Development of Lentiviral Vectors Pseudotyped With Influenza B Hemagglutinins: Application in Vaccine Immunogenicity, mAb Potency, and Sero-Surveillance Studies

Francesca Ferrara, Joanne Marie M Del Rosario, Kelly A S da Costa, Rebecca Kinsley, Simon Scott, Sasan Fereidouni, Craig Thompson, Paul Kellam, Sarah Gilbert, George Carnell, Nigel Temperton, Francesca Ferrara, Joanne Marie M Del Rosario, Kelly A S da Costa, Rebecca Kinsley, Simon Scott, Sasan Fereidouni, Craig Thompson, Paul Kellam, Sarah Gilbert, George Carnell, Nigel Temperton

Abstract

Influenza B viruses (IBV) cause respiratory disease epidemics in humans and are therefore components of seasonal influenza vaccines. Serological methods are employed to evaluate vaccine immunogenicity prior to licensure. However, classical methods to assess influenza vaccine immunogenicity such as the hemagglutination inhibition assay (HI) and the serial radial hemolysis assay (SRH), have been proven to have many limitations. As such, there is a need to develop innovative methods that can improve on these traditional assays and provide advantages such as ease of production and access, safety, reproducibility, and specificity. It has been previously demonstrated that the use of replication-defective viruses, such as lentiviral vectors pseudotyped with influenza A hemagglutinins in microneutralization assays (pMN) is a safe and sensitive alternative to study antibody responses elicited by natural influenza infection or vaccination. Consequently, we have produced Influenza B hemagglutinin-pseudotypes (IBV PV) using plasmid-directed transfection. To activate influenza B hemagglutinin, we have explored the use of proteases in increasing PV titers via their co-transfection during pseudotype virus production. When tested for their ability to transduce target cells, the influenza B pseudotypes produced exhibit tropism for different cell lines. The pseudotypes were evaluated as alternatives to live virus in microneutralization assays using reference sera standards, mouse and human sera collected during vaccine immunogenicity studies, surveillance sera from seals, and monoclonal antibodies (mAbs) against IBV. The influenza B pseudotype pMN was found to effectively detect neutralizing and cross-reactive responses in all assays and shows promise as an effective and versatile tool in influenza research.

Trial registration: ClinicalTrials.gov NCT00942071.

Keywords: influenza B; lentiviral (LV) vector; pseudotype neutralization; pseudotype viral particles; serosurveillance.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Ferrara, Del Rosario, da Costa, Kinsley, Scott, Fereidouni, Thompson, Kellam, Gilbert, Carnell and Temperton.

Figures

Figure 1
Figure 1
Phylogenetic tree of influenza B HA used for pseudotype production. Phylogenetic tree shows influenza B diverging into two distinct lineages, B/Yamagata (blue) and B/Victoria (red). Accession numbers are reported with the strain name on the tree tips. Nodes are shown at the ends of branches which represent sequences or hypothetical sequences at various points in evolutionary history. Branch lengths indicate the extent of genetic change. The tree generated was constructed with PhyML (48) and graphically elaborated with Archaeopteryx.js (https://sites.google.com/site/cmzmasek/home/software/archaeopteryx-js).
Figure 2
Figure 2
Production of influenza B pseudotypes employing different proteases. Various quantities of proteases were co-transfected with gag-pol packaging construct, firefly luciferase and influenza B HA-encoding plasmids in pI.18 (A) or pEVAC (B). (A) For transfection employing pI.18 HA plasmids, HAT, TMPRSS2 and TMPRSS4 were used in 250 and 125 ng quantities. Pseudotypes produced in the absence of protease (Δprotease) and those activated post-transfection and pre-titration with TPCK-Trypsin are also included. Plates were read using the Bright-Glo™ assay system (Promega) and GloMax Multi detection system luminometer (Promega). (B) For transfection employing pEVAC plasmids, only HAT and TMPRSS4 proteases in the range of 2.5 ng-10 ng were used. Titration plates were then read using the GloMax® Navigator (ProMega) using the Promega GloMax® Luminescence Quick-Read protocol. For both (A, B), pseudotype virus titers are expressed in relative luminescence units/mL (RLU/mL). Significant differences between titers obtained for each PV grouped into protease used and Δprotease are shown via brackets (*p=<0.05). Error bars show mean and standard deviation of four replicates.
