AAV Vectored Immunoprophylaxis for Filovirus Infections
Abstract
:1. Introduction
2. Filoviruses
2.1. A Brief History of Filovirus Outbreaks
2.2. Filovirus Taxonomy
2.3. Filovirus Molecular Biology and Pathogenesis
3. Filovirus Vaccine Development
4. Filovirus Monoclonal Antibodies and the Potential of Antibody Therapy
4.1. Murine Derived Monoclonal Antibodies
4.2. Human Derived Monoclonal Antibodies
4.3. Pan-Ebolavirus Monoclonal Antibodies
5. AAV-Mediated Monoclonal Antibody Expression for Filovirus Infections
5.1. AAV-Mediated Monoclonal Antibody Expression
5.2. Pre-Clinical Models of AAV-Mediated Monoclonal Antibody Expression and Protection against Filovirus Infections
5.3. Clinical Relevance
5.4. Benefits of the AAV VIP Approach
5.5. AAV VIP Challenges and Potential Solutions
6. Future Directions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- MacNeil, A.; Rollin, P.E. Ebola and Marburg hemorrhagic fevers: Neglected tropical diseases? PLoS. Negl. Trop. Dis. 2012, 6. [Google Scholar] [CrossRef] [Green Version]
- Martini, G.A.; Knauff, H.G.; Schmidt, H.A.; Mayer, G.; Baltzer, G. A hitherto unknown infectious disease contracted from monkeys. “Marburg-virus” disease. Ger. Med. Mon. 1968, 13, 457–470. [Google Scholar] [PubMed]
- Gear, J.S.; Cassel, G.A.; Gear, A.J.; Trappler, B.; Clausen, L.; Meyers, A.M.; Kew, M.C.; Bothwell, T.H.; Sher, R.; Miller, G.B.; et al. Outbreake of Marburg virus disease in Johannesburg. Br. Med. J. 1975, 4, 489–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Outbreaks Chronology: Marburg Hemorrhagic Fever. Available online: https://www.cdc.gov/vhf/marburg/outbreaks/chronology.html (accessed on 26 February 2020).
- Emond, R.T.; Evans, B.; Bowen, E.T.; Lloyd, G. A case of Ebola virus infection. Br. Med. J. 1977, 2, 541–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- New Ebola Outbreak Detected in Northwest Democratic Republic of the Congo; WHO Surge Team Supporting the Response. Available online: https://www.who.int/news-room/detail/01-06-2020-new-ebola-outbreak-detected-in-northwest-democratic-republic-of-the-congo-who-surge-team-supporting-the-response (accessed on 30 September 2020).
- Marí Saéz, A.; Weiss, S.; Nowak, K.; Lapeyre, V.; Zimmermann, F.; Düx, A.; Kühl, H.S.; Kaba, M.; Regnaut, S.; Merkel, K.; et al. Investigating the zoonotic origin of the West African Ebola epidemic. EMBO. Mol. Med. 2015, 7, 17–23. [Google Scholar] [CrossRef] [PubMed]
- 2014–2016 Ebola Outbreak in West Africa. Available online: https://www.cdc.gov/vhf/ebola/history/2014-2016-outbreak/index.html (accessed on 2 March 2020).
- Alexander, K.A.; Sanderson, C.E.; Marathe, M.; Lewis, B.L.; Rivers, C.M.; Shaman, J.; Drake, J.M.; Lofgren, E.; Dato, V.M.; Eisenberg, M.C.; et al. What factors might have led to the emergence of Ebola in West Africa? PLoS. Negl. Trop. Dis. 2015, 9. [Google Scholar] [CrossRef] [Green Version]
- Factors That Contributed to Undetected Spread of the Ebola Virus and Impeded Rapid Containment. Available online: http://www.who.int/entity/csr/disease/ebola/one-year-report/factors/en/index.html (accessed on 28 May 2020).
- Coltart, C.E.M.; Lindsey, B.; Ghinai, I.; Johnson, A.M.; Heymann, D.L. The Ebola outbreak, 2013–2016: Old lessons for new epidemics. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2017, 372, 20160297. [Google Scholar] [CrossRef]
- Wilson, H.W.; Amo-Addae, M.; Kenu, E.; Ilesanmi, O.S.; Ameme, D.K.; Sackey, S.O. Post-Ebola syndrome among Ebola virus disease survivors in Montserrado County, Liberia 2016. Biomed. Res. Int. 2018, 1909410. [Google Scholar] [CrossRef]
- Cost of the Ebola Epidemic. Available online: https://www.cdc.gov/vhf/ebola/history/2014-2016-outbreak/cost-of-ebola.html (accessed on 9 March 2020).
- Parpia, A.S.; Ndeffo-Mbah, M.L.; Wenzel, N.S.; Galvani, A.P. Effects of response to 2014–2015 Ebola outbreak on deaths from malaria, HIV/AIDS, and tuberculosis, West Africa. Emerg. Infect. Dis. 2016, 22, 433–441. [Google Scholar] [CrossRef]
- Mononegavirales. Available online: https://talk.ictvonline.org/ictv-reports/ictv_online_report/negative-sense-rna-viruses/mononegavirales/w/filoviridae (accessed on 29 May 2020).
- Hume, A.J.; Mühlberger, E. Distinct genome replication and transcription strategies within the growing filovirus family. J. Mol. Biol. 2019, 431, 4290–4320. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.-L.; Tan, C.W.; Anderson, D.E.; Jiang, R.-D.; Li, B.; Zhang, W.; Zhu, Y.; Lim, X.F.; Zhou, P.; Liu, X.-L.; et al. Characterization of a filovirus (Měnglà virus) from Rousettus bats in China. Nat. Microbiol. 2019, 4, 390–395. [Google Scholar] [CrossRef] [PubMed]
- Goldstein, T.; Anthony, S.J.; Gbakima, A.; Bird, B.H.; Bangura, J.; Tremeau-Bravard, A.; Belaganahalli, M.N.; Wells, H.L.; Dhanota, J.K.; Liang, E.; et al. The discovery of Bombali virus adds further support for bats as hosts of ebolaviruses. Nat. Microbiol. 2018, 3, 1084–1089. [Google Scholar] [CrossRef]
- Brauburger, K.; Hume, A.J.; Mühlberger, E.; Olejnik, J. Forty-five years of Marburg virus research. Viruses 2012, 4, 1878–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Negredo, A.; Palacios, G.; Vázquez-Morón, S.; González, F.; Dopazo, H.; Molero, F.; Juste, J.; Quetglas, J.; Savji, N.; de la Cruz Martínez, M.; et al. Discovery of an Ebolavirus-like Filovirus in Europe. PLoS. Pathog. 2011, 7, e1002304. [Google Scholar] [CrossRef] [Green Version]
- Languon, S.; Quaye, O. Filovirus disease outbreaks: A chronological overview. Virology 2019, 10. [Google Scholar] [CrossRef]
- Ebola Virus Disease. Available online: https://www.who.int/news-room/fact-sheets/detail/ebola-virus-disease (accessed on 12 May 2020).
- Marburg Virus Disease. Available online: https://www.who.int/news-room/fact-sheets/detail/marburg-virus-disease (accessed on 12 May 2020).
