WO2024220043A1 - A transmission-blocking composition against plasmodium vivax - Google Patents
A transmission-blocking composition against plasmodium vivax Download PDFInfo
- Publication number
- WO2024220043A1 WO2024220043A1 PCT/TH2023/000006 TH2023000006W WO2024220043A1 WO 2024220043 A1 WO2024220043 A1 WO 2024220043A1 TH 2023000006 W TH2023000006 W TH 2023000006W WO 2024220043 A1 WO2024220043 A1 WO 2024220043A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- polynucleotides
- vaccine composition
- malaria
- subject
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 41
- 241000223810 Plasmodium vivax Species 0.000 title claims description 35
- 229960005486 vaccine Drugs 0.000 claims abstract description 46
- 108091033319 polynucleotide Proteins 0.000 claims abstract description 44
- 102000040430 polynucleotide Human genes 0.000 claims abstract description 44
- 239000002157 polynucleotide Substances 0.000 claims abstract description 44
- 201000004792 malaria Diseases 0.000 claims abstract description 28
- 208000015181 infectious disease Diseases 0.000 claims abstract description 23
- 230000028993 immune response Effects 0.000 claims abstract description 17
- 239000011241 protective layer Substances 0.000 claims abstract description 16
- 150000002632 lipids Chemical class 0.000 claims abstract description 14
- 239000002105 nanoparticle Substances 0.000 claims abstract description 14
- 230000001939 inductive effect Effects 0.000 claims abstract description 12
- 239000002671 adjuvant Substances 0.000 claims abstract description 10
- 239000007791 liquid phase Substances 0.000 claims abstract description 8
- NRJAVPSFFCBXDT-HUESYALOSA-N 1,2-distearoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCCCC NRJAVPSFFCBXDT-HUESYALOSA-N 0.000 claims description 24
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 claims description 24
- 238000000034 method Methods 0.000 claims description 15
- DRTQHJPVMGBUCF-XVFCMESISA-N Uridine Chemical group O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-XVFCMESISA-N 0.000 claims description 14
- 229920001223 polyethylene glycol Polymers 0.000 claims description 14
- -1 cationic lipid Chemical class 0.000 claims description 13
- 235000012000 cholesterol Nutrition 0.000 claims description 12
- 229930185560 Pseudouridine Natural products 0.000 claims description 8
- PTJWIQPHWPFNBW-UHFFFAOYSA-N Pseudouridine C Natural products OC1C(O)C(CO)OC1C1=CNC(=O)NC1=O PTJWIQPHWPFNBW-UHFFFAOYSA-N 0.000 claims description 8
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 claims description 8
- WGDUUQDYDIIBKT-UHFFFAOYSA-N beta-Pseudouridine Natural products OC1OC(CN2C=CC(=O)NC2=O)C(O)C1O WGDUUQDYDIIBKT-UHFFFAOYSA-N 0.000 claims description 8
- PTJWIQPHWPFNBW-GBNDHIKLSA-N pseudouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1C1=CNC(=O)NC1=O PTJWIQPHWPFNBW-GBNDHIKLSA-N 0.000 claims description 8
- UVBYMVOUBXYSFV-XUTVFYLZSA-N 1-methylpseudouridine Chemical compound O=C1NC(=O)N(C)C=C1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 UVBYMVOUBXYSFV-XUTVFYLZSA-N 0.000 claims description 7
- UVBYMVOUBXYSFV-UHFFFAOYSA-N 1-methylpseudouridine Natural products O=C1NC(=O)N(C)C=C1C1C(O)C(O)C(CO)O1 UVBYMVOUBXYSFV-UHFFFAOYSA-N 0.000 claims description 7
- DRTQHJPVMGBUCF-PSQAKQOGSA-N beta-L-uridine Natural products O[C@H]1[C@@H](O)[C@H](CO)O[C@@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-PSQAKQOGSA-N 0.000 claims description 7
- DRTQHJPVMGBUCF-UHFFFAOYSA-N uracil arabinoside Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-UHFFFAOYSA-N 0.000 claims description 7
- 229940045145 uridine Drugs 0.000 claims description 7
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 claims description 6
- 239000003795 chemical substances by application Substances 0.000 claims description 5
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 claims description 4
- 238000011144 upstream manufacturing Methods 0.000 claims description 4
- 239000003242 anti bacterial agent Substances 0.000 claims description 3
- 239000003963 antioxidant agent Substances 0.000 claims description 3
- 239000006172 buffering agent Substances 0.000 claims description 3
- 239000002738 chelating agent Substances 0.000 claims description 3
- 239000002577 cryoprotective agent Substances 0.000 claims description 3
- 238000007918 intramuscular administration Methods 0.000 claims description 3
- 239000011780 sodium chloride Substances 0.000 claims description 3
- 238000007920 subcutaneous administration Methods 0.000 claims description 3
- 239000007864 aqueous solution Substances 0.000 claims description 2
- OGBUMNBNEWYMNJ-UHFFFAOYSA-N batilol Chemical class CCCCCCCCCCCCCCCCCCOCC(O)CO OGBUMNBNEWYMNJ-UHFFFAOYSA-N 0.000 claims description 2
- 235000011187 glycerol Nutrition 0.000 claims description 2
- 239000000243 solution Substances 0.000 claims description 2
- 240000009188 Phyllostachys vivax Species 0.000 abstract 1
- 108090000623 proteins and genes Proteins 0.000 description 32
- 108020004999 messenger RNA Proteins 0.000 description 30
- 102000004169 proteins and genes Human genes 0.000 description 28
- 238000002255 vaccination Methods 0.000 description 27
- 241000699670 Mus sp. Species 0.000 description 24
- 230000005540 biological transmission Effects 0.000 description 19
- 108090000765 processed proteins & peptides Proteins 0.000 description 14
- 241000255925 Diptera Species 0.000 description 10
- 230000011664 signaling Effects 0.000 description 10
- 238000011161 development Methods 0.000 description 8
- 230000005875 antibody response Effects 0.000 description 7
- 230000000903 blocking effect Effects 0.000 description 7
- 108010084884 GDP-mannose transporter Proteins 0.000 description 6
- 238000002649 immunization Methods 0.000 description 6
- 230000003053 immunization Effects 0.000 description 6
- 210000003936 merozoite Anatomy 0.000 description 6
- 230000004048 modification Effects 0.000 description 6
- 238000012986 modification Methods 0.000 description 6
- 230000001603 reducing effect Effects 0.000 description 6
- 108010002350 Interleukin-2 Proteins 0.000 description 5
- 241000223960 Plasmodium falciparum Species 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 244000045947 parasite Species 0.000 description 5
- 108010058597 HLA-DR Antigens Proteins 0.000 description 4
- 102000006354 HLA-DR Antigens Human genes 0.000 description 4
- 108091028043 Nucleic acid sequence Proteins 0.000 description 4
- 108010076504 Protein Sorting Signals Proteins 0.000 description 4
- 102000036639 antigens Human genes 0.000 description 4
- 108091007433 antigens Proteins 0.000 description 4
- 230000036039 immunity Effects 0.000 description 4
- 229940038694 mRNA-based vaccine Drugs 0.000 description 4
- 230000001681 protective effect Effects 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 241000256186 Anopheles <genus> Species 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- 206010035503 Plasmodium vivax infection Diseases 0.000 description 3
- 201000009976 Plasmodium vivax malaria Diseases 0.000 description 3
- 208000005469 Vivax Malaria Diseases 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 210000004027 cell Anatomy 0.000 description 3
- 230000036755 cellular response Effects 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 150000007523 nucleic acids Chemical class 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 210000004988 splenocyte Anatomy 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- GJOHLWZHWQUKAU-UHFFFAOYSA-N 5-azaniumylpentan-2-yl-(6-methoxyquinolin-8-yl)azanium;dihydrogen phosphate Chemical compound OP(O)(O)=O.OP(O)(O)=O.N1=CC=CC2=CC(OC)=CC(NC(C)CCCN)=C21 GJOHLWZHWQUKAU-UHFFFAOYSA-N 0.000 description 2
- 102100031126 6-phosphogluconolactonase Human genes 0.000 description 2
- 108010029731 6-phosphogluconolactonase Proteins 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 2
- 208000035473 Communicable disease Diseases 0.000 description 2
- 241000256113 Culicidae Species 0.000 description 2
- 108010018962 Glucosephosphate Dehydrogenase Proteins 0.000 description 2
- 102000043131 MHC class II family Human genes 0.000 description 2
- 108091054438 MHC class II family Proteins 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- LBHLFPGPEGDCJG-UHFFFAOYSA-N N(4)-{2,6-dimethoxy-4-methyl-5-[3-(trifluoromethyl)phenoxy]quinolin-8-yl}pentane-1,4-diamine Chemical compound COC=1C=C(NC(C)CCCN)C2=NC(OC)=CC(C)=C2C=1OC1=CC=CC(C(F)(F)F)=C1 LBHLFPGPEGDCJG-UHFFFAOYSA-N 0.000 description 2
- 101710160102 Outer membrane protein B Proteins 0.000 description 2
- 102000002067 Protein Subunits Human genes 0.000 description 2
- 108010001267 Protein Subunits Proteins 0.000 description 2
- 208000007502 anemia Diseases 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 229940028617 conventional vaccine Drugs 0.000 description 2
- 238000012937 correction Methods 0.000 description 2
- 230000005574 cross-species transmission Effects 0.000 description 2
- 231100000676 disease causative agent Toxicity 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000011835 investigation Methods 0.000 description 2
- 108700021021 mRNA Vaccine Proteins 0.000 description 2
- 229940126582 mRNA vaccine Drugs 0.000 description 2
- 229940124735 malaria vaccine Drugs 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- LXCFILQKKLGQFO-UHFFFAOYSA-N methylparaben Chemical compound COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000003472 neutralizing effect Effects 0.000 description 2
- 238000001543 one-way ANOVA Methods 0.000 description 2
- 210000003250 oocyst Anatomy 0.000 description 2
- 244000052769 pathogen Species 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 229960005179 primaquine Drugs 0.000 description 2
