Formulation, inflammation, and RNA sensing impact the immunogenicity of self-amplifying RNA vaccines
To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects...
Saved in:
Published in | Molecular therapy. Nucleic acids Vol. 31; pp. 29 - 42 |
---|---|
Main Authors | , , , , , , , , , , , , , |
Format | Journal Article |
Language | English |
Published |
United States
Elsevier Inc
14.03.2023
American Society of Gene & Cell Therapy Elsevier |
Subjects | |
Online Access | Get full text |
Cover
Loading…
Abstract | To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response.
[Display omitted]
RNA vaccines were a vital part of the pandemic response. But further research is required to broaden their usage. In the current study, we investigated the interplay of RNA vaccine-induced inflammation and the downstream adaptive response. We show that acute inflammation after immunization is required for protective antibody responses. |
---|---|
AbstractList | To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response. To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response.To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response. To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response. [Display omitted] RNA vaccines were a vital part of the pandemic response. But further research is required to broaden their usage. In the current study, we investigated the interplay of RNA vaccine-induced inflammation and the downstream adaptive response. We show that acute inflammation after immunization is required for protective antibody responses. To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response. RNA vaccines were a vital part of the pandemic response. But further research is required to broaden their usage. In the current study, we investigated the interplay of RNA vaccine-induced inflammation and the downstream adaptive response. We show that acute inflammation after immunization is required for protective antibody responses. To be effective, RNA vaccines require both translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response. |
Author | Fox, Christopher B. Stirling, David C. Flight, Katie E. Beattie, Mitchell Blakney, Anna K. Brown, Jonathan C. Tregoning, John S. McKay, Paul F. Murugaiah, Valarmathy Tam, Ying K. Cunliffe, Robert F. Shattock, Robin J. Johansson, Cecilia Wang, Ziyin |
Author_xml | – sequence: 1 givenname: John S. orcidid: 0000-0001-8093-8741 surname: Tregoning fullname: Tregoning, John S. email: john.tregoning@imperial.ac.uk organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 2 givenname: David C. surname: Stirling fullname: Stirling, David C. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 3 givenname: Ziyin surname: Wang fullname: Wang, Ziyin organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 4 givenname: Katie E. surname: Flight fullname: Flight, Katie E. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 5 givenname: Jonathan C. surname: Brown fullname: Brown, Jonathan C. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 6 givenname: Anna K. surname: Blakney fullname: Blakney, Anna K. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 7 givenname: Paul F. surname: McKay fullname: McKay, Paul F. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 8 givenname: Robert F. surname: Cunliffe fullname: Cunliffe, Robert F. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 9 givenname: Valarmathy surname: Murugaiah fullname: Murugaiah, Valarmathy organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK – sequence: 10 givenname: Christopher B. surname: Fox fullname: Fox, Christopher B. organization: IDRI, Seattle, WA, USA – sequence: 11 givenname: Mitchell surname: Beattie fullname: Beattie, Mitchell organization: Acuitas Therapeutics, 6190 Agronomy Road, Ste 405, Vancouver, BC, Canada – sequence: 12 givenname: Ying K. surname: Tam fullname: Tam, Ying K. organization: Acuitas Therapeutics, 6190 Agronomy Road, Ste 405, Vancouver, BC, Canada – sequence: 13 givenname: Cecilia surname: Johansson fullname: Johansson, Cecilia organization: National Heart and Lung Institute, Imperial College London, St. Mary’s Campus, London, UK – sequence: 14 givenname: Robin J. surname: Shattock fullname: Shattock, Robin J. organization: Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK |
BackLink | https://www.ncbi.nlm.nih.gov/pubmed/36589712$$D View this record in MEDLINE/PubMed |
BookMark | eNp9kU9r3DAQxUVJadI0X6CH4mMPtauRZdmGUgih-QOhhZK7kKXRRostbSV7Yb99tN1tSC7VRSPpvZ-Gee_JiQ8eCfkItAIK4uu6CtPsK0YZqwAqyvgbcsZAsJI1NZy8qE_JRUprmpegwAR7R05r0XR9C-yMmOsQp2VUswv-S-G8HdU0HU_Km-L3z8sioU_Orwo3bZSei_kRczktPqzQO-3mXRFsFo22VNNmdHa3F--NW6W185g-kLdWjQkvjvs5ebj-8XB1W97_urm7urwvddO1c2laYRgow3mrG1HXrVW16FB3ONjBdgYM7W3bKN43QwvCmLbrm6ZByqGv9VCfk7sD1gS1lpvoJhV3Mign_16EuJIqzk6PKKkRquZWWUUNF7wbGABo4DUfLBUDz6zvB9ZmGSY0Gv0c1fgK-vrFu0e5ClvZtz3vqciAz0dADH8WTLOcXNI4jspjWJJkbU5DdD2DLGUHqY4hpYj2-Rugch-2zM3nsOU-bAkgc9jZ9Ollg8-Wf9FmwbeDAPPEtw6jTNqh12hcRD3nmbj_8Z8AzfW-NQ |
CitedBy_id | crossref_primary_10_1016_j_ymthe_2023_07_006 crossref_primary_10_3389_fmicb_2023_1247041 crossref_primary_10_1073_pnas_2309472120 crossref_primary_10_3390_ph16081088 crossref_primary_10_1016_j_vaccine_2023_12_052 crossref_primary_10_1002_mabi_202300275 crossref_primary_10_1016_j_ymthe_2023_10_006 crossref_primary_10_3390_vaccines11091481 crossref_primary_10_1248_bpb_b23_00689 crossref_primary_10_3390_vaccines12030318 crossref_primary_10_1080_15476286_2024_2333123 crossref_primary_10_1002_jgm_3672 crossref_primary_10_1016_j_ymthe_2023_06_017 crossref_primary_10_37349_emed_2023_00168 |