- Yuan, J.; Zhang, Y.; Li, J.; Zhang, Y.; Wang, L.-F.; Shi, Z. Serological evidence of ebolavirus infection in bats, China. Virol. J. 2012, 9, 236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Formenty, P.; Hatz, C.; Le Guenno, B.; Stoll, A.; Rogenmoser, P.; Widmer, A. Human infection due to Ebola virus, subtype Côte d’Ivoire: Clinical and biologic presentation. J. Infect. Dis. 1999, 179 (Suppl. 1), S48–S53. [Google Scholar] [CrossRef] [Green Version]
- Geisbert, T.W.; Jahrling, P.B. Differentiation of filoviruses by electron microscopy. Virus Res. 1995, 39, 129–150. [Google Scholar] [CrossRef] [Green Version]
- Emanuel, J.; Marzi, A.; Feldmann, H. Filoviruses: Ecology, molecular biology, and evolution. Adv. Virus Res. 2018, 100, 189–221. [Google Scholar]
- Mühlberger, E. Filovirus replication and transcription. Future Virol. 2007, 2, 205–215. [Google Scholar] [CrossRef] [Green Version]
- Neumann, G.; Watanabe, S.; Kawaoka, Y. Characterization of Ebolavirus regulatory Genomic regions. Virus Res. 2009, 144, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Hofmann-Winkler, H.; Kaup, F.; Pöhlmann, S. Host cell factors in Filovirus entry: Novel players, new insights. Viruses 2012, 4, 3336–3362. [Google Scholar] [CrossRef]
- Groseth, A.; Marzi, A.; Hoenen, T.; Herwig, A.; Gardner, D.; Becker, S.; Ebihara, H.; Feldmann, H. The Ebola virus glycoprotein contributes to but is not sufficient for virulence in vivo. PLoS. Pathog. 2012, 8. [Google Scholar] [CrossRef] [Green Version]
- Leroy, E.M.; Kumulungui, B.; Pourrut, X.; Rouquet, P.; Hassanin, A.; Yaba, P.; Délicat, A.; Paweska, J.T.; Gonzalez, J.-P.; Swanepoel, R. Fruit bats as reservoirs of Ebola virus. Nature 2005, 438, 575–576. [Google Scholar] [CrossRef]
- Vetter, P.; Fischer, W.A.; Schibler, M.; Jacobs, M.; Bausch, D.G.; Kaiser, L. Ebola virus shedding and transmission: Review of current evidence. J. Infect. Dis. 2016, 214, S177–S184. [Google Scholar] [CrossRef]
- Eichner, M.; Dowell, S.F.; Firese, N. Incubation period of Ebola hemorrhagic virus subtype Zaire. Osong. Public Health Res. Perspect. 2011, 2, 3–7. [Google Scholar] [CrossRef] [Green Version]
- Breman, J.G.; Heymann, D.L.; Lloyd, G.; McCormick, J.B.; Miatudila, M.; Murphy, F.A.; Muyembé-Tamfun, J.-J.; Piot, P.; Ruppol, J.-F.; Sureau, P.; et al. Discovery and description of Ebola Zaire virus in 1976 and relevance to the West African epidemic during 2013–2016. J. Infect. Dis. 2016, 214, S93–S101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bwaka, M.A.; Bonnet, M.J.; Calain, P.; Colebunders, R.; De Roo, A.; Guimard, Y.; Katwiki, K.R.; Kibadi, K.; Kipasa, M.A.; Kuvula, K.J.; et al. Ebola hemorrhagic fever in Kikwit, Democratic Republic of the Congo: Clinical observations in 103 patients. J. Infect. Dis. 1999, 179 (Suppl. 1), S1–S7. [Google Scholar] [CrossRef] [Green Version]
- Lekone, P.E.; Finkenstädt, B.F. Statistical inference in a stochastic epidemic SEIR model with control intervention: Ebola as a case study. Biometrics 2006, 62, 1170–1177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martini, G.A.; Siegert, R. Marburg Virus Disease; Springer: New York, NY, USA, 1971; pp. 166–176. [Google Scholar]
- Slenczka, W.G. The Marburg virus outbreak of 1967 and subsequent episodes. Curr. Top. Microbiol. Immunol. 1999, 235, 49–75. [Google Scholar] [CrossRef]
- Van Kerkhove, M.D.; Bento, A.I.; Mills, H.L.; Ferguson, N.M.; Donnelly, C.A. A review of epidemiological parameters from Ebola outbreaks to inform early public health decision-making. Sci. Data 2015, 2, 150019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoenen, T.; Groseth, A.; Falzarano, D.; Feldmann, H. Ebola virus: Unravelling pathogenesis to combat a deadly disease. Trends. Mol. Med. 2006, 12, 206–215. [Google Scholar] [CrossRef]
- Geisbert, T.W.; Jaax, N.K. Marburg hemorrhagic fever: Report of a case studied by immunohistochemistry and electron microscopy. Ultrastruct. Pathol. 1998, 22, 3–17. [Google Scholar] [CrossRef]
- Mehedi, M.; Groseth, A.; Feldmann, H.; Ebihara, H. Clinical aspects of Marburg hemorrhagic fever. Future Virol. 2011, 6, 1091–1106. [Google Scholar] [CrossRef] [Green Version]
- Gautier, E.L.; Jakubzick, C.; Randolph, G.J. Regulation of the migration and survival of monocyte subsets by chemokine receptors and its relevance to atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1412–1418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morris, D.L.; Singer, K.; Lumeng, C.N. Adipose tissue macrophages: Phenotypic plasticity and diversity in lean and obese states. Curr. Opin. Clin. Nutr. Metab. Care 2011, 14, 341–346. [Google Scholar] [CrossRef] [Green Version]
- Robinson, M.J.; Sancho, D.; Slack, E.C.; LeibundGut-Landmann, S.; Sousa, C.R.E. Myeloid C-type lectins in innate immunity. Nat. Immunol. 2006, 7, 1258–1265. [Google Scholar] [CrossRef]
- Shabo, I.; Svanvik, J. Expression of macrophage antigens by tumor cells. Adv. Exp. Med. Biol. 2011, 714, 141–150. [Google Scholar] [CrossRef]
- Bosio, C.M.; Aman, M.J.; Grogan, C.; Hogan, R.; Ruthel, G.; Negley, D.; Mohamadzadeh, M.; Bavari, S.; Schmaljohn, A. Ebola and Marburg viruses replicate in monocyte-derived dendritic cells without inducing the production of cytokines and full maturation. J. Infect. Dis. 2003, 188, 1630–1638. [Google Scholar] [CrossRef] [Green Version]
- Lubaki, N.M.; Ilinykh, P.; Pietzsch, C.; Tigabu, B.; Freiberg, A.N.; Koup, R.A.; Bukreyev, A. The lack of maturation of Ebola virus-infected dendritic cells results from the cooperative effect of at least two viral domains. J. Virol. 2013, 87, 7471–7485. [Google Scholar] [CrossRef] [Green Version]
- Geisbert, T.W.; Hensley, L.E.; Gibb, T.R.; Steele, K.E.; Jaax, N.K.; Jahrling, P.B. Apoptosis induced in vitro and in vivo during infection by Ebola and Marburg viruses. Lab. Invest. 2000, 80, 171–186. [Google Scholar] [CrossRef] [Green Version]
- Bradfute, S.B.; Braun, D.R.; Shamblin, J.D.; Geisbert, J.B.; Paragas, J.; Garrison, A.; Hensley, L.E.; Geisbert, T.W. Lymphocyte death in a mouse model of Ebola virus infection. J. Infect. Dis. 2007, 196, S296–S304. [Google Scholar] [CrossRef] [PubMed]
- Gupta, M.; Spiropoulou, C.; Rollin, P.E. Ebola virus infection of human PBMCs causes massive death of macrophages, CD4 and CD8 T cell sub-populations in vitro. Virology 2007, 364, 45–54. [Google Scholar] [CrossRef] [Green Version]
- Baize, S.; Leroy, E.M.; Georges-Courbot, M.C.; Capron, M.; Lansoud-Soukate, J.; Debré, P.; Fisher-Hoch, S.P.; McCormick, J.B.; Georges, A.J. Defective humoral responses and extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-infected patients. Nat. Med. 1999, 5, 423–426. [Google Scholar] [CrossRef]
- McElroy, A.K.; Akondy, R.S.; Davis, C.W.; Ellebedy, A.H.; Mehta, A.K.; Kraft, C.S.; Lyon, G.M.; Ribner, B.S.; Varkey, J.; Sidney, J.; et al. Human Ebola virus infection results in substantial immune activation. Proc. Natl. Acad. Sci. USA 2015, 112, 4719–4724. [Google Scholar] [CrossRef] [Green Version]
