WO2018160690A1 - Rna pharmaceutical formulations for prophylactic and therapeutic treatment of zika virus infection - Google Patents
Rna pharmaceutical formulations for prophylactic and therapeutic treatment of zika virus infection Download PDFInfo
- Publication number
- WO2018160690A1 WO2018160690A1 PCT/US2018/020220 US2018020220W WO2018160690A1 WO 2018160690 A1 WO2018160690 A1 WO 2018160690A1 US 2018020220 W US2018020220 W US 2018020220W WO 2018160690 A1 WO2018160690 A1 WO 2018160690A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- composition
- sirna
- mammal
- zikv
- sirna molecule
- Prior art date
Links
- 208000020329 Zika virus infectious disease Diseases 0.000 title claims abstract description 35
- 208000001455 Zika Virus Infection Diseases 0.000 title claims abstract description 20
- 239000008194 pharmaceutical composition Substances 0.000 title claims abstract description 12
- 230000001225 therapeutic effect Effects 0.000 title abstract description 17
- 238000011282 treatment Methods 0.000 title abstract description 14
- 230000000069 prophylactic effect Effects 0.000 title abstract description 10
- 208000035332 Zika virus disease Diseases 0.000 title abstract description 9
- 108020004459 Small interfering RNA Proteins 0.000 claims abstract description 187
- 108020004999 messenger RNA Proteins 0.000 claims abstract description 50
- 238000000034 method Methods 0.000 claims abstract description 39
- 239000000203 mixture Substances 0.000 claims description 81
- 241000907316 Zika virus Species 0.000 claims description 78
- 239000002502 liposome Substances 0.000 claims description 50
- 108090000623 proteins and genes Proteins 0.000 claims description 50
- 239000002105 nanoparticle Substances 0.000 claims description 47
- 241000124008 Mammalia Species 0.000 claims description 42
- 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 32
- 241000282414 Homo sapiens Species 0.000 claims description 25
- 102000004169 proteins and genes Human genes 0.000 claims description 18
- 235000012000 cholesterol Nutrition 0.000 claims description 16
- 239000004472 Lysine Substances 0.000 claims description 14
- 229920000642 polymer Polymers 0.000 claims description 14
- 229920001577 copolymer Polymers 0.000 claims description 13
- 108091034117 Oligonucleotide Proteins 0.000 claims description 12
- 239000003937 drug carrier Substances 0.000 claims description 12
- 208000015181 infectious disease Diseases 0.000 claims description 11
- 241000282693 Cercopithecidae Species 0.000 claims description 9
- 241000283984 Rodentia Species 0.000 claims description 9
- 230000009385 viral infection Effects 0.000 claims description 9
- 108020005345 3' Untranslated Regions Proteins 0.000 claims description 8
- 101800001019 Non-structural protein 4B Proteins 0.000 claims description 8
- 101710144121 Non-structural protein 5 Proteins 0.000 claims description 8
- 208000036142 Viral infection Diseases 0.000 claims description 8
- 241000700199 Cavia porcellus Species 0.000 claims description 7
- 101710091045 Envelope protein Proteins 0.000 claims description 7
- 241000282341 Mustela putorius furo Species 0.000 claims description 7
- 101710188315 Protein X Proteins 0.000 claims description 7
- 101710158312 DNA-binding protein HU-beta Proteins 0.000 claims description 5
- 101710128560 Initiator protein NS1 Proteins 0.000 claims description 5
- 101710144127 Non-structural protein 1 Proteins 0.000 claims description 5
- 101710144111 Non-structural protein 3 Proteins 0.000 claims description 5
- 238000002347 injection Methods 0.000 claims description 5
- 239000007924 injection Substances 0.000 claims description 5
- 238000001990 intravenous administration Methods 0.000 claims description 5
- 238000007920 subcutaneous administration Methods 0.000 claims description 5
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 claims description 4
- 108091036066 Three prime untranslated region Proteins 0.000 claims description 4
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 claims description 4
- 108091023045 Untranslated Region Proteins 0.000 claims description 3
- 101150033828 NS1 gene Proteins 0.000 claims description 2
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 claims description 2
- 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 claims 6
- 101710174009 Suppressor of RNA silencing p3 Proteins 0.000 claims 2
- 102100021696 Syncytin-1 Human genes 0.000 claims 2
- 125000003275 alpha amino acid group Chemical group 0.000 claims 2
- 102100032091 ALK and LTK ligand 2 Human genes 0.000 claims 1
- 101000776351 Homo sapiens ALK and LTK ligand 2 Proteins 0.000 claims 1
- 230000002265 prevention Effects 0.000 abstract description 4
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 48
- 238000012384 transportation and delivery Methods 0.000 description 36
- 210000004027 cell Anatomy 0.000 description 31
- 241000699670 Mus sp. Species 0.000 description 23
- 241000700605 Viruses Species 0.000 description 19
- 230000014509 gene expression Effects 0.000 description 16
- 230000003612 virological effect Effects 0.000 description 15
- 239000002245 particle Substances 0.000 description 14
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 13
- 108700021021 mRNA Vaccine Proteins 0.000 description 13
- 229940126582 mRNA vaccine Drugs 0.000 description 13
- 241000725619 Dengue virus Species 0.000 description 12
- -1 NS2B Proteins 0.000 description 12
- 238000009472 formulation Methods 0.000 description 12
- 230000008685 targeting Effects 0.000 description 12
- 238000003556 assay Methods 0.000 description 11
- 210000001519 tissue Anatomy 0.000 description 11
- 101710204837 Envelope small membrane protein Proteins 0.000 description 10
- 101710145006 Lysis protein Proteins 0.000 description 10
- 230000030279 gene silencing Effects 0.000 description 10
- 239000000243 solution Substances 0.000 description 10
- PFNFFQXMRSDOHW-UHFFFAOYSA-N spermine Chemical compound NCCCNCCCCNCCCN PFNFFQXMRSDOHW-UHFFFAOYSA-N 0.000 description 10
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 9
- 230000009368 gene silencing by RNA Effects 0.000 description 9
- 241000699666 Mus <mouse, genus> Species 0.000 description 8
- 125000002091 cationic group Chemical group 0.000 description 8
- 230000003472 neutralizing effect Effects 0.000 description 8
- 230000000694 effects Effects 0.000 description 7
- 230000028993 immune response Effects 0.000 description 7
- 238000001727 in vivo Methods 0.000 description 7
- 206010022000 influenza Diseases 0.000 description 7
- 150000002632 lipids Chemical class 0.000 description 7
- 230000036515 potency Effects 0.000 description 7
- 238000012360 testing method Methods 0.000 description 7
- 238000001890 transfection Methods 0.000 description 7
- 229960005486 vaccine Drugs 0.000 description 7
- 150000001413 amino acids Chemical group 0.000 description 6
- 238000010171 animal model Methods 0.000 description 6
- 108090000765 processed proteins & peptides Proteins 0.000 description 6
- 239000000047 product Substances 0.000 description 6
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 5
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 5
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 238000013459 approach Methods 0.000 description 5
- 230000027455 binding Effects 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 210000000805 cytoplasm Anatomy 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 201000010099 disease Diseases 0.000 description 5
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 5
- 210000004072 lung Anatomy 0.000 description 5
- 208000004141 microcephaly Diseases 0.000 description 5
- 239000002773 nucleotide Substances 0.000 description 5
- 125000003729 nucleotide group Chemical group 0.000 description 5
- 230000008569 process Effects 0.000 description 5
- 229940063675 spermine Drugs 0.000 description 5
- 230000014616 translation Effects 0.000 description 5
- 210000003501 vero cell Anatomy 0.000 description 5
- 238000002965 ELISA Methods 0.000 description 4
- 241000710831 Flavivirus Species 0.000 description 4
- 101710172711 Structural protein Proteins 0.000 description 4
- 230000000840 anti-viral effect Effects 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- 230000005540 biological transmission Effects 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 239000003814 drug Substances 0.000 description 4
- 230000004927 fusion Effects 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 238000007912 intraperitoneal administration Methods 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 229920001606 poly(lactic acid-co-glycolic acid) Polymers 0.000 description 4
- 102000004196 processed proteins & peptides Human genes 0.000 description 4
- 230000001681 protective effect Effects 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 230000010076 replication Effects 0.000 description 4
- 238000013519 translation Methods 0.000 description 4
- 230000029812 viral genome replication Effects 0.000 description 4
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- 241000710781 Flaviviridae Species 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- 108700005077 Viral Genes Proteins 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000013461 design Methods 0.000 description 3
- 238000011156 evaluation Methods 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 238000012226 gene silencing method Methods 0.000 description 3
- 229920001477 hydrophilic polymer Polymers 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000010172 mouse model Methods 0.000 description 3
- 210000003097 mucus Anatomy 0.000 description 3
- PGZUMBJQJWIWGJ-ONAKXNSWSA-N oseltamivir phosphate Chemical compound OP(O)(O)=O.CCOC(=O)C1=C[C@@H](OC(CC)CC)[C@H](NC(C)=O)[C@@H](N)C1 PGZUMBJQJWIWGJ-ONAKXNSWSA-N 0.000 description 3
- 239000013612 plasmid Substances 0.000 description 3
- 229920001184 polypeptide Polymers 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 239000013589 supplement Substances 0.000 description 3
- 229940061367 tamiflu Drugs 0.000 description 3
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 2
- LDGWQMRUWMSZIU-LQDDAWAPSA-M 2,3-bis[(z)-octadec-9-enoxy]propyl-trimethylazanium;chloride Chemical compound [Cl-].CCCCCCCC\C=C/CCCCCCCCOCC(C[N+](C)(C)C)OCCCCCCCC\C=C/CCCCCCCC LDGWQMRUWMSZIU-LQDDAWAPSA-M 0.000 description 2
- KSXTUUUQYQYKCR-LQDDAWAPSA-M 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium;chloride Chemical compound [Cl-].CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KSXTUUUQYQYKCR-LQDDAWAPSA-M 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 108020004635 Complementary DNA Proteins 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- 208000001490 Dengue Diseases 0.000 description 2
- 206010012310 Dengue fever Diseases 0.000 description 2
- 108700039887 Essential Genes Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 102000016844 Immunoglobulin-like domains Human genes 0.000 description 2
- 108050006430 Immunoglobulin-like domains Proteins 0.000 description 2
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 101800001030 Non-structural protein 2A Proteins 0.000 description 2
- 101800001020 Non-structural protein 4A Proteins 0.000 description 2
- 101150038760 Ns3 gene Proteins 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 102100040283 Peptidyl-prolyl cis-trans isomerase B Human genes 0.000 description 2
- 229930182556 Polyacetal Natural products 0.000 description 2
- 229920000954 Polyglycolide Polymers 0.000 description 2
- 238000011529 RT qPCR Methods 0.000 description 2
- ATHGHQPFGPMSJY-UHFFFAOYSA-N Spermidine Natural products NCCCCNCCCN ATHGHQPFGPMSJY-UHFFFAOYSA-N 0.000 description 2
- 108010067390 Viral Proteins Proteins 0.000 description 2
- 108020000999 Viral RNA Proteins 0.000 description 2
- 206010052428 Wound Diseases 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 230000004888 barrier function Effects 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 210000004556 brain Anatomy 0.000 description 2
- 210000000234 capsid Anatomy 0.000 description 2
- 229920006317 cationic polymer Polymers 0.000 description 2
- 238000012512 characterization method Methods 0.000 description 2
- 108010048032 cyclophilin B Proteins 0.000 description 2
- 229940124447 delivery agent Drugs 0.000 description 2
- 208000025729 dengue disease Diseases 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 238000006471 dimerization reaction Methods 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 241001493065 dsRNA viruses Species 0.000 description 2
- 238000002296 dynamic light scattering Methods 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 229920000578 graft copolymer Polymers 0.000 description 2
- 210000002216 heart Anatomy 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 230000034217 membrane fusion Effects 0.000 description 2
- 239000000693 micelle Substances 0.000 description 2
- 238000002156 mixing Methods 0.000 description 2
- 108010044156 peptidyl-prolyl cis-trans isomerase b Proteins 0.000 description 2
- 238000013081 phylogenetic analysis Methods 0.000 description 2
- 229920000962 poly(amidoamine) Polymers 0.000 description 2
- 229920000747 poly(lactic acid) Polymers 0.000 description 2
- 229920006324 polyoxymethylene Polymers 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000000241 respiratory effect Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 238000012883 sequential measurement Methods 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 229940063673 spermidine Drugs 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 230000002195 synergetic effect Effects 0.000 description 2
- 210000001550 testis Anatomy 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 210000001215 vagina Anatomy 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- 206010000234 Abortion spontaneous Diseases 0.000 description 1
- 241000256111 Aedes <genus> Species 0.000 description 1
- 208000006820 Arthralgia Diseases 0.000 description 1
- 241001502567 Chikungunya virus Species 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 208000032170 Congenital Abnormalities Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 206010010741 Conjunctivitis Diseases 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 238000001712 DNA sequencing Methods 0.000 description 1
- 241000710829 Dengue virus group Species 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 108090000331 Firefly luciferases Proteins 0.000 description 1
- 208000007212 Foot-and-Mouth Disease Diseases 0.000 description 1
- 241000710198 Foot-and-mouth disease virus Species 0.000 description 1
- 206010017533 Fungal infection Diseases 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
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 102100022662 Guanylyl cyclase C Human genes 0.000 description 1
- 101710198293 Guanylyl cyclase C Proteins 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108010076039 Polyproteins Proteins 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 108010052090 Renilla Luciferases Proteins 0.000 description 1
- 241000725643 Respiratory syncytial virus Species 0.000 description 1
- 102000004389 Ribonucleoproteins Human genes 0.000 description 1
- 108010081734 Ribonucleoproteins Proteins 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 241000315672 SARS coronavirus Species 0.000 description 1
- 108091081021 Sense strand Proteins 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- 108010022999 Serine Proteases Proteins 0.000 description 1
- 102000012479 Serine Proteases Human genes 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 206010054094 Tumour necrosis Diseases 0.000 description 1
- 206010058874 Viraemia Diseases 0.000 description 1
- 108091005605 Vitamin K-dependent proteins Proteins 0.000 description 1
- 241000710886 West Nile virus Species 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- HIHOWBSBBDRPDW-PTHRTHQKSA-N [(3s,8s,9s,10r,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1h-cyclopenta[a]phenanthren-3-yl] n-[2-(dimethylamino)ethyl]carbamate Chemical compound C1C=C2C[C@@H](OC(=O)NCCN(C)C)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 HIHOWBSBBDRPDW-PTHRTHQKSA-N 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000001476 alcoholic effect Effects 0.000 description 1
- 210000002821 alveolar epithelial cell Anatomy 0.000 description 1
- 125000000129 anionic group Chemical group 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 229920000229 biodegradable polyester Polymers 0.000 description 1
- 239000004622 biodegradable polyester Substances 0.000 description 1
- 230000007698 birth defect Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000000621 bronchi Anatomy 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 230000006806 disease prevention Effects 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 230000002222 downregulating effect Effects 0.000 description 1
- 239000003596 drug target Substances 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 238000001493 electron microscopy Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 210000001163 endosome Anatomy 0.000 description 1
- 230000026502 entry into host cell Effects 0.000 description 1
- 239000002532 enzyme inhibitor Substances 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 238000007306 functionalization reaction Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 230000003054 hormonal effect Effects 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000008105 immune reaction Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 229960003971 influenza vaccine Drugs 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 238000013383 initial experiment Methods 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 210000001865 kupffer cell Anatomy 0.000 description 1
- 150000002605 large molecules Chemical class 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 239000002082 metal nanoparticle Substances 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 208000030247 mild fever Diseases 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 208000015994 miscarriage Diseases 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 239000006199 nebulizer Substances 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000000633 nuclear envelope Anatomy 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 230000009437 off-target effect Effects 0.000 description 1
- 125000001117 oleyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])/C([H])=C([H])\C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 230000007110 pathogen host interaction Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 230000000149 penetrating effect Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 230000035409 positive regulation of cell proliferation Effects 0.000 description 1
- 230000035935 pregnancy Effects 0.000 description 1
- 238000002331 protein detection Methods 0.000 description 1
- 238000000164 protein isolation Methods 0.000 description 1
- 238000000734 protein sequencing Methods 0.000 description 1
- 206010037844 rash Diseases 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000009256 replacement therapy Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000002864 sequence alignment Methods 0.000 description 1
- 230000001568 sexual effect Effects 0.000 description 1
- 230000001743 silencing effect Effects 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000012798 spherical particle Substances 0.000 description 1
- 208000000995 spontaneous abortion Diseases 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000031068 symbiosis, encompassing mutualism through parasitism Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 239000010409 thin film Substances 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 108010087967 type I signal peptidase Proteins 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 238000010200 validation analysis Methods 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 230000007444 viral RNA synthesis Effects 0.000 description 1
- 244000000009 viral human pathogen Species 0.000 description 1
- 230000024058 virion binding Effects 0.000 description 1
- 238000003260 vortexing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- 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
- A61K9/10—Dispersions; Emulsions
- A61K9/127—Synthetic bilayered vehicles, e.g. liposomes or liposomes with cholesterol as the only non-phosphatidyl surfactant
- A61K9/1271—Non-conventional liposomes, e.g. PEGylated liposomes or liposomes coated or grafted with polymers
- A61K9/1273—Polymersomes; Liposomes with polymerisable or polymerised bilayer-forming substances
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
- C12N15/1131—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against viruses
-
- 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
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
- A61K2039/541—Mucosal route
-
- 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
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering nucleic acids [NA]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/30—Special therapeutic applications
- C12N2320/31—Combination therapy
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/30—Special therapeutic applications
- C12N2320/32—Special delivery means, e.g. tissue-specific
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/24011—Flaviviridae
- C12N2770/24111—Flavivirus, e.g. yellow fever virus, dengue, JEV
- C12N2770/24134—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
-
- 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
- This invention presents novel prophylactic and therapeutic agents for prevention and treatment of Zika virus (ZIKV) infection and methods for their use.