- 230000005180 public health Effects 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 229950000856 tafenoquine Drugs 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-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
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- 150000005012 8-aminoquinolines Chemical class 0.000 description 1
- 206010000234 Abortion spontaneous Diseases 0.000 description 1
- 239000012103 Alexa Fluor 488 Substances 0.000 description 1
- 238000011725 BALB/c mouse Methods 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 208000025721 COVID-19 Diseases 0.000 description 1
- 206010010538 Congenital malaria Diseases 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 206010018910 Haemolysis Diseases 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- 206010024769 Local reaction Diseases 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-N 0.000 description 1
- 241000209140 Triticum Species 0.000 description 1
- 235000021307 Triticum Nutrition 0.000 description 1
- 241000907316 Zika virus Species 0.000 description 1
- 208000020329 Zika virus infectious disease Diseases 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000009824 affinity maturation Effects 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 1
- 230000008350 antigen-specific antibody response Effects 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000033077 cellular process Effects 0.000 description 1
- 150000001860 citric acid derivatives Chemical class 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 210000000172 cytosol Anatomy 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 238000002716 delivery method Methods 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 230000000694 effects Effects 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 210000001163 endosome Anatomy 0.000 description 1
- 230000003325 follicular Effects 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 230000008588 hemolysis Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 239000012678 infectious agent Substances 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 238000002743 insertional mutagenesis Methods 0.000 description 1
- 229940047124 interferons Drugs 0.000 description 1
- 230000004073 interleukin-2 production Effects 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 208000018773 low birth weight Diseases 0.000 description 1
- 231100000533 low birth weight Toxicity 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 230000008774 maternal effect Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 210000001806 memory b lymphocyte Anatomy 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- 238000001000 micrograph Methods 0.000 description 1
- 208000015994 miscarriage Diseases 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 238000012758 nuclear staining Methods 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 150000003833 nucleoside derivatives Chemical class 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 230000020477 pH reduction Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000035935 pregnancy Effects 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 229940023143 protein vaccine Drugs 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 230000007420 reactivation Effects 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 229940126583 recombinant protein vaccine Drugs 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 230000000979 retarding effect Effects 0.000 description 1
- 238000011808 rodent model Methods 0.000 description 1
- 230000001568 sexual effect Effects 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 208000000995 spontaneous abortion Diseases 0.000 description 1
- 230000007480 spreading Effects 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 229940031626 subunit vaccine Drugs 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 238000001262 western blot Methods 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
- A61K39/002—Protozoa antigens
- A61K39/015—Hemosporidia antigens, e.g. Plasmodium 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
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/53—DNA (RNA) vaccination
-
- 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
- A61K2039/55555—Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
-
- 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
-
- 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
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
- A61K2039/575—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/51—Nanocapsules; Nanoparticles
- A61K9/5107—Excipients; Inactive ingredients
- A61K9/5123—Organic compounds, e.g. fats, sugars
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
- A61P33/02—Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
- A61P33/06—Antimalarials
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- the present disclosure relates to a vaccine against transmission of malaria infection.
- the disclosed vaccine is a mRNA-based vaccine which is effective in blocking transmission or spread of malaria infection between two species, such as transmission between a human host and mosquitos.
- P. vivax Malaria represents the global public health concern.
- the causative agents of this disease are Plasmodium parasites which are transmitted by female Anopheles mosquitos. Plasmodium falciparum and Plasmodium vivax play the roles as the major parasite species for human malaria. 3.5 billion people are currently at risk of P. vivax infection [1].
- Growing number of studies have indicated that P. vivax can potentially cause complications, and death particularly in pregnant women and young children [2].
- infection with P. vivax is associated with chronic anemia and can lead to maternal anemia, miscarriage, low birth weight and congenital malaria [3].
- vivax infection is more complicated due to the parasite’s ability to become dormant in the liver of infected individuals for months to years before reactivation [4].
- the reservoir of these dormant parasites known as hypnozoites, not only extends the clinical attack but also sustains the transmission causing the great epidemiological success around the globe [4].
- P. vivax is more efficient at transmitting to mosquitos [5,6].
- drug applicable for killing hypnozoites are limited to 8 -aminoquinolines primaquine and tafenoquine, which are known to result in acute hemolysis in people with glucose-6-phosphate dehydrogenase (G6PD)- deficiency.
- G6PD glucose-6-phosphate dehydrogenase
- primaquine and tafenoquine are contraindicated during pregnancy and breast-feeding. These drugs are not recommended in children less than six months old. Alternative methods to control, cure and/or even to curb spreading of vivax malaria are thus needed [7].
- Vaccines are one of the most successful and cost-effective public health tools for eradicating, containing, and reducing infectious diseases.
- a malaria vaccine that targets sexual or mosquito stage parasites aiming to reduce man-to-mosquito transmission, i.e. a transmission blocking vaccine (TBV), is thus considered an important tool for P. vivax elimination [8]. Because an effective TBV is expected to reduce the overall malaria transmission, it will also have an impact on the intensity of mosquito-to-man transmission as well as the size of hypnozoite reservoir in the population.
- Pvs25 The Plasmodium vivax Ookinete Surface Protein, Pvs25, a protein antigen expressed on the surface of ookinetes, has been proposed as a leading TBV candidate [9-11] due to its strong transmission blocking efficacy [12, 13].
- Study assessing a recombinant Pvs25 formulated with anhydro gel targeting P. vivax malaria transmission revealed that the vaccine significantly reduced the parasite number in mosquitoes but the titer was not sufficient for being an effective vaccine [14].
- Another study conducted to test Pvs25/Montanide ISA 51 based transmission blocking vaccine observed a high and functionally active antibody response. Nonetheless, this study encountered the concern of local reactions that ceases further development of the formulation [15]. In view of that, a new and novel vaccination platform is therefore urgently needed, particularly a Pvs25-based transmission blocking vaccine capable of inducing potent and long-lasting transmission blocking immunity yet being safe for clinical application.
- nucleoside-modified mRNA-LNP vaccine elicited potent and sustained protective neutralizing antibody responses against Zika and influenza viruses in mice and non-human primates [18- 20]. Moreover, comparative studies confirmed that the nucleoside-modified mRNA-LNP vaccine outperformed conventional vaccine formats such as MF59-adjuvanted protein subunit and inactivated pathogen vaccines [18].
- the nucleoside-modified mRNA-LNP vaccine has the unique ability to potently induce T follicular helper cells [18] that are critical drivers of antibody affinity maturation and the generation of protective neutralizing antibodies [21].
- nucleoside modified mRNA vaccines encoding P. falciparum CSP was found to be immunogenic in mice and protective in homologous and heterologous transgenic rodent models making this a compelling platform for further malaria vaccine development [22], particularly a vaccine effective against P. vivax infection.
- One object of the present disclosure is to provide a composition capable of eliciting an immune response in a subject against infection of malaria, particularly caused by P. vivax, upon administrating the disclosed composition to the subject through the predetermined route.