Figure 3
Figure 3
Influenza B pseudotypes are expressed in different cell lines. (A) Titers of influenza B pseudotypes expressing firefly luciferase in HEK293T/17, MDCK, and A549 cells are shown in RLU/mL as the mean and standard deviation of 4 replicates. A ΔHA control representing non-HA pseudotyped virus was included for comparisons. No significant difference between PV titers and ΔHA is indicated by brackets (n.s. p>0.05). (B) Influenza B lentiviral pseudotype B/Florida/4/2006 containing an emerald GFP reporter is shown to successfully transduce HEK293T/17, MDCK and A549 cells. Scale bar = 6 µm.
Figure 4
Figure 4
Neutralization of influenza B pseudotypes by reference antisera and mouse sera from influenza vaccination studies. (A) Neutralization of pseudotypes from pI.18, B/Brisbane/60/2008, B/Florida/4/2006, and B/Hong Kong/8/1973 against reference antisera anti-B/Brisbane/60/2008 (NIBSC 11/36) was measured by a luciferase reporter assay. Reference sera was serially diluted two-fold from a starting dilution of 1:100. 1.0x106 RLU of IBV PV was then added to each well. For all plots, each point represents the mean and standard error of four replicates per dilution. (B)In vitro neutralization of influenza B pseudotypes from pEVAC, B/Phuket/3073/2013 and B/Washington/2/2019 by mouse sera vaccinated with Influenza B HA from B/Singapore/222/1972, B/Colorado/6/2017, B/Phuket/3073/2013, and PBS (negative control). Neutralization was determined via a luciferase-reporter assay and given as IC50 dilution values (In this case, IC50 is half maximal inhibitory serum dilution). Significant difference between the means of the IC50 values of the groups vaccinated with IBV HA against the PBS negative control group are shown via brackets (*p<0.05). Interleaved box plots show minimum, maximum, and mean values. Error bars show standard deviation of replicates. For mice vaccinated with B/Singapore/222/1972 HA, n = 5 mice/group; n = 6 mice/group for mice vaccinated with B/Colorado/6/2017 HA, B/Phuket/3073/2013 HA, and PBS.
Figure 5
Figure 5
Evaluation of IBV neutralization capacity of NCT0942071 clinical trial sera using influenza B pseudotype microneutralization assay (pMN). The neutralization capacity of human sera from clinical trial (NCT0942071) was assessed pre- and post-vaccination with Trivalent Influenza Vaccine (TIV) and then either a placebo saline solution or an MVA-NP fusion protein (n=17). All sera were serially diluted and incubated with each PV: (i) B/Brisbane/60/2008, (ii) B/Hong Kong/8/1973, and (iii) B/Florida/4/2006 PV before addition of HEK293T/17 cells. Residual luciferase activity was determined and % neutralization was calculated relative to the PV only control. From this, IC50 values were calculated as the reciprocal of the dilution required to neutralize 50% of initial input PV. Significant differences between the means of pre- and post-vaccination sera IC50 when tested against PV are indicated in brackets (*p(A). After data stratification on the basis of post-TIV vaccine regimens; TIV + placebo (n=8), TIV+MVA+NP+M1 (n=9), the fold-increase from pre-vaccination IC50 titers were compared to post-vaccination IC50(B). Data is compared using Tukey plots showing the median of the 25th and 75th percentile with outliers plotted as those which exceeded 1.5x quartile range. Virus pseudotype from B/Victoria lineage is indicated in red and B/Yamagata lineage in blue.
Figure 6
Figure 6
Neutralization of influenza B strains by lineage-specific monoclonal antibodies. Neutralization of influenza B pseudotypes B/Phuket/3073/2013 and B/Brisbane/60/2008 by monoclonal antibodies 3E8, 2F11, and 5A1, was measured by a luciferase reporter assay. Monoclonal antibodies were serially diluted two-fold from a starting concentration ranging from 2000 ng/mL to 0.0625 ng/mL against all pseudotypes tested. Influenza B pseudotypes (1.0x106 RLU) were then added to each well. For all plots, each point represents the mean and standard error of two replicates per dilution.

