A Scoping Review of Preclinical Research on Monoclonal Antibody Development for Prophylaxis and Treatment of West Nile Virus Infections
Abstract
1. Introduction
2. Materials and Methods
3. Results
3.1. MAbs Targeting the Envelope Protein
3.2. MAbs Targeting Other Viral Proteins
3.3. Flaviviral MAbs Evaluated for Cross-Protection from WNV Challenge
3.4. Novel Antibody Conformations and Delivery Methods
4. Discussion
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Mackenzie, J.S.; Gubler, D.J.; Petersen, L.R. Emerging flaviviruses: The spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat. Med. 2004, 10 (Suppl. S12), S98–109. [Google Scholar] [CrossRef] [PubMed]
- CDC. ArboNET West Nile Historical Data Maps (1999–2023). 2024. Available online: https://www.cdc.gov/west-nile-virus/data-maps/historic-data.html (accessed on 16 May 2025).
- Staples, J.E.; Shankar, M.B.; Sejvar, J.J.; Meltzer, M.I.; Fischer, M. Initial and long-term costs of patients hospitalized with West Nile virus disease. Am. J. Trop. Med. Hyg. 2014, 90, 402–409. [Google Scholar] [CrossRef] [PubMed]
- Ronca, S.E.; Ruff, J.C.; Murray, K.O. A 20-year historical review of West Nile virus since its initial emergence in North America: Has West Nile virus become a neglected tropical disease? PLoS Neglected Trop. Dis. 2021, 15, e0009190. [Google Scholar] [CrossRef]
- Mbonde, A.A.; Gritsch, D.; Harahsheh, E.Y.; Kasule, S.N.; Hasan, S.; Parsons, A.M.; Zhang, N.; Butterfield, R.; Shiue, H.; Norville, K.A.; et al. Neuroinvasive West Nile Virus Infection in Immunosuppressed and Immunocompetent Adults. JAMA Netw. Open 2024, 7, e244294. [Google Scholar] [CrossRef] [PubMed]
- Bureau, U.S.C. 2020 Census Results. 2020. Available online: https://www.census.gov/programs-surveys/decennial-census/decade/2020/2020-census-results.html (accessed on 1 November 2024).
- Wallace, B.I.; Kenney, B.; Malani, P.N.; Clauw, D.J.; Nallamothu, B.K.; Waljee, A.K. Prevalence of Immunosuppressive Drug Use Among Commercially Insured US Adults, 2018–2019. JAMA Netw. Open 2021, 4, e214920. [Google Scholar] [CrossRef]
- Gould, C.V.; Staples, J.E.; Huang, C.Y.; Brault, A.C.; Nett, R.J. Combating West Nile Virus Disease—Time to Revisit Vaccination. N. Engl. J. Med. 2023, 388, 1633–1636. [Google Scholar] [CrossRef]
- Curren, E.J.; Shankar, M.B.; Fischer, M.; Meltzer, M.I.; Erin Staples, J.; Gould, C.V. Cost-Effectiveness and Impact of a Targeted Age- and Incidence-based West Nile Virus Vaccine Strategy. Clin. Infect. Dis. 2021, 73, 1565–1570. [Google Scholar] [CrossRef]
- Kothari, M.; Wanjari, A.; Acharya, S.; Karwa, V.; Chavhan, R.; Kumar, S.; Kadu, A.; Patil, R. A Comprehensive Review of Monoclonal Antibodies in Modern Medicine: Tracing the Evolution of a Revolutionary Therapeutic Approach. Cureus 2024, 16, e61983. [Google Scholar] [CrossRef]
- Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef]
- Zhang, S.; Vogt, M.R.; Oliphant, T.; Engle, M.; Bovshik, E.I.; Diamond, M.S.; Beasley, D.W. Development of resistance to passive therapy with a potently neutralizing humanized monoclonal antibody against West Nile virus. J. Infect. Dis. 2009, 200, 202–205. [Google Scholar] [CrossRef]
- Vogt, M.R.; Moesker, B.; Goudsmit, J.; Jongeneelen, M.; Austin, S.K.; Oliphant, T.; Nelson, S.; Pierson, T.C.; Wilschut, J.; Throsby, M.; et al. Human monoclonal antibodies against West Nile virus induced by natural infection neutralize at a postattachment step. J. Virol. 2009, 83, 6494–6507. [Google Scholar] [CrossRef] [PubMed]
- Vogt, M.R.; Dowd, K.A.; Engle, M.; Tesh, R.B.; Johnson, S.; Pierson, T.C.; Diamond, M.S. Poorly neutralizing cross-reactive antibodies against the fusion loop of West Nile virus envelope protein protect in vivo via Fcgamma receptor and complement-dependent effector mechanisms. J. Virol. 2011, 85, 11567–11580. [Google Scholar] [CrossRef] [PubMed]
- Throsby, M.; Geuijen, C.; Goudsmit, J.; Bakker, A.Q.; Korimbocus, J.; Kramer, R.A.; Clijsters-van der Horst, M.; de Jong, M.; Jongeneelen, M.; Thijsse, S.; et al. Isolation and characterization of human monoclonal antibodies from individuals infected with West Nile Virus. J. Virol. 2006, 80, 6982–6992. [Google Scholar] [CrossRef] [PubMed]
- Smeraski, C.A.; Siddharthan, V.; Morrey, J.D. Treatment of spatial memory impairment in hamsters infected with West Nile virus using a humanized monoclonal antibody MGAWN1. Antivir. Res. 2011, 91, 43–49. [Google Scholar] [CrossRef]