- ZIKV Zika virus
- the Zika virus (a member of the flaviviridae family including West Nile Virus, Dengue and Chikungunya viruses) has recently garnered great interest because of a link between infection in pregnant women and miscarriage or birth defects in their infants. Transmission is via the bite of an infected Aedes mosquito, and a rapid increase in transmission observed in Brazil since late 2014 preceded an increase in the number of cases of microcephaly in infants reported to the Brazil Ministry of Health in 2015 [1, 2]. As of November 2016, a total of more than 4, 100 suspected microcephaly cases had been reported, many of which occurred in infants born to women who lived in or had visited areas where Zika virus transmission was observed [3].
- ZIKA virus infections have now been identified and validated in the mainland US in Florida and gulf coast states, and the need for suitable prophylactic or therapeutic options is significant.
- ZIKV is a single-strand positive RNA virus, belonging to family Flaviviridae; genus Flavivirus.
- the genome of ZIKV consists of about 10K nucleotides, which code at least three structural proteins (capsid (C), membrane (prM; processed to M) and envelope (E)), and seven non-structural proteins (NSl, NS2A, NS2B, NS3, NS4A, NS4B and NS5).
- C structural proteins
- prM membrane
- E envelope
- NSl seven non-structural proteins
- NS2A, NS2B, NS3, NS4A, NS4B and NS5 seven non-structural proteins
- the single ORF is flanked by 5'- and 3'- untranslated regions (UTRs), respectively ( Figure 1).
- RNA elements of the 3' UTR greatly enhance viral RNA synthesis.
- ZIKV infection initiates from envelope (E) glycoproteins on the surface of the ZIKV virion binding to a host cell receptor, such as AXL and broadly neutralizing antibodies have been demonstrated against this E protein region [5].
- E envelope
- AXL broadly neutralizing antibodies have been demonstrated against this E protein region [5].
- ZIKV enters the host cell by endocytosis, and then its genome escapes from the endosome into the cytoplasm after viral-host membrane fusion driven by the fusion loop.
- the viral genome is first translated into a large polyprotein, and then cleaved by host signal peptidases and a viral serine protease (during and after translation) to generate the viral proteins. Also in the cytoplasm, the positive strand RNA genome produces a negative strand RNA template to generate new copies of the genome.
- AXL a member of the receptor tyrosine kinase subfamily, has been identified as the major receptor for ZIKV binding.
- the study showed that blocking or silencing AXL dramatically reduced ZIKV replication in fibroblasts and alveolarepithelial cells [6].
- the function of AXL. includes transducing signals from the extracellular matrix into the cytoplasm by binding growth factors, such as vitamin K-dependent protein growth-arrest-specific gene 6 (GAS6), as well as being involved in the stimulation of cell proliferation.
- GAS6 vitamin K-dependent protein growth-arrest-specific gene 6
- Zika strains and similarities For a prophylactic or therapeutic to have viability it is important to ensure that it is able to function against a wide array (if not all) strains of a virus.
- ZIKV has circulated in both Africa and Asia since at least the 1950s. Haddow et al determined the nucleotide sequences of the open reading frames of five isolates from
- mRNA is a nontoxic molecule that allows transient protein expression of a desired protein product in nearly all transducable cell types. Compared with traditional plasmid and viral-based approaches, this approach allows design of patient personalized mRNAs that also benefit by not needing to pass through the nuclear membrane (as DNA does) and thus carries little to no risk of genomic integration. mRNA vaccines have also been demonstrated to promote immunogenicity in both very young and very old mice. If this translates to humans, then this would be beneficial since these demographics typically do not mount a robust immunological reaction to vaccines. Recently, self-amplifying mRNA vaccines have been demonstrated to be safe and efficacious against other viruses (e.g.
- mRNA vaccines may benefit in that they have been demonstrated to induce balanced, long-lived and protective immunity to influenza A virus infections in even very young and very old mice.
- Vaccines based on mRNA or RNA replicons have also been shown to be immunogenic in a variety of animal models, including nonhuman primates [10]. Target selection for mRNA vaccine against ZIKV
- the ZIKV E protein contains three distinct domains (shown in Figure 1C): a central ⁇ -barrel domain (Domain I; aa. 1-51, 132-192 and 280-295), an extended dimerization domain (Domain II; aa. 52-131 and 193-279), and an immunoglobulin-like domain (Domain III; aa. 297-406) ( Figure 1C.) [5].
- the fusion loop a hydrophobic sequence located in Domain II (aa.
- the fusion loop of Envelope protein Domain II may stimulate Antibody Dependent Enhancement (ADE) of immune response (when non-neutralizing antiviral proteins facilitate virus entry into host cells and increase infectivity in these cells resulting in higher viremia and more severe symptoms).
- ADE Antibody Dependent Enhancement
- stains of different DENV serotypes totally 4 identified serotypes
- the antibodies to the old strain interfered with the immune response to the new strain and resulted in more virus uptake [11].
- siRNA a flexible molecular platform for prophylactic and therapeutic efficacy against ZIKA virus RNA interference (RNAi) is a naturally occurring, highly specific mode of gene regulation that has broad potential applications in both research and therapeutic settings. The mechanics of RNAi are both extraordinar and highly discriminating.
- RNAi short (19-21bp) double stranded RNA sequences (referred to as short interfering RNAs, siRNAs) associate with the cytoplasmically localized RNA Interference Silencing Complex (RISC).
- RISC cytoplasmically localized RNA Interference Silencing Complex
- the resultant ribonucleoprotein complex searches the resident population of messenger RNAs (mRNAs) for complementary sequences, eventually degrading (and/or attenuating translation of) these transcripts and effectively down-regulating the expression of the targeted gene.
- mRNAs messenger RNAs
- RNAi represents a highly flexible platform by which researchers and clinicians can control diseases, including infectious diseases.
- RNA interference has previously been employed to target a range of human pathogenic viruses, including HIV, hepatitis, respiratory syncytial virus, polio virus, SARS coronavirus, Marburg, dengue, foot-and-mouth disease and Influenza.
- RNAi has the ability to (1) efficiently limit viral replication without reliance on host immune functions, and (2) target multiple genes and/or sequences simultaneously, making this an ideal therapeutic approach for viruses like Zika (where a large number of strains may need to be targeted and no other options currently exist) or against other viruses (like Influenza), which have rapidly evolving genomes.
- siRNAs By performing recursive analyses of siRNAs against 28 sequenced Zika strains (including both African and Asian lineages), we identified siRNAs that were able to cover an extensive number of strains (Table 2). Furthermore, careful selection of multiple siRNAs can be seen to provide complete coverage against all strains used in our predictions (Table 3).
- siRNA #62 and siRNA #62A can provide 100% coverage as seen by combining siRNA #62 and siRNA #62A (Table 3).
- siRNAs targeting different regions of the viral genome can produce synergistic silencing and antiviral efficacy compared with multiple siRNAs targeting the same gene. Therefore, while we can see complete coverage with certain combinations, we may see higher potency against the selected strains using a combination (such as 13 & 62) targeting distinct regions of the virus.
- a combination such as 13 & 62
- siRNA #62 and siRNA #62a can provide 100% coverage as seen by combining siRNA #62 and siRNA #62a.
- siRNAs targeting different regions of the viral genome can produce synergistic silencing and antiviral efficacy compared with multiple siRNAs targeting the same gene. Therefore, while we can see complete coverage with certain combinations, we may see higher potency against the selected strains using a combination (such as 13 & 62) targeting distinct regions of the virus.
- a combination such as 13 & 62
- ZIKV is a single-strand positive RNA virus, belonging to family Flaviviridae; genus Flavivirus.
- the genome of ZIKV consists of about 10K nucleotides, which code at least three structural proteins (capsid (C), membrane (prM; processed to M) and envelope (E)), and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5).
- C structural proteins
- prM membrane
- E envelope
- NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5 seven non-structural proteins
- the single ORF is flanked by 5'- and 3'- untranslated regions (UTRs), respectively.
- the ZIKV E protein contains three distinct domains (shown in Figure 1): a central ⁇ -barrel domain (Domain I; aa. 1-51, 132-192 and 280-295), an extended dimerization domain (Domain II; aa. 52-131 and 193-279), and an
- HKP/siRNA can self-assemble into nanoparticles (average 150nm in diameter).
- the nanoparticles can be dissolved in aqueous solution, can be lyophilized into dry powder, and can be redissovled and mixed with methylcellulose, or with RNAse free water.
- C HKP/siRNA nanoparticle delivery to mouse respiratory track: upper airway, bronchi, alveoli.
- FIG. 3 Comparison of target knockdown of lung endogenous gene among HKP, DOTAP and D5W after oral tracheal deliveries of siRNA with three different dosing regimens. HKP demonstrated the efficient siRNA-mediated knockdown of the target gene at the 20 ⁇ g dose.
- FIG. 5 The structures of Spermine-Lipid Conjugates (SLiC) species.
- the synthesis route for each of the five molecules are listed with the specific liposome chain, such as, Ri, R 2 , R 3 , R 4 and R 5 , conjugated at the location of RiH, R 2 H, R 3 H, R 4 H and R 5 H respectively.
- the structures of the five SLiC species are illustrated with a spermine head and two lipid legs.
- TM4-packaged siRNA specific to cyclophilin-B was selected for being tested in a Balb/c mouse model through a respiratory route of delivery.
- a HKP package cyclophilin-B siRNA was used as a positive control.
- Three different dosage: 25, 40 and 50 ⁇ g were tested. Both 40 and 50 ⁇ g siRNA dosages achieved significant target gene silencing (N 3, *P ⁇ 0.05).
- FIG. 7 Evaluation of the cytokine response in the mouse lung after HKP-siRNA nanoparticles delivery. HKP-siRNA at different dosages were oraltracheally administrated in the mouse lungs. The total lung tissue was harvested for protein isolation and cytokine measurements by ELIS A assay.
- FIG. 1 Standard curve to measure IFN-a concentration was prepared according to in-house SOP (Lowry Method); B. IFN-a concentration in each sample was determined and normalized to total protein.
- the present invention provides RN A pharmaceutical formulations comprising either siRNA or mRNA sequences for the prevention and treatment of Zika virus infection.
- One formulation comprises siRNA sequences targeting the viral genome RNA and mRNA for knocking down the viral activity, and a carrier, such as Histidine-Lysine Co-polymers (HKP) or Spermine-liposome Conjugates (SLiC).
- Another formulation comprises mRNA sequences coding viral proteins critical for Zika viral infection and replication, and a carrier serving as an adjuvant for amplifying an immune response against the Zika virus within the host.
- the invention also provides methods of use for the pharmaceutical formulations for the prevention and therapeutic treatment of Zika virus Infection.
- siRNA molecule or an “siRNA duplex” is a duplex
- oligonucleotide that is a short, double-stranded polynucleotide, that interferes with the expression of a gene in a cell, after the molecule is introduced into the cell, or interferes with the expression of a viral gene.
- it targets and binds to a complementary nucleotide sequence in a single stranded (ss) target RNA molecule.
- siRNA molecules are chemically synthesized or otherwise constructed by techniques known to those skilled in the art. Such techniques are described in U.S. Pat. Nos. 5, 898,031, 6,107,094, 6,506,559, 7,056,704 and in European Pat. Nos. 1214945 and 1230375, which are incorporated herein by reference in their entireties.
- the sequence refers to the sense strand of the duplex molecule.
- One or more of the ribonucleotides comprising the molecule can be chemically modified by techniques known in the art.
- the backbone of the oligonucleotide can be modified.
- siRNA molecules of the invention target a conserved region of the genome of a ZIKV.
- target or “targets” means that the molecule binds to a
- a "conserved region" of a ZIKV gene is a nucleotide sequence that is found in more than one strain of the virus, is identical among the strains, rarely mutates, and is critical for viral infection and/or replication and/or release from the infected cell.
- the targeted conserved region of the genome comprises sequences coding for envelope protein or nonstructural proteins NS1, NS3, NS4B, or NS5.
- the siRNA molecule can target envelope gene expression, NS1 gene expression, NS3 gene expression, NS4B gene expression, or NS5 gene expression.
- the targeted conserved region of the genome is the 3' untranslated region (3'-UTR) of the virus.
- the siRNA molecule can target the 3' untranslated region (3'-UTR) of the virus.
- the siRNA molecule is a double-stranded oligonucleotide with a length of about 19 to about 29 base pairs. In one aspect of this embodiment, the molecule is a double-stranded oligonucleotide with a length of 19 to 21 base pairs. In another aspect of this embodiment, it is a double-stranded oligonucleotide with a length of 25 base pairs. In all of these aspects, the molecule may have blunt ends at both ends, or sticky ends with overhangs at both ends (unpaired bases extending beyond the main strand), or a blunt end at one end and a sticky end at the other. In one particular aspect, it has blunt ends at both ends. In another particular aspect, the molecule has a length of 25 base pairs (25 mer) and has blunt ends at both ends. In another particular aspect, the siRNA molecule is one of those identified in Table 4.
- the siRNA molecules of the invention also include ones derived from those listed in Table 4 or otherwise disclosed herein.
