More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation
Abstract
:1. At a Glance: History of the Evolution of the Lentiviral Pseudotype System
2. Applications in Serological Studies
2.1. Evaluation of Vaccine Efficacy
2.2. Serosurveillance Studies
2.3. Cross-Reactive Antibody Responses
2.4. Antigenic Site Analysis
2.5. Selection of Therapeutic Antivirals
3. Production of Pseudotyped LVs
3.1. Factors towards Optimising Titres
3.2. Quantification of LV Yield
3.3. Quality Control of LVs
4. Correlation with Live Virus Assays
5. Alternative Vector Systems
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Rubin, H. Genetic control of cellular susceptibility to pseudotypes of rous sarcoma virus. Virology 1965, 26, 270–276. [Google Scholar] [CrossRef]
- Li, Q.; Liu, Q.; Huang, W.; Li, X.; Wang, Y. Current status on the development of pseudoviruses for enveloped viruses. Rev. Med. Virol. 2018, 28, e1963. [Google Scholar] [CrossRef] [PubMed]
- Mather, S.; Scott, S.; Temperton, N.; Wright, E.; King, B.; Daly, J. Current progress with serological assays for exotic emerging/re-emerging viruses. Future Virol. 2013, 8, 745–755. [Google Scholar] [CrossRef] [Green Version]
- Munis, A.M. Gene therapy applications of non-human lentiviral vectors. Viruses 2020, 12, 1106. [Google Scholar] [CrossRef] [PubMed]
- Duvergé, A.; Negroni, M. Pseudotyping Lentiviral Vectors: When the Clothes Make the Virus. Viruses 2020, 12, 1311. [Google Scholar] [CrossRef]
- Joglekar, A.V.; Sandoval, S. Pseudotyped Lentiviral Vectors: One Vector, Many Guises. Hum. Gene Ther. Methods 2017, 28, 291–301. [Google Scholar] [CrossRef] [Green Version]
- Frankel, A.; Young, J. HIV-1: Fifteen Proteins and an RNA. Annu. Rev. Biochem. 1998, 67, 1–25. [Google Scholar] [CrossRef] [Green Version]
- Terwilliger, E.F.; Godin, B.; Sodroski, J.G.; Haseltine, W.A. Construction and use of a replication-competent human immunodeficiency virus (HIV-1) that expresses the chloramphenicol acetyltransferase enzyme. Proc. Natl. Acad. Sci. USA 1989, 86, 3857–3861. [Google Scholar] [CrossRef] [Green Version]
- Sakuma, T.; Barry, M.A.; Ikeda, Y. Lentiviral vectors: Basic to translational. Biochem. J. 2012, 443, 603–618. [Google Scholar] [CrossRef] [Green Version]
- Boulanger, P.; Jones, I. Use of heterologous expression systems to study retroviral morphogenesis. Curr. Top. Microbiol. Immunol. 1996, 214, 236–260. [Google Scholar] [CrossRef]
- Ako-Adjei, D.; Johnson, M.C.; Vogt, V.M. The Retroviral Capsid Domain Dictates Virion Size, Morphology, and Coassembly of Gag into Virus-Like Particles. J. Virol. 2005, 79, 13463–13472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Landau, N.R.; Page, K.A.; Littman, D.R. Pseudotyping with human T-cell leukemia virus type I broadens the human immunodeficiency virus host range. J. Virol. 1991, 65, 162–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steffen, I.; Simmons, G. Pseudotyping Viral Vectors with Emerging Virus Envelope Proteins. Curr. Gene Ther. 2016, 16, 47–55. [Google Scholar] [CrossRef] [PubMed]
- Urbanowicz, R.A.; McClure, C.P.; King, B.; Mason, C.P.; Ball, J.K.; Tarr, A.W. Novel functional hepatitis C virus glycoprotein isolates identified using an optimized viral pseudotype entry assay. J. Gen. Virol. 2016, 97, 2265–2279. [Google Scholar] [CrossRef]
- Collins, M.H. Serologic tools and strategies to support intervention trials to combat Zika virus infection and disease. Trop. Med. Infect. Dis. 2019, 4, 68. [Google Scholar] [CrossRef] [Green Version]
- Winter, A.K.; Hegde, S.T. The important role of serology for COVID-19 control. Lancet Infect. Dis. 2020, 20, 758–759. [Google Scholar] [CrossRef]
- GeurtsvanKessel, C.H.; Okba, N.M.A.; Igloi, Z.; Bogers, S.; Embregts, C.W.E.; Laksono, B.M.; Leijten, L.; Rokx, C.; Rijnders, B.; Rahamat-Langendoen, J.; et al. An evaluation of COVID-19 serological assays informs future diagnostics and exposure assessment. Nat. Commun. 2020, 11, 1–5. [Google Scholar] [CrossRef]
- Berry, N.; Kempster, S.; Ham, C.; Jenkins, A.; Hall, J.; Page, M.; Mattiuzzo, G.; Adedeji, Y.; Hewson, R.; Giles, E.; et al. Passive immunisation of convalescent human anti-Zika plasma protects against challenge with New World Zika virus in cynomolgus macaques. NPJ Vaccines 2020, 5, 1–7. [Google Scholar] [CrossRef]
- Dowall, S.D.; Bosworth, A.; Watson, R.; Bewley, K.; Taylor, I.; Rayner, E.; Hunter, L.; Pearson, G.; Easterbrook, L.; Pitman, J.; et al. Chloroquine inhibited ebola virus replication in vitro but failed to protect against infection and disease in the in vivo guinea pig model. J. Gen. Virol. 2015, 96, 3484–3492. [Google Scholar] [CrossRef]
- El-Duah, P.; Meyer, B.; Sylverken, A.; Owusu, M.; Gottula, L.T.; Yeboah, R.; Lamptey, J.; Frimpong, Y.O.; Burimuah, V.; Folitse, R.; et al. Development of a whole-virus elisa for serological evaluation of domestic livestock as possible hosts of human coronavirus nl63. Viruses 2019, 11, 43. [Google Scholar] [CrossRef] [Green Version]
- Jiang, J.; Banglore, P.; Cashman, K.A.; Schmaljohn, C.S.; Schultheis, K.; Pugh, H.; Nguyen, J.; Humeau, L.M.; Broderick, K.E.; Ramos, S.J. Immunogenicity of a protective intradermal DNA vaccine against lassa virus in cynomolgus macaques. Hum. Vaccines Immunother. 2019, 15, 2066–2074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madrid, P.B.; Chopra, S.; Manger, I.D.; Gilfillan, L.; Keepers, T.R.; Shurtleff, A.C.; Green, C.E.; Iyer, L.V.; Dilks, H.H.; Davey, R.A.; et al. A Systematic Screen of FDA-Approved Drugs for Inhibitors of Biological Threat Agents. PLoS ONE 2013, 8, e60579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munster, V.J.; Wells, D.; Lambe, T.; Wright, D.; Fischer, R.J.; Bushmaker, T.; Saturday, G.; Van Doremalen, N.; Gilbert, S.C.; De Wit, E.; et al. Protective efficacy of a novel simian adenovirus vaccine against lethal MERS-CoV challenge in a transgenic human DPP4 mouse model. NPJ Vaccines 2017, 2, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Folegatti, P.M.; Bittaye, M.; Flaxman, A.; Lopez, F.R.; Bellamy, D.; Kupke, A.; Mair, C.; Makinson, R.; Sheridan, J.; Rohde, C.; et al. Safety and immunogenicity of a candidate Middle East respiratory syndrome coronavirus viral-vectored vaccine: A dose-escalation, open-label, non-randomised, uncontrolled, phase 1 trial. Lancet Infect. Dis. 2020, 20, 816–826. [Google Scholar] [CrossRef]
- Alharbi, N.K.; Qasim, I.; Almasoud, A.; Aljami, H.A.; Alenazi, M.W.; Alhafufi, A.; Aldibasi, O.S.; Hashem, A.M.; Kasem, S.; Albrahim, R.; et al. Humoral Immunogenicity and Efficacy of a Single Dose of ChAdOx1 MERS Vaccine Candidate in Dromedary Camels. Sci. Rep. 2019, 9, 1–11. [Google Scholar] [CrossRef]