- Oliphant, T.; Nybakken, G.E.; Engle, M.; Xu, Q.; Nelson, C.A.; Sukupolvi-Petty, S.; Marri, A.; Lachmi, B.E.; Olshevsky, U.; Fremont, D.H.; et al. Antibody recognition and neutralization determinants on domains I and II of West Nile Virus envelope protein. J. Virol. 2006, 80, 12149–12159. [Google Scholar] [CrossRef]
- Oliphant, T.; Engle, M.; Nybakken, G.E.; Doane, C.; Johnson, S.; Huang, L.; Gorlatov, S.; Mehlhop, E.; Marri, A.; Chung, K.M.; et al. Development of a humanized monoclonal antibody with therapeutic potential against West Nile virus. Nat. Med. 2005, 11, 522–530. [Google Scholar] [CrossRef]
- Morrey, J.D.; Siddharthan, V.; Wang, H.; Hall, J.O.; Skirpstunas, R.T.; Olsen, A.L.; Nordstrom, J.L.; Koenig, S.; Johnson, S.; Diamond, M.S. West Nile virus-induced acute flaccid paralysis is prevented by monoclonal antibody treatment when administered after infection of spinal cord neurons. J. Neurovirology 2008, 14, 152–163. [Google Scholar] [CrossRef]
- Morrey, J.D.; Siddharthan, V.; Olsen, A.L.; Roper, G.Y.; Wang, H.; Baldwin, T.J.; Koenig, S.; Johnson, S.; Nordstrom, J.L.; Diamond, M.S. Humanized monoclonal antibody against West Nile virus envelope protein administered after neuronal infection protects against lethal encephalitis in hamsters. J. Infect. Dis. 2006, 194, 1300–1308. [Google Scholar] [CrossRef]
- Gould, L.H.; Sui, J.; Foellmer, H.; Oliphant, T.; Wang, T.; Ledizet, M.; Murakami, A.; Noonan, K.; Lambeth, C.; Kar, K.; et al. Protective and therapeutic capacity of human single-chain Fv-Fc fusion proteins against West Nile virus. J. Virol. 2005, 79, 14606–14613. [Google Scholar] [CrossRef]
- Goo, L.; Debbink, K.; Kose, N.; Sapparapu, G.; Doyle, M.P.; Wessel, A.W.; Richner, J.M.; Burgomaster, K.E.; Larman, B.C.; Dowd, K.A.; et al. A protective human monoclonal antibody targeting the West Nile virus E protein preferentially recognizes mature virions. Nat. Microbiol. 2019, 4, 71–77. [Google Scholar] [CrossRef]
- Duan, T.; Ferguson, M.; Yuan, L.; Xu, F.; Li, G. Human Monoclonal Fab Antibodies Against West Nile Virus and its Neutralizing Activity Analyzed in Vitro and in Vivo. J. Antivir. Antiretrovir. 2009, 1, 36–42. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.J.; Luo, H.R.; Fan, Z.Y.; Feng, Y.X.; Wei, N.; Zhu, B.B.; Ye, J.; Cao, S.B.; Si, Y.H. Development and evaluation of neutralizing antibodies for cross-protection against West Nile virus and Japanese encephalitis virus. Infect. Med. 2023, 2, 212–223. [Google Scholar] [CrossRef]
- Wessel, A.W.; Doyle, M.P.; Engdahl, T.B.; Rodriguez, J.; Crowe, J.E., Jr.; Diamond, M.S. Human Monoclonal Antibodies against NS1 Protein Protect against Lethal West Nile Virus Infection. mBio 2021, 12, e0244021. [Google Scholar] [CrossRef]
- Chung, K.M.; Liszewski, M.K.; Nybakken, G.; Davis, A.E.; Townsend, R.R.; Fremont, D.H.; Atkinson, J.P.; Diamond, M.S. West Nile virus nonstructural protein NS1 inhibits complement activation by binding the regulatory protein factor H. Proc. Natl. Acad. Sci. USA 2006, 103, 19111–19116. [Google Scholar] [CrossRef]
- Calvert, A.E.; Kalantarov, G.F.; Chang, G.J.; Trakht, I.; Blair, C.D.; Roehrig, J.T. Human monoclonal antibodies to West Nile virus identify epitopes on the prM protein. Virology 2011, 410, 30–37. [Google Scholar] [CrossRef] [PubMed]
- Ozawa, T.; Masaki, H.; Takasaki, T.; Aoyama, I.; Yumisashi, T.; Yamanaka, A.; Konishi, E.; Ohnuki, Y.; Muraguchi, A.; Kishi, H. Human monoclonal antibodies against West Nile virus from Japanese encephalitis-vaccinated volunteers. Antivir. Res. 2018, 154, 58–65. [Google Scholar] [CrossRef]
- Modhiran, N.; Song, H.; Liu, L.; Bletchly, C.; Brillault, L.; Amarilla, A.A.; Xu, X.; Qi, J.; Chai, Y.; Cheung, S.T.M.; et al. A broadly protective antibody that targets the flavivirus NS1 protein. Science 2021, 371, 190–194. [Google Scholar] [CrossRef] [PubMed]
- Gupta, A.K.; Lad, V.J.; Koshy, A.A. Survival of mice immunized with monoclonal antibodies against glycoprotein E of Japanese encephalitis virus before or after infection with Japanese encephalitis, West Nile, and Dengue viruses. Acta Virol. 2008, 52, 219–224. [Google Scholar]
- Deng, Y.Q.; Dai, J.X.; Ji, G.H.; Jiang, T.; Wang, H.J.; Yang, H.O.; Tan, W.L.; Liu, R.; Yu, M.; Ge, B.X.; et al. A broadly flavivirus cross-neutralizing monoclonal antibody that recognizes a novel epitope within the fusion loop of E protein. PLoS ONE 2011, 6, e16059. [Google Scholar] [CrossRef]
- Pierson, T.C.; Xu, Q.; Nelson, S.; Oliphant, T.; Nybakken, G.E.; Fremont, D.H.; Diamond, M.S. The stoichiometry of antibody-mediated neutralization and enhancement of West Nile virus infection. Cell Host Microbe 2007, 1, 135–145. [Google Scholar] [CrossRef]
