (D) Bar graph of cytokines with more than 2 folds increase compared to pre-immunization sample. Cytokine array performed for wild type(pre), 4 weeks treated (N4), and 12 weeks treated (N12) mice sera.
Figure 5. Clone 6H3 mouse monoclonal antibody binds to SARS-CoV-2 N- protein with good affinity. ELISA was done to analyzed the binding affinity of peptides & N-protein (SARS-CoV2) to in house produced mouse SARS-CoVAb (clone 6H3). ELISA plate was coated with 5ng & 20ng/ well of different peptides & N-protein (SARS-CoV2). mouse 6H3 antibodies were diluted at 1 :1000 & 1 :5000 dilution. The binding of antibody was detected by anti-mouse IgG (HRP). The Optical Density (OD) was measured.
Definitions
As used herein, the term “adjuvant” refers to any substance or combination of substances which non-specifically enhances the immune response to an antigen.
As used herein, the term “comprising” or “including” is to be interpreted as specifying the presence of the stated features, integers, steps or components as referred to, but does not preclude the presence or addition of one or more features, integers, steps or components, or groups thereof. However, in context with the present disclosure, the term “comprising” or “including” also includes “consisting of. The variations of the word “comprising”, such as “comprise” and “comprises”, and “including”, such as “include” and “includes”, have correspondingly varied meanings.
An immunogenic fragment is defined as a part of an antigen which is capable of inducing/eliciting an immune response in a host. An immunogenic fragment of a protein/polypeptide preferably comprises one or more epitopes of said protein/polypeptide. An epitope of a protein/polypeptide is defined as a fragment of said protein/polypeptide of at least about 4 or 5 amino acids in length, capable of eliciting a specific antibody and/or an immune cell (e.g., a T cell or B cell) bearing a receptor capable of specifically binding said epitope. Two different kinds of epitopes exist: linear epitopes and conformational epitopes. A linear epitope comprises a stretch of consecutive amino acids. A conformational epitope is typically formed by several stretches of consecutive amino acids that are folded in position and together form an epitope in a properly folded protein. An immunogenic fragment as used herein refers to either one, or both, of said types of epitopes.
As used herein, the term “vaccine” refers to a composition comprising an antigen capable of stimulating an immune response specifically against the antigen and preferably to engender immunological memory that leads to mounting of a protective immune response should the subject encounter that antigen at some future time.
Detailed description of the invention Bibliographic references mentioned in the present specification are for convenience listed in the form of a list of references and added at the end of the examples. The whole content of such bibliographic references is herein incorporated by reference.
According to a first aspect, the present invention relates to an isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof.
The sequence of the SARS-CoV-2 N-protein comprises or consists of:
Another aspect of the present invention includes an isolated nucleic acid molecule encoding the SARS-CoV2 N-protein and/or an immunogenic fragment thereof.
The SARS-CoV-2 N-protein and/or immunogenic fragment thereof may be prepared by recombinant DNA technology or chemically synthesised. The nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may also be prepared by recombinant DNA technology or chemically synthesised.
A further aspect of the invention includes a vector comprising a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof. The invention further includes a host cell comprising a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof. The invention also includes a host cell comprising a vector comprising a nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof. The SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment may be for use as a vaccine.
The immunogenic fragment of the SARS-CoV-2 N-protein comprises or consists of a sequence selected from the group consisting of:
CIRQGTDYKHWPQIAQFAPSASAFFGMSRIG (SEQ ID NO: 2);
CIAQFAPSASAFFGMSRIGMEVTPSGTWLTY (SEQ ID NO: 3); CVILLNKHIDAYKTFPPTEPKKDKKKKADET (SEQ ID NO: 4).
In particular, the immunogenic fragment of the SARS-CoV-2 N-protein comprises SEQ ID NO: 4. More in particular, the immunogenic fragment of the SARS-CoV-2 N-protein consists of SEQ ID NO: 4.
The invention includes an immunogenic combination and/or immunogenic composition comprising two or more components as described herein according to any aspect of the invention. It will be appreciated that the components of an immunogenic combination are administered in combination, for example, they may be combined together before administration or may be administered simultaneously or sequentially. For example, the immunogenic combination and/or immunogenic composition may comprise any two or more components selected from the group consisting of:
(i) an isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); (ii) the immunogenic fragment(s) as described herein.
In particular, the immumogenic combination and/or immunogenic composition may comprise two or more immunogenic fragments as described herein.
Accordingly, the invention includes the use of SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the
SARS-CoV-2 N-protein and/or immunogenic fragment thereof in the preparation of a vaccine.
The invention includes a pharmaceutical composition comprising a SARS-CoV- 2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof. The pharmaceutical composition may comprise at least one pharmaceutically acceptable excipient.
The invention further includes a vaccine comprising a SARS-CoV-2 N-protein and/or immunogenic fragment thereof and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment thereof. The vaccine may also comprise at least one pharmaceutically acceptable excipient. The vaccine may further comprise at least one adjuvant.
The vaccine may be for immunizing a subject against a viral infection.
The invention includes a method for immunizing a subject against a viral infection, comprising administering to the subject the isolated SARS-CoV-2 N- protein and/or immunogenic fragment thereof, the immunogenic combination and/or the immunogenic composition and/or the nucleic acid molecule encoding the SARS-CoV-2 N-protein and/or immunogenic fragment and/or the vector; as described herein.
The viral infection may be a Coronavirus infection. For example, the vaccine may be for immunising against severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS- CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)]. In particular, the vaccine is for immunising against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
According to a further aspect, the invention includes an antibody capable of binding to the SARS-CoV-2 N-protein and/or an immunogenic fragment thereof or an antigen-binding fragment of the antibody. The immunogenic fragment thereof is also capable of binding to SARS-CoV-2 N-protein. In particular, the antibody binds to SARS-CoV-2 N-protein. The antigenic-binding fragment also binds to the SARS-CoV-2 N-protein.
The antibody capable of binding to the to the SARS-CoV-2 N-protein may be a monoclonal antibody. The monoclonal antibody may be a chimeric or humanised antibody. It will be appreciated that the monoclonal antibody may be produced by any method, for example hybridoma technology or recombinant DNA technology.
According to one embodiment, the antibody comprises a heavy chain comprising SEQ ID NO: 5:
MDWLWNLLFLMAAAQSAQAQIQLVQSGPELKKPGE TVKISCKASGYTFTNYGMNWVKQA PGKGLKWMGWINTYTGEPTYADDFKGRFAFSLE TSASTAYLQINNLKNEDMAKYFCTRP LYYDYDGHAMDYWGQGTSVTVSS
The regions of the heavy chain are arranged in the following order Signal oeotide-FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4· with the framework regions FR1 , FR2 and FR3 in bold and the complementary regions CDR1 , CDR2 and CDR3 underlined.
According to one embodiment, the antibody comprises a heavy chain comprising a variable region comprising SEQ ID NO: 6:
QIQLVQSGPELKKPGETVKISCKASGYT FTNYGMNWVKQAPGKGLKWMGWINTY TGEPTYADDFKGRFAFSLETSASTAYLQINNLKNEDMAKYFCTRPLYYDYDGHA MDYWGQGTSVTVSS
The complementary determining CDR regions are underlined.
According to a further embodiment, the antibody comprises a heavy chain comprising a CDR region 1 (CDR1) comprising NYGMN (SEQ ID NO: 7), a CDR region 2 (CDR2) comprising WINTYTGEPTYADDFKG (SEQ ID NO: 8), and a CDR region 3 (CDR3) comprising PLYYDYDGHAMDY (SEQ ID NO: 9). The invention includes an isolated nucleic acid encoding the heavy chain of an antibody as described herein.
The invention includes an isolated nucleic acid molecule encoding the heavy chain of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In particular, the isolated nucleic acid molecule encoding the heavy chain of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) comprises SEQ ID NO: 10:
In SEQ ID NO: 9 above, the nucleotide regions encoding the regions of the heavy chain are shown in the following order, Signal sequence-FR1-CDR1- FR2-CDR2-FR3-CDR3-FR4 with framework regions FR1 , FR2 and FR3 in bold and the complementary regions CDR1, CDR2 and CDR3 underlined.
