Generation of DelNS1 Influenza Viruses: a Strategy for Optimizing Live Attenuated Influenza Vaccines

Pui Wang, Min Zheng, Siu-Ying Lau, Pin Chen, Bobo Wing-Yee Mok, Siwen Liu, Honglian Liu, Xiaofeng Huang, Conor J Cremin, Wenjun Song, Yixin Chen, Yik-Chun Wong, Haode Huang, Kelvin Kai-Wong To, Zhiwei Chen, Ningshao Xia, Kwok-Yung Yuen, Honglin Chen, Pui Wang, Min Zheng, Siu-Ying Lau, Pin Chen, Bobo Wing-Yee Mok, Siwen Liu, Honglian Liu, Xiaofeng Huang, Conor J Cremin, Wenjun Song, Yixin Chen, Yik-Chun Wong, Haode Huang, Kelvin Kai-Wong To, Zhiwei Chen, Ningshao Xia, Kwok-Yung Yuen, Honglin Chen

Abstract

Nonstructural protein 1 (NS1) of influenza virus is a key virulence element with multifunctional roles in virus replication and a potent antagonist of host immune response. Deletion of NS1 (DelNS1) would create a safer and more extensively immunogenic live attenuated influenza virus (LAIV) vaccine. However, DelNS1 viruses are very difficult to grow in regular vaccine-producing systems, which has hampered the application of DelNS1 LAIV vaccines in humans. We have developed two master backbones of deleted-NS1 (DelNS1) viral genomes from influenza A or B viruses which contain novel adaptive mutations to support DelNS1-LAIV replication. These DelNS1-LAIVs are highly attenuated in human cells in vitro and nonpathogenic in mice but replicate well in vaccine-producing cells. Both influenza A and influenza B DelNS1 LAIVs grow better at 33°C than at 37 to 39°C. Vaccination with DelNS1 LAIV performed once is enough to provide potent protection against lethal challenge with homologous virus and strong long-lasting cross protection against heterosubtypic or antigenically distantly related influenza viruses in mice. Mechanistic investigations revealed that DelNS1-LAIVs induce cross protective neutralizing antibody and CD8+ and CD4+ T cell immunities. Importantly, it has been shown that DelNS1-LAIV can be used to enhance specific anti-influenza immunity through expression of additional antigens from the deleted-NS1 site. Generation of DelNS1 viruses which are nonpathogenic and able to grow in vaccine-producing systems is an important strategy for making highly immunogenic LAIV vaccines that induce broad cross protective immunity against seasonal and emerging influenza.IMPORTANCE Current seasonal influenza vaccines are suboptimal and low in immunogenicity and do not provide long-lasting immunity and cross protection against influenza virus strains that have antigenically drifted. More-effective influenza vaccines which can induce both humoral immunity and T cell immunity are needed. The NS1 protein of influenza virus is a virulence element and the critical factor for regulation of the host immune response during virus infection. Deletion of the NS1 protein is a strategy to make an optimal LAIV vaccine. However, DelNS1 viruses are very difficult to grow in regular vaccine-producing systems, hampering the application of DelNS1 LAIV vaccines in humans. We have generated a panel of both influenza A and influenza B DelNS1 LAIVs which are able to grow in regular vaccine-producing cells. These DelNS1 LAIV vaccines are completely nonpathogenic, exhibit potent and long-lasting immunity, and can be used to express extra viral antigen to induce cross protective immunity against seasonal and emerging influenza.

Keywords: NS1; influenza vaccines; live attenuated vaccine.

Copyright © 2019 Wang et al.