- the derived molecules can have less than the 25 base pairs shown for each molecule, down to 17 base pairs, so long as the "core" contiguous base pairs remain. That is, once given the specific sequences shown herein, a person skilled in the art can synthesize molecules that, in effect, "remove” one or more base pairs from either or both ends in any order, leaving the remaining contiguous base pairs, creating shorter molecules that are 24, 23, 22, 21, 20, 19, 18, or 17 base pairs in length, if starting with the 25 base pair molecule.
- the derived molecules of the 25 mer molecules disclosed in Table 4 includes: a) 24 contiguous base pairs of any one or more of the molecules; b) 23 contiguous base pairs of any one or more of the molecules; c) 22 contiguous base pairs of any one or more of the molecules; b) 21 contiguous base pairs of any one or more of the molecules; d) 20 contiguous base pairs of any one or more of the molecules; e) 19 contiguous base pairs of any one or more of the molecules; f) 18 contiguous base pairs of any one or more of the molecules; and g) 17 contiguous base pairs of any one or more of the molecules.
- the derived molecules can have more than the 25 base pairs shown for each molecule, so long as the initial 25 contiguous base pairs remain. That is, once given the specific sequences disclosed herein, a person skilled in the art can synthesize molecules that, in effect, "add" one or more base pairs to either or both ends in any order, creating molecules that are 26 or more base pairs in length and containing the original 25 contiguous base pairs.
- compositions comprising One or More siRNA Molecules
- the invention includes a pharmaceutical composition
- a pharmaceutical composition comprising an siRNA molecule that targets a conserved region of the genome of a ZIKV and a pharmaceutically acceptable carrier.
- the carrier condenses the molecules to form a nanoparticle.
- the composition may be formulated into nanoparticles.
- the compositions may be lyophilized into a dry powder.
- the pharmaceutically acceptable carrier comprises a polymeric nanoparticle or a liposomal nanoparticle.
- the composition comprises at least two different siRNA molecules that target one or more conserved regions of the genome of a ZIKV and a pharmaceutically acceptable carrier.
- compositions are sometimes referred to herein as a "cocktail.”
- the gene sequences in the conserved regions of the ZIKV are critical for the viral infection of a mammal.
- the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
- the mammal is a human.
- composition can include one or more additional siRNA molecules that target still other conserved regions of the ZIKV genome.
- composition comprises siRNA molecules that target the
- Envelope (E) gene of the ZIKV genome the molecules are selected from the group consisting of:
- ZIKV14 CCUUGACAAGCAAUCAGACACUCAA
- ZIKV17 CCGGAACUCCACACUGGAACAACAA .
- the composition comprises siRNA molecules that target the NSl gene of the ZIKV genome.
- the molecule comprises ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA.
- the composition comprises siRNA molecules that target the NS3 gene of the ZIKV genome.
- the molecules are selected from the group consisting of:
- ZIKV62 GCCUAUCAGGUUGCAUCUGCCGGAA ,
- ZIKV63 CCUAUCAGGUUGCAUCUGCCGGAAU ,
- the composition comprises siRNA molecules that target the NS4B gene of the ZIKV genome.
- the molecule comprises ZIKV74: CCACUUCAUACAACAACUACUCCUU.
- the composition comprises siRNA molecules that target the NS5 gene of the ZIKV genome.
- the molecule comprises ZIKV103 : GGUGCGCAGGAUCAUAGGUGAUGAA.
- the composition comprises siRNA molecules that target the 3'-UTR region of the ZIKV genome.
- the molecule comprises: ZIKV105: C C GAG A AC GC C AUGGC AC GG A AG A A .
- the composition comprises a cocktail, MSTZIKV13, wherein a first siRNA molecule comprises ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU and a second siRNA molecule comprises ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA.
- the composition comprises a cocktail, MSTZIKV62, wherein a first siRNA molecule comprises ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA and a second siRNA molecule comprises ZIKV74: CCACUUCAUACAACAACUACUCCUU.
- composition comprises a cocktail, MSTZIKV62B, wherein a first siRNA molecule comprises ZIKV62B:
- a second siRNA molecule comprises ZIKV17: CCGGAACUCCACACUGGAACAACAA.
- composition comprises a cocktail, MSTZIKV103, wherein a first siRNA molecule comprises ZIKV103 :
- a second siRNA molecule comprises KIKV63 : CCUAUCAGGUUGCAUCUGCCGGAAU
- a third siRNA molecule comprises ZIKVl 05 : CCGAGAACGCCAUGGC ACGGAAGAA.
- a pharmaceutically acceptable carrier comprises a polymeric nanoparticle or a liposomal nanoparticle.
- the siRNA molecules comprise 25 mer blunt-end siRNA molecules and the carrier comprises a Histidine-Lysine copolymer or Spermine-Lipid Conjugate (SLiC) and cholesterol.
- Pharmaceutically Acceptable Carriers for the siRNA Molecules include saline, sugars, polypeptides, polymers, lipids, creams, gels, micelle materials, and metal nanoparticles.
- the carrier comprises at least one of the following: a glucose solution, a polycationic binding agent, a cationic lipid, a cationic micelle, a cationic polypeptide, a hydrophilic polymer grafted polymer, a non-natural cationic polymer, a cationic polyacetal, a hydrophilic polymer grafted polyacetal, a ligand functionalized cationic polymer, a ligand
- the polymers comprise a biodegradable histidine-lysine polymer, a biodegradable polyester, such as poly(lactic acid) (PLA), poly(glycolic acid) (PGA), and poly(lactic-co-glycolic acid) (PLGA), a polyamidoamine (PAMAM) dendrimer, a cationic lipid, or a PEGylated PEL Cationic lipids include DOTAP, DOPE, DC-Chol/DOPE, DOTMA, and DOTMA/DOPE.
- PLA poly(lactic acid)
- PGA poly(glycolic acid)
- PLGA poly(lactic-co-glycolic acid)
- PAMAM polyamidoamine dendrimer
- a cationic lipid or a PEGylated PEL Cationic lipids
- PEGylated PEL Cationic lipids include DOTAP, DOPE, DC-Chol/DOPE, DOTMA, and DOTMA/DOPE
- the carrier is a polymer.
- the polymer comprises a histidine-lysine copolymer (HKP).
- HTP histidine-lysine copolymer
- Such copolymers are described in U.S. Pat. Nos. 7,070,807 B2, 7,163,695 B2, and 7,772,201 B2, which are incorporated herein by reference in their entireties.
- the HKP forms a nanoparticle with the siRNA molecule, wherein the diameter of the nanoparticle is about lOOnm to about 400 nm.
- the HKP and the siRNA molecules self-assemble into nanoparticles or can be formulated into nanoparticles.
- the carrier is a liposome.
- the liposome comprises a cationic lipid conjugated with cholesterol.
- the cationic lipid comprises a spermine head and one or two oleyl alcoholic tails.
- the liposome comprises a Spermine-Liposome-Conjugate (SLiC) and cholesterol. Examples of such molecules are disclosed in Figure 5.
- the liposome and the siRNA molecules self-assemble into nanoparticles or can be formulated into nanoparticles.
- the invention also includes methods of using the siRNA molecules and
- compositions containing them to prevent or treat ZIKV disease As used herein "treat” or “treatment” refers to reducing the severity of or curing ZIKV disease.
- a therapeutically effective amount of the composition of the invention is administered to a mammal.
- the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
- rodent e.g., rat, mouse, or guinea pig
- ferret e.g., a monkey
- non-human primate e.g., a monkey
- the mammal is a laboratory animal, such as a rodent.
- the mammal is a non-human primate, such as a monkey.
- the mammal is a human.
- a "therapeutically effective amount” is an amount that prevents, reduces the severity of, or cures ZIKV disease. Such amounts are determinable by persons skilled in the art, given the teachings contained herein.
- a therapeutically effective amount of the pharmaceutical composition administered to a human comprises about 1 mg of the siRNA molecules per kilogram of body weight of the human to about 5 mg of the siRNA molecules per kilogram of body weight of the human.
- mRNA Vaccines The invention provides a vaccine comprising an mRNA molecule that codes for an amino acid sequence encoded by a conserved region of the genome of a ZIKV and a pharmaceutically acceptable carrier comprising a polymer or a liposome.
- a "conserved region" of a ZIKV gene is a nucleotide sequence that is found in more than one strain of the virus, is identical among the strains, rarely mutates, and is critical for viral infection and/or replication and/or release from the infected cell.
- the gene sequence in the conserved region of the ZIKV genome is critical for the viral infection of a mammal.
- the conserved region of the genome comprises gene sequences coding for the Envelope protein of ZIKV.
- the gene sequences code for amino acid sequences within Domain III of the Envelope protein.
- the polymer comprises a Histidine-Lysine co-polymer (HKP).
- the HKP and the mRNA molecules self-assemble into nanoparticles.
- the HKP and mRNA molecules are formulated into nanoparticles.
- the liposome comprises a Spermine-Lipid Conjugate (SLiC) and cholesterol.
- the SLiC and cholesterol and the siRNA self-assemble into nanoparticles.
- the SLiC and cholesterol and the mRNA molecules are formulated into nanoparticles.
- the SLiC and cholesterol also acts as an adjuvant for amplifying the immune response.
- the mRNA vaccines of the invention are used to prevent a ZIKV infection or reduce its severity.
- the invention includes method of preventing or reducing the severity of a ZIKV infection in a mammal comprising administering to the mammal a therapeutically effective amount of the vaccine prior to infection.
- the vaccine is administered to the mammal through injection instillation or intradermal, intravenous, intraperitoneal, intravenous, intravaginal, or subcutaneous administration.
- the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
- the mammal is a laboratory animal, such as a rodent.
- the mammal is a non-human primate, such as a monkey.
- the mammal is a human.
- Example 1 Design mRNA targeting the envelope protein of ZIKV
- ZIKV E protein represents a major target for development of a neutralizing antibody.
- the ZIKV E protein contains three distinct domains (shown in Figure 1).
- the mRNA constructs containing Envelope Domain II of ZIKV will be transfected into human cells in vitro using a variety of commercially available transfection agent.
- Cells to be used for these studies included HEK293T, VERO cells, A549 cells and others.
- electroporation using MaxCyte technology
- the various delivery processes are aimed at determining which will give good uptake into a variety of cells and to see subsequent expression of the construct. The aim is to determine protein production by each construct and also to determine whether the product is secreted from the cells. This process is not necessarily identified a clinically viable delivery process.
- mRNA will be detected in live cells using SmartFlare probes (Millipore) or through use of QRTPCR. mRNA of GFP was taken as the positive control ( Figure 9).
- Example 3 Detection of mRNA uptaking into cells using SmartFlare technology
- These smart flares are beads that have a sequence attached that, when recognizing the RNA sequence in the cell, produce an increase in fluorescence. Smartflares will be designed against several regions along the constructs in case steric hindrance reduces signal from one region.
- the protein expressed by the mRNA construct will be identified and quantitated by RPHPLC using an analytical C18 column (250mm x 2.1mm; Phenomenex). Protein detection will use a dual wavelength detector. A gradient of 0.1%TFA/Acetonitrile will be adjusted over time to allow analytical separation of protein peaks. In initial experiments, fractions will be collected and submitted for Mass Spectrometry to determine the presence of the expected sequence. The secreted product and the product manufactured within the cells will be compared using protein sequencing. To mitigate enzyme degradation of the sample, we will use enzyme inhibitors in the media and concentrated media from multiple wells in order to detect the product on HPLC.
- Example 5 Examination of siRNAs against Zika in a CPE assay
- siRNAs for testing will be provided to Immuquest (Frederick, MD) for analysis in their Zika CPE assay.
- control and test siRNAs will be administered at select intervals ahead of the virus and the degree of effect monitored by examining the change in the CPE value.
- Example 6 Determine best nanoparticle for delivery
- branched polypeptides e.g. Histidine Lysine Ploymer or HKP
- various derivatives that, like the HKP, have been modified with targeting ligands to allow tissue specific delivery or PEGylated varieties that should assist in uptake across the mucosa of the vagina.
- HKP Histidine Lysine Ploymer
- a spermine/spermidine co-polymer carrier and a liposomal delivery agent that, like the HKP, protect the siRNA and provide high efficiency delivery to tissues.
- the various peptide/lipid formulations will be evaluated for their ability to form nanoparticles with single siRNAs or combinations of siRNAs against different targets. Binding with the nucleic acid will be evaluated by gel electrophoresis. Nanoparticle formation will be studied by particle size measurements using DLS (Dynamic Light Scattering) and the charge/size distribution measured using a nanoparticle size/charge instrument (Malvern instruments D9000).
- TEM electron microscopy
- Example 7 SLiC/mRNA/siR A nanoparticle SLiC Liposome Preparation Regular methods will be tried at first to prepare liposomes with newly synthesized SLiC molecules, such as thin film method, solvent injection and so on without much success.
- Norbert Maurer et al reported a method of liposome preparation in which siRNA or oligonucleotide solution will be slowly added under vortexing to the 50% ethanol solution (v/v) of liposome and ethanol was later removed by dialysis.
- the nanoparticles thus derived will be small in size and homogeneous.
- mRNA and siRNA will be directly wrapped by cationic lipids during formation of liposome, while in most other methods mRNA or siRNA are loaded (or entrapped) into preformed liposome, such as Lipofectamine 2000. Lipids dissolved in ethanol are in so-called metastable state in which liposomes are not very stable and tend to aggregate. We will then prepare un-loaded or pre-formed liposomes using modified Norbert Maurer's method. (Maurer, N., A., Mori, L., Palmer, M. A., Monck, K. W. C, Mok, B., Mui, Q. F., Akhong, and P. R., Cullis. 1999.
- Liposomes were prepared by addition of lipids (cationic SLiC /cholesterol, 50:50, mol %) dissolved in ethanol to sterile dd-LbO.
- the ethanolic lipid solution need to be added slowly under rapid mixing. Slow addition of ethanol and rapid mixing were critical for the success in making
- SLiC liposomes as the process allows formation of small and more homogeneous liposomes. Unlike conventional methods in which mRNA or siRNAs are loaded during the process of liposome formulation and ethanol or other solvent is removed at end of manufacturing, our SLiC liposomes were formulated with remaining ethanol still in the solution so that liposomes were thought to be still in metastable state. When mRNA or siRNA solution was mixed/loaded with liposome solution cationic groups lipids will interact with anionic siRNA and condense to form core. SLiC liposomes' metastable state helped or facilitated liposome structure transformation to entrap mRNA or siRNA more effectively. Because of the entrapment of mRNA/siRNA, SLiC liposomes become more compact and homogeneous.
- SLiC liposome formation After the liposome formation, we have developed an array of assays to characterize the physicochemical properties of SLiC liposome, including particle size, surface potential, morphology study, mRNA or siRNA loading efficiency and biological activity, etc.
- the particle size and zeta-potentials of SLiC liposomes were measured with Nano ZS Zeta Sizer (Malvern Instruments, UK). Each new SLiC liposome was tested for particle size and zeta-potential when ethanol contents changed from 50% to 25% and to 12.5%. Data were derived from formulations of different ethanol contents. All SLiC liposomes were prepared at lmg/ml in concentration and loaded with siRNA (2: 1, w/w).
- SLiC Liposomes were composed of cationic SLiC and cholesterol dissolved in ethanol at 12.5%, e.g. TM2 (12.5).
- TM2 cationic SLiC
- the average particle sizes of three sequential measurements and the average zeta-potentials of three sequential measurements were illustrated in Table 5.
- liposomes particle sizes became much smaller when they were loaded with mRNA or siRNA at 2: 1 (w/w) resulting in particle sizes in the range of 110 to 190nm in diameter and much lower PDI values.