References

    1. World Health Organization . Influenza Virus Infection in Humans (2014). Available at: .
    1. Petrova VN, Russell CA. The Evolution of Seasonal Influenza Viruses. Nat Rev Microbiol (2018) 16(1):47–60. 10.1038/nrmicro.2017.118
    1. Hause BM, Collin EA, Liu R, Huang B, Sheng Z, Lu W, et al. . Characterization of a Novel Influenza Virus in Cattle and Swine: Proposal for a New Genus in the Orthomyxoviridae Family. mBio (2014) 5(2):1–10. 10.1128/mBio.00031-14
    1. Tan J, Asthagiri Arunkumar G, Krammer F. Universal Influenza Virus Vaccines and Therapeutics: Where Do We Stand With Influenza B Virus? Curr Opin Immunol (2018) 53:45–50. 10.1016/j.coi.2018.04.002
    1. Osterhaus AD, Rimmelzwaan GF, Martina BE, Bestebroer TM, Fouchier RAM. Influenza B Virus in Seals. Sci (New York NY) (2000) 288(5468):1051 – 3. 10.1126/science.288.5468.1051
    1. Ran Z, Shen H, Lang Y, Kolb EA, Turan N, Zhu L, et al. . Domestic Pigs are Susceptible to Infection With Influenza B Viruses. J Virol (2015) 89(9):4818. 10.1128/JVI.00059-15
    1. Chen R, Holmes EC. The Evolutionary Dynamics of Human Influenza B Virus. J Mol Evol (2008) 66(6):655–63. 10.1007/s00239-008-9119-z
    1. Rota PA, Wallis TR, Harmon MW, Rota JS, Kendal AP, Nerome K. Cocirculation of Two Distinct Evolutionary Lineages of Influenza Type B Virus Since 1983. Virology (1990) 175(1):59–68. 10.1016/0042-6822(90)90186-U
    1. Caini S, Kusznierz G, Garate VV, Wangchuk S, Thapa B, de Paula Júnior FJ, et al. . The Epidemiological Signature of Influenza B Virus and Its B/Victoria and B/Yamagata Lineages in the 21st Century. PloS One (2019) 14(9):e0222381–e. 10.1371/journal.pone.0222381
    1. Skehel JJ, Wiley DC. Receptor Binding and Membrane Fusion in Virus Entry: The Influenza Hemagglutinin. Annu Rev Biochem (2000) 69(1):531–69. 10.1146/annurev.biochem.69.1.531
    1. Wiley DC, Skehel JJ. The Structure and Function of the Hemagglutinin Membrane Glycoprotein of Influenza Virus. Annu Rev Biochem (1987) 56(1):365–94. 10.1146/annurev.bi.56.070187.002053
    1. Varki A. Diversity in the Sialic Acids. Glycobiology (1992) 2(1):25–40. 10.1093/glycob/2.1.25
    1. Chernomordik LV, Frolov VA, Leikina E, Bronk P, Zimmerberg J. The Pathway of Membrane Fusion Catalyzed by Influenza Hemagglutinin: Restriction of Lipids, Hemifusion, and Lipidic Fusion Pore Formation. J Cell Biol (1998) 140(6):1369–82. 10.1083/jcb.140.6.1369
    1. Garten W, Klenk H. Cleavage Activation of the Influenza Virus Hemagglutinin and Its Role in Pathogenesis. .Monogr Virol (2008) 27:156–67. 10.1159/000151618
    1. Laporte M, Stevaert A, Raeymaekers V, Boogaerts T, Nehlmeier I, Chiu W, et al. . Hemagglutinin Cleavability, Acid-Stability and Temperature Dependence Optimize Influenza B Virus for Replication in Human Airways. J Virol (2019) 94(1):JVI.01430–19. 10.1128/JVI.01430-19
    1. Wang Y-F, Chang C-F, Chi C-Y, Wang H-C, Wang J-R, Su I-J. Characterization of Glycan Binding Specificities of Influenza B Viruses With Correlation With Hemagglutinin Genotypes and Clinical Features. J Med Virol (2012) 84(4):679–85. 10.1002/jmv.23219
    1. Klenk H-D, Garten W. Host Cell Proteases Controlling Virus Pathogenicity. Trends Microbiol (1994) 2(2):39–43. 10.1016/0966-842X(94)90123-6