Figures

FIG 1
FIG 1
Generation and characterization of CA4-DelNS1 virus. (A) Full-length and NS1 gene-deleted NS segments. NCR, noncoding region. (B) Confirmation of NS1 deletion in rescued CA4-DelNS1 virus by RT-PCR and agarose gel electrophoresis. -ve, negative control. (C) Newly rescued CA4-DelNS1 virus was passaged in MDCK cells 20 times to promote growth adaptation. After the virus titers stabilized, the entire genome of the CA4-DelNS1 virus was analyzed and adaptive mutations were identified. (D) Effect of adaptive mutations on the efficiency of CA4-DelNS1 virus rescue. DelNS1 pHW2000 plasmids with individual or combined adaptive mutations or the original DelNS1 plasmid (DelNS1-WT) were transfected together with plasmids encoding the other 7 viral segments into 293T/MDCK cell mixtures and incubated at 33°C or 37°C. After 72 h, viral supernatants were collected and titrated. (E) Growth of CA4-DelNS1 at 33°C or 37°C was analyzed. MDCK cells or embryonated chicken eggs were infected with CA4-DelNS1 or WT virus at 0.1 MOI or 1,000 PFU, respectively. Viral supernatants or allantoic fluids were collected at the indicated time points and titrated by plaque assay in MDCK cells. Data represent mean values ± standard deviations of results from three independent experiments. Statistical comparisons between means were performed by Student's t test. ***, P < 0.001; **, P < 0.01; *, P < 0.05; NS, not significant.
FIG 2
FIG 2
Avirulence of CA4-DelNS1 virus. (A) A549 cells were infected with CA4-WT virus, with ca-LAIV, or with CA4-DelNS1 virus at 0.01 MOI and cultured at 33°C or 37°C. ca-LAIV is a cold-adapted 2009 H1N1 virus that was isolated from the licensed LAIV vaccine (17). Viral supernatants were collected at different time points and titrated by plaque assay in MDCK cells. Data represent mean values ± standard deviations of results from three independent experiments. Statistical comparisons between means were performed by Student's t test. ***, P < 0.001; **, P < 0.01. (B) DelNS1 virus infection in mice. Groups of six 6-week-old mice were inoculated with 106 PFU of CA4-DelNS1, H7N9-DelNS1, WSN-DelNS1, or H7N9 WT virus. PBS was used as an inoculation control. Body weight and survival were observed for 14 days. Data represent mean body weight values ± standard deviations of results from 6 mice.
FIG 3
FIG 3
CA4-DelNS1 LAIV vaccination protects mice from both homologous and heterologous virus challenge. (A) Timelines for vaccination and virus challenge. Mice were intranasally vaccinated once with CA4-DelNS1 virus (2 × 106 PFU), ca-LAIV (2 × 106 PFU), or PBS and then challenged with virus after 3 weeks. (B to D) Vaccinated mice were challenged with 10 MLD50 CA4 (H1N1) mouse-adapted virus (B), 10 MLD50 H7N9 virus (C), or 100 MLD50 H5N1 virus (D). Body weights and survival were observed for 14 days. Body weight data represent mean values ± standard deviations of results from 6 mice. (E) HI analysis of sera from mice vaccinated with CA4-DelNS1, ca-LAIV, or PBS, collected prior to virus challenge.
FIG 4
FIG 4
CA4-DelNS1 as a master backbone for construction of DelNS1 H3N2 LAIV. (A) Generation of CA4-DelNS1-HK4801 (H3N2) by plasmid transfection. CA4-DelNS1-HK4801 (H3N2) virus was rescued with HA and NA plasmids derived from the A/Hong Kong/4801/2014 (H3N2) strain and 6 internal gene plasmids from the CA4-DelNS1 virus containing adaptive mutations, as described for Fig. 1. (B) CA4-DelNS1-HK4801 cannot replicate in IFN-competent A549 cells. (C) CA4-DelNS1-HK4801 replication efficiency varied with temperature in MDCK cells. (D) CA4-DelNS1-HK68 (H3N2) protects mice from challenge with homologous HK68-MA virus. Mice were vaccinated with CA4-DelNS1-HK68 (HK68-WT) virus (2 × 106 PFU) or PBS and after 3 weeks were challenged with HK68-MA (mouse-adapted) virus. (E) CA4-DelNS1-HK4801 protects mice from challenge with heterologous HK68-MA virus. Mice were vaccinated with CA4-DelNS1-HK4801 (H3N2) virus (3 × 107 PFU) or PBS and were challenged later with HK68-MA virus. Body weight and survival were observed for 14 days. Body weight data represent mean values ± standard deviations of results from 6/7 mice. Statistical comparisons between means were performed by Student's t test. ***, P < 0.001; **, P < 0.01; *, P < 0.05.
