Skip to main content
Top
Published in: Molecular Cancer 1/2021

01-12-2021 | COVID-19 | Review

mRNA vaccine: a potential therapeutic strategy

Authors: Yang Wang, Ziqi Zhang, Jingwen Luo, Xuejiao Han, Yuquan Wei, Xiawei Wei

Published in: Molecular Cancer | Issue 1/2021

Login to get access

Abstract

mRNA vaccines have tremendous potential to fight against cancer and viral diseases due to superiorities in safety, efficacy and industrial production. In recent decades, we have witnessed the development of different kinds of mRNAs by sequence optimization to overcome the disadvantage of excessive mRNA immunogenicity, instability and inefficiency. Based on the immunological study, mRNA vaccines are coupled with immunologic adjuvant and various delivery strategies. Except for sequence optimization, the assistance of mRNA-delivering strategies is another method to stabilize mRNAs and improve their efficacy. The understanding of increasing the antigen reactiveness gains insight into mRNA-induced innate immunity and adaptive immunity without antibody-dependent enhancement activity. Therefore, to address the problem, scientists further exploited carrier-based mRNA vaccines (lipid-based delivery, polymer-based delivery, peptide-based delivery, virus-like replicon particle and cationic nanoemulsion), naked mRNA vaccines and dendritic cells-based mRNA vaccines. The article will discuss the molecular biology of mRNA vaccines and underlying anti-virus and anti-tumor mechanisms, with an introduction of their immunological phenomena, delivery strategies, their importance on Corona Virus Disease 2019 (COVID-19) and related clinical trials against cancer and viral diseases. Finally, we will discuss the challenge of mRNA vaccines against bacterial and parasitic diseases.
Literature
1.
go back to reference Rabinovich NR, McInnes P, Klein DL, Hall BF. Vaccine technologies: view to the future. Science. 1994;265:1401–4.PubMedCrossRef Rabinovich NR, McInnes P, Klein DL, Hall BF. Vaccine technologies: view to the future. Science. 1994;265:1401–4.PubMedCrossRef
3.
go back to reference Malagón T, Drolet M, Boily MC, Franco EL, Jit M, Brisson J, Brisson M. Cross-protective efficacy of two human papillomavirus vaccines: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12:781–9.PubMedCrossRef Malagón T, Drolet M, Boily MC, Franco EL, Jit M, Brisson J, Brisson M. Cross-protective efficacy of two human papillomavirus vaccines: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12:781–9.PubMedCrossRef
5.
go back to reference Rodrigues CMC, Pinto MV, Sadarangani M, Plotkin SA. Whither vaccines? J Inf Secur. 2017;74(Suppl 1):S2–s9. Rodrigues CMC, Pinto MV, Sadarangani M, Plotkin SA. Whither vaccines? J Inf Secur. 2017;74(Suppl 1):S2–s9.
6.
go back to reference Pollard C, De Koker S, Saelens X, Vanham G, Grooten J. Challenges and advances towards the rational design of mRNA vaccines. Trends Mol Med. 2013;19:705–13.PubMedCrossRef Pollard C, De Koker S, Saelens X, Vanham G, Grooten J. Challenges and advances towards the rational design of mRNA vaccines. Trends Mol Med. 2013;19:705–13.PubMedCrossRef
7.
go back to reference Linares-Fernández S, Lacroix C, Exposito JY, Verrier B. Tailoring mRNA vaccine to balance innate/adaptive immune response. Trends Mol Med. 2020;26:311–23.PubMedCrossRef Linares-Fernández S, Lacroix C, Exposito JY, Verrier B. Tailoring mRNA vaccine to balance innate/adaptive immune response. Trends Mol Med. 2020;26:311–23.PubMedCrossRef
8.
go back to reference Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL. Direct gene transfer into mouse muscle in vivo. Science. 1990;247:1465–8.PubMedCrossRef Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL. Direct gene transfer into mouse muscle in vivo. Science. 1990;247:1465–8.PubMedCrossRef
9.
go back to reference Jirikowski GF, Sanna PP, Maciejewski-Lenoir D, Bloom FE. Reversal of diabetes insipidus in Brattleboro rats: intrahypothalamic injection of vasopressin mRNA. Science. 1992;255:996–8.PubMedCrossRef Jirikowski GF, Sanna PP, Maciejewski-Lenoir D, Bloom FE. Reversal of diabetes insipidus in Brattleboro rats: intrahypothalamic injection of vasopressin mRNA. Science. 1992;255:996–8.PubMedCrossRef
10.
go back to reference Pardi N, Hogan MJ, Weissman D. Recent advances in mRNA vaccine technology. Curr Opin Immunol. 2020;65:14–20.PubMedCrossRef Pardi N, Hogan MJ, Weissman D. Recent advances in mRNA vaccine technology. Curr Opin Immunol. 2020;65:14–20.PubMedCrossRef
11.
go back to reference Naik R, Peden K. Regulatory considerations on the development of mRNA vaccines. Curr Top Microbiol Immunol. 2020. Epub ahead of print. Naik R, Peden K. Regulatory considerations on the development of mRNA vaccines. Curr Top Microbiol Immunol. 2020. Epub ahead of print.
12.
go back to reference Rice AM, Morales AC, Ho AT, Mordstein C, Mühlhausen S, Watson S, Cano L, Young B, Kudla G, Hurst LD. Evidence for strong mutation bias towards, and selection against, U content in SARS-CoV-2: implications for vaccine design. Mol Biol Evol. 2020;38:67–83.CrossRefPubMedCentral Rice AM, Morales AC, Ho AT, Mordstein C, Mühlhausen S, Watson S, Cano L, Young B, Kudla G, Hurst LD. Evidence for strong mutation bias towards, and selection against, U content in SARS-CoV-2: implications for vaccine design. Mol Biol Evol. 2020;38:67–83.CrossRefPubMedCentral
13.
go back to reference Brito LA, Kommareddy S, Maione D, Uematsu Y, Giovani C, Berlanda Scorza F, Otten GR, Yu D, Mandl CW, Mason PW, et al. Self-amplifying mRNA vaccines. Adv Genet. 2015;89:179–233.PubMedCrossRef Brito LA, Kommareddy S, Maione D, Uematsu Y, Giovani C, Berlanda Scorza F, Otten GR, Yu D, Mandl CW, Mason PW, et al. Self-amplifying mRNA vaccines. Adv Genet. 2015;89:179–233.PubMedCrossRef
14.
go back to reference Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G, Gottardo R, Bica MA, Garofano A, Koch SD, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390:1511–20.PubMedCrossRef Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G, Gottardo R, Bica MA, Garofano A, Koch SD, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390:1511–20.PubMedCrossRef
15.
go back to reference Kirschman JL, Bhosle S, Vanover D, Blanchard EL, Loomis KH, Zurla C, Murray K, Lam BC, Santangelo PJ. Characterizing exogenous mRNA delivery, trafficking, cytoplasmic release and RNA-protein correlations at the level of single cells. Nucleic Acids Res. 2017;45:e113.PubMedPubMedCentralCrossRef Kirschman JL, Bhosle S, Vanover D, Blanchard EL, Loomis KH, Zurla C, Murray K, Lam BC, Santangelo PJ. Characterizing exogenous mRNA delivery, trafficking, cytoplasmic release and RNA-protein correlations at the level of single cells. Nucleic Acids Res. 2017;45:e113.PubMedPubMedCentralCrossRef
17.
go back to reference He S, Fan W, Wu N, Zhu J, Miao Y, Miao X, Li F, Zhang X, Gan Y. Lipid-based liquid crystalline nanoparticles facilitate cytosolic delivery of siRNA via structural transformation. Nano Lett. 2018;18:2411–9.PubMedCrossRef He S, Fan W, Wu N, Zhu J, Miao Y, Miao X, Li F, Zhang X, Gan Y. Lipid-based liquid crystalline nanoparticles facilitate cytosolic delivery of siRNA via structural transformation. Nano Lett. 2018;18:2411–9.PubMedCrossRef
18.
19.
go back to reference Zhu G, Zhang F. Efficient nanovaccine delivery in cancer immunotherapy. ACS Nano. 2017;11:2387–92.PubMedCrossRef Zhu G, Zhang F. Efficient nanovaccine delivery in cancer immunotherapy. ACS Nano. 2017;11:2387–92.PubMedCrossRef
20.
go back to reference Blakney AK, Zhu Y. Big is beautiful: enhanced saRNA delivery and immunogenicity by a higher molecular weight, bioreducible, cationic polymer. ACS Nano. 2020;14:5711–27.PubMedPubMedCentralCrossRef Blakney AK, Zhu Y. Big is beautiful: enhanced saRNA delivery and immunogenicity by a higher molecular weight, bioreducible, cationic polymer. ACS Nano. 2020;14:5711–27.PubMedPubMedCentralCrossRef
21.
go back to reference Egan KP, Hook LM. An HSV-2 nucleoside-modified mRNA genital herpes vaccine containing glycoproteins gC, gD, and gE protects mice against HSV-1 genital lesions and latent infection. PLoS Pathog. 2020;16:e1008795.PubMedPubMedCentralCrossRef Egan KP, Hook LM. An HSV-2 nucleoside-modified mRNA genital herpes vaccine containing glycoproteins gC, gD, and gE protects mice against HSV-1 genital lesions and latent infection. PLoS Pathog. 2020;16:e1008795.PubMedPubMedCentralCrossRef
22.
go back to reference Wang F, Xiao W, Elbahnasawy MA, Bao X, Zheng Q, Gong L, Zhou Y, Yang S, Fang A, Farag MMS, et al. Optimization of the linker length of mannose-cholesterol conjugates for enhanced mRNA delivery to dendritic cells by liposomes. Front Pharmacol. 2018;9:980.PubMedPubMedCentralCrossRef Wang F, Xiao W, Elbahnasawy MA, Bao X, Zheng Q, Gong L, Zhou Y, Yang S, Fang A, Farag MMS, et al. Optimization of the linker length of mannose-cholesterol conjugates for enhanced mRNA delivery to dendritic cells by liposomes. Front Pharmacol. 2018;9:980.PubMedPubMedCentralCrossRef
23.
go back to reference Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, Dowd KA, Sutherland LL, Scearce RM, Parks R, et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature. 2017;543:248–51.PubMedPubMedCentralCrossRef Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, Dowd KA, Sutherland LL, Scearce RM, Parks R, et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature. 2017;543:248–51.PubMedPubMedCentralCrossRef
24.
go back to reference Corbett KS, Edwards DK, Leist SR, Abiona OM, Boyoglu-Barnum S, Gillespie RA, Himansu S, Schäfer A, Ziwawo CT, DiPiazza AT, Dinnon KH. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature. 2020;586:567–71.PubMedPubMedCentralCrossRef Corbett KS, Edwards DK, Leist SR, Abiona OM, Boyoglu-Barnum S, Gillespie RA, Himansu S, Schäfer A, Ziwawo CT, DiPiazza AT, Dinnon KH. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature. 2020;586:567–71.PubMedPubMedCentralCrossRef
25.
go back to reference Corbett KS, Flynn B, Foulds KE, Francica JR, Boyoglu-Barnum S, Werner AP, Flach B, O'Connell S, Bock KW, Minai M, et al. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N Engl J Med. 2020;383:1544–55.PubMedCrossRef Corbett KS, Flynn B, Foulds KE, Francica JR, Boyoglu-Barnum S, Werner AP, Flach B, O'Connell S, Bock KW, Minai M, et al. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N Engl J Med. 2020;383:1544–55.PubMedCrossRef
26.
go back to reference Tai W, Zhang X, Drelich A, Shi J, Hsu JC, Luchsinger L, Hillyer CD, Tseng CK, Jiang S, Du L. A novel receptor-binding domain (RBD)-based mRNA vaccine against SARS-CoV-2. Cell Res. 2020;30:932–5.PubMedCrossRef Tai W, Zhang X, Drelich A, Shi J, Hsu JC, Luchsinger L, Hillyer CD, Tseng CK, Jiang S, Du L. A novel receptor-binding domain (RBD)-based mRNA vaccine against SARS-CoV-2. Cell Res. 2020;30:932–5.PubMedCrossRef
27.
go back to reference Shin MD, Shukla S, Chung YH. COVID-19 vaccine development and a potential nanomaterial path forward. Nat Nanotechnol. 2020;15:646–55.PubMedCrossRef Shin MD, Shukla S, Chung YH. COVID-19 vaccine development and a potential nanomaterial path forward. Nat Nanotechnol. 2020;15:646–55.PubMedCrossRef
30.
go back to reference Liang F, Lindgren G, Lin A, Thompson EA, Ols S, Röhss J, John S, Hassett K, Yuzhakov O, Bahl K, et al. Efficient targeting and activation of antigen-presenting cells in vivo after modified mRNA vaccine aministration in rhesus macaques. Mol Ther. 2017;25:2635–47.PubMedPubMedCentralCrossRef Liang F, Lindgren G, Lin A, Thompson EA, Ols S, Röhss J, John S, Hassett K, Yuzhakov O, Bahl K, et al. Efficient targeting and activation of antigen-presenting cells in vivo after modified mRNA vaccine aministration in rhesus macaques. Mol Ther. 2017;25:2635–47.PubMedPubMedCentralCrossRef
31.
go back to reference Pickering BM, Willis AE. The implications of structured 5′ untranslated regions on translation and disease. Semin Cell Dev Biol. 2005;16:39–47.PubMedCrossRef Pickering BM, Willis AE. The implications of structured 5′ untranslated regions on translation and disease. Semin Cell Dev Biol. 2005;16:39–47.PubMedCrossRef
32.
go back to reference Chatterjee S, Pal JK. Role of 5′- and 3′-untranslated regions of mRNAs in human diseases. Biol Cell. 2009;101:251–62.PubMedCrossRef Chatterjee S, Pal JK. Role of 5′- and 3′-untranslated regions of mRNAs in human diseases. Biol Cell. 2009;101:251–62.PubMedCrossRef
33.