- Further object of the present disclosure aims to offer a vaccine composition effective in curbing, prohibiting and/or reducing the likelihood of cross-species transmission of malaria which, but not limited to, P. vivax is the causative agent.
- the disclosed composition can induce an immune response in a subject received the vaccine for retarding, prohibiting and/or arresting development of merozoites and/or hypnozoites in the subject and/or mosquitoes fed on the subject through one or more antibodies generated by the subject in association with the vaccine composition received.
- the subject may not have infected with malaria.
- More object of the present disclosure associates to a mRNA-based vaccine composition designed to arrest development of merozoites and/or hypnozoites in a mammalian subject upon administrating one or more pharmaceutically effective dosage of the vaccine composition within a period of time.
- Another object of the present disclosure is to offer a method for inducing an immune response in a subject by way of administrating a mRNA-based vaccine composition prepared using polynucleotide sequence of Plasmodium vivax ookinete surface protein antigen, namely Pvs25 protein.
- the present disclosure in which one of the embodiments of the present disclosure is a vaccine composition for prohibiting cross-species infection of malaria caused by P. vivax in a subject, or even mosquitoes fed on the subject potentially carrying merozoites and/or hypnozoites of P. vivax.
- the vaccine composition comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 or SEQ ID No.
- the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
- the polynucleotides of the disclosed composition have uridine substituted or replaced by any one or combination of pseudouridine and 1-methyl- pseudouridine.
- the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. More preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40- 60: 5-15: 30-50: 1-5.
- the polynucleotide further comprises a region encoding for at least one signaling peptide, which is incorporated upstream of the sequence of SEQ ID No. 1 or SEQ ID No. 2 in an expression construct bearing the polynucleotide sequence of SEQ ID No. 1 or SEQ ID No. 2.
- the incorporation of the signaling peptide may facilitate better recognition of the expressed peptide by the host or subject receiving the vaccine in some embodiments.
- the signaling peptide derived from either one of signaling peptide of MHC class II. Some examples of the signaling peptide used are detailed in SEQ ID No. 5 or SEQ ID No. 6.
- sequence of SEQ ID No. 1 and/or SEQ ID No. 2 is derived from strain Sal I of P. vivax.
- the disclosed method comprises administrating a vaccine composition to the subject by way of intramuscular route or subcutaneous route that the vaccine composition comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 and/or SEQ ID No. 2, the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
- the polynucleotides have uridine substituted by any one or combination of pseudouridine and 1- methy 1-p seudouridine .
- the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG.
- DSPC distearoylphosphatidylcholine
- cholesterol and PEG are in a molar ratio of 40-60: 5-15: 30-50: 1-5.
- Fig. 1 is a gel picture showing various constructs with different modifications as explained in Example 1 were successfully expressed for investigation through subsequent experiments;
- Fig. 2 are graphs showing results about antigen specific antibody response in mice immunized with nucleoside-modified constructs incorporated with the polynucleotides sequence of Pvs25;
- Fig. 3 are micrographs showing the results obtained from testing the pooled plasma of the immunized mice against native antigen on the surface of P. vivax ookinetes that the uppermost panels show nuclear staining with DAPI followed by the second row showing staining with anti-mouse IgG Alexa Fluor 488 secondary antibody, the third row presenting the merged images, and the bottom row showing the differential Interference Contrast (DIC) images (points represents individual mice and horizontal lines denote geometric mean with 95% CI as well as vertical lines represent error bar)
- DIC differential Interference Contrast
- Fig. 4 is a graph showing the functional activity of antisera induced by Pvs25 mRNA-ENP vaccines with TRA represents the % reduction in the mean oocyst density in the presence of each specified immune-serum relative to the non-immune serum (using One-way ANOVA with Bonferroni correction, *p ⁇ 0.05);
- Fig. 6 includes graphs showing (a) activity indices of the anti-Pvs25 and (b) IgG2a/IgGl ratio of the anti-Pvs25 (using One-way ANOVA with Bonferroni correction, *p ⁇ 0.05);
- Fig. 7 includes graphs showing the frequency of production of different interleukins and interferons by (a) CD4-T cells and (b) CD8-T cells of the mice immunized using different constructs prepared;
- Fig. 8 is a graph showing the immune response induced in mice immunized with different constructs prepared over a period of 7 months.
- Fig. 9 includes graphs showing transmission reducing activity (TRA) induced by different immunization regimens at the first month and the seventh month;
- Fig. 10 is a graph showing relationship between TRA (from 9) and total IgG produced.
- Fig. 11 is a listing showing SEQ ID No. 1 and SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No.5, and SEQ ID No.6.
- polynucleotide or “nucleic acid” as used herein designates mRNA, RNA, cRNA, cDNA or DNA.
- the term typically refers to oligonucleotides greater than 30 nucleotide residues in length.
- gene may refer to a DNA sequence with functional significance. It can be a native nucleic acid sequence, or a recombinant nucleic acid sequence derived from natural source or synthetic construct. The term “gene” may also be used to refer to, for example and without limitation, a cDNA and/or an mRNA encoded by or derived from, directly or indirectly, genomic DNA sequence.
- a composition for prohibiting cross-species infection of malaria caused by P. vivax in a subject is provided.
- the composition is configured to inhibit, arrest, prohibit and/or cease development of merozoites and/or hypnozoites potentially carried by a human subject and/or a transmitting agent such as female mosquitoes of the genus Anopheles.
- the disclosed vaccine composition generally comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 and/or SEQ ID No.
- the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
- the inventors of the present disclosure employ a polynucleotide sequence of Plasmodium vivax ookinete surface protein antigen, namely Pvs25, protein as the template or base template for preparing the polynucleotide sequence SEQ ID No. 1 in the form of mRNA.
- mRNA is the minimal genetic vector; therefore, anti-vector immunity can be avoided, and mRNA vaccines can be administered repeatedly.
- the mRNA platform of the present disclosure is degradable by normal cellular processes, and it in vivo half-life can be regulated through various modifications introduced and delivery methods used to further reduce the likelihood of the occurrence of any undesired outcome.
- the polynucleotide used in the present disclosure for preparing the vaccine composition can be of natural, preferably further processed by one or more purification steps, or synthetic origin.
- the Pvs25 sequence used can be with or without modification depending on the designated outcome to be attained as further described in the following.
- the polynucleotide has uridine, which is found in natural or general mRNA template, substituted by any one or combination of pseudouridine and 1-methyl-pseudouridine.
- pseudouridine and/or 1-methyl- pseudouridine incorporated in the disclosed composition ensures higher level of protein expression in the subject.
- substitution of uridine by pseudouridine and/or 1-methyl- pseudouridine also lowers the risk of serious inflammatory responses in the subject after receiving the disclosed vaccine composition.
- the polynucleotide used may comprise a I130T mutation as an additional modification made towards the polynucleotide sequence used to improve overall performance of the disclosed composition.
- the sequence of the mentioned polynucleotide having the intended mutation is disclosed in SEQ ID No. 2 of Fig 11.
- the included mutation enhances the transmission blocking activity towards other variants of Plasmodium parasites, particularly a variant of Plasmodium vivax carrying the aforesaid mutation.
- sequences of the peptides correspondingly encoded by the SEQ ID No. 1 and SEQ ID No. 2 are shown in SEQ ID No. 3 and SEQ ID No. 4.
- the polynucleotide further comprises a region encoding for at least one signaling peptide, which is incorporated upstream of the sequence of SEQ ID No. 1 or SEQ ID No. 2 in an expression construct bearing the polynucleotide sequence of SEQ ID No. 1 or SEQ ID No. 2.
- the incorporation of the signaling peptide may facilitate better recognition of the expressed peptide by the host or subject receiving the vaccine in some embodiments.
- the signaling peptide derived from either one of signaling peptide of MHC class II.
- Some examples of the signaling peptide used are detailed in SEQ ID No. 5 or SEQ ID No. 6.
- each of the plurality of polynucleotides further comprises SEQ ID No. 5 and/or SEQ ID No. 6 located upstream or downstream of SEQ ID No. 1 or SEQ ID No. 2.
- a liquid phase composed of LNPs are used to form a protective layer to encapsulate, enclose, or wrap the plurality.
- the LNPs self-assemble into 80- 100 nm particles.
- the protective layer established though the LNPs creates a neutral surface in relation to the cellular environment allowing the encapsulated polynucleotides to shunt extensive binding to serum proteins prior to expression.
- the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. More preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5-15: 30-50: 1-5.