The invention also includes an isolated nucleic acid molecule encoding the heavy chain variable region of an antibody capable of binding to the isolated N- protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
The invention includes an isolated nucleic acid molecule encoding a heavy chain antibody capable of binding to the isolated N-protein from severe acute
respiratory syndrome coronavirus 2 (SARS-CoV-2) comprising a variable region comprising SEQ ID NO: 6.
The invention includes an isolated nucleic acid molecule encoding a heavy chain antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) comprising a CDR region 1 (CDR1) comprising NYGMN (SEQ ID NO: 7), a CDR region 2 (CDR2) comprising WINTYTGEPTYADDFKG (SEQ ID NO: 8), and a CDR region 3 (CDR3) comprising PLYYDYDGHAMDY (SEQ ID NO: 9).
The invention includes an isolated nucleic acid encoding the light chain of an antibody as described herein.
The invention includes an isolated nucleic acid molecule encoding the light chain of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
The invention also includes an isolated nucleic acid molecule encoding the light chain variable region of an antibody capable of binding to the isolated N-protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
The antibody and/or antigenic fragment thereof may be for use as a medicament. The antibody may be formulated into a pharmaceutical composition. The pharmaceutical composition may comprise at least one pharmaceutically acceptable excipient.
Accordingly, the invention includes the antibody and/or antigen-binding fragment thereof for use in treating a viral infection.
The invention also includes the use of the antibody and/or antigen-binding fragment thereof as described herein in the preparation of a pharmaceutical composition for treating a viral infection.
The invention further includes a method for treating a viral infection in a subject comprising administering the antibody and/or antigenic fragment as described herein thereof to the subject.
The viral infection may be a Coronavirus infection. For example, the viral infection may be severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
Having now generally described the invention, the same will be more readily understood through reference to the following examples which are provided by way of illustration, and are not intended to be limiting of the present invention.
Examples
Standard molecular biology techniques known in the art and not specifically described were generally followed as described in Green and Sambrook, Molecular Cloning: A Laboratory Manual, Cold Springs Harbor Laboratory, New York (2012).
Example 1: Vaccination of mice with SARS-CoV-2 nucleocapsid protein (N- protein) or immunogenic peptides derived from SARS-CoV-2 nucleocapsid protein (N-protein) Materials and methods
Preparation of GST-N protein:
SARS-CoV-2 GST-N protein bacterial clone was kindly supplied by Dr Yee Joo TAN (Monoclonal Antibody Unit, IMCB, A*STAR, Singapore). Bacterial clone was inoculated in 5 mL of Luria-Bertani (LB media) with 100 μg/mL ampicillin and cultured overnight, added to 500 mL of Luria-Bertani/ampicillin and grown until its OD reached 0.6 to 0.8 at A600 nm. Isopropyl-L-thio-h-D-
galactopyranoside was added to the culture at 0.5 mmol/L/mL and the culture was shaken overnight at room temperature. The culture was then centrifuged at 5,000 rpm for 10 minutes. The pellet was in 25 mL GST extraction buffer [1 mg/mL lysozyme, 5 mmol/L DTT and 0.5 mmol/L phenylmethylsulfonyl fluoride in GST buffer PBS, 50 mmol/L Tris (pH 8) and 0.5 mmol/L MgCI2], The lysate was incubated on ice for 15 minutes and sonicated for 5 minutes followed by centrifugation at 15,000 rpm for 30 minutes at 4°C. The supernatant was passed through a 0.45-μΜ filter. One milliliter of glutathione slurry (Pharmacia, Piscataway, NJ) was packed into a column, which was washed several times with PBS. The extract was incubated with the column at 4°C for 1 hour. The unbound extract was drained out and the column was washed with GST buffer for 3 times. The GST fusion proteins were eluted with elution buffer [20 mmol/L of reduced glutathione, 100 mmol/L Tris-HCI (pH 8.0), and 120 mmol/L NaCI] and the fractions were collected and then analyzed by SDS-PAGE. It will be appreciated that the SARS-CoV-2 N-protein from another source may be used for the present invention.
Immunization:
Animal·. All animal experiments were approved by Institutional Animal Care and Use Committee (IACUC) of A*STAR. 8 weeks old female BALB/c mice were purchased from In Vivos, Singapore.
Vaccine preparation & injection:
GST-Nucleocapsid protein (GST-N- protein) was purified from GST-N protein bacterial clone. Nucleocapsid peptide with or without KLH tagged, Peptide#1 , Peptide#2, Peptide#3 were synthesized by Genemed Synthesis, Inc (USA). GST-N-protein (75ug), KLH-Peptide#3 (50ug) and Peptide#3 vaccine (20ug) in 100 ul of PBS were mixed thoroughly with 100 μL of Freund’s adjuvant (complete adjuvant for 1st immunization and incomplete adjuvant for subsequent
immunization, Pierce). The mice were immunized by intraperitoneal injection of each vaccine in 2 week intervals for 3 or 4 times. The vaccine without any protein or peptide (Adjuvant only) was injected in a group of mice as control. Blood samples (20 μL each time) were taken by tail bleed in Eppendorf tube, and serum was prepared. The antibody titer was measured by ELISA.
The peptides are:
Peptide#1 (P1): CIRQGTDYKHWPQIAQFAPSASAFFGMSRIG (SEQ ID NO: 2)
Peptide#2 (P2): CIAQFAPSASAFFGMSRIGMEVTPSGTWLTY (SEQ ID NO: 3)
Peptide#3 (P3): CVILLNKHIDAYKTFPPTEPKKDKKKKADET (SEQ ID NO: 4) Preparation of serum samples for Elisa:
Collected blood at different time points were centrifuged at 5000 rpm for 15 minutes. The supernatant serum was collected and stored at -70°C. 2-fold serial dilution of serum was done in PBS starting from dilution of 2 pL serum in 1024 pL of PBS (which will be the same as 2 fold per steps in 10 steps). Classified specific antibody subtypes induced in mice vaccinated with whole N Protein or Peptide#3:
Ninety-six-well plates (IWAKI, Japan) were coated with 100 pL of solution containing 50 ng of GST-N-protein, KLH-peptides or 20ng of peptides in PBS overnight at 4°C. Coated plates were blocked with 3% bovine serum albumin (BSA) for 1 hr room temperature and washed with PBS-t (PBS with 0.05% Tween-20). 0.1 ml of diluted mouse serum (2-fold serial dilution) was added to each well, and incubated for 1.5 hours at 37°C. After extensive washing, different subtypes of bound antibody were detected with horseradish peroxidase (HRP)-conjugated different antibodies by incubating for 1 hour at 37°C. Anti-N Protein IgM antibody was detected by goat anti-mouse IgM-HRP Antibody
(Invitrogen 626820). Subtype of anti N Protein IgG, lgG2a & lgG1, were detected by goat anti-mouse lgG2a-HRP (Invitrogen A10685) & goat antimouse lgG1-HRP (Invitrogen A10551) secondary antibodies. Anti N-protein IgG (whole IgG) were detected by goat anti-mouse IgG-HRP (H+L) secondary antibody (Invitrogen 31430). The plates were washed with PBST subsequently and 100 μL of tetramethylbenzidine (TMB) peroxidase substrate (Pierce) was added. The reaction was stopped by adding 100 pL of 2 M H2S04. Optical Density (OD) was measured at 450 nm using a plate reader (Tecan). Normal mouse serum at 10 steps dilution is used as control. OD > 3 times of normal mouse serum was considered as positive signal. The positive signal at specific steps of dilution was considered as the titer of that sample.