- Conventional consideration of liposomal structure dictates that mRNA or siRNA is loaded or interacted with cationic lipids through electrostatic forces and liposomes wraps mRNA or siRNA to form spherical particles in shape in order to reduce surface tension. As the result, the liposomes particle sizes became much smaller after loaded with mRNA or siRNA. Liposomes formulated with mRNA or siRNA also have lower surface charge, which could be explained by neutralizing effect from loaded mRNA or siRNA.
- Example 8 Mouse model study for mRNA vaccine
- mice will be five-week old at the beginning of this study.
- 20 mice of prophylactic group will be intravenously injected with mRNA combination encapsidated with HKP-SLiC nanoparticle system from the tail vein.
- the other 20 mice of control group will be injected with PBS. 14 days later, the serum of all mice will be collected, and the mice from both prophylactic group and control group will be intravenously injected with ZIKV.
- mice All mice will be weighed and the survival of each group will be counted daily.
- the serum of infected mice will be taken at 1, 3, 5, 7, 9 and 14-day post-infection.
- the tumor necrosis factor alpha (TNF-a) will be detected using enzyme-linked immunosorbent assay (ELISA).
- ELISA enzyme-linked immunosorbent assay
- the tissues including testes, spleen, liver, heart, brain and kidney will be collected at 24 and 72 h post-infection.
- the total RNA from the tissues will be extracted, one-step quantitative real-time PCR and 5'-RACE assay will be performed as described in the above in vitro study part.
- the viral titers in the sera and tissues will be detected in Vero cells with serially dilution.
- the results from vaccination group will be compared with the control groups to evaluate the protective efficiency of the mRNA vaccine candidates.
- siRNAs By performing recursive analyses of siRNAs against 28 sequenced Zika strains (including both African and Asian lineages) we identified siRNAs that were able to cover an extensive number of strains (Table 2). Furthermore, careful selection of multiple siRNAs can be seen to provide complete coverage against all strains used in our predictions (Table 3). Further sequence analysis showed that the 11 elected anti-ZIKV siRNA target the highly homologous regions of Envelope (E) protein, non-structure protein NS1, NS3, NS4B, NS5 and 3'-UTR in all ZIKV strains (Table 4 and Figure 1). If the mRNA translation and genome replication of ZIKV mimic DENV and take place in the cytoplasm, the siRNA candidates should work efficiently to degrade both positive and negative strand of the viral RNAs in ZIKV.
- E Envelope
- Example 10 Cell Culture Based Screening for Potent Anti-ZIKV siRNA Oligos siRNAs will be tested for their silencing activities using a reporter assay where the regions of the ZIKV genome to be silenced are incorporated into an expression vector. An appropriate ZIKV gene segment will be cloned into psiCheckTM-2 (Promega). The cells will be co-transfected with reporter plasmids and siRNAs and the expression of renilla luciferase will be normalized to firefly luciferase expression (transfection efficiency control).
- RTPCR RT-PCR
- Example 11 Identify potent combinations of siRNAs to improve strain coverage.
- siRNA mixtures for evaluation in the CPE assay.
- siRNAs will selecte the siRNAs to include in the mixtures based on their efficacy as well as the degree of overlap between Zika strains.
- Example 12 Develop and characterize nanoparticles for in vivo delivery of siRNAs.
- the most critical and challenging aspect of developing an siRNA-based therapeutic is the delivery of the siRNA into the cells where ZIKV infection can be transmitted. Since ZIKV has been shown to be transmitted by sexual intercourse we have elected to pursue intravaginal siRNA delivery as a means of validating this as a preliminary approach to therapy. Once more is understood about the etiology of the disease we may be able to adjust the delivery approach to treat therapeutically.
- the vaginal tract is a suitable site for the administration of both local and systemic acting drugs.
- vaginal products on the market such as those approved for contraception, treatment of yeast infection, hormonal replacement therapy, and feminine hygiene. However, there are many biological barriers and factors that protect the vagina from foreign particles, such as a thick and elastic mucus layer that may bind and inhibit access of these agents.
- PLGA, PEG or PEG-PEI to this HKP nanoparticle or evaluation of a spermine/spermidine copolymer that exhibits suitable molecular characteristics for delivery including size, charge and hydrophobicity.
- Previous studies have shown significant success with PLGA, PEG and other modifications to existing nanoparticle formulations and we have demonstrated functionalization of our nanoparticles with these moieties while retaining this optimal size, ability to carry and protect the siRNAs while penetrating the mucus layer. Finally, others have shown that spray dried powders can enhance intravaginal siRNA delivery [16]. We have also demonstrated that we can lyophilize our HKP nanoparticle complexed with siRNA without adversely affecting its characteristic size or delivery capacity.
- Example 13 Mice model study for anti-ZIKV siRNA
- AG129 mice will be used as the animal model. We will perform all mouse studies in Biosafety level-2 conditions. All AG129 mice will be five-week old at the beginning of this study. 20 mice of treatment group will be intravenously injected with siRNA combination encapsidated with HKP-SLiC nanoparticle system from the tail vein. The other 20 mice of control group will be injected with PBS. 24 h later, mice from treatment group and control group will be infected intravenously with ZIKV, respectively. siRNA or PBS will be intravenously injected into the treatment or control group at 0, 24, 48 and 72 h post-infection.
- mice All mice will be weighed and the survival of each group was counted daily.
- the serum of infected mice will be taken at 1, 3, 5, 7, 9 and 14-day post-infection.
- the tumor necrosis factor alpha (TNF-a) will be detected using enzyme-linked immunosorbent assay (ELISA).
- ELISA enzyme-linked immunosorbent assay
- the studies are expected to demonstrate that the cell cultures infected with ZIKV exhibit up-regulated expression of tumor necrosis factor-a (TNF-a), and interleukin- ⁇ (IL- ⁇ ).
- the tissues including testes, spleen, liver, heart, brain and kidney will be collected at 24, 28 and 72 h post-infection.
- the total RNA from the tissues were extracted, one-step quantitative real-time PCR and 5 '-RACE assay will be performed.
- the viral titers in the sera and tissues will be detected in Vero cells with serially dilution.
- the results from treatment groups will be compared with the control
- Table 2 Table showing strains used for siRNA prediction and siRNA sequence overlap.
- Table 4 The list of sequence and targets for anti-ZIKV siRNA candidates Table 5 Characterization indexes of five SLiC species and five SLiC-siRNA nanoparticles, including particle sizes, poly-dispersity index (PDI) and Zeta-potential.
- PDI poly-dispersity index
- Zika vs Dengue virus serotype 3 Zika vs Dengue virus serotype 4
- Zanluca C Melo VC, Mosimann AL, Santos GI, Santos CN, Luz K. First report of autochthonous transmission of Zika virus in Brazil. Mem Inst Oswaldo Cruz.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Genetics & Genomics (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Molecular Biology (AREA)
- Virology (AREA)
- Organic Chemistry (AREA)
- Epidemiology (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Microbiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Physics & Mathematics (AREA)
- General Chemical & Material Sciences (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Dispersion Chemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Immunology (AREA)
- Mycology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicinal Preparation (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention provides prophylactic and therapeutic pharmaceutical compositions comprising either siRNA or mRNA molecules for prevention and treatment of Zika virus infection and provides methods for their use.
Description
RNA PHARMACEUTICAL FORMULATIONS FOR PROPHYLACTIC AND THERAPEUTIC TREATMENT OF ZIKA VIRUS INFECTION
CROSS-REFERENCE TO RELATED PATENT APPLICATIONS
This application claims the benefit of and priority to U.S. Provisional Patent
Application No. 62/465,096, filed February 28, 2017, which is incorporated herein by reference in its entirety.
FIELD OF INVENTION
This invention presents novel prophylactic and therapeutic agents for prevention and treatment of Zika virus (ZIKV) infection and methods for their use. BACKGROUND
Zika virus infection: Biology and Pathology
Initially identified in 1947 in Uganda, the Zika virus (a member of the flaviviridae family including West Nile Virus, Dengue and Chikungunya viruses) has recently garnered great interest because of a link between infection in pregnant women and miscarriage or birth defects in their infants. Transmission is via the bite of an infected Aedes mosquito, and a rapid increase in transmission observed in Brazil since late 2014 preceded an increase in the number of cases of microcephaly in infants reported to the Brazil Ministry of Health in 2015 [1, 2]. As of November 2016, a total of more than 4, 100 suspected microcephaly cases had been reported, many of which occurred in infants born to women who lived in or had visited areas where Zika virus transmission was observed [3]. There is now evidence of an association between Zika virus infection during early pregnancy and the occurrence of microcephaly. Viral infection has typically only resulted in patients presenting symptoms of mild fever, rash, conjunctivitis and arthralgia [4]. In the outbreak in Brazil, Zika virus (ZIKV) was detected by reverse transcription-polymerase chain reaction from the sera of eight patients and the result was confirmed by DNA sequencing. Phylogenetic analysis suggested that the ZIKV identified in this region belonged to the Asian clade [4]. ZIKA virus infections have now been identified and validated in the mainland US in Florida and gulf coast states, and the need for suitable
prophylactic or therapeutic options is significant.
ZIKV is a single-strand positive RNA virus, belonging to family Flaviviridae; genus Flavivirus. The genome of ZIKV consists of about 10K nucleotides, which code at least three structural proteins (capsid (C), membrane (prM; processed to M) and envelope (E)), and seven non-structural proteins (NSl, NS2A, NS2B, NS3, NS4A, NS4B and NS5). In ZIKV, the single ORF is flanked by 5'- and 3'- untranslated regions (UTRs), respectively (Figure 1). Both the 5' and the 3' UTRs form several additional RNA elements that also play important roles in regulation of mRNA translation and are required for viral RNA replication, as well as in interactions with viral and cellular proteins. RNA elements of the 3' UTR greatly enhance viral RNA synthesis. ZIKV infection initiates from envelope (E) glycoproteins on the surface of the ZIKV virion binding to a host cell receptor, such as AXL and broadly neutralizing antibodies have been demonstrated against this E protein region [5]. ZIKV enters the host cell by endocytosis, and then its genome escapes from the endosome into the cytoplasm after viral-host membrane fusion driven by the fusion loop. In the cytoplasm, the viral genome is first translated into a large polyprotein, and then cleaved by host signal peptidases and a viral serine protease (during and after translation) to generate the viral proteins. Also in the cytoplasm, the positive strand RNA genome produces a negative strand RNA template to generate new copies of the genome.
AXL, a member of the receptor tyrosine kinase subfamily, has been identified as the major receptor for ZIKV binding. The study showed that blocking or silencing AXL dramatically reduced ZIKV replication in fibroblasts and alveolarepithelial cells [6]. The function of AXL. includes transducing signals from the extracellular matrix into the cytoplasm by binding growth factors, such as vitamin K-dependent protein growth-arrest-specific gene 6 (GAS6), as well as being involved in the stimulation of cell proliferation. AXL is broadly distributed in different cell types in humans.
Zika strains and similarities
For a prophylactic or therapeutic to have viability it is important to ensure that it is able to function against a wide array (if not all) strains of a virus. We used bioinformatics to identify regions of sequence identity in the regions to be used as epitopes for vaccine or targets for therapeutic siRNA development across an extensive number of strains. ZIKV has circulated in both Africa and Asia since at least the 1950s. Haddow et al determined the nucleotide sequences of the open reading frames of five isolates from
Cambodia, Malaysia, Nigeria, Uganda, and Senegal collected between 1947 and 2010. Using phylogenetic analyses of these (and previously published) ZIKV sequences, the authors identified the existence of two main virus lineages (African and Asian) [7]. In 2013, Baronti et al were able to obtain the complete coding sequence of a Zika virus strain belonging to the Asian lineage, isolated from an infected patient returning from French Polynesia. Based on nucleotide and amino acid sequence composition, the African and Asian strains were most divergent while strains within a geographical area were least divergent [8]. More than 38 strains have now been sequenced and can be assigned to the African or Asian lineages. Advantages of mRNA vaccines
As a carrier for genetic information that specifies the amino acid sequence of the encoded protein, mRNA is a nontoxic molecule that allows transient protein expression of a desired protein product in nearly all transducable cell types. Compared with traditional plasmid and viral-based approaches, this approach allows design of patient personalized mRNAs that also benefit by not needing to pass through the nuclear membrane (as DNA does) and thus carries little to no risk of genomic integration. mRNA vaccines have also been demonstrated to promote immunogenicity in both very young and very old mice. If this translates to humans, then this would be beneficial since these demographics typically do not mount a robust immunological reaction to vaccines. Recently, self-amplifying mRNA vaccines have been demonstrated to be safe and efficacious against other viruses (e.g. Influenza). Generation of a robust immune response in infants and the elderly has always been an issue for influenza vaccines. However, mRNA
vaccines may benefit in that they have been demonstrated to induce balanced, long-lived and protective immunity to influenza A virus infections in even very young and very old mice. Vaccines based on mRNA or RNA replicons have also been shown to be immunogenic in a variety of animal models, including nonhuman primates [10]. Target selection for mRNA vaccine against ZIKV
Amongst all ZIKV proteins, E protein is responsible for the host cell receptor binding and virus entry into the cells. Therefore, E protein represents a major target for development of a neutralizing antibody. The ZIKV E protein contains three distinct domains (shown in Figure 1C): a central β-barrel domain (Domain I; aa. 1-51, 132-192 and 280-295), an extended dimerization domain (Domain II; aa. 52-131 and 193-279), and an immunoglobulin-like domain (Domain III; aa. 297-406) (Figure 1C.) [5]. The fusion loop, a hydrophobic sequence located in Domain II (aa. 98-109), is responsible for the pH-dependent conformational changes during viral-host membrane fusion. In several immunology studies, the results suggested that Domain III of E protein contains the determinants that can be recognized by a number of flavivirus type-specific neutralizing monoclonal antibodies (mAb) [5]. Cross- neutralizing mAbs targeting fusion loops (in Domain II) exhibit lower affinity (Table 1). The protective efficiency of those type-specific mAb has been proven in the animal model. However, ZIKV and dengue virus (DENV) share the same properties by which antibodies against select regions of the virus (e.g. the fusion loop of Envelope protein Domain II) may stimulate Antibody Dependent Enhancement (ADE) of immune response (when non-neutralizing antiviral proteins facilitate virus entry into host cells and increase infectivity in these cells resulting in higher viremia and more severe symptoms). Some people suffered a more severe form of DENV because of the cross-serotypic immune response, or ADE. When such a person was infected with stains of different DENV serotypes (totally 4 identified serotypes), the antibodies to the old strain interfered with the immune response to the new strain and resulted in more virus uptake [11]. siRNA: a flexible molecular platform for prophylactic and therapeutic efficacy against ZIKA virus
RNA interference (RNAi) is a naturally occurring, highly specific mode of gene regulation that has broad potential applications in both research and therapeutic settings. The mechanics of RNAi are both exquisite and highly discriminating. At the onset, short (19-21bp) double stranded RNA sequences (referred to as short interfering RNAs, siRNAs) associate with the cytoplasmically localized RNA Interference Silencing Complex (RISC). The resultant ribonucleoprotein complex then searches the resident population of messenger RNAs (mRNAs) for complementary sequences, eventually degrading (and/or attenuating translation of) these transcripts and effectively down-regulating the expression of the targeted gene.
Scientists have co-opted the endogenous RNAi machinery to advance a wide range of studies involving gene function analysis, pathway mapping, drug target validation, and host-pathogen interactions. As siRNAs can be designed to target virtually any gene and can be introduced into cells by a variety of methods, RNAi represents a highly flexible platform by which researchers and clinicians can control diseases, including infectious diseases.
