Short-Lived Immunity After 17DD Yellow Fever Single Dose Indicates That Booster Vaccination May Be Required to Guarantee Protective Immunity in Children

Ana Carolina Campi-Azevedo, Laise Rodrigues Reis, Vanessa Peruhype-Magalhães, Jordana Grazziela Coelho-Dos-Reis, Lis Ribeiro Antonelli, Cristina Toscano Fonseca, Christiane Costa-Pereira, Elaine Maria Souza-Fagundes, Ismael Artur da Costa-Rocha, Juliana Vaz de Melo Mambrini, Jandira Aparecida Campos Lemos, José Geraldo Leite Ribeiro, Iramaya Rodrigues Caldas, Luiz Antônio Bastos Camacho, Maria de Lourdes de Sousa Maia, Tatiana Guimarães de Noronha, Sheila Maria Barbosa de Lima, Marisol Simões, Marcos da Silva Freire, Reinaldo de Menezes Martins, Akira Homma, Pedro Luiz Tauil, Pedro Fernando Costa Vasconcelos, Alessandro Pecego Martins Romano, Carla Magda Domingues, Andréa Teixeira-Carvalho, Olindo Assis Martins-Filho, Ana Carolina Campi-Azevedo, Laise Rodrigues Reis, Vanessa Peruhype-Magalhães, Jordana Grazziela Coelho-Dos-Reis, Lis Ribeiro Antonelli, Cristina Toscano Fonseca, Christiane Costa-Pereira, Elaine Maria Souza-Fagundes, Ismael Artur da Costa-Rocha, Juliana Vaz de Melo Mambrini, Jandira Aparecida Campos Lemos, José Geraldo Leite Ribeiro, Iramaya Rodrigues Caldas, Luiz Antônio Bastos Camacho, Maria de Lourdes de Sousa Maia, Tatiana Guimarães de Noronha, Sheila Maria Barbosa de Lima, Marisol Simões, Marcos da Silva Freire, Reinaldo de Menezes Martins, Akira Homma, Pedro Luiz Tauil, Pedro Fernando Costa Vasconcelos, Alessandro Pecego Martins Romano, Carla Magda Domingues, Andréa Teixeira-Carvalho, Olindo Assis Martins-Filho

Abstract

The Yellow Fever (YF) vaccination is recommended for people living in endemic areas and represents the most effective strategy to reduce the risk of infection. Previous studies have warned that booster regimens should be considered to guarantee the long-term persistence of 17DD-YF-specific memory components in adults living in areas with YF-virus circulation. Considering the lower seroconversion rates observed in children (9-12 months of age) as compared to adults, this study was designed in order to access the duration of immunity in single-dose vaccinated children in a 10-years cross-sectional time-span. The levels of neutralizing antibodies (PRNT) and the phenotypic/functional memory status of T and B-cells were measured at a baseline, 30-45 days, 1, 2, 4, 7, and 10 years following primary vaccination. The results revealed that a single dose induced 85% of seropositivity at 30-45 days and a progressive time-dependent decrease was observed as early as 2 years and declines toward critical values (below 60%) at time-spans of ≥4-years. Moreover, short-lived YF-specific cellular immunity, mediated by memory T and B-cells was also observed after 4-years. Predicted probability and resultant memory analysis emphasize that correlates of protection (PRNT; effector memory CD8+ T-cells; non-classical memory B-cells) wane to critical values within ≥4-years after primary vaccination. Together, these results clearly demonstrate the decline of 17DD-YF-specific memory response along time in children primarily vaccinated at 9-12 months of age and support the need of booster regimen to guarantee the long-term persistence of memory components for children living in areas with high risk of YF transmission.

Keywords: 17DD vaccine; cellular memory; children; neutralizing antibodies; yellow fever.

Copyright © 2019 Campi-Azevedo, Reis, Peruhype-Magalhães, Coelho-dos-Reis, Antonelli, Fonseca, Costa-Pereira, Souza-Fagundes, Costa-Rocha, Mambrini, Lemos, Ribeiro, Caldas, Camacho, Maia, de Noronha, de Lima, Simões, Freire, Martins, Homma, Tauil, Vasconcelos, Romano, Domingues, Teixeira-Carvalho and Martins-Filho.

