Orthoflavivirus Vaccine Platforms: Current Strategies and Challenges
Abstract
1. Introduction to Orthoflaviviruses
1.1. Orthoflavivirus Disease Spectrum
1.2. Orthoflavivirus Epidemiology: A Growing and Global Threat
1.3. Orthoflavivirus Life Cycle and Antigenic Targets
2. Orthoflavivirus Vaccine Platforms: From Historical to Next-Generation Approaches
2.1. Live Attenuated Vaccines
2.2. Inactivated Vaccines
2.3. Nucleic Acid Vaccines: Messenger RNA (mRNA) and Self-Amplifying (saRNA)
2.3.1. mRNA and DNA Vaccines
2.3.2. Self-Amplifying RNA (saRNA) Vaccines
2.4. Virus-like Particles (VLPs) Vaccines
2.5. Insect-Specific Flaviviruses (ISFVs) Vaccines
2.6. Viral Vector Platforms
2.6.1. Adenovirus Vector Vaccines
2.6.2. Lentiviral Vector Vaccines
| Platform | Examples | Advantages | Disadvantages | Development Stage | Ref. |
|---|---|---|---|---|---|
| Live Attenuated Vaccines | YFV-17D (YFV), SA14–14–2 (JEV) | Long-lasting immunity; strong humoral and cellular responses; single dose; Preservation of antigenic structure | Safety concerns; ADE risks; Cold-chain requirements | Several licensed (e.g., YFV-17D, TAK-003) | [133,134,135,136,137,138,139,142,143,144] |
| Inactivated vaccines | Ixiaro (JEV), Ticovac (TBEV) | Safe for vulnerable individuals; No viral replication; Preservation of particle structure | Limited immunity; Requires adjuvant and multiple doses; Potential ADE risks; Potential epitope disruption | From Phase I/II trials (TDENV PIV; HydroVax 001) to licensed (Ixiaro) | [153,154,155,156,157,158,159,160,161,162,163,164] |
| mRNA vaccines | mRNA-1893 (ZIKV prM–E) | Rapid design and deployment; Potent systemic immunity | Limited durability of serum neutralizing responses; Cold-chain requirements; Authentic epitope presentation; ADE risks | From preclinical to Phase I/II (Zika mRNA-1325 and 1893) | [172,174,175,176,177,178] |
| DNA vaccines | GLS-5700 (ZIKV prM-E DNA plasmid) | Thermostability, versatility in genetic modifications | Low immunogenicity; High doses for injection; Authentic epitope presentation; Electroporation needed to enhance immunogenicity | From preclinical to Phase I trials | [183,184,185,186,187] |
| saRNA vaccines | Bivalent saRNA (ZIKV + YFV prM-E) | High antigen expression; More durable serum neutralizing responses compared to mRNA vaccines; Self-replication enhances adaptive immune activation; Multivalence and single-dose potential | Need to balance activation and evasion of cell-intrinsic immunity; Manufacturing challenges; ADE risks if monovalent; Authentic epitope presentation | Preclinical | [191,192,196,197,198] |
| VLPs | VLPs ZIKV prM-E | Presentation of authentic, membrane-anchored E protein; strong safety profile; potent immune responses | Manufacturing challenges because of low-titer production; ADE risks; require boost; Authentic epitope presentation | Preclinical | [214,215,221,222,223,224,225] |
| ISFVs | BinJ/WNVKUN-prME (WNV prM-E) | Grows to high titer in insect cells; Non-replicative in vertebrates (safe); Strong humoral and cellular responses | Requires insect-cell production; no GMP production capabilities | Preclinical | [237,240,241,242,243] |
| Adenoviral Vectors | ChAdOx1-ZIKV; Ad26.ZIKV.001 | Potent humoral and cellular responses with single dose with no evidence of ADE in animal models | Safety concerns; Authentic epitope presentation; Durability and efficacy concerns in humans; Pre-existing immunity | From preclinical to Phase I (Ad26.ZIKV.001) | [255,256,257] |
| Lentiviral Vectors | TRIP/sE_WNV (WNV prM-E) | Single-dose sterilizing immunity in mice; potent neutralizing antibodies | Integration into host genome; manufacturing complexity; Authentic epitope presentation | Preclinical | [266,267,274,275] |
2.7. Anti-Vector Vaccines
3. Orthoflavivirus Multivalent Vaccine Platforms
4. Key Attributes of a Robust Orthoflavivirus Vaccine
4.1. Must-Have Attributes
4.2. Desirable Attributes
5. Advancing Orthoflavivirus Vaccine Research
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Pierson, T.C.; Diamond, M.S. The continued threat of emerging flaviviruses. Nat. Microbiol. 2020, 5, 796–812. [Google Scholar] [CrossRef]
- Anis, H.; Basha Shaik, A.; Karabulut, E.; Uzun, M.; Tiwari, A.; Nazir, A.; Uwishema, O.; Alemayehu, A. Upsurge of Powassan virus disease in northeastern United States: A public health concern-a short communication. Ann. Med. Surg. 2023, 85, 5823–5826. [Google Scholar] [CrossRef]
- Pustijanac, E.; Bursic, M.; Talapko, J.; Skrlec, I.; Mestrovic, T.; Lisnjic, D. Tick-Borne Encephalitis Virus: A Comprehensive Review of Transmission, Pathogenesis, Epidemiology, Clinical Manifestations, Diagnosis, and Prevention. Microorganisms 2023, 11, 1634. [Google Scholar] [CrossRef]
- Gupta, P.C.; Satapathy, P.; Gupta, A.; Asumah, M.N.; Padhi, B.K. Usutu virus: A Flavivirus on the rise amid COVID-19 and monkeypox. Int. J. Surg. 2023, 109, 614–615. [Google Scholar] [CrossRef]
- Yakob, L.; Hu, W.; Frentiu, F.D.; Gyawali, N.; Hugo, L.E.; Johnson, B.; Lau, C.; Furuya-Kanamori, L.; Magalhaes, R.S.; Devine, G. Japanese Encephalitis Emergence in Australia: The Potential Population at Risk. Clin. Infect. Dis. 2023, 76, 335–337. [Google Scholar] [CrossRef]
- van den Hurk, A.F.; Ritchie, S.A.; Mackenzie, J.S. Ecology and geographical expansion of Japanese encephalitis virus. Annu. Rev. Entomol. 2009, 54, 17–35. [Google Scholar] [CrossRef] [PubMed]
- Dziadek, K.; Niczyporuk, J.S.; Stys-Fijol, N.; Czujkowska, A.; Smietanka, K.; Domanska-Blicharz, K. Usutu virus continues to spread across Europe: First report of multiple molecular detections of the USUV Africa 2 and Africa 3 lineages in free-living and captive birds in Poland, July–November 2023. Vet. Res. 2025, 56, 43. [Google Scholar] [CrossRef] [PubMed]
- Munger, E.; Atama, N.C.; van Irsel, J.; Blom, R.; Krol, L.; van Mastrigt, T.; van den Berg, T.J.; Braks, M.; de Vries, A.; van der Linden, A.; et al. One Health approach uncovers emergence and dynamics of Usutu and West Nile viruses in the Netherlands. Nat. Commun. 2025, 16, 7883. [Google Scholar] [CrossRef] [PubMed]
- Schilling, M.; Lawson, B.; Spiro, S.; Jagdev, M.; Vaux, A.G.C.; Bruce, R.C.; Johnston, C.J.; Abbott, A.J.; Wrigglesworth, E.; Pearce-Kelly, P.; et al. Vertical transmission in field-caught mosquitoes identifies a mechanism for the establishment of Usutu virus in a temperate country. Sci. Rep. 2025, 15, 25252. [Google Scholar] [CrossRef]
- Gould, E.A.; Solomon, T. Pathogenic flaviviruses. Lancet 2008, 371, 500–509. [Google Scholar] [CrossRef]
- Ryan, S.J.; Carlson, C.J.; Mordecai, E.A.; Johnson, L.R. Global expansion and redistribution of Aedes-borne virus transmission risk with climate change. PLoS Negl. Trop. Dis. 2019, 13, e0007213. [Google Scholar] [CrossRef] [PubMed]
- Gibb, R.; Colon-Gonzalez, F.J.; Lan, P.T.; Huong, P.T.; Nam, V.S.; Duoc, V.T.; Hung, D.T.; Dong, N.T.; Chien, V.C.; Trang, L.T.T.; et al. Interactions between climate change, urban infrastructure and mobility are driving dengue emergence in Vietnam. Nat. Commun. 2023, 14, 8179. [Google Scholar] [CrossRef] [PubMed]
- Worden-Sapper, E.R.; Gendler, P.; Lange, R.E.; Kuhn, J.H.; Sawyer, S.L. Future threats: Animal orthoflaviviruses that currently infect fewer than 100 people per year. Cell Rep. 2025, 44, 115700. [Google Scholar] [CrossRef]
- Swaminathan, S.; Schlaberg, R.; Lewis, J.; Hanson, K.E.; Couturier, M.R. Fatal Zika Virus Infection with Secondary Nonsexual Transmission. N. Engl. J. Med. 2016, 375, 1907–1909. [Google Scholar] [CrossRef]
- Karimi, O.; Goorhuis, A.; Schinkel, J.; Codrington, J.; Vreden, S.G.S.; Vermaat, J.S.; Stijnis, C.; Grobusch, M.P. Thrombocytopenia and subcutaneous bleedings in a patient with Zika virus infection. Lancet 2016, 387, 939–940. [Google Scholar] [CrossRef]
- van Leur, S.W.; Heunis, T.; Munnur, D.; Sanyal, S. Pathogenesis and virulence of flavivirus infections. Virulence 2021, 12, 2814–2838. [Google Scholar] [CrossRef]
- Bazer, D.A.; Orwitz, M.; Koroneos, N.; Syritsyna, O.; Wirkowski, E. Powassan Encephalitis: A Case Report from New York, USA. Case Rep. Neurol. Med. 2022, 2022, 8630349. [Google Scholar] [CrossRef] [PubMed]
- Aye, K.S.; Charngkaew, K.; Win, N.; Wai, K.Z.; Moe, K.; Punyadee, N.; Thiemmeca, S.; Suttitheptumrong, A.; Sukpanichnant, S.; Prida, M.; et al. Pathologic highlights of dengue hemorrhagic fever in 13 autopsy cases from Myanmar. Hum. Pathol. 2014, 45, 1221–1233. [Google Scholar] [CrossRef]
- Wills, B.A.; Oragui, E.E.; Stephens, A.C.; Daramola, O.A.; Dung, N.M.; Loan, H.T.; Chau, N.V.; Chambers, M.; Stepniewska, K.; Farrar, J.J.; et al. Coagulation abnormalities in dengue hemorrhagic Fever: Serial investigations in 167 Vietnamese children with Dengue shock syndrome. Clin. Infect. Dis. 2002, 35, 277–285. [Google Scholar] [CrossRef]
- Rodrigo, C.; Sigera, C.; Fernando, D.; Rajapakse, S. Plasma leakage in dengue: A systematic review of prospective observational studies. BMC Infect. Dis. 2021, 21, 1082. [Google Scholar] [CrossRef]
- Surabotsophon, M.; Laohachavalit, P.; Ponglikitmongkol, S.; Chuncharunee, S.; Sudsang, T.; Thanachartwet, V.; Sahassananda, D.; Hunsawong, T.; Klungthong, C.; Fernandez, S.; et al. Secondary dengue serotype 1 infection causing dengue shock syndrome with rhombencephalitis and bleeding associated with refractory thrombocytopenia: A case report. Heliyon 2023, 9, e17419. [Google Scholar] [CrossRef] [PubMed]
- Sejvar, J.J.; Bode, A.V.; Marfin, A.A.; Campbell, G.L.; Ewing, D.; Mazowiecki, M.; Pavot, P.V.; Schmitt, J.; Pape, J.; Biggerstaff, B.J.; et al. West Nile virus-associated flaccid paralysis. Emerg. Infect. Dis. 2005, 11, 1021–1027. [Google Scholar] [CrossRef]
- Centers for Disease Control and Prevention (CDC). Acute flaccid paralysis syndrome associated with West Nile virus infection—Mississippi and Louisiana, July–August 2002. MMWR Morb. Mortal. Wkly. Rep. 2002, 51, 825–828. [Google Scholar]
- Hart, J., Jr.; Tillman, G.; Kraut, M.A.; Chiang, H.S.; Strain, J.F.; Li, Y.; Agrawal, A.G.; Jester, P.; Gnann, J.W., Jr.; Whitley, R.J.; et al. West Nile virus neuroinvasive disease: Neurological manifestations and prospective longitudinal outcomes. BMC Infect. Dis. 2014, 14, 248. [Google Scholar] [CrossRef] [PubMed]
- Creswell, A.; Connor, C.M.; Ko, R.; Tu, S.; Karim, S.; Lui, F. Acute Flaccid Myelitis Caused by West Nile Virus: A Case Report and Neuroimaging Correlate. Cureus 2024, 16, e70107. [Google Scholar] [CrossRef]
- Kallas, E.G.; Wilder-Smith, A. Managing severe yellow fever in the intensive care: Lessons learnt from Brazil. J. Travel Med. 2019, 26, taz043. [Google Scholar] [CrossRef]
- de Avila, R.E.; Jose Fernandes, H.; Barbosa, G.M.; Araujo, A.L.; Gomes, T.C.C.; Barros, T.G.; Moreira, R.L.F.; Silva, G.L.C.; de Oliveira, N.R. Clinical profiles and factors associated with mortality in adults with yellow fever admitted to an intensive care unit in Minas Gerais, Brazil. Int. J. Infect. Dis. 2020, 93, 90–97. [Google Scholar] [CrossRef]