- the adjuvant can be any one or combination of an aqueous solution, a saline solution, Ringer’s solution, isotonic sodium chloride, synthetic monoglycerides, synthetic diglycerides, polyethylene glycols, glycerin, propylene glycol, antibacterial agents, antioxidants, chelating agents, buffering agents, tonicity modifying agents, and cryoprotectants.
- the antibacterial agents can be benzyl alcohol, methyl paraben, and the like.
- the antioxidants can be ascorbic acid, sodium bisulfite, and the like.
- the buffering agents can be acetates, citrates or phosphates, and the like.
- the tonicity modifying agents can be sodium chloride, dextrose, and the like.
- the cryoprotectants can be sucrose, trehalose, and the like.
- the chelating agents such as ethylenediaminetetraacetic acid can be used as well.
- Another aspect of the present disclosure relates to a method of inducing an immune response in a subject reactive against infection of malaria caused by P. vivax.
- the immune response induced shall result in production of an antibody reactive against Pvs25 antigen which in turn arrests or inhibits development of P. vivax, particularly merozoites and/or hypnozoites, in the subject.
- the antibodies produced can be transferred to malaria transmitting agents, such as female mosquitoes of the genus Anopheles, happened to feed on the blood of the subject carrying the antibodies. These antibodies are configured to attain the same outcome in the mosquitoes to arrest or inhibit development of the P. vivax merozoites and/or hypnozoites.
- the mosquitoes bearing these antibodies shall fail to effectuate transmission of malaria subsequently shrinking the pool of malaria infectious agents available.
- the disclosed method is able to reduce or prevent the crossspecies transmission of malaria particularly caused by P. vivax.
- the disclosed method preferably comprises the steps of administrating a vaccine composition to the subject by way of intramuscular route or subcutaneous route. More specifically, the vaccine composition used in the disclosed method comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No.
- the polynucleotides being configured to express in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
- the polynucleotides of the vaccine composition used in the disclosed method has uridine substituted by any one or combination of pseudouridine and 1-methyl-pseudouridine to improve expression of the antigenic protein in the subject.
- the lipid nanoparticles found in the vaccine composition used for the disclosed method comprises cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. More preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5-15: 30-50: 1-5.
- the other two (Pvs25A IL-2 SP and Pvs25A HLA-DR SP) have exogenous signal peptide sequences (human IL-2 and HLA-DR) which are known to enhance protein expression.
- the other four constructs encode Pvs25 with the C terminal GPI anchor.
- Pvs25F encodes the full-length sequence of the Pvs25 gene from Sal I.
- the remaining three constructs (Pvs25F I130T, Pvs25F IL-2 SP, and Pvs25F HLA-DR SP) contain the full-length sequence of Pvs25 with a) the I130T mutation, b) the IL-2 signal peptide, or c) the HLA-DR signal peptide.
- Cells transfected with polycytidine (PolyC) RNA-LNP was used as the negative control.
- Recombinant protein (Pvs25A) produced by the wheat germ cell free system was used as the positive control.
- mice were immunized using the various Pvs25 mRNA-LNPs constructs prepared.
- the results are present in Fig. 2.
- the direct membrane feeding assay was performed to evaluate transmission reducing activity (TRA) of immunized mouse antisera at different dilutions.
- mice were vaccinated with homologous and heterologous prime-boost regimens (10 pg mRNA or 10 pg recombinant protein vaccine, 4 weeks between the prime and boost).
- 10 pg Poly(C) RNA-LNP or Montanide ISA-51 VG was administered as negative controls for each vaccination group.
- Sera from the control groups had no detectable Pvs25-specific antibody response by ELISA (Fig. 5).
- the mRNA/mRNA homologous prime-boost vaccination generated the strongest Pvs25-specific antibody response with GMT of -140,000.
- the protein/protein homologous vaccination had GMT of -40,000.
- the protein/mRNA vaccination had GMT of -80,000 and the mRNA/protein vaccination yielded GMT of -46,000.
- the present disclosure further characterized the quality of Pvs25-specific antibodies.
- IgGl and IgG2a subclasses expressed as the IgG2a/IgGl ratio and antigen- antibody avidity index.
- pooled serum was used to determine the IgG2a/IgGl ratio and the avidity index (Fig. 6a and 6b).
- the IgG subclass patterns of all vaccination regimens were similar with the IgG2a/IgGl ratio of -0.75.
- the avidity indices were also similar across the groups.
- mouse splenocytes from the homologous and heterologous prime -boost experiment were evaluated for cellular immune responses.
- the mRNA/mRNA homologous prime-boost vaccination induced the most robust Pvs25-specific CD4+ and CD8+ T cell responses as measured by IFN-y and IL-2 production, while the protein/protein vaccination barely induced T cells.
- the results were summarized respectively in Fig. 7a and 7b.
- the mRNA/mRNA homologous vaccination elicited the strongest memory B cell response whereas this was almost absent in the protein/protein homologous vaccination.
- the protein/mRNA heterologous vaccination gave positive but intermediate cellular responses while the mRNA/protein vaccination elicited very low cellular response similar to the protein/protein vaccination. The result is presented in Fig 7.
- the present disclosure also used ELISA to determine Pvs25 antibodies response in the mice immunized by the four different constructs prepared over a period of 7 months with measurements being conducted at each of the month.
- Nine mice per group were maintained for 7 months after the booster dose at Month 0.
- the results are summarized in Fig. 8.
- Pvs25 antibody levels were followed monthly for 7 months post boost vaccination to assess the durability of antibody responses (Fig. 8).
- the antibody levels peaked at 1 month post boost in all vaccination groups and declined over the subsequent months.
- GMTs were highest in the mRNA/mRNA group and the lowest in the protein/protein group.
- the antibody level had a GMT of 36,000 for mRNA/mRNA, 2,300 for protein/protein, 25,000 for protein/mRNA, and 18,000 for mRNA/protein.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Immunology (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
The present disclosure relates to a composition for prohibiting cross-species infection of malaria caused by P. vivax in a subject. Preferably, the disclosed composition is prepared in the form of a vaccine, which comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 or SEQ ID No. 2, the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
Description
A TRANSMISSION-BLOCKING COMPOSITION AGAINST PLASMODIUM VIVAX
Technical Field
The present disclosure relates to a vaccine against transmission of malaria infection. In more specific, the disclosed vaccine is a mRNA-based vaccine which is effective in blocking transmission or spread of malaria infection between two species, such as transmission between a human host and mosquitos.
Background
Malaria represents the global public health concern. The causative agents of this disease are Plasmodium parasites which are transmitted by female Anopheles mosquitos. Plasmodium falciparum and Plasmodium vivax play the roles as the major parasite species for human malaria. 3.5 billion people are currently at risk of P. vivax infection [1]. Growing number of studies have indicated that P. vivax can potentially cause complications, and death particularly in pregnant women and young children [2]. Particularly, infection with P. vivax is associated with chronic anemia and can lead to maternal anemia, miscarriage, low birth weight and congenital malaria [3]. Compared to Plasmodium falciparum, management of P. vivax infection is more complicated due to the parasite’s ability to become dormant in the liver of infected individuals for months to years before reactivation [4]. The reservoir of these dormant parasites, known as hypnozoites, not only extends the clinical attack but also sustains the transmission causing the great epidemiological success around the globe [4]. In addition, P. vivax is more efficient at transmitting to mosquitos [5,6]. Currently, drug applicable for killing hypnozoites are limited to 8 -aminoquinolines primaquine and tafenoquine, which are known to result in acute hemolysis in people with glucose-6-phosphate dehydrogenase (G6PD)- deficiency. In addition, primaquine and tafenoquine are contraindicated during pregnancy and breast-feeding. These drugs are not recommended in children less than six months old. Alternative methods to control, cure and/or even to curb spreading of vivax malaria are thus needed [7].
Vaccines are one of the most successful and cost-effective public health tools for eradicating, containing, and reducing infectious diseases. A malaria vaccine that targets sexual or mosquito stage parasites aiming to reduce man-to-mosquito transmission, i.e. a transmission blocking vaccine (TBV), is thus considered an important tool for P. vivax elimination [8]. Because an
effective TBV is expected to reduce the overall malaria transmission, it will also have an impact on the intensity of mosquito-to-man transmission as well as the size of hypnozoite reservoir in the population. The Plasmodium vivax Ookinete Surface Protein, Pvs25, a protein antigen expressed on the surface of ookinetes, has been proposed as a leading TBV candidate [9-11] due to its strong transmission blocking efficacy [12, 13]. Study assessing a recombinant Pvs25 formulated with anhydro gel targeting P. vivax malaria transmission revealed that the vaccine significantly reduced the parasite number in mosquitoes but the titer was not sufficient for being an effective vaccine [14]. Another study conducted to test Pvs25/Montanide ISA 51 based transmission blocking vaccine observed a high and functionally active antibody response. Nonetheless, this study encountered the concern of local reactions that ceases further development of the formulation [15]. In view of that, a new and novel vaccination platform is therefore urgently needed, particularly a Pvs25-based transmission blocking vaccine capable of inducing potent and long-lasting transmission blocking immunity yet being safe for clinical application.