Cites_doi | 10.1016/j.immuni.2020.03.017 10.1371/journal.ppat.1003118 10.1016/j.immuni.2007.07.013 10.1016/j.ymthe.2017.08.006 10.1016/j.celrep.2021.109504 10.1016/j.immuni.2021.11.001 10.7554/eLife.46149 10.1371/journal.ppat.1005754 10.1016/j.vaccine.2012.01.070 10.1016/j.molimm.2009.11.003 10.1038/s41434-020-00204-y 10.1016/j.cyto.2017.08.009 10.1111/cei.13517 10.1038/s41541-017-0032-6 10.3389/fimmu.2018.00126 10.1038/s41467-020-17409-9 10.3390/vaccines8030433 10.1128/JVI.01675-10 10.1016/j.omtn.2019.08.001 10.4049/jimmunol.1601877 10.1080/21645515.2015.1070998 10.1111/imm.12742 10.1016/j.immuni.2006.08.014 10.1038/s41577-021-00592-1 10.1128/JVI.00333-14 10.1111/cei.13234 10.1016/j.ymthe.2017.11.017 10.1021/acsnano.0c00326 10.1038/s41590-022-01163-9 10.1016/j.eclinm.2021.101262 10.1016/j.ymthe.2018.07.010 10.1084/jem.20181899 10.1016/j.addr.2021.113900 10.1016/j.antiviral.2013.05.007 10.1016/j.omtn.2021.03.008 10.1093/oxfimm/iqac004 10.1016/j.jconrel.2021.08.029 10.1016/j.ymthe.2020.11.011 |
ContentType | Journal Article |
Copyright | 2022 The Authors 2022 The Authors. 2022 The Authors 2022 |
Copyright_xml | – notice: 2022 The Authors – notice: 2022 The Authors. – notice: 2022 The Authors 2022 |
DBID | 6I. AAFTH NPM AAYXX CITATION 7X8 5PM DOA |
DOI | 10.1016/j.omtn.2022.11.024 |
DatabaseName | ScienceDirect Open Access Titles Elsevier:ScienceDirect:Open Access PubMed CrossRef MEDLINE - Academic PubMed Central (Full Participant titles) Directory of Open Access Journals |
DatabaseTitle | PubMed CrossRef MEDLINE - Academic |
DatabaseTitleList | MEDLINE - Academic PubMed |
Database_xml | – sequence: 1 dbid: DOA name: Directory of Open Access Journals url: https://www.doaj.org/ sourceTypes: Open Website – sequence: 2 dbid: NPM name: PubMed url: https://proxy.k.utb.cz/login?url=http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=PubMed sourceTypes: Index Database |
DeliveryMethod | fulltext_linktorsrc |
Discipline | Anatomy & Physiology |
EISSN | 2162-2531 |
EndPage | 42 |
ExternalDocumentID | oai_doaj_org_article_0d6a34fafa0d4648b2111c1434bf06b4 10_1016_j_omtn_2022_11_024 36589712 S216225312200316X |
Genre | Journal Article |
GroupedDBID | 0R~ 0SF 53G 5VS 6I. 7X7 8FE 8FH 8FI AACTN AAEDW AAFTH AALRI AAXUO ABMAC ACGFS ADBBV AEXQZ AFKRA AFTJW AITUG ALMA_UNASSIGNED_HOLDINGS AMRAJ AOIJS AZQEC BBNVY BCNDV BENPR BHPHI BPHCQ BVXVI DIK EBS FDB FYUFA GROUPED_DOAJ HCIFZ KQ8 LK8 M2P M41 M48 M7P M~E NCXOZ O9- OK1 PIMPY PQQKQ PROAC RNTTT ROL RPM SSZ AAMRU ADVLN ALIPV NPM 3V. 88A 88I 8FJ AAYXX ABUWG ADRAZ CCPQU CITATION DWQXO EJD GNUQQ HMCUK HYE IPNFZ M0L RIG UKHRP 7X8 5PM |
ID | FETCH-LOGICAL-c587t-d76d21ad447c56337fa368ec8ebfbf8d1d09f75a495b716dd789555e04193cb3 |
IEDL.DBID | RPM |
ISSN | 2162-2531 |
IngestDate | Tue Oct 22 14:52:14 EDT 2024 Tue Sep 17 21:30:31 EDT 2024 Sat Oct 26 04:01:40 EDT 2024 Thu Nov 21 23:30:54 EST 2024 Sat Nov 02 11:56:26 EDT 2024 Fri Feb 23 02:39:41 EST 2024 |
IsDoiOpenAccess | true |
IsOpenAccess | true |
IsPeerReviewed | true |
IsScholarly | true |
Keywords | influenza MT: Oligonucleotides: Therapies and Applications sensing vaccine inflammation formulation pandemic |
Language | English |
License | This is an open access article under the CC BY license. 2022 The Authors. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). |
LinkModel | DirectLink |
MergedId | FETCHMERGED-LOGICAL-c587t-d76d21ad447c56337fa368ec8ebfbf8d1d09f75a495b716dd789555e04193cb3 |
Notes | ObjectType-Article-1 SourceType-Scholarly Journals-1 ObjectType-Feature-2 content type line 23 Present address: UBC, Canada Present address: UCL, UK |
ORCID | 0000-0001-8093-8741 |
OpenAccessLink | https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9794906/ |
PMID | 36589712 |
PQID | 2760168921 |
PQPubID | 23479 |
PageCount | 14 |
ParticipantIDs | doaj_primary_oai_doaj_org_article_0d6a34fafa0d4648b2111c1434bf06b4 pubmedcentral_primary_oai_pubmedcentral_nih_gov_9794906 proquest_miscellaneous_2760168921 crossref_primary_10_1016_j_omtn_2022_11_024 pubmed_primary_36589712 elsevier_sciencedirect_doi_10_1016_j_omtn_2022_11_024 |
PublicationCentury | 2000 |
PublicationDate | 2023-03-14 |
PublicationDateYYYYMMDD | 2023-03-14 |
PublicationDate_xml | – month: 03 year: 2023 text: 2023-03-14 day: 14 |
PublicationDecade | 2020 |
PublicationPlace | United States |
PublicationPlace_xml | – name: United States |
PublicationTitle | Molecular therapy. Nucleic acids |
PublicationTitleAlternate | Mol Ther Nucleic Acids |
PublicationYear | 2023 |
Publisher | Elsevier Inc American Society of Gene & Cell Therapy Elsevier |
Publisher_xml | – name: Elsevier Inc – name: American Society of Gene & Cell Therapy – name: Elsevier |