- Ribeiro, A.F.; Cavalin, R.F.; Abdul Hamid Suleiman, J.M.; Alves da Costa, J.; Januaria de Vasconcelos, M.; Sant’Ana Malaque, C.M.; Sztajnbok, J. Yellow Fever: Factors Associated with Death in a Hospital of Reference in Infectious Diseases, Sao Paulo, Brazil, 2018. Am. J. Trop. Med. Hyg. 2019, 101, 180–188. [Google Scholar] [CrossRef]
- Kallas, E.G.; D’Elia Zanella, L.; Moreira, C.H.V.; Buccheri, R.; Diniz, G.B.F.; Castineiras, A.C.P.; Costa, P.R.; Dias, J.Z.C.; Marmorato, M.P.; Song, A.T.W.; et al. Predictors of mortality in patients with yellow fever: An observational cohort study. Lancet Infect. Dis. 2019, 19, 750–758. [Google Scholar] [CrossRef] [PubMed]
- Solomon, T.; Dung, N.M.; Kneen, R.; Thao, L.T.T.; Gainsborough, M.; Nisalak, A.; Day, N.P.; Kirkham, F.J.; Vaughn, D.W.; Smith, S.; et al. Seizures and raised intracranial pressure in Vietnamese patients with Japanese encephalitis. Brain 2002, 125, 1084–1093. [Google Scholar] [CrossRef]
- Van Tan, L.; Qui, P.T.; Ha, D.Q.; Hue, N.B.; Bao, L.Q.; Van Cam, B.; Khanh, T.H.; Hien, T.T.; Chau, N.V.V.; Tram, T.T.; et al. Viral etiology of encephalitis in children in southern Vietnam: Results of a one-year prospective descriptive study. PLoS Neglected Trop. Dis. 2010, 4, e854. [Google Scholar] [CrossRef] [PubMed]
- Quan, T.M.; Thao, T.T.N.; Duy, N.M.; Nhat, T.M.; Clapham, H. Estimates of the global burden of Japanese encephalitis and the impact of vaccination from 2000-2015. eLife 2020, 9, e51027. [Google Scholar] [CrossRef]
- Sunwoo, J.S.; Lee, S.T.; Jung, K.H.; Park, K.I.; Moon, J.; Jung, K.Y.; Kim, M.; Lee, S.K.; Chu, K. Clinical Characteristics of Severe Japanese Encephalitis: A Case Series from South Korea. Am. J. Trop. Med. Hyg. 2017, 97, 369–375. [Google Scholar] [CrossRef]
- Kalita, J.; Misra, U.K.; Pandey, S.; Dhole, T.N. A comparison of clinical and radiological findings in adults and children with Japanese encephalitis. Arch. Neurol. 2003, 60, 1760–1764. [Google Scholar] [CrossRef]
- Misra, U.K.; Kalita, J. Seizures in Japanese encephalitis. J. Neurol. Sci. 2001, 190, 57–60. [Google Scholar] [CrossRef]
- Brasil, P.; Pereira, J.P., Jr.; Moreira, M.E.; Ribeiro Nogueira, R.M.; Damasceno, L.; Wakimoto, M.; Rabello, R.S.; Valderramos, S.G.; Halai, U.A.; Salles, T.S.; et al. Zika Virus Infection in Pregnant Women in Rio de Janeiro. N. Engl. J. Med. 2016, 375, 2321–2334. [Google Scholar] [CrossRef]
- Calvet, G.; Aguiar, R.S.; Melo, A.S.O.; Sampaio, S.A.; de Filippis, I.; Fabri, A.; Araujo, E.S.M.; de Sequeira, P.C.; de Mendonca, M.C.L.; de Oliveira, L.; et al. Detection and sequencing of Zika virus from amniotic fluid of fetuses with microcephaly in Brazil: A case study. Lancet Infect. Dis. 2016, 16, 653–660. [Google Scholar] [CrossRef] [PubMed]
- Marban-Castro, E.; Gonce, A.; Fumado, V.; Romero-Acevedo, L.; Bardaji, A. Zika virus infection in pregnant women and their children: A review. Eur. J. Obstet. Gynecol. Reprod. Biol. 2021, 265, 162–168. [Google Scholar] [CrossRef]
- Souza, J.P.; Meio, M.; de Andrade, L.M.; Figueiredo, M.R.; Gomes Junior, S.C.; Pereira Junior, J.P.; Brickley, E.; Lopes Moreira, M.E. Adverse fetal and neonatal outcomes in pregnancies with confirmed Zika Virus infection in Rio de Janeiro, Brazil: A cohort study. PLoS Neglected Trop. Dis. 2021, 15, e0008893. [Google Scholar] [CrossRef]
- Grant, R.; Flechelles, O.; Tressieres, B.; Dialo, M.; Elenga, N.; Mediamolle, N.; Mallard, A.; Hebert, J.C.; Lachaume, N.; Couchy, E.; et al. In utero Zika virus exposure and neurodevelopment at 24 months in toddlers normocephalic at birth: A cohort study. BMC Med. 2021, 19, 12. [Google Scholar] [CrossRef] [PubMed]
- Hoen, B.; Schaub, B.; Funk, A.L.; Ardillon, V.; Boullard, M.; Cabie, A.; Callier, C.; Carles, G.; Cassadou, S.; Cesaire, R.; et al. Pregnancy Outcomes after ZIKV Infection in French Territories in the Americas. N. Engl. J. Med. 2018, 378, 985–994. [Google Scholar] [CrossRef] [PubMed]
- Pomar, L.; Vouga, M.; Lambert, V.; Pomar, C.; Hcini, N.; Jolivet, A.; Benoist, G.; Rousset, D.; Matheus, S.; Malinger, G.; et al. Maternal-fetal transmission and adverse perinatal outcomes in pregnant women infected with Zika virus: Prospective cohort study in French Guiana. BMJ 2018, 363, k4431. [Google Scholar] [CrossRef] [PubMed]
- Davies, A.J.; Lleixa, C.; Siles, A.M.; Gourlay, D.S.; Berridge, G.; Dejnirattisai, W.; Ramirez-Santana, C.; Anaya, J.M.; Falconar, A.K.; Romero-Vivas, C.M.; et al. Guillain-Barre Syndrome Following Zika Virus Infection Is Associated With a Diverse Spectrum of Peripheral Nerve Reactive Antibodies. Neurol. Neuroimmunol. Neuroinflammation 2023, 10, e200047. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.; Beckham, J.D.; Piquet, A.L.; Tyler, K.L.; Pastula, D.M. An Overview of Powassan Virus Disease. Neurohospitalist 2019, 9, 181–182. [Google Scholar] [CrossRef]
- Bogovic, P.; Kastrin, A.; Lotric-Furlan, S.; Ogrinc, K.; Avsic Zupanc, T.; Korva, M.; Knap, N.; Resman Rus, K.; Strle, K.; Strle, F. Comparison of laboratory and immune characteristics of the initial and second phase of tick-borne encephalitis. Emerg. Microbes Infect. 2022, 11, 1647–1656. [Google Scholar] [CrossRef]
- Bogovic, P.; Logar, M.; Avsic-Zupanc, T.; Strle, F.; Lotric-Furlan, S. Quantitative evaluation of the severity of acute illness in adult patients with tick-borne encephalitis. BioMed Res. Int. 2014, 2014, 841027. [Google Scholar] [CrossRef]
- Kaiser, R. Tick-borne encephalitis (TBE) in Germany and clinical course of the disease. Int. J. Med. Microbiol. 2002, 291 (Suppl. 33), 58–61. [Google Scholar] [CrossRef]
- Piantadosi, A.; Rubin, D.B.; McQuillen, D.P.; Hsu, L.; Lederer, P.A.; Ashbaugh, C.D.; Duffalo, C.; Duncan, R.; Thon, J.; Bhattacharyya, S.; et al. Emerging Cases of Powassan Virus Encephalitis in New England: Clinical Presentation, Imaging, and Review of the Literature. Clin. Infect. Dis. 2016, 62, 707–713. [Google Scholar] [CrossRef]
- Mendoza, M.A.; Hass, R.M.; Vaillant, J.; Johnson, D.R.; Theel, E.S.; Toledano, M.; Abu Saleh, O. Powassan Virus Encephalitis: A Tertiary Center Experience. Clin. Infect. Dis. 2024, 78, 80–89. [Google Scholar] [CrossRef]
- Ndukwe, C.; Melville, A.C.; Osman, M.; Mohammed, Y.; Oduro, M.; Ankrah, P.K. Neurological Complications Associated With the Powassan Virus and Treatment Interventions. Cureus 2024, 16, e71780. [Google Scholar] [CrossRef]
- Charrel, R.N.; Attoui, H.; Butenko, A.M.; Clegg, J.C.; Deubel, V.; Frolova, T.V.; Gould, E.A.; Gritsun, T.S.; Heinz, F.X.; Labuda, M.; et al. Tick-borne virus diseases of human interest in Europe. Clin. Microbiol. Infect. 2004, 10, 1040–1055. [Google Scholar] [CrossRef] [PubMed]
- Baasandavga, U.; Badrakh, B.; Burged, N.; Davaajav, O.; Khurelsukh, T.; Barnes, A.; Ulaankhuu, U.; Nyamdorj, T. A case series of fatal meningoencephalitis in Mongolia: Epidemiological and molecular characteristics of tick-borne encephalitis virus. West. Pac. Surveill. Response J. 2019, 10, 25–31. [Google Scholar] [CrossRef] [PubMed]
- Varnaite, R.; Gredmark-Russ, S.; Klingstrom, J. Deaths from Tick-Borne Encephalitis, Sweden. Emerg. Infect. Dis. 2022, 28, 1471–1474. [Google Scholar] [CrossRef]
- Mladinich, M.C.; Himmler, G.E.; Conde, J.N.; Gorbunova, E.E.; Schutt, W.R.; Sarkar, S.; Tsirka, S.A.; Kim, H.K.; Mackow, E.R. Age-dependent Powassan virus lethality is linked to glial cell activation and divergent neuroinflammatory cytokine responses in a murine model. J. Virol. 2024, 98, e0056024. [Google Scholar] [CrossRef]
- Kemenesi, G.; Banyai, K. Tick-Borne Flaviviruses, with a Focus on Powassan Virus. Clin. Microbiol. Rev. 2019, 32, e00106-17. [Google Scholar] [CrossRef]
- Hall, C.; Khodr, Z.G.; Chang, R.N.; Bukowinski, A.T.; Gumbs, G.R.; Conlin, A.M.S. Safety of yellow fever vaccination in pregnancy: Findings from a cohort of active duty US military women. J. Travel. Med. 2020, 27, taaa138. [Google Scholar] [CrossRef]
- Roukens, A.H.; Soonawala, D.; Joosten, S.A.; de Visser, A.W.; Jiang, X.; Dirksen, K.; de Gruijter, M.; van Dissel, J.T.; Bredenbeek, P.J.; Visser, L.G. Elderly subjects have a delayed antibody response and prolonged viraemia following yellow fever vaccination: A prospective controlled cohort study. PLoS ONE 2011, 6, e27753. [Google Scholar] [CrossRef]
- Khodr, Z.G.; Hall, C.; Chang, R.N.; Bukowinski, A.T.; Gumbs, G.R.; Conlin, A.M.S. Japanese encephalitis vaccination in pregnancy among U.S. active duty military women. Vaccine 2020, 38, 4529–4535. [Google Scholar] [CrossRef] [PubMed]
- Hadinegoro, S.R.; Arredondo-Garcia, J.L.; Capeding, M.R.; Deseda, C.; Chotpitayasunondh, T.; Dietze, R.; Muhammad Ismail, H.I.; Reynales, H.; Limkittikul, K.; Rivera-Medina, D.M.; et al. Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease. N. Engl. J. Med. 2015, 373, 1195–1206. [Google Scholar] [CrossRef]
- Sridhar, S.; Luedtke, A.; Langevin, E.; Zhu, M.; Bonaparte, M.; Machabert, T.; Savarino, S.; Zambrano, B.; Moureau, A.; Khromava, A.; et al. Effect of Dengue Serostatus on Dengue Vaccine Safety and Efficacy. N. Engl. J. Med. 2018, 379, 327–340. [Google Scholar] [CrossRef]
- Caminade, C.; McIntyre, K.M.; Jones, A.E. Impact of recent and future climate change on vector-borne diseases. Ann. N. Y. Acad. Sci. 2019, 1436, 157–173. [Google Scholar] [CrossRef] [PubMed]
- Harrigan, R.J.; Thomassen, H.A.; Buermann, W.; Smith, T.B. A continental risk assessment of West Nile virus under climate change. Glob. Change Biol. 2014, 20, 2417–2425. [Google Scholar] [CrossRef]
- Liu-Helmersson, J.; Quam, M.; Wilder-Smith, A.; Stenlund, H.; Ebi, K.; Massad, E.; Rocklov, J. Climate Change and Aedes Vectors: 21st Century Projections for Dengue Transmission in Europe. eBioMedicine 2016, 7, 267–277. [Google Scholar] [CrossRef]
- Butterworth, M.K.; Morin, C.W.; Comrie, A.C. An Analysis of the Potential Impact of Climate Change on Dengue Transmission in the Southeastern United States. Environ. Health Perspect. 2017, 125, 579–585. [Google Scholar] [CrossRef] [PubMed]
- Lourenco, J.; Maia de Lima, M.; Faria, N.R.; Walker, A.; Kraemer, M.U.; Villabona-Arenas, C.J.; Lambert, B.; Marques de Cerqueira, E.; Pybus, O.G.; Alcantara, L.C.; et al. Epidemiological and ecological determinants of Zika virus transmission in an urban setting. eLife 2017, 6, e29820. [Google Scholar] [CrossRef] [PubMed]
- Iwamura, T.; Guzman-Holst, A.; Murray, K.A. Accelerating invasion potential of disease vector Aedes aegypti under climate change. Nat. Commun. 2020, 11, 2130. [Google Scholar] [CrossRef]
- Haider, N.; Hasan, M.N.; Onyango, J.; Billah, M.; Khan, S.; Papakonstantinou, D.; Paudyal, P.; Asaduzzaman, M. Global dengue epidemic worsens with record 14 million cases and 9000 deaths reported in 2024. Int. J. Infect. Dis. 2025, 158, 107940. [Google Scholar] [CrossRef]
- Sansone, N.M.S.; Boschiero, M.N.; Marson, F.A.L. Dengue outbreaks in Brazil and Latin America: The new and continuing challenges. Int. J. Infect. Dis. 2024, 147, 107192. [Google Scholar] [CrossRef]
- Moore, S.M. The current burden of Japanese encephalitis and the estimated impacts of vaccination: Combining estimates of the spatial distribution and transmission intensity of a zoonotic pathogen. PLoS Neglected Trop. Dis. 2021, 15, e0009385. [Google Scholar] [CrossRef]