- Hemida, M.G.; Perera, R.A.; Wang, P.; Alhammadi, M.A.; Siu, L.Y.; Li, M.; Poon, L.L.; Saif, L.; Alnaeem, A.; Peiris, M. Middle east respiratory syndrome (MERS) coronavirus seroprevalence in domestic livestock in Saudi Arabia, 2010 to 2013. Eurosurveillance 2013, 18, 1–7. [Google Scholar] [CrossRef]
- Perera, R.A.; Wang, P.; Gomaa, M.R.; El-Shesheny, R.; Kandeil, A.; Bagato, O.; Siu, L.Y.; Shehata, M.M.; Kayed, A.S.; Moatasim, Y.; et al. Seroepidemiology for MERS coronavirus using microneutralisation and pseudoparticle virus neutralisation assays reveal a high prevalence of antibody in dromedary camels in Egypt, June 2013. Eurosurveillance 2013, 18, 1–7. [Google Scholar] [CrossRef]
- Gierer, S.; Hofmann-Winkler, H.; Albuali, W.H.; Bertram, S.; Al-Rubaish, A.M.; Yousef, A.A.; Al-Nafaie, A.N.; Al-Ali, A.K.; Obeid, O.E.; Alkharsah, K.R.; et al. Lack of MERS coronavirus neutralizing antibodies in humans, Eastern Province, Saudi Arabia. Emerg. Infect. Dis. 2013, 19, 2034–2036. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Wang, G.; Li, J.; Nie, Y.; Shi, X.; Lian, G.; Wang, W.; Yin, X.; Zhao, Y.; Qu, X.; et al. Identification of an Antigenic Determinant on the S2 Domain of the Severe Acute Respiratory Syndrome Coronavirus Spike Glycoprotein Capable of Inducing Neutralizing Antibodies. J. Virol. 2004, 78, 6938–6945. [Google Scholar] [CrossRef] [Green Version]
- Yang, Z.; Kong, W.; Huang, Y.; Roberts, A.; Murphy, B.R.; Subbarao, K.; Nabel, G.J. A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice. Nature 2004, 428, 561–564. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.; Agudelo, J.; Lu, K.; Goetz, D.H.; Hansell, E.; Chen, Y.T.; Roush, W.R.; McKerrow, J.; Craik, C.S.; Amberg, S.M.; et al. Inhibitors of SARS-CoV entry—Identification using an internally-controlled dual envelope pseudovirion assay. Antiviral Res. 2011, 92, 187–194. [Google Scholar] [CrossRef] [PubMed]
- Folegatti, P.M.; Ewer, K.J.; Aley, P.K.; Angus, B.; Becker, S.; Belij-Rammerstorfer, S.; Bellamy, D.; Bibi, S.; Bittaye, M.; Clutterbuck, E.A.; et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: A preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 2020, 396, 467–478. [Google Scholar] [CrossRef]
- Muruato, A.E.; Fontes-Garfias, C.R.; Ren, P.; Garcia-Blanco, M.A.; Menachery, V.D.; Xie, X.; Shi, P.Y. A high-throughput neutralizing antibody assay for COVID-19 diagnosis and vaccine evaluation. Nat. Commun. 2020, 11, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Yang, R.; Huang, B.; Ruhan, A.; Li, W.; Wang, W.; Deng, Y.; Tan, W. Development and effectiveness of Pseudotyped SARS-CoV-2 system as determined by neutralizing efficiency and entry inhibition test in vitro. Biosaf. Health 2020. [Google Scholar] [CrossRef] [PubMed]
- Thompson, C.P.; Grayson, N.E.; Paton, R.S.; Bolton, J.S.; Lourenço, J.; Penman, B.S.; Lee, L.N.; Odon, V.; Mongkolsapaya, J.; Chinnakannan, S.; et al. Detection of neutralising antibodies to SARS-CoV-2 to determine population exposure in Scottish blood donors between March and May 2020. Eurosurveillance 2020, 25, 2000685. [Google Scholar] [CrossRef] [PubMed]
- Tandon, R.; Mitra, D.; Sharma, P.; McCandless, M.G.; Stray, S.J.; Bates, J.T.; Marshall, G.D. Effective screening of SARS-CoV-2 neutralizing antibodies in patient serum using lentivirus particles pseudotyped with SARS-CoV-2 spike glycoprotein. Sci. Rep. 2020, 10. [Google Scholar] [CrossRef] [PubMed]
- Henss, L.; Scholz, T.; von Rhein, C.; Wieters, I.; Borgans, F.; Eberhardt, F.J.; Zacharowski, K.; Ciesek, S.; Rohde, G.; Vehreschild, M.; et al. Analysis of Humoral Immune Responses in Patients With Severe Acute Respiratory Syndrome Coronavirus 2 Infection. J. Infect. Dis. 2020. [Google Scholar] [CrossRef]
- Zeng, C.; Evans, J.P.; Pearson, R.; Qu, P.; Zheng, Y.M.; Robinson, R.T.; Hall-Stoodley, L.; Yount, J.; Pannu, S.; Mallampalli, R.K.; et al. Neutralizing antibody against SARS-CoV-2 spike in COVID-19 patients, health care workers, and convalescent plasma donors. JCI Insight 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed]
- Wang, K.; Long, Q.-X.; Deng, H.-J.; Hu, J.; Gao, Q.-Z.; Zhang, G.-J.; He, C.-L.; Huang, L.-Y.; Hu, J.-L.; Chen, J.; et al. Longitudinal Dynamics of the Neutralizing Antibody Response to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection. Clin. Infect. Dis. 2020. [Google Scholar] [CrossRef] [PubMed]
- Kemp, S.A.; Collier, D.A.; Datir, R.; Ferreira, I.; Gayed, S.; Jahun, A.; Hosmillo, M.; Rees-Spear, C.; Mlcochova, P.; Lumb, I.U.; et al. Neutralising antibodies in Spike mediated SARS-CoV-2 adaptation. medRxiv Prepr. Serv. Health Sci. 2020. [Google Scholar] [CrossRef]
- Kemp, S.A.; Harvey, W.T.; Lytras, S.; Carabelli, A.M.; Robertson, D.L.; Gupta, R.K. Recurrent emergence and transmission of a SARS-CoV-2 Spike deletion H69/V70. bioRxiv 2021. [Google Scholar] [CrossRef]
- Greaney, A.J.; Loes, A.N.; Crawford, K.H.D.; Starr, T.N.; Malone, K.D.; Chu, H.Y.; Bloom, J.D. Comprehensive mapping of mutations to the SARS-CoV-2 receptor-binding domain that affect recognition by polyclonal human serum antibodies. bioRxiv 2021. [Google Scholar] [CrossRef]
- Collier, D.; Meng, B.; Ferreira, I.; Datir, R.; Temperton, N.J.; Elmer, A.; Graves, B.; Kingston, N.; McCoy, L.; Smith, K.; et al. Impact of SARS-CoV-2 B.1.1.7 Spike variant on neutralisation potency of sera from individuals vaccinated with Pfizer vaccine BNT162b2. medRxiv 2021. [Google Scholar] [CrossRef]
- Starr, T.N.; Greaney, A.J.; Hilton, S.K.; Ellis, D.; Crawford, K.H.D.; Dingens, A.S.; Navarro, M.J.; Bowen, J.E.; Tortorici, M.A.; Walls, A.C.; et al. Deep Mutational Scanning of SARS-CoV-2 Receptor Binding Domain Reveals Constraints on Folding and ACE2 Binding. Cell 2020, 182, 1295–1310.e20. [Google Scholar] [CrossRef] [PubMed]
- Wibmer, C.K.; Ayres, F.; Hermanus, T.; Madzivhandila, M.; Kgagudi, P.; Lambson, B.E.; Vermeulen, M.; van den Berg, K.; Rossouw, T.; Boswell, M.; et al. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. bioRxiv 2021. [Google Scholar] [CrossRef]
- Zost, S.J.; Gilchuk, P.; Case, J.B.; Binshtein, E.; Chen, R.E.; Nkolola, J.P.; Schäfer, A.; Reidy, J.X.; Trivette, A.; Nargi, R.S.; et al. Potently neutralizing and protective human antibodies against SARS-CoV-2. Nature 2020, 584, 443–449. [Google Scholar] [CrossRef]
- AZD7442—A Potential Combination Therapy for the Prevention and Treatment of COVID-19. Available online: https://clinicaltrials.gov/ct2/show/NCT04507256?term=NCT04507256&draw=2&rank=1 (accessed on 19 December 2020).