References | Blakney, Zhu, McKay, Bouton, Yeow, Tang, Hu, Samnuan, Grigsby, Shattock, Stevens (bib12) 2020; 14 McKay, Cizmeci, Aldon, Maertzdorf, Weiner, Kaufmann, Lewis, van den Berg, Del Giudice, Shattock (bib32) 2019; 8 Tregoning, Brown, Cheeseman, Flight, Higham, Lemm, Pierce, Stirling, Wang, Pollock (bib2) 2020; 202 Ndeupen, Qin, Jacobsen, Estanbouli, Bouteau, Igyártó (bib22) 2021 Shirai, Kawai, Shibuya, Munakata, Omata, Suzuki, Yoshioka (bib23) 2020; 8 Fischetti, Zhong, Pinder, Tregoning, Shattock (bib31) 2017; 99 Zhong, Haltalli, Holder, Rice, Donaldson, O'Driscoll, Le-Doare, Kampmann, Tregoning (bib41) 2019; 195 Minnaert, Vanluchene, Verbeke, Lentacker, De Smedt, Raemdonck, Sanders, Remaut (bib9) 2021; 176 Groves, McDonald, Langat, Kinnear, Kellam, McCauley, Ellis, Thompson, Elderfield, Parker (bib15) 2018; 9 Tregoning, Flight, Higham, Wang, Pierce (bib1) 2021; 21 Huysmans, Zhong, De Temmerman, Mui, Tam, Mc Cafferty, Gitsels, Vanrompay, Sanders (bib26) 2019; 17 Kandasamy, Suryawanshi, Tundup, Perez, Schmolke, Manicassamy, Manicassamy (bib18) 2016; 12 Liu, Gack (bib7) 2020; 53 Pepini, Pulichino, Carsillo, Carlson, Sari-Sarraf, Ramsauer, Debasitis, Maruggi, Otten, Geall (bib21) 2017; 198 Goritzka, Durant, Pereira, Salek-Ardakani, Openshaw, Johansson (bib39) 2014; 88 Lazear, Lancaster, Wilkins, Suthar, Huang, Vick, Clepper, Thackray, Brassil, Virgin (bib27) 2013; 9 Veazey, Siddiqui, Klein, Buffa, Fischetti, Doyle-Meyers, King, Tregoning, Shattock (bib33) 2015; 11 Liang, Lindgren, Lin, Thompson, Ols, Röhss, John, Hassett, Yuzhakov, Bahl (bib20) 2017; 25 Li, Lee, Grigoryan, Arunachalam, Scott, Trisal, Wimmers, Sanyal, Weidenbacher, Feng (bib34) 2022; 23 McKay, Zhou, Frise, Blakney, Bouton, Wang, Hu, Samnuan, Brown, Kugathasan (bib38) 2022; 3 Blakney, McKay, Bouton, Hu, Samnuan, Shattock (bib29) 2021; 29 Erasmus, Khandhar, Guderian, Granger, Archer, Archer, Gage, Fuerte-Stone, Larson, Lin (bib13) 2018; 26 Maruggi G, Ulmer JB, Rappuoli R, Yu D. Self-amplifying mRNA-Based Vaccine Technology and its Mode of Action. Springer Berlin; 1-40. Lutz, Lazzaro, Habbeddine, Schmidt, Baumhof, Mui, Tam, Madden, Hope, Heidenreich, Fotin-Mleczek (bib19) 2017; 2 Blakney, McKay, Hu, Samnuan, Jain, Brown, Thomas, Rogers, Polra, Sallah (bib14) 2021; 338 Bloom, van den Berg, Arbuthnot (bib11) 2021; 28 Liu, Xu, Hsu, Luo, Xiang, Chuang (bib37) 2010; 47 van Boxel-Dezaire, Rani, Stark (bib8) 2006; 25 Arimori, Nakamura, Yamada, Shibata, Maeda, Kase, Yoshikai (bib16) 2013; 99 Zhang, You, Wang, Cui, Wang, Xu, Li, Yang, Liu, Huang (bib28) 2021; 118 Pollock, Cheeseman, Szubert, Libri, Boffito, Owen, Bern, O'Hara, McFarlane, Lemm (bib5) 2022; 44 Watson, Endsley, Huang (bib24) 2012; 30 Bergamaschi, Terpos, Rosati, Angel, Bear, Stellas, Karaliota, Apostolakou, Bagratuni, Patseas (bib30) 2021; 36 Vogel, Lambert, Kinnear, Busse, Erbar, Reuter, Wicke, Perkovic, Beissert, Haas (bib3) 2018; 26 McDonald, Zhong, Groves, Tregoning (bib10) 2017; 151 Alameh, Tombácz, Bettini, Lederer, Sittplangkoon, Wilmore, Gaudette, Soliman, Pine, Hicks (bib25) 2021; 54 Kumagai, Takeuchi, Kato, Kumar, Matsui, Morii, Aozasa, Kawai, Akira (bib40) 2007; 27 Barrat (bib36) 2018; 215 McKay, Hu, Blakney, Samnuan, Brown, Penn, Zhou, Bouton, Rogers, Polra (bib4) 2020; 11 Seo, Kwon, Song, Byun, Seong, Kawai, Akira, Kweon (bib17) 2010; 84 Davies, Hovdal, Edmunds, Nordberg, Dahlén, Dabkowska, Arteta, Radulescu, Kjellman, Höijer (bib35) 2021; 24 Lutz (10.1016/j.omtn.2022.11.024_bib19) 2017; 2 Davies (10.1016/j.omtn.2022.11.024_bib35) 2021; 24 Bloom (10.1016/j.omtn.2022.11.024_bib11) 2021; 28 Blakney (10.1016/j.omtn.2022.11.024_bib12) 2020; 14 Kumagai (10.1016/j.omtn.2022.11.024_bib40) 2007; 27 Fischetti (10.1016/j.omtn.2022.11.024_bib31) 2017; 99 Vogel (10.1016/j.omtn.2022.11.024_bib3) 2018; 26 Liang (10.1016/j.omtn.2022.11.024_bib20) 2017; 25 van Boxel-Dezaire (10.1016/j.omtn.2022.11.024_bib8) 2006; 25 Alameh (10.1016/j.omtn.2022.11.024_bib25) 2021; 54 Minnaert (10.1016/j.omtn.2022.11.024_bib9) 2021; 176 Groves (10.1016/j.omtn.2022.11.024_bib15) 2018; 9 Ndeupen (10.1016/j.omtn.2022.11.024_bib22) 2021 Kandasamy (10.1016/j.omtn.2022.11.024_bib18) 2016; 12 Barrat (10.1016/j.omtn.2022.11.024_bib36) 2018; 215 Lazear (10.1016/j.omtn.2022.11.024_bib27) 2013; 9 Blakney (10.1016/j.omtn.2022.11.024_bib29) 2021; 29 Tregoning (10.1016/j.omtn.2022.11.024_bib2) 2020; 202 Seo (10.1016/j.omtn.2022.11.024_bib17) 2010; 84 Shirai (10.1016/j.omtn.2022.11.024_bib23) 2020; 8 McDonald (10.1016/j.omtn.2022.11.024_bib10) 2017; 151 Pollock (10.1016/j.omtn.2022.11.024_bib5) 2022; 44 Arimori (10.1016/j.omtn.2022.11.024_bib16) 2013; 99 McKay (10.1016/j.omtn.2022.11.024_bib38) 2022; 3 Blakney (10.1016/j.omtn.2022.11.024_bib14) 2021; 338 Bergamaschi (10.1016/j.omtn.2022.11.024_bib30) 2021; 36 Li (10.1016/j.omtn.2022.11.024_bib34) 2022; 23 Zhang (10.1016/j.omtn.2022.11.024_bib28) 2021; 118 Liu (10.1016/j.omtn.2022.11.024_bib37) 2010; 47 Watson (10.1016/j.omtn.2022.11.024_bib24) 2012; 30 10.1016/j.omtn.2022.11.024_bib6 Tregoning (10.1016/j.omtn.2022.11.024_bib1) 2021; 21 Erasmus (10.1016/j.omtn.2022.11.024_bib13) 2018; 26 Veazey (10.1016/j.omtn.2022.11.024_bib33) 2015; 11 Huysmans (10.1016/j.omtn.2022.11.024_bib26) 2019; 17 Goritzka (10.1016/j.omtn.2022.11.024_bib39) 2014; 88 Pepini (10.1016/j.omtn.2022.11.024_bib21) 2017; 198 McKay (10.1016/j.omtn.2022.11.024_bib32) 2019; 8 Liu (10.1016/j.omtn.2022.11.024_bib7) 2020; 53 Zhong (10.1016/j.omtn.2022.11.024_bib41) 2019; 195 McKay (10.1016/j.omtn.2022.11.024_bib4) 2020; 11 |