go back to reference Furuichi Y, LaFiandra A, Shatkin AJ. 5′-terminal structure and mRNA stability. Nature. 1977;266:235–9.PubMedCrossRef Furuichi Y, LaFiandra A, Shatkin AJ. 5′-terminal structure and mRNA stability. Nature. 1977;266:235–9.PubMedCrossRef
34.
go back to reference Brenner S, Jacob F, Meselson M. An unstable intermediate carrying information from genes to ribosomes for protein synthesis. Nature. 1961;190:576–81.PubMedCrossRef Brenner S, Jacob F, Meselson M. An unstable intermediate carrying information from genes to ribosomes for protein synthesis. Nature. 1961;190:576–81.PubMedCrossRef
36.
go back to reference Deering RP, Kommareddy S, Ulmer JB, Brito LA, Geall AJ. Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin Drug Deliv. 2014;11:885–99.PubMedCrossRef Deering RP, Kommareddy S, Ulmer JB, Brito LA, Geall AJ. Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin Drug Deliv. 2014;11:885–99.PubMedCrossRef
37.
go back to reference Berglund P, Smerdou C, Fleeton MN, Tubulekas I, Liljeström P. Enhancing immune responses using suicidal DNA vaccines. Nat Biotechnol. 1998;16:562–5.PubMedCrossRef Berglund P, Smerdou C, Fleeton MN, Tubulekas I, Liljeström P. Enhancing immune responses using suicidal DNA vaccines. Nat Biotechnol. 1998;16:562–5.PubMedCrossRef
38.
go back to reference Biddlecome A, Habte HH, McGrath KM, Sambanthamoorthy S, Wurm M, Sykora MM, Knobler CM, Lorenz IC. Delivery of self-amplifying RNA vaccines in in vitro reconstituted virus-like particles. PLoS One. 2019;14:e0215031.PubMedPubMedCentralCrossRef Biddlecome A, Habte HH, McGrath KM, Sambanthamoorthy S, Wurm M, Sykora MM, Knobler CM, Lorenz IC. Delivery of self-amplifying RNA vaccines in in vitro reconstituted virus-like particles. PLoS One. 2019;14:e0215031.PubMedPubMedCentralCrossRef
39.
go back to reference Englezou PC, Sapet C, Démoulins T, Milona P, Ebensen T, Schulze K, Guzman CA, Poulhes F, Zelphati O, Ruggli N, McCullough KC. Self-amplifying replicon RNA delivery to dendritic cells by cationic lipids. Mol Ther Nucleic Acids. 2018;12:118–34.PubMedPubMedCentralCrossRef Englezou PC, Sapet C, Démoulins T, Milona P, Ebensen T, Schulze K, Guzman CA, Poulhes F, Zelphati O, Ruggli N, McCullough KC. Self-amplifying replicon RNA delivery to dendritic cells by cationic lipids. Mol Ther Nucleic Acids. 2018;12:118–34.PubMedPubMedCentralCrossRef
40.
go back to reference Beissert T, Perkovic M, Vogel A, Erbar S, Walzer KC, Hempel T, Brill S, Haefner E, Becker R, Türeci Ö, Sahin U. A trans-amplifying RNA vaccine strategy for induction of potent protective immunity. Mol Ther. 2020;28:119–28.PubMedCrossRef Beissert T, Perkovic M, Vogel A, Erbar S, Walzer KC, Hempel T, Brill S, Haefner E, Becker R, Türeci Ö, Sahin U. A trans-amplifying RNA vaccine strategy for induction of potent protective immunity. Mol Ther. 2020;28:119–28.PubMedCrossRef
41.
go back to reference Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics--developing a new class of drugs. Nat Rev Drug Discov. 2014;13:759–80.PubMedCrossRef Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics--developing a new class of drugs. Nat Rev Drug Discov. 2014;13:759–80.PubMedCrossRef
42.
go back to reference Pozzi B, Bragado L, Mammi P, Torti MF, Gaioli N, Gebhard LG, García Solá ME, Vaz-Drago R, Iglesias NG, García CC, et al. Dengue virus targets RBM10 deregulating host cell splicing and innate immune response. Nucleic Acids Res. 2020;48:6824–38.PubMedPubMedCentralCrossRef Pozzi B, Bragado L, Mammi P, Torti MF, Gaioli N, Gebhard LG, García Solá ME, Vaz-Drago R, Iglesias NG, García CC, et al. Dengue virus targets RBM10 deregulating host cell splicing and innate immune response. Nucleic Acids Res. 2020;48:6824–38.PubMedPubMedCentralCrossRef
43.
go back to reference Hornung V, Barchet W, Schlee M, Hartmann G. RNA recognition via TLR7 and TLR8. Handb Exp Pharmacol. 2008;(183):71-86. Hornung V, Barchet W, Schlee M, Hartmann G. RNA recognition via TLR7 and TLR8. Handb Exp Pharmacol. 2008;(183):71-86.
44.
go back to reference Thess A, Grund S, Mui BL, Hope MJ, Baumhof P, Fotin-Mleczek M, Schlake T. Sequence-engineered mRNA without chemical nucleoside modifications enables an effective protein therapy in large animals. Mol Ther. 2015;23:1456–64.PubMedPubMedCentralCrossRef Thess A, Grund S, Mui BL, Hope MJ, Baumhof P, Fotin-Mleczek M, Schlake T. Sequence-engineered mRNA without chemical nucleoside modifications enables an effective protein therapy in large animals. Mol Ther. 2015;23:1456–64.PubMedPubMedCentralCrossRef
45.
go back to reference Weng Y, Li C, Yang T, Hu B, Zhang M, Guo S, Xiao H, Liang XJ, Huang Y. The challenge and prospect of mRNA therapeutics landscape. Biotechnol Adv. 2020;40:107534.PubMedCrossRef Weng Y, Li C, Yang T, Hu B, Zhang M, Guo S, Xiao H, Liang XJ, Huang Y. The challenge and prospect of mRNA therapeutics landscape. Biotechnol Adv. 2020;40:107534.PubMedCrossRef
48.
go back to reference Weissman D, Pardi N, Muramatsu H, Karikó K. HPLC purification of in vitro transcribed long RNA. Methods Mol Biol. 2013;969:43–54.PubMedCrossRef Weissman D, Pardi N, Muramatsu H, Karikó K. HPLC purification of in vitro transcribed long RNA. Methods Mol Biol. 2013;969:43–54.PubMedCrossRef
49.
go back to reference Pardi N, Secreto AJ, Shan X, Debonera F, Glover J, Yi Y, Muramatsu H, Ni H, Mui BL, Tam YK, et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat Commun. 2017;8:14630.PubMedPubMedCentralCrossRef Pardi N, Secreto AJ, Shan X, Debonera F, Glover J, Yi Y, Muramatsu H, Ni H, Mui BL, Tam YK, et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat Commun. 2017;8:14630.PubMedPubMedCentralCrossRef
50.
go back to reference Conry RM, LoBuglio AF, Wright M, Sumerel L, Pike MJ, Johanning F, Benjamin R, Lu D, Curiel DT. Characterization of a messenger RNA polynucleotide vaccine vector. Cancer Res. 1995;55:1397–400.PubMed Conry RM, LoBuglio AF, Wright M, Sumerel L, Pike MJ, Johanning F, Benjamin R, Lu D, Curiel DT. Characterization of a messenger RNA polynucleotide vaccine vector. Cancer Res. 1995;55:1397–400.PubMed
51.
go back to reference Kore AR, Shanmugasundaram M, Charles I, Vlassov AV, Barta TJ. Locked nucleic acid (LNA)-modified dinucleotide mRNA cap analogue: synthesis, enzymatic incorporation, and utilization. J Am Chem Soc. 2009;131:6364–5.PubMedCrossRef Kore AR, Shanmugasundaram M, Charles I, Vlassov AV, Barta TJ. Locked nucleic acid (LNA)-modified dinucleotide mRNA cap analogue: synthesis, enzymatic incorporation, and utilization. J Am Chem Soc. 2009;131:6364–5.PubMedCrossRef
52.
go back to reference Jani B, Fuchs R. In vitro transcription and capping of Gaussia luciferase mRNA followed by HeLa cell transfection. J Vis Exp. 2012;(61):3702. Jani B, Fuchs R. In vitro transcription and capping of Gaussia luciferase mRNA followed by HeLa cell transfection. J Vis Exp. 2012;(61):3702.
53.
go back to reference Stepinski J, Waddell C, Stolarski R, Darzynkiewicz E, Rhoads RE. Synthesis and properties of mRNAs containing the novel “anti-reverse” cap analogs 7-methyl(3'-O-methyl)GpppG and 7-methyl (3'-deoxy)GpppG. RNA. 2001;7:1486–95.PubMedPubMedCentral Stepinski J, Waddell C, Stolarski R, Darzynkiewicz E, Rhoads RE. Synthesis and properties of mRNAs containing the novel “anti-reverse” cap analogs 7-methyl(3'-O-methyl)GpppG and 7-methyl (3'-deoxy)GpppG. RNA. 2001;7:1486–95.PubMedPubMedCentral
54.
go back to reference Urbina F, Morales-Pison S. Enzymatic protein biopolymers as a tool to synthetize eukaryotic messenger ribonucleic acid (mRNA) with uses in vaccination, immunotherapy and nanotechnology. Polymers (Basel). 2020;12:1633.PubMedCentralCrossRef Urbina F, Morales-Pison S. Enzymatic protein biopolymers as a tool to synthetize eukaryotic messenger ribonucleic acid (mRNA) with uses in vaccination, immunotherapy and nanotechnology. Polymers (Basel). 2020;12:1633.PubMedCentralCrossRef
55.
go back to reference Whitelaw E, Coates A, Proudfoot NJ. Globin gene transcripts can utilize histone gene 3′ end processing signals. Nucleic Acids Res. 1986;14:7059–70.PubMedPubMedCentralCrossRef Whitelaw E, Coates A, Proudfoot NJ. Globin gene transcripts can utilize histone gene 3′ end processing signals. Nucleic Acids Res. 1986;14:7059–70.PubMedPubMedCentralCrossRef
56.
go back to reference Doel MT, Carey NH. The translational capacity of deadenylated ovalbumin messenger RNA. Cell. 1976;8:51–8.PubMedCrossRef Doel MT, Carey NH. The translational capacity of deadenylated ovalbumin messenger RNA. Cell. 1976;8:51–8.PubMedCrossRef
57.
go back to reference Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, Türeci O, Sahin U. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood. 2006;108:4009–17.PubMedCrossRef Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, Türeci O, Sahin U. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood. 2006;108:4009–17.PubMedCrossRef
58.
go back to reference Shabalina SA, Spiridonov NA, Kashina A. Sounds of silence: synonymous nucleotides as a key to biological regulation and complexity. Nucleic Acids Res. 2013;41:2073–94.PubMedPubMedCentralCrossRef Shabalina SA, Spiridonov NA, Kashina A. Sounds of silence: synonymous nucleotides as a key to biological regulation and complexity. Nucleic Acids Res. 2013;41:2073–94.PubMedPubMedCentralCrossRef
59.
60.
go back to reference Homma K, Noguchi T, Fukuchi S. Codon usage is less optimized in eukaryotic gene segments encoding intrinsically disordered regions than in those encoding structural domains. Nucleic Acids Res. 2016;44:10051–61.PubMedPubMedCentral Homma K, Noguchi T, Fukuchi S. Codon usage is less optimized in eukaryotic gene segments encoding intrinsically disordered regions than in those encoding structural domains. Nucleic Acids Res. 2016;44:10051–61.PubMedPubMedCentral
61.
go back to reference Zhong C, Wei P, Zhang YP. Enhancing functional expression of codon-optimized heterologous enzymes in Escherichia coli BL21(DE3) by selective introduction of synonymous rare codons. Biotechnol Bioeng. 2017;114:1054–64.PubMedCrossRef Zhong C, Wei P, Zhang YP. Enhancing functional expression of codon-optimized heterologous enzymes in Escherichia coli BL21(DE3) by selective introduction of synonymous rare codons. Biotechnol Bioeng. 2017;114:1054–64.PubMedCrossRef
62.
go back to reference Yokokawa H, Higashino A, Suzuki S, Moriyama M, Nakamura N, Suzuki T, Suzuki R, Ishii K, Kobiyama K, Ishii KJ, et al. Induction of humoural and cellular immunity by immunisation with HCV particle vaccine in a non-human primate model. Gut. 2018;67:372–9.PubMedCrossRef Yokokawa H, Higashino A, Suzuki S, Moriyama M, Nakamura N, Suzuki T, Suzuki R, Ishii K, Kobiyama K, Ishii KJ, et al. Induction of humoural and cellular immunity by immunisation with HCV particle vaccine in a non-human primate model. Gut. 2018;67:372–9.PubMedCrossRef
63.
go back to reference Monslow MA, Elbashir S, Sullivan NL, Thiriot DS, Ahl P, Smith J, Miller E, Cook J, Cosmi S, Thoryk E, et al. Immunogenicity generated by mRNA vaccine encoding VZV gE antigen is comparable to adjuvanted subunit vaccine and better than live attenuated vaccine in nonhuman primates. Vaccine. 2020;38:5793–802.PubMedCrossRef Monslow MA, Elbashir S, Sullivan NL, Thiriot DS, Ahl P, Smith J, Miller E, Cook J, Cosmi S, Thoryk E, et al. Immunogenicity generated by mRNA vaccine encoding VZV gE antigen is comparable to adjuvanted subunit vaccine and better than live attenuated vaccine in nonhuman primates. Vaccine. 2020;38:5793–802.PubMedCrossRef
64.
go back to reference Tateshita N, Miura N, Tanaka H, Masuda T, Ohtsuki S, Tange K, Nakai Y, Yoshioka H, Akita H. Development of a lipoplex-type mRNA carrier composed of an ionizable lipid with a vitamin E scaffold and the KALA peptide for use as an ex vivo dendritic cell-based cancer vaccine. J Control Release. 2019;310:36–46.PubMedCrossRef Tateshita N, Miura N, Tanaka H, Masuda T, Ohtsuki S, Tange K, Nakai Y, Yoshioka H, Akita H. Development of a lipoplex-type mRNA carrier composed of an ionizable lipid with a vitamin E scaffold and the KALA peptide for use as an ex vivo dendritic cell-based cancer vaccine. J Control Release. 2019;310:36–46.PubMedCrossRef
65.