- Harvala, H.; Robb, M.; Watkins, N.; Ijaz, S.; Dicks, S.; Patel, M.; Supasa, P.; Dejnirattisai, W.; Liu, C.; Mongkolsapaya, J.; et al. Convalescent plasma therapy for the treatment of patients with COVID-19: Assessment of methods available for antibody detection and their correlation with neutralising antibody levels. Transfus. Med. 2020, 1–9. [Google Scholar] [CrossRef]
- Ewer, K.; Rampling, T.; Venkatraman, N.; Bowyer, G.; Wright, D.; Lambe, T.; Imoukhuede, E.B.; Payne, R.; Fehling, S.K.; Strecker, T.; et al. A Monovalent Chimpanzee Adenovirus Ebola Vaccine Boosted with MVA. N. Engl. J. Med. 2016, 374, 1635–1646. [Google Scholar] [CrossRef]
- O’Hearn, A.E.; Voorhees, M.A.; Fetterer, D.P.; Wauquier, N.; Coomber, M.R.; Bangura, J.; Fair, J.N.; Gonzalez, J.-P.; Schoepp, R.J. Serosurveillance of viral pathogens circulating in West Africa. Virol. J. 2016, 13, 163. [Google Scholar] [CrossRef] [Green Version]
- Glynn, J.R.; Bower, H.; Johnson, S.; Houlihan, C.F.; Montesano, C.; Scott, J.T.; Semple, M.G.; Bangura, M.S.; Kamara, A.J.; Kamara, O.; et al. Asymptomatic infection and unrecognised Ebola virus disease in Ebola-affected households in Sierra Leone: A cross-sectional study using a new non-invasive assay for antibodies to Ebola virus. Lancet Infect. Dis. 2017, 17, 645–653. [Google Scholar] [CrossRef] [Green Version]
- Steffen, I.; Lu, K.; Yamamoto, L.K.; Hoff, N.A.; Mulembakani, P.; Wemakoy, E.O.; Muyembe-Tamfum, J.J.; Ndembi, N.; Brennan, C.A.; Hackett, J.; et al. Serologic prevalence of ebola virus in equatorial Africa. Emerg. Infect. Dis. 2019, 25, 911–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luczkowiak, J.; Lasala, F.; Mora-Rillo, M.; Arribas, J.R.; Delgado, R. Broad Neutralizing Activity Against Ebolaviruses Lacking the Mucin-Like Domain in Convalescent Plasma Specimens From Patients With Ebola Virus Disease. J. Infect. Dis. 2018, 218, S574–S581. [Google Scholar] [CrossRef] [PubMed]
- Long, J.; Wright, E.; Molesti, E.; Temperton, N.; Barclay, W. Antiviral therapies against Ebola and other emerging viral diseases using existing medicines that block virus entry. F1000Research 2015, 4, 30. [Google Scholar] [CrossRef] [Green Version]
- Chen, F.; Nagy, K.; Chavez, D.; Willis, S.; McBride, R.; Giang, E.; Honda, A.; Bukh, J.; Ordoukhanian, P.; Zhu, J.; et al. Antibody Responses to Immunization With HCV Envelope Glycoproteins as a Baseline for B-Cell–Based Vaccine Development. Gastroenterology 2020, 158, 1058–1071.e6. [Google Scholar] [CrossRef] [PubMed]
- Netski, D.M.; Mosbruger, T.; Depla, E.; Maertens, G.; Ray, S.C.; Hamilton, R.G.; Roundtree, S.; Thomas, D.L.; McKeating, J.; Cox, A. Humoral Immune Response in Acute Hepatitis C Virus Infection. Clin. Infect. Dis. 2005, 41, 667–675. [Google Scholar] [CrossRef]
- Urbanowicz, R.A.; McClure, C.P.; Brown, R.J.P.; Tsoleridis, T.; Persson, M.A.A.; Krey, T.; Irving, W.L.; Ball, J.K.; Tarr, A.W. A Diverse Panel of Hepatitis C Virus Glycoproteins for Use in Vaccine Research Reveals Extremes of Monoclonal Antibody Neutralization Resistance. J. Virol. 2016, 90, 3288–3301. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.W.; Cheng, T.J.R.; Huang, Y.; Jan, J.T.; Ma, S.H.; Yu, A.L.; Wong, C.H.; Ho, D.D. A consensus—Hemagglutinin-based DNA vaccine that protects mice against divergent H5N1 influenza viruses. Proc. Natl. Acad. Sci. USA 2008, 105, 13538–13543. [Google Scholar] [CrossRef] [Green Version]
- Rao, S.; Kong, W.P.; Wei, C.J.; Yang, Z.Y.; Nason, M.; Styles, D.; DeTolla, L.J.; Sorrell, E.M.; Song, H.; Wan, H.; et al. Multivalent HA DNA vaccination protects against highly pathogenic H5N1 avian influenza infection in chickens and mice. PLoS ONE 2008, 3, e2432. [Google Scholar] [CrossRef]
- Rao, S.S.; Kong, W.-P.; Wei, C.-J.; Van Hoeven, N.; Gorres, J.P.; Nason, M.; Andersen, H.; Tumpey, T.M.; Nabel, G.J. Comparative Efficacy of Hemagglutinin, Nucleoprotein, and Matrix 2 Protein Gene-Based Vaccination against H5N1 Influenza in Mouse and Ferret. PLoS ONE 2010, 5, e9812. [Google Scholar] [CrossRef] [Green Version]
- Tsai, C.; Caillet, C.; Hu, H.; Zhou, F.; Ding, H.; Zhang, G.; Zhou, B.; Wang, S.; Lu, S.; Buchy, P.; et al. Measurement of neutralizing antibody responses against H5N1 clades in immunized mice and ferrets using pseudotypes expressing influenza hemagglutinin and neuraminidase. Vaccine 2009, 27, 6777–6790. [Google Scholar] [CrossRef]
- Yang, Z.; Wei, C.; Kong, W.; Wu, L.; Xu, L.; Smith, D.F.; Nabel, G.J. Immunization by Avian H5 Influenza Hemagglutinin Mutants with Altered Receptor Binding Specificity. Science 2007, 317, 825–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, C.J.; Boyington, J.C.; McTamney, P.M.; Kong, W.P.; Pearce, M.B.; Xu, L.; Andersen, H.; Rao, S.; Tumpey, T.M.; Yang, Z.Y.; et al. Induction of broadly neutralizing H1N1 influenza antibodies by vaccination. Science 2010, 329, 1060–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, W.; Bhide, Y.; Sicca, F.; Meijerhof, T.; Guilfoyle, K.; Engelhardt, O.G.; Boon, L.; De Haan, C.A.M.; Carnell, G.; Temperton, N.; et al. Cross-protective immune responses induced by sequential influenza virus infection and by sequential vaccination with inactivated influenza vaccines. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