References_xml | – volume: 9 start-page: 126 year: 2018 ident: bib15 article-title: Mouse models of influenza infection with circulating strains to test seasonal vaccine efficacy. Original research frontiers in immunology publication-title: Front. Immunol. contributor: fullname: Parker – volume: 47 start-page: 1083 year: 2010 end-page: 1090 ident: bib37 article-title: A five-amino-acid motif in the undefined region of the TLR8 ectodomain is required for species-specific ligand recognition publication-title: Mol. Immunol. contributor: fullname: Chuang – volume: 25 start-page: 2635 year: 2017 end-page: 2647 ident: bib20 article-title: Efficient targeting and activation of antigen-presenting cells in vivo after modified mRNA vaccine administration in rhesus macaques publication-title: Mol. Ther. contributor: fullname: Bahl – volume: 99 start-page: 230 year: 2013 end-page: 237 ident: bib16 article-title: Type I interferon limits influenza virus-induced acute lung injury by regulation of excessive inflammation in mice publication-title: Antiviral Res. contributor: fullname: Yoshikai – volume: 36 start-page: 109504 year: 2021 ident: bib30 article-title: Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients publication-title: Cell Rep. contributor: fullname: Patseas – volume: 12 start-page: e100574 year: 2016 ident: bib18 article-title: RIG-I signaling is critical for efficient polyfunctional T cell responses during influenza virus infection publication-title: PLoS Pathog. contributor: fullname: Manicassamy – volume: 8 start-page: 433 year: 2020 ident: bib23 article-title: Lipid nanoparticle acts as a potential adjuvant for influenza split vaccine without inducing inflammatory responses publication-title: Vaccines contributor: fullname: Yoshioka – volume: 28 start-page: 117 year: 2021 end-page: 129 ident: bib11 article-title: Self-amplifying RNA vaccines for infectious diseases publication-title: Gene Ther. contributor: fullname: Arbuthnot – volume: 53 start-page: 26 year: 2020 end-page: 42 ident: bib7 article-title: Distinct and orchestrated functions of RNA sensors in innate immunity publication-title: Immunity contributor: fullname: Gack – volume: 27 start-page: 240 year: 2007 end-page: 252 ident: bib40 article-title: Alveolar macrophages are the primary interferon-alpha producer in pulmonary infection with RNA viruses publication-title: Immunity contributor: fullname: Akira – volume: 11 start-page: 3523 year: 2020 ident: bib4 article-title: Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice publication-title: Nat. Commun. contributor: fullname: Polra – volume: 30 start-page: 2256 year: 2012 end-page: 2272 ident: bib24 article-title: Design considerations for liposomal vaccines: influence of formulation parameters on antibody and cell-mediated immune responses to liposome associated antigens publication-title: Vaccine contributor: fullname: Huang – volume: 24 start-page: 369 year: 2021 end-page: 384 ident: bib35 article-title: Functionalized lipid nanoparticles for subcutaneous administration of mRNA to achieve systemic exposures of a therapeutic protein publication-title: Mol. Ther. Nucleic Acids contributor: fullname: Höijer – volume: 99 start-page: 287 year: 2017 end-page: 296 ident: bib31 article-title: The synergistic effects of combining TLR ligand based adjuvants on the cytokine response are dependent upon p38/JNK signalling publication-title: Cytokine contributor: fullname: Shattock – volume: 84 start-page: 12713 year: 2010 end-page: 12722 ident: bib17 article-title: MyD88 signaling is indispensable for primary influenza A virus infection but dispensable for secondary infection publication-title: J. Virol. contributor: fullname: Kweon – volume: 202 start-page: 162 year: 2020 end-page: 192 ident: bib2 article-title: Vaccines for COVID-19 publication-title: Clin. Exp. Immunol. contributor: fullname: Pollock – volume: 151 start-page: 451 year: 2017 end-page: 463 ident: bib10 article-title: Inflammatory responses to influenza vaccination at the extremes of age publication-title: Immunology contributor: fullname: Tregoning – volume: 88 start-page: 6128 year: 2014 end-page: 6136 ident: bib39 article-title: Alpha/beta interferon receptor signaling amplifies early proinflammatory cytokine production in the lung during respiratory syncytial virus infection publication-title: J. Virol. contributor: fullname: Johansson – volume: 26 start-page: 446 year: 2018 end-page: 455 ident: bib3 article-title: Self-amplifying RNA vaccines give equivalent protection against influenza to mRNA vaccines but at much lower doses publication-title: Mol. Ther. contributor: fullname: Haas – volume: 176 start-page: 113900 year: 2021 ident: bib9 article-title: Strategies for controlling the innate immune activity of conventional and self-amplifying mRNA therapeutics: getting the message across publication-title: Adv. Drug Deliv. Rev. contributor: fullname: Remaut – volume: 54 start-page: 2877 year: 2021 end-page: 2892.e7 ident: bib25 article-title: Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses publication-title: Immunity contributor: fullname: Hicks – volume: 29 start-page: 1174 year: 2021 end-page: 1185 ident: bib29 article-title: Innate inhibiting proteins enhance expression and immunogenicity of self-amplifying RNA publication-title: Mol. Ther. contributor: fullname: Shattock – volume: 23 start-page: 543 year: 2022 end-page: 555 ident: bib34 article-title: Mechanisms of innate and adaptive immunity to the Pfizer-BioNTech BNT162b2 vaccine publication-title: Nat. Immunol. contributor: fullname: Feng – volume: 215 start-page: 2964 year: 2018 end-page: 2966 