- Kaufmann, B.; Nybakken, G.E.; Chipman, P.R.; Zhang, W.; Diamond, M.S.; Fremont, D.H.; Kuhn, R.J.; Rossmann, M.G. West Nile virus in complex with the Fab fragment of a neutralizing monoclonal antibody. Proc. Natl. Acad. Sci. USA 2006, 103, 12400–12404. [Google Scholar] [CrossRef] [PubMed]
- Morrey, J.D.; Siddharthan, V.; Olsen, A.L.; Wang, H.; Julander, J.G.; Hall, J.O.; Li, H.; Nordstrom, J.L.; Koenig, S.; Johnson, S.; et al. Defining limits of treatment with humanized neutralizing monoclonal antibody for West Nile virus neurological infection in a hamster model. Antimicrob. Agents Chemother. 2007, 51, 2396–2402. [Google Scholar] [CrossRef]
- Pereboev, A.; Borisevich, V.; Tsuladze, G.; Shakhmatov, M.; Hudman, D.; Kazachinskaia, E.; Razumov, I.; Svyatchenko, V.; Loktev, V.; Yamshchikov, V. Genetically delivered antibody protects against West Nile virus. Antivir. Res. 2008, 77, 6–13. [Google Scholar] [CrossRef]
- Lai, H.; Engle, M.; Fuchs, A.; Keller, T.; Johnson, S.; Gorlatov, S.; Diamond, M.S.; Chen, Q. Monoclonal antibody produced in plants efficiently treats West Nile virus infection in mice. Proc. Natl. Acad. Sci. USA 2010, 107, 2419–2424. [Google Scholar] [CrossRef]
- Lai, H.; He, J.; Hurtado, J.; Stahnke, J.; Fuchs, A.; Mehlhop, E.; Gorlatov, S.; Loos, A.; Diamond, M.S.; Chen, Q. Structural and functional characterization of an anti-West Nile virus monoclonal antibody and its single-chain variant produced in glycoengineered plants. Plant Biotechnol. J. 2014, 12, 1098–1107. [Google Scholar] [CrossRef] [PubMed]
- He, J.; Lai, H.; Engle, M.; Gorlatov, S.; Gruber, C.; Steinkellner, H.; Diamond, M.S.; Chen, Q. Generation and analysis of novel plant-derived antibody-based therapeutic molecules against West Nile virus. PLoS ONE 2014, 9, e93541. [Google Scholar] [CrossRef] [PubMed]
- Lyu, X.; Zhao, Q.; Hui, J.; Wang, T.; Lin, M.; Wang, K.; Zhang, J.; Shentu, J.; Dalby, P.A.; Zhang, H.; et al. The global landscape of approved antibody therapies. Antib. Ther. 2022, 5, 233–257. [Google Scholar] [CrossRef]
- Society, T.A. Therapeutic Monoclonal Antibodies Approved or in Regulatory Review. 2025. Available online: https://www.antibodysociety.org/antibody-therapeutics-product-data (accessed on 16 May 2025).
- Yetmar, Z.A.; Khodadadi, R.B.; Seville, M.T.; Brumble, L.; O’Horo, J.C.; Ganesh, R.; Razonable, R.R. Outcomes of B-Cell-Depleted Patients with Coronavirus Disease 2019 Treated with Antispike Monoclonal Antibodies. Open Forum Infect. Dis. 2022, 9, ofac204. [Google Scholar] [CrossRef]
- Domachowske, J.; Hamren, U.W.; Banu, I.; Baronio, R.; Basavaraju, B.; Koen, A.; Leach, A.; Mankad, V.S.; Pannaraj, P.S.; Soler-Palacin, P.; et al. Safety and Pharmacokinetics of Nirsevimab in Immunocompromised Children. Pediatrics 2024, 154, e2024066508. [Google Scholar] [CrossRef]
- FDA. Product Development Under the Animal Rule Guidelines for Industry. 2015. Available online: http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htm (accessed on 16 May 2025).
- Baric, T.J.; Reneer, Z.B. Animal Models, Therapeutics, and Vaccine Approaches to Emerging and Re-Emerging Flaviviruses. Viruses 2024, 17, 1. [Google Scholar] [CrossRef]
- Rey, F.A.; Heinz, F.X.; Mandl, C.; Kunz, C.; Harrison, S.C. The envelope glycoprotein from tick-borne encephalitis virus at 2 A resolution. Nature 1995, 375, 291–298. [Google Scholar] [CrossRef] [PubMed]
- Modis, Y.; Ogata, S.; Clements, D.; Harrison, S.C. A ligand-binding pocket in the dengue virus envelope glycoprotein. Proc. Natl. Acad. Sci. USA 2003, 100, 6986–6991. [Google Scholar] [CrossRef] [PubMed]
- Mandl, C.W.; Guirakhoo, F.; Holzmann, H.; Heinz, F.X.; Kunz, C. Antigenic structure of the flavivirus envelope protein E at the molecular level, using tick-borne encephalitis virus as a model. J. Virol. 1989, 63, 564–571. [Google Scholar] [CrossRef] [PubMed]
- Thompson, B.S.; Moesker, B.; Smit, J.M.; Wilschut, J.; Diamond, M.S.; Fremont, D.H. A therapeutic antibody against west nile virus neutralizes infection by blocking fusion within endosomes. PLoS Pathog. 2009, 5, e1000453. [Google Scholar] [CrossRef]
- Beigel, J.H.; Nordstrom, J.L.; Pillemer, S.R.; Roncal, C.; Goldwater, D.R.; Li, H.; Holland, P.C.; Johnson, S.; Stein, K.; Koenig, S. Safety and pharmacokinetics of single intravenous dose of MGAWN1, a novel monoclonal antibody to West Nile virus. Antimicrob. Agents Chemother. 2010, 54, 2431–2436. [Google Scholar] [CrossRef]
- Treatment of West Nile Virus with MGAWN1 (PARADIGM). 2022. Available online: https://clinicaltrials.gov/study/NCT00927953?intr=mgawn1&rank=3 (accessed on 25 October 2024).