- Tully, C.M.; Chinnakannan, S.; Mullarkey, C.E.; Ulaszewska, M.; Ferrara, F.; Temperton, N.; Gilbert, S.C.; Lambe, T. Novel Bivalent Viral-Vectored Vaccines Induce Potent Humoral and Cellular Immune Responses Conferring Protection against Stringent Influenza A Virus Challenge. J. Immunol. 2017, 199, 1333–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andersen, T.K.; Zhou, F.; Cox, R.; Bogen, B.; Grødeland, G. A DNA Vaccine That Targets Hemagglutinin to Antigen-Presenting Cells Protects Mice against H7 Influenza. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valkenburg, S.A.; Mallajosyula, V.V.A.; Li, O.T.W.; Chin, A.W.H.; Carnell, G.; Temperton, N.; Varadarajan, R.; Poon, L.L.M. Stalking influenza by vaccination with pre-fusion headless HA mini-stem. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef] [Green Version]
- Mallajosyula, V.V.A.; Citron, M.; Ferrara, F.; Lu, X.; Callahan, C.; Heidecker, G.J.; Sarma, S.P.; Flynn, J.A.; Temperton, N.J.; Liang, X.; et al. Influenza hemagglutinin stem-fragment immunogen elicits broadly neutralizing antibodies and confers heterologous protection. Proc. Natl. Acad. Sci. USA 2014, 111, E2514. [Google Scholar] [CrossRef] [Green Version]
- Mallajosyula, V.V.A.; Citron, M.; Ferrara, F.; Temperton, N.J.; Liang, X.; Flynn, J.A.; Varadarajan, R. Hemagglutinin sequence conservation guided stem immunogen design from influenza A H3 subtype. Front. Immunol. 2015, 6, 329. [Google Scholar] [CrossRef] [Green Version]
- Cavailler, P.; Chu, S.; Ly, S.; Garcia, J.M.; Ha, D.Q.; Bergeri, I.; Som, L.; Ly, S.; Sok, T.; Vong, S.; et al. Seroprevalence of anti-H5 antibody in rural Cambodia, 2007. J. Clin. Virol. 2010, 48, 123–126. [Google Scholar] [CrossRef]
- Lo, C.-Y.; Strobl, S.L.; Dunham, K.; Wang, W.; Stewart, L.; Misplon, J.A.; Garcia, M.; Gao, J.; Ozawa, T.; Price, G.E.; et al. Surveillance Study of Influenza Occurrence and Immunity in a Wisconsin Cohort During the 2009 Pandemic. Open Forum Infect. Dis. 2017, 4, ofx023. [Google Scholar] [CrossRef] [Green Version]
- Buchy, P.; Vong, S.; Chu, S.; Garcia, J.M.; Hien, T.T.; Hien, V.M.; Channa, M.; Ha, D.Q.; van Chau, N.V.; Simmons, C.; et al. Kinetics of neutralizing antibodies in patients naturally infected by H5N1 virus. PLoS ONE 2010, 5, e10864. [Google Scholar] [CrossRef] [PubMed]
- Desvaux, S.; Garcia, J.M.; Nguyen, T.D.; Reid, S.A.; Bui, N.A.; Roger, F.; Fenwick, S.; Peiris, J.S.M.; Ellis, T. Evaluation of serological tests for H5N1 avian influenza on field samples from domestic poultry populations in Vietnam: Consequences for surveillance. Vet. Microbiol. 2012, 156, 277–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nefkens, I.; Garcia, J.M.; Ling, C.S.; Lagarde, N.; Nicholls, J.; Tang, D.J.; Peiris, M.; Buchy, P.; Altmeyer, R. Hemagglutinin pseudotyped lentiviral particles: Characterization of a new method for avian H5N1 influenza sero-diagnosis. J. Clin. Virol. 2007, 39, 27–33. [Google Scholar] [CrossRef] [PubMed]
- Kneissl, S.; Abel, T.; Rasbach, A.; Brynza, J.; Schneider-Schaulies, J.; Buchholz, C.J. Measles Virus Glycoprotein-Based Lentiviral Targeting Vectors That Avoid Neutralizing Antibodies. PLoS ONE 2012, 7, e46667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lévy, C.; Amirache, F.; Costa, C.; Frecha, C.; Muller, C.P.; Kweder, H.; Buckland, R.; Cosset, F.L.; Verhoeyen, E. Lentiviral vectors displaying modified measles virus gp overcome pre-existing immunity in in vivo-like transduction of human T and B cells. Mol. Ther. 2012, 20, 1699–1712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schoofs, T.; Klein, F.; Braunschweig, M.; Kreider, E.F.; Feldmann, A.; Nogueira, L.; Oliveira, T.; Lorenzi, J.C.C.; Parrish, E.H.; Learn, G.H.; et al. HIV-1 therapy with monoclonal antibody 3BNC117 elicits host immune responses against HIV-1. Science 2016, 352, 997–1001. [Google Scholar] [CrossRef] [Green Version]
- Mendoza, P.; Gruell, H.; Nogueira, L.; Pai, J.A.; Butler, A.L.; Millard, K.; Lehmann, C.; Suárez, I.; Oliveira, T.Y.; Lorenzi, J.C.C.; et al. Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature 2018, 561, 479–484. [Google Scholar] [CrossRef]
- Cohen, Y.Z.; Butler, A.L.; Millard, K.; Witmer-Pack, M.; Levin, R.; Unson-O’Brien, C.; Patel, R.; Shimeliovich, I.; Lorenzi, J.C.C.; Horowitz, J.; et al. Safety, pharmacokinetics, and immunogenicity of the combination of the broadly neutralizing anti-HIV-1 antibodies 3BNC117 and 10-1074 in healthy adults: A randomized, phase 1 study. PLoS ONE 2019, 14, e0219142. [Google Scholar] [CrossRef] [Green Version]
- Wright, E.; Temperton, N.J.; Marston, D.A.; McElhinney, L.M.; Fooks, A.R.; Weiss, R.A. Investigating antibody neutralization of lyssaviruses using lentiviral pseudotypes: A cross-species comparison. J. Gen. Virol. 2008, 89, 2204–2213. [Google Scholar] [CrossRef] [Green Version]
- Wright, E.; McNabb, S.; Goddard, T.; Horton, D.L.; Lembo, T.; Nel, L.H.; Weiss, R.A.; Cleaveland, S.; Fooks, A.R. A robust lentiviral pseudotype neutralisation assay for in-field serosurveillance of rabies and lyssaviruses in Africa. Vaccine 2009, 27, 7178–7186. [Google Scholar] [CrossRef] [Green Version]
- Wright, E.; Hayman, D.T.S.; Vaughan, A.; Temperton, N.J.; Wood, J.L.N.; Cunningham, A.A.; Suu-Ire, R.; Weiss, R.A.; Fooks, A.R. Virus neutralising activity of African fruit bat (Eidolon helvum) sera against emerging lyssaviruses. Virology 2010, 408, 183–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Benedictis, P.; Minola, A.; Rota Nodari, E.; Aiello, R.; Zecchin, B.; Salomoni, A.; Foglierini, M.; Agatic, G.; Vanzetta, F.; Lavenir, R.; et al. Development of broad-spectrum human monoclonal antibodies for rabies post-exposure prophylaxis. EMBO Mol. Med. 2016, 8, 407–421. [Google Scholar] [CrossRef] [PubMed]