    1. Glezen WP, Schmier JK, Kuehn CM, Ryan KJ, Oxford J. The Burden of Influenza B: A Structured Literature Review. Am J Public Health (2013) 103(3):e43–51. 10.2105/AJPH.2012.301137
    1. Dolin R. The Quadrivalent Approach to Influenza Vaccination. J Infect Dis (2013) 208(4):539–40. 10.1093/infdis/jit264
    1. Beran J, Peeters M, Dewé W, Raupachová J, Hobzová L, Devaster J-M. Immunogenicity and Safety of Quadrivalent Versus Trivalent Inactivated Influenza Vaccine: A Randomized, Controlled Trial in Adults. BMC Infect Dis (2013) 13(1):224. 10.1186/1471-2334-13-224
    1. Pepin S, Donazzolo Y, Jambrecina A, Salamand C, Saville M. Safety and Immunogenicity of a Quadrivalent Inactivated Influenza Vaccine in Adults. Vaccine (2013) 31(47):5572–8. 10.1016/j.vaccine.2013.08.069
    1. Tinoco JC, Pavia-Ruz N, Cruz-Valdez A, Doniz CA, Chandrasekaran V, Dewé W, et al. . Immunogenicity, Reactogenicity, and Safety of Inactivated Quadrivalent Influenza Vaccine Candidate Versus Inactivated Trivalent Influenza Vaccine in Healthy Adults Aged ≥18 Years: A Phase III, Randomized Trial. Vaccine (2014) 32(13):1480–7. 10.1016/j.vaccine.2014.01.022
    1. Eichner M, Schwehm M, Hain J, Uphoff H, Salzberger B, Knuf M, et al. . 4Flu - an Individual Based Simulation Tool to Study the Effects of Quadrivalent Vaccination on Seasonal Influenza in Germany. BMC Infect Dis (2014) 14(1):365. 10.1186/1471-2334-14-365
    1. World Health Organization . Manual for the Laboratory Diagnosis and Virological Surveillance of Influenza (2011). Available at: .
    1. Mancini G, Donatelli I, Arangio-Ruiz G, Rozera C, Macchia T. Comparison of Haemagglutination-Inhibition and Single Radial Haemolysis Techniques for Detecting Antibodies to Influenza A and B Viruses. J Hyg (1983) 91(1):157–62. 10.1017/S0022172400060137
    1. Trombetta CM, Remarque EJ, Mortier D, Montomoli E. Comparison of Hemagglutination Inhibition, Single Radial Hemolysis, Virus Neutralization Assays, and ELISA to Detect Antibody Levels Against Seasonal Influenza Viruses. Influenza Other Respir Viruses (2018) 12(6):675–86. 10.1111/irv.12591
    1. Wood JM, Gaines-Das RE, Taylor J, Chakraverty P. Comparison of Influenza Serological Techniques by International Collaborative Study. Vaccine (1994) 12(2):167–74. 10.1016/0264-410X(94)90056-6
    1. Kendal AP, Cate TR. Increased Sensitivity and Reduced Specificity of Hemagglutination Inhibition Tests With Ether-Treated Influenza B/Singapore/222/79. J Clin Microbiol (1983) 18(4):930. 10.1128/JCM.18.4.930-934.1983
    1. Monto AS, Maassab HF. Ether Treatment of Type B Influenza Virus Antigen for the Hemagglutination Inhibition Test. J Clin Microbiol (1981) 13(1):54. 10.1128/JCM.13.1.54-57.1981
    1. Pyhälä R, Kleemola M, Visakorpi R. The HI Test Modified by Ether Treatment in the Sero-Epidemiological Surveillance of Influenza B. J Hyg (1985) 94(03):341–8. 10.1017/S002217240006157X
    1. Ansaldi F, Bacilieri S, Amicizia D, Valle L, Banfi F, Durando P, et al. . Antigenic Characterisation of Influenza B Virus With a New Microneutralisation Assay: Comparison to Haemagglutination and Sequence Analysis. J Med Virol (2004) 74(1):141–6. 10.1002/jmv.20157