- Cherrier, M.V.; Kaufmann, B.; Nybakken, G.E.; Lok, S.M.; Warren, J.T.; Chen, B.R.; Nelson, C.A.; Kostyuchenko, V.A.; Holdaway, H.A.; Chipman, P.R.; et al. Structural basis for the preferential recognition of immature flaviviruses by a fusion-loop antibody. EMBO J. 2009, 28, 3269–3276. [Google Scholar] [CrossRef]
- Carpio, K.L.; Barrett, A.D.T. Flavivirus NS1 and Its Potential in Vaccine Development. Vaccines 2021, 9, 622. [Google Scholar] [CrossRef]
- Chung, K.M.; Thompson, B.S.; Fremont, D.H.; Diamond, M.S. Antibody recognition of cell surface-associated NS1 triggers Fc-gamma receptor-mediated phagocytosis and clearance of West Nile Virus-infected cells. J. Virol. 2007, 81, 9551–9555. [Google Scholar] [CrossRef]
- Austin, S.K.; Dowd, K.A. B cell response and mechanisms of antibody protection to West Nile virus. Viruses 2014, 6, 1015–1036. [Google Scholar] [CrossRef]
- Calisher, C.H.; Karabatsos, N.; Dalrymple, J.M.; Shope, R.E.; Porterfield, J.S.; Westaway, E.G.; Brandt, W.E. Antigenic relationships between flaviviruses as determined by cross-neutralization tests with polyclonal antisera. J. Gen. Virol. 1989, 70 Pt 1, 37–43. [Google Scholar] [CrossRef]
- Terstappen, G.C.; Meyer, A.H.; Bell, R.D.; Zhang, W. Strategies for delivering therapeutics across the blood-brain barrier. Nat. Rev. Drug Discov. 2021, 20, 362–383. [Google Scholar] [CrossRef] [PubMed]
- Stevens, D.; Claborn, M.K.; Gildon, B.L.; Kessler, T.L.; Walker, C. Onasemnogene Abeparvovec-xioi: Gene Therapy for Spinal Muscular Atrophy. Ann. Pharmacother. 2020, 54, 1001–1009. [Google Scholar] [CrossRef] [PubMed]
- Bajracharya, R.; Caruso, A.C.; Vella, L.J.; Nisbet, R.M. Current and Emerging Strategies for Enhancing Antibody Delivery to the Brain. Pharmaceutics 2021, 13, 2014. [Google Scholar] [CrossRef]
- Mohak, S.; Fabian, Z. Extracellular Vesicles as Precision Delivery Systems for Biopharmaceuticals: Innovations, Challenges, and Therapeutic Potential. Pharmaceutics 2025, 17, 641. [Google Scholar] [CrossRef] [PubMed]
- Soleimanizadeh, A.; Dinter, H.; Schindowski, K. Central Nervous System Delivery of Antibodies and Their Single-Domain Antibodies and Variable Fragment Derivatives with Focus on Intranasal Nose to Brain Administration. Antibodies 2021, 10, 47. [Google Scholar] [CrossRef]
- Verma, V.; Sinha, N.; Raja, A. Nanoscale warriors against viral invaders: A comprehensive review of Nanobodies as potential antiviral therapeutics. mAbs 2025, 17, 2486390. [Google Scholar] [CrossRef]
- Tien, J.; Leonoudakis, D.; Petrova, R.; Trinh, V.; Taura, T.; Sengupta, D.; Jo, L.; Sho, A.; Yun, Y.; Doan, E.; et al. Modifying antibody-FcRn interactions to increase the transport of antibodies through the blood-brain barrier. mAbs 2023, 15, 2229098. [Google Scholar] [CrossRef]
Study | MAb ID | Epitope Target | Dosage/Inoculation Routes | Prophylaxis/Therapy Evaluation | Animal Model | WNV Infectious Dose/Route | Baseline Survival After WNV Infection | Outcomes |
---|---|---|---|---|---|---|---|---|
Gould, et al. 2005 [21] | recombinant human IgG1 11, 15, 73, and 95 | E, DI/DII | single/SC | 100 µg 24 h before infection | C57BL/6 mice (4–6 weeks) | 100 PFU/IP | 0% | 100% survival rate |
recombinant human IgG1 11 and 15 | double/SC | 100 µg 24 h and 4 days pi | 80% survival rate | |||||
recombinant human IgG1 11 | single/SC | 250 µg 24 h pi, 500 µg 3 days pi, or 500 µg 5 days pi | 100%, 100%, and 60% survival rate, respectively | |||||
Oliphant, et al., 2005 [18] | murine Mabs E16, E24, E34 | E/DIII | single/IP | 100 µg administered 48 h pi | C57BL/6 mice (5 weeks) | 100 PFU/SC | 8–20% | >90% survival rate |
murine MAbs E16, E24 | 500 µg administered 4 days pi | 80–90% survival rate | ||||||
murine Mabs E16 | 2 mg administered 5 days pi | 90% survival rate, and cleared WNV from brain in 68% of mice by day 9 | ||||||
murine-human chimeric ChE16 IgG1 | 100 µg administered 48 h pi | 90% survival rate | ||||||
humanized hE16.1 IgG1 | 100 µg administered 48 h pi | 100% survival rate | ||||||
Morrey, et al., 2006 [20] | hE16 | E/DIII | single/IP | 60 mg/kg administered 48 h pi | Syrian golden hamsters (>7 weeks of age) | 0.1 mL of 106.3 TCID50/mL/SC | 20–35% | 95% survival rate |
100 mg/kg administered 5 days pi | 37% | 80% survival rate | ||||||
single/IC | 50 mg/kg administered 5 days pi | 22% | 88% survival rate | |||||
Oliphant et al., 2006 [17] | murine MAbs E60, E53, E18, E121, E113, E31, E48, E100 | E, DI/II | single/IP | 400 µg of MAb 24 h before infection | C57BL/6 mice (5 weeks) | 100 PFU/footpad | 13% | ≥75% survival rate with all MAbs, exept E100 |
murine MAbs E53, E60, E31, E113 | 500 of MAb 48 h pi | C57BL/6 mice (5 weeks) | ≥39% survival rate, 85% survival rate for E31 | |||||