- Schindell, B.G.; Webb, A.L.; Kindrachuk, J. Persistence and sexual transmission of Filoviruses. Viruses 2018, 10, 683. [Google Scholar] [CrossRef] [Green Version]
- Lupton, H.W.; Lambert, R.D.; Bumgardner, D.L.; Moe, J.B.; Eddy, G.A. Inactivated vaccine for Ebola virus efficacious in guineapig model. Lancet 1980, 2, 1294–1295. [Google Scholar] [CrossRef]
- Reynolds, P.; Marzi, A. Ebola and Marburg virus vaccines. Virus Genes. 2017, 53, 501–515. [Google Scholar] [CrossRef] [PubMed]
- Heald, A.E.; Charleston, J.S.; Iversen, P.L.; Warren, T.K.; Saoud, J.B.; Al-Ibrahim, M.; Wells, J.; Warfield, K.L.; Swenson, D.L.; Welch, L.S.; et al. AVI-7288 for Marburg virus in nonhuman primates and humans. N. Engl. J. Med. 2015, 373, 339–348. [Google Scholar] [CrossRef]
- Sarwar, U.N.; Costner, P.; Enama, M.E.; Berkowitz, N.; Hu, Z.; Hendel, C.S.; Sitar, S.; Plummer, S.; Mulangu, S.; Bailer, R.T.; et al. Safety and immunogenicity of DNA vaccines encoding Ebolavirus and Marburgvirus wild-type glycoproteins in a phase I clinical trial. J. Infect. Dis. 2015, 211, 549–557. [Google Scholar] [CrossRef] [PubMed]
- Kibuuka, H.; Berkowitz, N.M.; Millard, M.; Enama, M.E.; Tindikahwa, A.; Sekiziyivu, A.B.; Costner, P.; Sitar, S.; Glover, D.; Hu, Z.; et al. Safety and immunogenicity of Ebola virus and Marburg virus glycoprotein DNA vaccines assessed separately and concomitantly in healthy Ugandan adults: A phase 1b, randomised, double-blind, placebo-controlled clinical trial. Lancet 2015, 385, 1545–1554. [Google Scholar] [CrossRef]
- Henao-Restrepo, A.M.; Camacho, A.; Longini, I.M.; Watson, C.H.; Edmunds, W.J.; Egger, M.; Carroll, M.W.; Dean, N.E.; Diatta, I.; Doumbia, M.; et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: Final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Lancet 2017, 389, 505–518. [Google Scholar] [CrossRef] [Green Version]
- Mupapa, K.; Massamba, M.; Kibadi, K.; Kuvula, K.; Bwaka, A.; Kipasa, M.; Colebunders, R.; Muyembe-Tamfum, J.J. Treatment of Ebola hemorrhagic fever with blood transfusions from convalescent patients. J. Infect. Dis. 1999, 179, S18–S23. [Google Scholar] [CrossRef] [Green Version]
- Siragam, V.; Wong, G.; Qiu, X.-G. Animal models for filovirus infections. Zool. Res. 2018, 39, 15–24. [Google Scholar] [CrossRef]
- Kozak, R.; He, S.; Kroeker, A.; de La Vega, M.-A.; Audet, J.; Wong, G.; Urfano, C.; Antonation, K.; Embury-Hyatt, C.; Kobinger, G.P.; et al. Ferrets infected with Bundibugyo virus or Ebola virus recapitulate important aspects of human Filovirus disease. J. Virol. 2016, 90, 9209–9223. [Google Scholar] [CrossRef] [Green Version]
- Gibb, T.R.; Bray, M.; Geisbert, T.W.; Steele, K.E.; Kell, W.M.; Davis, K.J.; Jaax, N.K. Pathogenesis of experimental Ebola Zaire virus infection in BALB/c mice. J. Comp. Pathol. 2001, 125, 233–242. [Google Scholar] [CrossRef] [PubMed]
- Qiu, X.; Fernando, L.; Melito, P.L.; Audet, J.; Feldmann, H.; Kobinger, G.; Alimonti, J.B.; Jones, S.M. Ebola GP-specific monoclonal antibodies protect mice and guinea pigs from lethal Ebola virus infection. PLoS. Negl. Trop. Dis. 2012, 6, e1575. [Google Scholar] [CrossRef]
- Pascal, K.E.; Dudgeon, D.; Trefry, J.C.; Anantpadma, M.; Sakurai, Y.; Murin, C.D.; Turner, H.L.; Fairhurst, J.; Torres, M.; Rafique, A.; et al. Development of clinical-stage human monoclonal antibodies that treat advanced Ebola virus disease in nonhuman primates. J. Infect. Dis. 2018, 218, S612–S626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banadyga, L.; Wong, G.; Qiu, X. Small animal models for evaluating Filovirus countermeasures. ACS. Infect. Dis. 2018, 4, 673–685. [Google Scholar] [CrossRef] [PubMed]
- Dye, J.M.; Herbert, A.S.; Kuehne, A.I.; Barth, J.F.; Muhammad, M.A.; Zak, S.E.; Ortiz, R.A.; Prugar, L.I.; Pratt, W.D. Postexposure antibody prophylaxis protects nonhuman primates from filovirus disease. Proc. Natl. Acad. Sci. USA 2012, 109, 5034–5039. [Google Scholar] [CrossRef] [Green Version]
- Marzi, A.; Yoshida, R.; Miyamoto, H.; Ishijima, M.; Suzuki, Y.; Higuchi, M.; Matsuyama, Y.; Igarashi, M.; Nakayama, E.; Kuroda, M.; et al. Protective efficacy of neutralizing monoclonal antibodies in a nonhuman primate model of Ebola hemorrhagic fever. PLoS ONE 2012, 7, e36192. [Google Scholar] [CrossRef] [Green Version]
- Olinger, G.G.; Pettitt, J.; Kim, D.; Working, C.; Bohorov, O.; Bratcher, B.; Hiatt, E.; Hume, S.D.; Johnson, A.K.; Morton, J.; et al. Delayed treatment of Ebola virus infection with plant-derived monoclonal antibodies provides protection in rhesus macaques. Proc. Natl. Acad. Sci. USA 2012, 109, 18030–18035. [Google Scholar] [CrossRef] [Green Version]
- Pettitt, J.; Zeitlin, L.; Kim, D.H.; Working, C.; Johnson, J.C.; Bohorov, O.; Bratcher, B.; Hiatt, E.; Hume, S.D.; Johnson, A.K.; et al. Therapeutic intervention of Ebola virus infection in rhesus macaques with the MB-003 monoclonal antibody cocktail. Sci. Transl. Med. 2013, 5, 199ra113. [Google Scholar] [CrossRef] [Green Version]
- Marzi, A.; Engelmann, F.; Feldmann, F.; Haberthur, K.; Shupert, W.L.; Brining, D.; Scott, D.P.; Geisbert, T.W.; Kawaoka, Y.; Katze, M.G.; et al. Antibodies are necessary for rVSV/ZEBOV-GP–mediated protection against lethal Ebola virus challenge in nonhuman primates. Proc. Natl. Acad. Sci. USA 2013, 110, 1893–1898. [Google Scholar] [CrossRef] [Green Version]
- Wong, G.; Richardson, J.S.; Pillet, S.; Patel, A.; Qiu, X.; Alimonti, J.; Hogan, J.; Zhang, Y.; Takada, A.; Feldmann, H.; et al. Immune parameters correlate with protection against Ebola virus infection in rodents and nonhuman primates. Sci. Transl. Med. 2012, 4, 158ra146. [Google Scholar] [CrossRef] [Green Version]
- Qiu, X.; Alimonti, J.B.; Melito, P.L.; Fernando, L.; Ströher, U.; Jones, S.M. Characterization of Zaire ebolavirus glycoprotein-specific monoclonal antibodies. J. Clin. Immunol. 2011, 141, 218–227. [Google Scholar] [CrossRef]
- Martinez, O.; Tantral, L.; Mulherkar, N.; Chandran, K.; Basler, C.F. Impact of Ebola mucin-like domain on antiglycoprotein antibody responses induced by Ebola virus-like particles. J. Infect. Dis. 2011, 204, S825–S832. [Google Scholar] [CrossRef] [Green Version]
- Murin, C.D.; Fusco, M.L.; Bornholdt, Z.A.; Qiu, X.; Olinger, G.G.; Zeitlin, L.; Kobinger, G.P.; Ward, A.B.; Saphire, E.O. Structures of protective antibodies reveal sites of vulnerability on Ebola virus. Proc. Natl. Acad. Sci. USA 2014, 111, 17182–17187. [Google Scholar] [CrossRef] [Green Version]
- Qiu, X.; Audet, J.; Wong, G.; Pillet, S.; Bello, A.; Cabral, T.; Strong, J.E.; Plummer, F.; Corbett, C.R.; Alimonti, J.B.; et al. Successful treatment of ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci. Transl. Med. 2012, 4, 138ra81. [Google Scholar] [CrossRef] [Green Version]