go back to reference Joe PT, Christopoulou I, van Hoecke L, Schepens B, Ysenbaert T, Heirman C, Thielemans K, Saelens X, Aerts JL. Intranodal administration of mRNA encoding nucleoprotein provides cross-strain immunity against influenza in mice. J Transl Med. 2019;17:242.PubMedPubMedCentralCrossRef Joe PT, Christopoulou I, van Hoecke L, Schepens B, Ysenbaert T, Heirman C, Thielemans K, Saelens X, Aerts JL. Intranodal administration of mRNA encoding nucleoprotein provides cross-strain immunity against influenza in mice. J Transl Med. 2019;17:242.PubMedPubMedCentralCrossRef
66.
go back to reference Yang Y, Li CW, Chan LC, Wei Y, Hsu JM, Xia W, Cha JH, Hou J, Hsu JL, Sun L, Hung MC. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28:862–4.PubMedPubMedCentralCrossRef Yang Y, Li CW, Chan LC, Wei Y, Hsu JM, Xia W, Cha JH, Hou J, Hsu JL, Sun L, Hung MC. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28:862–4.PubMedPubMedCentralCrossRef
67.
go back to reference Cano F, Lehner PJ. A novel post-transcriptional role for ubiquitin in the differential regulation of MHC class I allotypes. Mol Immunol. 2013;55:135–8.PubMedPubMedCentralCrossRef Cano F, Lehner PJ. A novel post-transcriptional role for ubiquitin in the differential regulation of MHC class I allotypes. Mol Immunol. 2013;55:135–8.PubMedPubMedCentralCrossRef
68.
go back to reference Yamamoto K, Venida A, Yano J, Biancur DE, Kakiuchi M, Gupta S, Sohn ASW, Mukhopadhyay S, Lin EY, Parker SJ, et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature. 2020;581:100–5.PubMedPubMedCentralCrossRef Yamamoto K, Venida A, Yano J, Biancur DE, Kakiuchi M, Gupta S, Sohn ASW, Mukhopadhyay S, Lin EY, Parker SJ, et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature. 2020;581:100–5.PubMedPubMedCentralCrossRef
69.
go back to reference Campillo-Davo D, Versteven M, Roex G, Reu H, Heijden SV, Anguille S, Berneman ZN, Tendeloo V, Lion E. Rapid assessment of functional avidity of tumor-specific T cell receptors using an antigen-presenting tumor cell line electroporated with full-length tumor antigen mRNA. Cancers (Basel). 2020;12(2):256.CrossRef Campillo-Davo D, Versteven M, Roex G, Reu H, Heijden SV, Anguille S, Berneman ZN, Tendeloo V, Lion E. Rapid assessment of functional avidity of tumor-specific T cell receptors using an antigen-presenting tumor cell line electroporated with full-length tumor antigen mRNA. Cancers (Basel). 2020;12(2):256.CrossRef
70.
go back to reference Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunol Immunother. 2006;55:672–83.PubMedCrossRef Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunol Immunother. 2006;55:672–83.PubMedCrossRef
71.
go back to reference Lindgren G, Ols S, Liang F, Thompson EA, Lin A, Hellgren F, Bahl K, John S, Yuzhakov O, Hassett KJ, et al. Induction of robust B cell responses after influenza mRNA vaccination is accompanied by circulating hemagglutinin-specific ICOS+ PD-1+ CXCR3+ T follicular helper cells. Front Immunol. 2017;8:1539.PubMedPubMedCentralCrossRef Lindgren G, Ols S, Liang F, Thompson EA, Lin A, Hellgren F, Bahl K, John S, Yuzhakov O, Hassett KJ, et al. Induction of robust B cell responses after influenza mRNA vaccination is accompanied by circulating hemagglutinin-specific ICOS+ PD-1+ CXCR3+ T follicular helper cells. Front Immunol. 2017;8:1539.PubMedPubMedCentralCrossRef
72.
go back to reference Basso K, Dalla-Favera R. Germinal centres and B cell lymphomagenesis. Nat Rev Immunol. 2015;15:172–84.PubMedCrossRef Basso K, Dalla-Favera R. Germinal centres and B cell lymphomagenesis. Nat Rev Immunol. 2015;15:172–84.PubMedCrossRef
73.
go back to reference Heyman B. Regulation of antibody responses via antibodies, complement, and Fc receptors. Annu Rev Immunol. 2000;18:709–37.PubMedCrossRef Heyman B. Regulation of antibody responses via antibodies, complement, and Fc receptors. Annu Rev Immunol. 2000;18:709–37.PubMedCrossRef
74.
go back to reference Tam HH, Melo MB. Sustained antigen availability during germinal center initiation enhances antibody responses to vaccination. Proc Natl Acad Sci U S A. 2016;113:E6639–e6648.PubMedPubMedCentralCrossRef Tam HH, Melo MB. Sustained antigen availability during germinal center initiation enhances antibody responses to vaccination. Proc Natl Acad Sci U S A. 2016;113:E6639–e6648.PubMedPubMedCentralCrossRef
75.
go back to reference Bahl K, Senn JJ, Yuzhakov O, Bulychev A, Brito LA, Hassett KJ, Laska ME, Smith M, Almarsson Ö, Thompson J, et al. Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Mol Ther. 2017;25:1316–27.PubMedPubMedCentralCrossRef Bahl K, Senn JJ, Yuzhakov O, Bulychev A, Brito LA, Hassett KJ, Laska ME, Smith M, Almarsson Ö, Thompson J, et al. Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Mol Ther. 2017;25:1316–27.PubMedPubMedCentralCrossRef
76.
go back to reference Fleeton MN, Chen M, Berglund P, Rhodes G, Parker SE, Murphy M, Atkins GJ, Liljeström P. Self-replicative RNA vaccines elicit protection against influenza a virus, respiratory syncytial virus, and a tickborne encephalitis virus. J Infect Dis. 2001;183:1395–8.PubMedCrossRef Fleeton MN, Chen M, Berglund P, Rhodes G, Parker SE, Murphy M, Atkins GJ, Liljeström P. Self-replicative RNA vaccines elicit protection against influenza a virus, respiratory syncytial virus, and a tickborne encephalitis virus. J Infect Dis. 2001;183:1395–8.PubMedCrossRef
77.
go back to reference McCullough KC, Bassi I, Milona P, Suter R, Thomann-Harwood L, Englezou P, Démoulins T, Ruggli N. Self-replicating replicon-RNA delivery to dendritic cells by chitosan-nanoparticles for translation in vitro and in vivo. Mol Ther Nucleic Acids. 2014;3:e173.PubMedPubMedCentralCrossRef McCullough KC, Bassi I, Milona P, Suter R, Thomann-Harwood L, Englezou P, Démoulins T, Ruggli N. Self-replicating replicon-RNA delivery to dendritic cells by chitosan-nanoparticles for translation in vitro and in vivo. Mol Ther Nucleic Acids. 2014;3:e173.PubMedPubMedCentralCrossRef
78.
go back to reference Démoulins T, Milona P, Englezou PC, Ebensen T, Schulze K, Suter R, Pichon C, Midoux P, Guzmán CA, Ruggli N, McCullough KC. Polyethylenimine-based polyplex delivery of self-replicating RNA vaccines. Nanomedicine. 2016;12:711–22.PubMedCrossRef Démoulins T, Milona P, Englezou PC, Ebensen T, Schulze K, Suter R, Pichon C, Midoux P, Guzmán CA, Ruggli N, McCullough KC. Polyethylenimine-based polyplex delivery of self-replicating RNA vaccines. Nanomedicine. 2016;12:711–22.PubMedCrossRef
79.
go back to reference Liehl P, Zuzarte-Luís V, Chan J, Zillinger T, Baptista F, Carapau D, Konert M, Hanson KK, Carret C, Lassnig C, et al. Host-cell sensors for plasmodium activate innate immunity against liver-stage infection. Nat Med. 2014;20:47–53.PubMedCrossRef Liehl P, Zuzarte-Luís V, Chan J, Zillinger T, Baptista F, Carapau D, Konert M, Hanson KK, Carret C, Lassnig C, et al. Host-cell sensors for plasmodium activate innate immunity against liver-stage infection. Nat Med. 2014;20:47–53.PubMedCrossRef
80.
go back to reference Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181:1036–1045.e1039.PubMedPubMedCentralCrossRef Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181:1036–1045.e1039.PubMedPubMedCentralCrossRef
81.
go back to reference Jain A, Irizarry-Caro RA. T cells instruct myeloid cells to produce inflammasome-independent IL-1β and cause autoimmunity. Nat Immunol. 2020;21:65–74.PubMedCrossRef Jain A, Irizarry-Caro RA. T cells instruct myeloid cells to produce inflammasome-independent IL-1β and cause autoimmunity. Nat Immunol. 2020;21:65–74.PubMedCrossRef
82.
go back to reference Chow KT, Gale M Jr, Loo YM. RIG-I and other RNA sensors in antiviral immunity. Annu Rev Immunol. 2018;36:667–94.PubMedCrossRef Chow KT, Gale M Jr, Loo YM. RIG-I and other RNA sensors in antiviral immunity. Annu Rev Immunol. 2018;36:667–94.PubMedCrossRef
83.
go back to reference Medzhitov R, Preston-Hurlburt P, Janeway CA Jr. A human homologue of the Drosophila toll protein signals activation of adaptive immunity. Nature. 1997;388:394–7.PubMedCrossRef Medzhitov R, Preston-Hurlburt P, Janeway CA Jr. A human homologue of the Drosophila toll protein signals activation of adaptive immunity. Nature. 1997;388:394–7.PubMedCrossRef
84.
go back to reference Bergougnan C, Dittlein DC, Hümmer E, Riepl R, Eisenbart S, Böck D, Griesbaum L, Weigl A, Damialis A, Hartwig A, et al. Physical and immunological barrier of human primary nasal epithelial cells from non-allergic and allergic donors. World Allergy Organ J. 2020;13:100109.PubMedPubMedCentralCrossRef Bergougnan C, Dittlein DC, Hümmer E, Riepl R, Eisenbart S, Böck D, Griesbaum L, Weigl A, Damialis A, Hartwig A, et al. Physical and immunological barrier of human primary nasal epithelial cells from non-allergic and allergic donors. World Allergy Organ J. 2020;13:100109.PubMedPubMedCentralCrossRef
85.
86.
go back to reference Feng H, Lenarcic EM, Yamane D, Wauthier E, Mo J, Guo H, McGivern DR. NLRX1 promotes immediate IRF1-directed antiviral responses by limiting dsRNA-activated translational inhibition mediated by PKR. Nat Immunol. 2017;18:1299–309.PubMedPubMedCentralCrossRef Feng H, Lenarcic EM, Yamane D, Wauthier E, Mo J, Guo H, McGivern DR. NLRX1 promotes immediate IRF1-directed antiviral responses by limiting dsRNA-activated translational inhibition mediated by PKR. Nat Immunol. 2017;18:1299–309.PubMedPubMedCentralCrossRef
87.
go back to reference Ilan L, Osman F, Namer LS, Eliahu E, Cohen-Chalamish S, Ben-Asouli Y, Banai Y, Kaempfer R. PKR activation and eIF2α phosphorylation mediate human globin mRNA splicing at spliceosome assembly. Cell Res. 2017;27:688–704.PubMedPubMedCentralCrossRef Ilan L, Osman F, Namer LS, Eliahu E, Cohen-Chalamish S, Ben-Asouli Y, Banai Y, Kaempfer R. PKR activation and eIF2α phosphorylation mediate human globin mRNA splicing at spliceosome assembly. Cell Res. 2017;27:688–704.PubMedPubMedCentralCrossRef
88.
go back to reference Ghosh A, Shao L, Sampath P, Zhao B, Patel NV, Zhu J, Behl B, Parise RA, Beumer JH, O'Sullivan RJ, et al. Oligoadenylate-synthetase-family protein OASL inhibits activity of the DNA sensor cGAS during DNA virus infection to limit interferon production. Immunity. 2019;50:51–63.e55.PubMedPubMedCentralCrossRef Ghosh A, Shao L, Sampath P, Zhao B, Patel NV, Zhu J, Behl B, Parise RA, Beumer JH, O'Sullivan RJ, et al. Oligoadenylate-synthetase-family protein OASL inhibits activity of the DNA sensor cGAS during DNA virus infection to limit interferon production. Immunity. 2019;50:51–63.e55.PubMedPubMedCentralCrossRef
89.
go back to reference Lin H, Jiang M, Liu L, Yang Z, Ma Z, Liu S. The long noncoding RNA Lnczc3h7a promotes a TRIM25-mediated RIG-I antiviral innate immune response. Nat Immunol. 2019;20:812–23.PubMedCrossRef Lin H, Jiang M, Liu L, Yang Z, Ma Z, Liu S. The long noncoding RNA Lnczc3h7a promotes a TRIM25-mediated RIG-I antiviral innate immune response. Nat Immunol. 2019;20:812–23.PubMedCrossRef
90.
go back to reference Kaempfer R. Interferon-gamma mRNA attenuates its own translation by activating PKR: a molecular basis for the therapeutic effect of interferon-beta in multiple sclerosis. Cell Res. 2006;16:148–53.PubMedCrossRef Kaempfer R. Interferon-gamma mRNA attenuates its own translation by activating PKR: a molecular basis for the therapeutic effect of interferon-beta in multiple sclerosis. Cell Res. 2006;16:148–53.PubMedCrossRef
91.
go back to reference Bogers WM, Oostermeijer H, Mooij P, Koopman G, Verschoor EJ, Davis D, Ulmer JB, Brito LA, Cu Y, Banerjee K, et al. Potent immune responses in rhesus macaques induced by nonviral delivery of a self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsion. J Infect Dis. 2015;211:947–55.PubMedCrossRef Bogers WM, Oostermeijer H, Mooij P, Koopman G, Verschoor EJ, Davis D, Ulmer JB, Brito LA, Cu Y, Banerjee K, et al. Potent immune responses in rhesus macaques induced by nonviral delivery of a self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsion. J Infect Dis. 2015;211:947–55.PubMedCrossRef
92.