- McGuinness, S.L.; Lau, C.L.; Leder, K. The evolving Japanese encephalitis situation in Australia and implications for travel medicine. J. Travel Med. 2023, 30, taad029. [Google Scholar] [CrossRef]
- Bruce, R.C.; Abbott, A.J.; Jones, B.P.; Gardner, B.L.; Gonzalez, E.; Ionescu, A.M.; Jagdev, M.; Jenkins, A.; Santos, M.; Seilern-Macpherson, K.; et al. Detection of West Nile virus via retrospective mosquito arbovirus surveillance, United Kingdom, 2025. Eurosurveillance 2025, 30, 2500401. [Google Scholar] [CrossRef]
- Taheri, S.; Gonzalez, M.A.; Ruiz-Lopez, M.J.; Soriguer, R.; Figuerola, J. Patterns of West Nile virus vector co-occurrence and spatial overlap with human cases across Europe. One Health 2025, 20, 101041. [Google Scholar] [CrossRef] [PubMed]
- Laverdeur, J.; Amory, H.; Beckers, P.; Desmecht, D.; Francis, F.; Garigliany, M.-M.; Hayette, M.-P.; Linden, A.; Darcis, G. West Nile and Usutu viruses: Current spreading and future threats in a warming northern Europe. Front. Virol. 2025, 5, 2025. [Google Scholar] [CrossRef]
- Van Heuverswyn, J.; Hallmaier-Wacker, L.K.; Beaute, J.; Gomes Dias, J.; Haussig, J.M.; Busch, K.; Kerlik, J.; Markowicz, M.; Makela, H.; Nygren, T.M.; et al. Spatiotemporal spread of tick-borne encephalitis in the EU/EEA, 2012 to 2020. Eurosurveillance 2023, 28, 2200543. [Google Scholar] [CrossRef]
- Kelly, P.H.; Kwark, R.; Marick, H.M.; Davis, J.; Stark, J.H.; Madhava, H.; Dobler, G.; Moisi, J.C. Different environmental factors predict the occurrence of tick-borne encephalitis virus (TBEV) and reveal new potential risk areas across Europe via geospatial models. Int. J. Health Geogr. 2025, 24, 3. [Google Scholar] [CrossRef]
- Beerlage-de Jong, N.; Blanford, J. Mapping the risk of tick-borne encephalitis in Europe for informed vaccination decisions. J. Travel. Med. 2025, 32, taae153. [Google Scholar] [CrossRef] [PubMed]
- Ferrari, G.; Rosso, F.; Girardi, M.; Dagostin, F.; Arnoldi, D.; Zuccali, M.G.; Mocellin, C.; Molinaro, S.; Tagliapietra, V.; Rizzoli, A. A new hotspot of tick-borne encephalitis virus (TBEV) in the Autonomous Province of Trento, Italy. Ticks Tick Borne Dis. 2025, 16, 102513. [Google Scholar] [CrossRef] [PubMed]
- Kraemer, M.U.G.; Reiner, R.C., Jr.; Brady, O.J.; Messina, J.P.; Gilbert, M.; Pigott, D.M.; Yi, D.; Johnson, K.; Earl, L.; Marczak, L.B.; et al. Past and future spread of the arbovirus vectors Aedes aegypti and Aedes albopictus. Nat. Microbiol. 2019, 4, 854–863. [Google Scholar] [CrossRef]
- Samy, A.M.; Elaagip, A.H.; Kenawy, M.A.; Ayres, C.F.; Peterson, A.T.; Soliman, D.E. Climate Change Influences on the Global Potential Distribution of the Mosquito Culex quinquefasciatus, Vector of West Nile Virus and Lymphatic Filariasis. PLoS ONE 2016, 11, e0163863. [Google Scholar] [CrossRef]
- Tonk-Rugen, M.; Kratou, M.; Cabezas-Cruz, A. A Warming World, a Growing Threat: The Spread of Ticks and Emerging Tick-Borne Diseases. Pathogens 2025, 14, 213. [Google Scholar] [CrossRef]
- Smit, J.M.; Moesker, B.; Rodenhuis-Zybert, I.; Wilschut, J. Flavivirus cell entry and membrane fusion. Viruses 2011, 3, 160–171. [Google Scholar] [CrossRef]
- Kaufmann, B.; Rossmann, M.G. Molecular mechanisms involved in the early steps of flavivirus cell entry. Microbes Infect. 2011, 13, 1–9. [Google Scholar] [CrossRef]
- Krishnan, M.N.; Sukumaran, B.; Pal, U.; Agaisse, H.; Murray, J.L.; Hodge, T.W.; Fikrig, E. Rab 5 is required for the cellular entry of dengue and West Nile viruses. J. Virol. 2007, 81, 4881–4885. [Google Scholar] [CrossRef]
- Persaud, M.; Martinez-Lopez, A.; Buffone, C.; Porcelli, S.A.; Diaz-Griffero, F. Infection by Zika viruses requires the transmembrane protein AXL, endocytosis and low pH. Virology 2018, 518, 301–312. [Google Scholar] [CrossRef] [PubMed]
- Bai, F.; Town, T.; Pradhan, D.; Cox, J.; Ashish; Ledizet, M.; Anderson, J.F.; Flavell, R.A.; Krueger, J.K.; Koski, R.A.; et al. Antiviral peptides targeting the west nile virus envelope protein. J. Virol. 2007, 81, 2047–2055. [Google Scholar] [CrossRef]
- Mary, J.A.; Jittmittraphap, A.; Chattanadee, S.; Leaungwutiwong, P.; Shenbagarathai, R. A synthetic peptide derived from domain III envelope glycoprotein of Dengue virus induces neutralizing antibody. Virus Genes 2018, 54, 25–32. [Google Scholar] [CrossRef] [PubMed]
- Gallichotte, E.N.; Young, E.F.; Baric, T.J.; Yount, B.L.; Metz, S.W.; Begley, M.C.; de Silva, A.M.; Baric, R.S. Role of Zika Virus Envelope Protein Domain III as a Target of Human Neutralizing Antibodies. mBio 2019, 10, e01485-19. [Google Scholar] [CrossRef]
- Chen, Y.; Maguire, T.; Hileman, R.E.; Fromm, J.R.; Esko, J.D.; Linhardt, R.J.; Marks, R.M. Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nat. Med. 1997, 3, 866–871. [Google Scholar] [CrossRef]
- Chiu, M.W.; Yang, Y.L. Blocking the dengue virus 2 infections on BHK-21 cells with purified recombinant dengue virus 2 E protein expressed in Escherichia coli. Biochem. Biophys. Res. Commun. 2003, 309, 672–678. [Google Scholar] [CrossRef]
- Dutta, S.K.; Langenburg, T. A Perspective on Current Flavivirus Vaccine Development: A Brief Review. Viruses 2023, 15, 860. [Google Scholar] [CrossRef] [PubMed]
- Carbaugh, D.L.; Lazear, H.M. Flavivirus Envelope Protein Glycosylation: Impacts on Viral Infection and Pathogenesis. J. Virol. 2020, 94, e00104-20. [Google Scholar] [CrossRef]
- Beasley, D.W.; Whiteman, M.C.; Zhang, S.; Huang, C.Y.; Schneider, B.S.; Smith, D.R.; Gromowski, G.D.; Higgs, S.; Kinney, R.M.; Barrett, A.D. Envelope protein glycosylation status influences mouse neuroinvasion phenotype of genetic lineage 1 West Nile virus strains. J. Virol. 2005, 79, 8339–8347. [Google Scholar] [CrossRef]
- Fontes-Garfias, C.R.; Shan, C.; Luo, H.; Muruato, A.E.; Medeiros, D.B.A.; Mays, E.; Xie, X.; Zou, J.; Roundy, C.M.; Wakamiya, M.; et al. Functional Analysis of Glycosylation of Zika Virus Envelope Protein. Cell Rep. 2017, 21, 1180–1190. [Google Scholar] [CrossRef]
- Liang, J.J.; Chou, M.W.; Lin, Y.L. DC-SIGN Binding Contributed by an Extra N-Linked Glycosylation on Japanese Encephalitis Virus Envelope Protein Reduces the Ability of Viral Brain Invasion. Front. Cell. Infect. Microbiol. 2018, 8, 239. [Google Scholar] [CrossRef]
- Pierson, T.C.; Diamond, M.S. Degrees of maturity: The complex structure and biology of flaviviruses. Curr. Opin. Virol. 2012, 2, 168–175. [Google Scholar] [CrossRef]
- Zhang, X.; Zhang, Y.; Jia, R.; Wang, M.; Yin, Z.; Cheng, A. Structure and function of capsid protein in flavivirus infection and its applications in the development of vaccines and therapeutics. Vet. Res. 2021, 52, 98. [Google Scholar] [CrossRef]
- Donaldson, M.K.; Zanders, L.A.; Jose, J. Functional Roles and Host Interactions of Orthoflavivirus Non-Structural Proteins During Replication. Pathogens 2025, 14, 184. [Google Scholar] [CrossRef] [PubMed]
- van den Elsen, K.; Quek, J.P.; Luo, D. Molecular Insights into the Flavivirus Replication Complex. Viruses 2021, 13, 956. [Google Scholar] [CrossRef] [PubMed]
- Gillespie, L.K.; Hoenen, A.; Morgan, G.; Mackenzie, J.M. The endoplasmic reticulum provides the membrane platform for biogenesis of the flavivirus replication complex. J. Virol. 2010, 84, 10438–10447. [Google Scholar] [CrossRef] [PubMed]
- Youn, S.; Ambrose, R.L.; Mackenzie, J.M.; Diamond, M.S. Non-structural protein-1 is required for West Nile virus replication complex formation and viral RNA synthesis. Virol. J. 2013, 10, 339. [Google Scholar] [CrossRef]
- Lindenbach, B.D.; Rice, C.M. Genetic interaction of flavivirus nonstructural proteins NS1 and NS4A as a determinant of replicase function. J. Virol. 1999, 73, 4611–4621. [Google Scholar] [CrossRef]
- Ci, Y.; Liu, Z.Y.; Zhang, N.N.; Niu, Y.; Yang, Y.; Xu, C.; Yang, W.; Qin, C.F.; Shi, L. Zika NS1-induced ER remodeling is essential for viral replication. J. Cell Biol. 2020, 219, e201903062. [Google Scholar] [CrossRef]
- Apte-Sengupta, S.; Sirohi, D.; Kuhn, R.J. Coupling of replication and assembly in flaviviruses. Curr. Opin. Virol. 2014, 9, 134–142. [Google Scholar] [CrossRef]
- Zhang, Y.; Kaufmann, B.; Chipman, P.R.; Kuhn, R.J.; Rossmann, M.G. Structure of immature West Nile virus. J. Virol. 2007, 81, 6141–6145. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Corver, J.; Chipman, P.R.; Zhang, W.; Pletnev, S.V.; Sedlak, D.; Baker, T.S.; Strauss, J.H.; Kuhn, R.J.; Rossmann, M.G. Structures of immature flavivirus particles. EMBO J. 2003, 22, 2604–2613. [Google Scholar] [CrossRef] [PubMed]
- Heinz, F.X.; Stiasny, K.; Puschner-Auer, G.; Holzmann, H.; Allison, S.L.; Mandl, C.W.; Kunz, C. Structural changes and functional control of the tick-borne encephalitis virus glycoprotein E by the heterodimeric association with protein prM. Virology 1994, 198, 109–117. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Lok, S.M.; Yu, I.M.; Zhang, Y.; Kuhn, R.J.; Chen, J.; Rossmann, M.G. The flavivirus precursor membrane-envelope protein complex: Structure and maturation. Science 2008, 319, 1830–1834. [Google Scholar] [CrossRef]
- Yu, I.M.; Zhang, W.; Holdaway, H.A.; Li, L.; Kostyuchenko, V.A.; Chipman, P.R.; Kuhn, R.J.; Rossmann, M.G.; Chen, J. Structure of the immature dengue virus at low pH primes proteolytic maturation. Science 2008, 319, 1834–1837. [Google Scholar] [CrossRef]
- Yu, I.M.; Holdaway, H.A.; Chipman, P.R.; Kuhn, R.J.; Rossmann, M.G.; Chen, J. Association of the pr peptides with dengue virus at acidic pH blocks membrane fusion. J. Virol. 2009, 83, 12101–12107. [Google Scholar] [CrossRef]
- Renner, M.; Dejnirattisai, W.; Carrique, L.; Martin, I.S.; Karia, D.; Ilca, S.L.; Ho, S.F.; Kotecha, A.; Keown, J.R.; Mongkolsapaya, J.; et al. Flavivirus maturation leads to the formation of an occupied lipid pocket in the surface glycoproteins. Nat. Commun. 2021, 12, 1238. [Google Scholar] [CrossRef]
- Zheng, A.; Umashankar, M.; Kielian, M. In vitro and in vivo studies identify important features of dengue virus pr-E protein interactions. PLoS Pathog. 2010, 6, e1001157. [Google Scholar] [CrossRef]
- Zhang, X.; Ge, P.; Yu, X.; Brannan, J.M.; Bi, G.; Zhang, Q.; Schein, S.; Zhou, Z.H. Cryo-EM structure of the mature dengue virus at 3.5-A resolution. Nat. Struct. Mol. Biol. 2013, 20, 105–110. [Google Scholar] [CrossRef]
- Vaney, M.C.; Dellarole, M.; Duquerroy, S.; Medits, I.; Tsouchnikas, G.; Rouvinski, A.; England, P.; Stiasny, K.; Heinz, F.X.; Rey, F.A. Evolution and activation mechanism of the flavivirus class II membrane-fusion machinery. Nat. Commun. 2022, 13, 3718. [Google Scholar] [CrossRef]
- Crampon, E.; Covernton, E.; Vaney, M.C.; Dellarole, M.; Sommer, S.; Sharma, A.; Haouz, A.; England, P.; Lepault, J.; Duquerroy, S.; et al. New insight into flavivirus maturation from structure/function studies of the yellow fever virus envelope protein complex. mBio 2023, 14, e0070623. [Google Scholar] [CrossRef]
- Majowicz, S.A.; Narayanan, A.; Moustafa, I.M.; Bator, C.M.; Hafenstein, S.L.; Jose, J. Zika virus M protein latches and locks the E protein from transitioning to an immature state after prM cleavage. npj Viruses 2023, 1, 4. [Google Scholar] [CrossRef] [PubMed]