Figures

Figure 1
Figure 1
Neutralizing antibody titers after 17DD-YF primary vaccination in children. PRNT titers were measured in Ecteola-treated plasma samples (19) from non-vaccinated children at baseline NV(day 0)/(○, n = 47) and at different times after primary vaccination: PV(day 30–45)/(, n = 47), PV(year 1)/(, n = 141), PV(year 2)/(, n = 114), PV(year 4)/(, n = 128), PV(year 7)/(, n = 116), and PV(year 10)/(°, n = 127), as described previously by Simões et al. (20). (A) The PRNT levels were expressed in reverse of serum dilution. (B) Proportion of PRNT seropositivity (PRNT>1:10) were calculated for each group and the results expressed as seropositivity rates at baseline NV(day 0) [] and at different times after primary vaccination: PV(day 30–45) [], PV(year 1) [], PV(year 2) [], PV(year 4) [], PV(year 7) [], and PV(year 10) [], considering the serum dilution >1:10 as the cut-off (dashed line). (C) Correlation and logistic regression were employed to determine the wane of PRNT levels along time continuum and the results expressed as reverse of serum dilution and predicated probability, respectively. Statistical analysis was carried out as described in Methods. In all cases, significant differences at p < 0.05 were underscored by using letters “a,” “b,” “c,” and “d” for comparisons with NV(day 0), PV(day 30–45), PV(year 1), and PV(year 2), respectively and the p-values provide in the figure. Spearman correlation indices as well as Likelihood and Odds ratio are provided in the figure. Gray rectangle highlights the critical decrease of PRNT seropositivity rates ≥4 years after primary vaccination.
Figure 2
Figure 2
Phenotypic and functional memory biomarkers after 17DD-YF primary vaccination in children. The analysis of 17DD-YF-specific memory was measured upon in vitro 17DD-YF antigen recall as described previously by Campi-Azevedo et al. (8) for non-vaccinated children at baseline NV(day 0)/(, n = 47) and at different times after primary vaccination: PV(day 30–45)/(, n = 47), PV(year 1)/(, n = 141), PV(year 2)/(, n = 114), PV(year 4)/(, n = 128), PV(year 7)/(, n = 116), and PV(year 10)/(, n = 127). (A) Flow cytometric staining were used to quantify phenotypic features of T-cell memory subsets: Naïve T-cells/(NCD4;NCD8)/CD27+CD45RO−; early Effector Memory T-cells/(eEfCD4;eEfCD8)/CD27−CD45RO− Central Memory T-cells/(CMCD4;CMCD8)/CD27+CD45RO+ Effector Memory T-cells/(EMCD4;EMCD8)/CD27−CD45RO+ and B-cell memory subsets: Naïve B-cells/(NCD19)/CD27−IgD+; Non-classical Memory B-cells/(nCMCD19)/CD27+IgD+ and Classical Memory B-cells/(CMCD19)/CD27+IgD−. (B) Flow cytometric staining were also performed to quantity functional CD8+ T-cells producing TNF-α, IFN-γ and IL-5. The data were reported as median values ± inter-quartile range for 17DD-YF-stimulated Culture/non-stimulated Control Culture Index as described in Methods, highlighting the equivalence ratio by dashed line (Index = 1.0). Significant differences at p < 0.05 were underscored by using asterisk (*) to identify differences between NV(day 0) vs. PV(day 30–45) and intergroup differences identified by connecting lines.
Figure 3
Figure 3
Phenotypic and functional biomarker signatures after 17DD-YF primary vaccination in children. (A) Biomarker signatures of reference groups NV(day 0) [] and PV(day 30–45) [] were assembled to select biomarkers above the 75th percentile with proportions higher than the 25% in each group (white/black background rectangles). The selected biomarkers were underscored by asterisk (*) to identify differences between NV(day 0) vs. PV(day 30–45). (B) Venn diagram report was employed to identify the set of biomarkers selectively increased in [PV(day 30–45) vs. NV(day 0)]. The