- Both, L.; Dolleweerd, C.; Wright, E.; Banyard, A.C.; Bulmer-Thomas, B.; Selden, D.; Altmann, F.; Fooks, A.R.; Ma, J.K. Production, characterization, and antigen specificity of recombinant 62-71-3, a candidate monoclonal antibody for rabies prophylaxis in humans. FASEB J. 2013, 27, 2055–2065. [Google Scholar] [CrossRef] [PubMed]
- Evans, J.S.; Selden, D.; Wu, G.; Wright, E.; Horton, D.L.; Fooks, A.R.; Banyard, A.C. Antigenic site changes in the rabies virus glycoprotein dictates functionality and neutralizing capability against divergent lyssaviruses. J. Gen. Virol. 2018, 99, 169–180. [Google Scholar] [CrossRef]
- Munis, A.M.; Tijani, M.; Hassall, M.; Mattiuzzo, G.; Collins, M.K.; Takeuchi, Y. Characterization of Antibody Interactions with the G Protein of Vesicular Stomatitis Virus Indiana Strain and Other Vesiculovirus G Proteins. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [Green Version]
- Akahata, W.; Yang, Z.Y.; Andersen, H.; Sun, S.; Holdaway, H.A.; Kong, W.P.; Lewis, M.G.; Higgs, S.; Rossmann, M.G.; Rao, S.; et al. A virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat. Med. 2010, 16, 334–338. [Google Scholar] [CrossRef] [Green Version]
- Henss, L.; Yue, C.; Kandler, J.; Faddy, H.M.; Simmons, G.; Panning, M.; Lewis-Ximenez, L.L.; Baylis, S.A.; Schnierle, B.S. Establishment of an alphavirus-specific neutralization assay to distinguish infections with different members of the semliki forest complex. Viruses 2019, 11, 82. [Google Scholar] [CrossRef] [Green Version]
- Klasse, P.J. Neutralization of Virus Infectivity by Antibodies: Old Problems in New Perspectives. Adv. Biol. 2014, 2014, 1–24. [Google Scholar] [CrossRef] [Green Version]
- Feldmann, H.; Jones, S.; Klenk, H.; Schnittler, H. Ebola Virus: From discovery to vaccine. Nat. Rev. 2003, 3, 677–685. [Google Scholar] [CrossRef]
- Lambe, T.; Bowyer, G.; Ewer, K. A review of Phase I trials of Ebolavirus vaccines: What can we learn from the race to develop novel vaccines? Philos. Trans. R. Soc. B 2017, 372, 20160295. [Google Scholar] [CrossRef] [Green Version]
- Ye, L.; Yang, C. Development of vaccines for prevention of Ebola virus infection. Microbes Infect. 2015, 17, 98–108. [Google Scholar] [CrossRef] [PubMed]
- Carnell, G.W.; Ferrara, F.; Grehan, K.; Thompson, C.P.; Temperton, N.J. Pseudotype-based neutralization assays for influenza: A systematic analysis. Front. Immunol. 2015, 6, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bentley, E.M.; Mather, S.T.; Temperton, N.J. The use of pseudotypes to study viruses, virus sero-epidemiology and vaccination. Vaccine 2015, 33, 2955–2962. [Google Scholar] [CrossRef] [PubMed]
- Stephenson, I.; Das, R.G.; Wood, J.M.; Katz, J.M. Comparison of neutralising antibody assays for detection of antibody to influenza A/H3N2 viruses: An international collaborative study. Vaccine 2007, 25, 4056–4063. [Google Scholar] [CrossRef] [PubMed]
- Alberini, I.; Del Tordello, E.; Fasolo, A.; Temperton, N.J.; Galli, G.; Gentile, C.; Montomoli, E.; Hilbert, A.K.; Banzhoff, A.; Del Giudice, G.; et al. Pseudoparticle neutralization is a reliable assay to measure immunity and cross-reactivity to H5N1 influenza viruses. Vaccine 2009, 27, 5998–6003. [Google Scholar] [CrossRef]
- Prevato, M.; Cozzi, R.; Pezzicoli, A.; Taddei, A.R.; Ferlenghi, I.; Nandi, A.; Montomoli, E.; Settembre, E.C.; Bertholet, S.; Bonci, A.; et al. An innovative pseudotypes-based Enzyme-Linked Lectin Assay for the measurement of functional anti-neuraminidase antibodies. PLoS ONE 2015, 10, e0135383. [Google Scholar] [CrossRef]
- Biuso, F.; Palladino, L.; Manenti, A.; Stanzani, V.; Lapini, G.; Gao, J.; Couzens, L.; Eichelberger, M.C.; Montomoli, E. Use of lentiviral pseudotypes as an alternative to reassortant or Triton X-100-treated wild-type Influenza viruses in the neuraminidase inhibition enzyme-linked lectin assay. Influenza Other Respi. Viruses 2019, 13, 504–516. [Google Scholar] [CrossRef] [Green Version]
- Chigbu, D.I.; Loonawat, R.; Sehgal, M.; Patel, D.; Jain, P. Hepatitis C Virus Infection: Host–Virus Interaction and Mechanisms of Viral Persistence. Cells 2019, 8, 376. [Google Scholar] [CrossRef] [Green Version]
- Burm, R.; Collignon, L.; Mesalam, A.A.; Meuleman, P. Animal models to study hepatitis C virus infection. Front. Immunol. 2018. [Google Scholar] [CrossRef]
- Steffen, I.; Lu, K.; Hoff, N.A.; Mulembakani, P.; Okitolonda Wemakoy, E.; Muyembe-Tamfum, J.J.; Ndembi, N.; Brennan, C.A.; Hackett, J.; Switzer, W.M.; et al. Seroreactivity against Marburg or related filoviruses in West and Central Africa. Emerg. Microbes Infect. 2020, 9, 124–128. [Google Scholar] [CrossRef] [Green Version]
- Molesti, E.; Wright, E.; Terregino, C.; Rahman, R.; Cattoli, G.; Temperton, N.J. Multiplex Evaluation of Influenza Neutralizing Antibodies with Potential Applicability to In-Field Serological Studies. J. Immunol. Res. 2014, 2014. [Google Scholar] [CrossRef] [PubMed]
- WHO Director-General Summarizes the Outcome of the Emergency Committee Regarding Clusters of Microcephaly and Guillain-Barré Syndrome. Available online: https://www.who.int/news/item/01-02-2016-who-director-general-summarizes-the-outcome-of-the-emergency-committee-regarding-clusters-of-microcephaly-and-guillain-barré-syndrome (accessed on 19 December 2020).
