Safety and Immunogenicity of Malaria Vectored Vaccines Given with Routine Expanded Program on Immunization Vaccines in Gambian Infants and Neonates: A Randomized Controlled Trial

Victorine A Mensah, Sophie Roetynck, Ebrima K Kanteh, Georgina Bowyer, Amy Ndaw, Francis Oko, Carly M Bliss, Ya Jankey Jagne, Riccardo Cortese, Alfredo Nicosia, Rachel Roberts, Flavia D'Alessio, Odile Leroy, Babacar Faye, Beate Kampmann, Badara Cisse, Kalifa Bojang, Stephen Gerry, Nicola K Viebig, Alison M Lawrie, Ed Clarke, Egeruan B Imoukhuede, Katie J Ewer, Adrian V S Hill, Muhammed O Afolabi, Victorine A Mensah, Sophie Roetynck, Ebrima K Kanteh, Georgina Bowyer, Amy Ndaw, Francis Oko, Carly M Bliss, Ya Jankey Jagne, Riccardo Cortese, Alfredo Nicosia, Rachel Roberts, Flavia D'Alessio, Odile Leroy, Babacar Faye, Beate Kampmann, Badara Cisse, Kalifa Bojang, Stephen Gerry, Nicola K Viebig, Alison M Lawrie, Ed Clarke, Egeruan B Imoukhuede, Katie J Ewer, Adrian V S Hill, Muhammed O Afolabi

Abstract

Background: Heterologous prime-boost vaccination with chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) encoding multiple epitope string thrombospondin-related adhesion protein (ME-TRAP) has shown acceptable safety and promising immunogenicity in African adult and pediatric populations. If licensed, this vaccine could be given to infants receiving routine childhood immunizations. We therefore evaluated responses to ChAd63 MVA ME-TRAP when co-administered with routine Expanded Program on Immunization (EPI) vaccines.

Methods: We enrolled 65 Gambian infants and neonates, aged 16, 8, or 1 week at first vaccination and randomized them to receive either ME-TRAP and EPI vaccines or EPI vaccines only. Safety was assessed by the description of vaccine-related adverse events (AEs). Immunogenicity was evaluated using IFNγ enzyme-linked immunospot, whole-blood flow cytometry, and anti-TRAP IgG ELISA. Serology was performed to confirm all infants achieved protective titers to EPI vaccines.

Results: The vaccines were well tolerated in all age groups with no vaccine-related serious AEs. High-level TRAP-specific IgG and T cell responses were generated after boosting with MVA. CD8+ T cell responses, previously found to correlate with protection, were induced in all groups. Antibody responses to EPI vaccines were not altered significantly.

Conclusion: Malaria vectored prime-boost vaccines co-administered with routine childhood immunizations were well tolerated. Potent humoral and cellular immunity induced by ChAd63 MVA ME-TRAP did not reduce the immunogenicity of co-administered EPI vaccines, supporting further evaluation of this regimen in infant populations.

Clinical trial registration: The clinical trial was registered on http://Clinicaltrials.gov (NCT02083887) and the Pan-African Clinical Trials Registry (PACTR201402000749217).

Keywords: cellular immune response; clinical trials; cytokines; malaria; vaccines.