go back to reference Brito LA, Chan M, Shaw CA, Hekele A, Carsillo T, Schaefer M, Archer J, Seubert A, Otten GR, Beard CW, et al. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol Ther. 2014;22:2118–29.PubMedPubMedCentralCrossRef Brito LA, Chan M, Shaw CA, Hekele A, Carsillo T, Schaefer M, Archer J, Seubert A, Otten GR, Beard CW, et al. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol Ther. 2014;22:2118–29.PubMedPubMedCentralCrossRef
93.
go back to reference De Keersmaecker B, Claerhout S, Carrasco J, Bar I, Corthals J, Wilgenhof S, Neyns B, Thielemans K. TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: link between T-cell activation and clinical responses in advanced melanoma. J Immunother Cancer. 2020;8(1):e000329.PubMedPubMedCentralCrossRef De Keersmaecker B, Claerhout S, Carrasco J, Bar I, Corthals J, Wilgenhof S, Neyns B, Thielemans K. TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: link between T-cell activation and clinical responses in advanced melanoma. J Immunother Cancer. 2020;8(1):e000329.PubMedPubMedCentralCrossRef
94.
go back to reference Bialkowski L, van Weijnen A, Van der Jeught K, Renmans D, Daszkiewicz L, Heirman C, Stangé G, Breckpot K, Aerts JL, Thielemans K. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours. Sci Rep. 2016;6:22509.PubMedPubMedCentralCrossRef Bialkowski L, van Weijnen A, Van der Jeught K, Renmans D, Daszkiewicz L, Heirman C, Stangé G, Breckpot K, Aerts JL, Thielemans K. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours. Sci Rep. 2016;6:22509.PubMedPubMedCentralCrossRef
95.
go back to reference Leal L, Guardo AC, Morón-López S, Salgado M, Mothe B, Heirman C, Pannus P, Vanham G, van den Ham HJ, Gruters R, et al. Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection. AIDS. 2018;32:2533–45.PubMedCrossRef Leal L, Guardo AC, Morón-López S, Salgado M, Mothe B, Heirman C, Pannus P, Vanham G, van den Ham HJ, Gruters R, et al. Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection. AIDS. 2018;32:2533–45.PubMedCrossRef
96.
go back to reference de Jong W, Aerts J, Allard S, Brander C, Buyze J, Florence E, van Gorp E, Vanham G, Leal L, Mothe B, et al. iHIVARNA phase IIa, a randomized, placebo-controlled, double-blinded trial to evaluate the safety and immunogenicity of iHIVARNA-01 in chronically HIV-infected patients under stable combined antiretroviral therapy. Trials. 2019;20:361.PubMedPubMedCentralCrossRef de Jong W, Aerts J, Allard S, Brander C, Buyze J, Florence E, van Gorp E, Vanham G, Leal L, Mothe B, et al. iHIVARNA phase IIa, a randomized, placebo-controlled, double-blinded trial to evaluate the safety and immunogenicity of iHIVARNA-01 in chronically HIV-infected patients under stable combined antiretroviral therapy. Trials. 2019;20:361.PubMedPubMedCentralCrossRef
97.
go back to reference Jong W, Leal L, Buyze J, Pannus P, Guardo A, Salgado M, Mothe B, Molto J, Moron-Lopez S, Gálvez C, et al. Therapeutic vaccine in chronically HIV-1-infected patients: a randomized, double-blind, placebo-controlled phase IIa trial with HTI-TriMix. Vaccines (Basel). 2019;7(4):209.CrossRef Jong W, Leal L, Buyze J, Pannus P, Guardo A, Salgado M, Mothe B, Molto J, Moron-Lopez S, Gálvez C, et al. Therapeutic vaccine in chronically HIV-1-infected patients: a randomized, double-blind, placebo-controlled phase IIa trial with HTI-TriMix. Vaccines (Basel). 2019;7(4):209.CrossRef
98.
go back to reference Kübler H, Scheel B, Gnad-Vogt U, Miller K, Schultze-Seemann W, Vom Dorp F, Parmiani G, Hampel C, Wedel S, Trojan L, et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study. J Immunother Cancer. 2015;3:26.PubMedPubMedCentralCrossRef Kübler H, Scheel B, Gnad-Vogt U, Miller K, Schultze-Seemann W, Vom Dorp F, Parmiani G, Hampel C, Wedel S, Trojan L, et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study. J Immunother Cancer. 2015;3:26.PubMedPubMedCentralCrossRef
99.
go back to reference Fotin-Mleczek M, Zanzinger K, Heidenreich R, Lorenz C, Thess A, Duchardt KM, Kallen KJ. Highly potent mRNA based cancer vaccines represent an attractive platform for combination therapies supporting an improved therapeutic effect. J Gene Med. 2012;14:428–39.PubMedCrossRef Fotin-Mleczek M, Zanzinger K, Heidenreich R, Lorenz C, Thess A, Duchardt KM, Kallen KJ. Highly potent mRNA based cancer vaccines represent an attractive platform for combination therapies supporting an improved therapeutic effect. J Gene Med. 2012;14:428–39.PubMedCrossRef
100.
go back to reference Weide B, Carralot JP, Reese A, Scheel B, Eigentler TK, Hoerr I, Rammensee HG, Garbe C, Pascolo S. Results of the first phase I/II clinical vaccination trial with direct injection of mRNA. J Immunother. 2008;31:180–8.PubMedCrossRef Weide B, Carralot JP, Reese A, Scheel B, Eigentler TK, Hoerr I, Rammensee HG, Garbe C, Pascolo S. Results of the first phase I/II clinical vaccination trial with direct injection of mRNA. J Immunother. 2008;31:180–8.PubMedCrossRef
101.
go back to reference Sebastian M, Papachristofilou A, Weiss C, Früh M, Cathomas R, Hilbe W, Wehler T, Rippin G, Koch SD, Scheel B, et al. Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive®) combined with local radiation as consolidation and maintenance treatment for patients with stage IV non-small cell lung cancer. BMC Cancer. 2014;14:748.PubMedPubMedCentralCrossRef Sebastian M, Papachristofilou A, Weiss C, Früh M, Cathomas R, Hilbe W, Wehler T, Rippin G, Koch SD, Scheel B, et al. Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive®) combined with local radiation as consolidation and maintenance treatment for patients with stage IV non-small cell lung cancer. BMC Cancer. 2014;14:748.PubMedPubMedCentralCrossRef
102.
go back to reference Ziegler A, Soldner C, Lienenklaus S, Spanier J. A new RNA-based adjuvant enhances virus-specific vaccine responses by locally triggering TLR- and RLH-dependent effects. J Immunol. 2017;198:1595–605.PubMedCrossRef Ziegler A, Soldner C, Lienenklaus S, Spanier J. A new RNA-based adjuvant enhances virus-specific vaccine responses by locally triggering TLR- and RLH-dependent effects. J Immunol. 2017;198:1595–605.PubMedCrossRef
103.
go back to reference Mauriello A, Manolio C, Cavalluzzo B, Avallone A, Borrelli M, Morabito A, Iovine E, Chambery A, Russo R, Tornesello ML, et al. Immunological effects of adjuvants in subsets of antigen presenting cells of cancer patients undergoing chemotherapy. J Transl Med. 2020;18:34.PubMedPubMedCentralCrossRef Mauriello A, Manolio C, Cavalluzzo B, Avallone A, Borrelli M, Morabito A, Iovine E, Chambery A, Russo R, Tornesello ML, et al. Immunological effects of adjuvants in subsets of antigen presenting cells of cancer patients undergoing chemotherapy. J Transl Med. 2020;18:34.PubMedPubMedCentralCrossRef
104.
go back to reference Arvin AM, Fink K, Schmid MA, Cathcart A, Spreafico R, Havenar-Daughton C. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. Nature. 2020;584:353–63.PubMedCrossRef Arvin AM, Fink K, Schmid MA, Cathcart A, Spreafico R, Havenar-Daughton C. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. Nature. 2020;584:353–63.PubMedCrossRef
107.
go back to reference Laczkó D, Hogan MJ, Toulmin SA, Hicks P, Lederer K, Gaudette BT, Castaño D, Amanat F, Muramatsu H, Oguin TH 3rd, et al. A single immunization with nucleoside-modified mRNA vaccines elicits strong cellular and humoral immune responses against SARS-CoV-2 in mice. Immunity. 2020;53(4):724–732.e727.PubMedPubMedCentralCrossRef Laczkó D, Hogan MJ, Toulmin SA, Hicks P, Lederer K, Gaudette BT, Castaño D, Amanat F, Muramatsu H, Oguin TH 3rd, et al. A single immunization with nucleoside-modified mRNA vaccines elicits strong cellular and humoral immune responses against SARS-CoV-2 in mice. Immunity. 2020;53(4):724–732.e727.PubMedPubMedCentralCrossRef
108.
go back to reference Liu J, Chang J, Jiang Y, Meng X, Sun T, Mao L, Xu Q, Wang M. Fast and efficient CRISPR/Cas9 genome editing in vivo enabled by bioreducible lipid and messenger RNA nanoparticles. Adv Mater. 2019;31:e1902575.PubMedPubMedCentralCrossRef Liu J, Chang J, Jiang Y, Meng X, Sun T, Mao L, Xu Q, Wang M. Fast and efficient CRISPR/Cas9 genome editing in vivo enabled by bioreducible lipid and messenger RNA nanoparticles. Adv Mater. 2019;31:e1902575.PubMedPubMedCentralCrossRef
109.
go back to reference Lokugamage MP, Gan Z, Zurla C, Levin J, Islam FZ, Kalathoor S, Sato M, Sago CD, Santangelo PJ, Dahlman JE. Mild innate immune activation overrides efficient nanoparticle-mediated RNA delivery. Adv Mater. 2020;32:e1904905.PubMedCrossRef Lokugamage MP, Gan Z, Zurla C, Levin J, Islam FZ, Kalathoor S, Sato M, Sago CD, Santangelo PJ, Dahlman JE. Mild innate immune activation overrides efficient nanoparticle-mediated RNA delivery. Adv Mater. 2020;32:e1904905.PubMedCrossRef
110.
go back to reference Ramishetti S, Hazan-Halevy I, Palakuri R, Chatterjee S, Naidu Gonna S, Dammes N, Freilich I, Kolik Shmuel L, Danino D, Peer D. A combinatorial library of lipid nanoparticles for RNA delivery to leukocytes. Adv Mater. 2020;32:e1906128.PubMedCrossRef Ramishetti S, Hazan-Halevy I, Palakuri R, Chatterjee S, Naidu Gonna S, Dammes N, Freilich I, Kolik Shmuel L, Danino D, Peer D. A combinatorial library of lipid nanoparticles for RNA delivery to leukocytes. Adv Mater. 2020;32:e1906128.PubMedCrossRef
111.
go back to reference Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: current perspectives. Adv Drug Deliv Rev. 2020;154-155:37–63.PubMedCrossRef Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: current perspectives. Adv Drug Deliv Rev. 2020;154-155:37–63.PubMedCrossRef
112.
go back to reference Lou G, Anderluzzi G, Schmidt ST, Woods S, Gallorini S, Brazzoli M, Giusti F, Ferlenghi I, Johnson RN, Roberts CW, et al. Delivery of self-amplifying mRNA vaccines by cationic lipid nanoparticles: the impact of cationic lipid selection. J Control Release. 2020;325:370–9.PubMedCrossRef Lou G, Anderluzzi G, Schmidt ST, Woods S, Gallorini S, Brazzoli M, Giusti F, Ferlenghi I, Johnson RN, Roberts CW, et al. Delivery of self-amplifying mRNA vaccines by cationic lipid nanoparticles: the impact of cationic lipid selection. J Control Release. 2020;325:370–9.PubMedCrossRef
113.
go back to reference Tang B, Qian Y, Fang G. Development of lipid-polymer hybrid nanoparticles for improving oral absorption of enoxaparin. Pharmaceutics. 2020;12(7):607.PubMedCentralCrossRef Tang B, Qian Y, Fang G. Development of lipid-polymer hybrid nanoparticles for improving oral absorption of enoxaparin. Pharmaceutics. 2020;12(7):607.PubMedCentralCrossRef
114.
go back to reference Kedmi R, Ben-Arie N, Peer D. The systemic toxicity of positively charged lipid nanoparticles and the role of toll-like receptor 4 in immune activation. Biomaterials. 2010;31:6867–75.PubMedCrossRef Kedmi R, Ben-Arie N, Peer D. The systemic toxicity of positively charged lipid nanoparticles and the role of toll-like receptor 4 in immune activation. Biomaterials. 2010;31:6867–75.PubMedCrossRef
115.