FIG 5
FIG 5
Generation and characterization of DelNS1 B LAIV. (A) Generation of DelNS1-B8038 influenza B virus by reverse genetics. pHW2000 plasmids containing an NS1-deleted (DelNS1) segment and the seven remaining genome segments derived from B/Hong Kong/8038/2011 (Victoria) virus were transfected into a 293T/MDCK cell mixture. Rescued virus was passaged in MDCK cells until the virus titer stabilized. (B) Adaptive mutations in growth-adapted DelNS1-B8038 influenza B virus. Mutations in PA (T210C), NA (T1429C), NP (C182T), and M (G88C plus A280G) were identified. (C) Growth of DelNS1-B8038 influenza B virus in cell culture. MDCK or A549 cells were infected with either WT or DelNS1-B8038 virus at 0.01 MOI and incubated at 33°C or 37°C for 72 h. Virus supernatants were collected and titrated by plaque assay in MDCK cells. Data represent mean values ± standard deviations of results from three independent experiments. Statistical comparisons between means were performed by Student's t test. ***, P < 0.001. (D) DelNS1-B8038 (B-DelNS1) LAIV vaccination protected mice from challenge with both Victoria and Yamagata lineage mouse-adapted viruses. Mice were vaccinated with DelNS1-B8038 (2 × 106 PFU) or PBS. After 3 weeks, vaccinated mice were challenged with mouse-adapted B/Brisbane/60/2008 (Victoria) or B/Florida/4/2006 (Yamagata) influenza B virus at 10 MLD50. Body weight and survival were observed for 14 days. Body weight data represent mean values ± standard deviations of results from 6 mice.
FIG 6
FIG 6
DelNS1 LAIV induces cross protective T cell responses against influenza viruses. (A) Induction of antigen-specific CD8+ and CD4+ T cells by CA4-DelNS1 LAIV was estimated by intracellular cytokine staining (ICS) of ex vivo peptide-stimulated splenocytes, followed by flow cytometric analysis. (B) Schedule of T cell depletion and virus challenge. Vaccinated mice were injected intraperitoneally (i.p.) with 100 μg of anti-CD8α or anti-CD4 or with both anti-CD8α and anti-CD4 or with isotype control (IgG2b) antibodies on day 17 (D17), D19, and D21 after immunization and day 3 after virus challenge. (C) Mice were challenged with H7N9 virus (10 MLD50) on day 21 after vaccination and monitored for 14 days. (D) Mice were challenged with mouse-adapted CA4 (H1N1) virus (10 MLD50) and monitored for 14 days. Body weight data represent mean values ± standard deviations of results from 7 mice. Statistical comparisons between means were performed by Student's t test. ***, P < 0.001; **, P < 0.01.