RNA interference has previously been employed to target a range of human pathogenic viruses, including HIV, hepatitis, respiratory syncytial virus, polio virus, SARS coronavirus, Marburg, dengue, foot-and-mouth disease and Influenza. The ability to rapidly predict siRNA sequences with potency against a virus and the fact that siRNAs of the same length show similar molecular weight, charge and hydrophobicity, combined with highly efficient nanoparticle delivery systems that are agnostic for carrying these siRNAs independent of the sequence being targeted, makes RNAi a valuable tool in the arsenal against emerging viral threats and can act as an intermediate option while other therapeutic strategies, such as vaccines and small molecule therapeutics, are developed and validated. Additionally, RNAi has the ability to (1) efficiently limit viral replication without reliance on host immune functions, and (2) target multiple genes and/or sequences simultaneously, making this an ideal therapeutic approach for viruses like Zika (where a large number of strains may need to be targeted and no other options currently exist) or against other viruses (like Influenza), which have rapidly evolving genomes.
Target selection for therapeutic siRNA against ZIKV
By performing recursive analyses of siRNAs against 28 sequenced Zika strains (including both African and Asian lineages), we identified siRNAs that were able to cover an extensive number of strains (Table 2). Furthermore, careful selection of multiple siRNAs can be seen to provide complete coverage against all strains used in our predictions (Table 3).
Combining 2 siRNA sequences can provide 100% coverage as seen by combining siRNA #62 and siRNA #62A (Table 3). However, in our Influenza studies, we have identified that siRNAs targeting different regions of the viral genome can produce synergistic silencing and antiviral efficacy compared with multiple siRNAs targeting the same gene. Therefore, while we can see complete coverage with certain combinations, we may see higher potency against the selected strains using a combination (such as 13 & 62) targeting distinct regions of the virus. By comparing combinations of the siRNAs as shown in Table 1, we can determine the relative potencies of each combination and compare this with the combination of 62 and 62A or 62A and 63 (targeting all strains). If we see a marked increase in potency, combining 2 siRNA sequences can provide 100% coverage as seen by combining siRNA #62 and siRNA #62a. However, in our Influenza studies, we have identified that siRNAs targeting different regions of the viral genome can produce synergistic silencing and antiviral efficacy compared with multiple siRNAs targeting the same gene. Therefore, while we can see complete coverage with certain combinations, we may see higher potency against the selected strains using a combination (such as 13 & 62) targeting distinct regions of the virus. By comparing combinations of the siRNAs as shown in Table 1, we can determine the relative potencies of each combination and compare this with the combination of 62 and 62A or 62A and 63 (targeting all strains, Table 3).
BRIEF DESCRIPTION OF THE FIGURES Figure 1. The molecular basis and strategy of development of prophylactic and therapeutic RNA against zika virus infection. ZIKV is a single-strand positive RNA virus, belonging to family Flaviviridae; genus Flavivirus. The genome of ZIKV consists of about 10K nucleotides,
which code at least three structural proteins (capsid (C), membrane (prM; processed to M) and envelope (E)), and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5). In ZIKV, the single ORF is flanked by 5'- and 3'- untranslated regions (UTRs), respectively. Amongst all ZIKV proteins, E protein is responsible for the host cell receptor binding and virus entry into the cells. Therefore, E protein represents a major target for development of a neutralizing antibody. The ZIKV E protein contains three distinct domains (shown in Figure 1): a central β-barrel domain (Domain I; aa. 1-51, 132-192 and 280-295), an extended dimerization domain (Domain II; aa. 52-131 and 193-279), and an
immunoglobulin-like domain (Domain III; aa. 297-406). Figure 2. Histidine-Lysine co-polymer enhances topical and subcutaneous siRNA deliveries in vivo. (A) The HKP/siRNA can self-assemble into nanoparticles (average 150nm in diameter). (B) The nanoparticles can be dissolved in aqueous solution, can be lyophilized into dry powder, and can be redissovled and mixed with methylcellulose, or with RNAse free water. (C) HKP/siRNA nanoparticle delivery to mouse respiratory track: upper airway, bronchi, alveoli.
Figure 3. Comparison of target knockdown of lung endogenous gene among HKP, DOTAP and D5W after oral tracheal deliveries of siRNA with three different dosing regimens. HKP demonstrated the efficient siRNA-mediated knockdown of the target gene at the 20μg dose.
Figure 4. Intraperitoneal delivery of PAA-siRNA formulation demonstrated a therapeutic efficacy against H1N1 in the viral challenged mice (n = 15). The viral challenges through intranasal administrations of lx LD50 H1N1 (A/California/04/2009) were conducted on day 1 (red arrow) for the virus only, Tamiflu and siRNA treatment groups. The H1N1 challenged mice were treated with various dosages of PAA-siRNA combination
(siRNA89-siRNA103 with a 1 : 1 ratio), 1 mg/kg, 5 mg/kg and 10 mg/kg, through
intraperitoneal administration daily, from day 2 to day 6 (black arrows). Adapting the same route and dosing regimen, 25 mg/kg Tamiflu was also administrated daily on the H1N1 infected mice. The therapeutic efficacy of 10 mg/kg/day of PAA-siRNA combination
resulted in almost equal anti-influenza activity to that of 25 mg/kg/day of Tamiflu treatment.
Figure 5. The structures of Spermine-Lipid Conjugates (SLiC) species. The synthesis route for each of the five molecules are listed with the specific liposome chain, such as, Ri, R2, R3, R4 and R5, conjugated at the location of RiH, R2H, R3H, R4H and R5H respectively. The structures of the five SLiC species are illustrated with a spermine head and two lipid legs.
Figure 6. Target Gene Silencing by SLiC Liposome-Mediated siRNA Delivery In Vivo. TM4-packaged siRNA specific to cyclophilin-B was selected for being tested in a Balb/c mouse model through a respiratory route of delivery. In addition to Blank control and empty TM4 control, a HKP package cyclophilin-B siRNA was used as a positive control. Three different dosage: 25, 40 and 50 μg were tested. Both 40 and 50 μg siRNA dosages achieved significant target gene silencing (N=3, *P<0.05).
Figure 7. Evaluation of the cytokine response in the mouse lung after HKP-siRNA nanoparticles delivery. HKP-siRNA at different dosages were oraltracheally administrated in the mouse lungs. The total lung tissue was harvested for protein isolation and cytokine measurements by ELIS A assay.
Figure 8. A. Standard curve to measure IFN-a concentration was prepared according to in-house SOP (Lowry Method); B. IFN-a concentration in each sample was determined and normalized to total protein.
Figure 9. GFP mRNA delivery to HeLa or HepG2 cells with DDS or Lipofectamine
MessengerMAX reagent (ThermoFisher). Cells were plated into wells of a 24-well plates at a density of 5xl04cells per well in DMEM supplemented with 10% FBS. On the next day transfection complexes were prepared as follows: GFP mRNA (0.5ug) was diluted with 25ul of OPTI-MEM and added to DDS (2ug) or LipofectamineMessengerMAX reagent diluted with 25ul OPTI-MEM. Mixtures were incubated for 30min (for mGFP /DDS) or lOmin (mGFP/LipofectamineMessengerMAX) and then added to the cells. Fluorescent images were taken 24h after transfection using a Zeiss Axiovert inverted fluorescence microscope.
DESCRIPTION OF THE INVENTION
The present invention provides RN A pharmaceutical formulations comprising either siRNA or mRNA sequences for the prevention and treatment of Zika virus infection. One formulation comprises siRNA sequences targeting the viral genome RNA and mRNA for knocking down the viral activity, and a carrier, such as Histidine-Lysine Co-polymers (HKP) or Spermine-liposome Conjugates (SLiC). Another formulation comprises mRNA sequences coding viral proteins critical for Zika viral infection and replication, and a carrier serving as an adjuvant for amplifying an immune response against the Zika virus within the host. The invention also provides methods of use for the pharmaceutical formulations for the prevention and therapeutic treatment of Zika virus Infection. siRNA Molecules
As used herein, an "siRNA molecule" or an "siRNA duplex" is a duplex
oligonucleotide, that is a short, double-stranded polynucleotide, that interferes with the expression of a gene in a cell, after the molecule is introduced into the cell, or interferes with the expression of a viral gene. For example, it targets and binds to a complementary nucleotide sequence in a single stranded (ss) target RNA molecule. siRNA molecules are chemically synthesized or otherwise constructed by techniques known to those skilled in the art. Such techniques are described in U.S. Pat. Nos. 5, 898,031, 6,107,094, 6,506,559, 7,056,704 and in European Pat. Nos. 1214945 and 1230375, which are incorporated herein by reference in their entireties. By convention in the field, when an siRNA molecule is identified by a particular nucleotide sequence, the sequence refers to the sense strand of the duplex molecule.
One or more of the ribonucleotides comprising the molecule can be chemically modified by techniques known in the art. In addition to being modified at the level of one or more of its individual nucleotides, the backbone of the oligonucleotide can be modified.
Additional modifications include the use of small molecules (e.g. sugar molecules), amino acids, peptides, cholesterol, and other large molecules for conjugation onto the siRNA molecule.
The siRNA molecules of the invention target a conserved region of the genome of a ZIKV. As used herein, "target" or "targets" means that the molecule binds to a
complementary nucleotide sequence in a ZIKV gene, which is an RNA molecule, or it binds to mRNA produced by the gene. This inhibits or silences the expression of the viral gene and/or its replication. As used herein, a "conserved region" of a ZIKV gene is a nucleotide sequence that is found in more than one strain of the virus, is identical among the strains, rarely mutates, and is critical for viral infection and/or replication and/or release from the infected cell.
In one embodiment, the targeted conserved region of the genome comprises sequences coding for envelope protein or nonstructural proteins NS1, NS3, NS4B, or NS5. Thus, the siRNA molecule can target envelope gene expression, NS1 gene expression, NS3 gene expression, NS4B gene expression, or NS5 gene expression.
In another embodiment, the targeted conserved region of the genome is the 3' untranslated region (3'-UTR) of the virus. Thus, the siRNA molecule can target the
3'-UTR.
In one embodiment, the siRNA molecule is a double-stranded oligonucleotide with a length of about 19 to about 29 base pairs. In one aspect of this embodiment, the molecule is a double-stranded oligonucleotide with a length of 19 to 21 base pairs. In another aspect of this embodiment, it is a double-stranded oligonucleotide with a length of 25 base pairs. In all of these aspects, the molecule may have blunt ends at both ends, or sticky ends with overhangs at both ends (unpaired bases extending beyond the main strand), or a blunt end at one end and a sticky end at the other. In one particular aspect, it has blunt ends at both ends. In another particular aspect, the molecule has a length of 25 base pairs (25 mer) and has blunt ends at both ends. In another particular aspect, the siRNA molecule is one of those identified in Table 4.
The siRNA molecules of the invention also include ones derived from those listed in Table 4 or otherwise disclosed herein. The derived molecules can have less than the 25 base pairs shown for each molecule, down to 17 base pairs, so long as the "core" contiguous base
pairs remain. That is, once given the specific sequences shown herein, a person skilled in the art can synthesize molecules that, in effect, "remove" one or more base pairs from either or both ends in any order, leaving the remaining contiguous base pairs, creating shorter molecules that are 24, 23, 22, 21, 20, 19, 18, or 17 base pairs in length, if starting with the 25 base pair molecule. For example, the derived molecules of the 25 mer molecules disclosed in Table 4 includes: a) 24 contiguous base pairs of any one or more of the molecules; b) 23 contiguous base pairs of any one or more of the molecules; c) 22 contiguous base pairs of any one or more of the molecules; b) 21 contiguous base pairs of any one or more of the molecules; d) 20 contiguous base pairs of any one or more of the molecules; e) 19 contiguous base pairs of any one or more of the molecules; f) 18 contiguous base pairs of any one or more of the molecules; and g) 17 contiguous base pairs of any one or more of the molecules. It is not expected that molecules shorter than 17 base pairs would have sufficient activity or sufficiently low off-target effects to be pharmaceutically useful; however, if any such constructs did, they would be equivalents within the scope of this invention. Alternatively, the derived molecules can have more than the 25 base pairs shown for each molecule, so long as the initial 25 contiguous base pairs remain. That is, once given the specific sequences disclosed herein, a person skilled in the art can synthesize molecules that, in effect, "add" one or more base pairs to either or both ends in any order, creating molecules that are 26 or more base pairs in length and containing the original 25 contiguous base pairs.
Pharmaceutical Compositions Comprising One or More siRNA Molecules
The invention includes a pharmaceutical composition comprising an siRNA molecule that targets a conserved region of the genome of a ZIKV and a pharmaceutically acceptable carrier. In one embodiment, the carrier condenses the molecules to form a nanoparticle. Alternatively, the composition may be formulated into nanoparticles. The compositions may be lyophilized into a dry powder. In one particular embodiment, the pharmaceutically acceptable carrier comprises a polymeric nanoparticle or a liposomal nanoparticle.
In one embodiment, the composition comprises at least two different siRNA molecules that target one or more conserved regions of the genome of a ZIKV and a pharmaceutically acceptable carrier. Such compositions are sometimes referred to herein as a "cocktail." In one aspect of this embodiment, the gene sequences in the conserved regions of the ZIKV are critical for the viral infection of a mammal. In another aspect of this embodiment, the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey). In still another aspect, the mammal is a human.
The composition can include one or more additional siRNA molecules that target still other conserved regions of the ZIKV genome. In one embodiment, the composition comprises siRNA molecules that target the
Envelope (E) gene of the ZIKV genome. In one aspect of this embodiment, the molecules are selected from the group consisting of:
ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU ,
ZIKV14: CCUUGACAAGCAAUCAGACACUCAA , and ZIKV17: CCGGAACUCCACACUGGAACAACAA .
In one embodiment, the composition comprises siRNA molecules that target the NSl gene of the ZIKV genome. In one aspect of this embodiment, the molecule comprises ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA.
In one embodiment, the composition comprises siRNA molecules that target the NS3 gene of the ZIKV genome. In one aspect of this embodiment, the molecules are selected from the group consisting of:
ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA ,
ZIKV63 : CCUAUCAGGUUGCAUCUGCCGGAAU ,
ZIKV62A: CCUAUCAAGUAGCAUCUGCCGGAAU , and ZIKV62B: GCCUAUCAAGUAGCAUCUGCCGGAA .
In one embodiment, the composition comprises siRNA molecules that target the NS4B gene of the ZIKV genome. In one aspect of this embodiment, the molecule comprises ZIKV74: CCACUUCAUACAACAACUACUCCUU.
In one embodiment, the composition comprises siRNA molecules that target the NS5 gene of the ZIKV genome. In one aspect of this embodiment, the molecule comprises ZIKV103 : GGUGCGCAGGAUCAUAGGUGAUGAA.
In one embodiment, the composition comprises siRNA molecules that target the 3'-UTR region of the ZIKV genome. In one aspect of this embodiment, the molecule comprises: ZIKV105: C C GAG A AC GC C AUGGC AC GG A AG A A .
In another embodiment, the composition comprises a cocktail, MSTZIKV13, wherein a first siRNA molecule comprises ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU and a second siRNA molecule comprises ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA.
In another embodiment, the composition comprises a cocktail, MSTZIKV62, wherein a first siRNA molecule comprises ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA and a second siRNA molecule comprises ZIKV74: CCACUUCAUACAACAACUACUCCUU.
In another embodiment, the composition comprises a cocktail, MSTZIKV62B, wherein a first siRNA molecule comprises ZIKV62B:
GC CU AUC A AGU AGC AUCUGCC GGA A and a second siRNA molecule comprises ZIKV17: CCGGAACUCCACACUGGAACAACAA.