- Holoubek, J.; Salat, J.; Matkovic, M.; Bednar, P.; Novotny, P.; Hradilek, M.; Majerova, T.; Rosendal, E.; Eyer, L.; Fortova, A.; et al. Irreversible furin cleavage site exposure renders immature tick-borne flaviviruses fully infectious. Nat. Commun. 2025, 16, 7491. [Google Scholar] [CrossRef]
- Metz, S.W.; Thomas, A.; Brackbill, A.; Forsberg, J.; Miley, M.J.; Lopez, C.A.; Lazear, H.M.; Tian, S.; de Silva, A.M. Oligomeric state of the ZIKV E protein defines protective immune responses. Nat. Commun. 2019, 10, 4606. [Google Scholar] [CrossRef] [PubMed]
- Colpitts, T.M.; Rodenhuis-Zybert, I.; Moesker, B.; Wang, P.; Fikrig, E.; Smit, J.M. prM-antibody renders immature West Nile virus infectious in vivo. J. Gen. Virol. 2011, 92, 2281–2285. [Google Scholar] [CrossRef]
- Smith, S.A.; Nivarthi, U.K.; de Alwis, R.; Kose, N.; Sapparapu, G.; Bombardi, R.; Kahle, K.M.; Pfaff, J.M.; Lieberman, S.; Doranz, B.J.; et al. Dengue Virus prM-Specific Human Monoclonal Antibodies with Virus Replication-Enhancing Properties Recognize a Single Immunodominant Antigenic Site. J. Virol. 2016, 90, 780–789. [Google Scholar] [CrossRef] [PubMed]
- Slon-Campos, J.L.; Dejnirattisai, W.; Jagger, B.W.; Lopez-Camacho, C.; Wongwiwat, W.; Durnell, L.A.; Winkler, E.S.; Chen, R.E.; Reyes-Sandoval, A.; Rey, F.A.; et al. A protective Zika virus E-dimer-based subunit vaccine engineered to abrogate antibody-dependent enhancement of dengue infection. Nat. Immunol. 2019, 20, 1291–1298. [Google Scholar] [CrossRef] [PubMed]
- Shukla, R.; Shanmugam, R.K.; Ramasamy, V.; Arora, U.; Batra, G.; Acklin, J.A.; Krammer, F.; Lim, J.K.; Swaminathan, S.; Khanna, N. Zika virus envelope nanoparticle antibodies protect mice without risk of disease enhancement. eBioMedicine 2020, 54, 102738. [Google Scholar] [CrossRef] [PubMed]
- Calvert, A.E.; Huang, C.Y.; Blair, C.D.; Roehrig, J.T. Mutations in the West Nile prM protein affect VLP and virion secretion in vitro. Virology 2012, 433, 35–44. [Google Scholar] [CrossRef]
- Wang, Y.; Si, L.L.; Guo, X.L.; Cui, G.H.; Fang, D.Y.; Zhou, J.M.; Yan, H.J.; Jiang, L.F. Substitution of the precursor peptide prevents anti-prM antibody-mediated antibody-dependent enhancement of dengue virus infection. Virus Res. 2017, 229, 57–64. [Google Scholar] [CrossRef] [PubMed]
- Pinto, P.B.A.; Barros, T.A.C.; Lima, L.M.; Pacheco, A.R.; Assis, M.L.; Pereira, B.A.S.; Goncalves, A.J.S.; Azevedo, A.S.; Neves-Ferreira, A.G.C.; Costa, S.M.; et al. Combination of E- and NS1-Derived DNA Vaccines: The Immune Response and Protection Elicited in Mice against DENV2. Viruses 2022, 14, 1452. [Google Scholar] [CrossRef] [PubMed]
- Carpio, K.L.; Barrett, A.D.T. Flavivirus NS1 and Its Potential in Vaccine Development. Vaccines 2021, 9, 622. [Google Scholar] [CrossRef]
- Rey, F.A.; Stiasny, K.; Vaney, M.C.; Dellarole, M.; Heinz, F.X. The bright and the dark side of human antibody responses to flaviviruses: Lessons for vaccine design. EMBO Rep. 2018, 19, 206–224. [Google Scholar] [CrossRef]
- Akey, D.L.; Brown, W.C.; Dutta, S.; Konwerski, J.; Jose, J.; Jurkiw, T.J.; DelProposto, J.; Ogata, C.M.; Skiniotis, G.; Kuhn, R.J.; et al. Flavivirus NS1 structures reveal surfaces for associations with membranes and the immune system. Science 2014, 343, 881–885. [Google Scholar] [CrossRef]
- van den Elsen, K.; Chew, B.L.A.; Ho, J.S.; Luo, D. Flavivirus nonstructural proteins and replication complexes as antiviral drug targets. Curr. Opin. Virol. 2023, 59, 101305. [Google Scholar] [CrossRef]
- Verhaegen, M.; Vermeire, K. The endoplasmic reticulum (ER): A crucial cellular hub in flavivirus infection and potential target site for antiviral interventions. npj Viruses 2024, 2, 24. [Google Scholar] [CrossRef]
- Bailey, M.J.; Broecker, F.; Duehr, J.; Arumemi, F.; Krammer, F.; Palese, P.; Tan, G.S. Antibodies Elicited by an NS1-Based Vaccine Protect Mice against Zika Virus. mBio 2019, 10, e02861-18. [Google Scholar] [CrossRef]
- Tien, S.M.; Chang, P.C.; Lai, Y.C.; Chuang, Y.C.; Tseng, C.K.; Kao, Y.S.; Huang, H.J.; Hsiao, Y.P.; Liu, Y.L.; Lin, H.H.; et al. Therapeutic efficacy of humanized monoclonal antibodies targeting dengue virus nonstructural protein 1 in the mouse model. PLoS Pathog. 2022, 18, e1010469. [Google Scholar] [CrossRef]
- Chang, G.J.; Kuno, G.; Purdy, D.E.; Davis, B.S. Recent advancement in flavivirus vaccine development. Expert Rev. Vaccines 2004, 3, 199–220. [Google Scholar] [CrossRef]
- Mishra, N.; Boudewijns, R.; Schmid, M.A.; Marques, R.E.; Sharma, S.; Neyts, J.; Dallmeier, K. A Chimeric Japanese Encephalitis Vaccine Protects against Lethal Yellow Fever Virus Infection without Inducing Neutralizing Antibodies. mBio 2020, 11, e02494-19. [Google Scholar] [CrossRef] [PubMed]
- Collins, N.D.; Barrett, A.D. Live Attenuated Yellow Fever 17D Vaccine: A Legacy Vaccine Still Controlling Outbreaks In Modern Day. Curr. Infect. Dis. Rep. 2017, 19, 14. [Google Scholar] [CrossRef]
- Nasveld, P.E.; Ebringer, A.; Elmes, N.; Bennett, S.; Yoksan, S.; Aaskov, J.; McCarthy, K.; Kanesa-thasan, N.; Meric, C.; Reid, M. Long term immunity to live attenuated Japanese encephalitis chimeric virus vaccine: Randomized, double-blind, 5-year phase II study in healthy adults. Hum. Vaccines 2010, 6, 1038–1046. [Google Scholar] [CrossRef] [PubMed]
- Akondy, R.S.; Monson, N.D.; Miller, J.D.; Edupuganti, S.; Teuwen, D.; Wu, H.; Quyyumi, F.; Garg, S.; Altman, J.D.; Del Rio, C.; et al. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J. Immunol. 2009, 183, 7919–7930. [Google Scholar] [CrossRef]
- Salje, H.; Alera, M.T.; Chua, M.N.; Hunsawong, T.; Ellison, D.; Srikiatkhachorn, A.; Jarman, R.G.; Gromowski, G.D.; Rodriguez-Barraquer, I.; Cauchemez, S.; et al. Evaluation of the extended efficacy of the Dengvaxia vaccine against symptomatic and subclinical dengue infection. Nat. Med. 2021, 27, 1395–1400. [Google Scholar] [CrossRef] [PubMed]
- Halstead, S.B.; Thomas, S.J. New Japanese encephalitis vaccines: Alternatives to production in mouse brain. Expert Rev. Vaccines 2011, 10, 355–364. [Google Scholar] [CrossRef]
- Kum, D.B.; Mishra, N.; Boudewijns, R.; Gladwyn-Ng, I.; Alfano, C.; Ma, J.; Schmid, M.A.; Marques, R.E.; Schols, D.; Kaptein, S.; et al. A yellow fever-Zika chimeric virus vaccine candidate protects against Zika infection and congenital malformations in mice. npj Vaccines 2018, 3, 56. [Google Scholar] [CrossRef]
- Van Truong, L.; Thuy, L.T.; Hien, L.T.; Tran, T.Q.M.; Gad, A.; Tran, L.; Aziz, J.M.A.; Ahmed, O.; Mahabir, S.; Tiwari, R.; et al. From controversy to confidence: Strengthening dengue vaccines safety reporting. Vaccine 2025, 62, 127489. [Google Scholar] [CrossRef]
- Shukla, R.; Beesetti, H.; Brown, J.A.; Ahuja, R.; Ramasamy, V.; Shanmugam, R.K.; Poddar, A.; Batra, G.; Krammer, F.; Lim, J.K.; et al. Dengue and Zika virus infections are enhanced by live attenuated dengue vaccine but not by recombinant DSV4 vaccine candidate in mouse models. eBioMedicine 2020, 60, 102991. [Google Scholar] [CrossRef] [PubMed]
- Saez-Llorens, X.; DeAntonio, R.; Low, J.G.H.; Kosalaraksa, P.; Dean, H.; Sharma, M.; Tricou, V.; Biswal, S. TAK-003: Development of a tetravalent dengue vaccine. Expert Rev. Vaccines 2025, 24, 324–338. [Google Scholar] [CrossRef] [PubMed]
- Lee, M.F.; Long, C.M.; Poh, C.L. Current status of the development of dengue vaccines. Vaccine X 2025, 22, 100604. [Google Scholar] [CrossRef]
- Tricou, V.; Yu, D.; Reynales, H.; Biswal, S.; Saez-Llorens, X.; Sirivichayakul, C.; Lopez, P.; Borja-Tabora, C.; Bravo, L.; Kosalaraksa, P.; et al. Long-term efficacy and safety of a tetravalent dengue vaccine (TAK-003): 4.5-year results from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Glob. Health 2024, 12, e257–e270. [Google Scholar] [CrossRef] [PubMed]
- Monath, T.P. Stability of yellow fever vaccine. Dev. Biol. Stand. 1996, 87, 219–225. [Google Scholar]
- Wiggan, O.; Livengood, J.A.; Silengo, S.J.; Kinney, R.M.; Osorio, J.E.; Huang, C.Y.; Stinchcomb, D.T. Novel formulations enhance the thermal stability of live-attenuated flavivirus vaccines. Vaccine 2011, 29, 7456–7462. [Google Scholar] [CrossRef]
- Thomas, R.E.; Lorenzetti, D.L.; Spragins, W.; Jackson, D.; Williamson, T. The safety of yellow fever vaccine 17D or 17DD in children, pregnant women, HIV+ individuals, and older persons: Systematic review. Am. J. Trop. Med. Hyg. 2012, 86, 359–372. [Google Scholar] [CrossRef]
- Monath, T.P.; Vasconcelos, P.F. Yellow fever. J. Clin. Virol. Off. Publ. Pan Am. Soc. Clin. Virol. 2015, 64, 160–173. [Google Scholar] [CrossRef]
- Hernandez, N.; Bucciol, G.; Moens, L.; Le Pen, J.; Shahrooei, M.; Goudouris, E.; Shirkani, A.; Changi-Ashtiani, M.; Rokni-Zadeh, H.; Sayar, E.H.; et al. Inherited IFNAR1 deficiency in otherwise healthy patients with adverse reaction to measles and yellow fever live vaccines. J. Exp. Med. 2019, 216, 2057–2070. [Google Scholar] [CrossRef]
- Paul, L.M.; Carlin, E.R.; Jenkins, M.M.; Tan, A.L.; Barcellona, C.M.; Nicholson, C.O.; Michael, S.F.; Isern, S. Dengue virus antibodies enhance Zika virus infection. Clin. Transl. Immunol. 2016, 5, e117. [Google Scholar] [CrossRef] [PubMed]
- Garg, H.; Yeh, R.; Watts, D.M.; Mehmetoglu-Gurbuz, T.; Resendes, R.; Parsons, B.; Gonzales, F.; Joshi, A. Enhancement of Zika virus infection by antibodies from West Nile virus seropositive individuals with no history of clinical infection. BMC Immunol. 2021, 22, 5. [Google Scholar] [CrossRef]
- Katzelnick, L.C.; Narvaez, C.; Arguello, S.; Lopez Mercado, B.; Collado, D.; Ampie, O.; Elizondo, D.; Miranda, T.; Bustos Carillo, F.; Mercado, J.C.; et al. Zika virus infection enhances future risk of severe dengue disease. Science 2020, 369, 1123–1128. [Google Scholar] [CrossRef]
- Fan, Y.C.; Chiu, H.C.; Chen, L.K.; Chang, G.J.; Chiou, S.S. Formalin Inactivation of Japanese Encephalitis Virus Vaccine Alters the Antigenicity and Immunogenicity of a Neutralization Epitope in Envelope Protein Domain III. PLoS Neglected Trop. Dis. 2015, 9, e0004167. [Google Scholar] [CrossRef] [PubMed]
- Eckels, K.H.; Putnak, R. Formalin-inactivated whole virus and recombinant subunit flavivirus vaccines. Adv. Virus Res. 2003, 61, 395–418. [Google Scholar] [CrossRef]
- Fernandez, S.; Thomas, S.J.; De La Barrera, R.; Im-Erbsin, R.; Jarman, R.G.; Baras, B.; Toussaint, J.F.; Mossman, S.; Innis, B.L.; Schmidt, A.; et al. An adjuvanted, tetravalent dengue virus purified inactivated vaccine candidate induces long-lasting and protective antibody responses against dengue challenge in rhesus macaques. Am. J. Trop. Med. Hyg. 2015, 92, 698–708. [Google Scholar] [CrossRef] [PubMed]
- Stephenson, K.E.; Tan, C.S.; Walsh, S.R.; Hale, A.; Ansel, J.L.; Kanjilal, D.G.; Jaegle, K.; Peter, L.; Borducchi, E.N.; Nkolola, J.P.; et al. Safety and immunogenicity of a Zika purified inactivated virus vaccine given via standard, accelerated, or shortened schedules: A single-centre, double-blind, sequential-group, randomised, placebo-controlled, phase 1 trial. Lancet Infect. Dis. 2020, 20, 1061–1070. [Google Scholar] [CrossRef]