FIG 7
FIG 7
DelNS1 LAIV expression of B-HA1 antigen protected against lethal challenge with both influenza A and B viruses in mice. (A) Illustration of cloning of B-HAI region into the CA4-DelNS1 NS segment. (B) Mice were vaccinated once with CA4-DelNS1 and then challenged 3 weeks later with mouse-adapted H1N1 influenza A virus (A/California/04/2009, 10 MLD50) or influenza B virus (B/Brisbane/60/2008, 10 MLD50) and monitored for 14 days. (C) Mice were vaccinated twice, 1 month apart, with 106 PFU of either CA4-DelNS1 virus or CA4-DelNS1-BHA1, which contains the HA1 gene of influenza B virus (B/Brisbane/60/2008) at the site in the NS segment from which NS1 was deleted. Mice were challenged with mouse-adapted B/Brisbane/60/2008 (10 MLD50) 1 month after the second vaccination and monitored for 2 weeks. (D) Mice were vaccinated once with 1 × 107 PFU of either CA4-DelNS1-HK4801 (H3N2) virus or CA4-DelNS1-HK4801-BHA1 LAIV. Vaccinated mice were challenged with mouse-adapted B/Brisbane/60/2008 (10 MLD50) 1 month after the second dose of vaccination. Mice were monitored for body weight change and survival for 2 weeks. Body weight data represent mean values ± standard deviations of results from 6 (B and C) or 7 (D) mice.

References

    1. Molinari NA, Ortega-Sanchez IR, Messonnier ML, Thompson WW, Wortley PM, Weintraub E, Bridges CB. 2007. The annual impact of seasonal influenza in the US: measuring disease burden and costs. Vaccine 25:5086–5096. doi:10.1016/j.vaccine.2007.03.046.
    1. Saletti G, Gerlach T, Rimmelzwaan GF. 2018. Influenza vaccines: ‘tailor-made’ or ‘one fits all’. Curr Opin Immunol 53:102–110. doi:10.1016/j.coi.2018.04.015.
    1. Nachbagauer R, Palese P. 2018. Development of next generation hemagglutinin-based broadly protective influenza virus vaccines. Curr Opin Immunol 53:51–57. doi:10.1016/j.coi.2018.04.001.
    1. Petsch B, Schnee M, Vogel AB, Lange E, Hoffmann B, Voss D, Schlake T, Thess A, Kallen KJ, Stitz L, Kramps T. 2012. Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection. Nat Biotechnol 30:1210–1216. doi:10.1038/nbt.2436.
    1. Liebowitz D, Lindbloom JD, Brandl JR, Garg SJ, Tucker SN. 2015. High titre neutralising antibodies to influenza after oral tablet immunisation: a phase 1, randomised, placebo-controlled trial. Lancet Infect Dis 15:1041–1048. doi:10.1016/S1473-3099(15)00266-2.
    1. Baz M, Boonnak K, Paskel M, Santos C, Powell T, Townsend A, Subbarao K. 2015. Nonreplicating influenza A virus vaccines confer broad protection against lethal challenge. mBio 6:e01487-15. doi:10.1128/mBio.01487-15.
    1. Impagliazzo A, Milder F, Kuipers H, Wagner MV, Zhu X, Hoffman RMB, van Meersbergen R, Huizingh J, Wanningen P, Verspuij J, de Man M, Ding Z, Apetri A, Kükrer B, Sneekes-Vriese E, Tomkiewicz D, Laursen NS, Lee PS, Zakrzewska A, Dekking L, Tolboom J, Tettero L, van Meerten S, Yu W, Koudstaal W, Goudsmit J, Ward AB, Meijberg W, Wilson IA, Radošević K. 2015. A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen. Science 349:1301–1306. doi:10.1126/science.aac7263.
    1. Steel J, Lowen AC, Wang TT, Yondola M, Gao Q, Haye K, Garcia-Sastre A, Palese P. 2010. Influenza virus vaccine based on the conserved hemagglutinin stalk domain. mBio 1:e00018-10. doi:10.1128/mBio.00018-10.
    1. Chen YQ, Wohlbold TJ, Zheng NY, Huang M, Huang Y, Neu KE, Lee J, Wan H, Rojas KT, Kirkpatrick E, Henry C, Palm AE, Stamper CT, Lan LY, Topham DJ, Treanor J, Wrammert J, Ahmed R, Eichelberger MC, Georgiou G, Krammer F, Wilson PC. 2018. Influenza infection in humans induces broadly cross-reactive and protective neuraminidase-reactive antibodies. Cell 173:417–429.e410. doi:10.1016/j.cell.2018.03.030.