116.
go back to reference Hajj KA, Melamed JR, Chaudhary N, Lamson NG, Ball RL, Yerneni SS, Whitehead KA. A potent branched-tail lipid nanoparticle enables multiplexed mRNA delivery and gene editing in vivo. Nano Lett. 2020;20:5167–75.PubMedPubMedCentralCrossRef Hajj KA, Melamed JR, Chaudhary N, Lamson NG, Ball RL, Yerneni SS, Whitehead KA. A potent branched-tail lipid nanoparticle enables multiplexed mRNA delivery and gene editing in vivo. Nano Lett. 2020;20:5167–75.PubMedPubMedCentralCrossRef
117.
go back to reference Kauffman KJ, Dorkin JR, Yang JH, Heartlein MW, DeRosa F, Mir FF, Fenton OS, Anderson DG. Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs. Nano Lett. 2015;15:7300–6.PubMedCrossRef Kauffman KJ, Dorkin JR, Yang JH, Heartlein MW, DeRosa F, Mir FF, Fenton OS, Anderson DG. Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs. Nano Lett. 2015;15:7300–6.PubMedCrossRef
118.
go back to reference Zhang G, Guo B, Wu H, Tang T, Zhang BT, Zheng L, He Y, Yang Z, Pan X, Chow H, et al. A delivery system targeting bone formation surfaces to facilitate RNAi-based anabolic therapy. Nat Med. 2012;18:307–14.PubMedCrossRef Zhang G, Guo B, Wu H, Tang T, Zhang BT, Zheng L, He Y, Yang Z, Pan X, Chow H, et al. A delivery system targeting bone formation surfaces to facilitate RNAi-based anabolic therapy. Nat Med. 2012;18:307–14.PubMedCrossRef
119.
go back to reference Li B, Luo X, Deng B, Wang J, McComb DW, Shi Y, Gaensler KM, Tan X, Dunn AL, Kerlin BA, Dong Y. An orthogonal array optimization of lipid-like nanoparticles for mRNA delivery in vivo. Nano Lett. 2015;15:8099–107.PubMedPubMedCentralCrossRef Li B, Luo X, Deng B, Wang J, McComb DW, Shi Y, Gaensler KM, Tan X, Dunn AL, Kerlin BA, Dong Y. An orthogonal array optimization of lipid-like nanoparticles for mRNA delivery in vivo. Nano Lett. 2015;15:8099–107.PubMedPubMedCentralCrossRef
120.
go back to reference Suga K, Tanabe T, Tomita H, Shimanouchi T, Umakoshi H. Conformational change of single-stranded RNAs induced by liposome binding. Nucleic Acids Res. 2011;39:8891–900.PubMedPubMedCentralCrossRef Suga K, Tanabe T, Tomita H, Shimanouchi T, Umakoshi H. Conformational change of single-stranded RNAs induced by liposome binding. Nucleic Acids Res. 2011;39:8891–900.PubMedPubMedCentralCrossRef
122.
go back to reference Sato Y, Hashiba K, Sasaki K, Maeki M, Tokeshi M, Harashima H. Understanding structure-activity relationships of pH-sensitive cationic lipids facilitates the rational identification of promising lipid nanoparticles for delivering siRNAs in vivo. J Control Release. 2019;295:140–52.PubMedCrossRef Sato Y, Hashiba K, Sasaki K, Maeki M, Tokeshi M, Harashima H. Understanding structure-activity relationships of pH-sensitive cationic lipids facilitates the rational identification of promising lipid nanoparticles for delivering siRNAs in vivo. J Control Release. 2019;295:140–52.PubMedCrossRef
123.
go back to reference Miao L, Li L, Huang Y, Delcassian D. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat Biotechnol. 2019;37:1174–85.PubMedCrossRef Miao L, Li L, Huang Y, Delcassian D. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat Biotechnol. 2019;37:1174–85.PubMedCrossRef
124.
go back to reference Le Moignic A, Malard V, Benvegnu T, Lemiègre L, Berchel M, Jaffrès PA, Baillou C, Delost M, Macedo R, Rochefort J, et al. Preclinical evaluation of mRNA trimannosylated lipopolyplexes as therapeutic cancer vaccines targeting dendritic cells. J Control Release. 2018;278:110–21.PubMedCrossRef Le Moignic A, Malard V, Benvegnu T, Lemiègre L, Berchel M, Jaffrès PA, Baillou C, Delost M, Macedo R, Rochefort J, et al. Preclinical evaluation of mRNA trimannosylated lipopolyplexes as therapeutic cancer vaccines targeting dendritic cells. J Control Release. 2018;278:110–21.PubMedCrossRef
125.
go back to reference Goswami R, Chatzikleanthous D, Lou G. Mannosylation of LNP results in improved potency for self-amplifying RNA (SAM) vaccines. ACS Infect Dis. 2019;5:1546–58.PubMedCrossRef Goswami R, Chatzikleanthous D, Lou G. Mannosylation of LNP results in improved potency for self-amplifying RNA (SAM) vaccines. ACS Infect Dis. 2019;5:1546–58.PubMedCrossRef
126.
go back to reference Wang Y, Zhang L, Xu Z, Miao L, Huang L. mRNA vaccine with antigen-specific checkpoint blockade induces an enhanced immune response against established melanoma. Mol Ther. 2018;26:420–34.PubMedCrossRef Wang Y, Zhang L, Xu Z, Miao L, Huang L. mRNA vaccine with antigen-specific checkpoint blockade induces an enhanced immune response against established melanoma. Mol Ther. 2018;26:420–34.PubMedCrossRef
127.
go back to reference Verbeke R, Lentacker I. Broadening the message: a nanovaccine co-loaded with messenger RNA and α-GalCer induces antitumor immunity through conventional and natural killer T cells. ACS Nano. 2019;13:1655–69.PubMed Verbeke R, Lentacker I. Broadening the message: a nanovaccine co-loaded with messenger RNA and α-GalCer induces antitumor immunity through conventional and natural killer T cells. ACS Nano. 2019;13:1655–69.PubMed
128.
go back to reference Pardi N, Tuyishime S, Muramatsu H, Kariko K, Mui BL, Tam YK, Madden TD, Hope MJ, Weissman D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J Control Release. 2015;217:345–51.PubMedPubMedCentralCrossRef Pardi N, Tuyishime S, Muramatsu H, Kariko K, Mui BL, Tam YK, Madden TD, Hope MJ, Weissman D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J Control Release. 2015;217:345–51.PubMedPubMedCentralCrossRef
129.
go back to reference Fan YN, Li M, Luo YL, Chen Q, Wang L, Zhang HB. Cationic lipid-assisted nanoparticles for delivery of mRNA cancer vaccine. Biomater Sci. 2018;6:3009–18.PubMedCrossRef Fan YN, Li M, Luo YL, Chen Q, Wang L, Zhang HB. Cationic lipid-assisted nanoparticles for delivery of mRNA cancer vaccine. Biomater Sci. 2018;6:3009–18.PubMedCrossRef
130.
go back to reference Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol Ther. 2019;27:710–28.PubMedPubMedCentralCrossRef Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol Ther. 2019;27:710–28.PubMedPubMedCentralCrossRef
131.
go back to reference Dong Y, Dorkin JR, Wang W, Chang PH, Webber MJ, Tang BC, Yang J, Abutbul-Ionita I, Danino D, DeRosa F, et al. Poly(glycoamidoamine) brushes formulated nanomaterials for systemic siRNA and mRNA delivery in vivo. Nano Lett. 2016;16:842–8.PubMedPubMedCentralCrossRef Dong Y, Dorkin JR, Wang W, Chang PH, Webber MJ, Tang BC, Yang J, Abutbul-Ionita I, Danino D, DeRosa F, et al. Poly(glycoamidoamine) brushes formulated nanomaterials for systemic siRNA and mRNA delivery in vivo. Nano Lett. 2016;16:842–8.PubMedPubMedCentralCrossRef
132.
go back to reference Patel AK, Kaczmarek JC, Bose S, Kauffman KJ, Mir F, Heartlein MW, DeRosa F, Langer R, Anderson DG. Inhaled Nanoformulated mRNA polyplexes for protein production in lung epithelium. Adv Mater. 2019;31:e1805116.PubMedPubMedCentralCrossRef Patel AK, Kaczmarek JC, Bose S, Kauffman KJ, Mir F, Heartlein MW, DeRosa F, Langer R, Anderson DG. Inhaled Nanoformulated mRNA polyplexes for protein production in lung epithelium. Adv Mater. 2019;31:e1805116.PubMedPubMedCentralCrossRef
133.
go back to reference Kaczmarek JC, Patel AK, Kauffman KJ, Fenton OS, Webber MJ, Heartlein MW, DeRosa F, Anderson DG. Polymer-lipid nanoparticles for systemic delivery of mRNA to the lungs. Angew Chem Int Ed Engl. 2016;55:13808–12.PubMedPubMedCentralCrossRef Kaczmarek JC, Patel AK, Kauffman KJ, Fenton OS, Webber MJ, Heartlein MW, DeRosa F, Anderson DG. Polymer-lipid nanoparticles for systemic delivery of mRNA to the lungs. Angew Chem Int Ed Engl. 2016;55:13808–12.PubMedPubMedCentralCrossRef
134.
go back to reference Lei C, Cui Y, Zheng L, Chow PK, Wang CH. Development of a gene/drug dual delivery system for brain tumor therapy: potent inhibition via RNA interference and synergistic effects. Biomaterials. 2013;34:7483–94.PubMedCrossRef Lei C, Cui Y, Zheng L, Chow PK, Wang CH. Development of a gene/drug dual delivery system for brain tumor therapy: potent inhibition via RNA interference and synergistic effects. Biomaterials. 2013;34:7483–94.PubMedCrossRef
135.
go back to reference Vaidyanathan S, Orr BG, Banaszak Holl MM. Role of cell membrane-vector interactions in successful gene delivery. Acc Chem Res. 2016;49:1486–93.PubMedCrossRef Vaidyanathan S, Orr BG, Banaszak Holl MM. Role of cell membrane-vector interactions in successful gene delivery. Acc Chem Res. 2016;49:1486–93.PubMedCrossRef
136.
go back to reference Stefan J, Kus K, Wisniewska A, Lorkowska-Zawicka B, Kaminski K, Szczubialka K, Nowakowska M, Korbut R. The antiatherogenic effect of new biocompatible cationically modified polysaccharides: chitosan and pullulan - the comparison study. J Physiol Pharmacol. 2018;69(6). https://doi.org/10.26402/jpp.2018.6.15. Stefan J, Kus K, Wisniewska A, Lorkowska-Zawicka B, Kaminski K, Szczubialka K, Nowakowska M, Korbut R. The antiatherogenic effect of new biocompatible cationically modified polysaccharides: chitosan and pullulan - the comparison study. J Physiol Pharmacol. 2018;69(6). https://​doi.​org/​10.​26402/​jpp.​2018.​6.​15.
137.
go back to reference Li M, Zhao M, Fu Y, Li Y, Gong T, Zhang Z, Sun X. Enhanced intranasal delivery of mRNA vaccine by overcoming the nasal epithelial barrier via intra- and paracellular pathways. J Control Release. 2016;228:9–19.PubMedCrossRef Li M, Zhao M, Fu Y, Li Y, Gong T, Zhang Z, Sun X. Enhanced intranasal delivery of mRNA vaccine by overcoming the nasal epithelial barrier via intra- and paracellular pathways. J Control Release. 2016;228:9–19.PubMedCrossRef
138.
go back to reference Kannan S, Kolhe P, Raykova V, Glibatec M, Kannan RM, Lieh-Lai M, Bassett D. Dynamics of cellular entry and drug delivery by dendritic polymers into human lung epithelial carcinoma cells. J Biomater Sci Polym Ed. 2004;15:311–30.PubMedCrossRef Kannan S, Kolhe P, Raykova V, Glibatec M, Kannan RM, Lieh-Lai M, Bassett D. Dynamics of cellular entry and drug delivery by dendritic polymers into human lung epithelial carcinoma cells. J Biomater Sci Polym Ed. 2004;15:311–30.PubMedCrossRef
139.
go back to reference Chahal JS, Khan OF, Cooper CL, McPartlan JS, Tsosie JK, Tilley LD, Sidik SM, Lourido S. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and toxoplasma gondii challenges with a single dose. Proc Natl Acad Sci U S A. 2016;113:E4133–42.PubMedPubMedCentralCrossRef Chahal JS, Khan OF, Cooper CL, McPartlan JS, Tsosie JK, Tilley LD, Sidik SM, Lourido S. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and toxoplasma gondii challenges with a single dose. Proc Natl Acad Sci U S A. 2016;113:E4133–42.PubMedPubMedCentralCrossRef
140.
141.
go back to reference Islam MA, Xu Y, Tao W. Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat Biomed Eng. 2018;2:850–64.PubMedPubMedCentralCrossRef Islam MA, Xu Y, Tao W. Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat Biomed Eng. 2018;2:850–64.PubMedPubMedCentralCrossRef
142.
go back to reference Yang XZ, Dou S, Sun TM, Mao CQ, Wang HX, Wang J. Systemic delivery of siRNA with cationic lipid assisted PEG-PLA nanoparticles for cancer therapy. J Control Release. 2011;156:203–11.PubMedCrossRef Yang XZ, Dou S, Sun TM, Mao CQ, Wang HX, Wang J. Systemic delivery of siRNA with cationic lipid assisted PEG-PLA nanoparticles for cancer therapy. J Control Release. 2011;156:203–11.PubMedCrossRef
143.
go back to reference Yasar H, Biehl A, De Rossi C, Koch M, Murgia X, Loretz B, Lehr CM. Kinetics of mRNA delivery and protein translation in dendritic cells using lipid-coated PLGA nanoparticles. J Nanobiotechnology. 2018;16:72.PubMedPubMedCentralCrossRef Yasar H, Biehl A, De Rossi C, Koch M, Murgia X, Loretz B, Lehr CM. Kinetics of mRNA delivery and protein translation in dendritic cells using lipid-coated PLGA nanoparticles. J Nanobiotechnology. 2018;16:72.PubMedPubMedCentralCrossRef
144.
go back to reference Sharifnia Z, Bandehpour M, Hamishehkar H, Mosaffa N, Kazemi B, Zarghami N. In-vitro transcribed mRNA delivery using PLGA/PEI nanoparticles into human monocyte-derived dendritic cells. Iran J Pharm Res. 2019;18:1659–75.PubMedPubMedCentral Sharifnia Z, Bandehpour M, Hamishehkar H, Mosaffa N, Kazemi B, Zarghami N. In-vitro transcribed mRNA delivery using PLGA/PEI nanoparticles into human monocyte-derived dendritic cells. Iran J Pharm Res. 2019;18:1659–75.PubMedPubMedCentral
145.
go back to reference Zhao W, Zhang C, Li B, Zhang X, Luo X, Zeng C, Li W, Gao M, Dong Y. Lipid polymer hybrid nanomaterials for mRNA delivery. Cell Mol Bioeng. 2018;11:397–406.PubMedPubMedCentralCrossRef Zhao W, Zhang C, Li B, Zhang X, Luo X, Zeng C, Li W, Gao M, Dong Y. Lipid polymer hybrid nanomaterials for mRNA delivery. Cell Mol Bioeng. 2018;11:397–406.PubMedPubMedCentralCrossRef
146.
go back to reference Qiu Y, Man RCH, Liao Q, Kung KLK, Chow MYT, Lam JKW. Effective mRNA pulmonary delivery by dry powder formulation of PEGylated synthetic KL4 peptide. J Control Release. 2019;314:102–15.PubMedCrossRef Qiu Y, Man RCH, Liao Q, Kung KLK, Chow MYT, Lam JKW. Effective mRNA pulmonary delivery by dry powder formulation of PEGylated synthetic KL4 peptide. J Control Release. 2019;314:102–15.PubMedCrossRef
147.
go back to reference Udhayakumar VK, De Beuckelaer A, McCaffrey J, McCrudden CM, Kirschman JL, Vanover D, Van Hoecke L, Roose K, Deswarte K, De Geest BG, et al. Arginine-rich peptide-based mRNA nanocomplexes efficiently instigate cytotoxic T cell immunity dependent on the amphipathic organization of the peptide. Adv Healthc Mater. 2017;6(13). https://doi.org/10.1002/adhm.201601412. Udhayakumar VK, De Beuckelaer A, McCaffrey J, McCrudden CM, Kirschman JL, Vanover D, Van Hoecke L, Roose K, Deswarte K, De Geest BG, et al. Arginine-rich peptide-based mRNA nanocomplexes efficiently instigate cytotoxic T cell immunity dependent on the amphipathic organization of the peptide. Adv Healthc Mater. 2017;6(13). https://​doi.​org/​10.​1002/​adhm.​201601412.