In another embodiment, the composition comprises a cocktail, MSTZIKV103, wherein a first siRNA molecule comprises ZIKV103 :
GGUGCGCAGGAUCAUAGGUGAUGAA, a second siRNA molecule comprises KIKV63 : CCUAUCAGGUUGCAUCUGCCGGAAU, and a third siRNA molecule comprises ZIKVl 05 : CCGAGAACGCCAUGGC ACGGAAGAA.
In one aspect of all of these embodiments, a pharmaceutically acceptable carrier comprises a polymeric nanoparticle or a liposomal nanoparticle.
In one aspect of all of these embodiments, the siRNA molecules comprise 25 mer blunt-end siRNA molecules and the carrier comprises a Histidine-Lysine copolymer or Spermine-Lipid Conjugate (SLiC) and cholesterol.
Pharmaceutically Acceptable Carriers for the siRNA Molecules Pharmaceutically acceptable carriers include saline, sugars, polypeptides, polymers, lipids, creams, gels, micelle materials, and metal nanoparticles. In one embodiment, the carrier comprises at least one of the following: a glucose solution, a polycationic binding agent, a cationic lipid, a cationic micelle, a cationic polypeptide, a hydrophilic polymer grafted polymer, a non-natural cationic polymer, a cationic polyacetal, a hydrophilic polymer grafted polyacetal, a ligand functionalized cationic polymer, a ligand
functionalized-hydrophilic polymer grafted polymer, and a ligand functionalized liposome. In another embodiment, the polymers comprise a biodegradable histidine-lysine polymer, a biodegradable polyester, such as poly(lactic acid) (PLA), poly(glycolic acid) (PGA), and poly(lactic-co-glycolic acid) (PLGA), a polyamidoamine (PAMAM) dendrimer, a cationic lipid, or a PEGylated PEL Cationic lipids include DOTAP, DOPE, DC-Chol/DOPE, DOTMA, and DOTMA/DOPE.
In one embodiment, the carrier is a polymer. In one aspect of this embodiment, the polymer comprises a histidine-lysine copolymer (HKP). Such copolymers are described in U.S. Pat. Nos. 7,070,807 B2, 7,163,695 B2, and 7,772,201 B2, which are incorporated herein by reference in their entireties. In one aspect of this embodiment, the HKP forms a nanoparticle with the siRNA molecule, wherein the diameter of the nanoparticle is about lOOnm to about 400 nm. In another aspect of this embodiment, the HKP comprises the structure (R)K(R)-K(R)-(R)K(X), where R =KHHHKHHHKHHHKHHHK, K = lysine, and H = histidine. In another aspect, the HKP and the siRNA molecules self-assemble into nanoparticles or can be formulated into nanoparticles.
In another embodiment, the carrier is a liposome. In one aspect of this embodiment, the liposome comprises a cationic lipid conjugated with cholesterol. In a further aspect, the
cationic lipid comprises a spermine head and one or two oleyl alcoholic tails. In a further aspect, the liposome comprises a Spermine-Liposome-Conjugate (SLiC) and cholesterol. Examples of such molecules are disclosed in Figure 5. In another aspect, the liposome and the siRNA molecules self-assemble into nanoparticles or can be formulated into nanoparticles. Methods of Using the Pharmaceutical Compositions
The invention also includes methods of using the siRNA molecules and
pharmaceutical compositions containing them to prevent or treat ZIKV disease. As used herein "treat" or "treatment" refers to reducing the severity of or curing ZIKV disease. A therapeutically effective amount of the composition of the invention is administered to a mammal. In one embodiment, the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey). In one aspect of this embodiment, the mammal is a laboratory animal, such as a rodent. In another aspect of this embodiment, the mammal is a non-human primate, such as a monkey. In still another aspect of this embodiment, the mammal is a human. As used herein, a "therapeutically effective amount" is an amount that prevents, reduces the severity of, or cures ZIKV disease. Such amounts are determinable by persons skilled in the art, given the teachings contained herein. In one embodiment, a therapeutically effective amount of the pharmaceutical composition administered to a human comprises about 1 mg of the siRNA molecules per kilogram of body weight of the human to about 5 mg of the siRNA molecules per kilogram of body weight of the human.
Routes of administration are also determinable by persons skilled in the art, given the teachings contained herein. Such routes include intranasal administration and airway instillation, such as through use of an airway nebulizer. Such routes also include injection instillation and intraperitoneal, intravenous, intradermal, intravaginal, and subcutaneous administration. mRNA Vaccines
The invention provides a vaccine comprising an mRNA molecule that codes for an amino acid sequence encoded by a conserved region of the genome of a ZIKV and a pharmaceutically acceptable carrier comprising a polymer or a liposome. As used herein, a "conserved region" of a ZIKV gene is a nucleotide sequence that is found in more than one strain of the virus, is identical among the strains, rarely mutates, and is critical for viral infection and/or replication and/or release from the infected cell. In one embodiment, the gene sequence in the conserved region of the ZIKV genome is critical for the viral infection of a mammal. In another embodiment, the conserved region of the genome comprises gene sequences coding for the Envelope protein of ZIKV. In one aspect of this embodiment, the gene sequences code for amino acid sequences within Domain III of the Envelope protein.
In one embodiment, the polymer comprises a Histidine-Lysine co-polymer (HKP). In one aspect of this embodiment, the HKP and the mRNA molecules self-assemble into nanoparticles. In another aspect of this embodiment, the HKP and mRNA molecules are formulated into nanoparticles. In one embodiment, the liposome comprises a Spermine-Lipid Conjugate (SLiC) and cholesterol. In one aspect of this embodiment, the SLiC and cholesterol and the siRNA self-assemble into nanoparticles. In another aspect of this embodiment, the SLiC and cholesterol and the mRNA molecules are formulated into nanoparticles. The SLiC and cholesterol also acts as an adjuvant for amplifying the immune response. Use of the mRNA Vaccines
The mRNA vaccines of the invention are used to prevent a ZIKV infection or reduce its severity. Thus, the invention includes method of preventing or reducing the severity of a ZIKV infection in a mammal comprising administering to the mammal a therapeutically effective amount of the vaccine prior to infection. The vaccine is administered to the mammal through injection instillation or intradermal, intravenous, intraperitoneal, intravenous, intravaginal, or subcutaneous administration. In one embodiment, the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a
monkey). In one aspect of this embodiment, the mammal is a laboratory animal, such as a rodent. In another aspect of this embodiment, the mammal is a non-human primate, such as a monkey. In still another aspect of this embodiment, the mammal is a human.
The following examples illustrate certain aspects of the invention and should not be construed as limiting the scope thereof.
Example 1: Design mRNA targeting the envelope protein of ZIKV
Amongst all ZIKV proteins, E protein represents a major target for development of a neutralizing antibody. The ZIKV E protein contains three distinct domains (shown in Figure 1). We selected the sequence of Envelope Domain III of the virus to be the immunogenic regions of mRNA vaccine (Table 1). We used BLAST searches for identifying a region within
Domain III that should provide the necessary immune response without inducing Antibody Dependent Enhancement (ADE) in response to other flaviviruses (e.g. DENV; Supplement 1). To avoid ADE, we gave up Envelope Domain II as this region shares high homology with the regions in common with DENV (Supplement 1). Example 2: mRNA transfection into cells and measurement of mRNA expression in vitro.
The mRNA constructs containing Envelope Domain II of ZIKV will be transfected into human cells in vitro using a variety of commercially available transfection agent. Cells to be used for these studies included HEK293T, VERO cells, A549 cells and others. We will also examine electroporation (using MaxCyte technology) as an option for delivery. The various delivery processes are aimed at determining which will give good uptake into a variety of cells and to see subsequent expression of the construct. The aim is to determine protein production by each construct and also to determine whether the product is secreted from the cells. This process is not necessarily identified a clinically viable delivery process. mRNA will be detected in live cells using SmartFlare probes (Millipore) or through use of QRTPCR. mRNA of GFP was taken as the positive control (Figure 9).
Example 3: Detection of mRNA uptaking into cells using SmartFlare technology
These smart flares are beads that have a sequence attached that, when recognizing the RNA sequence in the cell, produce an increase in fluorescence. Smartflares will be designed against several regions along the constructs in case steric hindrance reduces signal from one region.
Example 4: Detection of the protein expression in the culture media
The protein expressed by the mRNA construct will be identified and quantitated by RPHPLC using an analytical C18 column (250mm x 2.1mm; Phenomenex). Protein detection will use a dual wavelength detector. A gradient of 0.1%TFA/Acetonitrile will be adjusted over time to allow analytical separation of protein peaks. In initial experiments, fractions will be collected and submitted for Mass Spectrometry to determine the presence of the expected sequence. The secreted product and the product manufactured within the cells will be compared using protein sequencing. To mitigate enzyme degradation of the sample, we will use enzyme inhibitors in the media and concentrated media from multiple wells in order to detect the product on HPLC. Example 5: Examination of siRNAs against Zika in a CPE assay
Specific siRNAs for testing will be provided to Immuquest (Frederick, MD) for analysis in their Zika CPE assay. We will first optimize siRNA delivery into the cells to be used for this study (Vero cells) by comparing multiple commercial and proprietary delivery agents (including lipofectamine, Oligofectamine, Spermine and PEG-PEI). These experiments will use siRNAs against housekeeping genes combined with qRTPCR measurements to show degree of silencing with each formulation. The optimal formulation will then be used to examine efficacy of the designed siRNAs against Zika in a CPE assay. In this experiment, control and test siRNAs will be administered at select intervals ahead of the virus and the degree of effect monitored by examining the change in the CPE value. We will provide test and control siRNAs for delivery at varying doses in the same delivery vehicle. These materials will be supplied as blinded samples.
Example 6: Determine best nanoparticle for delivery
We have developed several different varieties of nanoparticle for mRNA/siRNA delivery. These range from branched polypeptides (e.g. Histidine Lysine Ploymer or HKP) and various derivatives that, like the HKP, have been modified with targeting ligands to allow tissue specific delivery or PEGylated varieties that should assist in uptake across the mucosa of the vagina. We have demonstrated delivery topically to open wounds, and systemically to various tumors as well as specific targeted delivery to the lung and liver (avoiding uptake by Kupffer cells) using these formulations. We also have developed a spermine/spermidine co-polymer carrier and a liposomal delivery agent that, like the HKP, protect the siRNA and provide high efficiency delivery to tissues. Evaluate mRNA/siRNA-nanoparticle biological activity in cell culture. The various peptide/lipid formulations will be evaluated for their ability to form nanoparticles with single siRNAs or combinations of siRNAs against different targets. Binding with the nucleic acid will be evaluated by gel electrophoresis. Nanoparticle formation will be studied by particle size measurements using DLS (Dynamic Light Scattering) and the charge/size distribution measured using a nanoparticle size/charge instrument (Malvern instruments D9000). Morphology/size distribution of the nanoparticles will be confirmed by electron microscopy (TEM). The biological activity of these molecules will be evaluated in cell culture prior to in vivo studies. Particles formed at different N/P (Nitrogen/Phosphate) ratios will be tested in 2 different cell lines for their ability to deliver fluorescently labeled siRNA.
We will examine the ability of HKP and the other nanoparticles to deliver fluorescently labeled mRNA or siRNAs into the tissues of AG129 mice. We will then demonstrate efficacy by silencing housekeeping genes such as GAPDH in these local cells. These experiments will determine depth of penetration of the mRNA or siRNAs into the surrounding tissues. Studies to characterize the delivery vehicle will include 1) biophysical properties; 2) in vitro efficiency of mRNA and siRNA delivery; 3) delivery efficiency and toxicity in mice; and 4) the efficacy of siRNA-mediated gene silencing in Zika infected mice.
Example 7: SLiC/mRNA/siR A nanoparticle SLiC Liposome Preparation
Regular methods will be tried at first to prepare liposomes with newly synthesized SLiC molecules, such as thin film method, solvent injection and so on without much success. Norbert Maurer et al reported a method of liposome preparation in which siRNA or oligonucleotide solution will be slowly added under vortexing to the 50% ethanol solution (v/v) of liposome and ethanol was later removed by dialysis. The nanoparticles thus derived will be small in size and homogeneous. In this method mRNA and siRNA will be directly wrapped by cationic lipids during formation of liposome, while in most other methods mRNA or siRNA are loaded (or entrapped) into preformed liposome, such as Lipofectamine 2000. Lipids dissolved in ethanol are in so-called metastable state in which liposomes are not very stable and tend to aggregate. We will then prepare un-loaded or pre-formed liposomes using modified Norbert Maurer's method. (Maurer, N., A., Mori, L., Palmer, M. A., Monck, K. W. C, Mok, B., Mui, Q. F., Akhong, and P. R., Cullis. 1999. Lipid-based systems for the intracellular delivery of genetic drugs. Mol. Membr. Biol. 16, 129-140, incorporated herein by reference in its entirety.) We found that stable liposome solution could be made by simply diluting ethanol to the final concentration of 12.5% (v/v).
Liposomes were prepared by addition of lipids (cationic SLiC /cholesterol, 50:50, mol %) dissolved in ethanol to sterile dd-LbO. The ethanolic lipid solution need to be added slowly under rapid mixing. Slow addition of ethanol and rapid mixing were critical for the success in making
SLiC liposomes, as the process allows formation of small and more homogeneous liposomes. Unlike conventional methods in which mRNA or siRNAs are loaded during the process of liposome formulation and ethanol or other solvent is removed at end of manufacturing, our SLiC liposomes were formulated with remaining ethanol still in the solution so that liposomes were thought to be still in metastable state. When mRNA or siRNA solution was mixed/loaded with liposome solution cationic groups lipids will interact with anionic siRNA and condense to form core. SLiC liposomes' metastable state helped or facilitated liposome structure transformation to entrap mRNA or siRNA more effectively. Because of the
entrapment of mRNA/siRNA, SLiC liposomes become more compact and homogeneous.
Physiochemical Characterization of SLiC Liposome
After the liposome formation, we have developed an array of assays to characterize the physicochemical properties of SLiC liposome, including particle size, surface potential, morphology study, mRNA or siRNA loading efficiency and biological activity, etc. The particle size and zeta-potentials of SLiC liposomes were measured with Nano ZS Zeta Sizer (Malvern Instruments, UK). Each new SLiC liposome was tested for particle size and zeta-potential when ethanol contents changed from 50% to 25% and to 12.5%. Data were derived from formulations of different ethanol contents. All SLiC liposomes were prepared at lmg/ml in concentration and loaded with siRNA (2: 1, w/w). Each of SLiC Liposomes was composed of cationic SLiC and cholesterol dissolved in ethanol at 12.5%, e.g. TM2 (12.5). The average particle sizes of three sequential measurements and the average zeta-potentials of three sequential measurements were illustrated in Table 5.
Further analysis of the physiochemical perimeters of the SLiC liposome suggested that ethanol concentrations were positively proportional to particle sizes (the lower of ethanol concentration, the smaller of particle sizes), but negatively proportional to zeta-potential (the lower of ethanol concentration, the higher of zeta-potential at the same time). The higher surface potential will render particles more stable in solution. In addition to stability in solution, data shown later also indicated that toxicity was lower with lower ethanol concentration, too. Therefore, to put all factors together, ethanol concentration of 12.5% (v/v) was selected as solvent to suspend cholesterol as well as SLiC into the master working stock solution before they were used to make liposome formulations.