Elisa Assays:
Ninety-six-well plates (IWAKI, Japan) were coated with 100 pL of solution containing 50 ng of GST-N-protein, KLH-peptides or 20ng of peptides in PBS overnight at 4°C. Coated plates were blocked with 3% bovine serum albumin (BSA) in PBS containing 0.05% Tween 20 and washed with PBS. 0.1 ml of diluted mouse serum (2-fold serial dilution) was added to each well, and incubated for 1.5 hours at 37°C. After extensive washing, bound antibody was detected using horseradish peroxidase-conjugated anti-mouse antibody IgM, lgG2a, lgG1 , IgG-Fc by incubating for 1 hour at 37°C. Development was done using Turbo-TMB substrate (Pierce) and stopped by adding 100 pL of 2 M H2S04. Optical Density (OD) was measured at 450 nm using a plate reader (Tecan). Normal mouse serum at 10 steps dilution is used as control. OD > 3 times of normal mouse serum was considered as positive signal. The positive signal at specific steps of dilution was considered as the titer of that sample.
Classified specific antibody subtypes induced in mice vaccinated with whole N Protein or Peptide#3:
Ninety-six-well plates (IWAKI, Japan) were coated with 100 pL of solution containing 50 ng of GST-N-protein, KLH-peptides or 20ng of peptides in PBS overnight at 4°C. Coated plates were blocked with 3% bovine serum albumin (BSA) for 1 hr room temperature and washed with PBS-t (PBS with 0.05% Tween-20). 0.1 ml of diluted mouse serum (2-fold serial dilution) was added to each well, and incubated for 1.5 hours at 37°C. After extensive washing, different subtypes of bound antibody were detected with horseradish peroxidase (HRP)-conjugated different antibodies by incubating for 1 hour at 37°C. Anti-N Protein IgM antibody was detected by goat anti-mouse IgM-HRP Antibody (Invitrogen 626820). Subtype of anti N Protein IgG, lgG2a & lgG1 , were detected by goat anti-mouse lgG2a-HRP (Invitrogen A10685) & goat antimouse lgG1-HRP (Invitrogen A10551) secondary antibodies. Anti N-protein IgG (whole IgG) were detected by goat anti-mouse IgG-HRP (H+L) secondary antibody (Invitrogen 31430). The plates were washed with PBST subsequently and 100 pL of tetramethylbenzidine (TMB) peroxidase substrate (Pierce) was added. The reaction was stopped by adding 100 pL of 2 M H2S04. Optical Density (OD) was measured at 450 nm using a plate reader (Tecan). Normal mouse serum at 10 steps dilution is used as control. OD > 3 times of normal mouse serum was considered as positive signal. The positive signal at specific steps of dilution was considered as the titer of that sample.
Immuno-profiling of blood from unvaccinated and vaccinated mice:
Whole blood from mice was lysed with ACK Lysing Buffer (Gibco, A1049201) for 10 min at RT to remove RBCs. The remaining single-cell suspensions were then stained with Zombie UV Fixable Viability dye (BioLegend) for 30 min at 4 °C, approximately 300,000 - 500,000 cells were used per stain. Non-specific labelling was blocked with anti-CD16/32 (clone 2.4G2; BD Biosciences) for
30 min at 4 °C before multiplex labelling for 30 min at 4 °C with the following antibodies from BioLegend: Brilliant Violet 711 anti-mouse CD3e (clone 145- 2C11), PE-Cy7 anti-mouse CD4 (clone RM4-5), Brilliant Violet 786 anti-mouse CD8a (clone 53-6.7), PE/Dazzle 594 anti-mouse CD11b (clone M1/70), APC- Cy7 anti-mouse CD19 (clone 6D5), Brilliant Violet 510™ anti-mouse CD25 (clone PC61), AF488 anti-mouse CD45 (clone 30-F11), APC anti-mouse CD69 (clone H1.2F3) PE/Dazzle 594 anti-mouse CD127 (clone A7R34), Brilliant Violet 421 anti-mouse CD335 (clone 29A1.4), Brilliant Violet 421 anti-mouse IgG (clone Poly4053). And the following antibodies from BD Biosciences: BV711 Anti-Mouse CD3e (clone 145-2C11), APC-Cy7 Rat Anti-Mouse CD19 (clone 1 D3), PE Anti-Mouse CD44 (clone IM7), FITC Anti-Mouse CD45 (clone 30- F11), BV650 Anti-Mouse CD62L (clone MEL-14), PE-CF594 Anti-Mouse CD80 (clone 16-10A1), BV786 Anti-Mouse CD138 (clone 281-2), BV510 Anti-Mouse CD273 (clone TY25), APC Anti-Mouse IgD (clone 11-26c.2a). All samples were run on a BD LSR II flow cytometer and analysed using the FlowJo software 10.5.3 (FlowJo).
Results
N-Protein vaccination can produce high and sustainable anti N-protein different antibody subtypes Immunization of N-protein vaccine was done in BALB/c mice and the antibody response at different time intervals was analyzed. Serum IgM, IgG and the subclasses lgG1 and lgG2a were measured using ELISA to evaluate the profile of the immune response. lgG1 indicates a humoral immune response, whereas lgG2a indicates a cellular immune response. Antibody production was not detected after 1st dose of vaccination. IgM antibody was detected after 2nd vaccination, but it stays at the plateau phase, and at a sustainable level throughout the time point. IgG and its subclasses lgG1 & lgG2a antibodies can also be detected after the 2nd dose of vaccination. These antibodies gradually
increase upon subsequent vaccination and remain in the plateau phase throughout the time points. Peak antibody responses were detected after the 4th dose of vaccination in each BALB/c mouse (Figure 1A, B, C, D). The second dose of vaccine significantly boosted the concentrations of all antibody subtypes, IgM (p-value < 0.001), lgG2a (p-value <0.001), lgG1 (p-value <0.0001) and IgG (p-value <0.0001) comparison between 2 weeks (2weeks after 1st vaccination) and 4 weeks (2 weeks after 2nd vaccination) time point (Figure 1 E). All types of antibodies were not detected in the mice vaccinated with adjuvant only. For all mice, the second dose of vaccine elicited a greater increase in the lgG1 antibody concentration than in the lgG2a antibody concentration, which resulted in a lower lgG2a/lgG1 ratio compared to the ratio observed after the first vaccination.
Next, to confirm the above antibody production, we did immunization of N- protein in another mouse species, FVB mice. The same trend of antibody production was detected in FVB mice also (Figure 1 F). By ELISA assays, we showed that mice vaccinated with whole N protein produced high titer of specific anti-N antibody subtypes: IgM (p-value <0.045), IgG (p-value <0.0037), lgG1(p- value <0.045), lgG2a (p-value <0.029), suggesting that N-protein is an excellent antigen with high immunogenicity to evoke a protective immunity and produce anti-N specific antibodies at high titers.
Peptides selected on N-protein sequence could bind specifically to anti-N protein antibody
N protein vaccination resulted in a high and sustained production of different IgG subtypes in mice, indicating that N-protein is a good vaccine candidate. Based on N-protein sequences (419 amino acid sequence), we considered the development of a peptide vaccine of a specific epitope which possesses a higher accuracy in targeting N protein. We selected 3 different peptides: peptide 1 , peptide 2, and peptide 3 that are derived from N-protein, and 3 N-protein
peptides were synthesized, 30 amino acids in the length of each peptide (Figure 2A) with or without KLH carrier.
To test which synthetic peptides (Peptide #1 , #2, and #3) induces the host to produce the highest titer of anti-N-protein specific antibodies, we performed ELISA assays by coating the ELISA plate with different concentration, 5 ng or 50 ng per well of each peptide, incubated with serum sample taken after the 4th immunization of N-protein vaccine at 2-fold serial dilution at 10, 11 and 13 steps. N-protein coating was used as positive control. By the appearance of the ELISA assay, peptide #3 showed the best binding activity compared to other peptides. Quantitating the reaction by measuring Optical Density (OD) showed similar results (Figure 2B). The OD of Peptide #3 (with or without KLH carrier) were nearly 2/3 of N-protein OD at dilution step 10. The OD of other peptides, peptide #1 and Peptide #2 were nearly 10 times lower than peptide #3, indicating that Peptide 3 is the best candidate to represent N protein epitope to produce antibody which is specific to N-protein, and the peptide#3 could potentially be combined with the cocktail of traditional Influenza vaccine to be a general safe vaccine.