Conventional vaccine approaches, such as live attenuated and inactivated pathogens and protein subunit vaccines, employed for development of vaccine against P. vivax in the earlier studies have encountered critical setbacks as mentioned above. Nonetheless, in recent years, various forms of mRNA-based vaccines have proven to be highly effective against cancer and infectious diseases [16] compared to the conventional approaches. One of the most promising vaccine platforms comprises nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNPs). The nucleoside-modified mRNA-LNP vaccines developed by Pfizer/BioNTech and Moderna have demonstrated the effectiveness to curb Covid-19 [17]. A single dose of nucleoside-modified mRNA-LNP vaccine elicited potent and sustained protective neutralizing antibody responses against Zika and influenza viruses in mice and non-human primates [18- 20]. Moreover, comparative studies confirmed that the nucleoside-modified mRNA-LNP vaccine outperformed conventional vaccine formats such as MF59-adjuvanted protein subunit and inactivated pathogen vaccines [18]. The nucleoside-modified mRNA-LNP vaccine has the unique ability to potently induce T follicular helper cells [18] that are critical drivers of antibody affinity maturation and the generation of protective neutralizing antibodies [21]. Recently, nucleoside modified mRNA vaccines encoding P. falciparum CSP was found to be immunogenic in mice and protective in homologous and heterologous transgenic rodent
models making this a compelling platform for further malaria vaccine development [22], particularly a vaccine effective against P. vivax infection.
Summary
One object of the present disclosure is to provide a composition capable of eliciting an immune response in a subject against infection of malaria, particularly caused by P. vivax, upon administrating the disclosed composition to the subject through the predetermined route.
Further object of the present disclosure aims to offer a vaccine composition effective in curbing, prohibiting and/or reducing the likelihood of cross-species transmission of malaria which, but not limited to, P. vivax is the causative agent. Particularly, the disclosed composition can induce an immune response in a subject received the vaccine for retarding, prohibiting and/or arresting development of merozoites and/or hypnozoites in the subject and/or mosquitoes fed on the subject through one or more antibodies generated by the subject in association with the vaccine composition received. The subject may not have infected with malaria.
More object of the present disclosure associates to a mRNA-based vaccine composition designed to arrest development of merozoites and/or hypnozoites in a mammalian subject upon administrating one or more pharmaceutically effective dosage of the vaccine composition within a period of time.
Still, another object of the present disclosure is to offer a method for inducing an immune response in a subject by way of administrating a mRNA-based vaccine composition prepared using polynucleotide sequence of Plasmodium vivax ookinete surface protein antigen, namely Pvs25 protein.
At least one of the preceding objects is met, in whole or in part, by the present disclosure, in which one of the embodiments of the present disclosure is a vaccine composition for prohibiting cross-species infection of malaria caused by P. vivax in a subject, or even mosquitoes fed on the subject potentially carrying merozoites and/or hypnozoites of P. vivax. Preferably, the vaccine composition comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 or SEQ ID No. 2, the polynucleotides being
expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
In some embodiments, the polynucleotides of the disclosed composition have uridine substituted or replaced by any one or combination of pseudouridine and 1-methyl- pseudouridine.
In more embodiments, the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. More preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40- 60: 5-15: 30-50: 1-5.
For some embodiments, the polynucleotide further comprises a region encoding for at least one signaling peptide, which is incorporated upstream of the sequence of SEQ ID No. 1 or SEQ ID No. 2 in an expression construct bearing the polynucleotide sequence of SEQ ID No. 1 or SEQ ID No. 2. The incorporation of the signaling peptide may facilitate better recognition of the expressed peptide by the host or subject receiving the vaccine in some embodiments. According to several preferred embodiments, the signaling peptide derived from either one of signaling peptide of MHC class II. Some examples of the signaling peptide used are detailed in SEQ ID No. 5 or SEQ ID No. 6.
For more embodiments, the sequence of SEQ ID No. 1 and/or SEQ ID No. 2 is derived from strain Sal I of P. vivax.
Another aspect of the present disclosure involves a method of inducing an immune response in a subject reactive against infection of malaria caused by P. vivax. Essentially, the disclosed method comprises administrating a vaccine composition to the subject by way of intramuscular route or subcutaneous route that the vaccine composition comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 and/or SEQ ID No. 2, the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles
configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant. Preferably, the polynucleotides have uridine substituted by any one or combination of pseudouridine and 1- methy 1-p seudouridine .
According to several embodiments of the disclosed method, the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. Preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5-15: 30-50: 1-5.
Brief Description of the Drawings
Fig. 1 is a gel picture showing various constructs with different modifications as explained in Example 1 were successfully expressed for investigation through subsequent experiments;
Fig. 2 are graphs showing results about antigen specific antibody response in mice immunized with nucleoside-modified constructs incorporated with the polynucleotides sequence of Pvs25;
Fig. 3 are micrographs showing the results obtained from testing the pooled plasma of the immunized mice against native antigen on the surface of P. vivax ookinetes that the uppermost panels show nuclear staining with DAPI followed by the second row showing staining with anti-mouse IgG Alexa Fluor 488 secondary antibody, the third row presenting the merged images, and the bottom row showing the differential Interference Contrast (DIC) images (points represents individual mice and horizontal lines denote geometric mean with 95% CI as well as vertical lines represent error bar)
Fig. 4 is a graph showing the functional activity of antisera induced by Pvs25 mRNA-ENP vaccines with TRA represents the % reduction in the mean oocyst density in the presence of each specified immune-serum relative to the non-immune serum (using One-way ANOVA with Bonferroni correction, *p < 0.05);
Fig. 5 is a graph showing the Pvs25-specific antibody response in the mice using EEISA (n=17 per group) at 1 month after the prime vaccination (Month 0) and 1 month after boost vaccination (Month 1);
Fig. 6 includes graphs showing (a) activity indices of the anti-Pvs25 and (b) IgG2a/IgGl ratio of the anti-Pvs25 (using One-way ANOVA with Bonferroni correction, *p < 0.05);
Fig. 7 includes graphs showing the frequency of production of different interleukins and interferons by (a) CD4-T cells and (b) CD8-T cells of the mice immunized using different constructs prepared;
Fig. 8 is a graph showing the immune response induced in mice immunized with different constructs prepared over a period of 7 months; and
Fig. 9 includes graphs showing transmission reducing activity (TRA) induced by different immunization regimens at the first month and the seventh month;
Fig. 10 is a graph showing relationship between TRA (from 9) and total IgG produced; and
Fig. 11 is a listing showing SEQ ID No. 1 and SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No.5, and SEQ ID No.6.
Detailed Description
Hereinafter, the disclosure shall be described according to the preferred embodiments and by referring to the accompanying description and drawings. However, it is to be understood that referring the description to the preferred embodiments of the disclosure and to the drawings is merely to facilitate discussion of the various disclosed embodiments and it is envisioned that those skilled in the art may devise various modifications without departing from the scope of the appended claim.
The term "polynucleotide" or "nucleic acid" as used herein designates mRNA, RNA, cRNA, cDNA or DNA. The term typically refers to oligonucleotides greater than 30 nucleotide residues in length.
The term "gene" as used herein may refer to a DNA sequence with functional significance. It can be a native nucleic acid sequence, or a recombinant nucleic acid sequence derived from natural source or synthetic construct. The term "gene" may also be used to refer to, for example
and without limitation, a cDNA and/or an mRNA encoded by or derived from, directly or indirectly, genomic DNA sequence.