    1. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno A, Jarrossay D, et al. . A Neutralizing Antibody Selected From Plasma Cells That Binds to Group 1 and Group 2 Influenza A Hemagglutinins. Sci (New York NY) (2011) 333(6044):850–6. 10.1126/science.1205669
    1. Dreyfus C, Laursen NS, Kwaks T, Zuijdgeest D, Khayat R, Ekiert DC, et al. . Highly Conserved Protective Epitopes on Influenza B Viruses. Science (2012) 337(6100):1343–8. 10.1126/science.1222908
    1. Ekiert DC, Bhabha G, Elsliger MA, Friesen RHE, Jongeneelen M, Throsby M, et al. . Antibody Recognition of a Highly Conserved Influenza Virus Epitope. Science (2009) 324(5924):246–51. 10.1126/science.1171491
    1. Ekiert DC, Friesen RHE, Bhabha G, Kwaks T, Jongeneelen M, Yu W, et al. . A Highly Conserved Neutralizing Epitope on Group 2 Influenza A Viruses. Science (2011) 333(6044):843–50. 10.1126/science.1204839
    1. Sui J, Hwang WC, Perez S, Wei G, Aird D, Chen L-M, et al. . Structural and Functional Bases for Broad-Spectrum Neutralization of Avian and Human Influenza A Viruses. Nat Struct Mol Biol (2009) 16(3):265–73. 10.1038/nsmb.1566
    1. Krammer F, Palese P, Steel J. Advances in Universal Influenza Virus Vaccine Design and Antibody Mediated Therapies Based on Conserved Regions of the Hemagglutinin. In: Oldstone MBA, Compans RW, editors. Influenza Pathogenesis and Control vol. II. Cham: Springer International Publishing; (2015). p. 301–21.
    1. Nachbagauer R, Feser J, Naficy A, Bernstein DI, Guptill J, Walter EB, et al. . A Chimeric Hemagglutinin-Based Universal Influenza Virus Vaccine Approach Induces Broad and Long-Lasting Immunity in a Randomized, Placebo-Controlled Phase I Trial. Nat Med (2021) 27(1):106–14. 10.1038/s41591-020-1118-7
    1. Wiersma LCM, Rimmelzwaan GF, de Vries RD. Developing Universal Influenza Vaccines: Hitting the Nail, Not Just on the Head. Vaccines (2015) 3(2):239–62. 10.3390/vaccines3020239
    1. Yasugi M, Kubota-Koketsu R, Yamashita A, Kawashita N, Du A, Sasaki T, et al. . Human Monoclonal Antibodies Broadly Neutralizing Against Influenza B Virus. PloS Pathog (2013) 9(2):e1003150. 10.1371/journal.ppat.1003150
    1. Carnell GW, Ferrara F, Grehan K, Thompson CP, Temperton NJ. Pseudotype-Based Neutralization Assays for Influenza: A Systematic Analysis. Front Immunol (2015) 6:161. 10.3389/fimmu.2015.00161
    1. Del Rosario JMM, Smith M, Zaki K, Risley P, Temperton N, Engelhardt OG, et al. . Protection From Influenza by Intramuscular Gene Vector Delivery of a Broadly Neutralizing Nanobody Does Not Depend on Antibody Dependent Cellular Cytotoxicity. Front Immunol (2020) 11:627. 10.3389/fimmu.2020.00627
    1. Corti D, Suguitan Jr AL, Pinna D, Silacci C, Fernandez-Rodriguez MF, Vanzetta F, et al. . Heterosubtypic Neutralizing Antibodies are Produced by Individuals Immunized With a Seasonal Influenza Vaccine. J Clin Invest (2010) 120: (5):1663–73. 10.1172/JCI41902
    1. Liu L, Nachbagauer R, Zhu L, Huang Y, Xie X, Jin S, et al. . Induction of Broadly Cross-Reactive Stalk-Specific Antibody Responses to Influenza Group 1 and Group 2 Hemagglutinins by Natural H7n9 Virus Infection in Humans. J Infect Dis (2017) 215(4):518–28. 10.1093/infdis/jiw608