murine MAbs, E53, E60, E121 | 400 µg of MAb 24 h before infection | Fcy receptor-deficient C57BL/6 mice (8 weeks) | ≥61% survival rate | |||||
Throsby et al., 2006 [15] | recombinant human IgG1 CR4374 | E, DIII | single/IP | 15 mg/kg of MAb 24 h before infection | BALB/c mice (4 weeks) | 20 × 9.5 TCID50/IP | 0% | 100% survival rate |
Morrey, et al., 2008 [19] | hE16 | E/DIII | single/IP | 32 mg/kg administered 2-, 3-, 4-, or 5 days pi | Syrian golden hamsters (>7 weeks of age) | 101.8 PFU/spinal cord | 0% | 100% survival rate 2 days pi, 83% survival rate 3 days pi, 29% survival rate 4 days pi, 0% survival rate 5 days pi |
Duan, et al., 2009 [23] | human Fab 1 | E/DIII | single/IP | 200 µl Fab 1 administered 24 h before infection, concurrently, or 24 h pi | C57BL/6 mice (4–6 weeks) | 103PFU/IP | 0% | 25% survival rate |
Vogt et al., 2009 [13] | recombinant human IgG1 CR4348 and CR4354 | E, DI/DII | single/IP | 50 µg 24 h before infection | NIH Swiss mice (3–4 weeks) | 300 PFU/IP | 25% | 94% survival rate with CR4354 |
0.014 to 14 µg 24 h before infection | C57BL/6 mice (5 weeks) | 100 PFU/SC | 20% | 100% survival rate in mice given ≥1.4 and ≥0.42 µg of CR4348 and CR4354, respectively | ||||
Zhang, et al., 2009 [12] | hE16 | E/DIII | single/IP | 100 µg administered 24 h before infection | Swiss Webster (3–4 weeks) | 100 PFU WNV variants/SC | 0–20% | 30%, 10%, and 0% survival rate with WNV NY99 variants T332K, K307E, or WNV SA58, respectively |
100 µg administered 48 h pi | congenic RAG C57BL/6 mice (5 weeks) | 0–30% | 100% survival rate with WNV NY99 variants T332A, T332M, T330I, or WNV SA58 K332T 10% survival rate with WNV SA58 | |||||
Smeraski, et al., 2011 [16] | MGAWN1 (hE16) | E/DIII | single/IP | 32 mg/kg 4.5 days pi | Syrian golden hamsters (9–10 weeks) | 104PFU/SC | 56% maintained spatial memory | MGAWN1 improved survival weight, weight loss, and anorexia in WNV-infected hamsters. MGAWN1-treated infected hamsters showed a 92% retention in long-term spatial memory |
Vogt, et al., 2011 [14] | murine-human chimeric IgG1 MAbs E28 and E53 | E, DII-fusion loop | single/IP | 40 µg 24 h before infection | C57BL/6J mice (4–5 weeks) | 100 PFU/footpad | 14% | 60% and 43% survival rate with E28 and E53, respectively |
Goo et al., 2019 [22] | human MAb WNV-86 | E, DII | single/IP | 100 µg 48 h pi | C57BL/6 mice (5 weeks) | 100 FFU/SC | 10% | 100% survival rate |
Yang et al., 2023 [24] | C9-G11-F3 and B2-D1-H6 | E | triple/IP | 5 mg/kg three consecutive days pi | C57BL/6 mice (4 weeks) | 106 PFU/IP | 0% | 20%, 40% and 66.7% survival rate with B2-D1-H6, C9-G11-F3, and B2-D1-H6 + C9-G11-F3 combined, respectively |
Study | MAb ID | Epitope Target | Dosage/Inoculation Routes | Prophylaxis/Therapy Evaluation | Animal Model | WNV infectious Dose/Route | Baseline Survival after WNV Infection | Outcomes |
---|---|---|---|---|---|---|---|---|
Chung et al., 2006 [26] | murine MAbs 10NS1, 14NS1, 16NS1, and 17NS1 | NS1 | single/IP | 500 µg administered concurrently with infection | C57BL/6 mice (6 weeks) | 100 PFU/footpad | 17% | 75–95% survived rate |
murine MAb 17NS1 | dose response (0.8 to 500 µg) administered concurrently with infection | C57BL/6 mice (4 weeks) | 45% survival rate with 4 µg | |||||
murine MAb 14NS1 | 1 mg administered 7 or 14 days before infection | C57BL/6 mice (4 weeks) | 70% and 80% survival rate 7 and 14 days before infection, respectively. | |||||
murine MAbs 14NS1, 16NS1 | 500 µg administered 2- or 4- days pi | C57BL/6 mice (5 weeks) | 10% | 68% and 47% survival rate with 14NS1 and 16NS1, respectively, 2 days pi. 60% survival rate with 14NS1 and 16NS1 4 days pi. | ||||
Calvert, et al., 2011 [27] | human MAbs 8G8, 5G12 | prM | single/IP | 500 µg 24 h before infection | Swiss Webster (3–4 weeks) | 100 PFU/IP | 6% | 10–30% survival rate |
AG129 IFN-receptor deficient mice (5–8 weeks) | 0% | 0% survival rate | ||||||
Wessel, et al., 2021 [25] | human MAbs WNV-103, WNV-95, WNV-96, WNV-97, WNV-99 WNV-100, WNV-104, WNV-117, WNV-98, WNV-113, WNV-116 | NS1 | single/footpad | 200 µg administered concurrently with infection | C57BL/6 mice (4–5 weeks) | 100 FFU/footpad | 10–15% | 43–75% survival rates |
Study | MAb ID | Epitope Target | Dosage/Inoculation Routes | Prophylaxis/Therapy Evaluation | Animal Model | WNV Infectious Dose/Route | Baseline Survival After WNV Infection | Outcomes |
---|---|---|---|---|---|---|---|---|
Gupta et al., 2008 [30] | murine MAb Hx-1, Hx-2, Hs-3, Hx-5 | JEV-E | single/IP | 100 µl ascitic fluid administered 24 h before infection | Swiss mice (3–4 weeks) | 100 LD50/IC | 0% | 45%, 70%, 0%, and 25% survival rate for Hx-1, Hx-2, Hs-3, Hx-5, respectively |
100 µl ascitic fluid administered 48 h pi | 40%, 0%, 55%, and 35% survival rate for Hx-1, Hx-2, Hs-3, Hx-5, respectively | |||||||