ident: bib36 article-title: TLR8: No gain, no pain publication-title: J. Exp. Med. contributor: fullname: Barrat – volume: 25 start-page: 361 year: 2006 end-page: 372 ident: bib8 article-title: Complex modulation of cell type-specific signaling in response to type I interferons publication-title: Immunity contributor: fullname: Stark – volume: 338 start-page: 201 year: 2021 end-page: 210 ident: bib14 article-title: Polymeric and lipid nanoparticles for delivery of self-amplifying RNA vaccines publication-title: J. Control. Release contributor: fullname: Sallah – volume: 17 start-page: 867 year: 2019 end-page: 878 ident: bib26 article-title: Expression kinetics and innate immune response after electroporation and LNP-mediated delivery of a self-amplifying mRNA in the skin publication-title: Mol. Ther. Nucleic Acids contributor: fullname: Sanders – volume: 9 start-page: e1003118 year: 2013 ident: bib27 article-title: IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling publication-title: PLoS Pathog. contributor: fullname: Virgin – volume: 118 year: 2021 ident: bib28 article-title: Delivery of mRNA vaccine with a lipid-like material potentiates antitumor efficacy through Toll-like receptor 4 signaling publication-title: Proc. Natl. Acad. Sci. USA contributor: fullname: Huang – volume: 3 start-page: iqac004 year: 2022 ident: bib38 article-title: Polymer formulated self-amplifying RNA vaccine is partially protective against influenza virus infection in ferrets publication-title: Oxford Open Immunol. contributor: fullname: Kugathasan – volume: 26 start-page: 2507 year: 2018 end-page: 2522 ident: bib13 article-title: A nanostructured lipid carrier for delivery of a replicating viral RNA provides single, low-dose protection against Zika publication-title: Mol. Ther. contributor: fullname: Lin – volume: 8 start-page: e46149 year: 2019 ident: bib32 article-title: Identification of potential biomarkers of vaccine inflammation in mice publication-title: Elife contributor: fullname: Shattock – volume: 21 start-page: 626 year: 2021 end-page: 636 ident: bib1 article-title: Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape publication-title: Nat. Rev. Immunol. contributor: fullname: Pierce – volume: 195 start-page: 139 year: 2019 end-page: 152 ident: bib41 article-title: The impact of timing of maternal influenza immunization on infant antibody levels at birth publication-title: Clin. Exp. Immunol. contributor: fullname: Tregoning – volume: 14 start-page: 5711 year: 2020 end-page: 5727 ident: bib12 article-title: Big is beautiful: enhanced saRNA delivery and immunogenicity by a higher molecular weight, bioreducible, cationic polymer publication-title: ACS Nano contributor: fullname: Stevens – volume: 2 start-page: 29 year: 2017 ident: bib19 article-title: Unmodified mRNA in LNPs constitutes a competitive technology for prophylactic vaccines publication-title: NPJ Vaccines contributor: fullname: Fotin-Mleczek – volume: 198 start-page: 4012 year: 2017 end-page: 4024 ident: bib21 article-title: Induction of an IFN-mediated antiviral response by a self-amplifying RNA vaccine: implications for vaccine design publication-title: J. Immunol. contributor: fullname: Geall – volume: 11 start-page: 2913 year: 2015 end-page: 2922 ident: bib33 article-title: Evaluation of mucosal adjuvants and immunization routes for the induction of systemic and mucosal humoral immune responses in macaques publication-title: Hum. Vaccin. Immunother. contributor: fullname: Shattock – volume: 44 start-page: 101262 year: 2022 ident: bib5 article-title: Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial publication-title: EClinicalMedicine contributor: fullname: Lemm – year: 2021 ident: bib22 article-title: The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory publication-title: bioRxiv contributor: fullname: Igyártó – volume: 53 start-page: 26 year: 2020 ident: 10.1016/j.omtn.2022.11.024_bib7 article-title: Distinct and orchestrated functions of RNA sensors in innate immunity publication-title: Immunity doi: 10.1016/j.immuni.2020.03.017 contributor: fullname: Liu – year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib22 article-title: The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory publication-title: bioRxiv contributor: fullname: Ndeupen – volume: 9 start-page: e1003118 year: 2013 ident: 10.1016/j.omtn.2022.11.024_bib27 article-title: IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling publication-title: PLoS Pathog. doi: 10.1371/journal.ppat.1003118 contributor: fullname: Lazear – volume: 27 start-page: 240 year: 2007 ident: 10.1016/j.omtn.2022.11.024_bib40 article-title: Alveolar macrophages are the primary interferon-alpha producer in pulmonary infection with RNA viruses publication-title: Immunity doi: 10.1016/j.immuni.2007.07.013 contributor: fullname: Kumagai – volume: 25 start-page: 2635 year: 2017 ident: 10.1016/j.omtn.2022.11.024_bib20 article-title: Efficient targeting and activation of antigen-presenting cells in vivo after modified mRNA vaccine administration in rhesus macaques publication-title: Mol. Ther. doi: 10.1016/j.ymthe.2017.08.006 contributor: fullname: Liang – volume: 36 start-page: 109504 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib30 article-title: Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients publication-title: Cell Rep. doi: 10.1016/j.celrep.2021.109504 contributor: fullname: Bergamaschi – volume: 118 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib28 article-title: Delivery of mRNA vaccine with a lipid-like material potentiates antitumor efficacy through Toll-like receptor 4 signaling publication-title: Proc. Natl. Acad. Sci. USA contributor: fullname: Zhang – volume: 54 start-page: 2877 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib25 article-title: Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses publication-title: Immunity doi: 10.1016/j.immuni.2021.11.001 contributor: fullname: Alameh – volume: 8 start-page: e46149 year: 2019 ident: 10.1016/j.omtn.2022.11.024_bib32 article-title: Identification of potential biomarkers of vaccine inflammation in mice publication-title: Elife doi: 10.7554/eLife.46149 contributor: fullname: McKay – ident: 10.1016/j.omtn.2022.11.024_bib6 – volume: 12 start-page: e100574 year: 2016 ident: 10.1016/j.omtn.2022.11.024_bib18 article-title: RIG-I signaling is critical for efficient polyfunctional T cell responses during influenza virus infection publication-title: PLoS Pathog. doi: 10.1371/journal.ppat.1005754 contributor: fullname: Kandasamy – volume: 30 start-page: 2256 year: 2012 ident: 10.1016/j.omtn.2022.11.024_bib24 article-title: Design considerations for liposomal vaccines: influence of formulation parameters on antibody and cell-mediated immune responses to liposome associated antigens publication-title: Vaccine doi: 10.1016/j.vaccine.2012.01.070 contributor: fullname: Watson – volume: 47 start-page: 1083 year: 2010 ident: 10.1016/j.omtn.2022.11.024_bib37 article-title: A five-amino-acid motif in the undefined region of the TLR8 ectodomain is required for species-specific ligand recognition publication-title: Mol. Immunol. doi: 10.1016/j.molimm.2009.11.003 contributor: fullname: Liu – volume: 28 start-page: 117 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib11 article-title: Self-amplifying RNA vaccines for infectious diseases publication-title: Gene Ther. doi: 10.1038/s41434-020-00204-y contributor: fullname: Bloom – volume: 99 start-page: 287 year: 2017 ident: 10.1016/j.omtn.2022.11.024_bib31 article-title: The synergistic effects of combining TLR ligand based adjuvants on the cytokine response are dependent upon p38/JNK signalling publication-title: Cytokine doi: 10.1016/j.cyto.2017.08.009 contributor: fullname: Fischetti – volume: 202 start-page: 162 year: 2020 ident: 10.1016/j.omtn.2022.11.024_bib2 article-title: Vaccines for COVID-19 publication-title: Clin. Exp. Immunol. doi: 10.1111/cei.13517 contributor: fullname: Tregoning – volume: 2 start-page: 29 year: 2017 ident: 10.1016/j.omtn.2022.11.024_bib19 article-title: Unmodified mRNA in LNPs constitutes a competitive technology for prophylactic vaccines publication-title: NPJ Vaccines doi: 10.1038/s41541-017-0032-6 contributor: fullname: Lutz – volume: 9 start-page: 126 year: 2018 ident: 10.1016/j.omtn.2022.11.024_bib15 article-title: Mouse models of influenza infection with circulating strains to test seasonal vaccine efficacy. Original research frontiers in immunology publication-title: Front. Immunol. doi: 10.3389/fimmu.2018.00126 contributor: fullname: Groves – volume: 11 start-page: 3523 year: 2020 ident: 10.1016/j.omtn.2022.11.024_bib4 article-title: Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice publication-title: Nat. Commun. doi: 10.1038/s41467-020-17409-9 contributor: fullname: McKay – volume: 8 start-page: 433 year: 2020 ident: 10.1016/j.omtn.2022.11.024_bib23 article-title: Lipid nanoparticle acts as a potential adjuvant for influenza split vaccine without inducing inflammatory responses publication-title: Vaccines doi: 10.3390/vaccines8030433 contributor: fullname: Shirai – volume: 84 start-page: 12713 year: 2010 ident: 10.1016/j.omtn.2022.11.024_bib17 article-title: MyD88 signaling is indispensable for primary influenza A virus infection but dispensable for secondary infection publication-title: J. Virol. doi: 10.1128/JVI.01675-10 contributor: fullname: Seo – volume: 17 start-page: 867 year: 2019 ident: 10.1016/j.omtn.2022.11.024_bib26 article-title: Expression kinetics and innate immune response after electroporation and LNP-mediated delivery of a self-amplifying mRNA in the skin publication-title: Mol. Ther. Nucleic Acids doi: 10.1016/j.omtn.2019.08.001 contributor: fullname: Huysmans – volume: 198 start-page: 4012 year: 2017 ident: 10.1016/j.omtn.2022.11.024_bib21 article-title: Induction of an IFN-mediated antiviral response by a self-amplifying RNA vaccine: implications for vaccine design publication-title: J. Immunol. doi: 10.4049/jimmunol.1601877 contributor: fullname: Pepini – volume: 11 start-page: 2913 year: 2015 ident: 10.1016/j.omtn.2022.11.024_bib33 article-title: Evaluation of mucosal adjuvants and immunization routes for the induction of systemic and mucosal humoral immune responses in macaques publication-title: Hum. Vaccin. Immunother. doi: 10.1080/21645515.2015.1070998 contributor: fullname: Veazey – volume: 151 start-page: 451 year: 2017 ident: 10.1016/j.omtn.2022.11.024_bib10 article-title: Inflammatory responses to influenza vaccination at the extremes of age publication-title: Immunology doi: 10.1111/imm.12742 contributor: fullname: McDonald – volume: 25 start-page: 361 year: 2006 ident: 