Peptide vaccination could induce high and sustainable antibody production similar to N protein vaccine. Among the 3 peptides synthesized, most polyclonal anti-N antibodies react highly with peptide#3, but not peptide 1 and peptide 2, suggesting that peptide 3 alone could be a good immunogen for vaccination to evoke host immune system to produce antibody specific to N protein. By mixing with Freund adjuvant, the immunization was done on BALB/c mice for 3 times in a 2-week interval. The blood collection (20 μL each) was done before each immunization and every 2 weeks after immunization. Similar to N-protein vaccination, serum IgM, IgG and the subclasses lgG1 and lgG2a were measured using ELISA to evaluate the profile of the immune response.
Antibody production can be seen after the 2nd vaccine dose. The pattern and quantity of rise in antibody titer is similar to N-protein vaccine (Figure 2C, D).
To test the binding of anti-Peptide#3 antibody in mouse serum and N-protein, ELISA assay was performed by coating the ELISA plate with different concentration, 5ng or 50 ng per well, of N protein, incubated with serum sample taken after the 3rd immunization of Peptide#3 vaccine at 2-fold serial dilution at 10, 11 and 13 steps. By the appearance of the ELISA reaction, anti-Peptide#3 antibody bound to N-protein with good affinity (Figure 2E). Quantitating the reaction by measuring Optical Density (OD) showed similar results (Figure 2F). N protein vaccination results in the accumulation of memory T cells
To investigate if N protein vaccination can result in the accumulation of memory immune cells, 8-week-old BALB/c mice were vaccinated once weekly with a combination of Freund’s adjuvant and N protein (Vaccinated mice) for four weeks. Vaccinated mice were then bled and sacrificed eight weeks after the last vaccination to determine if they had elevated levels of memory cells compared to unvaccinated mice.
Circulating live CD45+CD3+CD335 CD4+CD8 CD44+CD62I_- (Figure 3A, 3B) and CD45+CD3+CD335 CD4 CD8+CD44+CD62L- (Figure 3D, 3E) memory T cell frequencies were significantly (CD4+, p-value = 0.0000129; CD8+, p-value = 0.000306, one way-ANOVA) increased in vaccinated BALB/c mice compared with unvaccinated WT controls, suggesting that our vaccination protocol can successfully induce a robust and lasting memory CD4* and CD8* T cell population.
Additional phenotypic analysis of T cell subpopulations reveal a corresponding decrease in the proportion of circulating live CD45+CD3+CD335 CD4+CD8" CD44 CD62L* (Figure 3A, 3C) and CD45+CD3+CD335 CD4 CD8+CD44 CD62L+ (Figure 3D, 3F) naive T cell levels (CD4+, p-value = 0.00817; CD8+ p-value =
0.0160) in vaccinated BALB/c mice compared with unvaccinated WT controls, supporting our observation that our vaccination protocol results in a decrease in antigen naive T cells and an elevated frequency of antigen experienced memory CD4+ and CD8+ T cells. In contrast, the change in live CD45+CD19+CD138lgD+lgG' naive B cells and CD45+CD19+CD138lgDlgG+ class-switched memory B cells is less distinct. The frequency of both IgG class-switched memory B cells in vaccinated mice is similar to their unvaccinated counterparts, while the frequency of naive B cells is elevated in vaccinated BALB/c mice (Figure 3G-I). The lack of a permanent large increase in the frequency of memory B cells in our vaccinated mice may indicate that T cells may be more important for mediating long term immunity against SARS-CoV-2 and may explain why some former SARS-CoV-2 patients experience a decline in antibodies several months after recovery [Marot et al., (2021); Self et al., 2020], Example 2: Cvtokine production in vaccinated mice
Materials and methods
Cytokine Array:
Mouse serum cytokines from unvaccinated and vaccinated mice were analysed with the RayBiotech mouse cytokine array C1 (Cat: #AMM-CYT-1-8) using the provided experimental protocol unless otherwise indicated. In brief, blots were blocked with 2m L of provided blocking buffer and incubated for 30m ins at room temperature. 6pL of each serum sample was diluted to a total volume of 500μL with blocking buffer and arrays were incubated overnight at 4°C with dilute serum samples. Arrays were then washed with provided washing buffers according to the standard protocol. Next arrays were incubated with 500μL of pre-diluted biotinylated antibody cocktail for 4hrs at room temperature. Arrays were then washed with provided washing buffers according to the standard
protocol. Arrays were then incubated with 500μΙ_ of x1 HRP-Streptavidin for 2hrs at room temperature. Arrays were washed and incubated with detection buffers for chemiluminescence detection.
Chemiluminescence Detection: The fold change of N4 and N12 vaccinated mice was calculated for all the cytokine proteins against untreated mice (see Figure 5). A significant cut-off fold change > 2 is used and highlighted in red. Seven proteins (IFN gamma, CCL2, GCSF, IL-10, CCL5, TNFR1 and TNFalpha) highlighted in yellow exhibits both significant and steady rise in the fold change in both groups. Results
Vaccination N protein with complete Freund’s adjuvant can induce the
secretion of pro-inflammatory memory cell and TH1 associated cytokines.
Cytokine array studies of mouse serum suggests that our vaccinated mice have elevated levels of pro-inflammatory cytokines and chemokines such as CCL2, CCL5, IFN-y, TNF-α, TNF-RI, GCSF, IL-4, and IL-10 compared to unvaccinated mice (Figure 4A-4D). A subsequent cycle of vaccination (4th immunization) also results in an increased level of these cytokines compared to a prior cycle (2nd immunization), suggesting that repeated vaccinations with N protein can result in progressively elevated cytokine levels in mice (Figure 4C-4D) and likely enhanced immune responses during subsequent vaccinations due to the accumulation of memory immune cells.
Example 3: Generation of Anti N -protein antibody against Nucleocapsid N protein for therapeutic
Materials and methods
Generation of mouse anti-N protein monoclonal antibody: N-protein hybridoma clone generated by fusion of splenocytes from N -protein immunized BALB/c mice and BALB/c parental myeloma SP2/0 cells was kindly provided by Dr Yee Joo TAN (Monoclonal Antibody Unit, IMCB, A*STAR, Singapore). Hybridoma cells (5 x 105) were suspended in 200 μL of Phosphate Buffered Saline (PBS) and injected into the peritoneal cavity and wait until the mouse developed a large quantity of ascitic fluid. The mouse was sacrificed and ascitic fluid was collected, centrifuged and frozen at -70°C until further use.
Total RNA was isolated from the hybridoma cells following the technical manual of RNeasy Plus Micro Kit. Total RNA was then reverse-transcribed into cDNA using either isotype-specific anti-sense primers or universal primers following the technical manual of SMARTScribe Reverse Transcriptase. Antibody fragments of heavy chain procedure (SOP) of rapid amplification of cDNA ends (RACE). Amplified antibody fragments were cloned into a standard cloning vector separately. Colony PCR was performed to screen for clones with inserts of correct sizes. Generation of Ascetic Fluids:
Hybridoma cells (5 x 106) were suspended in 200 L of serum-free DMEM medium and injected with a 26-gauge needle into the peritoneal cavity to BALB/c mice. After 10 days, the mouse developed a large quantity of ascitic fluid, and the abdomen was greatly distended. The mouse was sacrificed and a small shallow was cut to open the abdominal cavity. The ascitic fluid was drawn with a 10-mL syringe fitted with an 18-gauge needle. The fluid was centrifuged
at 200 x g for 10 minutes at 4°C. The supernatant fluid was collected and frozen at -70°C until further use.
Results
In 2003, we generated a monoclonal antibody (clone 6H3) against SARS-CoV. This SARS-CoV antibody binds to SAR-CoV2 N-protein with good affinity (Figure 5). We further demonstrated 6H3 cross-reacting with SAR-CoV-2, using ELISA assay to access the binding affinity. ELISA plate was coated with 5 & 20 ng/well of Peptide #1 , Peptide #2, Peptide #3 & N-protein (SARS-CoV2). Anti- SARS-CoV-N protein antibody clone (6H3) was diluted at 1:1000 & 1 :5000, followed by goat anti-mouse IgG-HRP secondary antibody. The measurement of Optical Density showed Mouse SAR-CoV-N-protein antibody could not bind well to Peptide #1 , Peptide #2, and Peptide #3 but bind strongly to SARS-CoV2 N-protein in a concentration dependent manner (Figure 5), suggesting clone 6H3 epitope presents in the whole N protein but not in all 3 peptides. The mouse antibody (6H3) can be developed for the First in Class humanized antibody to treat patients infected with coronavirus.