    1. Nachbagauer R, Salaun B, Stadlbauer D, Behzadi MA, Friel D, Rajabhathor A, et al. . Pandemic Influenza Virus Vaccines Boost Hemagglutinin Stalk-Specific Antibody Responses in Primed Adult and Pediatric Cohorts. NPJ Vaccines (2019) 4(1):51. 10.1038/s41541-019-0147-z
    1. Cox RJ, Mykkeltvedt E, Robertson J, Haaheim LR. Non-Lethal Viral Challenge of Influenza Haemagglutinin and Nucleoprotein Dna Vaccinated Mice Results in Reduced Viral Replication. Scandinavian J Immunol (2002) 55(1):14–23. 10.1046/j.1365-3083.2002.01015.x
    1. Temperton NJ, Hoschler K, Major D, Nicolson C, Manvell R, Hien VM, et al. . A Sensitive Retroviral Pseudotype Assay for Influenza H5N1-Neutralizing Antibodies. Influenza Other Respir Viruses (2007) 1(3):105–12. 10.1111/j.1750-2659.2007.00016.x
    1. Guindon S, Gascuel OA. Simple, Fast, and Accurate Algorithm to Estimate Large Phylogenies by Maximum Likelihood. Syst Biol (2003) 52(5):696–704. 10.1080/10635150390235520
    1. Böttcher E, Matrosovich T, Beyerle M, Klenk HD, Garten W, Matrosovich M. Proteolytic Activation of Influenza Viruses by Serine Proteases TMPRSS2 and HAT From Human Airway Epithelium. J Virol (2006) 80(19):9896–8. 10.1128/JVI.01118-06
    1. Bertram S, Glowacka I, Blazejewska P, Soilleux E, Allen P, Danisch S, et al. . TMPRSS2 and TMPRSS4 Facilitate Trypsin-Independent Spread of Influenza Virus in Caco-2 Cells. J Virol (2010) 84(19):10016–25. 10.1128/JVI.00239-10
    1. Naldini L, Blömer U, Gallay P, Ory D, Mulligan R, Gage FH, et al. . In Vivo Gene Delivery and Stable Transduction of Nondividing Cells by a Lentiviral Vector. Science (1996) 272(5259):263. 10.1126/science.272.5259.263
    1. Ferrara F, Temperton N. Pseudotype Neutralization Assays: From Laboratory Bench to Data Analysis. Methods Protoc (2018) 1(1):1–16. 10.3390/mps1010008
    1. Antrobus RD, Berthoud TK, Mullarkey CE, Hoschler K, Coughlan L, Zambon M, et al. . Coadministration of Seasonal Influenza Vaccine and MVA-NP+M1 Simultaneously Achieves Potent Humoral and Cell-Mediated Responses. Mol Ther (2013) 22(1):233–8. 10.1038/mt.2013.162
    1. Berthoud TK, Hamill M, Lillie PJ, Hwenda L, Collins KA, Ewer KJ, et al. . Potent CD8+ T-Cell Immunogenicity in Humans of a Novel Heterosubtypic Influenza A Vaccine, MVA-NP+M1. Clin Infect Dis (2011) 52(1):1–7. 10.1093/cid/ciq015
    1. Verma S, Soto J, Vasudevan A, Schmeisser F, Alvarado-Facundo E, Wang W, et al. . Determination of Influenza B Identity and Potency in Quadrivalent Inactivated Influenza Vaccines Using Lineage-Specific Monoclonal Antibodies. PloS One (2017) 12(4):e0175733–e. 10.1371/journal.pone.0175733
    1. Waterhouse AM, Procter JB, Martin DMA, Clamp M, Barton GJ. Jalview Version 2–a Multiple Sequence Alignment Editor and Analysis Workbench. Bioinf (Oxford England) (2009) 25(9):1189 – 91. 10.1093/bioinformatics/btp033
    1. Edgar RC. MUSCLE: Multiple Sequence Alignment With High Accuracy and High Throughput. Nucleic Acids Res (2004) 32(5):1792–7. 10.1093/nar/gkh340
    1. Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S. MEGA5: Molecular Evolutionary Genetics Analysis Using Maximum Likelihood, Evolutionary Distance, and Maximum Parsimony Methods. Mol Biol Evol (2011) 28(10):2731–9. 10.1093/molbev/msr121