- Davison, E.; Bryan, C.; Fong, R.H.; Barnes, T.; Pfaff, J.M.; Mabila, M.; Rucker, J.B.; Doranz, B.J. Mechanism of binding to Ebola virus glycoprotein by the ZMapp, ZMAb, and MB-003 cocktail antibodies. J. Virol. 2015, 89, 10982–10992. [Google Scholar] [CrossRef] [Green Version]
- Budzianowski, J. Tobacco against Ebola virus disease. Prz. Lek. 2015, 72, 567–571. [Google Scholar]
- Wilson, J.A.; Hevey, M.; Bakken, R.; Guest, S.; Bray, M.; Schmaljohn, A.L.; Hart, M.K. Epitopes involved in antibody-mediated protection from Ebola Virus. Science 2000, 287, 1664–1666. [Google Scholar] [CrossRef]
- Qiu, X.; Wong, G.; Audet, J.; Bello, A.; Fernando, L.; Alimonti, J.B.; Fausther-Bovendo, H.; Wei, H.; Aviles, J.; Hiatt, E.; et al. Reversion of advanced Ebola virus disease in nonhuman primates with ZMappTM. Nature 2014, 514, 47–53. [Google Scholar] [CrossRef] [Green Version]
- Group, The PREVAIL II Writing Group for the Multi-National PREVAIL II Study Team. A Randomized, Controlled Trial of ZMapp for Ebola Virus Infection. New Engl. J. Med. 2016, 375, 1448–1456. [Google Scholar] [CrossRef] [PubMed]
- LoBuglio, A.F.; Wheeler, R.H.; Trang, J.; Haynes, A.; Rogers, K.; Harvey, E.B.; Sun, L.; Ghrayeb, J.; Khazaeli, M.B. Mouse/human chimeric monoclonal antibody in man: Kinetics and immune response. Proc. Natl. Acad. Sci. USA 1989, 86, 4220–4224. [Google Scholar] [CrossRef] [Green Version]
- Lu, R.-M.; Hwang, Y.-C.; Liu, I.-J.; Lee, C.-C.; Tsai, H.-Z.; Li, H.-J.; Wu, H.-C. Development of therapeutic antibodies for the treatment of diseases. J. Biomed. Sci. 2020, 27, 1. [Google Scholar] [CrossRef] [PubMed]
- Harding, F.A.; Stickler, M.M.; Razo, J.; DuBridge, R.B. The immunogenicity of humanized and fully human antibodies. MAbs 2010, 2, 256–265. [Google Scholar] [CrossRef] [Green Version]
- Hwang, W.Y.K.; Foote, J. Immunogenicity of engineered antibodies. Methods 2005, 36, 3–10. [Google Scholar] [CrossRef]
- Weiner, L.M.; Surana, R.; Wang, S. Monoclonal antibodies: Versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010, 10, 317–327. [Google Scholar] [CrossRef] [Green Version]
- Bruhns, P.; Jönsson, F. Mouse and human FcR effector functions. Immunol. Rev. 2015, 268, 25–51. [Google Scholar] [CrossRef] [Green Version]
- Corti, D.; Misasi, J.; Mulangu, S.; Stanley, D.A.; Kanekiyo, M.; Wollen, S.; Ploquin, A.; Doria-Rose, N.A.; Staupe, R.P.; Bailey, M.; et al. Protective monotherapy against lethal Ebola virus infection by a potently neutralizing antibody. Science 2016, 351, 1339–1342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gilchuk, P.; Mire, C.E.; Geisbert, J.B.; Agans, K.N.; Deer, D.J.; Cross, R.W.; Slaughter, J.C.; Flyak, A.I.; Mani, J.; Pauly, M.H.; et al. Efficacy of human monoclonal antibody monotherapy against Bundibugyo virus infection in nonhuman primates. J. Infect. Dis. 2018, 218, S565–S573. [Google Scholar] [CrossRef] [Green Version]
- Muyembe-Tamfum, J.J.; Kipasa, M.; Kiyungu, C.; Colebunders, R. Ebola outbreak in Kikwit, Democratic Republic of the Congo: Discovery and control measures. J. Infect. Dis. 1999, 179 (Suppl. 1), S259–S262. [Google Scholar] [CrossRef] [Green Version]
- Gaudinski, M.R.; Coates, E.E.; Novik, L.; Widge, A.; Houser, K.V.; Burch, E.; Holman, L.A.; Gordon, I.J.; Chen, G.L.; Carter, C.; et al. Safety, tolerability, pharmacokinetics, and immunogenicity of mAb114: A phase 1 trial of a therapeutic monoclonal antibody targeting Ebola virus glycoprotein. Lancet 2019, 393, 889–898. [Google Scholar] [CrossRef] [Green Version]
- Mire, C.E.; Geisbert, J.B.; Borisevich, V.; Fenton, K.A.; Agans, K.N.; Flyak, A.I.; Deer, D.J.; Steinkellner, H.; Bohorov, O.; Bohorova, N.; et al. Therapeutic treatment of Marburg and Ravn virus infection in nonhuman primates with a human monoclonal antibody. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [Green Version]
- Flyak, A.I.; Ilinykh, P.A.; Murin, C.D.; Garron, T.; Shen, X.; Fusco, M.L.; Hashiguchi, T.; Bornholdt, Z.A.; Slaughter, J.C.; Sapparapu, G.; et al. Mechanism of human antibody-mediated neutralization of Marburg virus. Cell 2015, 160, 893–903. [Google Scholar] [CrossRef] [Green Version]
- Ledgerwood, J.E.; DeZure, A.D.; Stanley, D.A.; Coates, E.E.; Novik, L.; Enama, M.E.; Berkowitz, N.M.; Hu, Z.; Joshi, G.; Ploquin, A.; et al. Chimpanzee adenovirus vector Ebola vaccine. N. Engl. J. Med. 2017, 376, 928–938. [Google Scholar] [CrossRef]
- Fusco, M.L.; Hashiguchi, T.; Cassan, R.; Biggins, J.E.; Murin, C.D.; Warfield, K.L.; Li, S.; Holtsberg, F.W.; Shulenin, S.; Vu, H.; et al. Protective mAbs and cross-reactive mAbs raised by immunization with engineered Marburg virus GPs. PLoS. Pathog. 2015, 11, e1005016. [Google Scholar] [CrossRef]
- Brannan, J.M.; He, S.; Howell, K.A.; Prugar, L.I.; Zhu, W.; Vu, H.; Shulenin, S.; Kailasan, S.; Raina, H.; Wong, G.; et al. Post-exposure immunotherapy for two ebolaviruses and Marburg virus in nonhuman primates. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
- Holtsberg, F.W.; Shulenin, S.; Vu, H.; Howell, K.A.; Patel, S.J.; Gunn, B.; Karim, M.; Lai, J.R.; Frei, J.C.; Nyakatura, E.K.; et al. Pan-ebolavirus and pan-filovirus mouse monoclonal antibodies: Protection against Ebola and Sudan viruses. J. Virol. 2016, 90, 266–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, X.; Howell, K.A.; He, S.; Brannan, J.M.; Wec, A.Z.; Davidson, E.; Turner, H.L.; Chiang, C.-I.; Lei, L.; Fels, J.M.; et al. Immunization-elicited broadly protective antibody reveals Ebolavirus fusion loop as a site of vulnerability. Cell 2017, 169, 891–904. [Google Scholar] [CrossRef]
- Howell, K.A.; Qiu, X.; Brannan, J.M.; Bryan, C.; Davidson, E.; Holtsberg, F.W.; Wec, A.Z.; Shulenin, S.; Biggins, J.E.; Douglas, R.; et al. Antibody treatment of Ebola and Sudan virus infection via a uniquely exposed epitope within the glycoprotein receptor-binding site. Cell. Rep. 2016, 15, 1514–1526. [Google Scholar] [CrossRef] [Green Version]
- Bornholdt, Z.A.; Turner, H.L.; Murin, C.D.; Li, W.; Sok, D.; Souders, C.A.; Piper, A.E.; Goff, A.; Shamblin, J.D.; Wollen, S.E.; et al. Isolation of potent neutralizing antibodies from a survivor of the 2014 Ebola virus outbreak. Science 2016, 351, 1078–1083. [Google Scholar] [CrossRef] [Green Version]
- Wec, A.Z.; Herbert, A.S.; Murin, C.D.; Nyakatura, E.K.; Abelson, D.M.; Fels, J.M.; He, S.; James, R.M.; de La Vega, M.-A.; Zhu, W.; et al. Antibodies from a human survivor define sites of vulnerability for broad protection against Ebolaviruses. Cell 2017, 169, 878–890. [Google Scholar] [CrossRef]
- Flyak, A.I.; Shen, X.; Murin, C.D.; Turner, H.L.; David, J.A.; Fusco, M.L.; Lampley, R.; Kose, N.; Ilinykh, P.A.; Kuzmina, N.; et al. Cross-reactive and potent neutralizing antibody responses in human survivors of natural Ebola virus infection. Cell 2016, 164, 392–405. [Google Scholar] [CrossRef] [Green Version]