According to one aspect of the present disclosure, a composition for prohibiting cross-species infection of malaria caused by P. vivax in a subject is provided. Preferably, the composition is configured to inhibit, arrest, prohibit and/or cease development of merozoites and/or hypnozoites potentially carried by a human subject and/or a transmitting agent such as female mosquitoes of the genus Anopheles. The disclosed vaccine composition generally comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 and/or SEQ ID No. 2, the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
Referring to sequence listing of SEQ ID No. 1, the inventors of the present disclosure employ a polynucleotide sequence of Plasmodium vivax ookinete surface protein antigen, namely Pvs25, protein as the template or base template for preparing the polynucleotide sequence SEQ ID No. 1 in the form of mRNA. With the utilization of mRNA instead of conventional proteinbased vaccine, the disclosed composition is relatively safer to use considering that mRNA is a non-infectious and non-integrating platform without exposing the subject to the risk of infection or insertional mutagenesis. Moreover, mRNA is the minimal genetic vector; therefore, anti-vector immunity can be avoided, and mRNA vaccines can be administered repeatedly. Additionally, the mRNA platform of the present disclosure is degradable by normal cellular processes, and it in vivo half-life can be regulated through various modifications introduced and delivery methods used to further reduce the likelihood of the occurrence of any undesired outcome. It is important to note that the polynucleotide used in the present disclosure for preparing the vaccine composition can be of natural, preferably further processed by one or more purification steps, or synthetic origin. The Pvs25 sequence used can be with or without modification depending on the designated outcome to be attained as further described in the following. For instance, in some embodiments, the polynucleotide has uridine, which is found in natural or general mRNA template, substituted by any one or combination of pseudouridine and 1-methyl-pseudouridine. The employment of pseudouridine and/or 1-methyl- pseudouridine incorporated in the disclosed composition ensures higher level of protein
expression in the subject. The substitution of uridine by pseudouridine and/or 1-methyl- pseudouridine also lowers the risk of serious inflammatory responses in the subject after receiving the disclosed vaccine composition.
Pursuant to more embodiments of the disclosed composition, the polynucleotide used may comprise a I130T mutation as an additional modification made towards the polynucleotide sequence used to improve overall performance of the disclosed composition. The sequence of the mentioned polynucleotide having the intended mutation is disclosed in SEQ ID No. 2 of Fig 11. Preferably, the included mutation enhances the transmission blocking activity towards other variants of Plasmodium parasites, particularly a variant of Plasmodium vivax carrying the aforesaid mutation. Further, the sequences of the peptides correspondingly encoded by the SEQ ID No. 1 and SEQ ID No. 2 are shown in SEQ ID No. 3 and SEQ ID No. 4.
For some embodiments, the polynucleotide further comprises a region encoding for at least one signaling peptide, which is incorporated upstream of the sequence of SEQ ID No. 1 or SEQ ID No. 2 in an expression construct bearing the polynucleotide sequence of SEQ ID No. 1 or SEQ ID No. 2. The incorporation of the signaling peptide may facilitate better recognition of the expressed peptide by the host or subject receiving the vaccine in some embodiments. According to several preferred embodiments, the signaling peptide derived from either one of signaling peptide of MHC class II. Some examples of the signaling peptide used are detailed in SEQ ID No. 5 or SEQ ID No. 6. Particularly, in some embodiments, each of the plurality of polynucleotides further comprises SEQ ID No. 5 and/or SEQ ID No. 6 located upstream or downstream of SEQ ID No. 1 or SEQ ID No. 2.
As described in the setting forth, a liquid phase composed of LNPs are used to form a protective layer to encapsulate, enclose, or wrap the plurality. By mixing the LNP along with the polynucleotides under a regulated and controlled condition, the LNPs self-assemble into 80- 100 nm particles. The protective layer established though the LNPs creates a neutral surface in relation to the cellular environment allowing the encapsulated polynucleotides to shunt extensive binding to serum proteins prior to expression. More specifically, the mRNA-loaded LNPs are taken up via endocytosis then go through the endosomal pathway, get disrupted by endosomal acidification, and a fraction of the RNA escapes from the endosomes to enter the cytosol, where protein production from mRNA occurs [35-37]. In some embodiments, the lipid
nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. More preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5-15: 30-50: 1-5.
For several embodiments, the adjuvant can be any one or combination of an aqueous solution, a saline solution, Ringer’s solution, isotonic sodium chloride, synthetic monoglycerides, synthetic diglycerides, polyethylene glycols, glycerin, propylene glycol, antibacterial agents, antioxidants, chelating agents, buffering agents, tonicity modifying agents, and cryoprotectants. Preferably, the antibacterial agents can be benzyl alcohol, methyl paraben, and the like. Preferably, the antioxidants can be ascorbic acid, sodium bisulfite, and the like. Preferably, the buffering agents can be acetates, citrates or phosphates, and the like. Preferably, the tonicity modifying agents can be sodium chloride, dextrose, and the like. Preferably, the cryoprotectants can be sucrose, trehalose, and the like. Also, the chelating agents such as ethylenediaminetetraacetic acid can be used as well.
Another aspect of the present disclosure relates to a method of inducing an immune response in a subject reactive against infection of malaria caused by P. vivax. The immune response induced shall result in production of an antibody reactive against Pvs25 antigen which in turn arrests or inhibits development of P. vivax, particularly merozoites and/or hypnozoites, in the subject. It is crucial to note that the antibodies produced can be transferred to malaria transmitting agents, such as female mosquitoes of the genus Anopheles, happened to feed on the blood of the subject carrying the antibodies. These antibodies are configured to attain the same outcome in the mosquitoes to arrest or inhibit development of the P. vivax merozoites and/or hypnozoites. Thus, the mosquitoes bearing these antibodies shall fail to effectuate transmission of malaria subsequently shrinking the pool of malaria infectious agents available. Through the inhibition mechanism, the disclosed method is able to reduce or prevent the crossspecies transmission of malaria particularly caused by P. vivax. The disclosed method preferably comprises the steps of administrating a vaccine composition to the subject by way of intramuscular route or subcutaneous route. More specifically, the vaccine composition used in the disclosed method comprises a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1, the polynucleotides being configured to express in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the
pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
Corresponding to the vaccine composition mentioned above, the polynucleotides of the vaccine composition used in the disclosed method has uridine substituted by any one or combination of pseudouridine and 1-methyl-pseudouridine to improve expression of the antigenic protein in the subject.
Likewise, the lipid nanoparticles found in the vaccine composition used for the disclosed method comprises cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG. More preferably, the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5-15: 30-50: 1-5.
The following example is intended to further illustrate the disclosure, without any intent for the disclosure to be limited to the specific embodiments described therein.
Example 1
Western blotting was performed with Pvs25-specific antibodies confirming the expression of Pvs25 from all mRNA-LNPs in human monocyte-derived dendritic cells (mo-Dcs). The results are present in Fig. 1. All eight constructs were designed based on the sequence of the Pvs25 gene from the reference P. vivax strain Sal I. Four constructs express Pvs25 without the C- terminal GPI anchor. Two of them (Pvs25A and Pvs25A I130T) have the wild type signal peptide sequence; one of which (Pvs25A I130T) contains the I130T substitution predominant in the Asian P. vivax isolates. The other two (Pvs25A IL-2 SP and Pvs25A HLA-DR SP) have exogenous signal peptide sequences (human IL-2 and HLA-DR) which are known to enhance protein expression. The other four constructs encode Pvs25 with the C terminal GPI anchor. Pvs25F encodes the full-length sequence of the Pvs25 gene from Sal I. The remaining three constructs (Pvs25F I130T, Pvs25F IL-2 SP, and Pvs25F HLA-DR SP) contain the full-length sequence of Pvs25 with a) the I130T mutation, b) the IL-2 signal peptide, or c) the HLA-DR signal peptide. Cells transfected with polycytidine (PolyC) RNA-LNP was used as the negative control. Recombinant protein (Pvs25A) produced by the wheat germ cell free system was used as the positive control.
Example 2
Experiments was performed to assess immunity response after administrating the various constructs prepared in example 1 to mice. The mice were immunized using the various Pvs25 mRNA-LNPs constructs prepared. Particularly, BALB/C mice were immunized 3 pg, 10 pg or 30 pg of Pvs25 mRNA-LNPs via intramuscular injection in a 4week interval prime: boost regimen (N = 6 per group). In the control group, mice received 30 pg of PolyC RNA-LNP (N=6). The results are present in Fig. 2.
Further, pooled plasma from the immunized BALB/c mice, after the final immunization with 30 pg of Pvs25F mRNA-LNP (Pvs25F) or Poly C mRNA-LNP (PolyC), was used to assess reactivity towards the native antigen on the surface of P. vivax ookinetes by immunofluorescence assay (IFA). The results are shown in Fig. 3.