- Modjarrad, K.; Lin, L.; George, S.L.; Stephenson, K.E.; Eckels, K.H.; De La Barrera, R.A.; Jarman, R.G.; Sondergaard, E.; Tennant, J.; Ansel, J.L.; et al. Preliminary aggregate safety and immunogenicity results from three trials of a purified inactivated Zika virus vaccine candidate: Phase 1, randomised, double-blind, placebo-controlled clinical trials. Lancet 2018, 391, 563–571. [Google Scholar] [CrossRef]
- Woods, C.W.; Sanchez, A.M.; Swamy, G.K.; McClain, M.T.; Harrington, L.; Freeman, D.; Poore, E.A.; Slifka, D.K.; Poer DeRaad, D.E.; Amanna, I.J.; et al. An observer blinded, randomized, placebo-controlled, phase I dose escalation trial to evaluate the safety and immunogenicity of an inactivated West Nile virus Vaccine, HydroVax-001, in healthy adults. Vaccine 2019, 37, 4222–4230. [Google Scholar] [CrossRef]
- Vorovitch, M.F.; Tuchynskaya, K.K.; Kruglov, Y.A.; Peunkov, N.S.; Mostipanova, G.F.; Kholodilov, I.S.; Ivanova, A.L.; Fedina, M.P.; Gmyl, L.V.; Morozkin, E.S.; et al. An Inactivated West Nile Virus Vaccine Candidate Based on the Lineage 2 Strain. Vaccines 2024, 12, 1398. [Google Scholar] [CrossRef] [PubMed]
- Yonekawa, M.; Watanabe, T.; Kogawara, O.; Yoshii, C.; Yamaji, M.; Aizawa, M.; Erber, W.; Ito, S.; Jug, B.; Koelch, D.; et al. Phase 3 immunogenicity and safety study of a tick-borne encephalitis vaccine in healthy Japanese participants 1 year of age and older. Vaccine 2024, 42, 3180–3189. [Google Scholar] [CrossRef]
- Pugh, S.J.; Moisi, J.C.; Kundi, M.; Santonja, I.; Erber, W.; Angulo, F.J.; Jodar, L. Effectiveness of two doses of tick-borne encephalitis (TBE) vaccine. J. Travel. Med. 2022, 29, taab193. [Google Scholar] [CrossRef]
- Loew-Baselli, A.; Konior, R.; Pavlova, B.G.; Fritsch, S.; Poellabauer, E.; Maritsch, F.; Harmacek, P.; Krammer, M.; Barrett, P.N.; Ehrlich, H.J.; et al. Safety and immunogenicity of the modified adult tick-borne encephalitis vaccine FSME-IMMUN: Results of two large phase 3 clinical studies. Vaccine 2006, 24, 5256–5263. [Google Scholar] [CrossRef]
- Harabacz, I.; Bock, H.; Jungst, C.; Klockmann, U.; Praus, M.; Weber, R. A randomized phase II study of a new tick-borne encephalitis vaccine using three different doses and two immunization regimens. Vaccine 1992, 10, 145–150. [Google Scholar] [CrossRef]
- Zavadska, D.; Freimane, Z.; Karelis, G.; Ermina, I.; Harper, L.R.; Bender, C.; Zhang, P.; Angulo, F.J.; Erber, W.; Bormane, A.; et al. Effectiveness of Tick-borne Encephalitis Vaccines in Children, Latvia, 2018–2020. Pediatr. Infect. Dis. J. 2023, 42, 927–931. [Google Scholar] [CrossRef]
- Friberg, H.; Gargulak, M.; Kong, A.; Lin, L.; Martinez, L.J.; Schmidt, A.C.; Paris, R.M.; Jarman, R.G.; Diaz, C.; Thomas, S.J.; et al. Characterization of B-cell and T-cell responses to a tetravalent dengue purified inactivated vaccine in healthy adults. npj Vaccines 2022, 7, 132. [Google Scholar] [CrossRef] [PubMed]
- Rendi-Wagner, P.; Paulke-Korinek, M.; Kundi, M.; Wiedermann, U.; Laaber, B.; Kollaritsch, H. Antibody persistence following booster vaccination against tick-borne encephalitis: 3-year post-booster follow-up. Vaccine 2007, 25, 5097–5101. [Google Scholar] [CrossRef]
- Wollner, C.J.; Richner, J.M. mRNA Vaccines against Flaviviruses. Vaccines 2021, 9, 148. [Google Scholar] [CrossRef] [PubMed]
- Sun, N.; Su, Z.; Zheng, X. Research progress of mosquito-borne virus mRNA vaccines. Mol. Ther. Methods Clin. Dev. 2025, 33, 101398. [Google Scholar] [CrossRef]
- Chaudhary, N.; Weissman, D.; Whitehead, K.A. mRNA vaccines for infectious diseases: Principles, delivery and clinical translation. Nat. Rev. Drug Discov. 2021, 20, 817–838. [Google Scholar] [CrossRef] [PubMed]
- Maruggi, G.; Zhang, C.; Li, J.; Ulmer, J.B.; Yu, D. mRNA as a Transformative Technology for Vaccine Development to Control Infectious Diseases. Mol. Ther. 2019, 27, 757–772. [Google Scholar] [CrossRef]
- Kariko, K.; Buckstein, M.; Ni, H.; Weissman, D. Suppression of RNA recognition by Toll-like receptors: The impact of nucleoside modification and the evolutionary origin of RNA. Immunity 2005, 23, 165–175. [Google Scholar] [CrossRef] [PubMed]
- Wollner, C.J.; Richner, M.; Hassert, M.A.; Pinto, A.K.; Brien, J.D.; Richner, J.M. A Dengue Virus Serotype 1 mRNA-LNP Vaccine Elicits Protective Immune Responses. J. Virol. 2021, 95, e02482-20. [Google Scholar] [CrossRef] [PubMed]
- He, L.; Sun, W.; Yang, L.; Liu, W.; Li, J. A multiple-target mRNA-LNP vaccine induces protective immunity against experimental multi-serotype DENV in mice. Virol. Sin. 2022, 37, 746–757. [Google Scholar] [CrossRef]
- VanBlargan, L.A.; Himansu, S.; Foreman, B.M.; Ebel, G.D.; Pierson, T.C.; Diamond, M.S. An mRNA Vaccine Protects Mice against Multiple Tick-Transmitted Flavivirus Infections. Cell Rep. 2018, 25, 3382–3392.E3. [Google Scholar] [CrossRef]
- Richner, J.M.; Himansu, S.; Dowd, K.A.; Butler, S.L.; Salazar, V.; Fox, J.M.; Julander, J.G.; Tang, W.W.; Shresta, S.; Pierson, T.C.; et al. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell 2017, 168, 1114–1125.E10. [Google Scholar] [CrossRef]
- Richner, J.M.; Jagger, B.W.; Shan, C.; Fontes, C.R.; Dowd, K.A.; Cao, B.; Himansu, S.; Caine, E.A.; Nunes, B.T.D.; Medeiros, D.B.A.; et al. Vaccine Mediated Protection Against Zika Virus-Induced Congenital Disease. Cell 2017, 170, 273–283.E12. [Google Scholar] [CrossRef] [PubMed]
- Bollman, B.; Nunna, N.; Bahl, K.; Hsiao, C.J.; Bennett, H.; Butler, S.; Foreman, B.; Burgomaster, K.E.; Aleshnick, M.; Kong, W.P.; et al. An optimized messenger RNA vaccine candidate protects non-human primates from Zika virus infection. npj Vaccines 2023, 8, 58. [Google Scholar] [CrossRef]
- Essink, B.; Chu, L.; Seger, W.; Barranco, E.; Le Cam, N.; Bennett, H.; Faughnan, V.; Pajon, R.; Paila, Y.D.; Bollman, B.; et al. The safety and immunogenicity of two Zika virus mRNA vaccine candidates in healthy flavivirus baseline seropositive and seronegative adults: The results of two randomised, placebo-controlled, dose-ranging, phase 1 clinical trials. Lancet Infect. Dis. 2023, 23, 621–633. [Google Scholar] [CrossRef]
- Whitley, J.; Zwolinski, C.; Denis, C.; Maughan, M.; Hayles, L.; Clarke, D.; Snare, M.; Liao, H.; Chiou, S.; Marmura, T.; et al. Development of mRNA manufacturing for vaccines and therapeutics: mRNA platform requirements and development of a scalable production process to support early phase clinical trials. Transl. Res. 2022, 242, 38–55. [Google Scholar] [CrossRef]
- Uddin, M.N.; Roni, M.A. Challenges of Storage and Stability of mRNA-Based COVID-19 Vaccines. Vaccines 2021, 9, 1033. [Google Scholar] [CrossRef]
- Korzun, T.; Moses, A.S.; Diba, P.; Sattler, A.L.; Taratula, O.R.; Sahay, G.; Taratula, O.; Marks, D.L. From Bench to Bedside: Implications of Lipid Nanoparticle Carrier Reactogenicity for Advancing Nucleic Acid Therapeutics. Pharmaceuticals 2023, 16, 1088. [Google Scholar] [CrossRef]
- Korzun, T.; Moses, A.S.; Jozic, A.; Grigoriev, V.; Newton, S.; Kim, J.; Diba, P.; Sattler, A.; Levasseur, P.R.; Le, N.; et al. Lipid Nanoparticles Elicit Reactogenicity and Sickness Behavior in Mice Via Toll-Like Receptor 4 and Myeloid Differentiation Protein 88 Axis. ACS Nano 2024, 18, 24842–24859. [Google Scholar] [CrossRef]
- Muthumani, K.; Griffin, B.D.; Agarwal, S.; Kudchodkar, S.B.; Reuschel, E.L.; Choi, H.; Kraynyak, K.A.; Duperret, E.K.; Keaton, A.A.; Chung, C.; et al. In vivo protection against ZIKV infection and pathogenesis through passive antibody transfer and active immunisation with a prMEnv DNA vaccine. npj Vaccines 2016, 1, 16021. [Google Scholar] [CrossRef]
- Tebas, P.; Roberts, C.C.; Muthumani, K.; Reuschel, E.L.; Kudchodkar, S.B.; Zaidi, F.I.; White, S.; Khan, A.S.; Racine, T.; Choi, H.; et al. Safety and Immunogenicity of an Anti-Zika Virus DNA Vaccine. N. Engl. J. Med. 2021, 385, e35. [Google Scholar] [CrossRef]
- Davis, B.S.; Chang, G.J.; Cropp, B.; Roehrig, J.T.; Martin, D.A.; Mitchell, C.J.; Bowen, R.; Bunning, M.L. West Nile virus recombinant DNA vaccine protects mouse and horse from virus challenge and expresses in vitro a noninfectious recombinant antigen that can be used in enzyme-linked immunosorbent assays. J. Virol. 2001, 75, 4040–4047. [Google Scholar] [CrossRef]
- Martin, J.E.; Pierson, T.C.; Hubka, S.; Rucker, S.; Gordon, I.J.; Enama, M.E.; Andrews, C.A.; Xu, Q.; Davis, B.S.; Nason, M.; et al. A West Nile virus DNA vaccine induces neutralizing antibody in healthy adults during a phase 1 clinical trial. J. Infect. Dis. 2007, 196, 1732–1740. [Google Scholar] [CrossRef]
- Ledgerwood, J.E.; Pierson, T.C.; Hubka, S.A.; Desai, N.; Rucker, S.; Gordon, I.J.; Enama, M.E.; Nelson, S.; Nason, M.; Gu, W.; et al. A West Nile virus DNA vaccine utilizing a modified promoter induces neutralizing antibody in younger and older healthy adults in a phase I clinical trial. J. Infect. Dis. 2011, 203, 1396–1404. [Google Scholar] [CrossRef]
- McCracken, M.K.; Kuklis, C.H.; Kannadka, C.B.; Barvir, D.A.; Sanborn, M.A.; Waickman, A.T.; Siegfried, H.C.; Victor, K.A.; Hatch, K.L.; De La Barrera, R.; et al. Enhanced dengue vaccine virus replication and neutralizing antibody responses in immune primed rhesus macaques. npj Vaccines 2021, 6, 77. [Google Scholar] [CrossRef] [PubMed]
- Zellweger, R.M.; Eddy, W.E.; Tang, W.W.; Miller, R.; Shresta, S. CD8+ T cells prevent antigen-induced antibody-dependent enhancement of dengue disease in mice. J. Immunol. 2014, 193, 4117–4124. [Google Scholar] [CrossRef] [PubMed]
- Ballesteros-Briones, M.C.; Silva-Pilipich, N.; Herrador-Canete, G.; Vanrell, L.; Smerdou, C. A new generation of vaccines based on alphavirus self-amplifying RNA. Curr. Opin. Virol. 2020, 44, 145–153. [Google Scholar] [CrossRef] [PubMed]
- Lu, H.H.; Dos Santos Alves, R.P.; Li, Q.H.; Eder, L.; Timis, J.; Madany, H.; Chuensirikulchai, K.; Varghese, K.V.; Singh, A.; Le Tran, L.; et al. Enhanced durability of a Zika virus self-amplifying RNA vaccine through combinatorial OX40 and 4-1BB agonism. JCI Insight 2025, 10, e187405. [Google Scholar] [CrossRef] [PubMed]
- Luisi, K.; Morabito, K.M.; Burgomaster, K.E.; Sharma, M.; Kong, W.P.; Foreman, B.M.; Patel, S.; Fisher, B.; Aleshnick, M.A.; Laliberte, J.; et al. Development of a potent Zika virus vaccine using self-amplifying messenger RNA. Sci. Adv. 2020, 6, eaba5068. [Google Scholar] [CrossRef]
- Oda, Y.; Kumagai, Y.; Kanai, M.; Iwama, Y.; Okura, I.; Minamida, T.; Yagi, Y.; Kurosawa, T.; Greener, B.; Zhang, Y.; et al. Immunogenicity and safety of a booster dose of a self-amplifying RNA COVID-19 vaccine (ARCT-154) versus BNT162b2 mRNA COVID-19 vaccine: A double-blind, multicentre, randomised, controlled, phase 3, non-inferiority trial. Lancet Infect. Dis. 2024, 24, 351–360. [Google Scholar] [CrossRef]
- Ho, N.T.; Hughes, S.G.; Ta, V.T.; Phan, L.T.; Do, Q.; Nguyen, T.V.; Pham, A.T.V.; Thi Ngoc Dang, M.; Nguyen, L.V.; Trinh, Q.V.; et al. Safety, immunogenicity and efficacy of the self-amplifying mRNA ARCT-154 COVID-19 vaccine: Pooled phase 1, 2, 3a and 3b randomized, controlled trials. Nat. Commun. 2024, 15, 4081. [Google Scholar] [CrossRef]