- King, L.B.; West, B.R.; Moyer, C.L.; Gilchuk, P.; Flyak, A.; Ilinykh, P.A.; Bombardi, R.; Hui, S.; Huang, K.; Bukreyev, A.; et al. Cross-reactive neutralizing human survivor monoclonal antibody BDBV223 targets the ebolavirus stalk. Nat. Commun. 2019, 10, 1788. [Google Scholar] [CrossRef] [Green Version]
- Flyak, A.I.; Kuzmina, N.; Murin, C.D.; Bryan, C.; Davidson, E.; Gilchuk, P.; Gulka, C.P.; Ilinykh, P.A.; Shen, X.; Huang, K.; et al. Broadly neutralizing antibodies from human survivors target a conserved site in the Ebola virus glycoprotein HR2/MPER region. Nat. Microbiol. 2018, 3, 670–677. [Google Scholar] [CrossRef] [Green Version]
- Keck, Z.-Y.; Enterlein, S.G.; Howell, K.A.; Vu, H.; Shulenin, S.; Warfield, K.L.; Froude, J.W.; Araghi, N.; Douglas, R.; Biggins, J.; et al. Macaque monoclonal antibodies targeting novel conserved epitopes within Filovirus glycoprotein. J. Virol. 2015, 90, 279–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wec, A.Z.; Bornholdt, Z.A.; He, S.; Herbert, A.S.; Goodwin, E.; Wirchnianski, A.S.; Gunn, B.M.; Zhang, Z.; Zhu, W.; Liu, G.; et al. Development of a human antibody cocktail that deploys multiple functions to confer pan-Ebolavirus protection. Cell Host Microbe. 2019, 25, 39–48. [Google Scholar] [CrossRef] [Green Version]
- Russell, S.; Bennett, J.; Wellman, J.A.; Chung, D.C.; Yu, Z.-F.; Tillman, A.; Wittes, J.; Pappas, J.; Elci, O.; McCague, S.; et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: A randomised, controlled, open-label, phase 3 trial. Lancet 2017, 390, 849–860. [Google Scholar] [CrossRef]
- Mendell, J.R.; Al-Zaidy, S.; Shell, R.; Arnold, W.D.; Rodino-Klapac, L.R.; Prior, T.W.; Lowes, L.; Alfano, L.; Berry, K.; Church, K.; et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N. Engl. J. Med. 2017, 377, 1713–1722. [Google Scholar] [CrossRef]
- Daya, S.; Berns, K.I. Gene therapy using adeno-associated virus vectors. Clin. Microbiol. Rev. 2008, 21, 583–593. [Google Scholar] [CrossRef] [Green Version]
- Penaud-Budloo, M.; Le Guiner, C.; Nowrouzi, A.; Toromanoff, A.; Chérel, Y.; Chenuaud, P.; Schmidt, M.; von Kalle, C.; Rolling, F.; Moullier, P.; et al. Adeno-associated virus vector genomes persist as episomal chromatin in primate muscle. J. Virol. 2008, 82, 7875–7885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toromanoff, A.; Chérel, Y.; Guilbaud, M.; Penaud-Budloo, M.; Snyder, R.O.; Haskins, M.E.; Deschamps, J.-Y.; Guigand, L.; Podevin, G.; Arruda, V.R.; et al. Safety and efficacy of regional intravenous (RI) versus intramuscular (IM) delivery of rAAV1 and rAAV8 to nonhuman primate skeletal muscle. Mol. Ther. 2008, 16, 1291–1299. [Google Scholar] [CrossRef]
- Balazs, A.B.; Chen, J.; Hong, C.M.; Rao, D.S.; Yang, L.; Baltimore, D. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature 2011, 481, 81–84. [Google Scholar] [CrossRef] [Green Version]
- Chng, J.; Wang, T.; Nian, R.; Lau, A.; Hoi, K.M.; Ho, S.C.; Gagnon, P.; Bi, X.; Yang, Y. Cleavage efficient 2A peptides for high level monoclonal antibody expression in CHO cells. MAbs 2015, 7, 403–412. [Google Scholar] [CrossRef] [Green Version]
- Van Lieshout, L.P.; Soule, G.; Sorensen, D.; Frost, K.L.; He, S.; Tierney, K.; Safronetz, D.; Booth, S.A.; Kobinger, G.P.; Qiu, X.; et al. Intramuscular adeno-associated virus–mediated expression of monoclonal antibodies provides 100% protection against Ebola virus infection in mice. J. Infect. Dis. 2018, 217, 916–925. [Google Scholar] [CrossRef]
- Balazs, A.B.; Ouyang, Y.; Hong, C.M.; Chen, J.; Nguyen, S.M.; Rao, D.S.; An, D.S.; Baltimore, D. Vectored immunoprophylaxis protects humanized mice from mucosal HIV transmission. Nat. Med. 2014, 20, 296–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnson, P.R.; Schnepp, B.C.; Zhang, J.; Connell, M.J.; Greene, S.M.; Yuste, E.; Desrosiers, R.C.; Clark, K.R. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat. Med. 2009, 15, 901–906. [Google Scholar] [CrossRef] [Green Version]
- Martinez-Navio, J.M.; Fuchs, S.P.; Pantry, S.N.; Lauer, W.A.; Duggan, N.N.; Keele, B.F.; Rakasz, E.G.; Gao, G.; Lifson, J.D.; Desrosiers, R.C. Adeno-associated virus delivery of anti-HIV monoclonal antibodies can drive long-term virologic suppression. Immunity 2019, 50, 567–575. [Google Scholar] [CrossRef] [Green Version]
- Fuchs, S.P.; Martinez-Navio, J.M.; Piatak , M., Jr.; Lifson, J.D.; Gao, G.; Desrosiers, R.C. AAV-Delivered antibody mediates significant protective effects against SIVmac239 challenge in the absence of neutralizing activity. PLoS. Pathog. 2015, 11, e1005090. [Google Scholar] [CrossRef] [PubMed]
- Balazs, A.B.; Bloom, J.D.; Hong, C.M.; Rao, D.S.; Baltimore, D. Broad protection against influenza infection by vectored immunoprophylaxis in mice. Nat. Biotechnol. 2013, 31, 647–652. [Google Scholar] [CrossRef]
- Deal, C.; Balazs, A.B.; Espinosa, D.A.; Zavala, F.; Baltimore, D.; Ketner, G. Vectored antibody gene delivery protects against Plasmodium falciparum sporozoite challenge in mice. Proc. Natl. Acad. Sci. USA 2014, 111, 12528–12532. [Google Scholar] [CrossRef] [Green Version]
- De Jong, Y.P.; Dorner, M.; Mommersteeg, M.C.; Xiao, J.W.; Balazs, A.B.; Robbins, J.B.; Winer, B.Y.; Gerges, S.; Vega, K.; Labitt, R.N.; et al. Broadly neutralizing antibodies abrogate established hepatitis C virus infection. Sci. Transl. Med. 2014, 6, 254ra129. [Google Scholar] [CrossRef] [Green Version]
- Priddy, F.H.; Lewis, D.J.M.; Gelderblom, H.C.; Hassanin, H.; Streatfield, C.; LaBranche, C.; Hare, J.; Cox, J.H.; Dally, L.; Bendel, D.; et al. Adeno-associated virus vectored immunoprophylaxis to prevent HIV in healthy adults: A phase 1 randomised controlled trial. Lancet HIV 2019, 6, e230–e239. [Google Scholar] [CrossRef] [Green Version]
- Lewis, A.D.; Chen, R.; Montefiori, D.C.; Johnson, P.R.; Clark, K.R. Generation of neutralizing activity against human immunodeficiency virus type 1 in serum by antibody gene transfer. J. Virol. 2002, 76, 8769–8775. [Google Scholar] [CrossRef] [Green Version]
- Limberis, M.P.; Adam, V.S.; Wong, G.; Gren, J.; Kobasa, D.; Ross, T.M.; Kobinger, G.P.; Tretiakova, A.; Wilson, J.M. Intranasal antibody gene transfer in mice and ferrets elicits broad protection against pandemic influenza. Sci. Transl. Med. 2013, 5, 187ra72. [Google Scholar] [CrossRef] [Green Version]
- Limberis, M.P.; Tretiakova, A.; Nambiar, K.; Wong, G.; Racine, T.; Crosariol, M.; Xiangguo, Q.; Kobinger, G.; Wilson, J.M. Adeno-associated virus serotype 9-expressed ZMapp in mice confers protection against systemic and airway-acquired Ebola virus infection. J. Infect. Dis. 2016, 214, 1975–1979. [Google Scholar] [CrossRef]
- Robert, M.-A.; Nassoury, N.; Chahal, P.S.; Venne, M.-H.; Racine, T.; Qiu, X.; Kobinger, G.; Kamen, A.; Gilbert, R.; Gaillet, B. Gene transfer of ZMapp antibodies mediated by recombinant adeno-associated virus protects against Ebola infections. Hum. Gene Ther. 2017, 29, 452–466. [Google Scholar] [CrossRef]