In contrast to bare SLiC liposome formulation, liposomes particle sizes became much smaller when they were loaded with mRNA or siRNA at 2: 1 (w/w) resulting in particle sizes in the range of 110 to 190nm in diameter and much lower PDI values. Conventional consideration of liposomal structure dictates that mRNA or siRNA is loaded or interacted with cationic lipids through electrostatic forces and liposomes wraps mRNA or siRNA to
form spherical particles in shape in order to reduce surface tension. As the result, the liposomes particle sizes became much smaller after loaded with mRNA or siRNA. Liposomes formulated with mRNA or siRNA also have lower surface charge, which could be explained by neutralizing effect from loaded mRNA or siRNA. Example 8: Mouse model study for mRNA vaccine
To investigate the protective efficiency of mRNA vaccine against ZIKV infection in vivo, AG129 mice will be used as the animal model. We will perform all mouse studies in Biosafety level-2 conditions.
All AG129 mice will be five-week old at the beginning of this study. 20 mice of prophylactic group will be intravenously injected with mRNA combination encapsidated with HKP-SLiC nanoparticle system from the tail vein. The other 20 mice of control group will be injected with PBS. 14 days later, the serum of all mice will be collected, and the mice from both prophylactic group and control group will be intravenously injected with ZIKV.
All mice will be weighed and the survival of each group will be counted daily. The serum of infected mice will be taken at 1, 3, 5, 7, 9 and 14-day post-infection. The tumor necrosis factor alpha (TNF-a) will be detected using enzyme-linked immunosorbent assay (ELISA). The tissues including testes, spleen, liver, heart, brain and kidney will be collected at 24 and 72 h post-infection. The total RNA from the tissues will be extracted, one-step quantitative real-time PCR and 5'-RACE assay will be performed as described in the above in vitro study part. The viral titers in the sera and tissues will be detected in Vero cells with serially dilution. The results from vaccination group will be compared with the control groups to evaluate the protective efficiency of the mRNA vaccine candidates.
Example 9: Design of siRNAs against ZIKV infection
By performing recursive analyses of siRNAs against 28 sequenced Zika strains (including both African and Asian lineages) we identified siRNAs that were able to cover an extensive number of strains (Table 2). Furthermore, careful selection of multiple siRNAs can be seen to provide complete coverage against all strains used in our predictions (Table 3).
Further sequence analysis showed that the 11 elected anti-ZIKV siRNA target the highly homologous regions of Envelope (E) protein, non-structure protein NS1, NS3, NS4B, NS5 and 3'-UTR in all ZIKV strains (Table 4 and Figure 1). If the mRNA translation and genome replication of ZIKV mimic DENV and take place in the cytoplasm, the siRNA candidates should work efficiently to degrade both positive and negative strand of the viral RNAs in ZIKV.
Example 10: Cell Culture Based Screening for Potent Anti-ZIKV siRNA Oligos siRNAs will be tested for their silencing activities using a reporter assay where the regions of the ZIKV genome to be silenced are incorporated into an expression vector. An appropriate ZIKV gene segment will be cloned into psiCheck™-2 (Promega). The cells will be co-transfected with reporter plasmids and siRNAs and the expression of renilla luciferase will be normalized to firefly luciferase expression (transfection efficiency control). Using RTPCR we will examine the ability of the siRNAs (both individually and in combinations) to silence their respective sequences of interest by transfecting in the plasmid containing the reporter construct, administering the siRNA and monitoring the reporter output at 24, 48 and 72h post transfection. Specifically, Vero cells will be transfected with single siRNA at 20 nM and infected (5 hours post transfection) with a strain of Zika. At T= 24 or 48hrs post infection, global expression profiling of the viral genome and viral titering will be performed using standard protocols (RT-PCR). The siRNAs with high activity will be further validated by performing a dose response of the siRNAs.
Example 11: Identify potent combinations of siRNAs to improve strain coverage.
Using similar experimental conditions to those used in Example 10, we will supply siRNA mixtures for evaluation in the CPE assay. We will selecte the siRNAs to include in the mixtures based on their efficacy as well as the degree of overlap between Zika strains.
Essentially, we believe that testing the combinations shown in Table 1 will allow identification of suitable combinations for subsequent in vivo tests based on potency in the CPE assay and/or strain coverage (for equipotent combinations strain coverage will be a selection criteria). We
will identify either single siRNAs or combinations that demonstrate the greatest efficacy in silencing and inhibiting the viral genes and which produce the greatest effect on ZIKV in the CPE assay.
Example 12: Develop and characterize nanoparticles for in vivo delivery of siRNAs. The most critical and challenging aspect of developing an siRNA-based therapeutic is the delivery of the siRNA into the cells where ZIKV infection can be transmitted. Since ZIKV has been shown to be transmitted by sexual intercourse we have elected to pursue intravaginal siRNA delivery as a means of validating this as a preliminary approach to therapy. Once more is understood about the etiology of the disease we may be able to adjust the delivery approach to treat therapeutically. The vaginal tract is a suitable site for the administration of both local and systemic acting drugs. There are numerous vaginal products on the market such as those approved for contraception, treatment of yeast infection, hormonal replacement therapy, and feminine hygiene. However, there are many biological barriers and factors that protect the vagina from foreign particles, such as a thick and elastic mucus layer that may bind and inhibit access of these agents.
One of the biggest barriers to successful siRNA delivery is the mucus layer [12]. Cross-linked mucin fibers limit the size and hydrophobicity of reagents that can penetrate it. In regards to optimal nanoparticle size for siRNA delivery the upper limit for effective access and uptake is 0.5um and the lower limit appears to be 0.2um [12, 13]. However, despite these reported limitations, others have successfully demonstrated siRNA delivery intravaginally [14, 15] using other carriers. We have several nanoparticles that are below this size - including the branched peptide Histidine Lysine copolymer (HKP) that has shown excellent delivery characteristics to wounds and systemically. This will allow exploration of other modifications e.g. PLGA, PEG or PEG-PEI to this HKP nanoparticle or evaluation of a spermine/spermidine copolymer that exhibits suitable molecular characteristics for delivery including size, charge and hydrophobicity. Previous studies have shown significant success with PLGA, PEG and other modifications to existing nanoparticle formulations and we have
demonstrated functionalization of our nanoparticles with these moieties while retaining this optimal size, ability to carry and protect the siRNAs while penetrating the mucus layer. Finally, others have shown that spray dried powders can enhance intravaginal siRNA delivery [16]. We have also demonstrated that we can lyophilize our HKP nanoparticle complexed with siRNA without adversely affecting its characteristic size or delivery capacity.
Example 13: Mice model study for anti-ZIKV siRNA
To investigate the efficiency of siRNA combination against ZIKV virus infection in vivo, AG129 mice will be used as the animal model. We will perform all mouse studies in Biosafety level-2 conditions. All AG129 mice will be five-week old at the beginning of this study. 20 mice of treatment group will be intravenously injected with siRNA combination encapsidated with HKP-SLiC nanoparticle system from the tail vein. The other 20 mice of control group will be injected with PBS. 24 h later, mice from treatment group and control group will be infected intravenously with ZIKV, respectively. siRNA or PBS will be intravenously injected into the treatment or control group at 0, 24, 48 and 72 h post-infection.
All mice will be weighed and the survival of each group was counted daily. The serum of infected mice will be taken at 1, 3, 5, 7, 9 and 14-day post-infection. The tumor necrosis factor alpha (TNF-a) will be detected using enzyme-linked immunosorbent assay (ELISA). The studies are expected to demonstrate that the cell cultures infected with ZIKV exhibit up-regulated expression of tumor necrosis factor-a (TNF-a), and interleukin-ΐβ (IL-Ιβ). The tissues including testes, spleen, liver, heart, brain and kidney will be collected at 24, 28 and 72 h post-infection. The total RNA from the tissues were extracted, one-step quantitative real-time PCR and 5 '-RACE assay will be performed. The viral titers in the sera and tissues will be detected in Vero cells with serially dilution. The results from treatment groups will be compared with the control groups to evaluate the anti-viral efficiency of the therapeutic siRNA candidates.
Table 1. Characteristics of ZIKV mRNA vaccine candidate proteins.
Table 4. The list of sequence and targets for anti-ZIKV siRNA candidates
Table 5 Characterization indexes of five SLiC species and five SLiC-siRNA nanoparticles, including particle sizes, poly-dispersity index (PDI) and Zeta-potential.
Supplement 1: The Envelope gene amino acid sequence alignment results of ZIKV with all four serotypes of DENV
High homology was indicated in red frame, while low homology was indicated in green frame.
Dengue virus serotype 1
REFERENCES
1. CDC bulletin on Zika (htt :// w ww , cdc. gov/zi ka )
2. MMWR CDC on Zika and Microcephaly
3. Microcephaly in Brazil potentially linked to the Zika virus epidemic, ECDC assesses the risk. News and Media. European Centre for Disease Prevention and Control. 25 November 2015.
4. Zanluca C, Melo VC, Mosimann AL, Santos GI, Santos CN, Luz K. First report of autochthonous transmission of Zika virus in Brazil. Mem Inst Oswaldo Cruz.
2015; 110:569-72.
5. Dai L, Song J, Lu X, Deng YQ, Musyoki AM, Cheng H, Zhang Y, Yuan Y, Song H, Haywood J, Xiao H, Yan J, Shi Y, Qin CF, Qi J, Gao GF. Structures of the Zika Virus Envelope Protein and Its Complex with a Flavivirus Broadly Protective Antibody. Cell Host & Microbe 2016: 19, 696-704.
6. Hamel R, Dejarnac O, Wichit S, Ekchariyawat P, Neyret A, Luplertlop N,
Perera-Lecoin M, Surasombatpattana P, Talignani L, Thomas F, Cao-Lormeau VM, Choumet V, Briant L, Despres P, Amara A, Yssel H, Misse D. Biology of Zika Virus Infection in Human Skin Cells. Journal of Virology. 2015: 89(17):8880-96.
7. Ladner JT, Wiley MR, Prieto K, Yasuda CY, Nagle E, Kasper MR, Reyes D,
Vasilakis N, Heang V, Weaver SC, Haddow A, Tesh RB, Sovann L, Palacios G. Structures of the Zika Virus Envelope Protein and Its Complex with a Flavivirus Broadly Protective Antibody. Cell Host Microbe. 2016: 19(5):696-704.
8. Baronti C, Piorkowski G, Charrel RN, Boubis L, Leparc-Goffart I, de Lamballerie X.
Complete coding sequence of zika virus from a French polynesia outbreak in 2013. Genome Announc. 2014: 2(3). pii: e00500-14. doi: 10.1128/genomeA.00500-14.
9. Liang, H., Lee, M., and Jin, X. Guiding dengue vaccine development using
knowledge gained from the success of the yellow fever vaccine. Cellular & Molecular Immunology. 2015: 13, 36-46.
10. Petsch B, Schnee M, Vogel AB, Lange E, Hoffmann B, Voss D, Schlake T, Thess A, Kallen KJ, Stitz L, Kramps T. Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection. Nat Biotechnol. 2012:
30(12): 1210-6.
11. Charles AS, Christofferson RC. Utility of a Dengue-Derived Monoclonal Antibody to Enhance Zika Infection In Vitro. PLoS Curr. 2016 Jul 5;8.
12. Yang,S., Chen, Y.,Ahmadie, R., Ho, EA. Advancements in the Field of
IntravaginalsiRNA Delivery. Journal of Controlled Release. 2013 : 167; 29-39
13. Lai SK, O'Hanlon DE, Harrold S, Man ST, Wang YY, Cone R, Hanes J.. Rapid
transport of large polymeric particles in fresh undiluted human mucus. Proc Nat AcdSci. 2007: 104: 1482-1487.
14. Palliser D, Chowdhury D, Wang QY, Lee SJ, Bronson RT, Knipe DM, Lieberman J. An SiRNA-based microbicide protects mice from lethal Herpes Simplex 2 virus infection. Nature. 2006: 439; 89-94.
15. Woodrow KA, Cu Y, Booth CJ, Saucier- Sawyer JK, Wood MJ, Saltzman WM.
Intravaginal gene silencing using biodegradabalepolyer nanoparticles densely loaded with small interfering RNA. Nat Mater. 2009: 8; 526-533.
16. Wu N, Zhang X, Li F, Zhang T, Gan Y, Li J. Spray-dried powders enhance vaginal siRNAdelivery by potentially modulating the mucus molecular sieve structure.
International Journal of Nanomedicine. 2015: 10 5383-5396. The disclosures of all publications identified herein, including issued patents and published patent applications, and all database entries identified herein by url addresses or accession numbers are incorporated herein by reference in their entirety.
Although this invention has been described in relation to certain embodiments thereof, and many details have been set forth for purposes of illustration, it will be apparent to those skilled in the art that the invention is susceptible to additional embodiments and that certain of the details described herein may be varied considerably without departing from the basic principles of the invention.
Claims
1. A pharmaceutical composition comprising at least two different siRNA molecules that target one or more conserved regions of the genome of a Zika Virus (ZIKV) and a pharmaceutically acceptable carrier, wherein the pharmaceutically acceptable carrier comprises a polymer or a liposome and wherein the siRNA molecules and the carrier form a nanoparticle.
2. The composition of claim 1, wherein the gene sequences in the conserved regions of the ZIKV are critical for the viral infection of a mammal.
3. The composition of claim 2, wherein the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
4. The composition of any one of claims 1-3, wherein the targeted conserved regions of the genome comprise the 3'-UTR (untranslated region) and gene sequences coding for ZIKV proteins selected from the group consisting of Envelope (E) protein and non-structure proteins NS1, NS3, NS4B, and NS5.
5. The composition of claim 4, wherein the siRNA molecules are selected from the group consisting of:
ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU , ZIKV14: CCUUGACAAGCAAUCAGACACUCAA , ZIKV17: CCGGAACUCCACACUGGAACAACAA , ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA, ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA , ZIKV63 : CCUAUCAGGUUGCAUCUGCCGGAAU , ZIKV62A: CCUAUCAAGUAGCAUCUGCCGGAAU , ZIKV62B: GCCUAUCAAGUAGCAUCUGCCGGAA , ZIKV74: CCACUUCAUACAACAACUACUCCUU ,
ZIKV103 : GGUGCGCAGGAUCAUAGGUGAUGAA , and
ZIKVl 05 : CCGAGAACGCC AUGGCACGGAAGAA
6. The composition of claim 4, wherein the siRNA molecules that target the Envelope (E) gene are selected from the group consisting of:
ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU ,
ZIKV14: CCUUGACAAGCAAUCAGACACUCAA , and
ZIKV17: CCGGAACUCCACACUGGAACAACAA .
7. The composition of claim 4, wherein the siRNA molecule that targets the non-structure protein NS1 gene comprises:
ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA .
8. The composition of claim 4, wherein the siRNA molecules that target the non-structure protein NS3 gene are selected from the group consisting of:
ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA , ZIKV63 : CCUAUCAGGUUGCAUCUGCCGGAAU , ZIKV62A: CCUAUCAAGUAGCAUCUGCCGGAAU , and ZIKV62B: GCCUAUCAAGUAGCAUCUGCCGGAA .
9. The composition of claim 4, wherein the siRNA molecule that targets the non-structure protein NS4B gene comprises:
ZIKV74: CCACUUCAUACAACAACUACUCCUU .
10. The composition of claim 4, wherein the siRNA molecule that targets the non-structure protein NS5 gene comprises:
ZIKV103 : GGUGCGCAGGAUCAUAGGUGAUGAA .
11. The composition of claim 4, wherein the siRNA molecule that targets the 3 ' -UTR
comprises:
ZIKV105: CCGAGAACGCCAUGGCACGGAAGAA .