10.1016/j.omtn.2022.11.024_bib8 article-title: Complex modulation of cell type-specific signaling in response to type I interferons publication-title: Immunity doi: 10.1016/j.immuni.2006.08.014 contributor: fullname: van Boxel-Dezaire – volume: 21 start-page: 626 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib1 article-title: Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape publication-title: Nat. Rev. Immunol. doi: 10.1038/s41577-021-00592-1 contributor: fullname: Tregoning – volume: 88 start-page: 6128 year: 2014 ident: 10.1016/j.omtn.2022.11.024_bib39 article-title: Alpha/beta interferon receptor signaling amplifies early proinflammatory cytokine production in the lung during respiratory syncytial virus infection publication-title: J. Virol. doi: 10.1128/JVI.00333-14 contributor: fullname: Goritzka – volume: 195 start-page: 139 year: 2019 ident: 10.1016/j.omtn.2022.11.024_bib41 article-title: The impact of timing of maternal influenza immunization on infant antibody levels at birth publication-title: Clin. Exp. Immunol. doi: 10.1111/cei.13234 contributor: fullname: Zhong – volume: 26 start-page: 446 year: 2018 ident: 10.1016/j.omtn.2022.11.024_bib3 article-title: Self-amplifying RNA vaccines give equivalent protection against influenza to mRNA vaccines but at much lower doses publication-title: Mol. Ther. doi: 10.1016/j.ymthe.2017.11.017 contributor: fullname: Vogel – volume: 14 start-page: 5711 year: 2020 ident: 10.1016/j.omtn.2022.11.024_bib12 article-title: Big is beautiful: enhanced saRNA delivery and immunogenicity by a higher molecular weight, bioreducible, cationic polymer publication-title: ACS Nano doi: 10.1021/acsnano.0c00326 contributor: fullname: Blakney – volume: 23 start-page: 543 year: 2022 ident: 10.1016/j.omtn.2022.11.024_bib34 article-title: Mechanisms of innate and adaptive immunity to the Pfizer-BioNTech BNT162b2 vaccine publication-title: Nat. Immunol. doi: 10.1038/s41590-022-01163-9 contributor: fullname: Li – volume: 44 start-page: 101262 year: 2022 ident: 10.1016/j.omtn.2022.11.024_bib5 article-title: Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial publication-title: EClinicalMedicine doi: 10.1016/j.eclinm.2021.101262 contributor: fullname: Pollock – volume: 26 start-page: 2507 year: 2018 ident: 10.1016/j.omtn.2022.11.024_bib13 article-title: A nanostructured lipid carrier for delivery of a replicating viral RNA provides single, low-dose protection against Zika publication-title: Mol. Ther. doi: 10.1016/j.ymthe.2018.07.010 contributor: fullname: Erasmus – volume: 215 start-page: 2964 year: 2018 ident: 10.1016/j.omtn.2022.11.024_bib36 article-title: TLR8: No gain, no pain publication-title: J. Exp. Med. doi: 10.1084/jem.20181899 contributor: fullname: Barrat – volume: 176 start-page: 113900 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib9 article-title: Strategies for controlling the innate immune activity of conventional and self-amplifying mRNA therapeutics: getting the message across publication-title: Adv. Drug Deliv. Rev. doi: 10.1016/j.addr.2021.113900 contributor: fullname: Minnaert – volume: 99 start-page: 230 year: 2013 ident: 10.1016/j.omtn.2022.11.024_bib16 article-title: Type I interferon limits influenza virus-induced acute lung injury by regulation of excessive inflammation in mice publication-title: Antiviral Res. doi: 10.1016/j.antiviral.2013.05.007 contributor: fullname: Arimori – volume: 24 start-page: 369 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib35 article-title: Functionalized lipid nanoparticles for subcutaneous administration of mRNA to achieve systemic exposures of a therapeutic protein publication-title: Mol. Ther. Nucleic Acids doi: 10.1016/j.omtn.2021.03.008 contributor: fullname: Davies – volume: 3 start-page: iqac004 year: 2022 ident: 10.1016/j.omtn.2022.11.024_bib38 article-title: Polymer formulated self-amplifying RNA vaccine is partially protective against influenza virus infection in ferrets publication-title: Oxford Open Immunol. doi: 10.1093/oxfimm/iqac004 contributor: fullname: McKay – volume: 338 start-page: 201 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib14 article-title: Polymeric and lipid nanoparticles for delivery of self-amplifying RNA vaccines publication-title: J. Control. Release doi: 10.1016/j.jconrel.2021.08.029 contributor: fullname: Blakney – volume: 29 start-page: 1174 year: 2021 ident: 10.1016/j.omtn.2022.11.024_bib29 article-title: Innate inhibiting proteins enhance expression and immunogenicity of self-amplifying RNA publication-title: Mol. Ther. doi: 10.1016/j.ymthe.2020.11.011 contributor: fullname: Blakney |
SSID | ssj0000601262 |
Score | 2.4277146 |
Snippet | To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These... To be effective, RNA vaccines require both translation and the induction of an immune response to recruit cells to the site of immunization. These factors can... To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These... |
SourceID | doaj pubmedcentral proquest crossref pubmed elsevier |
SourceType | Open Website Open Access Repository Aggregation Database Index Database Publisher |
StartPage | 29 |
SubjectTerms | formulation inflammation influenza MT: Oligonucleotides: Therapies and Applications Original pandemic sensing vaccine |