    1. Wohlbold TJ, Podolsky KA, Chromikova V, Kirkpatrick E, Falconieri V, Meade P, Amanat F, Tan J, tenOever BR, Tan GS, Subramaniam S, Palese P, Krammer F. 2017. Broadly protective murine monoclonal antibodies against influenza B virus target highly conserved neuraminidase epitopes. Nat Microbiol 2:1415–1424. doi:10.1038/s41564-017-0011-8.
    1. El Bakkouri K, Descamps F, De Filette M, Smet A, Festjens E, Birkett A, Van Rooijen N, Verbeek S, Fiers W, Saelens X. 2011. Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection. J Immunol 186:1022–1031. doi:10.4049/jimmunol.0902147.
    1. Kolpe A, Schepens B, Fiers W, Saelens X. 2017. M2-based influenza vaccines: recent advances and clinical potential. Expert Rev Vaccines 16:123–136. doi:10.1080/14760584.2017.1240041.
    1. Belshe RB, Mendelman PM, Treanor J, King J, Gruber WC, Piedra P, Bernstein DI, Hayden FG, Kotloff K, Zangwill K, Iacuzio D, Wolff M. 1998. The efficacy of live attenuated, cold-adapted, trivalent, intranasal influenzavirus vaccine in children. N Engl J Med 338:1405–1412. doi:10.1056/NEJM199805143382002.
    1. Du Y, Xin L, Shi Y, Zhang TH, Wu NC, Dai L, Gong D, Brar G, Shu S, Luo J, Reiley W, Tseng YW, Bai H, Wu TT, Wang J, Shu Y, Sun R. 2018. Genome-wide identification of interferon-sensitive mutations enables influenza vaccine design. Science 359:290–296. doi:10.1126/science.aan8806.
    1. Si L, Xu H, Zhou X, Zhang Z, Tian Z, Wang Y, Wu Y, Zhang B, Niu Z, Zhang C, Fu G, Xiao S, Xia Q, Zhang L, Zhou D. 2016. Generation of influenza A viruses as live but replication-incompetent virus vaccines. Science 354:1170–1173. doi:10.1126/science.aah5869.
    1. Wang L, Liu SY, Chen HW, Xu J, Chapon M, Zhang T, Zhou F, Wang YE, Quanquin N, Wang G, Tian X, He Z, Liu L, Yu W, Sanchez DJ, Liang Y, Jiang T, Modlin R, Bloom BR, Li Q, Deng JC, Zhou P, Qin FX, Cheng G. 2017. Generation of a live attenuated influenza vaccine that elicits broad protection in mice and ferrets. Cell Host Microbe 21:334–343. doi:10.1016/j.chom.2017.02.007.
    1. Krug RM. 2015. Functions of the influenza A virus NS1 protein in antiviral defense. Curr Opin Virol 12:1–6. doi:10.1016/j.coviro.2015.01.007.
    1. Ayllon J, Garcia-Sastre A. 2015. The NS1 protein: a multitasking virulence factor. Curr Top Microbiol Immunol 386:73–107. doi:10.1007/82_2014_400.
    1. Fernandez-Sesma A, Marukian S, Ebersole BJ, Kaminski D, Park MS, Yuen T, Sealfon SC, Garcia-Sastre A, Moran TM. 2006. Influenza virus evades innate and adaptive immunity via the NS1 protein. J Virol 80:6295–6304. doi:10.1128/JVI.02381-05.
    1. Geiss GK, Salvatore M, Tumpey TM, Carter VS, Wang X, Basler CF, Taubenberger JK, Bumgarner RE, Palese P, Katze MG, Garcia-Sastre A. 2002. Cellular transcriptional profiling in influenza A virus-infected lung epithelial cells: the role of the nonstructural NS1 protein in the evasion of the host innate defense and its potential contribution to pandemic influenza. Proc Natl Acad Sci U S A 99:10736–10741. doi:10.1073/pnas.112338099.