Deng, et al., 2011 [31] | murine MAb 2A10G6 | DENV2 E, DII-fusion loop | single/IP | 200 µg 24 h before infection | BALB/c mice (4 weeks) | 40 PFU/IC | 0% | 80% of mice survived WNV challenge when treated prophylactically |
200 µg 24 h pi | 37.5% survived challenge when treated therapeutically | |||||||
Ozawa, et al., 2018 [28] | human Fab WN_11, WN_39, WN_83 | WNV-E from JEV-vaccinated volunteers | double/SC | 350 µg administered 24 h before and concurrently with infection | C57BL/6 mice (5 weeks) | 5 × LD50/IP | 0% | 50%, 0%, and 0% survival rate with WN_83, WN_39 and WN_11, respectively |
Modhiran et al., 2021 [29] | murine-human chimeric 10G5.3 IgG1 | DENV-NS1 | single/IP | 200 µg administered 24 h pi | CD1 mice (18–19 days) | 1000 FFU/IP | 0% | 0% survival rate |
Study | MAb ID | Epitope Target | Dosage/Inoculation Routes | Prophylaxis/Therapy Evaluation | Animal Model | WNV Infectious Dose/Route | Baseline Survival After WNV Infection | Outcomes |
---|---|---|---|---|---|---|---|---|
Morrey, et al., 2007 [34] | hE16 | E/DIII | single/IP | 32 mg/kg administered 5- and 6- days pi | Syrian golden hamsters (>7 weeks of age) | 107.1 TCID50/SC | <50% | >75% and <60% survival rate at 5- and 6- days pi, respectively |
single/convection enhanced delivery (CED) | 43 mg/kg administered 5 days pi | 105.3 TCID50/SC | 25% | >75% survival rate | ||||
31 mg/kg administered 6 days pi | 107.1 TCID50/SC | 25% | >75% survival rate | |||||
48 mg/kg administered 8 days after challenge | 55% | <45% survival rate | ||||||
Pereboev, et al., 2008 [35] | Ad/Fc-9E2 | E/DIII | single/IP | 109 PFU 24 h before, concurrently, or 24 h pi | Swiss Webster mice (4 weeks) | 200 PFU/IP | 20% | 83.3% survival rate when administered 24 h before infection. 100% survival rate when administered concurrently or 24 h pi |
Lai, et al., 2010 [36] | pHu-E16 | E/DIII | single/IP | 0.001 to 10 µg administered same day as infection | C57BL/6 mice (5 weeks) | 100 PFU/SC | 20% | 80% survival rate with 10 µg of pHu-E16 |
4 to 100 µg administered 48 h pi | 30% | 90% and 100% survival rate with 20 and 100 µg, respectively | ||||||
50 and 500 µg administered 4 days pi | 10% | 70% and 90% survival with 50 and 500 µg, respectively | ||||||
Lai, et al., 2014 [37] | ΔXFpE16 or ΔXFpE16scFv-CH plant-derived | E/DIII | single/IP | 1 ng to 1 µg administered concurrently with infection | C57BL/6 mice (5 weeks) | 100 PFU/SC | 20% | 100% and 70% survival rate with 100 ng of ΔXFpE16 or ΔXFpE16scFv-CH, respectively. |
500 µg administered 4 days pi | 10–30% | 85% survival rate with 100 µg of ΔXFpE16scFv-CH. | ||||||
He, et al., 2014 [38] | pHu-E16scFv-CH1-3 | E/DIII | single/IP | 1 to 100 ng same day as infection | C57BL/6 mice (5 weeks) | 100 PFU/SC | 20% | 40%, 60%, and 80% survival rates with 1 ng, 10 ng, 100 ng, respectively. |
pHu-E16scFv-CH1-3 and Tetra pHu-E16 | 50 µg 4 days pi | 75% and 90% survival rates with pHu-E16 scFv-CH1-3 and Tetra pHu-E16, respectively. |
Study | Summary of Main Findings |
---|---|
Gould, et al., 2005 [21] | A panel of human single-chain variable fragments (scFvs) specific to WNV-E protein was developed to determine protective efficacy in a mouse model. Five scFvs offered significant protection when administered before or after WNV infection, making them promising candidates for therapeutics. Addition of the Fc region to scFvs increased the half-lives of antibody molecules as well as their protective efficacy. |
Oliphant, et al., 2005 [18] | A panel of murine MAbs against E-DIII strongly neutralized WNV and recognized epitopes on the lateral ridge of DIII. MAb E16 was effective at treating mice from lethal WNV infection when administered as a single dose 5 days pi. A single dose of a humanized version, hE16, was effective at protecting mice from lethal WNV infection. Humanization of E16 will likely increase half-life in humans, and Fc mediated functions including complement fixation and cytotoxicity improving its efficacy as a therapeutic. |
Chung et al., 2006 [26] | This study investigated the therapeutic potential of a panel of 22 anti-NS1 MAbs for treatment of WNV infection. A single dose of MAb 14NS1 provided significant long-lasting protection when administered up to 14 days before infection, and 2–4 days pi. Anti-NS1 MAbs may offer protection via complement activation or other complement-independent pathways. Inclusion of anti-NS1 MAbs in MAb cocktails for therapuetics may increase potency and help to prevent emergence of treatment resistance. |
Morrey, et al., 2006 [20] | Humanized E16, hE16, was evaluated as a therapeutic in Syrian golden hamster model. A single dose of hE16 protected hamsters when administered 48 h or 5 days pi and reduced viral load in the brain and spleen. HE16 was effective at treating WNV infection in a second lethal animal model, demonstrating its potential utility as a treatment for WNV infection in humans. |