- Prat, C.M.; Flusin, O.; Panella, A.; Tenebray, B.; Lanciotti, R.; Leparc-Goffart, I. Evaluation of commercially available serologic diagnostic tests for chikungunya virus. Emerg. Infect. Dis. 2014, 20, 2129–2132. [Google Scholar] [CrossRef] [PubMed]
- Partidos, C.D.; Paykel, J.; Weger, J.; Borland, E.M.; Powers, A.M.; Seymour, R.; Weaver, S.C.; Stinchcomb, D.T.; Osorio, J.E. Cross-protective immunity against o’nyong-nyong virus afforded by a novel recombinant chikungunya vaccine. Vaccine 2012, 30, 4638–4643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Page, K.A.; Landau, N.R.; Littman, D.R. Construction and use of a human immunodeficiency virus vector for analysis of virus infectivity. J. Virol. 1990, 64, 5270–5276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kishishita, N.; Takeda, N.; Anuegoonpipat, A.; Anantapreecha, S. Development of a pseudotyped-lentiviral-vector-based neutralization assay for chikungunya virus infection. J. Clin. Microbiol. 2013, 51, 1389–1395. [Google Scholar] [CrossRef] [Green Version]
- Wei, C.J.; Boyington, J.C.; Dai, K.; Houser, K.V.; Pearce, M.B.; Kong, W.P.; Yang, Z.Y.; Tumpey, T.M.; Nabel, G.J. Cross-neutralization of 1918 and 2009 influenza viruses: Role of glycans in viral evolution and vaccine design. Sci. Transl. Med. 2010, 2, 24ra21. [Google Scholar] [CrossRef] [Green Version]
- Bender, R.R.; Muth, A.; Schneider, I.C.; Friedel, T.; Hartmann, J.; Plückthun, A.; Maisner, A.; Buchholz, C.J. Receptor-Targeted Nipah Virus Glycoproteins Improve Cell-Type Selective Gene Delivery and Reveal a Preference for Membrane-Proximal Cell Attachment. PLoS Pathog. 2016, 12, e1005641. [Google Scholar] [CrossRef]
- Wang, J.; Cheng, H.; Ratia, K.; Varhegyi, E.; Hendrickson, W.G.; Li, J.; Rong, L. A Comparative High-Throughput Screening Protocol to Identify Entry Inhibitors of Enveloped Viruses. J. Biomol. Screen. 2014, 19, 100–107. [Google Scholar] [CrossRef] [Green Version]
- Hu, S.; Li, M.; Akkina, R. Generation of high-titer pseudotyped lentiviral vectors. Methods Mol. Biol. 2019, 1937, 125–134. [Google Scholar] [CrossRef]
- Kumar, M.; Keller, B.; Makalou, N.; Sutton, R.E. Systematic determination of the packaging limit of lentiviral vectors. Hum. Gene Ther. 2001, 12, 1893–1905. [Google Scholar] [CrossRef]
- Jiang, W.; Hua, R.; Wei, M.; Li, C.; Qiu, Z.; Yang, X.; Zhang, C. An optimized method for high-titer lentivirus preparations without ultracentrifugation. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Addgene: Lentivirus Production Protocol. Available online: https://www.addgene.org/protocols/lentivirus-production/ (accessed on 19 December 2020).
- Greg’s RetroPrep in 293T cells by Fugene-6 Transfection. Available online: https://www.ucl.ac.uk/towers-lab/sites/towers-lab/files/gregsretroprep.pdf (accessed on 19 December 2020).
- Storck, A.; Ludtke, J.; Kopp, L.; Juckem, L. Development and optimization of a high titer recombinant lentivirus system. Biotechniques 2017, 63, 136–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grehan, K.; Ferrara, F.; Temperton, N. An optimised method for the production of MERS-CoV spike expressing viral pseudotypes. MethodsX 2015, 2, 379–384. [Google Scholar] [CrossRef] [PubMed]
- Carnell, G.; Grehan, K.; Ferrara, F.; Molesti, E.; Temperton, N. An Optimized Method for the Production Using PEI, Titration and Neutralization of SARS-CoV Spike Luciferase Pseudotypes. Bio-Protocol 2017, 7. [Google Scholar] [CrossRef] [Green Version]
- Crawford, K.H.D.; Eguia, R.; Dingens, A.S.; Loes, A.N.; Malone, K.D.; Wolf, C.R.; Chu, H.Y.; Tortorici, M.A.; Veesler, D.; Murphy, M.; et al. Protocol and Reagents for Pseudotyping Lentiviral Particles with SARS-CoV-2 Spike Protein for Neutralization Assays. Viruses 2020, 12, 513. [Google Scholar] [CrossRef]
- Johnson, M.C.; Lyddon, T.D.; Suarez, R.; Salcedo, B.; LePique, M.; Graham, M.; Ricana, C.; Robinson, C.; Ritter, D.G. Optimized Pseudotyping Conditions for the SARS-COV-2 Spike Glycoprotein. J. Virol. 2020, 94. [Google Scholar] [CrossRef]
- Schmidt, F.; Weisblum, Y.; Muecksch, F.; Hoffmann, H.H.; Michailidis, E.; Lorenzi, J.C.C.; Mendoza, P.; Rutkowska, M.; Bednarski, E.; Gaebler, C.; et al. Measuring SARS-CoV-2 neutralizing antibody activity using pseudotyped and chimeric viruses. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef]
- Khetawat, D.; Broder, C.C. A functional henipavirus envelope glycoprotein pseudotyped lentivirus assay system. Virol. J. 2010, 7, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Frecha, C.; Costa, C.; Nègre, D.; Gauthier, E.; Russell, S.J.; Cosset, F.-L.; Verhoeyen, E. Stable transduction of quiescent T cells without induction of cycle progression by a novel lentiviral vector pseudotyped with measles virus glycoproteins. Blood 2008, 112, 4843–4852. [Google Scholar] [CrossRef] [Green Version]
- Carpentier, D.C.J.; Vevis, K.; Trabalza, A.; Georgiadis, C.; Ellison, S.M.; Asfahani, R.I.; Mazarakis, N.D. Enhanced pseudotyping efficiency of HIV-1 lentiviral vectors by a rabies/vesicular stomatitis virus chimeric envelope glycoprotein. Gene Ther. 2011, 19, 761–774. [Google Scholar] [CrossRef]
- Sandrin, V.; Boson, B.; Salmon, P.; Gay, W.; Nègre, D.; Le Grand, R.; Trono, D.; Cosset, F.L. Lentiviral vectors pseudotyped with a modified RD114 envelope glycoprotein show increased stability in sera and augmented transduction of primary lymphocytes and CD34+ cells derived from human and nonhuman primates. Blood 2002, 100, 823–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chojnacki, J.; Staudt, T.; Glass, B.; Bingen, P.; Engelhardt, J.; Anders, M.; Schneider, J.; Müller, B.; Hell, S.W.; Kräusslich, H.-G. Maturation-dependent HIV-1 surface protein redistribution revealed by fluorescence nanoscopy. Science 2012, 338, 524–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohan, G.S.; Ye, L.; Li, W.; Monteiro, A.; Lin, X.; Sapkota, B.; Pollack, B.P.; Compans, R.W.; Yang, C. Less is More: Ebola Surface Glycoprotein Expression Levels Regulate Virus Production and Infectivity. J. Virol. 2015, 89, 1205–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferrara, F.; Molesti, E.; Böttcher-Friebertshäuser, E.; Cattoli, G.; Corti, D.; Scott, S.D.; Temperton, N.J. The human Transmembrane Protease Serine 2 is necessary for the production of Group 2 influenza A virus pseudotypes. J. Mol. Genet. Med. 2012, 7, 309–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Temperton, N.J.; Wright, E.; Scott, S.D.; Temperton, N.J.; Wright, E.; Scott, S.D. Retroviral Pseudotypes—From Scientific Tools to Clinical Utility. eLS 2015, 1–11. [Google Scholar] [CrossRef]