    1. Zanin M, Wong SS, Barman S, Kaewborisuth C, Vogel P, Rubrum A, Darnell D, Marinova-Petkova A, Krauss S, Webby RJ, Webster RG. 2017. Molecular basis of mammalian transmissibility of avian H1N1 influenza viruses and their pandemic potential. Proc Natl Acad Sci U S A 114:11217–11222. doi:10.1073/pnas.1713974114.
    1. Richt JA, Garcia-Sastre A. 2009. Attenuated influenza virus vaccines with modified NS1 proteins. Curr Top Microbiol Immunol 333:177–195. doi:10.1007/978-3-540-92165-3_9.
    1. Mueller SN, Langley WA, Carnero E, Garcia-Sastre A, Ahmed R. 2010. Immunization with live attenuated influenza viruses that express altered NS1 proteins results in potent and protective memory CD8+ T-cell responses. J Virol 84:1847–1855. doi:10.1128/JVI.01317-09.
    1. Pica N, Langlois RA, Krammer F, Margine I, Palese P. 2012. NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol 86:10293–10301. doi:10.1128/JVI.01131-12.
    1. Richt JA, Lekcharoensuk P, Lager KM, Vincent AL, Loiacono CM, Janke BH, Wu WH, Yoon KJ, Webby RJ, Solorzano A, Garcia-Sastre A. 2006. Vaccination of pigs against swine influenza viruses by using an NS1-truncated modified live-virus vaccine. J Virol 80:11009–11018. doi:10.1128/JVI.00787-06.
    1. Kappes MA, Sandbulte MR, Platt R, Wang C, Lager KM, Henningson JN, Lorusso A, Vincent AL, Loving CL, Roth JA, Kehrli ME Jr.. 2012. Vaccination with NS1-truncated H3N2 swine influenza virus primes T cells and confers cross-protection against an H1N1 heterosubtypic challenge in pigs. Vaccine 30:280–288. doi:10.1016/j.vaccine.2011.10.098.
    1. Vincent AL, Ma W, Lager KM, Richt JA, Janke BH, Sandbulte MR, Gauger PC, Loving CL, Webby RJ, Garcia-Sastre A. 2012. Live attenuated influenza vaccine provides superior protection from heterologous infection in pigs with maternal antibodies without inducing vaccine-associated enhanced respiratory disease. J Virol 86:10597–10605. doi:10.1128/JVI.01439-12.
    1. Mossler C, Groiss F, Wolzt M, Wolschek M, Seipelt J, Muster T. 2013. Phase I/II trial of a replication-deficient trivalent influenza virus vaccine lacking NS1. Vaccine 31:6194–6200. doi:10.1016/j.vaccine.2013.10.061.
    1. Wacheck V, Egorov A, Groiss F, Pfeiffer A, Fuereder T, Hoeflmayer D, Kundi M, Popow-Kraupp T, Redlberger-Fritz M, Mueller CA, Cinatl J, Michaelis M, Geiler J, Bergmann M, Romanova J, Roethl E, Morokutti A, Wolschek M, Ferko B, Seipelt J, Dick-Gudenus R, Muster T. 2010. A novel type of influenza vaccine: safety and immunogenicity of replication-deficient influenza virus created by deletion of the interferon antagonist NS1. J Infect Dis 201:354–362. doi:10.1086/649428.
    1. Egorov A, Brandt S, Sereinig S, Romanova J, Ferko B, Katinger D, Grassauer A, Alexandrova G, Katinger H, Muster T. 1998. Transfectant influenza A viruses with long deletions in the NS1 protein grow efficiently in Vero cells. J Virol 72:6437–6441.