Example 4: Discussion
Severe Acute Respiratory Syndrome Coronavirus 2 (SAR-CoV-2) caused the global pandemic of the Coronavirus disease in late 2019 (COVID-19). Vaccine development efforts have predominantly been aimed at ‘Extra-viral’ Spike (S) mRNA as vaccine vehicles but there are concerns regarding ‘viral immune escape’ since multiple mutations may enable the mutated virus strains to escape from immunity against S protein. The ‘Intra-viral’ Nucleocapsid (N- protein) is relatively conserved among mutant strains of coronaviruses during spread and evolution. Herein, we demonstrate novel vaccine candidates against SARS-CoV-2 by using the whole conserved N-protein or its fragment/peptides. Using ELISA assay, we showed that high titers of specific anti-N antibodies (IgG, lgG1 , lgG2a, IgM) were maintained > 5 months, suggesting that N-protein
is an excellent immunogen to stimulate host immune system and robust B cell activation. We synthesized 3 peptides located at the conserved regions of N- protein among CoVs. One peptide showed as a good immunogen for vaccination as well. Cytokine arrays on post-immunization mouse sera showed progressive upregulation of various cytokines such as IFN-γ and CCL5, suggesting that TH1 associated responses are also stimulated. Furthermore, vaccinated mice exhibited an elevated memory T cells population. Here, we propose an unconventional vaccine strategy targeting the conserved N-protein as an alternative ‘Universal vaccine’ for coronaviruses. Moreover, we generated a mouse monoclonal antibody specifically against an epitope shared between SAR-CoV and SAR-CoV-2, and we are currently developing the First-in-Class humanized anti-N-protein antibody to potentially treat patients infected by various CoVs in the future.
In this study, we have demonstrated that mice vaccinated with N protein/or N protein fragment/peptides had sustainably high titers of anti-N antibodies (IgG, lgG1 , lgG2a, IgM). Interestingly, vaccination with peptide #3 gave similar results as that of the whole N protein, suggesting that peptide #3 is not only the major epitope in the N-protein but also sufficient to elicit protective immunity in the host. We also observed a robust induction of CD4+ and CD8+ memory T cells along with the induction of pro-inflammatory and TH1 associated cytokines.
A major challenge in the early development of SARS coronavirus vaccines has been the discovery that double-inactivated SARS-CoV whole viral vaccines have low efficacy and resulted in enhanced immune pathology especially in aged animal model [Bolles et al., (2011)]. Interestingly, a further study demonstrated that while vaccination with Venezuelan equine encephalitis virus replicon particles (VRP) containing the SARS-CoV strain spike (S) glycoprotein could provide protection against viral challenges, vaccination with nucleocapsid (N) protein resulted in enhanced immunopathology with increased eosinophilic lung infiltrates in challenged mice [Deming et al., (2006)]. Another study also
reported that SARS-CoV N protein vaccination in mice resulted in severe pneumonia upon viral challenge, suggesting that excessive host immune response against N protein may cause the severe acute lung injury observed in SARS-CoV infection [Yasui etal., (2008)]. In addition, only mice immunized with S protein exhibited reduced viral titers after vaccination, in contrast to vaccination with other SARS-CoV structural proteins, such as the N, membrane (M), and envelop (E) proteins [Yasui etal., (2008)].
Clinically, patients with both SARS-CoV and SARS-CoV-2 first exhibit antibodies against N protein and antibodies against N protein are the most sensitive for serologic diagnosis [Tan et al., (2004); Wu et al., (2004); Leung et al., (2004); Zhu etal., (2006); Burbelo etal., (2020a)]. Interestingly patients with elevated levels of antibodies against N protein and lower levels of anti-S protein antibodies have a higher risk of admission to the intensive care unit and longer hospitalization stays [Batra et al., (2021); Roltgen et al., (2020)]. This may suggest that N protein antibodies may also favor a stronger inflammatory response in human patients.
Similar to the SARS coronavirus (SARS-CoV), SARS-CoV-2 infects target cells via spike protein receptor binding domain (RBD) and ACE2 receptor interactions [Hoffmann et al., (2020); Zhou etal., (2020)]. To generate effective neutralizing antibodies to block SARS-CoV-2 viral entry, the SARS-CoV-2 spike protein and its RBD were selected as the leading target antigens in vaccine development [Chen et al., (2020); Pang et al., (2020)]. Wang et al., (2021) reported that volunteers injected with either the Modema (mRNA-1273) or Pfizer-BioNTech (BNT162b2) vaccine against SARS-CoV-2 demonstrated high litres of IgM and IgG antibodies against SARS-CoV-2 S protein and RBD [Wang et al., (2021)]. The plasma neutralizing activity and relative numbers of RBD- specific memory B cells of vaccinated individuals is also reported to be similar to patients who recovered from natural infection [Wang et al., (2021); Gaebler et
al., (2021); Robbiani et al., (2020)]. A study group involving approximately 600,000 individuals in Israel also demonstrated that the BNT162b2 vaccine has an 92% effectiveness of preventing SARS-CoV-2 infection [Dagan et al., (2021)]. However, the presence of neutralizing antibodies against SARS-CoV-2 S protein and its RBD does not confer complete protection against SARS-CoV-2 infection in all vaccinated individuals, even in recently vaccinated individuals. Surprisingly, a subset of recently vaccinated individuals still contract SARS- CoV-2 despite multiple vaccinations which should have induced robust levels of neutralizing antibodies. In addition, it has been reported that the titer of SARS- CoV-2 neutralizing antibodies decline fairly rapidly, with some individuals reporting close to baseline neutralizing antibody levels as soon as two months post-infection [Marot et al., (2021); Seow et al., (2020); Yamayoshi et al., (2021)]. The SARS-CoV-2 S gene also has a relatively lower amino acid similarity (76%) compared to the SARS-CoV S gene with a higher rate of mutation compared to the more conserved (90)% N gene [Dutta et al., (2020); Grifoni et al., (2020a); Marra et al., (2003); Drosten et al., (2003); Zhu et al., (2005)]. This data suggests that while anti-S protein antibodies may be key for controlling viral litres during an ongoing infection, other immune mediators may be responsible for conferring long-term immunity to SARS-CoV-2.
The N protein is highly immunogenic and is the most abundant viral protein during coronavirus infections [Dai et al., (2021); Long et al., (2020)]. It is also a major target for antibody and T cell responses [Sariol and Perlman (2020)]. Importantly, non-neutralizing antibodies against N protein can protect mice against some other viruses, such as the mouse hepatitis virus [Nakanaga et al., (1986); Lecomte et al., (1987)] and influenza A virus [Fujimoto et al., (2016)]. N protein is also commonly externalized on the cell surface membrane of infected cells, and can act as a potential target for both antibody and T cell responses [Fujimoto et al., (2016)].
Furthermore, memory T cells may play a critical role in conferring long-term immunity to SARS-CoV-2. Grifoni et al (2020b) reports the induction of robust CD4* and CD8* T cells in convalescent SARS-CoV-2 patients. Surprisingly even some non-exposed individuals demonstrate T cell reactivity against SARS-CoV-2 epitopes, suggesting that prior infections in these individuals could also enhance immunity against SARS-CoV-2 [Grifoni et al (2020b)]. Le Bert et al also demonstrates that former SARS-CoV patients possess long lasting memory T cells which are reactive to N protein over 17 years after the SARS epidemic in 2003 [Le Bert et al, (2020)]. These memory T cells were also highly cross-reactive to the SARS-CoV-2 N protein, suggesting that these individuals may be less susceptible to SARS-CoV-2 infection and other similar coronavirus [Le Bert et al, (2020)]. Other animal model studies involving vaccination with SARS-CoV N protein have also demonstrated robust SARS- specific T cell proliferation and cytotoxic responses [Gao et al., (2003); Okada et al., (2003)]. N protein specific CD8* T cells also protect against infectious bronchitis virus model in chickens. This data suggests that T cells are essential for mediating long-term immunity.