- Condit, R.C. Principles of Virology. In Fields Virology; Knipe, D.M., Howley, P.M., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2001; pp. 19–51. [Google Scholar]
- Reed, L.; Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Hyg. 1938, 27, 493–497. [Google Scholar] [CrossRef]
- Zufferey, R.; Dull, T.; Mandel, R.J.; Bukovsky, A.; Quiroz, D.; Naldini, L.; Trono, D. Self-inactivating lentivirus vector for safe and efficient in vivo gene delivery. J. Virol. 1998, 72, 9873–9880. [Google Scholar] [CrossRef] [Green Version]
- Dautzenberg, I.J.C.; Rabelink, M.J.W.E.; Hoeben, R.C. The stability of envelope-pseudotyped lentiviral vectors. Gene Ther. 2020. [Google Scholar] [CrossRef]
- Pizzato, M.; Erlwein, O.; Bonsall, D.; Kaye, S.; Muir, D.; McClure, M.O. A one-step SYBR Green I-based product-enhanced reverse transcriptase assay for the quantitation of retroviruses in cell culture supernatants. J. Virol. Methods 2009, 156, 1–7. [Google Scholar] [CrossRef]
- Vermeire, J.; Naessens, E.; Vanderstraeten, H.; Landi, A.; Iannucci, V.; Van Nuffel, A.; Taghon, T.; Pizzato, M.; Verhasselt, B. Quantification of Reverse Transcriptase Activity by Real-Time PCR as a Fast and Accurate Method for Titration of HIV, Lenti- and Retroviral Vectors. PLoS ONE 2012, 7, e50859. [Google Scholar] [CrossRef]
- Ferrer, M.; Henriet, S.; Chamontin, C.; Lainé, S.; Mougel, M. From cells to virus particles: Quantitative methods to monitor RNA packaging. Viruses 2016, 8, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vamva, E.; Lever, A.M.L.; Vink, C.A.; Kenyon, J.C. Development of a Novel Competitive qRT-PCR Assay to Measure Relative Lentiviral Packaging Efficiency. Mol. Ther. Methods Clin. Dev. 2020, 19, 307–319. [Google Scholar] [CrossRef]
- Filipe, V.; Hawe, A.; Jiskoot, W. Critical evaluation of nanoparticle tracking analysis (NTA) by NanoSight for the measurement of nanoparticles and protein aggregates. Pharm. Res. 2010, 27, 796–810. [Google Scholar] [CrossRef] [Green Version]
- Heider, S.; Metzner, C. Quantitative real-time single particle analysis of virions. Virology 2014, 462–463, 199–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rossi, C.A.; Kearney, B.J.; Olschner, S.P.; Williams, P.L.; Robinson, C.G.; Heinrich, M.L.; Zovanyi, A.M.; Ingram, M.F.; Norwood, D.A.; Schoepp, R.J. Evaluation of ViroCyt® Virus Counter for rapid filovirus quantitation. Viruses 2015, 7, 857–872. [Google Scholar] [CrossRef] [Green Version]
- Geraerts, M.; Willems, S.; Baekelandt, V.; Debyser, Z.; Gijsbers, R. Comparison of lentiviral vector titration methods. BMC Biotechnol. 2006, 6, 34. [Google Scholar] [CrossRef] [Green Version]
- Lizée, G.; Aerts, J.L.; Gonzales, M.I.; Chinnasamy, N.; Morgan, R.A.; Topalian, S.L. Real-time quantitative reverse transcriptase-polymerase chain reaction as a method for determining lentiviral vector titers and measuring transgene expression. Hum. Gene Ther. 2003, 14, 497–507. [Google Scholar] [CrossRef] [PubMed]
- Sastry, L.; Johnson, T.; Hobson, M.J.; Smucker, B.; Cornetta, K. Titering lentiviral vectors: Comparison of DNA, RNA and marker expression methods. Gene Ther. 2002, 9, 1155–1162. [Google Scholar] [CrossRef] [Green Version]
- Scherr, M.; Battmer, K.; Blömer, U.; Ganser, A.; Grez, M. Quantitative determination of lentiviral vector particle numbers by real-time PCR. Biotechniques 2001, 31, 520–526. [Google Scholar] [CrossRef] [Green Version]
- Committee for Medicinal Products For Human Use (CHMP) Guideline on Development and Manufacture of Lentiviral Vectors Discussion in Biotechnology Working Party. Available online: http://www.emea.eu.int (accessed on 21 December 2020).
- FDA Testing of Retroviral Vector-Based Human Gene Therapy Products for Replication Competent Retrovirus during Product Manufacture and Patient Follow-up. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/testing-retroviral-vector-based-human-gene-therapy-products-replication-competent-retrovirus-during (accessed on 21 December 2020).
- Escarpe, P.; Zayek, N.; Chin, P.; Borellini, F.; Zufferey, R.; Veres, G.; Kiermer, V. Development of a sensitive assay for detection of replication-competent recombinant lentivirus in large-scale HIV-based vector preparations. Mol. Ther. 2003, 8, 332–341. [Google Scholar] [CrossRef]
- Sastry, L.; Cornetta, K. Detection of replication competent retrovirus and lentivirus. Methods Mol. Biol. 2009, 506, 243–263. [Google Scholar] [CrossRef]
- Gándara, C.; Affleck, V.; Stoll, E.A. Manufacture of Third-Generation Lentivirus for Preclinical Use, with Process Development Considerations for Translation to Good Manufacturing Practice. Hum. Gene Ther. Methods 2018, 29, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baylor College of Medicine Standard Operating Procedures. Available online: https://www.bcm.edu/research/research-services/service-labs/vector-development/technical-information/standard-operating-procedures (accessed on 21 December 2020).
- Valkama, A.J.; Oruetxebarria, I.; Lipponen, E.M.; Leinonen, H.M.; Käyhty, P.; Hynynen, H.; Turkki, V.; Malinen, J.; Miinalainen, T.; Heikura, T.; et al. Development of Large-Scale Downstream Processing for Lentiviral Vectors. Mol. Ther. Methods Clin. Dev. 2020, 17, 717–730. [Google Scholar] [CrossRef] [PubMed]
- Martínez-Molina, E.; Chocarro-Wrona, C.; Martínez-Moreno, D.; Marchal, J.A.; Boulaiz, H. Large-Scale Production of Lentiviral Vectors: Current Perspectives and Challenges. Pharmaceutics 2020, 12, 1051. [Google Scholar] [CrossRef] [PubMed]
- European Medicines Agency Reflection paper for laboratories that perform the analysis or evaluation of clinical trial samples. Available online: https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/reflection-paper-laboratories-perform-analysis-evaluation-clinical-trial-samples_en.pdf (accessed on 21 December 2020).
- World Health Organisation Medical Device Regulations: Global overview and guiding principles. Available online: https://apps.who.int/iris/handle/10665/42744 (accessed on 21 December 2020).
- Zhao, Y.; Stepto, H.; Schneider, C.K. Development of the First World Health Organization Lentiviral Vector Standard: Toward the Production Control and Standardization of Lentivirus-Based Gene Therapy Products. Hum. Gene Ther. Methods 2017, 28, 205–214. [Google Scholar] [CrossRef] [PubMed]
- World Health Organisation WHO Expert Committee on Biological Standardization Seventieth Report. Available online: https://apps.who.int/iris/handle/10665/332102 (accessed on 21 December 2020).
- World Health Organisation WHO Expert Committee on Biological Standardization: Fifty-Fifth Report. Available online: https://apps.who.int/iris/handle/10665/43278 (accessed on 21 December 2020).