- Flotte, T.R.; Trapnell, B.C.; Humphries, M.; Carey, B.; Calcedo, R.; Rouhani, F.; Campbell-Thompson, M.; Yachnis, A.T.; Sandhaus, R.A.; McElvaney, N.G.; et al. Phase 2 clinical trial of a recombinant adeno-associated viral vector expressing α1-antitrypsin: Interim results. Hum. Gene Ther. 2011, 22, 1239–1247. [Google Scholar] [CrossRef] [Green Version]
- Weber, A.; Engelmaier, A.; Voelkel, D.; Pachlinger, R.; Scheiflinger, F.; Monahan, P.E.; Rottensteiner, H. Development of methods for the selective measurement of the single amino acid exchange variant coagulation factor IX Padua. Mol. Ther. Methods Clin. Dev. 2018, 10, 29–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miesbach, W.; Meijer, K.; Coppens, M.; Kampmann, P.; Klamroth, R.; Schutgens, R.; Tangelder, M.; Castaman, G.; Schwäble, J.; Bonig, H.; et al. Gene therapy with adeno-associated virus vector 5–human factor IX in adults with hemophilia B. Blood 2018, 131, 1022–1031. [Google Scholar] [CrossRef]
- Martinez-Navio, J.M.; Fuchs, S.P.; Mendes, D.E.; Rakasz, E.G.; Gao, G.; Lifson, J.D.; Desrosiers, R.C. Long-term delivery of an anti-SIV monoclonal antibody with AAV. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
- Van Lieshout, L.P.; Domm, J.M.; Rindler, T.N.; Frost, K.L.; Sorensen, D.L.; Medina, S.J.; Booth, S.A.; Bridges, J.P.; Wootton, S.K. A novel triple-mutant AAV6 capsid induces rapid and potent transgene expression in the muscle and respiratory tract of mice. Mol. Ther. Methods Clin. Dev. 2018, 9, 323–329. [Google Scholar] [CrossRef] [Green Version]
- Croyle, M.A.; Cheng, X.; Wilson, J.M. Development of formulations that enhance physical stability of viral vectors for gene therapy. Gene Ther. 2001, 8, 1281–1290. [Google Scholar] [CrossRef] [Green Version]
- Martinez-Navio, J.M.; Fuchs, S.P.; Pedreño-López, S.; Rakasz, E.G.; Gao, G.; Desrosiers, R.C. Host anti-antibody responses following adeno-associated virus-mediated delivery of antibodies against HIV and SIV in rhesus monkeys. Mol. Ther. 2016, 24, 76–86. [Google Scholar] [CrossRef] [Green Version]
- Saunders, K.O.; Wang, L.; Joyce, M.G.; Yang, Z.Y.; Balazs, A.B.; Cheng, C.; Ko, S.-Y.; Kong, W.-P.; Rudicell, R.S.; Georgiev, I.S.; et al. Broadly neutralizing human immunodeficiency virus type 1 antibody gene transfer protects nonhuman primates from mucosal simian-human immunodeficiency virus infection. J. Virol. 2015, 89, 8334–8345. [Google Scholar] [CrossRef] [Green Version]
- Scharf, L.; West, A.P.; Sievers, S.A.; Chen, C.; Jiang, S.; Gao, H.; Gray, M.D.; McGuire, A.T.; Scheid, J.F.; Nussenzweig, M.C.; et al. Structural basis for germline antibody recognition of HIV-1 immunogens. eLife 2016, 5. [Google Scholar] [CrossRef] [Green Version]
- Wu, X.; Zhou, T.; Zhu, J.; Zhang, B.; Georgiev, I.; Wang, C.; Chen, X.; Longo, N.S.; Louder, M.; McKee, K.; et al. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 2011, 333, 1593–1602. [Google Scholar] [CrossRef] [Green Version]
- Kwong, P.D.; Mascola, J.R. Human antibodies that neutralize HIV-1: Identification, structures, and B cell ontogenies. Immunity 2012, 37, 412–425. [Google Scholar] [CrossRef] [Green Version]
- Moody, M.A.; Zhang, R.; Walter, E.B.; Woods, C.W.; Ginsburg, G.S.; McClain, M.T.; Denny, T.N.; Chen, X.; Munshaw, S.; Marshall, D.J.; et al. H3N2 influenza infection elicits more cross-reactive and less clonally expanded anti-hemagglutinin antibodies than influenza vaccination. PLoS ONE 2011, 6, e25797. [Google Scholar] [CrossRef]
- Kalinke, U.; Bucher, E.M.; Ernst, B.; Oxenius, A.; Roost, H.P.; Geley, S.; Kofler, R.; Zinkernagel, R.M.; Hengartner, H. The role of somatic mutation in the generation of the protective humoral immune response against vesicular stomatitis virus. Immunity 1996, 5, 639–652. [Google Scholar] [CrossRef] [Green Version]
- Walker, L.M.; Phogat, S.K.; Chan-Hui, P.-Y.; Wagner, D.; Phung, P.; Goss, J.L.; Wrin, T.; Simek, M.D.; Fling, S.; Mitcham, J.L.; et al. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 2009, 326, 285–289. [Google Scholar] [CrossRef] [Green Version]
- Shcherbakov, D.N.; Bakulina, A.Y.; Karpenko, L.I.; Ilyichev, A.A. Broadly neutralizing antibodies against HIV-1 as a novel aspect of the immune response. Acta Nat. 2015, 7, 11–21. [Google Scholar] [CrossRef]
- Huang, J.; Ofek, G.; Laub, L.; Louder, M.K.; Doria-Rose, N.A.; Longo, N.S.; Imamichi, H.; Bailer, R.T.; Chakrabarti, B.; Sharma, S.K.; et al. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 2012, 491, 406–412. [Google Scholar] [CrossRef] [Green Version]
- Gardner, M.R.; Fetzer, I.; Kattenhorn, L.M.; Davis-Gardner, M.E.; Zhou, A.S.; Alfant, B.; Weber, J.A.; Kondur, H.R.; Martinez-Navio, J.M.; Fuchs, S.P.; et al. Anti-drug antibody responses impair prophylaxis mediated by AAV-delivered HIV-1 broadly neutralizing antibodies. Mol. Ther. 2019, 27, 650–660. [Google Scholar] [CrossRef] [Green Version]
- Kishimoto, T.K.; Ferrari, J.D.; LaMothe, R.A.; Kolte, P.N.; Griset, A.P.; O’Neil, C.; Chan, V.; Browning, E.; Chalishazar, A.; Kuhlman, W.; et al. Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat. Nanotechnol. 2016, 11, 890–899. [Google Scholar] [CrossRef]
- Maldonado, R.A.; LaMothe, R.A.; Ferrari, J.D.; Zhang, A.-H.; Rossi, R.J.; Kolte, P.N.; Griset, A.P.; O’Neil, C.; Altreuter, D.H.; Browning, E.; et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl. Acad. Sci. USA 2015, 112, E156–E165. [Google Scholar] [CrossRef] [Green Version]
- Meliani, A.; Boisgerault, F.; Hardet, R.; Marmier, S.; Collaud, F.; Ronzitti, G.; Leborgne, C.; Costa Verdera, H.; Simon Sola, M.; Charles, S.; et al. Antigen-selective modulation of AAV immunogenicity with tolerogenic rapamycin nanoparticles enables successful vector re-administration. Nat. Commun. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
- Shin, J.-H.; Yue, Y.; Smith, B.; Duan, D. Humoral immunity to AAV-6, 8, and 9 in normal and dystrophic dogs. Hum. Gene Ther. 2012, 23, 287–294. [Google Scholar] [CrossRef]
- Murphy, S.L.; Li, H.; Zhou, S.; Schlachterman, A.; High, K.A.; High, K. Prolonged susceptibility to antibody-mediated neutralization for adeno-associated vectors targeted to the liver. Mol. Ther. 2008, 16, 138–145. [Google Scholar] [CrossRef]
- Rapti, K.; Louis-Jeune, V.; Kohlbrenner, E.; Ishikawa, K.; Ladage, D.; Zolotukhin, S.; Hajjar, R.J.; Weber, T. Neutralizing antibodies against AAV serotypes 1, 2, 6, and 9 in sera of commonly used animal models. Mol. Ther. 2012, 20, 73–83. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Calcedo, R.; Bell, P.; Lin, J.; Grant, R.L.; Siegel, D.L.; Wilson, J.M. Impact of pre-existing immunity on gene transfer to nonhuman primate liver with adeno-associated virus 8 vectors. Hum. Gene Ther. 2011, 22, 1389–1401. [Google Scholar] [CrossRef] [Green Version]
- Elmore, Z.C.; Oh, D.K.; Simon, K.E.; Fanous, M.M.; Asokan, A. Rescuing AAV gene transfer from neutralizing antibodies with an IgG-degrading enzyme. JCI. Insight 2020, 5. [Google Scholar] [CrossRef]