12. The composition of claim 1, comprising a siRNA cocktail, MSTzn v13, wherein a first siRNA molecule comprises ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU and a second siRNA molecule comprises ZIKV30:
GCCAUGGCACAGUGAAGAGCUUGAA.
13. The composition of claim 1, comprising a siRNA cocktail, MSTzn v62, wherein a first siRNA molecule comprises ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA and a second siRNA molecule comprises ZIKV74:
CCACUUCAUACAACAACUACUCCUU.
14. The composition of claim 1, comprising a siRNA cocktail, MSTzn v62B, wherein a first siRNA molecule comprises ZIKV62B: GCCUAUCAAGUAGCAUCUGCCGGAA and a second siRNA molecule comprises ZIKV17:
CCGGAACUCCACACUGGAACAACAA.
15. The composition of claim 1, comprising a siRNA cocktail, MSTzn v103, wherein a first siRNA molecule comprises ZIKV103 : GGUGC GC AGG AUC AU AGGUG AUGA A, a second siRNA molecule comprises KTKV63 :
CCUAUCAGGUUGCAUCUGCCGGAAU, and a third siRNA molecule comprises ZIKVl 05 : CCGAGAACGCC AUGGCACGGAAGAA.
16. The composition of any one of claims 1-3, wherein the polymer comprises a
Histidine-Lysine co-polymer (HKP).
17. The composition of claim 4, wherein the polymer comprises a Histidine-Lysine
co-polymer (HKP).
18. The composition of any one of claims 5-15, wherein the polymer comprises a
Histidine-Lysine co-polymer (HKP).
19. The composition of any one of claims 16-18, wherein the HKP comprises the structure (R)K(R)-K(R)-(R)K(X), where R =KHHHKHHHKHHHKHHHK, K = lysine, and H = histidine.
20. The composition of any one of claims 1-3, wherein the liposome comprises a
Spermine-Lipid Conjugate (SLiC) and cholesterol.
21. The composition of claim 4, wherein the liposome comprises a Spermine-Lipid
Conjugate (SLiC) and cholesterol.
22. The composition of any one of claims 5-15, wherein the liposome comprises a
Spermine-Lipid Conjugate (SLiC) and cholesterol.
23. The composition of any one of claims 20-22, wherein the SLiC comprises one of the structures TM1, TM2, TM3, TM4, or TM5 shown in Figure 5.
24. The composition of claim 23, wherein the SLiC comprises the structure TM4 shown in Figure 5.
25. The composition of any one of claims 1-24, wherein the siRNA molecules comprise oligonucleotides with a length of 19-29 base pairs.
26. The composition of any one of claims 1-24, wherein the siRNA molecules comprise oligonucleotides with a length of 19-21 base pairs.
27. The composition of any one of claims 1-24, wherein the siRNA molecules comprise oligonucleotides with a length of 25 base pairs.
28. A method of treating a mammal with a ZIKV infection comprising administering to said mammal a therapeutically effective amount of the composition of claim 1.
29. A method of treating a mammal with a ZIKV infection comprising administering to said mammal a therapeutically effective amount of the composition of any one of claims 2-27.
30. The method of claims 28 or 29, wherein the composition is administered to the mammal through injection instillation.
31. The method of claims 28 or 29, wherein the composition is administered to the mammal through intranasal administration.
32. The method of claims 28 or 29, wherein the composition is administered to the mammal through intradermal administration.
33. The method of claims 28 or 29, wherein the composition is administered to the mammal through intravenous administration.
34. The method of claims 28 or 29, wherein the composition is administered to the mammal through subcutaneous administration.
35. The method of claims 28 or 29, wherein the composition is administered to the mammal through intravaginal administration.
36. The method of any one of claims 28-35, wherein the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
37. The method of claim 36, wherein the mammal is a human.
38. An siRNA molecule that targets a conserved region of the genome of a ZIKV.
39. The siRNA molecule of claim 38, wherein the targeted conserved region of the genome comprises the 3'-UTR and gene sequences coding for ZIKV proteins selected from the group consisting of Envelope (E) protein, non-structure protein NS1, non-structure protein NS3, non-structure protein NS4B, and non-structure protein NS5.
40. The siRNA molecule of claim 38, wherein the wherein the siRNA molecules are
selected from the group consisting of:
ZIKV13 : GGUGAAGCCUACCUUGACAAGCAAU ,
ZIKV14: CCUUGACAAGCAAUCAGACACUCAA ,
ZIKV17: CCGGAACUCCACACUGGAACAACAA ,
ZIKV30: GCCAUGGCACAGUGAAGAGCUUGAA,
ZIKV62: GCCUAUCAGGUUGCAUCUGCCGGAA ,
ZIKV63 : CCUAUCAGGUUGCAUCUGCCGGAAU ,
ZIKV62A: CCUAUCAAGUAGCAUCUGCCGGAAU ,
ZIKV62B: GCCUAUCAAGUAGCAUCUGCCGGAA ,
ZIKV74: CCACUUCAUACAACAACUACUCCUU ,
ZIKV103 : GGUGCGCAGGAUCAUAGGUGAUGAA , and
ZIKV105: CCGAGAACGCCAUGGCACGGAAGAA .
41. The siRNA molecule of any one of claims 38-40, wherein the siRNA molecule
comprises oligonucleotides with a length of 19-29 base pairs.
42. The siRNA molecule of any one of claims 38-40, wherein the siRNA molecule
comprises oligonucleotides with a length of 19-21 base pairs.
43. The siRNA molecule of any one of claims 38-40, wherein the siRNA molecule
comprises oligonucleotides with a length of 25 base pairs.
44. The siRNA molecule of claim 40, wherein the siRNA molecules comprise derivatives of the identified siRNA molecules, the derivatives having 17-24 contiguous base pairs of original 25 contiguous base pairs of the identified molecules or one or more base pairs in addition to the original 25 contiguous base pairs of the identified molecules.
45. A composition comprising the siRNA molecule of any one of claims 38-44 and a pharmaceutically acceptable carrier, wherein the pharmaceutically acceptable carrier comprises a polymer or a liposome and wherein the siRNA molecules and the carrier form a nanoparticle.
46. A method of treating a mammal with a ZIKV infection comprising administering to said mammal a therapeutically effective amount of the composition of claim 45.
47. The method of claim 46, wherein the mammal is a human.
48. A pharmaceutical composition comprising an mRNA molecule that codes for an amino acid sequence encoded by a conserved region of the genome of a Zika Virus (ZIKV) and a pharmaceutically acceptable carrier comprising a polymer or a liposome.
49. The composition of claim 48, wherein the gene sequence in the conserved region of the ZIKV is critical for the viral infection of a mammal.
50. The composition of claim 49, wherein the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
51. The composition of claim 48, wherein the conserved region of the genome comprises gene sequences coding for the Envelope protein of ZIKV.
52. The composition of claim 51, wherein the gene sequences code for amino acid
sequences within Domain III of the Envelope protein.
53. The composition of any one of claims 48-52, wherein the polymer comprises a
Histidine-Lysine co-polymer (HKP).
54. The composition of any one of claims 48-52, wherein the liposome comprises a
Spermine-Lipid Conjugate (SLiC) and cholesterol.
55. The composition of any one of claims 48-53, wherein the HKP and mRNA molecules are formulated into nanoparticles.
56. The composition of any one of claims 48-51, wherein the SLiC and the mRNA
molecules are formulated into nanoparticles.
57. The composition of claim 53, wherein the HKP and the mRNA molecules self-assemble into nanoparticles.
58. The composition of claim 54, wherein the SLiC and cholesterol and the siRNA
self-assemble into nanoparticles.
59. A method of preventing or reducing the severity of a ZIKV infection in a mammal comprising administering to said mammal a therapeutically effective amount of the composition of claim 48 prior to infection.
60. A method of preventing or reducing the severity of a ZIKV infection in a mammal comprising administering to said mammal a therapeutically effective amount of the composition of any one of claim 49-58 prior to infection.
61. The method of claims 59 or 60, wherein the composition is administered to the mammal through injection instillation or intradermal, intravenous, intravaginal, or subcutaneous administration.
62. The method of any one of claims 59-61, wherein the mammal is a human, rodent (e.g., rat, mouse, or guinea pig), ferret, or non-human primate (e.g., a monkey).
63. The method of claim 62, wherein the mammal is a human.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201880013887.4A CN110520155A (en) | 2017-02-28 | 2018-02-28 | Prevent and treat the RNA pharmaceutical preparation of zika virus infection |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201762465096P | 2017-02-28 | 2017-02-28 | |
US62/465,096 | 2017-02-28 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018160690A1 true WO2018160690A1 (en) | 2018-09-07 |
Family
ID=63370993
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2018/020220 WO2018160690A1 (en) | 2017-02-28 | 2018-02-28 | Rna pharmaceutical formulations for prophylactic and therapeutic treatment of zika virus infection |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN110520155A (en) |
WO (1) | WO2018160690A1 (en) |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022152109A3 (en) * | 2021-01-14 | 2022-08-25 | Suzhou Abogen Biosciences Co., Ltd. | Lipid compounds and lipid nanoparticle compositions |
US11964052B2 (en) | 2021-05-24 | 2024-04-23 | Suzhou Abogen Biosciences Co., Ltd. | Lipid compounds and lipid nanoparticle compositions |
EP4100066A4 (en) * | 2020-02-07 | 2024-05-01 | Rnaimmune, Inc. | COMPOSITION AND PROCESS OF MRNA VACCINES AGAINST NEW CORONAVIRUS INFECTIONS |
AU2022207550B2 (en) * | 2021-01-14 | 2024-12-12 | Suzhou Abogen Biosciences Co., Ltd. | Lipid compounds and lipid nanoparticle compositions |
US12280105B2 (en) | 2021-08-11 | 2025-04-22 | RNAimmune, Inc. | Compositions and methods of mRNA vaccines against novel coronavirus infection |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111574633B (en) * | 2020-05-15 | 2022-04-08 | 清华大学 | Application of protein CbAE with broad-spectrum antiviral function |
CN114934050B (en) * | 2022-03-31 | 2023-08-22 | 暨南大学 | miRNA-2 for inhibiting Zika virus and application thereof |
CN114908091B (en) * | 2022-03-31 | 2023-08-22 | 暨南大学 | miRNA-1 for inhibiting Zika virus and application thereof |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2016061549A1 (en) * | 2014-10-17 | 2016-04-21 | Sirnaomics, Inc. | Compositions comprising small interfering rna molecules for prevention and treatment of ebola virus disease |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2017015463A2 (en) * | 2015-07-21 | 2017-01-26 | Modernatx, Inc. | Infectious disease vaccines |
-
2018
- 2018-02-28 WO PCT/US2018/020220 patent/WO2018160690A1/en active Application Filing
- 2018-02-28 CN CN201880013887.4A patent/CN110520155A/en not_active Withdrawn
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2016061549A1 (en) * | 2014-10-17 | 2016-04-21 | Sirnaomics, Inc. | Compositions comprising small interfering rna molecules for prevention and treatment of ebola virus disease |
Non-Patent Citations (3)
Title |
---|
KUMAR, N. ET AL.: "RNA Interference Study for Gene Silencing of Flaviviruses: A Computational Approach. Poster", SYMPOSIUM ON CURRENT TRENDS IN LIFE SCIENCE AT CENTRAL UNIVERSITY OF BIHAR, 2014, pages 1 * |
PARDI, N. ET AL.: "Zika Virus Protection by a Single Low-Dose Nucleoside-Modified mRNA ' Vaccination", NATURE, vol. 543, 2 February 2017 (2017-02-02), pages 248 - 251, XP055370679 * |
SHAWAN, M. ET AL.: "Design and Prediction of Potential RNAi (siRNA) Molecules for 3' UTR PTGS of Different Strains of Zika Virus: A Computational Approach", NATURE AND SCIENCE OF SLEEP, vol. 13, February 2015 (2015-02-01), pages 37 - 50, XP055326136 * |
Cited By (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP4100066A4 (en) * | 2020-02-07 | 2024-05-01 | Rnaimmune, Inc. | COMPOSITION AND PROCESS OF MRNA VACCINES AGAINST NEW CORONAVIRUS INFECTIONS |
US12029786B2 (en) | 2020-02-07 | 2024-07-09 | RNAimmune, Inc. | Composition and method of mRNA vaccines against novel coronavirus infection |
WO2022152109A3 (en) * | 2021-01-14 | 2022-08-25 | Suzhou Abogen Biosciences Co., Ltd. | Lipid compounds and lipid nanoparticle compositions |
AU2022207550B2 (en) * | 2021-01-14 | 2024-12-12 | Suzhou Abogen Biosciences Co., Ltd. | Lipid compounds and lipid nanoparticle compositions |
US11964052B2 (en) | 2021-05-24 | 2024-04-23 | Suzhou Abogen Biosciences Co., Ltd. | Lipid compounds and lipid nanoparticle compositions |
US12280105B2 (en) | 2021-08-11 | 2025-04-22 | RNAimmune, Inc. | Compositions and methods of mRNA vaccines against novel coronavirus infection |
Also Published As
Publication number | Publication date |
---|---|
CN110520155A (en) | 2019-11-29 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2018160690A1 (en) | Rna pharmaceutical formulations for prophylactic and therapeutic treatment of zika virus infection | |
US20220145301A1 (en) | siRNA/Nanoparticle Formulations for Treatment of Middle-East Respiratory Syndrome Coronaviral Infection | |
US12029786B2 (en) | Composition and method of mRNA vaccines against novel coronavirus infection | |
JP2023513436A (en) | RNAi Prophylactic Therapeutic Compositions and Methods for Treatment of Severe Acute Respiratory Infection Caused by 2019 Novel Coronavirus (2019-nCoV) | |
US9506063B2 (en) | SiRNA compositions and methods for treatment of HPV and other infections | |
Qin et al. | Retinoic acid inducible gene-I and melanoma differentiation-associated gene 5 are induced but not essential for dengue virus induced type I interferon response | |
US9695421B2 (en) | Dengue virus-specific siRNA, double helix oligo-RNA structure comprising siRNA, and composition for suppressing proliferation of dengue virus comprising RNA structure | |
WO2013012835A2 (en) | Sirna useful in the treatment of flavivirus infection | |
EP4183879A1 (en) | Double-stranded oligonucleotide and composition for treating covid-19 containing same | |
Wang et al. | Construction of exosome-loaded LL-37 and its protection against zika virus infection | |
CN114870026A (en) | Preparation method of ACE2siRNA medicine for targeting delivery of shRNA by ACE2 | |
US11981714B2 (en) | CCR5 and CD4 siRNA-targeted phospholipid nanosomes therapeutics for treatment of HIV-1 and other diseases | |
TWI414301B (en) | Treating picornavirus infection by targeting microrna mir-141 | |
WO2022247817A1 (en) | Nucleic acid-polypeptide nano-pharmaceutical composition for treating and preventing human papilloma virus infection | |
KR101239492B1 (en) | Polysorbitol-based osmotically active transporter and gene therapy using the same as a gene carrier | |
JP4672654B2 (en) | SiRNA against VP-1 of JC virus and pharmaceutical composition comprising the same | |
WO2023083315A1 (en) | Ncovsirna drug for targeted delivery of shrna by using rbd, synthesis method therefor and use thereof | |
CN118812667A (en) | Varicella-Zoster RNA Vaccine | |
CN102533823A (en) | Antiviral preparation with HSV (Herpes Simplex Virus) amplicon as carrier |
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: 18761493 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
32PN | Ep: public notification in the ep bulletin as address of the adressee cannot be established |
Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 03/12/2019) |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18761493 Country of ref document: EP Kind code of ref document: A1 |