148.
go back to reference Hoerr I, Obst R, Rammensee HG, Jung G. In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies. Eur J Immunol. 2000;30:1–7.PubMedCrossRef Hoerr I, Obst R, Rammensee HG, Jung G. In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies. Eur J Immunol. 2000;30:1–7.PubMedCrossRef
149.
go back to reference Stitz L, Vogel A, Schnee M, Voss D, Rauch S, Mutzke T, Ketterer T, Kramps T, Petsch B. A thermostable messenger RNA based vaccine against rabies. PLoS Negl Trop Dis. 2017;11:e0006108.PubMedPubMedCentralCrossRef Stitz L, Vogel A, Schnee M, Voss D, Rauch S, Mutzke T, Ketterer T, Kramps T, Petsch B. A thermostable messenger RNA based vaccine against rabies. PLoS Negl Trop Dis. 2017;11:e0006108.PubMedPubMedCentralCrossRef
150.
go back to reference Scheel B, Braedel S, Probst J, Carralot JP, Wagner H, Schild H, Jung G, Rammensee HG, Pascolo S. Immunostimulating capacities of stabilized RNA molecules. Eur J Immunol. 2004;34:537–47.PubMedCrossRef Scheel B, Braedel S, Probst J, Carralot JP, Wagner H, Schild H, Jung G, Rammensee HG, Pascolo S. Immunostimulating capacities of stabilized RNA molecules. Eur J Immunol. 2004;34:537–47.PubMedCrossRef
152.
go back to reference Scheel B, Teufel R, Probst J, Carralot JP, Geginat J, Radsak M, Jarrossay D, Wagner H, Jung G, Rammensee HG, et al. Toll-like receptor-dependent activation of several human blood cell types by protamine-condensed mRNA. Eur J Immunol. 2005;35:1557–66.PubMedCrossRef Scheel B, Teufel R, Probst J, Carralot JP, Geginat J, Radsak M, Jarrossay D, Wagner H, Jung G, Rammensee HG, et al. Toll-like receptor-dependent activation of several human blood cell types by protamine-condensed mRNA. Eur J Immunol. 2005;35:1557–66.PubMedCrossRef
153.
go back to reference Scheel B, Aulwurm S, Probst J, Stitz L, Hoerr I, Rammensee HG, Weller M, Pascolo S. Therapeutic anti-tumor immunity triggered by injections of immunostimulating single-stranded RNA. Eur J Immunol. 2006;36:2807–16.PubMedCrossRef Scheel B, Aulwurm S, Probst J, Stitz L, Hoerr I, Rammensee HG, Weller M, Pascolo S. Therapeutic anti-tumor immunity triggered by injections of immunostimulating single-stranded RNA. Eur J Immunol. 2006;36:2807–16.PubMedCrossRef
154.
go back to reference Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, Dietz K, Pascolo S, Kuczyk M, Rammensee HG, Stenzl A. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate. 2009;69:917–27.PubMedCrossRef Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, Dietz K, Pascolo S, Kuczyk M, Rammensee HG, Stenzl A. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate. 2009;69:917–27.PubMedCrossRef
155.
go back to reference Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, Pawelec G, Hoerr I, Rammensee HG, Garbe C. Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients. J Immunother. 2009;32:498–507.PubMedCrossRef Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, Pawelec G, Hoerr I, Rammensee HG, Garbe C. Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients. J Immunother. 2009;32:498–507.PubMedCrossRef
156.
go back to reference Westdorp H, Creemers JHA, van Oort IM, Schreibelt G, Gorris MAJ, Mehra N, Simons M, de Goede AL, van Rossum MM, Croockewit AJ, et al. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer. J Immunother Cancer. 2019;7:302.PubMedPubMedCentralCrossRef Westdorp H, Creemers JHA, van Oort IM, Schreibelt G, Gorris MAJ, Mehra N, Simons M, de Goede AL, van Rossum MM, Croockewit AJ, et al. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer. J Immunother Cancer. 2019;7:302.PubMedPubMedCentralCrossRef
157.
go back to reference Coolen AL, Lacroix C, Mercier-Gouy P, Delaune E, Monge C, Exposito JY, Verrier B. Poly(lactic acid) nanoparticles and cell-penetrating peptide potentiate mRNA-based vaccine expression in dendritic cells triggering their activation. Biomaterials. 2019;195:23–37.PubMedCrossRef Coolen AL, Lacroix C, Mercier-Gouy P, Delaune E, Monge C, Exposito JY, Verrier B. Poly(lactic acid) nanoparticles and cell-penetrating peptide potentiate mRNA-based vaccine expression in dendritic cells triggering their activation. Biomaterials. 2019;195:23–37.PubMedCrossRef
158.
go back to reference Bell GD, Yang Y, Leung E, Krissansen GW. mRNA transfection by a Xentry-protamine cell-penetrating peptide is enhanced by TLR antagonist E6446. PLoS One. 2018;13:e0201464.PubMedPubMedCentralCrossRef Bell GD, Yang Y, Leung E, Krissansen GW. mRNA transfection by a Xentry-protamine cell-penetrating peptide is enhanced by TLR antagonist E6446. PLoS One. 2018;13:e0201464.PubMedPubMedCentralCrossRef
159.
go back to reference Lou B, De Koker S, Lau CYJ, Hennink WE. mRNA polyplexes with post-conjugated GALA peptides efficiently target, transfect, and activate antigen presenting cells. Bioconjug Chem. 2019;30:461–75.PubMedCrossRef Lou B, De Koker S, Lau CYJ, Hennink WE. mRNA polyplexes with post-conjugated GALA peptides efficiently target, transfect, and activate antigen presenting cells. Bioconjug Chem. 2019;30:461–75.PubMedCrossRef
160.
go back to reference Li W, Ma L, Guo LP, Wang XL, Zhang JW, Bu ZG, Hua RH. West Nile virus infectious replicon particles generated using a packaging-restricted cell line is a safe reporter system. Sci Rep. 2017;7:3286.PubMedPubMedCentralCrossRef Li W, Ma L, Guo LP, Wang XL, Zhang JW, Bu ZG, Hua RH. West Nile virus infectious replicon particles generated using a packaging-restricted cell line is a safe reporter system. Sci Rep. 2017;7:3286.PubMedPubMedCentralCrossRef
161.
go back to reference Erasmus JH, Khandhar AP. An Alphavirus-derived replicon RNA vaccine induces SARS-CoV-2 neutralizing antibody and T cell responses in mice and nonhuman primates. Sci Transl Med. 2020;12:eabc9396. Erasmus JH, Khandhar AP. An Alphavirus-derived replicon RNA vaccine induces SARS-CoV-2 neutralizing antibody and T cell responses in mice and nonhuman primates. Sci Transl Med. 2020;12:eabc9396.
162.
go back to reference White LJ, Sariol CA, Mattocks MD, Wahala MPBW, Yingsiwaphat V, Collier ML, Whitley J, Mikkelsen R, Rodriguez IV, Martinez MI, et al. An alphavirus vector-based tetravalent dengue vaccine induces a rapid and protective immune response in macaques that differs qualitatively from immunity induced by live virus infection. J Virol. 2013;87:3409–24.PubMedPubMedCentralCrossRef White LJ, Sariol CA, Mattocks MD, Wahala MPBW, Yingsiwaphat V, Collier ML, Whitley J, Mikkelsen R, Rodriguez IV, Martinez MI, et al. An alphavirus vector-based tetravalent dengue vaccine induces a rapid and protective immune response in macaques that differs qualitatively from immunity induced by live virus infection. J Virol. 2013;87:3409–24.PubMedPubMedCentralCrossRef
163.
go back to reference Hoang-Le D, Smeenk L, Anraku I, Pijlman GP, Wang XJ, de Vrij J, Liu WJ, Le TT, Schroder WA, Khromykh AA, Suhrbier A. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther. 2009;16:190–9.PubMedCrossRef Hoang-Le D, Smeenk L, Anraku I, Pijlman GP, Wang XJ, de Vrij J, Liu WJ, Le TT, Schroder WA, Khromykh AA, Suhrbier A. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther. 2009;16:190–9.PubMedCrossRef
164.
go back to reference Lundstrom K. Replicon RNA viral vectors as vaccines. Vaccines (Basel). 2016;4(4):39.CrossRef Lundstrom K. Replicon RNA viral vectors as vaccines. Vaccines (Basel). 2016;4(4):39.CrossRef
165.
go back to reference Morrison J, Plotkin S. Chapter 19 – viral vaccines: fighting viruses with vaccines. In: Viral pathogenesis; 2016. p. 253–69.CrossRef Morrison J, Plotkin S. Chapter 19 – viral vaccines: fighting viruses with vaccines. In: Viral pathogenesis; 2016. p. 253–69.CrossRef
166.
go back to reference Fuchs JD, Frank I, Elizaga ML, Allen M, Frahm N, Kochar N, Li S, Edupuganti S, Kalams SA, Tomaras GD, et al. First-in-human evaluation of the safety and immunogenicity of a recombinant vesicular stomatitis virus human immunodeficiency virus-1 gag vaccine (HVTN 090). Open Forum Infect Dis. 2015;2:ofv082.PubMedPubMedCentralCrossRef Fuchs JD, Frank I, Elizaga ML, Allen M, Frahm N, Kochar N, Li S, Edupuganti S, Kalams SA, Tomaras GD, et al. First-in-human evaluation of the safety and immunogenicity of a recombinant vesicular stomatitis virus human immunodeficiency virus-1 gag vaccine (HVTN 090). Open Forum Infect Dis. 2015;2:ofv082.PubMedPubMedCentralCrossRef
167.
go back to reference Tanaka H, Oasa S, Kinjo M, Tange K, Nakai Y, Harashima H, Akita H. Temperature and pH sensitivity of a stabilized self-nanoemulsion formed using an ionizable lipid-like material via an oil-to-surfactant transition. Colloids Surf B Biointerfaces. 2017;151:95–101.PubMedCrossRef Tanaka H, Oasa S, Kinjo M, Tange K, Nakai Y, Harashima H, Akita H. Temperature and pH sensitivity of a stabilized self-nanoemulsion formed using an ionizable lipid-like material via an oil-to-surfactant transition. Colloids Surf B Biointerfaces. 2017;151:95–101.PubMedCrossRef
168.
go back to reference Samsa MM, Dupuy LC, Beard CW, Six CM, Schmaljohn CS, Mason PW, Geall AJ, Ulmer JB, Yu D. Self-amplifying RNA vaccines for Venezuelan equine encephalitis virus induce robust protective immunogenicity in mice. Mol Ther. 2019;27:850–65.PubMedPubMedCentralCrossRef Samsa MM, Dupuy LC, Beard CW, Six CM, Schmaljohn CS, Mason PW, Geall AJ, Ulmer JB, Yu D. Self-amplifying RNA vaccines for Venezuelan equine encephalitis virus induce robust protective immunogenicity in mice. Mol Ther. 2019;27:850–65.PubMedPubMedCentralCrossRef
169.
go back to reference Luisi K, Morabito KM. Development of a potent Zika virus vaccine using self-amplifying messenger. RNA. 2020;6:eaba5068. Luisi K, Morabito KM. Development of a potent Zika virus vaccine using self-amplifying messenger. RNA. 2020;6:eaba5068.
170.
go back to reference Diken M, Kreiter S, Selmi A, Britten CM, Huber C, Türeci Ö, Sahin U. Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation. Gene Ther. 2011;18:702–8.PubMedCrossRef Diken M, Kreiter S, Selmi A, Britten CM, Huber C, Türeci Ö, Sahin U. Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation. Gene Ther. 2011;18:702–8.PubMedCrossRef
171.
go back to reference Selmi A, Vascotto F, Kautz-Neu K, Türeci Ö, Sahin U, von Stebut E, Diken M, Kreiter S. Uptake of synthetic naked RNA by skin-resident dendritic cells via macropinocytosis allows antigen expression and induction of T-cell responses in mice. Cancer Immunol Immunother. 2016;65:1075–83.PubMedCrossRef Selmi A, Vascotto F, Kautz-Neu K, Türeci Ö, Sahin U, von Stebut E, Diken M, Kreiter S. Uptake of synthetic naked RNA by skin-resident dendritic cells via macropinocytosis allows antigen expression and induction of T-cell responses in mice. Cancer Immunol Immunother. 2016;65:1075–83.PubMedCrossRef
172.
go back to reference Brachet J, Huez G, Hubert E. Microinjection of rabbit hemoglobin messenger RNA into amphibian oocytes and embryos. Proc Natl Acad Sci U S A. 1973;70:543–7.PubMedPubMedCentralCrossRef Brachet J, Huez G, Hubert E. Microinjection of rabbit hemoglobin messenger RNA into amphibian oocytes and embryos. Proc Natl Acad Sci U S A. 1973;70:543–7.PubMedPubMedCentralCrossRef
173.
go back to reference Gallie DR. The cap and poly(A) tail function synergistically to regulate mRNA translational efficiency. Genes Dev. 1991;5:2108–16.PubMedCrossRef Gallie DR. The cap and poly(A) tail function synergistically to regulate mRNA translational efficiency. Genes Dev. 1991;5:2108–16.PubMedCrossRef
174.
go back to reference Stewart MP, Langer R. Intracellular delivery by membrane disruption: mechanisms. Strateg Concepts. 2018;118:7409–531. Stewart MP, Langer R. Intracellular delivery by membrane disruption: mechanisms. Strateg Concepts. 2018;118:7409–531.