In short, our findings have shown that N protein vaccination provides sustainably long protective immunity. Much emphasis has been placed on the extra-viral spike (S) protein in vaccine development. This is due to its importance in the detection by host immune system and viral entry into host cells. However, the glycosylation and mutation of the S protein have posed challenges in vaccine development. Intra-viral nucleocapsid (N) protein, on the other hand, is more conserved [Surjit et al., (2008)] in sequence. Importantly, N protein can be detected by the host immune system as there is a presence of anti-N protein antibodies in the sera of SARS-CoV-2 infected patients [Burbelo et al., (2020b)]. On the potential of anti-N protein antibodies in the prevention of infection, dominant helper T-cell epitopes in the N protein of SARS-CoV have been identified to assist in antiviral neutralizing antibody production [Zhao et al.,
(2007). The anti-N protein antibodies have been previously shown to confer protection against several types of lethal influenza A viruses [La Mere et al., (2011); Fujimoto et al., (2016); Carragher et al., (2008)]. A combination of neutralizing antibodies targeting S protein and its RBD, anti-N protein antibodies, and memory T cells against N protein epitopes may be essential to confer long-term protection against SARS-CoV-2.
Our data showed that monoclonal anti-N protein antibody raised against N protein of SARS-CoV can recognize/bind to the SARS-CoV-2 N protein with good affinity. From this, it is hypothesized that a humanized anti-N protein antibody could potentially be used as a therapy in the eradication of infected host cells.
Similar strategies of Intra-viral protein unconventional immunotherapies could apply to other viral infections, such as HBV or HIV.
References
Any listing or discussion of an apparently prior-published document in this specification should not necessarily be taken as an acknowledgement that such document is part of the state of the art or is common general knowledge. Batra, M., et al., Role of IgG against N-protein of SARS-CoV2 in COVID19 clinical outcomes. Sci Rep, 2021. 11(1): p. 3455.
Bolles, M., et al., A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflam matory pulmonary response upon challenge. J Virol, 2011. 85(23): p. 12201-15.
Burbelo, P.D., et al., Sensitivity in Detection of Antibodies to Nucleocapsid and Spike Proteins of Severe Acute Respiratory Syndrome Coronavirus 2 in Patients With Coronavirus Disease 2019. J Infect Dis, 2020a. 222(2): p. 206- 213.
Burbelo, P.D., et al., Detection of Nucleocapsid Antibody to SARS-CoV-2 is More Sensitive than Antibody to Spike Protein in COVID-19 Patients. medRxiv : the preprint server for health sciences, 2020b: p. 2020.04.20.20071423.
Carragher, D.M., et al., A novel role for non-neutralizing antibodies against nucleoprotein in facilitating resistance to influenza virus. Journal of immunology (Baltimore, Md. : 1950), 2008. 181(6): p. 4168-4176.
Chen, W.H., et al., The SARS-CoV-2 Vaccine Pipeline: an Overview. Curr Trap Med Rep, 2020: p. 1-4.
Dagan, N., et al., BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Mass Vaccination Setting. N Engl J Med, 2021.
Dai, L. and G.F. Gao, Viral targets for vaccines against COVID-19. Nat Rev Immunol, 2021. 21(2): p. 73-82.
Deming, D., et al., Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med, 2006. 3(12): p. e525.
Drosten, C., et al., Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med, 2003. 348(20): p. 1967-76.
Dutta, N.K., K. Mazumdar, and J.T. Gordy, The Nucleocapsid Protein of SARS- CoV-2: a Target for Vaccine Development. J Virol, 2020a. 94(13).
Fujimoto, Y., et al., Cross-protective potential of anti-nucleoprotein human monoclonal antibodies against lethal influenza A virus infection. J Gen Virol, 2016. 97(9): p. 2104-2116.
Gaebler, C., et al., Evolution of antibody immunity to SARS-CoV-2. Nature, 2021.
Gao, W., et al., Effects of a SARS-associated coronavirus vaccine in monkeys. Lancet, 2003. 362(9399): p. 1895-6.
Grifoni, A., et al., A Sequence Homology and Bioinformatic Approach Can Predict Candidate Targets for Immune Responses to SARS-CoV-2. Cell Host Microbe, 2020a. 27(4): p. 671-680 e2.
Grifoni, A., et al., Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell, 2020b. 181(7): p. 1489-1501 e15.
Hoffmann, M., et al., SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell, 2020. 181(2): p. 271-280 e8.
Le Bert, N., et al., SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature, 2020. 584(7821): p. 457-462.
Lecomte, J., et al., Protection from mouse hepatitis virus type 3-induced acute disease by an anti-nucleoprotein monoclonal antibody. Brief report. Arch Virol, 1987. 97(1 -2): p. 123-30.
LaMere, M.W., et al., Regulation of Antinucleoprotein IgG by Systemic Vaccination and Its Effect on Influenza Virus Clearance. Journal of Virology, 2011. 85(10): p. 5027.
Leung, D.T., et al., Antibody response of patients with severe acute respiratory syndrome (SARS) targets the viral nucleocapsid. J Infect Dis, 2004. 190(2): p. 379-86.
Long, Q.X., etal., Antibody responses to SARS-CoV-2 in patients with COVID- 19. Nat Med, 2020. 26(6): p. 845-848.
Marot, S., et al., Rapid decline of neutralizing antibodies against SARS-CoV-2 among infected healthcare workers. Nat Commun, 2021. 12(1): p. 844.
Marra, M.A., et al., The Genome sequence of the SARS-associated coronavirus. Science, 2003. 300(5624): p. 1399-404.
Nakanaga, K., K. Yamanouchi, and K. Fujiwara, Protective effect of monoclonal antibodies on lethal mouse hepatitis virus infection in mice. J Virol, 1986. 59(1): p. 168-71.
Okada, M., et al., The development of vaccines against BARS corona virus in mice and SCID-PBL/hu mice. Vaccine, 2005. 23(17-18): p. 2269-72.
Pang, J., etal., Potential Rapid Diagnostics, Vaccine and Therapeutics for 2019 Novel Coronavirus (2019-nCoV): A Systematic Review. J Clin Med, 2020. 9(3).
Robbiani, D.F., et al., Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature, 2020. 584(7821 ): p. 437-442.
Roltgen, K., et al., Defining the features and duration of antibody responses to SARS-CoV-2 infection associated with disease severity and outcome. Sci Immunol, 2020. 5(54).
Sariol, A. and S. Perlman, Lessons for COVID-19 Immunity from Other Coronavirus Infections. Immunity, 2020. 53(2): p. 248-263.
Self, W.H., et al., Decline in SARS-CoV-2 Antibodies After Mild Infection Among Frontline Health Care Personnel in a Multistate Hospital Network - 12 States, April-August 2020. MMWR Morb Mortal Wkly Rep, 2020. 69(47): p. 1762-1766.
Seow, J., et al., Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans. Nat Microbiol, 2020. 5(12): p. 1598-1607.
Surjit, M. and S.K. Lai, The SARS-CoV nucleocapsid protein: a protein with multifarious activities. Infection, genetics and evolution : journal of molecular epidemiology and evolutionary genetics in infectious diseases, 2008. 8(4): p. 397-405.
Tan, Y.J., et al., Profiles of antibody responses against severe acute respiratory syndrome coronavirus recombinant proteins and their potential use as diagnostic markers. Clin Diagn Lab Immunol, 2004. 11(2): p. 362-71.
Wang, Z., et al., mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature, 2021.
WHO Coronavirus (COVID-19) Dashboard, https://covid19.who.int/ (assessed at 3rd April 2021).
Wu, H.S., et al., Early detection of antibodies against various structural proteins of the SARS-associated coronavirus in SARS patients. J Biomed Sci, 2004. 11(1): p. 117-26.
Yamayoshi, S., et al., Antibody titers against SARS-CoV-2 decline, but do not disappear for several months. EClinicalMedicine, 2021. 32: p. 100734.