    1. Zheng M, Wang P, Song W, Lau SY, Liu S, Huang X, Mok BW, Liu YC, Chen Y, Yuen KY, Chen H. 2015. An A14U substitution in the 3’ noncoding region of the M segment of viral RNA supports replication of influenza virus with an NS1 deletion by modulating alternative splicing of M segment mRNAs. J Virol 89:10273–10285. doi:10.1128/JVI.00919-15.
    1. Wressnigg N, Voss D, Wolff T, Romanova J, Ruthsatz T, Mayerhofer I, Reiter M, Nakowitsch S, Humer J, Morokutti A, Muster T, Egorov A, Kittel C. 2009. Development of a live-attenuated influenza B DeltaNS1 intranasal vaccine candidate. Vaccine 27:2851–2857. doi:10.1016/j.vaccine.2009.02.087.
    1. van Wielink R, Harmsen MM, Martens DE, Peeters BP, Wijffels RH, Moormann RJ. 2012. Mutations in the M-gene segment can substantially increase replication efficiency of NS1 deletion influenza A virus in MDCK cells. J Virol 86:12341–12350. doi:10.1128/JVI.01725-12.
    1. Gould PS, Easton AJ, Dimmock NJ. 2017. Live attenuated influenza vaccine contains substantial and unexpected amounts of defective viral genomic RNA. Viruses 9:269. doi:10.3390/v9100269.
    1. Brown EG, Liu H, Kit LC, Baird S, Nesrallah M. 2001. Pattern of mutation in the genome of influenza A virus on adaptation to increased virulence in the mouse lung: identification of functional themes. Proc Natl Acad Sci U S A 98:6883–6888. doi:10.1073/pnas.111165798.
    1. Valkenburg SA, Li OT, Mak PW, Mok CK, Nicholls JM, Guan Y, Waldmann TA, Peiris JS, Perera LP, Poon LL. 2014. IL-15 adjuvanted multivalent vaccinia-based universal influenza vaccine requires CD4+ T cells for heterosubtypic protection. Proc Natl Acad Sci U S A 111:5676–5681. doi:10.1073/pnas.1403684111.
    1. Shen C, Chen J, Li R, Zhang M, Wang G, Stegalkina S, Zhang L, Chen J, Cao J, Bi X, Anderson SF, Alefantis T, Zhang M, Cai X, Yang K, Zheng Q, Fang M, Yu H, Luo W, Zheng Z, Yuan Q, Zhang J, Wai-Kuo Shih J, Kleanthous H, Chen H, Chen Y, Xia N. 18 October 2017, posting date A multimechanistic antibody targeting the receptor binding site potently cross-protects against influenza B viruses. Sci Transl Med doi:10.1126/scitranslmed.aam5752.
    1. Belongia EA, Simpson MD, King JP, Sundaram ME, Kelley NS, Osterholm MT, McLean HQ. 2016. Variable influenza vaccine effectiveness by subtype: a systematic review and meta-analysis of test-negative design studies. Lancet Infect Dis 16:942–951. doi:10.1016/S1473-3099(16)00129-8.
    1. Houser K, Subbarao K. 2015. Influenza vaccines: challenges and solutions. Cell Host Microbe 17:295–300. doi:10.1016/j.chom.2015.02.012.
    1. Jin H, Subbarao K. 2015. Live attenuated influenza vaccine. Curr Top Microbiol Immunol 386:181–204. doi:10.1007/82_2014_410.
    1. Murphy BR, Coelingh K. 2002. Principles underlying the development and use of live attenuated cold-adapted influenza A and B virus vaccines. Viral Immunol 15:295–323. doi:10.1089/08828240260066242.
    1. Coelingh KL, Luke CJ, Jin H, Talaat KR. 2014. Development of live attenuated influenza vaccines against pandemic influenza strains. Expert Rev Vaccines 13:855–871. doi:10.1586/14760584.2014.922417.