Example 3
The direct membrane feeding assay (DMFA) was performed to evaluate transmission reducing activity (TRA) of immunized mouse antisera at different dilutions. Lab-reared uninfected mosquitoes were fed on P. vivax malaria patient blood (n = 5) suspended with immune (from mice that received 30 pg of each of the eight Pvs25 mRNA-LNPs) or non-immune (from mice received 30 pg of poly C RNA-LNP sera, then the mosquitoes were dissected 7 days post feeding and oocysts were counted under the light microscope to derive the results thereof as shown in Fig. 4.
Example 4
The present disclosure compared the immunogenicity and kinetics of the immune response of four different immunization regimens: a) Pvs25F mRNA-LNP homologous prime-boost vaccination b) Pvs25 protein (with ISA-51 adjuvant) homologous prime-boost vaccination, c) Pvs25F mRNA-LNP/Pvs25 protein heterologous vaccination and d) Pvs25 protein/Pvs25F mRNA-LNP heterologous vaccination. Mice were randomly assigned into 8 groups (n=17 mice per group). In 4 vaccination groups, mice were vaccinated with homologous and heterologous prime-boost regimens (10 pg mRNA or 10 pg recombinant protein vaccine, 4 weeks between the prime and boost). In the other 4 groups, 10 pg Poly(C) RNA-LNP or Montanide ISA-51 VG was administered as negative controls for each vaccination group.
Sera from the control groups had no detectable Pvs25-specific antibody response by ELISA (Fig. 5). The mRNA/mRNA homologous prime-boost vaccination generated the strongest Pvs25-specific antibody response with GMT of -140,000. The protein/protein homologous vaccination had GMT of -40,000. The protein/mRNA vaccination had GMT of -80,000 and the mRNA/protein vaccination yielded GMT of -46,000.
The present disclosure further characterized the quality of Pvs25-specific antibodies. IgGl and IgG2a subclasses expressed as the IgG2a/IgGl ratio and antigen- antibody avidity index. Here, pooled serum was used to determine the IgG2a/IgGl ratio and the avidity index (Fig. 6a and 6b). The IgG subclass patterns of all vaccination regimens were similar with the IgG2a/IgGl ratio of -0.75. The avidity indices were also similar across the groups.
Example 5
Experiment was performed to assess the production of IFN-y and IL-2 by CD4+ and CD8+ T cells. Particularly, splenocytes of the immunized mice were obtained at Month 1 after the immunization then subjecting the splenocytes to Pvs25 peptide stimulation to analyze the IFN- y and IL-2 produced by CD4+ and CD8+ T cells with controls (n = 2) selected from the 4 controls predetermined. The results were summarized respectively in Fig. 7a and 7b.
More specifically, mouse splenocytes from the homologous and heterologous prime -boost experiment were evaluated for cellular immune responses. The mRNA/mRNA homologous prime-boost vaccination induced the most robust Pvs25-specific CD4+ and CD8+ T cell responses as measured by IFN-y and IL-2 production, while the protein/protein vaccination barely induced T cells. The results were summarized respectively in Fig. 7a and 7b. Likewise, the mRNA/mRNA homologous vaccination elicited the strongest memory B cell response whereas this was almost absent in the protein/protein homologous vaccination. The protein/mRNA heterologous vaccination gave positive but intermediate cellular responses while the mRNA/protein vaccination elicited very low cellular response similar to the protein/protein vaccination. The result is presented in Fig 7.
Example 6
The present disclosure also used ELISA to determine Pvs25 antibodies response in the mice immunized by the four different constructs prepared over a period of 7 months with
measurements being conducted at each of the month. Nine mice per group were maintained for 7 months after the booster dose at Month 0. The results are summarized in Fig. 8. Particularly, Pvs25 antibody levels were followed monthly for 7 months post boost vaccination to assess the durability of antibody responses (Fig. 8). The antibody levels peaked at 1 month post boost in all vaccination groups and declined over the subsequent months. In agreement with the previous results, GMTs were highest in the mRNA/mRNA group and the lowest in the protein/protein group. By the 7th month, the antibody level had a GMT of 36,000 for mRNA/mRNA, 2,300 for protein/protein, 25,000 for protein/mRNA, and 18,000 for mRNA/protein.
Further, investigation was done towards transmission reducing activity of the immunized mice. Particularly, pooled sera obtained from the mice at the 1st month and 7th month were subjected to direct membrane feeding assay (DMFA) against P. mvu-infcctcd blood from four different patients. Transmission reducing activity (TRA) was determined at 1:2, 1:10, and 1:50 serum dilutions with the obtained results shown in Fig. 9. Particularly, after the booster immunization of the homologous and heterologous prime-boost experiment, the Pvs25 antibody levels in mice (8-9 per group) were followed monthly for 7 months to assess the durability of the antibody response by ELISA. The result is presented in Fig 9. A similar pattern was observed when the TRA was followed over time; the TRA was the highest one month after the boost (Fig. 9).
At this time, all vaccination regimens rendered the full efficacy (100% TRA) at serum dilution 1:2. However, for the same 1:2 dilution the TRA of protein/protein vaccination declined to 63% whereas the other three vaccination strategies retained high efficacy >99% at 7 months post boost. Among the 4 vaccination regimens, the mRNA/mRNA group exhibited the most durable functional response, as the TRA remained over 80% even at 1:50 dilution in this group. When the TRA data across all immunization regimens are pooled, there is a clear dose response relationship between TRA and total IgG as expected (Fig. 10). The half-maximal inhibition concentration (IC50) of Pvs25 total IgG was 781 (CI95: 564-1003) reciprocal titer unit.
It is to be understood that the present disclosure may be embodied in other specific forms and is not limited to the sole embodiment described above. However, modification and equivalents of the disclosed concepts such as those which readily occur to one skilled in the art are intended to be included within the scope of the claims which are appended thereto.
References
1. WHO . W orld malaria report 2019. 2019.
2. Naing C, Whittaker MA, Nyunt Wai V, Mak JW. Is Plasmodium vivax malaria a severe malaria?: a systematic review and meta- analysis. PLoS Negl Trop Dis. 2014;8(8):e3071.
3. Anstey NM, Douglas NM, Poespoprodjo JR, Price RN. Plasmodium vivax: clinical spectrum, risk factors and pathogenesis. Adv Parasitol. 2012;80:151-201.
4. Puji BS Asih DSaJKB . Challenges in the Control and Elimination of Plasmodium vivax Malaria. In: Dev SMaV, editor. Towards Malaria Elimination- A Leap Forward: IntechOpen; 2018.
5. Roth A, Maher SP, Conway AJ, Ubalee R, Chaumeau V, Andolina C, et al. A comprehensive model for assessment of liver stage therapies targeting Plasmodium vivax and Plasmodium falciparum. Nat Commun. 2018;9(1): 1837.
6. Bennett JW, Yadava A, Tosh D, Sattabongkot J, Komisar J, Ware LA, et al. Phase l/2a Trial of Plasmodium vivax Malaria Vaccine Candidate VMP001/AS01B in Malaria-Naive Adults: Safety, Immunogenicity, and Efficacy. PLoS Negl Trop Dis. 2016;10(2):e0004423.
7. Watson J, Taylor WRJ, Bancone G, Chu CS, Jittamala P, White NJ. Implications of current therapeutic restrictions for primaquine and tafenoquine in the radical cure of vivax malaria. PLoS Negl Trop Dis. 2018;12(4):e0006440.
8. Roth A, Maher SP, Conway AJ, Ubalee R, Chaumeau V, Andolina C, et al. A comprehensive model for assessment of liver stage therapies targeting Plasmodium vivax and Plasmodium falciparum. Nat Commun. 2018;9( 1): 1837.
9 Arakawa, T. et al. Tricomponent complex loaded with a mosquito-stage antigen of the malaria parasite induces potent transmission-blocking immunity. Clin Vaccine Immunol 21, 561-569, doi:10.1128/CVI.00053-14 (2014).
10 Blagborough, A. M. et al. Transmission blocking potency and immunogenicity of a plant-produced Pvs25-based subunit vaccine against Plasmodium vivax. Vaccine 34, 3252- 3259, doi:10.1016/j.vaccine.2016.05.007 (2016).
11 Mizutani, M. et al. Baculovirus-vectored multistage Plasmodium vivax vaccine induces both protective and transmission-blocking immunities against transgenic rodent malaria parasites. Infect Immun 82, 4348-4357, doi:10.1128/IAI.02040-14 (2014).
12 Hisaeda, H. et al. Antibodies to malaria vaccine candidates Pvs25 and Pvs28 completely block the ability of Plasmodium vivax to infect mosquitoes. Infect Immun 68, 6618- 6623 (2000).