Yasui, F., et al., Prior immunization with severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) nucleocapsid protein causes severe pneumonia in mice infected with SARS-CoV. J Immunol, 2008. 181(9): p. 6337-48.
Zhao, J., et al., Identification and characterization of dominant helper T-cell epitopes in the nucleocapsid protein of severe acute respiratory syndrome coronavirus. Journal of virology, 2007. 81 (11 ): p. 6079-6088.
Zhou, P., et al., A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature, 2020. 579(7798): p. 270-273.
Zhu, Y., et al., Isolation of virus from a SARS patient and genome-wide analysis of genetic mutations related to pathogenesis and epidemiology from 47 SARS- CoV isolates. Virus Genes, 2005. 30(1): p. 93-102.
Zhu, H., et al., Severe acute respiratory syndrome diagnostics using a coronavirus protein microarray. Proc Natl Acad Sci U S A, 2006. 103(11): p. 4011-6.
Claims
1. An isolated nucleocapsid protein (N-protein) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or an immunogenic fragment thereof.
2. The isolated N-protein according to claim 1, comprising SEQ ID NO: 1.
3. The isolated N-protein according to claim 1, consisting of SEQ ID NO: 1.
4. An immunogenic fragment according to claim 1 , comprising a sequence selected from the group consisting of:
SEQ ID NO: 2;
SEQ ID NO: 3;
SEQ ID NO: 4.
5. An immunogenic fragment according to claim 1, consisting of SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 4.
6. An immunogenic fragment according to claim 1 , comprising SEQ ID NO: 4.
7. An immunogenic fragment according to claim 1, consisting of SEQ ID NO:
4.
8. An immunogenic combination and/or or immunogenic composition comprising any two or more components selected from the group consisting of:
(i) the isolated nucleocapsid protein (N-protein) according to claim 1 ;
(ii) immunogenic fragment(s) according to any one of claims 1 to 7.
9. The immunogenic combination and/or immunogenic composition according to claim 5 comprising two or more immunogenic fragments according to any one of claims 1 to 7.
10. An isolated nucleic acid molecule encoding the SARS-CoV2 nucleocapsid
(N-protein) and/or an immunogenic fragment thereof.
11. A vector comprising the isolated nucleic acid according to claim 10.
12. A host cell comprising the isolated nucleic acid molecule according to claim 10 or the vector according to claim 11.
13 A vaccine comprising the isolated N-protein and/or immunogenic fragment thereof according to any one of claims 1 to 7, the isolated nucleic acid molecule according to claim 10 and/or the vector according to claim 11.
14. The vaccine according to claim 13, further comprising a pharmaceutically acceptable excipient.
15. The vaccine according to claim 13 or 14, further comprising an adjuvant.
16. The isolated N-protein and/or immunogenic fragment thereof according to any one of claims 1 to 7, the isolated nucleic acid molecule according to claim 10 and/or the vector according to claim 11 , for use as a vaccine.
17. Use of the isolated N-protein and/or immunogenic fragment thereof according to any one of claims 1 to 7, the isolated nucleic acid molecule according to claim 10 and/or the vector according to claim 10; in the preparation of a vaccine.
18. The use according to claim 17, in the preparation of a vaccine for immunizing against a viral infection.
19. The use according to claim 18, wherein the viral infection comprises a Coronavirus infection.
20. The use according to claim 19, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
21. The use according to claim 17, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus 2 (SARS-CoV- 2) infection [Coronavirus disease 2019 (COVID-19)].
22. A method of immunising a subject against a virus infection comprising administering to the subject the isolated N-protein and/or immunogenic fragment thereof according to any one of claims 1 to 7, the immunogenic combination and/or the immunogenic composition according to claim 8, the isolated nucleic acid molecule according to claim 9 and/or the vector according to claim 10.
23. The method according to claim 22, wherein the viral infection comprises a Coronavirus infection.
24. The method according to claim 23, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus (SARS-CoV)
infection, Middle East respiratory syndrome coronavirus (MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
25. The method according to claim 24, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus 2 (SARS-CoV- 2) infection [Coronavirus disease 2019 (COVID-19)].
26. An antibody capable of binding to the SARS-CoV-2 nucleocapsid protein (N-protein) and/or an immunogenic fragment thereof or an antigen-binding fragment of the antibody.
27. The antibody according to claim 26 wherein the antibody comprises a monoclonal antibody.
28 The monoclonal antibody according to claim 27, wherein the antibody comprises a chimeric or humanised antibody.
29. The antibody according to claim 26, comprising a heavy chain comprising SEQ ID NO: 5
30. The antibody according to claim 26, comprising a heavy chain variable region comprising SEQ ID NO: 6.
31. The antibody according to claim 26, comprising a heavy chain comprising a CDR region 1 (CDR1) comprising SEQ ID NO: 7, a CDR region 2 (CDR2) comprising SEQ ID NO: 8, and a CDR region 3 (CDR3) comprising SEQ ID NO: 9.
32. A pharmaceutical composition comprising the antibody according to any one of claims 26 to 31.
33. The pharmaceutical composition according to claim 32, further comprising a pharmaceutically acceptable excipient.
34. The antibody and/or antigen-binding fragment according to any one of claims 26 to 31 , for use as a medicament.
35. The antibody according to any one of claims 26 to 31 , for use in treating a viral infection.
36. The antibody for the use according to claim 35, wherein the viral infection comprises a Coronavirus infection.
37. The antibody for the use according to claim 36, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus
(MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
38. The antibody for the use according to claim 37, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
39. Use of the antibody and/or antigen-binding fragment according to any one of claims 26 to 31 in the preparation of a pharmaceutical composition for treating a viral infection.
40. Use according to claim 39, wherein the viral infection comprises a Coronavirus infection.
41. Use according to claim 40, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
42. Use according to claim 41, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
43. A method of treating a viral infection in a subject comprising administering the antibody and/or antigenic fragment according to any one of claims 26 to 31 to the subject.
44. The method according to claim 43, wherein the viral infection comprises a Coronavirus infection.
45. The method according to claim 44, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus (SARS-CoV) infection, Middle East respiratory syndrome coronavirus (MERS-CoV) and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [Coronavirus disease 2019 (COVID-19)].
46. The method according to claim 45, wherein the Coronavirus infection comprises severe acute respiratory syndrome coronavirus 2 (SARS-CoV- 2) infection [Coronavirus disease 2019 (COVID-19)].
47. An isolated nucleic acid molecule encoding the heavy chain of an antibody according to any one of claims 26 to 31.
48. The isolated nucleic acid molecule according to claim 47, comprising SEQ ID NO: 10.
49. An isolated nucleic acid molecule encoding the heavy chain variable region of an antibody according to any one of claims 26 to 31.
50. The isolated nucleic acid molecule according to claim 49, wherein the variable region of the antibody comprises SEQ ID NO: 6.
51. The isolated nucleic acid molecule according to claim 47, wherein the heavy chain of the antibody comprising a CDR region 1 (CDR1) comprises SEQ ID NO: 7, the CDR region 2 (CDR2) comprising SEQ ID NO: 8, and a CDR region 3 (CDR3) comprising SEQ ID NO: 9.