- Delacroix, N.; Ouengue Mbele, G.; Deroyer, J.; Deplaine, G.; Bauche, C. 671. Development of a Successful Lyophilization Process for Lentiviral Vector Clinical Batches. Mol. Ther. 2015, 23, S267. [Google Scholar] [CrossRef] [Green Version]
- Mather, S.T.; Wright, E.; Scott, S.D.; Temperton, N.J. Lyophilisation of influenza, rabies and Marburg lentiviral pseudotype viruses for the development and distribution of a neutralisation -assay-based diagnostic kit. J. Virol. Methods 2014, 210, 51–58. [Google Scholar] [CrossRef]
- Hyseni, I.; Molesti, E.; Benincasa, L.; Piu, P.; Casa, E.; Temperton, N.J.; Manenti, A.; Montomoli, E. Characterisation of SARS-CoV-2 Lentiviral Pseudotypes and Correlation between Pseudotype-Based Neutralisation Assays and Live Virus-Based Micro Neutralisation Assays. Viruses 2020, 12, 1011. [Google Scholar] [CrossRef]
- Qiu, C.; Huang, Y.; Wang, Q.; Tian, D.; Zhang, W.; Hu, Y.; Yuan, Z.; Zhang, X.; Xu, J. Boosting heterosubtypic neutralization antibodies in recipients of 2009 pandemic H1N1 influenza vaccine. Clin. Infect. Dis. 2012, 54, 17–24. [Google Scholar] [CrossRef]
- Steeds, K.; Hall, Y.; Slack, G.S.; Longet, S.; Strecker, T.; Fehling, S.K.; Wright, E.; Bore, J.A.; Koundouno, F.R.; Konde, M.K.; et al. Pseudotyping of VSV with Ebola virus glycoprotein is superior to HIV-1 for the assessment of neutralising antibodies. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef]
- Wilkinson, D.E.; Page, M.; Mattiuzzo, G.; Hassall, M.; Dougall, T.; Rigsby, P.; Stone, L.; Minor, P. Comparison of platform technologies for assaying antibody to Ebola virus. Vaccine 2017, 35, 1347–1352. [Google Scholar] [CrossRef] [PubMed]
- Jayakar, H.R.; Jeetendra, E.; Whitt, M.A. Rhabdovirus assembly and budding. Virus Res. 2004, 106, 117–132. [Google Scholar] [CrossRef] [PubMed]
- Stremlau, M.; Owens, C.M.; Perron, M.J.; Kiessling, M.; Autissier, P.; Sodroski, J. The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys. Nature 2004, 427, 848–853. [Google Scholar] [CrossRef] [PubMed]
- Cavrois, M.; Neidleman, J.; Greene, W.C. HIV-1 Fusion Assay. Bio-Protocol 2014, 4. [Google Scholar] [CrossRef]
- Sandrin, V.; Muriaux, D.; Darlix, J.; Inserm, U. Intracellular Trafficking of Gag and Env Proteins and Their Interactions Modulate Pseudotyping of Retroviruses. J. Virol. 2004, 78, 7153–7164. [Google Scholar] [CrossRef] [Green Version]
- Sandrin, V.; Cosset, F.-L. Intracellular versus cell surface assembly of retroviral pseudotypes is determined by the cellular localization of the viral glycoprotein, its capacity to interact with Gag, and the expression of the Nef protein. J. Biol. Chem. 2006, 281, 528–542. [Google Scholar] [CrossRef] [Green Version]
- Xiao, J.; Rijal, P.; Schimanski, L.; Tharkeshwar, A.K.; Wright, E.; Annaert, W.; Townsend, A. Characterization of an influenza virus pseudotyped with Ebolavirus glycoprotein. J. Virol. 2017, 92. [Google Scholar] [CrossRef] [Green Version]
- Pierson, T.C.; Sánchez, M.D.; Puffer, B.A.; Ahmed, A.A.; Geiss, B.J.; Valentine, L.E.; Altamura, L.A.; Diamond, M.S.; Doms, R.W. A rapid and quantitative assay for measuring antibody-mediated neutralization of West Nile virus infection. Virology 2006, 346, 53–65. [Google Scholar] [CrossRef] [Green Version]
- Ansarah-Sobrinho, C.; Nelson, S.; Jost, C.A.; Whitehead, S.S.; Pierson, T.C. Temperature-dependent production of pseudoinfectious dengue reporter virus particles by complementation. Virology 2008, 381, 67–74. [Google Scholar] [CrossRef] [Green Version]
- Matsuda, M.; Yamanaka, A.; Yato, K.; Yoshii, K.; Watashi, K.; Aizaki, H.; Konishi, E.; Takasaki, T.; Kato, T.; Muramatsu, M.; et al. High-throughput neutralization assay for multiple flaviviruses based on single-round infectious particles using dengue virus type 1 reporter replicon. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef]
Virus Family | Virus Envelope Protein | Serological Application |
---|---|---|
Arenaviridae | Lassa fever virus G-protein | Vaccine Efficacy [21] Therapeutic Antivirals [22] |
Coronaviridae | Middle East respiratory syndrome coronavirus S-protein | Vaccine Efficacy [23,24,25] Serosurveillance [26,27,28] |
Severe acute respiratory syndrome coronavirus S-protein | Vaccine Efficacy [29,30] Therapeutic Antivirals [31] | |
Sever acute respiratory syndrome coronavirus-2 S-protein | Vaccine Efficacy [32,33,34] Serosurveillance [35,36,37,38,39,40,41,42,43,44,45] Therapeutic Antivirals [46,47,48] | |
Filoviridae | Ebola virus G-protein | Vaccine Efficacy [49] Serosurveillance [50,51,52] |
Ebola, Sudan, Bundibugyo, Reston virus G-protein | Cross-Reactivity [53] | |
Ebola, Marburg virus G-protein | Therapeutic Antivirals [22,54] | |
Flaviviridae | Hepatitis C virus E1/E2-protein | Vaccine Efficacy [55] Serosurveillance [56] Therapeutic Antivirals [57] |
Orthomyxoviridae | Influenza A virus HA and NA-protein | Vaccine Efficacy [58,59,60,61,62,63,64,65,66,67,68,69] Serosurveillance [70,71,72,73,74] Therapeutic Antivirals [54] |
Paramyxoviridae | Measles virus H-protein | Antigenic Site Analysis [75,76] |
Retroviridae | Human immunodeficiency virus 1 G-protein | Therapeutic Antivirals [77,78,79] |
Rhabdoviridae | Rabies, European bat 1, European bat 2 lyssavirus G-protein | Vaccine Efficacy [80] |
Rabies, Lagos bat, Duvenhage, Mokola, West Caucasian bat lyssavirus G-protein | Serosurveillance [81,82] | |
Rabies lyssavirus, vesicular stomatitis virus G-protein | Antigenic Site Analysis [83,84,85,86] | |
Togaviridae | Chikungunya virus E1/E2-protein | Vaccine Efficacy [87] |
Chikungunya, Mayaro, Ross River, Semliki Forest, Barmah Forest, O’nyong nyong virus E1/E2-protein | Cross-Reactivity [88] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Toon, K.; Bentley, E.M.; Mattiuzzo, G. More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation. Viruses 2021, 13, 217. https://doi.org/10.3390/v13020217
Toon K, Bentley EM, Mattiuzzo G. More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation. Viruses. 2021; 13(2):217. https://doi.org/10.3390/v13020217
Chicago/Turabian StyleToon, Kamilla, Emma M. Bentley, and Giada Mattiuzzo. 2021. "More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation" Viruses 13, no. 2: 217. https://doi.org/10.3390/v13020217
APA StyleToon, K., Bentley, E. M., & Mattiuzzo, G. (2021). More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation. Viruses, 13(2), 217. https://doi.org/10.3390/v13020217