Effect of a Russian-backbone live-attenuated influenza vaccine with an updated pandemic H1N1 strain on shedding and immunogenicity among children in The Gambia: an open-label, observational, phase 4 study

Benjamin B Lindsey, Ya Jankey Jagne, Edwin P Armitage, Anika Singanayagam, Hadijatou J Sallah, Sainabou Drammeh, Elina Senghore, Nuredin I Mohammed, David Jeffries, Katja Höschler, John S Tregoning, Adam Meijer, Ed Clarke, Tao Dong, Wendy Barclay, Beate Kampmann, Thushan I de Silva, Benjamin B Lindsey, Ya Jankey Jagne, Edwin P Armitage, Anika Singanayagam, Hadijatou J Sallah, Sainabou Drammeh, Elina Senghore, Nuredin I Mohammed, David Jeffries, Katja Höschler, John S Tregoning, Adam Meijer, Ed Clarke, Tao Dong, Wendy Barclay, Beate Kampmann, Thushan I de Silva

Abstract

Background: The efficacy and effectiveness of the pandemic H1N1 (pH1N1) component in live attenuated influenza vaccine (LAIV) is poor. The reasons for this paucity are unclear but could be due to impaired replicative fitness of pH1N1 A/California/07/2009-like (Cal09) strains. We assessed whether an updated pH1N1 strain in the Russian-backbone trivalent LAIV resulted in greater shedding and immunogenicity compared with LAIV with Cal09.

Methods: We did an open-label, prospective, observational, phase 4 study in Sukuta, a periurban area in The Gambia. We enrolled children aged 24-59 months who were clinically well. Children received one dose of the WHO prequalified Russian-backbone trivalent LAIV containing either A/17/California/2009/38 (Cal09) or A/17/New York/15/5364 (NY15) based on their year of enrolment. Primary outcomes were the percentage of children with LAIV strain shedding at day 2 and day 7, haemagglutinin inhibition seroconversion, and an increase in influenza haemagglutinin-specific IgA and T-cell responses at day 21 after LAIV. This study is nested within a randomised controlled trial investigating LAIV-microbiome interactions (NCT02972957).

Findings: Between Feb 8, 2017, and April 12, 2017, 118 children were enrolled and received one dose of the Cal09 LAIV from 2016-17. Between Jan 15, 2018, and March 28, 2018, a separate cohort of 135 children were enrolled and received one dose of the NY15 LAIV from 2017-18, of whom 126 children completed the study. Cal09 showed impaired pH1N1 nasopharyngeal shedding (16 of 118 children [14%, 95% CI 8·0-21·1] with shedding at day 2 after administration of LAIV) compared with H3N2 (54 of 118 [46%, 36·6-55·2]; p<0·0001) and influenza B (95 of 118 [81%, 72·2-87·2]; p<0·0001), along with suboptimal serum antibody (seroconversion in six of 118 [5%, 1·9-10·7]) and T-cell responses (CD4+ interferon γ-positive and/or CD4+ interleukin 2-positive responses in 45 of 111 [41%, 31·3-50·3]). After the switch to NY15, a significant increase in pH1N1 shedding was seen (80 of 126 children [63%, 95% CI 54·4-71·9]; p<0·0001 compared with Cal09), along with improvements in seroconversion (24 of 126 [19%, 13·2-26·8]; p=0·011) and influenza-specific CD4+ T-cell responses (73 of 111 [66%, 60·0-75·6; p=0·00028]). The improvement in pH1N1 seroconversion with NY15 was even greater in children who were seronegative at baseline (24 of 64 children [38%, 95% CI 26·7-49·8] vs six of 79 children with Cal09 [8%, 2·8-15·8]; p<0·0001). Persistent shedding to day 7 was independently associated with both seroconversion (odds ratio 12·69, 95% CI 4·1-43·6; p<0·0001) and CD4+ T-cell responses (odds ratio 7·83, 95% CI 2·99-23·5; p<0·0001) by multivariable logistic regression.

Interpretation: The pH1N1 component switch that took place between 2016 and 2018 might have overcome the poor efficacy and effectiveness reported with previous LAIV formulations. LAIV effectiveness against pH1N1 should, therefore, improve in upcoming influenza seasons. Our data highlight the importance of assessing replicative fitness, in addition to antigenicity, when selecting annual LAIV components.

Funding: The Wellcome Trust.

Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study profile Overview of participants who received (A) the 2016–17 northern hemisphere Russian-backbone LAIV formulation and (B) the 2017–18 northern hemisphere Russian-backbone LAIV formulation. LAIV=live attenuated influenza vaccine. pH1N1=pandemic H1N1. HAI=haemagglutinin inhibition. *The study was nested within a larger randomised controlled trial (NCT02972957; appendix pp 3,4). †Sparse cell populations seen on flow cytometry. ‡Total IgA not detected in sample. §No pH1N1 data for one sample in 2016–17 cohort and no pH1N1 data for four samples and H3N2 data for three samples in 2017–18 cohort because of inadequate sample volume.
Figure 2
Figure 2
Shedding of strains in the nasopharynx after vaccination (A) Percentage of children shedding vaccine virus with 2016–17 LAIV formulation compared with the 2017–18 LAIV formulation, at day 2 and day 7. Error bars represent the upper 95% CI. (B) Viral load in the nasopharynx is indicated by ct values from RT-PCR. Red bars indicate median ct values. Lower ct values indicate higher viral loads. (C) Quantitative RT-PCR viral load in children from the 2018 cohort for each strain. Red bars indicate median values. p values are Bonferroni-adjusted for multiplicity within each group of analyses. LAIV=live attenuated influenza vaccine. pH1N1=pandemic H1N1. H3N2=A/17/Hong Kong/2014/8296. B/Vic=B/Texas/02/2013 (Victoria lineage). ct=cycle threshold. RT-PCR=reverse-transcriptase PCR. EID50eq=50% egg infectious dose equivalents.
Figure 3
Figure 3
Effect of baseline serum antibody on LAIV strain shedding in the nasopharynx and replicative ability of viruses in primary epithelial cell cultures (A–D) Predicted probability from logistic regression of vaccine strain shedding at day 2 after LAIV at a given baseline serum HAI titre to each matched strain. Dots show predicted proportions and shaded areas show 95% CIs. Data shown for Cal09 pH1N1 (A), NY15 pH1N1 (B), H3N2 (C), and B/Vic (D). Upper limit is based on maximum observed HAI titre in the dataset. When data from 2017 and 2018 were combined for H3N2 and B/Vic, results were adjusted for year (appendix p 12). (E) Nasopharyngeal viral load at day 2 and day 7 after 2017–18 LAIV, with participants stratified by baseline serostatus to vaccine haemagglutinin-matched and neuraminidase-matched influenza strains. Red bars indicate median values. (F and G) Replication of pH1N1 (F) or H3N2 and B/Vic (G) vaccine strains in primary nasal epithelium. Dots denote mean values and errors bars the SD. In (F), p

Figure 4

Immunogenicity to pH1N1 with the…

Figure 4

Immunogenicity to pH1N1 with the 2016–17 and 2017–18 LAIV formulations p values are…

Figure 4
Immunogenicity to pH1N1 with the 2016–17 and 2017–18 LAIV formulations p values are Bonferroni-adjusted for multiplicity within each group of analyses. (A) Percentage of children seroconverting to each LAIV strain, comparing 2016–17 and 2017–18 formulations. Error bars represent the upper 95% CI. (B) Geometric mean fold change in serum haemagglutinin inhibition titre from baseline to day 21, comparing children seronegative at baseline given 2016–17 and 2017–18 LAIVs. Dotted line depicts a fold change of four. y axis is a logarithmic scale. (C) Influenza-specific CD4+ T-cell responses to vaccine strain-matched pH1 haemagglutinin (Cal09 in 2016–17 or NY15 in 2017–18), H3 haemagglutinin, influenza A matrix and nucleoprotein (both matched to LAIV backbone) peptide pools, comparing 2016–17 and 2017–18 LAIVs. Error bars represent the upper 95% CI. (D) Percentage of children with a twofold rise in influenza-specific CD4+ T-cell responses at day 21 after 2016–17 and 2017–18 LAIVs. y axis is a logarithmic scale. (E) Percentage of influenza-specific mucosal IgA responders given the 2016–17 and 2017–18 LAIVs. Error bars represent the upper 95% CI. pH1N1=pandemic H1N1. LAIV=live attenuated influenza vaccine. Cal09=A/17/California/2009/38. NY15=A/17/New York/15/5364. H3N2=A/17/Hong Kong/2014/8296. B/Vic=B/Texas/02/2013 (Victoria lineage). IFNγ=interferon γ.
Figure 4
Figure 4
Immunogenicity to pH1N1 with the 2016–17 and 2017–18 LAIV formulations p values are Bonferroni-adjusted for multiplicity within each group of analyses. (A) Percentage of children seroconverting to each LAIV strain, comparing 2016–17 and 2017–18 formulations. Error bars represent the upper 95% CI. (B) Geometric mean fold change in serum haemagglutinin inhibition titre from baseline to day 21, comparing children seronegative at baseline given 2016–17 and 2017–18 LAIVs. Dotted line depicts a fold change of four. y axis is a logarithmic scale. (C) Influenza-specific CD4+ T-cell responses to vaccine strain-matched pH1 haemagglutinin (Cal09 in 2016–17 or NY15 in 2017–18), H3 haemagglutinin, influenza A matrix and nucleoprotein (both matched to LAIV backbone) peptide pools, comparing 2016–17 and 2017–18 LAIVs. Error bars represent the upper 95% CI. (D) Percentage of children with a twofold rise in influenza-specific CD4+ T-cell responses at day 21 after 2016–17 and 2017–18 LAIVs. y axis is a logarithmic scale. (E) Percentage of influenza-specific mucosal IgA responders given the 2016–17 and 2017–18 LAIVs. Error bars represent the upper 95% CI. pH1N1=pandemic H1N1. LAIV=live attenuated influenza vaccine. Cal09=A/17/California/2009/38. NY15=A/17/New York/15/5364. H3N2=A/17/Hong Kong/2014/8296. B/Vic=B/Texas/02/2013 (Victoria lineage). IFNγ=interferon γ.

References

    1. Rhorer J, Ambrose CS, Dickinson S. Efficacy of live attenuated influenza vaccine in children: a meta-analysis of nine randomized clinical trials. Vaccine. 2009;27:1101–1110.
    1. Flannery B, Chung J. Influenza vaccine effectiveness, including LAIV vs IIV in children and adolescents, US flu VE network, 2015–16. June 22, 2016.
    1. Singanayagam A, Zambon M, Lalvani A, Barclay W. Urgent challenges in implementing live attenuated influenza vaccine. Lancet Infect Dis. 2018;18:e25–e32.
    1. Victor JC, Lewis KD, Diallo A. Efficacy of a Russian-backbone live attenuated influenza vaccine among children in Senegal: a randomised, double-blind, placebo-controlled trial. Lancet Glob Health. 2016;4:e955–e965.
    1. Lafond KE, Nair H, Rasooly MH. Global role and burden of influenza in pediatric respiratory hospitalizations, 1982–2012: a systematic analysis. PLoS Med. 2016;13
    1. Lindsey BB, Armitage EP, Kampmann B, de Silva TI. The efficacy, effectiveness, and immunogenicity of influenza vaccines in Africa: a systematic review. Lancet Infect Dis. 2019;19:e110–e119.
    1. Niang MN, Barry MA, Talla C. Estimation of the burden of flu-association influenza-like illness visits on total clinic visits through the sentinel influenza monitoring system in Senegal during the 2013-2015 influenza seasons. Epidemiol Infect. 2018;146:2049–2055.
    1. Rudenko L, van den Bosch H, Kiseleva I. Live attenuated pandemic influenza vaccine: clinical studies on A/17/California/2009/38 (H1N1) and licensing of the Russian-developed technology to WHO for pandemic influenza preparedness in developing countries. Vaccine. 2011;29(suppl 1):A40–A44.
    1. Dhere R, Yeolekar L, Kulkarni P. A pandemic influenza vaccine in India: from strain to sale within 12 months. Vaccine. 2011;29(suppl 1):A16–A21.
    1. Curtis D, Ning MF, Armon C, Li S, Weinberg A. Safety, immunogenicity and shedding of LAIV4 in HIV-infected and uninfected children. Vaccine. 2015;33:4790–4797.
    1. Ellis JS, Zambon MC. Molecular analysis of an outbreak of influenza in the United Kingdom. Eur J Epidemiol. 1997;13:369–372.
    1. de Silva TI, Gould V, Mohammed NI. Comparison of mucosal lining fluid sampling methods and influenza-specific IgA detection assays for use in human studies of influenza immunity. J Immunol Methods. 2017;449:1–6.
    1. Ambrose CS, Wu X, Jones T, Mallory RM. The role of nasal IgA in children vaccinated with live attenuated influenza vaccine. Vaccine. 2012;30:6794–6801.
    1. de Silva TI, Peng Y, Leligdowicz A. Correlates of T-cell-mediated viral control and phenotype of CD8(+) T cells in HIV-2, a naturally contained human retroviral infection. Blood. 2013;121:4330–4339.
    1. Roederer M, Nozzi JL, Nason MC. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A. 2011;79:167–174.
    1. Shcherbik S, Sergent SB, Davis WG. Application of real time RT-PCR for the genetic homogeneity and stability tests of the seed candidates for live attenuated influenza vaccine production. J Virol Methods. 2014;195:18–25.
    1. Mallory R, Nyborg AC, Kalyani R, Tsai L-F, Block SL, Dubovsky F. 1954: a randomized study to evaluate the shedding and immunogenicity of H1N1 strains in trivalent and quadrivalent formulations of FluMist in children 24 to 48 months of age. Open Forum Infect Dis. 2018;5(suppl 1):S564–S565.
    1. Caspard H, Gaglani M, Clipper L. Effectiveness of live attenuated influenza vaccine and inactivated influenza vaccine in children 2-17 years of age in 2013–2014 in the United States. Vaccine. 2016;34:77–82.
    1. Mallory RM, Yi T, Ambrose CS. Shedding of Ann Arbor strain live attenuated influenza vaccine virus in children 6-59 months of age. Vaccine. 2011;29:4322–4327.
    1. Russell CJ, Hu M, Okda FA. Influenza hemagglutinin protein stability, activation, and pandemic risk. Trends Microbiol. 2018;26:841–853.
    1. Cotter CR, Jin H, Chen Z. A single amino acid in the stalk region of the H1N1pdm influenza virus HA protein affects viral fusion, stability and infectivity. PLoS Pathog. 2014;10
    1. Ilyushina NA, Ikizler MR, Kawaoka Y. Comparative study of influenza virus replication in MDCK cells and in primary cells derived from adenoids and airway epithelium. J Virol. 2012;86:11725–11734.
    1. Bandell A, Woo J, Coelingh K. Protective efficacy of live-attenuated influenza vaccine (multivalent, Ann Arbor strain): a literature review addressing interference. Expert Rev Vaccines. 2011;10:1131–1141.
    1. Forrest BD, Pride MW, Dunning AJ. Correlation of cellular immune responses with protection against culture-confirmed influenza virus in young children. Clin Vaccine Immunol. 2008;15:1042–1053.
    1. Lewis KDC, Ortiz JR, Rahman MZ. Immunogenicity and viral shedding of Russian-backbone seasonal trivalent, live-attenuated influenza vaccine in a phase II randomized placebo-controlled trial among pre-school aged children in urban Bangladesh. Clin Infect Dis. 2018 doi: 10.1093/cid/ciy1003. published online Nov 27.
    1. Public Health England Influenza vaccine effectiveness (VE) in adults and children in primary care in the United Kingdom (UK): provisional end-of-season results 2017–18.

Source: PubMed

3
Předplatit