175.
go back to reference Ringer S. Regarding the action of hydrate of soda, hydrate of ammonia, and hydrate of potash on the ventricle of the frog’s heart. J Physiol. 1882;3:195–202.196.PubMedPubMedCentralCrossRef Ringer S. Regarding the action of hydrate of soda, hydrate of ammonia, and hydrate of potash on the ventricle of the frog’s heart. J Physiol. 1882;3:195–202.196.PubMedPubMedCentralCrossRef
176.
177.
go back to reference Probst J, Weide B, Scheel B, Pichler BJ, Hoerr I, Rammensee HG, Pascolo S. Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent. Gene Ther. 2007;14:1175–80.PubMedCrossRef Probst J, Weide B, Scheel B, Pichler BJ, Hoerr I, Rammensee HG, Pascolo S. Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent. Gene Ther. 2007;14:1175–80.PubMedCrossRef
178.
go back to reference Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Löwer M, Bukur V, Tadmor AD, Luxemburger U, Schrörs B, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–6.CrossRefPubMed Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Löwer M, Bukur V, Tadmor AD, Luxemburger U, Schrörs B, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–6.CrossRefPubMed
179.
go back to reference Edwards DK, Jasny E, Yoon H, Horscroft N, Schanen B, Geter T, Fotin-Mleczek M, Petsch B, Wittman V. Adjuvant effects of a sequence-engineered mRNA vaccine: translational profiling demonstrates similar human and murine innate response. J Transl Med. 2017;15:1.PubMedPubMedCentralCrossRef Edwards DK, Jasny E, Yoon H, Horscroft N, Schanen B, Geter T, Fotin-Mleczek M, Petsch B, Wittman V. Adjuvant effects of a sequence-engineered mRNA vaccine: translational profiling demonstrates similar human and murine innate response. J Transl Med. 2017;15:1.PubMedPubMedCentralCrossRef
180.
go back to reference Kreiter S, Diken M, Selmi A, Diekmann J, Attig S, Hüsemann Y, Koslowski M, Huber C, Türeci Ö, Sahin U. FLT3 ligand enhances the cancer therapeutic potency of naked RNA vaccines. Cancer Res. 2011;71:6132–42.PubMedCrossRef Kreiter S, Diken M, Selmi A, Diekmann J, Attig S, Hüsemann Y, Koslowski M, Huber C, Türeci Ö, Sahin U. FLT3 ligand enhances the cancer therapeutic potency of naked RNA vaccines. Cancer Res. 2011;71:6132–42.PubMedCrossRef
181.
go back to reference Van Lint S, Goyvaerts C, Maenhout S, Goethals L, Disy A, Benteyn D, Pen J, Bonehill A, Heirman C, Breckpot K, Thielemans K. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res. 2012;72:1661–71.PubMedCrossRef Van Lint S, Goyvaerts C, Maenhout S, Goethals L, Disy A, Benteyn D, Pen J, Bonehill A, Heirman C, Breckpot K, Thielemans K. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res. 2012;72:1661–71.PubMedCrossRef
182.
go back to reference Hewitt SL, Bailey D, Zielinski J, Apte A, Musenge F. Intratumoral interleukin-12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin Cancer Res. 2020;26:6284–98.PubMedCrossRef Hewitt SL, Bailey D, Zielinski J, Apte A, Musenge F. Intratumoral interleukin-12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin Cancer Res. 2020;26:6284–98.PubMedCrossRef
183.
go back to reference Van der Jeught K, Joe PT, Bialkowski L, Heirman C, Daszkiewicz L, Liechtenstein T, Escors D, Thielemans K, Breckpot K. Intratumoral administration of mRNA encoding a fusokine consisting of IFN-β and the ectodomain of the TGF-β receptor II potentiates antitumor immunity. Oncotarget. 2014;5:10100–13.PubMedPubMedCentralCrossRef Van der Jeught K, Joe PT, Bialkowski L, Heirman C, Daszkiewicz L, Liechtenstein T, Escors D, Thielemans K, Breckpot K. Intratumoral administration of mRNA encoding a fusokine consisting of IFN-β and the ectodomain of the TGF-β receptor II potentiates antitumor immunity. Oncotarget. 2014;5:10100–13.PubMedPubMedCentralCrossRef
184.
go back to reference Van Lint S, Renmans D, Broos K, Goethals L, Maenhout S, Benteyn D, Goyvaerts C, Du Four S, Van der Jeught K, Bialkowski L, et al. Intratumoral delivery of TriMix mRNA results in T-cell activation by cross-presenting dendritic cells. Cancer Immunol Res. 2016;4:146–56.PubMedCrossRef Van Lint S, Renmans D, Broos K, Goethals L, Maenhout S, Benteyn D, Goyvaerts C, Du Four S, Van der Jeught K, Bialkowski L, et al. Intratumoral delivery of TriMix mRNA results in T-cell activation by cross-presenting dendritic cells. Cancer Immunol Res. 2016;4:146–56.PubMedCrossRef
186.
go back to reference Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol. 2014;14:719–30.PubMedCrossRef Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol. 2014;14:719–30.PubMedCrossRef
187.
go back to reference Ding Y, Guo Z, Liu Y, Li X, Zhang Q, Xu X, Gu Y, Zhang Y, Zhao D, Cao X. The lectin Siglec-G inhibits dendritic cell cross-presentation by impairing MHC class I-peptide complex formation. Nat Immunol. 2016;17:1167–75.PubMedCrossRef Ding Y, Guo Z, Liu Y, Li X, Zhang Q, Xu X, Gu Y, Zhang Y, Zhao D, Cao X. The lectin Siglec-G inhibits dendritic cell cross-presentation by impairing MHC class I-peptide complex formation. Nat Immunol. 2016;17:1167–75.PubMedCrossRef
188.
go back to reference Qiao J, Liu Z, Dong C, Luan Y, Zhang A, Moore C, Fu K, Peng J, Wang Y, Ren Z, et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8(+) T cell apoptosis. Cancer Cell. 2019;35:901–915.e904.PubMedCrossRef Qiao J, Liu Z, Dong C, Luan Y, Zhang A, Moore C, Fu K, Peng J, Wang Y, Ren Z, et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8(+) T cell apoptosis. Cancer Cell. 2019;35:901–915.e904.PubMedCrossRef
189.
go back to reference Inaba K, Metlay JP, Crowley MT, Steinman RM. Dendritic cells pulsed with protein antigens in vitro can prime antigen-specific, MHC-restricted T cells in situ. J Exp Med. 1990;172:631–40.PubMedCrossRef Inaba K, Metlay JP, Crowley MT, Steinman RM. Dendritic cells pulsed with protein antigens in vitro can prime antigen-specific, MHC-restricted T cells in situ. J Exp Med. 1990;172:631–40.PubMedCrossRef
190.
go back to reference De Temmerman ML, Dewitte H, Vandenbroucke RE, Lucas B, Libert C, Demeester J, De Smedt SC, Lentacker I, Rejman J. mRNA-Lipoplex loaded microbubble contrast agents for ultrasound-assisted transfection of dendritic cells. Biomaterials. 2011;32:9128–35.PubMedCrossRef De Temmerman ML, Dewitte H, Vandenbroucke RE, Lucas B, Libert C, Demeester J, De Smedt SC, Lentacker I, Rejman J. mRNA-Lipoplex loaded microbubble contrast agents for ultrasound-assisted transfection of dendritic cells. Biomaterials. 2011;32:9128–35.PubMedCrossRef
191.
go back to reference Tuyaerts S, Noppe SM, Corthals J, Breckpot K, Heirman C, De Greef C, Van Riet I, Thielemans K. Generation of large numbers of dendritic cells in a closed system using cell factories. J Immunol Methods. 2002;264:135–51.PubMedCrossRef Tuyaerts S, Noppe SM, Corthals J, Breckpot K, Heirman C, De Greef C, Van Riet I, Thielemans K. Generation of large numbers of dendritic cells in a closed system using cell factories. J Immunol Methods. 2002;264:135–51.PubMedCrossRef
192.
go back to reference Ahmed R, Sayegh N, Graciotti M, Kandalaft LE. Electroporation as a method of choice to generate genetically modified dendritic cell cancer vaccines. Curr Opin Biotechnol. 2020;65:142–55.PubMedCrossRef Ahmed R, Sayegh N, Graciotti M, Kandalaft LE. Electroporation as a method of choice to generate genetically modified dendritic cell cancer vaccines. Curr Opin Biotechnol. 2020;65:142–55.PubMedCrossRef
193.
go back to reference Hiasa M, Abe M, Nakano A, Oda A, Amou H, Kido S, Takeuchi K, Kagawa K, Yata K, Hashimoto T, et al. GM-CSF and IL-4 induce dendritic cell differentiation and disrupt osteoclastogenesis through M-CSF receptor shedding by up-regulation of TNF-alpha converting enzyme (TACE). Blood. 2009;114:4517–26.PubMedCrossRef Hiasa M, Abe M, Nakano A, Oda A, Amou H, Kido S, Takeuchi K, Kagawa K, Yata K, Hashimoto T, et al. GM-CSF and IL-4 induce dendritic cell differentiation and disrupt osteoclastogenesis through M-CSF receptor shedding by up-regulation of TNF-alpha converting enzyme (TACE). Blood. 2009;114:4517–26.PubMedCrossRef
194.
go back to reference Fernández-Delgado I, Calzada-Fraile D. Immune regulation by dendritic cell extracellular vesicles in cancer immunotherapy and vaccines. Cancers (Basel). 2020;12. Fernández-Delgado I, Calzada-Fraile D. Immune regulation by dendritic cell extracellular vesicles in cancer immunotherapy and vaccines. Cancers (Basel). 2020;12.
195.
go back to reference Carreno BM, Becker-Hapak M, Huang A, Chan M, Alyasiry A, Lie WR, Aft RL, Cornelius LA, Trinkaus KM, Linette GP. IL-12p70-producing patient DC vaccine elicits Tc1-polarized immunity. J Clin Invest. 2013;123:3383–94.PubMedPubMedCentralCrossRef Carreno BM, Becker-Hapak M, Huang A, Chan M, Alyasiry A, Lie WR, Aft RL, Cornelius LA, Trinkaus KM, Linette GP. IL-12p70-producing patient DC vaccine elicits Tc1-polarized immunity. J Clin Invest. 2013;123:3383–94.PubMedPubMedCentralCrossRef
196.
go back to reference Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol. 2005;6:769–76.PubMedPubMedCentralCrossRef Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol. 2005;6:769–76.PubMedPubMedCentralCrossRef
197.
go back to reference Dewitte H, Van Lint S, Heirman C, Thielemans K, De Smedt SC, Breckpot K, Lentacker I. The potential of antigen and TriMix sonoporation using mRNA-loaded microbubbles for ultrasound-triggered cancer immunotherapy. J Control Release. 2014;194:28–36.PubMedCrossRef Dewitte H, Van Lint S, Heirman C, Thielemans K, De Smedt SC, Breckpot K, Lentacker I. The potential of antigen and TriMix sonoporation using mRNA-loaded microbubbles for ultrasound-triggered cancer immunotherapy. J Control Release. 2014;194:28–36.PubMedCrossRef
198.
go back to reference Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534:396–401.PubMedCrossRef Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534:396–401.PubMedCrossRef
199.
go back to reference Gandhi RT, Kwon DS, Macklin EA, Shopis JR, McLean AP, McBrine N, Flynn T, Peter L, Sbrolla A, Kaufmann DE, et al. Immunization of HIV-1-infected persons with autologous dendritic cells transfected with mRNA encoding HIV-1 gag and Nef: results of a randomized, placebo-controlled clinical trial. J Acquir Immune Defic Syndr. 2016;71:246–53.PubMedPubMedCentralCrossRef Gandhi RT, Kwon DS, Macklin EA, Shopis JR, McLean AP, McBrine N, Flynn T, Peter L, Sbrolla A, Kaufmann DE, et al. Immunization of HIV-1-infected persons with autologous dendritic cells transfected with mRNA encoding HIV-1 gag and Nef: results of a randomized, placebo-controlled clinical trial. J Acquir Immune Defic Syndr. 2016;71:246–53.PubMedPubMedCentralCrossRef
200.
go back to reference Jacobson JM, Routy JP, Welles S, DeBenedette M, Tcherepanova I, Angel JB, Asmuth DM, Stein DK, Baril JG, McKellar M, et al. Dendritic cell immunotherapy for HIV-1 infection using autologous HIV-1 RNA: a randomized, double-blind, placebo-controlled clinical trial. J Acquir Immune Defic Syndr. 2016;72:31–8.PubMedPubMedCentralCrossRef Jacobson JM, Routy JP, Welles S, DeBenedette M, Tcherepanova I, Angel JB, Asmuth DM, Stein DK, Baril JG, McKellar M, et al. Dendritic cell immunotherapy for HIV-1 infection using autologous HIV-1 RNA: a randomized, double-blind, placebo-controlled clinical trial. J Acquir Immune Defic Syndr. 2016;72:31–8.PubMedPubMedCentralCrossRef
201.