- Oda, Y.; Kumagai, Y.; Kanai, M.; Iwama, Y.; Okura, I.; Minamida, T.; Yagi, Y.; Kurosawa, T.; Chivukula, P.; Zhang, Y.; et al. Persistence of immune responses of a self-amplifying RNA COVID-19 vaccine (ARCT-154) versus BNT162b2. Lancet Infect. Dis. 2024, 24, 341–343. [Google Scholar] [CrossRef]
- Battisti, P.; Ykema, M.R.; Kasal, D.N.; Jennewein, M.F.; Beaver, S.; Weight, A.E.; Hanson, D.; Singh, J.; Bakken, J.; Cross, N.; et al. A bivalent self-amplifying RNA vaccine against yellow fever and Zika viruses. Front. Immunol. 2025, 16, 1569454. [Google Scholar] [CrossRef]
- Zhong, Z.; Portela Catani, J.P.; Mc Cafferty, S.; Couck, L.; Van Den Broeck, W.; Gorle, N.; Vandenbroucke, R.E.; Devriendt, B.; Ulbert, S.; Cnops, L.; et al. Immunogenicity and Protection Efficacy of a Naked Self-Replicating mRNA-Based Zika Virus Vaccine. Vaccines 2019, 7, 96. [Google Scholar] [CrossRef] [PubMed]
- Chahal, J.S.; Fang, T.; Woodham, A.W.; Khan, O.F.; Ling, J.; Anderson, D.G.; Ploegh, H.L. An RNA nanoparticle vaccine against Zika virus elicits antibody and CD8+ T cell responses in a mouse model. Sci. Rep. 2017, 7, 252. [Google Scholar] [CrossRef]
- Brito, L.A.; Chan, M.; Shaw, C.A.; Hekele, A.; Carsillo, T.; Schaefer, M.; Archer, J.; Seubert, A.; Otten, G.R.; Beard, C.W.; et al. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol. Ther. 2014, 22, 2118–2129. [Google Scholar] [CrossRef] [PubMed]
- Blakney, A.K.; McKay, P.F.; Hu, K.; Samnuan, K.; Jain, N.; Brown, A.; Thomas, A.; Rogers, P.; Polra, K.; Sallah, H.; et al. Polymeric and lipid nanoparticles for delivery of self-amplifying RNA vaccines. J. Control Release 2021, 338, 201–210. [Google Scholar] [CrossRef]
- Geall, A.J.; Verma, A.; Otten, G.R.; Shaw, C.A.; Hekele, A.; Banerjee, K.; Cu, Y.; Beard, C.W.; Brito, L.A.; Krucker, T.; et al. Nonviral delivery of self-amplifying RNA vaccines. Proc. Natl. Acad. Sci. USA 2012, 109, 14604–14609. [Google Scholar] [CrossRef]
- Gerhardt, A.; Voigt, E.; Archer, M.; Reed, S.; Larson, E.; Van Hoeven, N.; Kramer, R.; Fox, C.; Casper, C. A flexible, thermostable nanostructured lipid carrier platform for RNA vaccine delivery. Mol. Ther. Methods Clin. Dev. 2022, 25, 205–214. [Google Scholar] [CrossRef]
- Voigt, E.A.; Gerhardt, A.; Hanson, D.; Jennewein, M.F.; Battisti, P.; Reed, S.; Singh, J.; Mohamath, R.; Bakken, J.; Beaver, S.; et al. A self-amplifying RNA vaccine against COVID-19 with long-term room-temperature stability. npj Vaccines 2022, 7, 136. [Google Scholar] [CrossRef]
- Pepini, T.; Pulichino, A.M.; Carsillo, T.; Carlson, A.L.; Sari-Sarraf, F.; Ramsauer, K.; Debasitis, J.C.; Maruggi, G.; Otten, G.R.; Geall, A.J.; et al. Induction of an IFN-Mediated Antiviral Response by a Self-Amplifying RNA Vaccine: Implications for Vaccine Design. J. Immunol. 2017, 198, 4012–4024. [Google Scholar] [CrossRef]
- Minnaert, A.K.; Vanluchene, H.; Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Raemdonck, K.; Sanders, N.N.; Remaut, K. Strategies for controlling the innate immune activity of conventional and self-amplifying mRNA therapeutics: Getting the message across. Adv. Drug Deliv. Rev. 2021, 176, 113900. [Google Scholar] [CrossRef] [PubMed]
- Frederickson, R.; Herzog, R.W. RNA-based vaccines and innate immune activation: Not too hot and not too cold. Mol. Ther. 2021, 29, 1365–1366. [Google Scholar] [CrossRef] [PubMed]
- Tregoning, J.S.; Stirling, D.C.; Wang, Z.; Flight, K.E.; Brown, J.C.; Blakney, A.K.; McKay, P.F.; Cunliffe, R.F.; Murugaiah, V.; Fox, C.B.; et al. Formulation, inflammation, and RNA sensing impact the immunogenicity of self-amplifying RNA vaccines. Mol. Ther. Nucleic Acids 2023, 31, 29–42. [Google Scholar] [CrossRef] [PubMed]
- Ong, E.Z.; Yee, J.X.; Ooi, J.S.G.; Syenina, A.; de Alwis, R.; Chen, S.; Sim, J.X.Y.; Kalimuddin, S.; Leong, Y.S.; Chan, Y.F.Z.; et al. Immune gene expression analysis indicates the potential of a self-amplifying COVID-19 mRNA vaccine. npj Vaccines 2022, 7, 154. [Google Scholar] [CrossRef]
- Pollock, K.M.; Cheeseman, H.M.; Szubert, A.J.; Libri, V.; Boffito, M.; Owen, D.; Bern, H.; O’Hara, J.; McFarlane, L.R.; Lemm, N.M.; et al. Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial. eClinicalMedicine 2022, 44, 101262. [Google Scholar] [CrossRef]
- McGee, J.E.; Kirsch, J.R.; Kenney, D.; Cerbo, F.; Chavez, E.C.; Shih, T.Y.; Douam, F.; Wong, W.W.; Grinstaff, M.W. Complete substitution with modified nucleotides in self-amplifying RNA suppresses the interferon response and increases potency. Nat. Biotechnol. 2025, 43, 720–726. [Google Scholar] [CrossRef]
- Gong, Y.; Yong, D.; Liu, G.; Xu, J.; Ding, J.; Jia, W. A Novel Self-Amplifying mRNA with Decreased Cytotoxicity and Enhanced Protein Expression by Macrodomain Mutations. Adv. Sci. 2024, 11, e2402936. [Google Scholar] [CrossRef] [PubMed]
- Kim, B.J.; Hosn, R.R.; Remba, T.K.; Dye, J.; Mak, H.H.; Jeong, J.Y.; Cornwall-Brady, M.; Abraham, W.; Maiorino, L.; Melo, M.B.; et al. Targeted suppression of type 1 interferon signaling during RNA delivery enhances vaccine-elicited immunity. bioRxiv 2025, bioRxiv:2025.03.09.642244. [Google Scholar] [CrossRef]
- Asghar, N.; Melik, W.; Paulsen, K.M.; Pedersen, B.N.; Bo-Granquist, E.G.; Vikse, R.; Stuen, S.; Andersson, S.; Strid, A.; Andreassen, A.K.; et al. Transient Expression of Flavivirus Structural Proteins in Nicotiana benthamiana. Vaccines 2022, 10, 1667. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Liang, M.; Gu, W.; Li, C.; Miao, F.; Wang, X.; Jin, C.; Zhang, L.; Zhang, F.; Zhang, Q.; et al. Vaccination with dengue virus-like particles induces humoral and cellular immune responses in mice. Virol. J. 2011, 8, 333. [Google Scholar] [CrossRef]
- Dai, S.; Zhang, T.; Zhang, Y.; Wang, H.; Deng, F. Zika Virus Baculovirus-Expressed Virus-Like Particles Induce Neutralizing Antibodies in Mice. Virol. Sin. 2018, 33, 213–226. [Google Scholar] [CrossRef]
- Hua, R.H.; Li, Y.N.; Chen, Z.S.; Liu, L.K.; Huo, H.; Wang, X.L.; Guo, L.P.; Shen, N.; Wang, J.F.; Bu, Z.G. Generation and characterization of a new mammalian cell line continuously expressing virus-like particles of Japanese encephalitis virus for a subunit vaccine candidate. BMC Biotechnol. 2014, 14, 62. [Google Scholar] [CrossRef]
- Sugrue, R.J.; Fu, J.; Howe, J.; Chan, Y.C. Expression of the dengue virus structural proteins in Pichia pastoris leads to the generation of virus-like particles. J. Gen. Virol. 1997, 78 Pt 8, 1861–1866. [Google Scholar] [CrossRef]
- Krol, E.; Brzuska, G.; Szewczyk, B. Production and Biomedical Application of Flavivirus-like Particles. Trends Biotechnol. 2019, 37, 1202–1216. [Google Scholar] [CrossRef]
- Metz, S.W.; Thomas, A.; White, L.; Stoops, M.; Corten, M.; Hannemann, H.; de Silva, A.M. Dengue virus-like particles mimic the antigenic properties of the infectious dengue virus envelope. Virol. J. 2018, 15, 60. [Google Scholar] [CrossRef]
- Abbo, S.R.; Yan, K.; Geertsema, C.; Hick, T.A.H.; Altenburg, J.J.; Nowee, G.; van Toor, C.; van Lent, J.W.; Nakayama, E.; Tang, B.; et al. Virus-like particle vaccine with authentic quaternary epitopes protects against Zika virus-induced viremia and testicular damage. J. Virol. 2025, 99, e0232224. [Google Scholar] [CrossRef] [PubMed]
- Thoresen, D.; Matsuda, K.; Urakami, A.; Ngwe Tun, M.M.; Nomura, T.; Moi, M.L.; Watanabe, Y.; Ishikawa, M.; Hau, T.T.T.; Yamamoto, H.; et al. A tetravalent dengue virus-like particle vaccine induces high levels of neutralizing antibodies and reduces dengue replication in non-human primates. J. Virol. 2024, 98, e0023924. [Google Scholar] [CrossRef]
- Garg, H.; Sedano, M.; Plata, G.; Punke, E.B.; Joshi, A. Development of Virus-Like-Particle Vaccine and Reporter Assay for Zika Virus. J. Virol. 2017, 91, e00834-17. [Google Scholar] [CrossRef]
- Wang, P.G.; Kudelko, M.; Lo, J.; Siu, L.Y.; Kwok, K.T.; Sachse, M.; Nicholls, J.M.; Bruzzone, R.; Altmeyer, R.M.; Nal, B. Efficient assembly and secretion of recombinant subviral particles of the four dengue serotypes using native prM and E proteins. PLoS ONE 2009, 4, e8325. [Google Scholar] [CrossRef]
- Tsai, W.Y.; Chen, H.L.; Tsai, J.J.; Dejnirattisai, W.; Jumnainsong, A.; Mongkolsapaya, J.; Screaton, G.; Crowe, J.E., Jr.; Wang, W.K. Potent Neutralizing Human Monoclonal Antibodies Preferentially Target Mature Dengue Virus Particles: Implication for Novel Strategy for Dengue Vaccine. J. Virol. 2018, 92, e00556-18. [Google Scholar] [CrossRef]
- Vang, L.; Morello, C.S.; Mendy, J.; Thompson, D.; Manayani, D.; Guenther, B.; Julander, J.; Sanford, D.; Jain, A.; Patel, A.; et al. Zika virus-like particle vaccine protects AG129 mice and rhesus macaques against Zika virus. PLoS Negl. Trop. Dis. 2021, 15, e0009195. [Google Scholar] [CrossRef]
- Gupta, R.; Arora, K.; Roy, S.S.; Joseph, A.; Rastogi, R.; Arora, N.M.; Kundu, P.K. Platforms, advances, and technical challenges in virus-like particles-based vaccines. Front. Immunol. 2023, 14, 1123805. [Google Scholar] [CrossRef]
- Vicente, T.; Roldao, A.; Peixoto, C.; Carrondo, M.J.; Alves, P.M. Large-scale production and purification of VLP-based vaccines. J. Invertebr. Pathol. 2011, 107, S42–S48. [Google Scholar] [CrossRef] [PubMed]
- Srivastava, V.; Nand, K.N.; Ahmad, A.; Kumar, R. Yeast-Based Virus-like Particles as an Emerging Platform for Vaccine Development and Delivery. Vaccines 2023, 11, 479. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.C.; Chen, J.M.; Chen, Y.Y.; Hsu, W.L.; Chang, G.J.; Chiou, S.S. Low-temperature culture enhances production of flavivirus virus-like particles in mammalian cells. Appl. Microbiol. Biotechnol. 2024, 108, 242. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Lai, H. Plant-derived virus-like particles as vaccines. Hum. Vaccines Immunother. 2013, 9, 26–49. [Google Scholar] [CrossRef]
- Cimica, V.; Galarza, J.M. Adjuvant formulations for virus-like particle (VLP) based vaccines. Clin. Immunol. 2017, 183, 99–108. [Google Scholar] [CrossRef]
- Salvo, M.A.; Kingstad-Bakke, B.; Salas-Quinchucua, C.; Camacho, E.; Osorio, J.E. Zika virus like particles elicit protective antibodies in mice. PLoS Neglected Trop. Dis. 2018, 12, e0006210. [Google Scholar] [CrossRef]
- Okamoto, S.; Yoshii, H.; Matsuura, M.; Kojima, A.; Ishikawa, T.; Akagi, T.; Akashi, M.; Takahashi, M.; Yamanishi, K.; Mori, Y. Poly-gamma-glutamic acid nanoparticles and aluminum adjuvant used as an adjuvant with a single dose of Japanese encephalitis virus-like particles provide effective protection from Japanese encephalitis virus. Clin. Vaccine Immunol. 2012, 19, 17–22. [Google Scholar] [CrossRef] [PubMed]
- Auguste, A.J.; Langsjoen, R.M.; Porier, D.L.; Erasmus, J.H.; Bergren, N.A.; Bolling, B.G.; Luo, H.; Singh, A.; Guzman, H.; Popov, V.L.; et al. Isolation of a novel insect-specific flavivirus with immunomodulatory effects in vertebrate systems. Virology 2021, 562, 50–62. [Google Scholar] [CrossRef]