- Tse, L.V.; Klinc, K.A.; Madigan, V.J.; Castellanos Rivera, R.M.; Wells, L.F.; Havlik, L.P.; Smith, J.K.; Agbandje-McKenna, M.; Asokan, A. Structure-guided evolution of antigenically distinct adeno-associated virus variants for immune evasion. Proc. Natl. Acad. Sci. USA 2017, 114, E4812–E4821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Wu, S.; Albright, B.; Hirsch, M.; Li, W.; Tseng, Y.-S.; Agbandje-McKenna, M.; McPhee, S.; Asokan, A.; Samulski, R.J. Development of patient-specific AAV vectors after neutralizing antibody selection for enhanced muscle gene transfer. Mol. Ther. 2016, 24, 53–65. [Google Scholar] [CrossRef] [Green Version]
- Auricchio, A.; O’Connor, E.; Weiner, D.; Gao, G.-P.; Hildinger, M.; Wang, L.; Calcedo, R.; Wilson, J.M. Noninvasive gene transfer to the lung for systemic delivery of therapeutic proteins. J. Clin. Investig. 2002, 110, 499–504. [Google Scholar] [CrossRef]
- Limberis, M.P.; Wilson, J.M. Adeno-associated virus serotype 9 vectors transduce murine alveolar and nasal epithelia and can be readministered. Proc. Natl. Acad. Sci. USA 2006, 103, 12993–12998. [Google Scholar] [CrossRef] [Green Version]
- Masat, E.; Pavani, G.; Mingozzi, F. Humoral immunity to AAV vectors in gene therapy: Challenges and potential solutions. Discov. Med. 2013, 15, 379–389. [Google Scholar]
- Manning, W.C.; Zhou, S.; Bland, M.P.; Escobedo, J.A.; Dwarki, V. Transient immunosuppression allows transgene expression following readministration of adeno-associated viral vectors. Hum. Gene Ther. 1998, 9, 477–485. [Google Scholar] [CrossRef]
- Stasi, R.; Provan, D. Management of immune thrombocytopenic purpura in adults. Mayo Clin. Proc. 2004, 79, 504–522. [Google Scholar] [CrossRef] [Green Version]
- Nathwani, A.C.; Gray, J.T.; Ng, C.Y.C.; Zhou, J.; Spence, Y.; Waddington, S.N.; Tuddenham, E.G.D.; Kemball-Cook, G.; McIntosh, J.; Boon-Spijker, M.; et al. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood 2006, 107, 2653–2661. [Google Scholar] [CrossRef]
- Corti, M.; Elder, M.; Falk, D.; Lawson, L.; Smith, B.; Nayak, S.; Conlon, T.; Clément, N.; Erger, K.; Lavassani, E.; et al. B-cell depletion is protective against anti-AAV capsid immune response: A human subject case study. Mol. Ther. Methods Clin. Dev. 2014, 1. [Google Scholar] [CrossRef]
- Mingozzi, F.; Chen, Y.; Edmonson, S.C.; Zhou, S.; Thurlings, R.M.; Tak, P.P.; High, K.A.; Vervoordeldonk, M.J. Prevalence and pharmacological modulation of humoral immunity to AAV vectors in gene transfer to synovial tissue. Gene Ther. 2013, 20, 417–424. [Google Scholar] [CrossRef]
- Halbert, C.L.; Standaert, T.A.; Wilson, C.B.; Miller, A.D. Successful readministration of adeno-associated virus vectors to the mouse lung requires transient immunosuppression during the initial exposure. J. Virol. 1998, 72, 9795–9805. [Google Scholar] [CrossRef] [Green Version]
- Bouvet, M.; Fang, B.; Ekmekcioglu, S.; Ji, L.; Bucana, C.D.; Hamada, K.; Grimm, E.A.; Roth, J.A. Suppression of the immune response to an adenovirus vector and enhancement of intratumoral transgene expression by low-dose etoposide. Gene Ther. 1998, 5, 189–195. [Google Scholar] [CrossRef] [Green Version]
- Guilbaud, M.; Devaux, M.; Couzinié, C.; Le Duff, J.; Toromanoff, A.; Vandamme, C.; Jaulin, N.; Gernoux, G.; Larcher, T.; Moullier, P.; et al. Five years of successful inducible transgene expression following locoregional adeno-associated virus delivery in nonhuman primates with no detectable immunity. Hum. Gene Ther. 2019, 30, 802–813. [Google Scholar] [CrossRef]
- Fisher, K.J.; Jooss, K.; Alston, J.; Yang, Y.; Haecker, S.E.; High, K.; Pathak, R.; Raper, S.E.; Wilson, J.M. Recombinant adeno-associated virus for muscle directed gene therapy. Nat. Med. 1997, 3, 306–312. [Google Scholar] [CrossRef]
- Herzog, R.W.; Hagstrom, J.N.; Kung, S.-H.; Tai, S.J.; Wilson, J.M.; Fisher, K.J.; High, K.A. Stable gene transfer and expression of human blood coagulation factor IX after intramuscular injection of recombinant adeno-associated virus. Proc. Natl. Acad. Sci. USA 1997, 94, 5804–5809. [Google Scholar] [CrossRef] [Green Version]
- Zhu, J.; Huang, X.; Yang, Y. The TLR9-MyD88 pathway is critical for adaptive immune responses to adeno-associated virus gene therapy vectors in mice. J. Clin. Investig. 2009, 119, 2388–2398. [Google Scholar] [CrossRef] [Green Version]
- Sudres, M.; Ciré, S.; Vasseur, V.; Brault, L.; Da Rocha, S.; Boisgérault, F.; Le Bec, C.; Gross, D.A.; Blouin, V.; Ryffel, B.; et al. MyD88 signaling in B cells regulates the production of Th1-dependent antibodies to AAV. Mol. Ther. 2012, 20, 1571–1581. [Google Scholar] [CrossRef] [Green Version]
- Rogers, G.L.; Suzuki, M.; Zolotukhin, I.; Markusic, D.M.; Morel, L.M.; Lee, B.; Ertl, H.C.J.; Herzog, R.W. Unique roles of TLR9- and MyD88-dependent and -independent pathways in adaptive immune responses to AAV-mediated gene transfer. JIN 2015, 7, 302–314. [Google Scholar] [CrossRef] [PubMed]
- Hösel, M.; Broxtermann, M.; Janicki, H.; Esser, K.; Arzberger, S.; Hartmann, P.; Gillen, S.; Kleeff, J.; Stabenow, D.; Odenthal, M.; et al. Toll-like receptor 2-mediated innate immune response in human nonparenchymal liver cells toward adeno-associated viral vectors. J. Hepatol. 2012, 55, 287–297. [Google Scholar] [CrossRef]
- Martino, A.T.; Suzuki, M.; Markusic, D.M.; Zolotukhin, I.; Ryals, R.C.; Moghimi, B.; Ertl, H.C.J.; Muruve, D.A.; Lee, B.; Herzog, R.W. The genome of self-complementary adeno-associated viral vectors increases Toll-like receptor 9-dependent innate immune responses in the liver. Blood 2011, 117, 6459–6468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faust, S.M.; Bell, P.; Cutler, B.J.; Ashley, S.N.; Zhu, Y.; Rabinowitz, J.E.; Wilson, J.M. CpG-depleted adeno-associated virus vectors evade immune detection. J. Clin. Investig. 2013, 123, 2994–3001. [Google Scholar] [CrossRef] [Green Version]
- Mou, H.; Zhong, G.; Gardner, M.R.; Wang, H.; Wang, Y.-W.; Cheng, D.; Farzan, M. Conditional regulation of gene expression by ligand-induced occlusion of a microRNA target qequence. Mol. Ther. 2018, 26, 1277–1286. [Google Scholar] [CrossRef] [Green Version]
- Marceau, C.D.; Negi, S.S.; Hernandez, H.; Callison, J.; Marzi, A.; Borisevich, V.; Berry, J.; Feldmann, H.; Rockx, B. Novel neutralizing monoclonal antibodies protect rodents against lethal filovirus challenges. Trials Vaccinol. 2014, 3, 89–94. [Google Scholar] [CrossRef] [Green Version]
- Strohl, W.R. Current progress in innovative engineered antibodies. Protein Cell 2018, 9, 86–120. [Google Scholar] [CrossRef] [Green Version]
- Gu, J.; Ghayur, T. Generation of dual-variable-domain immunoglobulin molecules for dual-specific targeting. Methods Enzymol. 2012, 502, 25–41. [Google Scholar] [CrossRef]
- Cerulli, R.A.; Kritzer, J.A. Stringing together a universal influenza antibody. Biochemistry 2019, 58, 1943–1944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Antibody and Source | Target | Competes with | Cross-Binds | Cross Neutralizes | Protection | Reference | |