attributes EMCD4, EMCD8, nCMCD19, TNFCD8, and IFNCD8 were underscored as PV (day 30–45)-selective biomarkers. These attributes were tagged in bold underline format and employed for follow-up analysis overtime after 17DD-YF primary vaccination. Biomarkers with proportion higher than the 25% were underscored by asterisk (*) to identify differences between NV(day 0) vs. PV(day 30–45). (C) Overlaid signatures of selected biomarkers were assembled to identify changes in the 17DD-YF specific phenotypic and functional features at different times after primary vaccination: PV(day 30–45) [], PV(year 1) [], PV(year 2) [], PV(year 4) [], PV(year 7) [], and PV(year 10) []. Dashed rectangles underscore the critical decline of selected biomarkers overtime after primary vaccination with absence of EMCD8 ≥ 4 years after primary vaccination.
Figure 4
Figure 4
Descriptive analysis of selected biomarkers after 17DD-YF primary vaccination in children. (A) Prevalence of Biomarkers (PRNT>1:10, EMCD4, EMCD8, nCMCD19, TNFCD8, and IFNCD8) above the 75th percentile cut-off. Data are presented as proportion of subjects with biomarkers above the cut-off at baseline NV(day 0) [] and at different times after primary vaccination: PV(day 30–45) [], PV(year 1) [], PV(year 2) [], PV(year 4) [], PV(year 7) [], and PV(year 10) []. (B) Number of Biomarkers (PRNT>1:10, EMCD4, EMCD8, nCMCD19, TNFCD8, and IFNCD8) above the 75th Quartile cut-off. Data are presented as mean (min to max) number of biomarkers above the cut-off at baseline NV(day 0) [] and at different times after primary vaccination: PV(day 30–45) [], PV(year 1) [], PV(year 2) [], PV(year 4) [], PV(year 7) [], and PV(year 10) [] as well as PV(≤year 2) [], PV(≥year 4) []. Significant differences at p < 0.05 are underscored by using letters “a,” “b,” “c,” and “d” for comparisons with NV(day 0), PV(day 30–45), PV(year 1), and PV(year 2), respectively. Intergroup differences are identified by connecting lines. The p-values are provided in the figure.
Figure 5
Figure 5
Predictive capacity of biomarkers to monitor the 17DD-YF memory after primary vaccination in children. The Receiver Operating Characteristic (ROC) curves were used to estimate the capacity of time as a predictor of changes in biomarker levels to monitor the 17DD-YF memory after primary vaccination in children. Logistic and multinomial regression models were constructed to evaluate the association between time after vaccination and changes in the biomarker levels. Following, the fitted regression model was employed to calculate the predicted probabilities for each biomarker (A) isolated or (B) combined along time continuum. The Area Under the ROC Curves (AUC) were employed for comparative analysis of predictive capacity amongst biomarkers and the values provided in the figure. The gray background highlights the top three isolated biomarkers and the best combination of predictor biomarkers to monitor the 17DD-YF memory after primary vaccination in children.
Figure 6
Figure 6
Resultant memory (PRNT, EMCD8, or nCMCD19) after 17DD-YF Primary vaccination in children. The resultant memory status (PRNT, EMCD8, or nCMCD19) were assessed at individual level to define the overall proportion of subjects presenting None (), PRNT (), cellular memory “EMCD8 and/or nCMCD19” () or both attributes “PRNT and cellular memory” () at distinct time-points before/after primary 17DD-YF vaccination, including: NV(day 0), PV(≤year 2), PV(≥year 4) as well as PV(day 30–45), PV(year 1), PV(year 2), PV(year 4), PV(year 7), and PV(year 10). Significant differences (p < 0.05) of resultant memory status amongst study groups were assessed by Chi-square test and represented by letter “a” as compared to NV(day 0).