- Elrefaey, A.M.; Abdelnabi, R.; Rosales Rosas, A.L.; Wang, L.; Basu, S.; Delang, L. Understanding the Mechanisms Underlying Host Restriction of Insect-Specific Viruses. Viruses 2020, 12, 964. [Google Scholar] [CrossRef] [PubMed]
- Tangudu, C.S.; Charles, J.; Nunez-Avellaneda, D.; Hargett, A.M.; Brault, A.C.; Blitvich, B.J. Chimeric Zika viruses containing structural protein genes of insect-specific flaviviruses cannot replicate in vertebrate cells due to entry and post-translational restrictions. Virology 2021, 559, 30–39. [Google Scholar] [CrossRef]
- Harrison, J.J.; Hobson-Peters, J.; Bielefeldt-Ohmann, H.; Hall, R.A. Chimeric Vaccines Based on Novel Insect-Specific Flaviviruses. Vaccines 2021, 9, 1230. [Google Scholar] [CrossRef]
- Dawurung, J.S.; Harrison, J.J.; Modhiran, N.; Hall, R.A.; Hobson-Peters, J.; de Malmanche, H. Serum-Free Suspension Culture of the Aedes albopictus C6/36 Cell Line for Chimeric Orthoflavivirus Vaccine Production. Viruses 2025, 17, 250. [Google Scholar] [CrossRef]
- Hall, R.A.; Nguyen, W.; Khromykh, A.A.; Suhrbier, A. Insect-specific virus platforms for arbovirus vaccine development. Front. Immunol. 2025, 16, 1521104. [Google Scholar] [CrossRef]
- Porier, D.L.; Wilson, S.N.; Auguste, D.I.; Leber, A.; Coutermarsh-Ott, S.; Allen, I.C.; Caswell, C.C.; Budnick, J.A.; Bassaganya-Riera, J.; Hontecillas, R.; et al. Enemy of My Enemy: A Novel Insect-Specific Flavivirus Offers a Promising Platform for a Zika Virus Vaccine. Vaccines 2021, 9, 1142. [Google Scholar] [CrossRef] [PubMed]
- Porier, D.L.; Adam, A.; Kang, L.; Michalak, P.; Tupik, J.; Santos, M.A.; Tanelus, M.; Lopez, K.; Auguste, D.I.; Lee, C.; et al. Humoral and T-cell-mediated responses to an insect-specific flavivirus-based Zika virus vaccine candidate. PLoS Pathog. 2024, 20, e1012566. [Google Scholar] [CrossRef] [PubMed]
- Vet, L.J.; Setoh, Y.X.; Amarilla, A.A.; Habarugira, G.; Suen, W.W.; Newton, N.D.; Harrison, J.J.; Hobson-Peters, J.; Hall, R.A.; Bielefeldt-Ohmann, H. Protective Efficacy of a Chimeric Insect-Specific Flavivirus Vaccine against West Nile Virus. Vaccines 2020, 8, 258. [Google Scholar] [CrossRef]
- Hazlewood, J.E.; Tang, B.; Yan, K.; Rawle, D.J.; Harrison, J.J.; Hall, R.A.; Hobson-Peters, J.; Suhrbier, A. The Chimeric Binjari-Zika Vaccine Provides Long-Term Protection against ZIKA Virus Challenge. Vaccines 2022, 10, 85. [Google Scholar] [CrossRef]
- Scott, R.M.; Shelton, A.L.; Eckels, K.H.; Bancroft, W.H.; Summers, R.J.; Russell, P.K. Human hypersensitivity to a sham vaccine prepared from mosquito-cell culture fluids. J. Allergy Clin. Immunol. 1984, 74, 808–811. [Google Scholar] [CrossRef]
- Carvalho, V.L.; Long, M.T. Perspectives on New Vaccines against Arboviruses Using Insect-Specific Viruses as Platforms. Vaccines 2021, 9, 263. [Google Scholar] [CrossRef]
- Zhang, X.; Sheng, J.; Plevka, P.; Kuhn, R.J.; Diamond, M.S.; Rossmann, M.G. Dengue structure differs at the temperatures of its human and mosquito hosts. Proc. Natl. Acad. Sci. USA 2013, 110, 6795–6799. [Google Scholar] [CrossRef]
- Fibriansah, G.; Ng, T.S.; Kostyuchenko, V.A.; Lee, J.; Lee, S.; Wang, J.; Lok, S.M. Structural changes in dengue virus when exposed to a temperature of 37 °C. J. Virol. 2013, 87, 7585–7592. [Google Scholar] [CrossRef] [PubMed]
- Gwon, Y.D.; Zusinaite, E.; Merits, A.; Overby, A.K.; Evander, M. N-glycosylation in the Pre-Membrane Protein Is Essential for the Zika Virus Life Cycle. Viruses 2020, 12, 925. [Google Scholar] [CrossRef] [PubMed]
- Hobson-Peters, J.; Harrison, J.J.; Watterson, D.; Hazlewood, J.E.; Vet, L.J.; Newton, N.D.; Warrilow, D.; Colmant, A.M.G.; Taylor, C.; Huang, B.; et al. A recombinant platform for flavivirus vaccines and diagnostics using chimeras of a new insect-specific virus. Sci. Transl. Med. 2019, 11, eaax7888. [Google Scholar] [CrossRef]
- Ewer, K.J.; Barrett, J.R.; Belij-Rammerstorfer, S.; Sharpe, H.; Makinson, R.; Morter, R.; Flaxman, A.; Wright, D.; Bellamy, D.; Bittaye, M.; et al. T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat. Med. 2021, 27, 270–278. [Google Scholar] [CrossRef]
- Tan, W.G.; Jin, H.T.; West, E.E.; Penaloza-MacMaster, P.; Wieland, A.; Zilliox, M.J.; McElrath, M.J.; Barouch, D.H.; Ahmed, R. Comparative analysis of simian immunodeficiency virus gag-specific effector and memory CD8+ T cells induced by different adenovirus vectors. J. Virol. 2013, 87, 1359–1372. [Google Scholar] [CrossRef]
- Schepp-Berglind, J.; Luo, M.; Wang, D.; Wicker, J.A.; Raja, N.U.; Hoel, B.D.; Holman, D.H.; Barrett, A.D.; Dong, J.Y. Complex adenovirus-mediated expression of West Nile virus C, PreM, E, and NS1 proteins induces both humoral and cellular immune responses. Clin. Vaccine Immunol. 2007, 14, 1117–1126. [Google Scholar] [CrossRef]
- Abbink, P.; Larocca, R.A.; De La Barrera, R.A.; Bricault, C.A.; Moseley, E.T.; Boyd, M.; Kirilova, M.; Li, Z.; Ng’ang’a, D.; Nanayakkara, O.; et al. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science 2016, 353, 1129–1132. [Google Scholar] [CrossRef]
- Larocca, R.A.; Mendes, E.A.; Abbink, P.; Peterson, R.L.; Martinot, A.J.; Iampietro, M.J.; Kang, Z.H.; Aid, M.; Kirilova, M.; Jacob-Dolan, C.; et al. Adenovirus Vector-Based Vaccines Confer Maternal-Fetal Protection against Zika Virus Challenge in Pregnant IFN-αβR−/−. Mice. Cell Host Microbe 2019, 26, 591–600.E4. [Google Scholar] [CrossRef]
- Bullard, B.L.; Corder, B.N.; Gorman, M.J.; Diamond, M.S.; Weaver, E.A. Efficacy of a T Cell-Biased Adenovirus Vector as a Zika Virus Vaccine. Sci. Rep. 2018, 8, 18017. [Google Scholar] [CrossRef]
- Lopez-Camacho, C.; Abbink, P.; Larocca, R.A.; Dejnirattisai, W.; Boyd, M.; Badamchi-Zadeh, A.; Wallace, Z.R.; Doig, J.; Velazquez, R.S.; Neto, R.D.L.; et al. Rational Zika vaccine design via the modulation of antigen membrane anchors in chimpanzee adenoviral vectors. Nat. Commun. 2018, 9, 2441. [Google Scholar] [CrossRef] [PubMed]
- Salisch, N.C.; Stephenson, K.E.; Williams, K.; Cox, F.; van der Fits, L.; Heerwegh, D.; Truyers, C.; Habets, M.N.; Kanjilal, D.G.; Larocca, R.A.; et al. A Double-Blind, Randomized, Placebo-Controlled Phase 1 Study of Ad26.ZIKV.001, an Ad26-Vectored Anti-Zika Virus Vaccine. Ann. Intern. Med. 2021, 174, 585–594. [Google Scholar] [CrossRef]
- Sumida, S.M.; Truitt, D.M.; Kishko, M.G.; Arthur, J.C.; Jackson, S.S.; Gorgone, D.A.; Lifton, M.A.; Koudstaal, W.; Pau, M.G.; Kostense, S.; et al. Neutralizing antibodies and CD8+ T lymphocytes both contribute to immunity to adenovirus serotype 5 vaccine vectors. J. Virol. 2004, 78, 2666–2673. [Google Scholar] [CrossRef] [PubMed]
- Barouch, D.H.; Pau, M.G.; Custers, J.H.; Koudstaal, W.; Kostense, S.; Havenga, M.J.; Truitt, D.M.; Sumida, S.M.; Kishko, M.G.; Arthur, J.C.; et al. Immunogenicity of recombinant adenovirus serotype 35 vaccine in the presence of pre-existing anti-Ad5 immunity. J. Immunol. 2004, 172, 6290–6297. [Google Scholar] [CrossRef]
- Frahm, N.; DeCamp, A.C.; Friedrich, D.P.; Carter, D.K.; Defawe, O.D.; Kublin, J.G.; Casimiro, D.R.; Duerr, A.; Robertson, M.N.; Buchbinder, S.P.; et al. Human adenovirus-specific T cells modulate HIV-specific T cell responses to an Ad5-vectored HIV-1 vaccine. J. Clin. Investig. 2012, 122, 359–367. [Google Scholar] [CrossRef]
- Cheng, C.; Gall, J.G.; Nason, M.; King, C.R.; Koup, R.A.; Roederer, M.; McElrath, M.J.; Morgan, C.A.; Churchyard, G.; Baden, L.R.; et al. Differential specificity and immunogenicity of adenovirus type 5 neutralizing antibodies elicited by natural infection or immunization. J. Virol. 2010, 84, 630–638. [Google Scholar] [CrossRef]
- Sobieszczyk, M.E.; Maaske, J.; Falsey, A.R.; Sproule, S.; Robb, M.L.; Frenck, R.W., Jr.; Tieu, H.V.; Mayer, K.H.; Corey, L.; Neuzil, K.M.; et al. Durability of protection and immunogenicity of AZD1222 (ChAdOx1 nCoV-19) COVID-19 vaccine over 6 months. J. Clin. Investig. 2022, 132, e160565. [Google Scholar] [CrossRef]
- Sadoff, J.; Gray, G.; Vandebosch, A.; Cardenas, V.; Shukarev, G.; Grinsztejn, B.; Goepfert, P.A.; Truyers, C.; Van Dromme, I.; Spiessens, B.; et al. Final Analysis of Efficacy and Safety of Single-Dose Ad26.COV2.S. N. Engl. J. Med. 2022, 386, 847–860. [Google Scholar] [CrossRef]
- Greinacher, A.; Thiele, T.; Warkentin, T.E.; Weisser, K.; Kyrle, P.A.; Eichinger, S. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N. Engl. J. Med. 2021, 384, 2092–2101. [Google Scholar] [CrossRef]
- Muir, K.L.; Kallam, A.; Koepsell, S.A.; Gundabolu, K. Thrombotic Thrombocytopenia after Ad26.COV2.S Vaccination. N. Engl. J. Med. 2021, 384, 1964–1965. [Google Scholar] [CrossRef]
- Iglesias, M.C.; Frenkiel, M.P.; Mollier, K.; Souque, P.; Despres, P.; Charneau, P. A single immunization with a minute dose of a lentiviral vector-based vaccine is highly effective at eliciting protective humoral immunity against West Nile virus. J. Gene Med. 2006, 8, 265–274. [Google Scholar] [CrossRef]
- de Wispelaere, M.; Ricklin, M.; Souque, P.; Frenkiel, M.P.; Paulous, S.; Garcia-Nicolas, O.; Summerfield, A.; Charneau, P.; Despres, P. A Lentiviral Vector Expressing Japanese Encephalitis Virus-like Particles Elicits Broad Neutralizing Antibody Response in Pigs. PLoS Neglected Trop. Dis. 2015, 9, e0004081. [Google Scholar] [CrossRef]
- Cousin, C.; Oberkampf, M.; Felix, T.; Rosenbaum, P.; Weil, R.; Fabrega, S.; Morante, V.; Negri, D.; Cara, A.; Dadaglio, G.; et al. Persistence of Integrase-Deficient Lentiviral Vectors Correlates with the Induction of STING-Independent CD8+ T Cell Responses. Cell Rep. 2019, 26, 1242–1257.e7. [Google Scholar] [CrossRef]
- Mahesh, S.; Li, J.; Travieso, T.; Psaradelli, D.; Negri, D.; Klotman, M.; Cara, A.; Blasi, M. Integrase Defective Lentiviral Vector Promoter Impacts Transgene Expression in Target Cells and Magnitude of Vector-Induced Immune Responses. Viruses 2023, 15, 2255. [Google Scholar] [CrossRef]
- Gallinaro, A.; Borghi, M.; Bona, R.; Grasso, F.; Calzoletti, L.; Palladino, L.; Cecchetti, S.; Vescio, M.F.; Macchia, D.; Morante, V.; et al. Integrase Defective Lentiviral Vector as a Vaccine Platform for Delivering Influenza Antigens. Front. Immunol. 2018, 9, 171. [Google Scholar] [CrossRef]
- Gallinaro, A.; Pirillo, M.F.; Aldon, Y.; Cecchetti, S.; Michelini, Z.; Tinari, A.; Borghi, M.; Canitano, A.; McKay, P.F.; Bona, R.; et al. Persistent immunogenicity of integrase defective lentiviral vectors delivering membrane-tethered native-like HIV-1 envelope trimers. npj Vaccines 2022, 7, 44. [Google Scholar] [CrossRef]
- Gallinaro, A.; Borghi, M.; Pirillo, M.F.; Cecchetti, S.; Bona, R.; Canitano, A.; Michelini, Z.; Di Virgilio, A.; Olvera, A.; Brander, C.; et al. Development and Preclinical Evaluation of an Integrase Defective Lentiviral Vector Vaccine Expressing the HIVACAT T Cell Immunogen in Mice. Mol. Ther. Methods Clin. Dev. 2020, 17, 418–428. [Google Scholar] [CrossRef]
- Blasi, M.; Negri, D.; LaBranche, C.; Alam, S.M.; Baker, E.J.; Brunner, E.C.; Gladden, M.A.; Michelini, Z.; Vandergrift, N.A.; Wiehe, K.J.; et al. IDLV-HIV-1 Env vaccination in non-human primates induces affinity maturation of antigen-specific memory B cells. Commun. Biol. 2018, 1, 134. [Google Scholar] [CrossRef]