---|---|---|---|---|---|---|---|
MR72 | Human IgG1; Survivor from 2014 MARV outbreak in Uganda | Receptor-binding site (RBS) near the hydrophobic trough and the apex of GP1 | MR78, MR82, MR191 | MARV (all strains), EBOV GP | MARV (Uganda), RAVV | 100% protective in mice (n = 5) against MARV (Uganda) in mice | [95] |
M78 | Human IgG1; Survivor from 2014 MARV outbreak in Uganda | Binds to the top and side of GP1 at a shallow angle relative to the central three-fold axis | MR72, MR82, MR191 | MARV (Uganda) and RAVV | MARV (Uganda and Ravn), potentially neutralizes EBOV entry by inhibiting viral membrane fusion downstream from virus receptor recognition | 100% protective in guinea pigs against MARV (Angola) and 100% against RAVV | [95,97] |
MR82 | Human IgG1; Survivor from 2014 MARV outbreak in Uganda | Binds toward the top and side of GP1 at a shallow angle relative to the central three-fold axis | MR72, MR78, MR191 | MARV (all strains) | MARV (all strains) | 40% protective in guinea pigs; however, it was 100% effective in mice | [95] |
MR191 | Human IgG1; Survivor from 2014 MARV outbreak in Uganda | RBS near the hydrophobic trough and the apex of GP1 | MR72, MR78, MR82 | MARV (Muskoke and Angola), RAVV, EBOV GP (does not neutralize) | MARV (Angola and Muskoke) | 100% protective against MARV (Angola) and RAVV in guinea pigs. 100% effective in NHPs when treated on day 4 and 7 post infection (MARV Angola). 80% protective on days 5 and 8 post infection of RAVV. Did not protect against GPA-SUDV in guinea pig models 3 dpi | [94,95,98] |
Antibody and Source | Target | Competes with | Cross-Binds | Cross Neutralizes | Protection | Reference | |
---|---|---|---|---|---|---|---|
CA45 | Macaque; Immunized macaque against EBOV, SUDV and MARV GPs | Conserved epitope in the internal fusion loop. Binds to the native GP but binds with way higher potency to the cleaved GP. Does not bind soluble GP (sGP) | KZ52, 2G4 and 4G7 | EBOV, SUDV and BDBV (does not bind MARV, Tai Forest or LLOV) | Potently neutralizes EBOV, SUDV, and BDBV and moderately neutralizes Reston. IC50 for EBOV and cleaved core ectodomain of EBOV (EBOV GPcl) was 4.63 and 0.05 nM, BDBV and GPcl was 4.24 and 0.007 nM, and SUDV and GPcl was 9.16 and 0.05 nM | In mice (n = 20), protection was as good as against EBOV and 80% against SUDV. In guinea pigs (n = 6) it was 100% protective against EBOV and SUDV. 100% protection in ferret model (n = 4) of BDBV. FVM04 + CA45 in mice against SUDV was 100% effective, mirrored in guinea pigs for EBOV | [100] |
BDBV223 | Human IgG3; Human survivor of 2007 Uganda BDBV outbreak | GP2 stalk (canonical heptad repeat 2 (HR2) domain near the MPER) (71% conserved amongst the five first Ebolaviruses, and 90% conserved amongst EBOV, BDBV, and SUDV), does not recognize BDBV sGP | BDBV317, BDBV340 | rVSV-GP of EBOV, BDBV, and SUDV | rVSV-GP of EBOV, BDBV, and SUDV | 100% in mice (n = 5), post-exposure (1 day) was 20% protection against lethal EBOV (Mayinga) challenge in guinea pigs (n = 5) and 50% effective in ferret models (n = 4), 3- and 6-days post-challenge | [104,105,106] |
FVM04 | Cloned as human IgG1 with kappa light chain; Macaque vaccinated with EBOV, SUDV, and MARV GP | The exposed tip of the receptor binding domain crest at the apex of the GP trimer and blocks interaction of GP with NPC1 in the late endosome. Glycans may moderately interfere with optimal binding. The RBD crest is highly conserved | M13C6, partially KZ52 | EBOV, SUDV, and BDBV (Binds WT, cleaved and sGP of all Ebola species), low affinity binding to MARV and Reston | EBOV and SUDV but not really BDBV or MARV. It is a weaker neutralization than some others: EC50 of 4.3 and 4.3 ug/mL for SUDV and EBOV | 100% in mice against EBOV and 75% in mice against SUDV. 100% protective in guinea pigs against SUDV and 40% protective against EBOV | [101,107] |
ADI-15742 | Human IgG; Human survivor of 2014 West Africa outbreak | A highly conserved region of GP2 within the internal fusion loop. Locks the GP into a pre-fusion state whether free or bound to NPC1. Does not bind sGP | ADI-15878, 100, KZ52, CA45 | Everything but MARV/LLOV (rVSV-GP for each Ebolavirus and Marburgvirus) | Potently neutralizes all five Ebola virus with IC50 of <2 nM for rVSV pseudotypes | 100% protection in lethal mouse model (100 μg against 100 pfu MA-EBOV) The two ADIs are clonal siblings with 90% and 95% chain identity. * | [102] |
ADI-15878 | Human IgG; Human survivor of 2014 West Africa outbreak | A highly conserved region of GP2 within the internal fusion loop. Locks the GP into a pre-fusion state whether free or bound to NPC1. Does not bind sGP. Seems to target a cleaved intermediate in endosomes while first generation base binders act further upstream | 100, KZ52, CA45 | Everything but MARV/LLOV | Potently neutralizes all five Ebola virus with IC50 of <2 nM for rVSV pseudotypes | 80% protective in mice (n = 10) against EBOV and 100% protective in mice (n = 20) against SUDV. 75% protective against BDBV in ferrets (n = 4) | [102] |
ADI-23374 | Derived from affinity maturation of a human mAb (Human survivor of 2014 West Africa outbreak ADI-15946) using yeast resulting in enhanced potency against SUDV. | ADI-15946 binds a highly conserved epitope shielded by the mobile β17–β18 loop of the glycan cap, which is where ADI-23774 binds, does not bind sGP of EBOV | EBOV, BDBV, SUDV | EBOV, BDBV, SUDV | rVSV-SUDV GP | 90% protective against EBOV in mice (n = 10) 3 days post exposure and was 100% protective against wt-SUDV in mice (IFNR-/- mice) (n = 10) at 1- and 4-days post exposure | [108] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rghei, A.D.; van Lieshout, L.P.; Santry, L.A.; Guilleman, M.M.; Thomas, S.P.; Susta, L.; Karimi, K.; Bridle, B.W.; Wootton, S.K. AAV Vectored Immunoprophylaxis for Filovirus Infections. Trop. Med. Infect. Dis. 2020, 5, 169. https://doi.org/10.3390/tropicalmed5040169
Rghei AD, van Lieshout LP, Santry LA, Guilleman MM, Thomas SP, Susta L, Karimi K, Bridle BW, Wootton SK. AAV Vectored Immunoprophylaxis for Filovirus Infections. Tropical Medicine and Infectious Disease. 2020; 5(4):169. https://doi.org/10.3390/tropicalmed5040169
Chicago/Turabian StyleRghei, Amira D., Laura P. van Lieshout, Lisa A. Santry, Matthew M. Guilleman, Sylvia P. Thomas, Leonardo Susta, Khalil Karimi, Byram W. Bridle, and Sarah K. Wootton. 2020. "AAV Vectored Immunoprophylaxis for Filovirus Infections" Tropical Medicine and Infectious Disease 5, no. 4: 169. https://doi.org/10.3390/tropicalmed5040169
APA StyleRghei, A. D., van Lieshout, L. P., Santry, L. A., Guilleman, M. M., Thomas, S. P., Susta, L., Karimi, K., Bridle, B. W., & Wootton, S. K. (2020). AAV Vectored Immunoprophylaxis for Filovirus Infections. Tropical Medicine and Infectious Disease, 5(4), 169. https://doi.org/10.3390/tropicalmed5040169