    1. Heaton NS, Sachs D, Chen CJ, Hai R, Palese P. 2013. Genome-wide mutagenesis of influenza virus reveals unique plasticity of the hemagglutinin and NS1 proteins. Proc Natl Acad Sci U S A 110:20248–20253. doi:10.1073/pnas.1320524110.
    1. Carrillo B, Choi JM, Bornholdt ZA, Sankaran B, Rice AP, Prasad BV. 2014. The influenza A virus protein NS1 displays structural polymorphism. J Virol 88:4113–4122. doi:10.1128/JVI.03692-13.
    1. Huang X, Zheng M, Wang P, Mok BW, Liu S, Lau SY, Chen P, Liu YC, Liu H, Chen Y, Song W, Yuen KY, Chen H. 2017. An NS-segment exonic splicing enhancer regulates influenza A virus replication in mammalian cells. Nat Commun 8:14751. doi:10.1038/ncomms14751.
    1. Fonville JM, Wilks SH, James SL, Fox A, Ventresca M, Aban M, Xue L, Jones TC, Le NMH, Pham QT, Tran ND, Wong Y, Mosterin A, Katzelnick LC, Labonte D, Le TT, van der Net G, Skepner E, Russell CA, Kaplan TD, Rimmelzwaan GF, Masurel N, de Jong JC, Palache A, Beyer WEP, Le QM, Nguyen TH, Wertheim HFL, Hurt AC, Osterhaus A, Barr IG, Fouchier RAM, Horby PW, Smith DJ. 2014. Antibody landscapes after influenza virus infection or vaccination. Science 346:996–1000. doi:10.1126/science.1256427.
    1. Smith DJ, Lapedes AS, de Jong JC, Bestebroer TM, Rimmelzwaan GF, Osterhaus AD, Fouchier RA. 2004. Mapping the antigenic and genetic evolution of influenza virus. Science 305:371–376. doi:10.1126/science.1097211.
    1. Bedford T, Riley S, Barr IG, Broor S, Chadha M, Cox NJ, Daniels RS, Gunasekaran CP, Hurt AC, Kelso A, Klimov A, Lewis NS, Li X, McCauley JW, Odagiri T, Potdar V, Rambaut A, Shu Y, Skepner E, Smith DJ, Suchard MA, Tashiro M, Wang D, Xu X, Lemey P, Russell CA. 2015. Global circulation patterns of seasonal influenza viruses vary with antigenic drift. Nature 523:217–220. doi:10.1038/nature14460.
    1. Hoffmann E, Neumann G, Kawaoka Y, Hobom G, Webster RG. 2000. A DNA transfection system for generation of influenza A virus from eight plasmids. Proc Natl Acad Sci U S A 97:6108–6113. doi:10.1073/pnas.100133697.
    1. Song W, Wang P, Mok BW, Lau SY, Huang X, Wu WL, Zheng M, Wen X, Yang S, Chen Y, Li L, Yuen KY, Chen H. 2014. The K526R substitution in viral protein PB2 enhances the effects of E627K on influenza virus replication. Nat Commun 5:5509. doi:10.1038/ncomms6509.
    1. Zheng B, Chan KH, Zhang AJ, Zhou J, Chan CC, Poon VK, Zhang K, Leung VH, Jin DY, Woo PC, Chan JF, To KK, Chen H, Yuen KY. 2010. D225G mutation in hemagglutinin of pandemic influenza H1N1 (2009) virus enhances virulence in mice. Exp Biol Med (Maywood) 235:981–988. doi:10.1258/ebm.2010.010071.
    1. Guo H, Santiago F, Lambert K, Takimoto T, Topham DJ. 2011. T cell-mediated protection against lethal 2009 pandemic H1N1 influenza virus infection in a mouse model. J Virol 85:448–455. doi:10.1128/JVI.01812-10.

Source: PubMed

3
購読する