- Ku, M.W.; Anna, F.; Souque, P.; Petres, S.; Prot, M.; Simon-Loriere, E.; Charneau, P.; Bourgine, M. A Single Dose of NILV-Based Vaccine Provides Rapid and Durable Protection against Zika Virus. Mol. Ther. 2020, 28, 1772–1782. [Google Scholar] [CrossRef]
- Coutant, F.; Frenkiel, M.P.; Despres, P.; Charneau, P. Protective antiviral immunity conferred by a nonintegrative lentiviral vector-based vaccine. PLoS ONE 2008, 3, e3973. [Google Scholar] [CrossRef]
- Lin, Y.Y.; Belle, I.; Blasi, M.; Huang, M.N.; Buckley, A.F.; Rountree, W.; Klotman, M.E.; Cara, A.; Negri, D. Skeletal Muscle Is an Antigen Reservoir in Integrase-Defective Lentiviral Vector-Induced Long-Term Immunity. Mol. Ther. Methods Clin. Dev. 2020, 17, 532–544. [Google Scholar] [CrossRef]
- Havlikova, S.; Roller, L.; Koci, J.; Trimnell, A.R.; Kazimirova, M.; Klempa, B.; Nuttall, P.A. Functional role of 64P, the candidate transmission-blocking vaccine antigen from the tick, Rhipicephalus appendiculatus. Int. J. Parasitol. 2009, 39, 1485–1494. [Google Scholar] [CrossRef]
- Labuda, M.; Trimnell, A.R.; Lickova, M.; Kazimirova, M.; Davies, G.M.; Lissina, O.; Hails, R.S.; Nuttall, P.A. An antivector vaccine protects against a lethal vector-borne pathogen. PLoS Pathog. 2006, 2, e27. [Google Scholar] [CrossRef]
- Hastings, A.K.; Uraki, R.; Gaitsch, H.; Dhaliwal, K.; Stanley, S.; Sproch, H.; Williamson, E.; MacNeil, T.; Marin-Lopez, A.; Hwang, J.; et al. Aedes aegypti NeSt1 Protein Enhances Zika Virus Pathogenesis by Activating Neutrophils. J. Virol. 2019, 93, e00395-19. [Google Scholar] [CrossRef]
- Uraki, R.; Hastings, A.K.; Marin-Lopez, A.; Sumida, T.; Takahashi, T.; Grover, J.R.; Iwasaki, A.; Hafler, D.A.; Montgomery, R.R.; Fikrig, E. Aedes aegypti AgBR1 antibodies modulate early Zika virus infection of mice. Nat. Microbiol. 2019, 4, 948–955. [Google Scholar] [CrossRef]
- Marin-Lopez, A.; Wang, Y.; Jiang, J.; Ledizet, M.; Fikrig, E. AgBR1 and NeSt1 antisera protect mice from Aedes aegypti-borne Zika infection. Vaccine 2021, 39, 1675–1679. [Google Scholar] [CrossRef]
- Jin, L.; Guo, X.; Shen, C.; Hao, X.; Sun, P.; Li, P.; Xu, T.; Hu, C.; Rose, O.; Zhou, H.; et al. Salivary factor LTRIN from Aedes aegypti facilitates the transmission of Zika virus by interfering with the lymphotoxin-beta receptor. Nat. Immunol. 2018, 19, 342–353. [Google Scholar] [CrossRef]
- Machain-Williams, C.; Reagan, K.; Wang, T.; Zeidner, N.S.; Blair, C.D. Immunization with Culex tarsalis mosquito salivary gland extract modulates West Nile virus infection and disease in mice. Viral Immunol. 2013, 26, 84–92. [Google Scholar] [CrossRef]
- Manning, J.E.; Oliveira, F.; Coutinho-Abreu, I.V.; Herbert, S.; Meneses, C.; Kamhawi, S.; Baus, H.A.; Han, A.; Czajkowski, L.; Rosas, L.A.; et al. Safety and immunogenicity of a mosquito saliva peptide-based vaccine: A randomised, placebo-controlled, double-blind, phase 1 trial. Lancet 2020, 395, 1998–2007. [Google Scholar] [CrossRef]
- Londono-Renteria, B.; Troupin, A.; Colpitts, T.M. Arbovirosis and potential transmission blocking vaccines. Parasites Vectors 2016, 9, 516. [Google Scholar] [CrossRef]
- Neelakanta, G.; Sultana, H. Transmission-Blocking Vaccines: Focus on Anti-Vector Vaccines against Tick-Borne Diseases. Arch. Immunol. Ther. Exp. 2015, 63, 169–179. [Google Scholar] [CrossRef]
- Muthuraman, K.R.; Boonyakida, J.; Matsuda, M.; Suzuki, R.; Kato, T.; Park, E.Y. Tetravalent Virus-like Particles Engineered To Display Envelope Domain IIIs of Four Dengue Serotypes in Silkworm as Vaccine Candidates. Biomacromolecules 2025, 26, 2003–2013. [Google Scholar] [CrossRef]
- Boonyakida, J.; Matsuda, M.; Suzuki, R.; Muthuraman, K.R.; Park, E.Y. Virus-Like Particle-Based Multiserotype Quartet Vaccine of Dengue Envelope Protein Domain III Elicited Potent Anti-Dengue Responses. Biomacromolecules 2025, 26, 4449–4463. [Google Scholar] [CrossRef]
- Phan, T.T.N.; Thiono, D.J.; Hvasta, M.G.; Shah, R.P.; Ajo, G.P.; Huang, W.C.; Lovell, J.F.; Tian, S.; de Silva, A.M.; Kuhlman, B. Multivalent administration of dengue E dimers on liposomes elicits type-specific neutralizing responses without immune interference. npj Vaccines 2025, 10, 119. [Google Scholar] [CrossRef]
- Kudlacek, S.T.; Metz, S.; Thiono, D.; Payne, A.M.; Phan, T.T.N.; Tian, S.; Forsberg, L.J.; Maguire, J.; Seim, I.; Zhang, S.; et al. Designed, highly expressing, thermostable dengue virus 2 envelope protein dimers elicit quaternary epitope antibodies. Sci. Adv. 2021, 7, eabg4084. [Google Scholar] [CrossRef]
- Garg, H.; Mehmetoglu-Gurbuz, T.; Joshi, A. Virus Like Particles (VLP) as multivalent vaccine candidate against Chikungunya, Japanese Encephalitis, Yellow Fever and Zika Virus. Sci. Rep. 2020, 10, 4017. [Google Scholar] [CrossRef]
- Erasmus, J.H.; Seymour, R.L.; Kaelber, J.T.; Kim, D.Y.; Leal, G.; Sherman, M.B.; Frolov, I.; Chiu, W.; Weaver, S.C.; Nasar, F. Novel Insect-Specific Eilat Virus-Based Chimeric Vaccine Candidates Provide Durable, Mono- and Multivalent, Single-Dose Protection against Lethal Alphavirus Challenge. J. Virol. 2018, 92, e01274-17. [Google Scholar] [CrossRef]
- Bonaldo, M.C.; Sequeira, P.C.; Galler, R. The yellow fever 17D virus as a platform for new live attenuated vaccines. Hum. Vaccines Immunother. 2014, 10, 1256–1265. [Google Scholar] [CrossRef]
- Carrera, J.; Aktepe, T.E.; Earnest, L.; Christiansen, D.; Wheatley, A.K.; Tan, H.X.; Chung, A.W.; Collett, S.; McPherson, K.; Torresi, J.; et al. Adenovirus vector produced Zika virus-like particles induce a long-lived neutralising antibody response in mice. Vaccine 2023, 41, 4888–4898. [Google Scholar] [CrossRef]
- Cuevas-Juarez, E.; Pando-Robles, V.; Palomares, L.A. Flavivirus vaccines: Virus-like particles and single-round infectious particles as promising alternatives. Vaccine 2021, 39, 6990–7000. [Google Scholar] [CrossRef]
- Henriquez, R.; Munoz-Barroso, I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024, 10, e34927. [Google Scholar] [CrossRef]
- Rothen, D.A.; Dutta, S.K.; Krenger, P.S.; Pardini, A.; Vogt, A.S.; Josi, R.; Lieknina, I.; Osterhaus, A.; Mohsen, M.O.; Vogel, M.; et al. Preclinical Development of a Novel Zika Virus-like Particle Vaccine in Combination with Tetravalent Dengue Virus-like Particle Vaccines. Vaccines 2024, 12, 1053. [Google Scholar] [CrossRef]
- Saito, Y.; Moi, M.L.; Takeshita, N.; Lim, C.K.; Shiba, H.; Hosono, K.; Saijo, M.; Kurane, I.; Takasaki, T. Japanese encephalitis vaccine-facilitated dengue virus infection-enhancement antibody in adults. BMC Infect. Dis. 2016, 16, 578. [Google Scholar] [CrossRef]
- Bielefeldt-Ohmann, H.; Prow, N.A.; Wang, W.; Tan, C.S.; Coyle, M.; Douma, A.; Hobson-Peters, J.; Kidd, L.; Hall, R.A.; Petrovsky, N. Safety and immunogenicity of a delta inulin-adjuvanted inactivated Japanese encephalitis virus vaccine in pregnant mares and foals. Vet. Res. 2014, 45, 130. [Google Scholar] [CrossRef]
- Yun, K.W.; Lee, H.J.; Park, J.Y.; Cho, H.K.; Kim, Y.J.; Kim, K.H.; Kim, N.H.; Hong, Y.J.; Kim, D.H.; Kim, H.M.; et al. Long-term immunogenicity of an initial booster dose of an inactivated, Vero cell culture-derived Japanese encephalitis vaccine (JE-VC) and the safety and immunogenicity of a second JE-VC booster dose in children previously vaccinated with an inactivated, mouse brain-derived Japanese encephalitis vaccine. Vaccine 2018, 36, 1398–1404. [Google Scholar] [CrossRef]
- Lobigs, M.; Pavy, M.; Hall, R.A.; Lobigs, P.; Cooper, P.; Komiya, T.; Toriniwa, H.; Petrovsky, N. An inactivated Vero cell-grown Japanese encephalitis vaccine formulated with Advax, a novel inulin-based adjuvant, induces protective neutralizing antibody against homologous and heterologous flaviviruses. J. Gen. Virol. 2010, 91, 1407–1417. [Google Scholar] [CrossRef]
- Wu, S.J.; Ewing, D.; Sundaram, A.K.; Chen, H.W.; Liang, Z.; Cheng, Y.; Jani, V.; Sun, P.; Gromowski, G.D.; De La Barrera, R.A.; et al. Enhanced Immunogenicity of Inactivated Dengue Vaccines by Novel Polysaccharide-Based Adjuvants in Mice. Microorganisms 2022, 10, 1034. [Google Scholar] [CrossRef]
- Dubischar-Kastner, K.; Eder, S.; Buerger, V.; Gartner-Woelfl, G.; Kaltenboeck, A.; Schuller, E.; Tauber, E.; Klade, C. Long-term immunity and immune response to a booster dose following vaccination with the inactivated Japanese encephalitis vaccine IXIARO, IC51. Vaccine 2010, 28, 5197–5202. [Google Scholar] [CrossRef]
- Friberg, H.; Martinez, L.J.; Lin, L.; Blaylock, J.M.; De La Barrera, R.A.; Rothman, A.L.; Putnak, J.R.; Eckels, K.H.; Thomas, S.J.; Jarman, R.G.; et al. Cell-Mediated Immunity Generated in Response to a Purified Inactivated Vaccine for Dengue Virus Type 1. mSphere 2020, 5, e00671-19. [Google Scholar] [CrossRef]
- Shrestha, B.; Ng, T.; Chu, H.J.; Noll, M.; Diamond, M.S. The relative contribution of antibody and CD8+ T cells to vaccine immunity against West Nile encephalitis virus. Vaccine 2008, 26, 2020–2033. [Google Scholar] [CrossRef]
- Yang, L.; Xiao, A.; Wang, H.; Zhang, X.; Zhang, Y.; Li, Y.; Wei, Y.; Liu, W.; Chen, C. A VLP-Based Vaccine Candidate Protects Mice against Japanese Encephalitis Virus Infection. Vaccines 2022, 10, 197. [Google Scholar] [CrossRef]
- Spohn, G.; Jennings, G.T.; Martina, B.E.; Keller, I.; Beck, M.; Pumpens, P.; Osterhaus, A.D.; Bachmann, M.F. A VLP-based vaccine targeting domain III of the West Nile virus E protein protects from lethal infection in mice. Virol. J. 2010, 7, 146. [Google Scholar] [CrossRef]
- Zhang, H.Q.; Li, N.; Zhang, Z.R.; Deng, C.L.; Xia, H.; Ye, H.Q.; Yuan, Z.M.; Zhang, B. A Chimeric Classical Insect-Specific Flavivirus Provides Complete Protection Against West Nile Virus Lethal Challenge in Mice. J. Infect. Dis. 2024, 229, 43–53. [Google Scholar] [CrossRef]
- Harrison, J.J.; Nguyen, W.; Morgan, M.S.; Tang, B.; Habarugira, G.; de Malmanche, H.; Freney, M.E.; Modhiran, N.; Watterson, D.; Cox, A.L.; et al. A chimeric vaccine derived from Australian genotype IV Japanese encephalitis virus protects mice from lethal challenge. npj Vaccines 2024, 9, 134. [Google Scholar] [CrossRef]
- Kalimuddin, S.; Tham, C.Y.L.; Chan, Y.F.Z.; Hang, S.K.; Kunasegaran, K.; Chia, A.; Chan, C.Y.Y.; Ng, D.H.L.; Sim, J.X.Y.; Tan, H.C.; et al. Vaccine-induced T cell responses control Orthoflavivirus challenge infection without neutralizing antibodies in humans. Nat. Microbiol. 2025, 10, 374–387. [Google Scholar] [CrossRef]



Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Unali, G.; Douam, F. Orthoflavivirus Vaccine Platforms: Current Strategies and Challenges. Vaccines 2025, 13, 1015. https://doi.org/10.3390/vaccines13101015
Unali G, Douam F. Orthoflavivirus Vaccine Platforms: Current Strategies and Challenges. Vaccines. 2025; 13(10):1015. https://doi.org/10.3390/vaccines13101015
Chicago/Turabian StyleUnali, Giulia, and Florian Douam. 2025. "Orthoflavivirus Vaccine Platforms: Current Strategies and Challenges" Vaccines 13, no. 10: 1015. https://doi.org/10.3390/vaccines13101015
APA StyleUnali, G., & Douam, F. (2025). Orthoflavivirus Vaccine Platforms: Current Strategies and Challenges. Vaccines, 13(10), 1015. https://doi.org/10.3390/vaccines13101015