Oliphant et al., 2006 [17] | MAbs targeting E-DII and E-DI were evaluated for their ability to protect mice from lethal WNV infection. These MAbs were not as potent neutralizers as their E-DIII counterparts but were able to protect mice from lethal WNV infection, albeit at lower efficacy than E-DIII MAbs. When evaluated in Fc-receptor deficiently mice, the protective efficacy of E-DII and E-DI MAbs was unaffected. |
Throsby et al., 2006 [15] | Human MAbs generated to WNV with phage display library constructed from peripheral blood lymphocytes of three patients with previous WNV infections were examined for their ability to protect mice from lethal challenge, with CR4374 offering the most protective efficacy. The human antibody repertoire to WNV is made up mostly of nonneutralizing or weakly neutralizing MAbs specific to E-DII, and E-DIII specific MAbs while rare, are the most potent neutralizers of WNV. |
Morrey, et al., 2006 [20] | This study investigated the use of hE16 for treating WNND and found that hE16 could protect hamsters from death even when administered 5 days pi with a dose of 0.32 mg/kg. Different administration routes may improve the efficacy of hE16. |
Gupta et al. 2008 [30] | The study investigated the cross-protective efficacy of anti-JEV E MAbs to WNV and DENV2. While some MAbs were able to cross-protect against WNV infection, their efficacy was limited and did not provide significant protection. |
Morrey, et al., 2008 [19] | Humanized E16 was evaluated for its ability to treat WNV paralysis in hamsters when administered various times after infection. MAb hE16 was able to significantly reduce paralysis and mortality when given before the onset of paralysis (approximately 48 h pi) and lowered viral load in the spinal cord. The study demonstrates the therapeutic efficacy of hE16 in reducing paralysis and mortality in an animal model. |
Pereboev, et al., 2008 [35] | This study investigated whether recombinant antibodies delivered via adenovirus vector could be used to treat WNV infection. After inoculation with Ad/Fc-9E2, recombinant antibody was detected in serum within 24 h and remained detectable up to 21 days post-inoculation. When used as a prophylactic Ad/Fc-9E2 protected mice from lethal WNV infection with a survival rate of 83.3% demonstrating a novel and effective prophylactic delivery method for WNV therapeutic MAbs. |
Duan, et al., 2009 [23] | Human anti-WNV Fabs were analyzed for their ability to neutralize in vitro and protect in vivo in a mouse model. Fab1 had strong neutralizing capacity in vitro but failed to protect mice in vivo implying that therapeutic MAb efficacy relies on factors beyond neutralization. |
Vogt et al., 2009 [13] | Two strongly neutralizing anti-WNV human MAbs, CR4348 and CR4354, recognize a unique epitope on the virion surface at the dimer interface in DII and hinge regions between DI and DII and inhibit viral infection at a postattachment step. Their ability to protect mice from lethal WNV infection demonstrates their utility as a therapeutic MAb that may be used in combination with anti-E-DIII MAbs to enhance therapeutic capacity. |
Zhang, et al., 2009 [12] | The study investigated prophylactic and therapeutic efficacy of hE16 to protect mice from WNV challenge, and the potential for treatment to select for neutralization escape variants. MAb hE16 protected mice from wild type WNV infection, but failed to protect mice infected with variants with mutations in the epitope of hE16 at positions 307 (K307E) and 332 (T332M). WNV resistant variants developed in mice treated with hE16 and exposed to high doses of virus. While hE16 offers strong protective efficacy to WNV infection, resistance may arise, particularly in immunocompromised hosts, and combinantions of MAb targeting different epitopes may improve therapeutic efficacy. |
Lai, et al., 2010 [36] | hE16 produced in plants (pHu-E16) was evaluated for its ability to treat WNND in vivo. pHU-E16 was effective at protecting mice from lethal WNV infection when administered either before or after infection, and was comparable to chimeric hE16 produced in mammalian cell culture. The study highlights the effectiveness of therapeutic MAb produced in plants that may be a more cost-effective method for production. |
Calvert, et al., 2011 [27] | This study describes the limited protective efficacy of non-neutralizing WNV MAbs targeting the prM protein. |
Deng, et al., 2011 [31] | This study details the characterization of flavivirus cross-reactive murine MAbs 2A10G6 recognizing an epitope in the fusion loop of E-DII. While not an effective neutralizer in vitro, the MAb did afford some protection before WNV infection in vivo. |