    1. Ferrara F, Molesti E, Böttcher-Friebertshäuser E, Cattoli G, Corti D, Scott SD, et al. . The Human Transmembrane Protease Serine 2 Is Necessary for the Production of Group 2 Influenza A Virus Pseudotypes. J Mol Genet Med (2012) 7:309–14. 10.4172/1747-0862.1000055
    1. Brassard DL, Lamb RA. Expression of Influenza B Virus Hemagglutinin Containing Multibasic Residue Cleavage Sites. Virology (1997) 236(2):234–48. 10.1006/viro.1997.8749
    1. Stech J, Garn H, Herwig A, Stech O, Dauber B, Wolff T, et al. . Influenza B Virus With Modified Hemagglutinin Cleavage Site as a Novel Attenuated Live Vaccine. J Infect Dis (2011) 204(10):1483–90. 10.1093/infdis/jir613
    1. Carnell GW, Trombetta CM, Ferrara F, Montomoli E, Temperton NJ. Correlation of Influenza B Haemagglutination Inhibiton, Single-Radial Haemolysis and Pseudotype-Based Microneutralisation Assays for Immunogenicity Testing of Seasonal Vaccines. Vaccines (Basel) (2021) 9(2):1–18. 10.3390/vaccines9020100
    1. Das DK, Govindan R, Nikić-Spiegel I, Krammer F, Lemke EA, Munro JB. Direct Visualization of the Conformational Dynamics of Single Influenza Hemagglutinin Trimers. Cell (2018) 174(4):926–37.e12. 10.1016/j.cell.2018.05.050
    1. Zhirnov OP, Golyando PB, Ovcharenko AV. Replication of Influenza B Virus in Chicken Embryos Is Suppressed by Exogenous Aprotinin. Arch Virol (1994) 135(1-2):209–16. 10.1007/BF01309780
    1. Lugovtsev VY, Melnyk D, Weir JP. Heterogeneity of the MDCK Cell Line and Its Applicability for Influenza Virus Research. PloS One (2013) 8(9):e75014. 10.1371/journal.pone.0075014
    1. Noma K, Kiyotani K, Kouchi H, Fujii Y, Egi Y, Tanaka K, et al. . Endogenous Protease-Dependent Replication of Human Influenza Viruses in Two MDCK Cell Lines. Arch Virol (1998) 143(10):1893–909. 10.1007/s007050050428
    1. Shibata S, Yamamoto-Goshima F, Maeno K, Hanaichi T, Fujita Y, Nakajima K, et al. . Characterization of a Temperature-Sensitive Influenza B Virus Mutant Defective in Neuraminidase. J Virol (1993) 67(6):3264–73. 10.1128/JVI.67.6.3264-3273.1993
    1. Yamamoto-Goshima F, Maeno K. Approach to the Involvement of Influenza B Neuraminidase in the Cleavage of HA by Host Cell Protease Using Low Ph-Induced Cell Fusion Reaction. Microbiol Immunol (1994) 38(10):819–22. 10.1111/j.1348-0421.1994.tb01864.x
    1. Limburg H, Harbig A, Bestle D, Stein DA, Moulton HM, Jaeger J, et al. . Tmprss2 Is the Major Activating Protease of Influenza A Virus in Primary Human Airway Cells and Influenza B Virus in Human Type II Pneumocytes. J Virol (2019) 93(21):e00649–19. 10.1128/JVI.00649-19
    1. Böttcher E, Freuer C, Steinmetzer T, Klenk H-D, Garten W. MDCK Cells That Express Proteases TMPRSS2 and HAT Provide a Cell System to Propagate Influenza Viruses in the Absence of Trypsin and to Study Cleavage of HA and its Inhibition. Vaccine (2009) 27(45):6324–9. 10.1016/j.vaccine.2009.03.029
    1. Walker JA, Sakaguchi T, Matsuda Y, Yoshida T, Kawaoka Y. Location and Character of the Cellular Enzyme That Cleaves the Hemagglutinin of a Virulent Avian Influenza Virus. Virology (1992) 190(1):278–87. 10.1016/0042-6822(92)91214-f