13 Sattabongkot, J. et al. Blocking of transmission to mosquitoes by antibody to Plasmodium vivax malaria vaccine candidates Pvs25 and Pvs28 despite antigenic polymorphism in field isolates. Am J Trop Med Hyg 69, 536-541 (2003)
14 Markin, E. M. et al. Phase 1 vaccine trial of Pvs25H: a transmission blocking vaccine for Plasmodium vivax malaria. Vaccine. 2005 May 2; 23(24): 3131-3138. doi: 10.1016/j.vaccine.2004.12.019
15 Wu, Y. et al. “Phase 1 trial of malaria transmission blocking vaccine candidates Pfs25 and Pvs25 formulated with montanide ISA 51” PloS one vol. 3,7 e2636. 9 Jul. 2008, doi: 10.1371/journal.pone.0002636
16. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261-79.
17. Chaudhary N, Weissman D, Whitehead KA. mRNA vaccines for infectious diseases: principles, delivery and clinical translation [published online ahead of print, 2021 Aug 25] [published correction appears in Nat Rev Drug Discov. 2021 Sep 21;:]. Nat Rev Drug Discov. 2021;l-22.
18. Pardi N, Hogan MJ, Naradikian MS, Parkhouse K, Cain DW, Jones L, et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med. 2018;215(6): 1571-88.
19. Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature. 2017;543(7644):248-51.
20. Pardi N, Parkhouse K, Kirkpatrick E, McMahon M, Zost SJ, Mui BL, et al. Nucleoside- modified mRNA immunization elicits influenza virus hemagglutinin stalk- specific antibodies. Nat Commun. 2018;9(l):3361.
21. Crotty S. T Follicular Helper Cell Biology: A Decade of Discovery and Diseases. Immunity. 2019;50(5):l 132-48
22. Mallory KL, Taylor JA, Zou X, et al. Messenger RNA expressing PfCSP induces functional, protective immune responses against malaria in mice. NPJ Vaccines. 2021;6(l):84.
Claims
1. A vaccine composition for prohibiting cross-species infection of malaria caused by Plasmodium vivax in a subject comprising: a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 or SEQ ID No. 2, the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
2. The vaccine composition of claim 1, wherein the polynucleotides have uridine substituted by any one or combination of pseudouridine and 1-methyl-pseudouridine.
3. The vaccine composition of claim 1, wherein the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG.
4. The vaccine composition of claim 3, wherein the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5- 15: 30-50: 1-5.
5. The vaccine composition of claim 1, wherein the sequence of SEQ ID No. 1 and/or SEQ ID No. 2 is derived from strain Sal I of P. vivax.
6. The vaccine composition of claim 1, wherein each of the plurality of polynucleotides further comprises SEQ ID No. 5 and/or SEQ ID No. 6 located upstream or downstream of SEQ ID No. 1 or SEQ ID No. 2.
7. The vaccine composition of claim 1, wherein the pharmaceutically acceptable adjuvant is any one or combination of an aqueous solution, a saline solution, Ringer’s solution, isotonic sodium chloride, synthetic monoglycerides, synthetic diglycerides, polyethylene glycols, glycerin, propylene glycol, antibacterial agents, antioxidants, chelating agents, buffering agents, tonicity modifying agents, and cryoprotectants.
8. A method of inducing an immune response in a subject reactive against infection of malaria caused by P. vivax comprising: administrating a vaccine composition to the subject by way of intramuscular route or subcutaneous route, the vaccine composition comprising a plurality of polynucleotides each comprising a sequence as setting forth in SEQ ID No. 1 or SEQ ID No. 2, the polynucleotides being expressed in a body of the subject for inducing an immune response reactive against the infection of malaria thereof; a liquid phase of lipid nanoparticles configured to form a protective layer encapsulating the pluralities of polynucleotides within the protective layer; and a pharmaceutically acceptable adjuvant.
9. The method of claim 8, wherein the polynucleotides have uridine substituted by any one or combination of pseudouridine and 1-methyl-pseudouridine.
10. The method of claim 8, wherein the lipid nanoparticles comprise cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG.
11. The method of claim 8, wherein the wherein the cationic lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG are in a molar ratio of 40-60: 5- 15: 30-50: 1-5.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/TH2023/000006 WO2024220043A1 (en) | 2023-04-20 | 2023-04-20 | A transmission-blocking composition against plasmodium vivax |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/TH2023/000006 WO2024220043A1 (en) | 2023-04-20 | 2023-04-20 | A transmission-blocking composition against plasmodium vivax |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024220043A1 true WO2024220043A1 (en) | 2024-10-24 |
Family
ID=93152862
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/TH2023/000006 WO2024220043A1 (en) | 2023-04-20 | 2023-04-20 | A transmission-blocking composition against plasmodium vivax |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024220043A1 (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2001055181A2 (en) * | 2000-01-31 | 2001-08-02 | The Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services, Centers For Disease Control And Prevention | Recombinant multivalent malarial vaccines against plasmodium vivax |
US20220040281A1 (en) * | 2018-12-21 | 2022-02-10 | Curevac Ag | Rna for malaria vaccines |
US20220257746A1 (en) * | 2015-10-22 | 2022-08-18 | Modernatx, Inc. | Tropical disease vaccines |
-
2023
- 2023-04-20 WO PCT/TH2023/000006 patent/WO2024220043A1/en unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2001055181A2 (en) * | 2000-01-31 | 2001-08-02 | The Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services, Centers For Disease Control And Prevention | Recombinant multivalent malarial vaccines against plasmodium vivax |
US20220257746A1 (en) * | 2015-10-22 | 2022-08-18 | Modernatx, Inc. | Tropical disease vaccines |
US20220040281A1 (en) * | 2018-12-21 | 2022-02-10 | Curevac Ag | Rna for malaria vaccines |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Cockburn et al. | Malaria prevention: from immunological concepts to effective vaccines and protective antibodies | |
Hoffman et al. | The march toward malaria vaccines | |
Epstein et al. | Safety, tolerability, and lack of antibody responses after administration of a Pf CSP DNA malaria vaccine via needle or needle-free jet injection, and comparison of intramuscular and combination intramuscular/intradermal routes | |
CN113215178B (en) | mRNA vaccine for 2019-nCoV coronavirus, preparation method and application thereof | |
Robinson et al. | DNA vaccines for viral infections: basic studies and applications | |
Wang et al. | Boosting of DNA vaccine-elicited gamma interferon responses in humans by exposure to malaria parasites | |
TWI620574B (en) | Foot-and-mouth disease synthetic peptide emergency vaccine | |
US10195260B2 (en) | Antigen for use in malaria | |
Coban et al. | Induction of Plasmodium falciparum transmission-blocking antibodies in nonhuman primates by a combination of DNA and protein immunizations | |
US11857611B2 (en) | Compositions and methods for generating an immune response to treat or prevent malaria | |
Bahloul et al. | Field trials of a very potent rabies DNA vaccine which induced long lasting virus neutralizing antibodies and protection in dogs in experimental conditions | |
US12233117B2 (en) | Immunogenic compositions, antigen screening methods, and methods of generating immune responses | |
US20250230197A1 (en) | Malaria vaccine | |
US20250186576A1 (en) | Sars-cov-2 subunit vaccine | |
US9913896B2 (en) | Attenuated parvovirus vaccine for muscovy duck parvovirus and goose parvovirus (derzsy's disease) | |
WO2024220043A1 (en) | A transmission-blocking composition against plasmodium vivax | |
US11969465B2 (en) | Toxoplasma gondii vaccines and their use | |
Sedegah et al. | Vaxfectin™ enhances immunogenicity and protective efficacy of P. yoelii circumsporozoite DNA vaccines | |
Kumar | A vaccine to prevent transmission of human malaria: a long way to travel on a dusty and often bumpy road | |
US20240299528A1 (en) | A dna plasmid sars-corona virus-2/covid-19 vaccine | |
WO2024226031A1 (en) | Compositions, devices, systems and methods relating to vaccination and sterile protection against malaria | |
Abaza | Spotlights on new publications | |
Tsoumani et al. | Malaria Vaccines: From the Past towards the mRNA Vaccine Era. Vaccines 2023, 11, 1452 | |
Mosqueda et al. | Molecular Mechanisms of Babesia Invasion: Potential Targets for Vaccine Development | |
Misra et al. | Vaccine Design, Nanoparticle Vaccines and Biomaterial Applications |
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: 23934251 Country of ref document: EP Kind code of ref document: A1 |