References

    1. Areas with Risk of Yellow fever Virus Transmission in Africa South America. (2019). Available online at:
    1. World Health Organization Yellow Fever Fact Sheet. (2019). Available online at:
    1. The Lancet Yellow fever: a major threat to public health. Lancet. (2018) 391:402 10.1016/S0140-6736(18)30152-1
    1. World Health Organization Eliminate Yellow fever Epidemics (EYE): A Global Strategy 2017–2026. (2017). Available online at:
    1. World Health Organization Vaccines and vaccination against yellow fever. WHO position paper – June 2013. Wkly Epidemiol Record. (2013) 88:269–83.
    1. World Health Organization Background paper on yellow fever vaccine. In: Vaccine Position Papers. Geneva, SAGE Working Group; (2013).
    1. Caldas IR, Camacho LA, Martins-Filho OA, Maia M de L, Freire M da S, Torres C de R, et al. Duration of post-vaccination immunity against yellow fever in adults. Vaccine. (2014) 32:4977–84. 10.1016/j.vaccine.2014.07.021
    1. Campi-Azevedo AC, Costa-Pereira C, Antonelli LR, Fonseca CT, Teixeira-Carvalho A, Villela-Rezende G, et al. . Booster dose after 10 years is recommended following 17DD-YF primary vaccination. Hum Vaccin Immunother. (2016) 12:491–502. 10.1080/21645515.2015.1082693
    1. Costa-Pereira C, Campi-Azevedo AC, Coelho-dos-Reis JG, Peruhype-Magalhāes V, Araújo MSS, Antonelli LRV, et al. . Multi-parameter approach to evaluate the timing of memory status after primary 17DD-YF vaccination. PLoS Neglec Trop Dis. (2018) 12:e0006462. 10.1371/journal.pntd.0006462
    1. Vasconcelos PF. Single shot of 17D vaccine may not confer life-long protection against yellow fever. Mem Inst Oswaldo Cruz. (2018) 113:135–7. 10.1590/0074-02760170347
    1. Patel D, Simons H. Yellow fever vaccination: is one dose always enough? Travel Med Infect Dis. (2013) 11:266–73. 10.1016/j.tmaid.2013.08.007
    1. Grobusch MP, Goorhuis A, Wieten RW, Verberk JD, Jonker EF, van Genderen PJ, et al. . Yellow fever revaccination guidelines change – a decision too feverish? Clin Microbiol Infect. (2013) 19:885–6. 10.1111/1469-0691.12332
    1. Amanna IJ, Slifka MK. Questions regarding the safety and duration of immunity following live yellow fever vaccination. Expert Rev Vaccines. (2016) 15:1519–33. 10.1080/14760584.2016.1198259
    1. Estofolete CF, Nogueira ML. Is a dose of 17D vaccine in the current context of Yellow Fever enough? Braz J Microbiol. (2018) 49:683–4. 10.1016/j.bjm.2018.02.003
    1. Nascimento Silva JR, Camacho LA, Siqueira MM, Freire Mde S, Castro YP, Maia Mde L, et al. . Mutual interference on the immune response to yellow fever vaccine and a combined vaccine against measles, mumps and rubella. Vaccine. (2011) 29:6327–34. 10.1016/j.vaccine.2011.05.019
    1. Niedrig M, Lademann M, Emmerich P, Lafrenz M. Assessment of IgG antibodies against yellow fever virus after vaccination with 17D by different assays: neutralization test, haemagglutination inhibition test, immunofluorescence assay and ELISA. Trop Med Int Health. (1999) 4:867–71. 10.1046/j.1365-3156.1999.00496.x
    1. Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. . Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. (2019) 10:116–25. 10.1038/ni.1688
    1. Ahmed R, Akondy RS. Insights into human CD8(+) T-cell memory using the yellow fever and smallpox vaccines. Immunol Cell Biol. (2011) 89:340–5. 10.1038/icb.2010.155
    1. Campi-Azevedo AC, Peruhype-Magalhães V, Coelho-Dos-Reis JG, Costa-Pereira C, Yamamura AY, Lima SMB, et al. . Heparin removal by ecteola-cellulose pre-treatment enables the use of plasma samples for accurate measurement of anti-Yellow fever virus neutralizing antibodies. J Immunol Methods. (2017) 448:9–20. 10.1016/j.jim.2017.05.002
    1. Simões M, Camacho LAB, Yamamura AMY, Miranda EH, Cajaraville ACRA, Freire MS. Evaluation of accuracy and reliability of the plaque reduction neutralization test (micro-PRNT) in detection of yellow fever virus antibodies. Biologicals. (2012) 40:399–404. 10.1016/j.biologicals.2012.09.005
    1. Poland JD, Calisher CH, Monath TP, Downs WG, Murphy K. Persistence of neutralizing antibody 30–35 years after immunization with 17D yellow fever vaccine. Bull World Health Organ. (1981) 59:895–900.