    1. Lu X, Humeau L, Slepushkin V, Binder G, Yu Q, Slepushkina T, et al. . Safe Two-Plasmid Production for the First Clinical Lentivirus Vector That Achieves >99% Transduction in Primary Cells Using a One-Step Protocol. J Gene Med (2004) 6(9):963–73. 10.1002/jgm.593
    1. Naldini L, Blömer U, Gage FH, Trono D, Verma IM. Efficient Transfer, Integration, and Sustained Long-Term Expression of the Transgene in Adult Rat Brains Injected With a Lentiviral Vector. Proc Natl Acad Sci (1996) 93(21):11382. 10.1073/pnas.93.21.11382
    1. Merten O-W, Charrier S, Laroudie N, Fauchille S, Dugué C, Jenny C, et al. . Large-Scale Manufacture and Characterization of a Lentiviral Vector Produced for Clinical Ex Vivo Gene Therapy Application. Hum Gene Ther (2010) 22(3):343–56. 10.1089/hum.2010.060
    1. Matrosovich MN, Gambaryan AS, Tuzikov AB, Byramova NE, Mochalova LV, Golbraikh AA, et al. . Probing of the Receptor-Binding Sites of the H1 and H3 Influenza A and Influenza B Virus Hemagglutinins by Synthetic and Natural Sialosides. Virology (1993) 196(1):111–21. 10.1006/viro.1993.1459
    1. Varki NM, Varki A. Diversity in Cell Surface Sialic Acid Presentations: Implications for Biology and Disease. Lab Invest (2007) 87(9):851–7. 10.1038/labinvest.3700656
    1. Sieben C, Kappel C, Zhu R, Wozniak A, Rankl C, Hinterdorfer P, et al. . Influenza Virus Binds its Host Cell Using Multiple Dynamic Interactions. Proc Natl Acad Sci (2012) 109(34):13626–31. 10.1073/pnas.1120265109
    1. Heaton NS, Randall G. Multifaceted Roles for Lipids in Viral Infection. Trends Microbiol (2011) 19(7):368–75. 10.1016/j.tim.2011.03.007
    1. Pizzato M, Marlow SA, Blair ED, Takeuchi Y. Initial Binding of Murine Leukemia Virus Particles to Cells Does Not Require Specific Env-Receptor Interaction. J Virol (1999) 73(10):8599. 10.1128/JVI.73.10.8599-8611.1999
    1. Voelkel C, Galla M, Dannhauser PN, Maetzig T, Sodeik B, Schambach A, et al. . Pseudotype-Independent Nonspecific Uptake of Gammaretroviral and Lentiviral Particles in Human Cells. Hum Gene Ther (2012) 23(3):274–86. 10.1089/hum.2011.011
    1. Skowronski DM, Hamelin M-E, Janjua NZ, Serres GD, Gardy JL, Rhéaume C, et al. . Cross-Lineage Influenza B and Heterologous Influenza A Antibody Responses in Vaccinated Mice: Immunologic Interactions and B/Yamagata Dominance. PloS One (2012) 7(6):e38929. 10.1371/journal.pone.0038929
    1. Francis T. (1960). On the Doctrine of Original Antigenic Sin. JSTOR, in: Proceedings of the American Philosophical Society. p. 572–8.
    1. Molesti E, Ferrara F, Lapini G, Montomoli E, Temperton NJ. Discordant Correlation Between Serological Assays Observed When Measuring Heterosubtypic Responses Against Avian Influenza H5 and H7 Viruses in Unexposed Individuals. BioMed Res Int (2014) 2014(5):231365–12. 10.1155/2014/231365
    1. Ramage W, Gaiotto T, Ball C, Risley P, Carnell GW, Temperton N, et al. . Cross-Reactive and Lineage-Specific Single Domain Antibodies Against Influenza B Hemagglutinin. Antibodies (Basel) (2019) 8(1):1–20. 10.3390/antib8010014
    1. Ferrara F, Temperton N. Chimeric Influenza Haemagglutinins: Generation and Use in Pseudotype Neutralization Assays. MethodsX (2017) 4:11–24. 10.1016/j.mex.2016.12.001

Source: PubMed

3
Abonneren