SummonAdditionalLinks | – databaseName: Directory of Open Access Journals dbid: DOA link: http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwrV1Lb9QwELZQT1wQUB7hJSMhLhDwO_FxQawqJHpARerNsmMbFrFOxe5W6r9nbGerDUhw4ZZ34syX8Tfx5xmEXjgPLq6P4P2Ctq1ggreOedGSQGjsrSehFIP5dKpOvoiP5_L8oNRX1oTV9MD1xb0lXlkuoo2WeKFE7yBioQP08sJFolzNBErYQTBVfTA43lJNlFHFWgZIm2bMVHHXuN7m5KeMvckpPJmY9Uolef-sc_qTfP6uoTzolJa30a2JTeJFbcUddCOku-h4kSCSXl_hl7joO8uP82Pkl0BPp2JdrzEAC7CwntZs8vjz6QJvspw9fcV17iQGcgiL610aAWarAQg7HiMc9CO2NivRyxypcuKlHfII_eYeOlt-OHt_0k41FtpB9t229Z3yjFovRDdIxXkXLVd9GPrgoou9p57o2EkLcZSD0Mr7rtdSykAEML_B8fvoKI0pPERYA1MKnQ_cSS0kxHFRUy2s5AGciB10g17tX7G5qJk0zF5i9t1kg5hsEAhJDBikQe-yFa6PzFmwywbAhpmwYf6FjQbJvQ3NRCgqUYBLrf568-d7gxv42vIQik1h3G0MyxIi1WtGG_SgAuD6ETmQOd1R1qBuBo1ZG-Z70upbyeitwStqoh79j0Y_RjehKTzr5Kh4go62P3fhKRCnrXtWvpFfvrkVUg priority: 102 providerName: Directory of Open Access Journals – databaseName: Scholars Portal Open Access Journals dbid: M48 link: http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1Jb9QwFLZKuXBBQFmGTUZCXCBV4i32oaqGilGF1B5QK_Vm2bFdpuokMAui_55nxymEVhy4ZbGz-C3-XvL5PYTeWgcuTgbwfl6ZghFGC0scK0pfVkEaV_pUDOboWByess9n_GwLDeWO8gCubg3tYj2p0-Xl7s_vV_tg8Hu_uVrdYh1zmRKyGzNyEnYH3YW7VpHidZThfu-ZwR0LktfO3N51ND-lNP6jaeomDP2bTfnH9DR7gO5nXImnvSI8RFu-fYR2pi3E1Isr_A4npmf6hL6D3AyAai7b9QHDS4NWLPKeaR3-cjzFq0hsb89xv4oSA0yEzcWm7UDh5g1Ad9wFaHQZChM56Wm1VOr4wzTxX_3qMTqZfTo5OCxytYWi4bJeF64WjlTGMVY3XFBaB0OF9I30NtggXeVKFWpuIKKyEGQ5V0vFOfclAwzYWPoEbbdd658hrAAz-dp5arliHCK6oCrFDKce3Ilp1AS9H4ZYf-tzauiBbHaho0B0FAgEJxoEMkEfoxSuW8Z82OlAtzzX2bx06YShLJhgSscEkxbi2qoBLMhsKIWFi_BBhjpDix4ywKXm_7z5m0HgGuwu_kwxre82K00imUhIRaoJetorwPUjUoB1qq7IBNUj1Ri9w_hMO_-acnsr8I-qFM__u-cLdA-en0aaXMVeou31cuNfAW5a29fJGH4Bcdsa0w priority: 102 providerName: Scholars Portal |
Title | Formulation, inflammation, and RNA sensing impact the immunogenicity of self-amplifying RNA vaccines |
URI | https://dx.doi.org/10.1016/j.omtn.2022.11.024 https://www.ncbi.nlm.nih.gov/pubmed/36589712 https://www.proquest.com/docview/2760168921 https://pubmed.ncbi.nlm.nih.gov/PMC9794906 https://doaj.org/article/0d6a34fafa0d4648b2111c1434bf06b4 |
Volume | 31 |
hasFullText | 1 |
inHoldings | 1 |
isFullTextHit | |
isPrint | |
link | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1Lb9QwELbanrggoDwWSmUkxAWymzi2Yx-XilWFtBVCRerN8rNs1ThVdxep_56xk1RdkDhwifKwHSczHn9jfx4j9N44MHEigPXzUheU0LowxNGi9GUVhHalz5vBLM_46Q_69YJd7CE2roXJpH1rVtN43U7j6mfmVt60djbyxGbflicSlEiWfLaP9qH7feCi9-YXbC4nwwKZnsvVtZsU65SQaYrYSehOJ5Rj9e_0RX9jzT8pkw_6oMUT9HgAj3jeV_Ip2vPxGTqcR3Cc2zv8AWc6Zx4nP0RuAWh02JvrEwY9AtG3w5WODn8_m-N1Yq_HS9wvlcSABeG03cYOtGplAZ_jLkCi61DoRDzPS6Jyxl_apgn59XN0vvhyfnJaDFsqFJaJZlO4hjtSaUdpYxmv6ybomgtvhTfBBOEqV8rQMA1ukwFPyrlGSMaYLykAPWvqF-ggdtG_QlgCMPKN87VhkjJw24KsJNWs9mAztJUT9HH8xeqmD5yhRkbZlUoCUUkg4IEoEMgEfU5SuE-Zgl7nG93tpRpEr0rHdU2DDrp0lFNhwHmtLAA-akLJDRTCRhmqAT_0uACKWv3z5e9GgStoXGnGREffbdeKJMYQF5JUE_SyV4D7KtaA3WRTkQlqdlRj5xt2n4A-5wDeg_6-_u-cb9CjtO194sJV9AgdbG63_i2Ao405zoMKcFxScZwbxm_9oRRX |
link.rule.ids | 230,314,727,780,784,864,885,2102,24318,27924,27925,53791,53793 |
linkProvider | National Library of Medicine |
linkToHtml | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1Lj9MwELaW5QAXBCyP8jQS4gJpEz_i-FhWVAW2FUJF2pvl51K0SVbbFol_z9hJVluQOHDLw07szHj8TfzNGKHXxoGJqwJYPy91xgijmSGOZbnPi1Bpl_u0GcxiWc6_sU-n_PQA8SEWJpH2rVmPm_N63Ky_J27lRW0nA09s8mVxLEGJZF5ObqCbnApZXHPSOwMMVrckfYhMx-Zq623MdkrIOObsJGxvGkrZ-vdmo7_R5p-kyWuz0OwuutPDRzztmnkPHfjmPjqaNuA617_wG5wInelP-RFyM8Cj_e5c7zBoEgi_7s904_DX5RRvIn-9OcNdsCQGNAiH9a5pQa_WFhA6bgMUOg-ZjtTzFBSVKv7UNi7Jbx6g1ezD6nie9ZsqZJZXYps5UTpSaMeYsLykVARNy8rbyptgQuUKl8sguAbHyYAv5ZyoJOfc5wygnjX0ITps2sY_RlgCNPLCeWq4ZBwctyALyTSnHqyGtnKE3g6fWF10qTPUwCn7oaJAVBQI-CAKBDJC76MUrkrGtNfpQnt5pnrhq9yVmrKgg84dK1llwH0tLEA-ZkJeGngIH2SoegTRIQN41PqfL381CFzB8IprJrrx7W6jSOQMlZUkxQg96hTgqokU0JsUBRkhsacae33YvwManVJ49xr85L9rvkS35qvFiTr5uPz8FN2GvtDIjCvYM3S4vdz55wCVtuZFGhi_AaJ5Fd0 |
linkToPdf | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1bb9MwFLZgSIgXBIxLNy5GQrxA2sSXJH4sg2pcVk1oSHuzfB1Fi1OtLRL_nmMnmVqQeOAt9zg5X46_E38-B6FX2oKLqz14PydUxgijmSaWZbnLC18rm7tUDOZkXh5_Y5_O-flWqa8k2jd6MQ6XzTgsvidt5bIxk0EnNjk9ORIAIpGXk6X1k5voFqcAsq1AvXPC4HlL0k-T6RRdbbOOGU8JGce8nYTtdEUpY_9Oj_Q34_xTOLnVE83uobs9hcTTrqn30Q0XHqD9aYDwufmFX-Mk6kx_y_eRnQEn7St0vcWAJgBA06-pYPHX-RSvooY9XOBuwiQGRgiLzSa0gK2FAZaOWw8HXfpMRfl5mhiVTvypTByWXz1EZ7MPZ0fHWV9YITO8rtaZrUpLCmUZqwwvKa28omXtTO201762hc2Fr7iC4ElDPGVtVQvOucsZ0D2j6SO0F9rgniAsgB65yjqquWAcgjcvCsEUpw48hzJihN4Mr1guu_QZctCV_ZDRIDIaBOIQCQYZoXfRCtdHxtTXaUN7dSF7AMjclooyr7zKLStZrSGELQzQPqZ9Xmq4CB9sKHsW0bEDuNTinzd_ORhcwicWx01UcO1mJUnUDZW1IMUIPe4AcN1ECgxOVAUZoWoHGjvPsLsHUJ3SePcoPvjvM1-g26fvZ_LLx_nnQ3QHHoVGcVzBnqK99dXGPQO2tNbP03fxG4p5FvA |
openUrl | ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Formulation%2C+inflammation%2C+and+RNA+sensing+impact+the+immunogenicity+of+self-amplifying+RNA+vaccines&rft.jtitle=Molecular+therapy.+Nucleic+acids&rft.au=Tregoning%2C+John+S.&rft.au=Stirling%2C+David+C.&rft.au=Wang%2C+Ziyin&rft.au=Flight%2C+Katie+E.&rft.date=2023-03-14&rft.pub=American+Society+of+Gene+%26+Cell+Therapy&rft.eissn=2162-2531&rft.volume=31&rft.spage=29&rft.epage=42&rft_id=info:doi/10.1016%2Fj.omtn.2022.11.024&rft.externalDBID=PMC9794906 |
thumbnail_l | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=2162-2531&client=summon |
thumbnail_m | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=2162-2531&client=summon |
thumbnail_s | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=2162-2531&client=summon |