    1. Gotuzzo E, Yactayo S, Córdova E. Review article: Efficacy and duration of immunity after yellow fever vaccination: systematic review on the need for a booster every 10 years. Am J Trop Med Hyg. (2013) 89:434–44. 10.4269/ajtmh.13-0264
    1. Wieten RW, Jonker EF, van Leeuwen EM, Remmerswaal EB, Ten Berge IJ, de Visser AW, et al. . A single 17D yellow fever vaccination provides lifelong immunity; characterization of yellow-fever-specific neutralizing antibody and T-cell responses after vaccination. PLoS One. (2016) 11:e0149871. 10.1371/journal.pone.0149871
    1. Muyanja E, Ssemaganda A, Ngauv P, Cubas R, Perrin H, Srinivasan D, et al. . Immune activation alters cellular and humoral responses to yellow fever 17D vaccine. J Clin Invest. (2014) 124:3147–58. 10.1172/JCI75429
    1. Gaucher D, Therrien R, Kettaf N, Angermann BR, Boucher G, Filali-Mouhim A, et al. . Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med. (2008) 205:3119–31. 10.1084/jem.20082292
    1. Collaborative Group for Studies with Yellow Fever Vaccine Randomized, double-blind, multicenter study of the immunogenicity and reactogenicity of 17DD and WHO 17D-213/77 yellow fever vaccines in children: implications for the Brazilian National Immunization Program. Vaccine. (2007) 25:3118–23. 10.1016/j.vaccine.2007.01.053
    1. Collaborative Group for Studies of Yellow Fever Vaccine A randomized double-blind clinical trial of two yellow fever vaccines prepared with substrains 17DD and 17D-213/77 in children nine-23 months old. Mem Inst Oswaldo Cruz. (2015) 110:771–80. 10.1590/0074-02760150176
    1. Luiza-Silva M, Campi-Azevedo AC, Batista MA, Martins MA, Avelar RS, da Silveira Lemos D, et al. . Cytokine signatures of innate and adaptive immunity in 17DD yellow fever vaccinated children and its association with the level of neutralizing antibody. J Infect Dis. (2011) 204:873–83. 10.1093/infdis/jir439
    1. Hepburn MJ, Kortepeter MG, Pittman PR, Boudreau EF, Mangiafico JA, Buck PA, et al. . Neutralizing antibody response to booster vaccination with the 17d yellow fever vaccine. Vaccine. (2006) 24:2843–9. 10.1016/j.vaccine.2005.12.055
    1. Kongsgaard M, Bassi MR, Rasmussen M, Skjødt K, Thybo S, Gabriel M, et al. . Adaptive immune responses to booster vaccination against yellow fever virus are much reduced compared to those after primary vaccination. Sci Rep. (2017) 7:662. 10.1038/s41598-017-00798-1
    1. Lindsey NP, Horiuchi KA, Fulton C, Panella AJ, Kosoy OI, Velez JO, et al. . Persistence of yellow fever virus-specific neutralizing antibodies after vaccination among US travellers. J Travel Med. (2018) 25:1–6. 10.1093/jtm/tay108
    1. Collaborative group for studies on yellow fever vaccines Duration of immunity in recipients of two doses of 17DD yellow fever vaccine. Vaccine. (2019) 37:5129–35. 10.1016/j.vaccine.2019.05.048
    1. Campi-Azevedo AC, Peruhype-Magalhāes V, Coelho-dos-Reis JG, Antonelli LRV, Costa-Pereira C, Speziali E, et al. 17DD-Yellow Fever re-vaccination regimens are required for long-term persistence of neutralizing antibodies and memory CD8+ T-cells in populations of endemic areas with high risk of yellow fever transmission. Emerg Infec Dis. (2019) 25:1511–21. 10.3201/eid2508.181432
    1. Campi-Azevedo AC, de Araújo-Porto LP, Luiza-Silva M, Batista MA, Martins MA, Sathler-Avelar R, et al. . 17DD and 17D-213/77 yellow fever substrains trigger a balanced cytokine profile in primary vaccinated children. PLoS ONE. (2012) 7:e49828. 10.1371/journal.pone.0049828
    1. Reinhardt B, Jaspert R, Niedrig M, Kostner C, L'age-Stehr J. Development of viremia and humoral and cellular parameters of immune activation after vaccination with yellow fever virus strain 17D: a model of human flavivirus infection. J Med Virol. (1998) 56:159–67. 10.1002/(SICI)1096-9071(199810)56:2<159::AID-JMV10>;2-B
    1. Sarkander J, Hojyo S, Tokoyoda K. Vaccination to gain humoral immune memory. Clin Transl Immunol. (2016) 5:e120. 10.1038/cti.2016.81
    1. Shi Y, Agematsu K, Ochs HD, Sugane K. Functional analysis of human memory B-cell subpopulations: IgD+CD27+ B cells are crucial in secondary immune response by producing high affinity IgM. Clin Immunol. (2003) 108:128–37. 10.1016/S1521-6616(03)00092-5

Source: PubMed

3
Suscribir