go back to reference Gay CL, DeBenedette MA, Tcherepanova IY, Gamble A, Lewis WE, Cope AB, Kuruc JD, McGee KS, Kearney MF, Coffin JM, et al. Immunogenicity of AGS-004 dendritic cell therapy in patients treated during acute HIV infection. AIDS Res Hum Retrovir. 2018;34:111–22.PubMedCrossRefPubMedCentral Gay CL, DeBenedette MA, Tcherepanova IY, Gamble A, Lewis WE, Cope AB, Kuruc JD, McGee KS, Kearney MF, Coffin JM, et al. Immunogenicity of AGS-004 dendritic cell therapy in patients treated during acute HIV infection. AIDS Res Hum Retrovir. 2018;34:111–22.PubMedCrossRefPubMedCentral
202.
go back to reference Derdelinckx J, Mansilla MJ, De Laere M, Lee WP, Navarro-Barriuso J, Wens I, Nkansah I, Daans J, De Reu H, Jolanta Keliris A, et al. Clinical and immunological control of experimental autoimmune encephalomyelitis by tolerogenic dendritic cells loaded with MOG-encoding mRNA. J Neuroinflammation. 2019;16:167.PubMedPubMedCentralCrossRef Derdelinckx J, Mansilla MJ, De Laere M, Lee WP, Navarro-Barriuso J, Wens I, Nkansah I, Daans J, De Reu H, Jolanta Keliris A, et al. Clinical and immunological control of experimental autoimmune encephalomyelitis by tolerogenic dendritic cells loaded with MOG-encoding mRNA. J Neuroinflammation. 2019;16:167.PubMedPubMedCentralCrossRef
203.
go back to reference Stewart MP, Sharei A, Ding X, Sahay G, Langer R, Jensen KF. In vitro and ex vivo strategies for intracellular delivery. Nature. 2016;538:183–92.PubMedCrossRef Stewart MP, Sharei A, Ding X, Sahay G, Langer R, Jensen KF. In vitro and ex vivo strategies for intracellular delivery. Nature. 2016;538:183–92.PubMedCrossRef
204.
go back to reference Derdelinckx J, Berneman ZN, Cools N. GMP-grade mRNA electroporation of dendritic cells for clinical use. Methods Mol Biol. 2016;1428:139–50.PubMedCrossRef Derdelinckx J, Berneman ZN, Cools N. GMP-grade mRNA electroporation of dendritic cells for clinical use. Methods Mol Biol. 2016;1428:139–50.PubMedCrossRef
205.
go back to reference Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med. 1996;184:465–72.PubMedCrossRef Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med. 1996;184:465–72.PubMedCrossRef
206.
go back to reference De Keersmaecker B, Heirman C, Corthals J, Empsen C, van Grunsven LA, Allard SD, Pen J, Lacor P, Thielemans K, Aerts JL. The combination of 4-1BBL and CD40L strongly enhances the capacity of dendritic cells to stimulate HIV-specific T cell responses. J Leukoc Biol. 2011;89:989–99.PubMedCrossRef De Keersmaecker B, Heirman C, Corthals J, Empsen C, van Grunsven LA, Allard SD, Pen J, Lacor P, Thielemans K, Aerts JL. The combination of 4-1BBL and CD40L strongly enhances the capacity of dendritic cells to stimulate HIV-specific T cell responses. J Leukoc Biol. 2011;89:989–99.PubMedCrossRef
207.
go back to reference Aerts-Toegaert C, Heirman C, Tuyaerts S, Corthals J, Aerts JL, Bonehill A, Thielemans K, Breckpot K. CD83 expression on dendritic cells and T cells: correlation with effective immune responses. Eur J Immunol. 2007;37:686–95.PubMedCrossRef Aerts-Toegaert C, Heirman C, Tuyaerts S, Corthals J, Aerts JL, Bonehill A, Thielemans K, Breckpot K. CD83 expression on dendritic cells and T cells: correlation with effective immune responses. Eur J Immunol. 2007;37:686–95.PubMedCrossRef
208.
go back to reference Dannull J, Nair S, Su Z, Boczkowski D, DeBeck C, Yang B, Gilboa E, Vieweg J. Enhancing the immunostimulatory function of dendritic cells by transfection with mRNA encoding OX40 ligand. Blood. 2005;105:3206–13.PubMedCrossRef Dannull J, Nair S, Su Z, Boczkowski D, DeBeck C, Yang B, Gilboa E, Vieweg J. Enhancing the immunostimulatory function of dendritic cells by transfection with mRNA encoding OX40 ligand. Blood. 2005;105:3206–13.PubMedCrossRef
209.
go back to reference Met O, Balslev E, Flyger H, Svane IM. High immunogenic potential of p53 mRNA-transfected dendritic cells in patients with primary breast cancer. Breast Cancer Res Treat. 2011;125:395–406.PubMedCrossRef Met O, Balslev E, Flyger H, Svane IM. High immunogenic potential of p53 mRNA-transfected dendritic cells in patients with primary breast cancer. Breast Cancer Res Treat. 2011;125:395–406.PubMedCrossRef
210.
go back to reference Do ASS, Amano T, Edwards LA, Zhang L, De Peralta-Venturina M, Yu JS. CD133 mRNA-loaded dendritic cell vaccination abrogates glioma stem cell propagation in humanized glioblastoma mouse model. Mol Ther Oncolytics. 2020;18:295–303.PubMedPubMedCentralCrossRef Do ASS, Amano T, Edwards LA, Zhang L, De Peralta-Venturina M, Yu JS. CD133 mRNA-loaded dendritic cell vaccination abrogates glioma stem cell propagation in humanized glioblastoma mouse model. Mol Ther Oncolytics. 2020;18:295–303.PubMedPubMedCentralCrossRef
211.
go back to reference Minkis K, Kavanagh DG, Alter G, Bogunovic D, O'Neill D, Adams S, Pavlick A, Walker BD, Brockman MA, Gandhi RT, Bhardwaj N. Type 2 bias of T cells expanded from the blood of melanoma patients switched to type 1 by IL-12p70 mRNA-transfected dendritic cells. Cancer Res. 2008;68:9441–50.PubMedPubMedCentralCrossRef Minkis K, Kavanagh DG, Alter G, Bogunovic D, O'Neill D, Adams S, Pavlick A, Walker BD, Brockman MA, Gandhi RT, Bhardwaj N. Type 2 bias of T cells expanded from the blood of melanoma patients switched to type 1 by IL-12p70 mRNA-transfected dendritic cells. Cancer Res. 2008;68:9441–50.PubMedPubMedCentralCrossRef
212.
go back to reference Bontkes HJ, Kramer D, Ruizendaal JJ, Meijer CJ, Hooijberg E. Tumor associated antigen and interleukin-12 mRNA transfected dendritic cells enhance effector function of natural killer cells and antigen specific T-cells. Clin Immunol. 2008;127:375–84.PubMedCrossRef Bontkes HJ, Kramer D, Ruizendaal JJ, Meijer CJ, Hooijberg E. Tumor associated antigen and interleukin-12 mRNA transfected dendritic cells enhance effector function of natural killer cells and antigen specific T-cells. Clin Immunol. 2008;127:375–84.PubMedCrossRef
213.
go back to reference Coosemans A, Tuyaerts S, Morias K, Corthals J, Heirman C, Thielemans K, Van Gool SW, Vergote I, Amant F. mRNA electroporation of dendritic cells with WT1, survivin, and TriMix (a mixture of caTLR4, CD40L, and CD70). Methods Mol Biol. 2016;1428:277–83.PubMedCrossRef Coosemans A, Tuyaerts S, Morias K, Corthals J, Heirman C, Thielemans K, Van Gool SW, Vergote I, Amant F. mRNA electroporation of dendritic cells with WT1, survivin, and TriMix (a mixture of caTLR4, CD40L, and CD70). Methods Mol Biol. 2016;1428:277–83.PubMedCrossRef
214.
go back to reference Bonehill A, Tuyaerts S, Van Nuffel AM, Heirman C, Bos TJ, Fostier K, Neyns B, Thielemans K. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Mol Ther. 2008;16:1170–80.PubMedCrossRef Bonehill A, Tuyaerts S, Van Nuffel AM, Heirman C, Bos TJ, Fostier K, Neyns B, Thielemans K. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Mol Ther. 2008;16:1170–80.PubMedCrossRef
215.
go back to reference Pen JJ, De Keersmaecker B, Maenhout SK, Van Nuffel AM, Heirman C, Corthals J, Escors D, Bonehill A, Thielemans K, Breckpot K, Aerts JL. Modulation of regulatory T cell function by monocyte-derived dendritic cells matured through electroporation with mRNA encoding CD40 ligand, constitutively active TLR4, and CD70. J Immunol. 2013;191:1976–83.PubMedCrossRef Pen JJ, De Keersmaecker B, Maenhout SK, Van Nuffel AM, Heirman C, Corthals J, Escors D, Bonehill A, Thielemans K, Breckpot K, Aerts JL. Modulation of regulatory T cell function by monocyte-derived dendritic cells matured through electroporation with mRNA encoding CD40 ligand, constitutively active TLR4, and CD70. J Immunol. 2013;191:1976–83.PubMedCrossRef
216.
go back to reference Wilgenhof S, Van Nuffel AM, Benteyn D, Corthals J, Aerts C, Heirman C, Van Riet I, Bonehill A, Thielemans K, Neyns B. A phase IB study on intravenous synthetic mRNA electroporated dendritic cell immunotherapy in pretreated advanced melanoma patients. Ann Oncol. 2013;24:2686–93.PubMedCrossRef Wilgenhof S, Van Nuffel AM, Benteyn D, Corthals J, Aerts C, Heirman C, Van Riet I, Bonehill A, Thielemans K, Neyns B. A phase IB study on intravenous synthetic mRNA electroporated dendritic cell immunotherapy in pretreated advanced melanoma patients. Ann Oncol. 2013;24:2686–93.PubMedCrossRef
217.
go back to reference Zeng C, Hou X, Yan J, Zhang C, Li W, Zhao W, Du S, Dong Y. Leveraging mRNA sequences and nanoparticles to deliver SARS-CoV-2 antigens in vivo. Adv Mater. 2020;32:e2004452.PubMedCrossRefPubMedCentral Zeng C, Hou X, Yan J, Zhang C, Li W, Zhao W, Du S, Dong Y. Leveraging mRNA sequences and nanoparticles to deliver SARS-CoV-2 antigens in vivo. Adv Mater. 2020;32:e2004452.PubMedCrossRefPubMedCentral
218.
go back to reference Lu J, Lu G, Tan S, Xia J, Xiong H, Yu X. A COVID-19 mRNA vaccine encoding SARS-CoV-2 virus-like particles induces a strong antiviral-like immune response in mice. Cell Res. 2020;30:936–9.PubMedCrossRef Lu J, Lu G, Tan S, Xia J, Xiong H, Yu X. A COVID-19 mRNA vaccine encoding SARS-CoV-2 virus-like particles induces a strong antiviral-like immune response in mice. Cell Res. 2020;30:936–9.PubMedCrossRef
219.
go back to reference Zhang NN, Li XF, Deng YQ, Zhao H, Huang YJ, Yang G, Huang WJ, Gao P, Zhou C, Zhang RR, et al. A thermostable mRNA vaccine against COVID-19. Cell. 2020;182:1271–1283.e1216.PubMedPubMedCentralCrossRef Zhang NN, Li XF, Deng YQ, Zhao H, Huang YJ, Yang G, Huang WJ, Gao P, Zhou C, Zhang RR, et al. A thermostable mRNA vaccine against COVID-19. Cell. 2020;182:1271–1283.e1216.PubMedPubMedCentralCrossRef
220.
go back to reference Mulligan MJ, Lyke KE, Kitchin N, Absalon J. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–93.PubMedCrossRef Mulligan MJ, Lyke KE, Kitchin N, Absalon J. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–93.PubMedCrossRef
221.
go back to reference Mulligan MJ, Lyke KE, Kitchin N, Absalon J. Phase 1/2 study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–93.PubMedCrossRef Mulligan MJ, Lyke KE, Kitchin N, Absalon J. Phase 1/2 study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–93.PubMedCrossRef
222.
go back to reference Sahin U, Muik A. COVID-19 vaccine BNT162b1 elicits human antibody and T(H)1 T cell responses. Nature. 2020;586:594–9.CrossRefPubMed Sahin U, Muik A. COVID-19 vaccine BNT162b1 elicits human antibody and T(H)1 T cell responses. Nature. 2020;586:594–9.CrossRefPubMed
223.
224.
go back to reference Maruggi G, Chiarot E, Giovani C, Buccato S, Bonacci S, Frigimelica E, Margarit I, Geall A, Bensi G, Maione D. Immunogenicity and protective efficacy induced by self-amplifying mRNA vaccines encoding bacterial antigens. Vaccine. 2017;35:361–8.PubMedCrossRef Maruggi G, Chiarot E, Giovani C, Buccato S, Bonacci S, Frigimelica E, Margarit I, Geall A, Bensi G, Maione D. Immunogenicity and protective efficacy induced by self-amplifying mRNA vaccines encoding bacterial antigens. Vaccine. 2017;35:361–8.PubMedCrossRef
225.
go back to reference Baeza Garcia A, Siu E, Sun T, Exler V, Brito L, Hekele A, Otten G, Augustijn K, Janse CJ, Ulmer JB, et al. Neutralization of the plasmodium-encoded MIF ortholog confers protective immunity against malaria infection. Nat Commun. 2018;9:2714.PubMedPubMedCentralCrossRef Baeza Garcia A, Siu E, Sun T, Exler V, Brito L, Hekele A, Otten G, Augustijn K, Janse CJ, Ulmer JB, et al. Neutralization of the plasmodium-encoded MIF ortholog confers protective immunity against malaria infection. Nat Commun. 2018;9:2714.PubMedPubMedCentralCrossRef
Metadata
Title
mRNA vaccine: a potential therapeutic strategy
Authors
Yang Wang
Ziqi Zhang
Jingwen Luo
Xuejiao Han
Yuquan Wei
Xiawei Wei
Publication date
01-12-2021
Publisher
BioMed Central
Keyword
COVID-19
Published in
Molecular Cancer / Issue 1/2021
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-021-01311-z

Other articles of this Issue 1/2021

Molecular Cancer 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine