HAI and NAI titer correlates of inactivated and live attenuated influenza vaccine efficacy

Peter B Gilbert, Youyi Fong, Michal Juraska, Lindsay N Carpp, Arnold S Monto, Emily T Martin, Joshua G Petrie, Peter B Gilbert, Youyi Fong, Michal Juraska, Lindsay N Carpp, Arnold S Monto, Emily T Martin, Joshua G Petrie

Abstract

Background: High hemagglutination inhibition (HAI) and neuraminidase inhibition (NAI) titers are generally associated with reduced influenza risk. While repeated influenza vaccination reduces seroresponse, vaccine effectiveness is not always reduced.

Methods: During the 2007-2008 influenza season, a randomized, placebo-controlled trial (FLUVACS) evaluated the efficacies of live-attenuated (LAIV) and inactivated influenza vaccines (IIV) among healthy adults aged 18-49 in Michigan; IIV vaccine efficacy (VE) and LAIV VE against influenza disease were estimated at 68% and 36%. Using the principal stratification/VE moderation framework, we analyzed data from this trial to assess how each VE varied by HAI or NAI responses to vaccination observed for vaccinated individuals and predicted counterfactually for placebo recipients. We also assessed how each VE varied with pre-vaccination/baseline variables including HAI titer, NAI titer, and vaccination history.

Results: IIV VE appeared to increase with Day 30 post-vaccination HAI titer, albeit not significantly (p=0.20 and estimated VE 14.4%, 70.5%, and 85.5% at titer below the assay lower quantification limit, 512, and 4096 (maximum)). Moreover, IIV VE increased significantly with Day 30 post-vaccination NAI titer (p=0.040), with estimated VE zero at titer 10 and 92.2% at highest titer 640. There was no evidence that fold-change in post-vaccination HAI or NAI titer associated with IIV VE (p=0.76, 0.38). For LAIV, there was no evidence that VE associated with post-vaccination or fold-rise HAI or NAI titers (p-values >0.40). For IIV, VE increased with increasing baseline NAI titer in those previously vaccinated, but VE decreased with increasing baseline NAI titer in those previously unvaccinated. In contrast, for LAIV, VE did not depend on previous vaccination or baseline HAI or NAI titer.

Conclusions: Future efficacy trials should measure baseline and post-vaccination antibody titers in both vaccine and control/placebo recipients, enabling analyses to better elucidate correlates of vaccine- and natural-protection.

Trial registration: ClinicalTrials.gov NCT00538512. October 1, 2007.

Keywords: FLUVACS trial; Hemagglutinin inhibition (HAI) titers; Immune correlates; Neuraminidase inhibition (NAI) titer; Principal stratification/vaccine efficacy moderation framework; Vaccine efficacy.

Conflict of interest statement

Arnold S. Monto declares grant support from Sanofi Pasteur and consultancy fees from Sanofi Pasteur, Novartis, and Novavax. Peter B. Gilbert declares grant support from Sanofi Pasteur.

Figures

Fig. 1
Fig. 1
Boxplots of Day 0, Day 30, and fold-rise HAI and NAI titers by treatment group (Placebo, LAIV, IIV) and influenza case-control outcome status. Boxplots of HAI and NAI titers by treatment group and influenza case-control outcome status. Day 0 titers (top row), Day 30 titers (middle row), and titer fold-rise (Day 30 / Day 0) are shown for participants assigned to placebo (left column), live attenuated influenza vaccine (middle column), or inactivated influenza vaccine (right column)
Fig. 2
Fig. 2
Distributions of Day 0 HAI and NAI titers by previous vaccination status and age. for all three treatment groups combined. Small perturbations are added to titer values to improve visibility. Spearman correlation coefficients r are shown in the scatterplots. Data shown are from all three treatment groups combined. Small perturbations are added to titer values to improve visibility. Spearman correlation coefficients r are shown in the scatterplots. For the HAI titer plots, 8192 means >4096 and 4 means <8 (the upper and lower limits of quantification of the HAI assay were 4096 and 8, respectively)
Fig. 3
Fig. 3
VE of the IIV vaccine depends on baseline NAI titer differently among the previously vaccinated and previously unvaccinated. VE-by-Day-0 NAI titer plots are shown for the IIV vaccine in (a) the previously vaccinated vs (b) unvaccinated
Fig. 4
Fig. 4
Evervax modifies the association between Day 0 NAI and influenza risk with decreasing strength in the placebo arm. The shaded area is the pointwise 95% confidence region for the risk function among Evervax=NO in the placebo group. The slopes of the other three risk functions are not significantly different from 0
Fig. 5
Fig. 5
Estimated IIV VE by Day 30 and fold-rise HAI and NAI titer variables. Estimated VE by a Day 30 HAI titer if assigned vaccine, b fold-rise in HAI titer if assigned vaccine, c Day 30 NAI titer if assigned vaccine, d fold-rise in NAI titer if assigned vaccine. Each solid black line is the estimated VE by the Juraska (2018) method [48]. Dashed and dot-dashed black lines are pointwise and simultaneous 95% confidence intervals, respectively. Each dotted red line is the estimated overall VE. P-values shown are 2-sided p-values for effect modification
Fig. 6
Fig. 6
Estimated LAIV VE by Day 30 and fold-rise HAI and NAI titer variables. Estimated VE by a Day 30 HAI titer if assigned vaccine, b fold-rise in HAI titer if assigned vaccine, c Day 30 NAI titer if assigned vaccine, d fold-rise in NAI titer if assigned vaccine. Each solid black line is the estimated VE by the Juraska (2018) method [48]. Dashed and dot-dashed black lines are pointwise and simultaneous 95% confidence intervals, respectively. Each dotted red line is the estimated overall VE. P-values shown are 2-sided p-values for effect modification

References

    1. Chen GL, Subbarao K. Attacking the flu: Neutralizing antibodies may lead to ‘universal’ vaccine. Nat Med. 2009;15(11):1251. doi: 10.1038/nm1109-1251.
    1. Houser K, Subbarao K. Influenza vaccines: Challenges and solutions. Cell Host Microbe. 2015;17(3):295–300. doi: 10.1016/j.chom.2015.02.012.
    1. Coudeville L, Bailleux F, Riche B, Megas F, Andre P, Ecochard R. Relationship between haemagglutination-inhibiting antibody titres and clinical protection against influenza: Development and application of a bayesian random-effects model. BMC Med Res Methodol. 2010;10:18. doi: 10.1186/1471-2288-10-18.
    1. Hobson D, Curry RL, Beare AS, Ward-Gardner A. The role of serum haemagglutination-inhibiting antibody in protection against challenge infection with influenza A2 and B viruses. J Hyg (Lond) 1972;70(4):767–77. doi: 10.1017/S0022172400022610.
    1. Potter CW, Oxford JS. Determinants of immunity to influenza infection in Man. Br Med Bull. 1979;35(1):69–75. doi: 10.1093/oxfordjournals.bmb.a071545.
    1. Al-Khayatt R, Jennings R, Potter CW. Interpretation of responses and protective levels of antibody against attenuated influenza A viruses using single radial haemolysis. J Hyg (Lond) 1984;93(2):301–12. doi: 10.1017/S0022172400064834.
    1. Evans AS. Serologic studies of acute respiratory infections in military personnel. Yale J Biol Med. 1975;48(3):201.
    1. Fox JP, Cooney MK, Hall CE, Foy HM. Influenza virus infections in Seattle families, 1975–1979 ii. Pattern of infection in invaded households and relation of age and prior antibody to occurrence of infection and related illness. Am J Epidemiol. 1982;116(2):228–42. doi: 10.1093/oxfordjournals.aje.a113408.
    1. Meiklejohn G, Kempe CH, Thalman WG, Lennette EH. Evaluation of monovalent influenza vaccines. ii. Observations during an influenza A-prime epidemic. Am J Hyg. 1952;55(1):12–21.
    1. Petrie JG, Ohmit SE, Johnson E, Cross RT, Monto AS. Efficacy studies of influenza vaccines: Effect of end points used and characteristics of vaccine failures. J Infect Dis. 2011;203(9):1309–15. doi: 10.1093/infdis/jir015.
    1. Black S, Nicolay U, Vesikari T, Knuf M, Del Giudice G, Della Chioppa G, et al. Hemagglutination inhibition antibody titers as a correlate of protection for inactivated influenza vaccines in children. Pediatr Infect Dis J. 2011;30(12):1081–5. doi: 10.1097/INF.0b013e3182367662.
    1. Ohmit SE, Petrie JG, Cross RT, Johnson E, Monto AS. Influenza hemagglutination-inhibition antibody titer as a correlate of vaccine-induced protection. J Infect Dis. 2011;204(12):1879–85. doi: 10.1093/infdis/jir661.
    1. Eichelberger MC, Wan H. Influenza neuraminidase as a vaccine antigen. Curr Top Microbiol Immunol. 2015;386:275–99.
    1. Monto AS, Petrie JG, Cross RT, Johnson E, Liu M, Zhong W, et al. Antibody to influenza virus neuraminidase: An independent correlate of protection. J Infect Dis. 2015;212(8):1191–9. doi: 10.1093/infdis/jiv195.
    1. Memoli MJ, Shaw PA, Han A, Czajkowski L, Reed S, Athota R, et al. Evaluation of antihemagglutinin and antineuraminidase antibodies as correlates of protection in an influenza A/H1N1 virus healthy human challenge model. MBio. 2016;7(2):00417–16. doi: 10.1128/mBio.00417-16.
    1. Plotkin JB, Dushoff J, Levin SA. Hemagglutinin sequence clusters and the antigenic evolution of influenza A virus. Proc Natl Acad Sci U S A. 2002;99(9):6263–8. doi: 10.1073/pnas.082110799.
    1. Krystal M, Young JF, Palese P, Wilson IA, Skehel JJ, Wiley DC. Sequential mutations in hemagglutinins of influenza B virus isolates: Definition of antigenic domains. Proc Natl Acad Sci U S A. 1983;80(14):4527–31. doi: 10.1073/pnas.80.14.4527.
    1. McDonald NJ, Smith CB, Cox NJ. Antigenic drift in the evolution of H1N1 influenza a viruses resulting from deletion of a single amino acid in the haemagglutinin gene. J Gen Virol. 2007;88(12):3209–13. doi: 10.1099/vir.0.83184-0.
    1. Carrat F., Flahault A. Influenza vaccine: The challenge of antigenic drift. Vaccine. 2007;25(39):6852–62. doi: 10.1016/j.vaccine.2007.07.027.
    1. Grohskopf LA, Sokolow LZ, Broder KR, Walter EB, Fry AM, Jernigan DB. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices —- United States, 2018–19 influenza season. MMWR Recomm Rep. 2018;67(3):1. doi: 10.15585/mmwr.rr6703a1.
    1. Petrie JG, Ohmit SE, Johnson E, Truscon R, Monto AS. Persistence of antibodies to influenza hemagglutinin and neuraminidase following one or two years of influenza vaccination. J Infect Dis. 2015;212(12):1914–22. doi: 10.1093/infdis/jiv313.
    1. Beyer W, Palache A, Sprenger M, Hendriksen E, Tukker J., Darioli R, et al. Effects of repeated annual influenza vaccination on vaccine sero-response in young and elderly adults. Vaccine. 1996;14(14):1331–9. doi: 10.1016/S0264-410X(96)00058-8.
    1. Sasaki S, He XS, Holmes TH, Dekker CL, Kemble GW, Arvin AM, et al. Influence of prior influenza vaccination on antibody and B-cell responses. PLoS One. 2008;3(8):2975. doi: 10.1371/journal.pone.0002975.
    1. Huijskens E, Rossen J, Mulder P, van Beek R, van Vugt H, Verbakel J, et al. Immunogenicity, boostability, and sustainability of the immune response after vaccination against influenza A virus (H1N1) 2009 in a healthy population. Clin Vaccine Immunol. 2011;18(9):1401–5. doi: 10.1128/CVI.05046-11.
    1. Künzel W, Glathe H, Engelmann H, Van Hoecke C. Kinetics of humoral antibody response to trivalent inactivated split influenza vaccine in subjects previously vaccinated or vaccinated for the first time. Vaccine. 1996;14(12):1108–10. doi: 10.1016/0264-410X(96)00061-8.
    1. Nabeshima S, Kashiwagi K, Murata M, Kanamoto Y, Furusyo N, Hayashi J. Antibody response to influenza vaccine in adults vaccinated with identical vaccine strains in consecutive years. J Med Virol. 2007;79(3):320–5. doi: 10.1002/jmv.20801.
    1. McLean HQ, Thompson MG, Sundaram ME, Meece JK, McClure DL, Friedrich TC, et al. Impact of repeated vaccination on vaccine effectiveness against influenza A (H3N2) and B during 8 seasons. Clin Infect Dis. 2014;59(10):1375–85. doi: 10.1093/cid/ciu680.
    1. Martínez-Baz I, Casado I, Navascués A, et al. Effect of repeated vaccination with the same vaccine component against influenza A (H1N1) virus. J Infect Dis. 2017;215:847–55. doi: 10.1093/infdis/jix055.
    1. Skowronski DM, Janjua NZ, Sabaiduc S, De Serres G, Winter AL, Gubbay JB, et al. Influenza A/subtype and B/lineage effectiveness estimates for the 2011-12 trivalent vaccine: Cross-season and cross-lineage protection with unchanged vaccine. J Infect Dis. 2014;210(1):126–137. doi: 10.1093/infdis/jiu048.
    1. Beyer WE, de Bruijn IA, Palache AM, Westendorp RG, Osterhaus AD. Protection against influenza after annually repeated vaccination: A meta-analysis of serologic and field studies. Arch Intern Med. 1999;159(2):182–8. doi: 10.1001/archinte.159.2.182.
    1. Keitel WA, Cate TR, Couch RB, Huggins LL, Hess KR. Efficacy of repeated annual immunization with inactivated influenza virus vaccines over a five year period. Vaccine. 1997;15(10):1114–22. doi: 10.1016/S0264-410X(97)00003-0.
    1. Ohmit SE, Petrie JG, Malosh RE, Cowling BJ, Thompson MG, Shay DK, et al. Influenza vaccine effectiveness in the community and the household. Clin Infect Dis. 2013;56(10):1363–9. doi: 10.1093/cid/cit060.
    1. Ohmit SE, Thompson MG, Petrie JG, Thaker SN, Jackson ML, Belongia EA, et al. Influenza vaccine effectiveness in the 2011–2012 season: Protection against each circulating virus and the effect of prior vaccination on estimates. Clin Infect Dis. 2014;58(3):319–27. doi: 10.1093/cid/cit736.
    1. Thompson MG, Li D-K, Shifflett P, Sokolow LZ, Ferber JR, Kurosky S, et al. Effectiveness of seasonal trivalent influenza vaccine for preventing influenza virus illness among pregnant women: A population-based case-control study during the 2010-2011 and 2011-2012 influenza seasons. Clin Infect Dis. 2013;58(4):449–57. doi: 10.1093/cid/cit750.
    1. Hoskins T, Davies J, Smith A, Miller C, Allchin A. Assessment of inactivated influenza-A vaccine after three outbreaks of influenza A at Christ’s Hospital. The Lancet. 1979;313(8106):33–5. doi: 10.1016/S0140-6736(79)90468-9.
    1. Gupta V, Earl DJ, Deem MW. Quantifying influenza vaccine efficacy and antigenic distance. Vaccine. 2006;24(18):3881–8. doi: 10.1016/j.vaccine.2006.01.010.
    1. Smith DJ, Forrest S, Ackley DH, Perelson AS. Variable efficacy of repeated annual influenza vaccination. Proc Natl Acad Sci U S A. 1999;96(24):14001–6. doi: 10.1073/pnas.96.24.14001.
    1. Frangakis CE, Rubin DB. Principal stratification in causal inference. Biometrics. 2002;58(1):21–9. doi: 10.1111/j.0006-341X.2002.00021.x.
    1. Follmann D. Augmented designs to assess immune response in vaccine trials. Biometrics. 2006;62(4):1161–9. doi: 10.1111/j.1541-0420.2006.00569.x.
    1. Gilbert PB, Hudgens MG. Evaluating candidate principal surrogate endpoints. Biometrics. 2008;64(4):1146–54. doi: 10.1111/j.1541-0420.2008.01014.x.
    1. Gilbert PB, Gabriel EE, Miao XP, Li X, Su SC, Parrino J, et al. Fold rise in antibody titers by measured by glycoprotein-based enzyme-linked immunosorbent assay is an excellent correlate of protection for a herpes zoster vaccine, demonstrated via the vaccine efficacy curve. J Infect Dis. 2014;210(10):1573–81. doi: 10.1093/infdis/jiu279.
    1. Moodie Z, Juraska M, Huang Y, Zhuang Y, Fong Y, Carpp L, et al. Neutralizing antibody correlates analysis of tetravalent dengue vaccine efficacy trials in Asia and Latin America, J Infect Dis. 2018;217(5):742–53. doi: 10.1093/infdis/jix609.
    1. Monto AS, Ohmit SE, Petrie JG, Johnson E, Truscon R, Teich E, et al. Comparative efficacy of inactivated and live attenuated influenza vaccines. N Engl J Med. 2009;361(13):1260–7. doi: 10.1056/NEJMoa0808652.
    1. Katz JM, Hancock K, Xu X. Serologic assays for influenza surveillance, diagnosis and vaccine evaluation. Expert Rev Anti Infect Ther. 2011;9(6):669–83. doi: 10.1586/eri.11.51.
    1. Couzens L, Gao J, Westgeest K, Sandbulte M, Lugovtsev V, Fouchier R, et al. An optimized enzyme-linked lectin assay to measure influenza A virus neuraminidase inhibition antibody titers in human sera. J Virol Methods. 2014;210:7–14. doi: 10.1016/j.jviromet.2014.09.003.
    1. Huang Y, Gilbert PB. Comparing biomarkers as principal surrogate endpoints. Biometrics. 2011;67(4):1442–51. doi: 10.1111/j.1541-0420.2011.01603.x.
    1. Prentice R, Pyke R. Logistic disease incidence models and case-control studies. Biometrika. 1979;66:403–11. doi: 10.1093/biomet/66.3.403.
    1. Juraska M, Huang Y, Gilbert PB. Inference on treatment effect modification by biomarker response in a three-phase sampling design. Biostatistics. 2018. 10.1093/biostatistics/kxy074.
    1. Hall P, Racine J, Li Q. Cross-validation and the estimation of conditional probability densities. J Am Stat Assoc. 2004;99:1015–26. doi: 10.1198/016214504000000548.
    1. Juraska M.pssmooth: Flexible and Efficient Evaluation of Principal Surrogates/Treatment Effect Modifiers. 2019. R package version 1.0.2. . Accessed 16 Apr 2019.
    1. Petrie JG, Ohmit SE, Truscon R, Johnson e, Braun TM, Levine MZ, et al. Modest waning of influenza vaccine efficacy and antibody titers during the 2007–2008 influenza season. J Infect Dis. 2016;214(8):1142–9. doi: 10.1093/infdis/jiw105.
    1. Coudeville L, Andre P, Bailleux F, Weber F, Plotkin S. A new approach to estimate vaccine efficacy based on immunogenicity data applied to influenza vaccines administered by the intradermal or intramuscular routes. Hum Vaccin. 2010;6(10):841–8. doi: 10.4161/hv.6.10.12636.
    1. Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng N-Y, et al. Limited efficacy of inactivated influenza vaccine in elderly individuals is associated with decreased production of vaccine-specific antibodies. J Clin Invest. 2011;121(8):3109–19. doi: 10.1172/JCI57834.
    1. Prentice R. Surrogate endpoints in clinical trials: definition and operational criteria. Stat Med. 1989;8:431–40. doi: 10.1002/sim.4780080407.
    1. Dunning AJ, Kensler J, Coudeville L, Bailleux F. Some extensions in continuous models for immunological correlates of protection. BMC Med Res Methodol. 2015;15(1):107. doi: 10.1186/s12874-015-0096-9.
    1. Dunning AJ, DiazGranados CA, Voloshen T, Hu B, Landolfi VA, Talbot HK. Correlates of protection against influenza in the elderly: results from an influenza vaccine efficacy trial. Clin Vaccine Immunol. 2016;23(3):228–35. doi: 10.1128/CVI.00604-15.
    1. Fiore AE, Shay DK, Broder K, Iskander JK, Uyeki TM, Mootrey G, Bresee JS, Cox NS. Prevention and control of influenza: recommendations of the advisory committee on immunization practices (acip), 2008. MMWR. Recommendations and reports: Morbidity and mortality weekly report. Recommendations and reports. 2008;57(RR-7):1–60.
    1. McElhaney JE, Coler RN, Baldwin SL. Immunologic correlates of protection and potential role for adjuvants to improve influenza vaccines in older adults. Expert review of vaccines. 2013;12(7):759–66. doi: 10.1586/14760584.2013.811193.
    1. Tsang JS, Schwartzberg PL, Kotliarov Y, Biancotto A, Xie Z, Germain RN, et al. Global analyses of human immune variation reveal baseline predictors of postvaccination responses. Cell. 2014;157(2):499–513. doi: 10.1016/j.cell.2014.03.031.

Source: PubMed

3
S'abonner