52. An isolated nucleic acid molecule encoding the light chain of an antibody according to any one of claims 26 to 31.
53. An isolated nucleic acid molecule encoding the light chain variable region of an antibody according to any one of claims 26 to 31.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/SG2021/050497 WO2022216223A1 (en) | 2020-04-09 | 2021-08-25 | Vaccine and/or antibody for viral infection |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
SG10202003282W | 2020-04-09 | ||
SG10202003282W | 2020-04-09 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2021206638A1 true WO2021206638A1 (en) | 2021-10-14 |
Family
ID=78026174
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/SG2021/050197 WO2021206638A1 (en) | 2020-04-09 | 2021-04-08 | Vaccine and/or antibody for viral infection |
PCT/SG2021/050497 WO2022216223A1 (en) | 2020-04-09 | 2021-08-25 | Vaccine and/or antibody for viral infection |
Family Applications After (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/SG2021/050497 WO2022216223A1 (en) | 2020-04-09 | 2021-08-25 | Vaccine and/or antibody for viral infection |
Country Status (1)
Country | Link |
---|---|
WO (2) | WO2021206638A1 (en) |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11564892B2 (en) | 2020-04-09 | 2023-01-31 | Finncure Oy | Virus-like particles for preventing the spreading and lowering the infection rate of viruses |
WO2023117742A1 (en) * | 2021-12-22 | 2023-06-29 | Osivax | Vaccine compositions and their use |
CN117659180A (en) * | 2022-09-06 | 2024-03-08 | 东莞市朋志生物科技有限公司 | Anti-novel coronavirus antibody or functional fragment thereof, reagent for detecting novel coronavirus and kit |
US12144898B2 (en) | 2020-04-09 | 2024-11-19 | Finncure Oy | Virus-like particles for preventing the spreading and lowering the infection rate of viruses |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060188519A1 (en) * | 2004-06-14 | 2006-08-24 | To Cheung | Peptides, antibodies, and methods for the diagnosis of SARS |
CN111647079A (en) * | 2020-07-03 | 2020-09-11 | 北部湾大学 | Neutralizing antibody for resisting novel coronavirus N protein |
CN111875700A (en) * | 2020-07-28 | 2020-11-03 | 武汉华美生物工程有限公司 | Single-chain antibody of anti SARS-COV-2 virus N protein and its use |
Family Cites Families (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR100832870B1 (en) * | 2006-04-19 | 2008-05-30 | 대한민국 | Monoclonal antibody against nucleocapsid protein of SARS coronavirus and the use thereof |
CN112961222B (en) * | 2020-02-04 | 2022-11-04 | 中国科学院微生物研究所 | 2019 novel coronavirus N protein linear epitope peptide, monoclonal antibody and application |
CN111153991A (en) * | 2020-02-26 | 2020-05-15 | 北京博奥森生物技术有限公司 | Human SARS-CoV-2 monoclonal antibody and its preparation method and use |
CN112225797B (en) * | 2020-09-24 | 2022-01-25 | 杭州医学院 | Monoclonal antibody for resisting SARS-CoV-2 nucleocapsid protein and application thereof |
CN112940110B (en) * | 2021-04-14 | 2022-09-20 | 中山大学 | Anti-novel coronavirus N protein monoclonal antibody and application thereof |
-
2021
- 2021-04-08 WO PCT/SG2021/050197 patent/WO2021206638A1/en active Application Filing
- 2021-08-25 WO PCT/SG2021/050497 patent/WO2022216223A1/en active Application Filing
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060188519A1 (en) * | 2004-06-14 | 2006-08-24 | To Cheung | Peptides, antibodies, and methods for the diagnosis of SARS |
CN111647079A (en) * | 2020-07-03 | 2020-09-11 | 北部湾大学 | Neutralizing antibody for resisting novel coronavirus N protein |
CN111875700A (en) * | 2020-07-28 | 2020-11-03 | 武汉华美生物工程有限公司 | Single-chain antibody of anti SARS-COV-2 virus N protein and its use |
Non-Patent Citations (4)
Title |
---|
AHMED S.F ET AL.: "Preliminary Identification of Potential Vaccine Targets for the COVID-19 Coronavirus (SARS-CoV-2) Based on SARS-CoV Immunological Studies", VIRUSES, vol. 12, no. 3, 25 February 2020 (2020-02-25), pages 254,1 - 15, XP055823903, [retrieved on 20210618], DOI: 10.3390/V12030254 * |
DATABASE Protein 18 March 2020 (2020-03-18), "Nucleocapsid phosphoprotein [Severe acute respiratory syndrome coronavirus 2]", XP055846541, retrieved from ncbi Database accession no. QHD43423.2 * |
GRIFONI ALBA; SIDNEY JOHN; ZHANG YUN; SCHEUERMANN RICHARD H.; PETERS BJOERN; SETTE ALESSANDRO: "A Sequence Homology and Bioinformatic Approach Can Predict Candidate Targets for Immune Responses to SARS-CoV-2", CELL HOST & MICROBE, ELSEVIER, NL, vol. 27, no. 4, 16 March 2020 (2020-03-16), NL , pages 671, XP086125935, ISSN: 1931-3128, DOI: 10.1016/j.chom.2020.03.002 * |
WANG HONGYE, WU XIAN, ZHANG XIAOMEI, HOU XIN, LIANG TE, WANG DAN, TENG FEI, DAI JIAYU, DUAN HU, GUO SHUBIN, LI YONGZHE, YU XIAOBO: "SARS-CoV-2 Proteome Microarray for Mapping COVID-19 Antibody Interactions at Amino Acid Resolution", ACS CENTRAL SCIENCE, vol. 6, no. 12, 23 December 2020 (2020-12-23), pages 2238 - 2249, XP055864645, ISSN: 2374-7943, DOI: 10.1021/acscentsci.0c00742 * |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11564892B2 (en) | 2020-04-09 | 2023-01-31 | Finncure Oy | Virus-like particles for preventing the spreading and lowering the infection rate of viruses |
US12144898B2 (en) | 2020-04-09 | 2024-11-19 | Finncure Oy | Virus-like particles for preventing the spreading and lowering the infection rate of viruses |
WO2023117742A1 (en) * | 2021-12-22 | 2023-06-29 | Osivax | Vaccine compositions and their use |
CN117659180A (en) * | 2022-09-06 | 2024-03-08 | 东莞市朋志生物科技有限公司 | Anti-novel coronavirus antibody or functional fragment thereof, reagent for detecting novel coronavirus and kit |
Also Published As
Publication number | Publication date |
---|---|
WO2022216223A1 (en) | 2022-10-13 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Mengist et al. | Mutations of SARS-CoV-2 spike protein: Implications on immune evasion and vaccine-induced immunity | |
JP6774149B2 (en) | Viral particles and their production as immunogens against enterovirus infection | |
TWI700295B (en) | Antibodies specific for enteroviruses that infect humans | |
CN116437951A (en) | SARS-COV-2 vaccine | |
EA027069B1 (en) | Monoclonal antibodies capable of reacting with a plurality of influenza virus a subtypes | |
WO2021206638A1 (en) | Vaccine and/or antibody for viral infection | |
Fink et al. | Dengue virus specific IgY provides protection following lethal dengue virus challenge and is neutralizing in the absence of inducing antibody dependent enhancement | |
WO2009094755A1 (en) | Monoclonal antibodies for ebola and marburg viruses | |
JP6942309B2 (en) | Flavivir virus-like particles | |
CA2548942A1 (en) | Anti-sars monoclonal antibodies | |
US8563305B2 (en) | Rapid generation of antibodies | |
JP2021072793A (en) | Novel paramyxovirus and use of the same | |
WO2020211801A1 (en) | Recombinant classical swine fever virus | |
CN114349853B (en) | Anti-H1N 1 influenza virus hemagglutinin protein neutralizing monoclonal antibody ZJU11-01 and application thereof | |
EP3522919A1 (en) | Vaccine | |
TW201938578A (en) | Enterovirus vaccine | |
CN105085672B (en) | 3D protein specific monoclonal immunoglobulin A antibodies and compositions thereof | |
Onodera et al. | Immune-focusing properties of virus-like particles improve protective IgA responses | |
CN110294802B (en) | Monoclonal antibody 10G12 and application thereof | |
CN105085671B (en) | Monoclonal immunoglobulin G antibodies against enterovirus 3D proteins and immunogenic compositions thereof | |
CN115819568B (en) | Enterovirus A71 monoclonal antibody and application thereof | |
Zhu et al. | Dual blockages of a broad and potent neutralizing IgM antibody targeting GH loop of EV‐As | |
EP2571519A1 (en) | Marker vaccine for classical swine fever | |
WO2016115664A1 (en) | 3d protein-specific monoclonal immunoglobulin a antibodies | |
WO2016115665A1 (en) | Immunoglobulin g monoclonal antibodies against 3d proteins of enteroviruses |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21784210 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21784210 Country of ref document: EP Kind code of ref document: A1 |