Smeraski, et al., 2011 [16] | The study investigated the effectiveness of MAb MGAWN1 (hE16) to treat WNV-infected hamsters and assessed the long-term memory retention of surviving mice using the Morris-Water Maze (MWM) test. MGAWN1 treatment prevented cognitive impairement and improved survival in treated hamsters highlighting its potential as a therapuetic MAb for the treatment of WNV by preventing long-term cognitive decline. |
Vogt, et al., 2011 [14] | Anti-E DII MAbs specific to the fusion loop were investigated for their use as therapeutic MAbs for WNV infection. While non-neutralizing in vitro, MAb E28 was able to protect mice in vivo from lethal WNV infection. This protection was dependent upon Fc-mediated mechanisms of protection. |
He, et al., 2014 [38] | The study investigated the efficacy of engineering pHu-E16 into a tetravalent form (tetra pHu-E16) as a bifunctional MAb for therapeutic use. Tetra pHu-E16 and other variants purified from plants retained binding and neutralization to WNV similar to hE16, and showed enhanced protection for WNND when administered therapeutically. The study demonstrates tetra pHu-E16’s utility as a potential therapeutic MAb for WNND. |
Lai, et al., 2014 [37] | This study expands on previous work (Lai et al., 2010) [36] with pE16 expression in Nicotiana benthamiana plants that lack specific N-glycan residues to more closely resemble the mammalian glycosylation profile. A single dose of pE16 plant derivatives protected mice from lethal WNV infection even when administered 4 days after infection. This plant production platform may be a more cost-effective way to produce antibody-based therapeutics. |
Ozawa, et al., 2018 [28] | Human anti-WNV MAbs were isolated using peripheral blood lymphocytes from JEV-vaccinated individuals and were shown to neutralize in vitro and in vivo offering some protection from WNV infection in mice when administered prophlactically. The study highlights the potential for cross-reactive antibodies to offer some protection to WNV. |
Goo et al., 2019 [22] | Highly neutralizing human MAb WNV-86 recognizes an epitope on E-DII in mature virions lacking prM protein and protected mice therapeutically from lethal WNV infection. Compared to hE16, WNV-86 offered more robust protection in vivo highlighting its potential as a therapeutic MAb for WNND. |
Modhiran et al., 2021 [29] | A broadly cross-reactive anti-NS1 DENV MAb was evaluated for its ability to confer protection in vivo against several flavivirueses including WNV. While the MAb offered some protection for DENV and ZIKV infection, no protective efficacy to WNV was observed. Even so, the study highlights the importance of a NS1 targeted vaccine and therapeutic approach for protection. |
Wessel, et al., 2021 [25] | The study describes the generation of human anti-NS1 MAbs and their protective efficacy in vivo in an Fc-dependent manner. While non-neuralizing, anti-NS1 MAbs may provide therapeutic protection for WNV infection. |
Yang et al., 2023 [24] | This study describes the development of WNV MAbs and evaluation for therapeutic efficacy in mice. While two MAbs offered some protection in vivo, their efficacy significantly increased when administered together. Overall, the study demonstrates the protective efficacy of MAbs for WNV infection and reduction in neuroinflammation of treated animals. |
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
Calvert, A.E.; Miazgowicz, K.L.; Atkinson, B.; Long, A.H.; Thrasher, E.; Brault, A.C.; Nett, R.J. A Scoping Review of Preclinical Research on Monoclonal Antibody Development for Prophylaxis and Treatment of West Nile Virus Infections. Viruses 2025, 17, 845. https://doi.org/10.3390/v17060845
Calvert AE, Miazgowicz KL, Atkinson B, Long AH, Thrasher E, Brault AC, Nett RJ. A Scoping Review of Preclinical Research on Monoclonal Antibody Development for Prophylaxis and Treatment of West Nile Virus Infections. Viruses. 2025; 17(6):845. https://doi.org/10.3390/v17060845
Chicago/Turabian StyleCalvert, Amanda E., Kerri L. Miazgowicz, Bailey Atkinson, Audrey H. Long, Elisa Thrasher, Aaron C. Brault, and Randall J. Nett. 2025. "A Scoping Review of Preclinical Research on Monoclonal Antibody Development for Prophylaxis and Treatment of West Nile Virus Infections" Viruses 17, no. 6: 845. https://doi.org/10.3390/v17060845
APA StyleCalvert, A. E., Miazgowicz, K. L., Atkinson, B., Long, A. H., Thrasher, E., Brault, A. C., & Nett, R. J. (2025). A Scoping Review of Preclinical Research on Monoclonal Antibody Development for Prophylaxis and Treatment of West Nile Virus Infections. Viruses, 17(6), 845. https://doi.org/10.3390/v17060845