Figures

Figure 1
Figure 1
Flow of study design and volunteer enrollment. Seventy-two infants and neonates were screened for eligibility across the three age groups and 65 eligible infants were enrolled, randomized, vaccinated according to randomization list and followed up for 252 days. Of 24 infants screened in the 16-week-old group, mothers of 3 infants withdrew consent before enrollment while 1 infant was excluded due to markedly raised alanine transaminase. Among those randomized to the vaccine arm, one infant had fatal SAE that was non-related to the study vaccines and another was lost to follow-up due to relocation of mother, while only one infant did not complete the follow-up in the control arm. Twenty-two infants were screened for the 8-week-old cohort and all were eligible. Only the first 20 were randomized into 15 vaccine group and 5 controls. Similarly, 26 mother-newborn pairs were screened for the 1-week-old group. A mother withdrew consent before enrollment and the remaining 25 neonates were randomized into 15 vaccinees and 10 controls. All study infants in the 8- and 1-week-old groups completed the study follow-up. SAE, serious adverse event.
Figure 2
Figure 2
Ex vivo IFNγ enzyme-linked immunospot responses to multiple epitope string thrombospondin-related adhesion protein (ME-TRAP) pre- and post-vaccination with ChAd63 and MVA ME-TRAP in Gambian infants. (A–C) Scatter plots show individual responses to ME-TRAP in both vaccinees (in black) and controls (in white) of each age group. (A) 16-week-old; (B) 8-week-old; (C) 1-week-old infants. (D) Geometric mean ME-TRAP responses in each age group over period of follow-up. LLD, lower limit of detection of the assay. Increases in responses over the time course were analyzed using two-tailed Friedman test. Responses between individual time points were compared within each age group using two-tailed Wilcoxon analysis, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. Lines and numbers in blue denote geometric mean. Numbers shown in black represent the percentage of responders among the vaccinated infants for each time point.
Figure 3
Figure 3
Peak enzyme-linked immunospot (ELISpot) responses to multiple epitope string thrombospondin-related adhesion protein (ME-TRAP) at D63, 7 days post-MVA. (A) Peak responses from vaccinated infants expressed per million PBMC. (B) Lymphocyte counts per milliliter of blood at D63. (C) ELISpot responses expressed per ml of blood. LLD, lower limit of detection of the assay. Peak responses and cell counts were compared across groups using Kruskal–Wallis test with Dunn’s multiple comparison post-test, *p < 0.05. Lines and numbers indicate geometric mean.
Figure 4
Figure 4
T cells induced by vaccination with thrombospondin-related adhesion protein (TRAP) antigen from P. falciparum T9/96 strain are highly cross-reactive. Scatter plots show individual ex vivo IFNγ enzyme-linked immunospot responses to TRAP peptides from T9/96 and 3D7 strains, respectively. (A) Responses pre-vaccination. (B) Responses 3 weeks post-prime with ChAd63 multiple epitope string thrombospondin-related adhesion protein (ME-TRAP). (C,D) Responses at day 63 and day 168, 7 and 112 days post-boost with MVA ME-TRAP, respectively. Numbers indicate the percentage of positive responses for each age group at each time point for both parasite strains. LLD, lower limit of detection of the assay. Lines represent geometric mean.
Figure 5
Figure 5
CD4+ and CD8+ T cell contribution to total IFNγ response to thrombospondin-related adhesion protein (TRAP). Pie charts display the proportion of IFNγ produced by CD4+ (black) and CD8+ (gray) T cells as a fraction of the total IFNγ response to TRAP post-prime and post-boost vaccination. Here, any negative response was excluded from analysis.
Figure 6
Figure 6
Multifunctionality of peak CD4+ and CD8+ T cell responses to thrombospondin-related adhesion protein (TRAP) at day 63 (1 week post-boosting). Cytokine production from CD4+(A) and CD8+(B) T cells was analyzed using Boolean gating analysis. Pie charts illustrate the relative frequency of each combination of cytokines, indicated in the bar chart below, as a fraction of the total cytokine response. Bar charts show the absolute frequency of each subset in response to stimulation with TRAP peptides, compared between each age group. Bars show median responses in this case as geometric mean is not available in SPICE. Any response which was less than or equal to the background (unstimulated) response was replaced by lower limit of detection value in bar chart and excluded from pie charts.
Figure 7
Figure 7
Antibody responses to Expanded Program on Immunization (EPI) and multiple epitope string thrombospondin-related adhesion protein (ME-TRAP) vaccination. (A) IgG responses to vaccination with ChAd63 ME-TRAP (day 0) and MVA ME-TRAP (day 56). Scatter plots show peak responses post-prime (day 21) and post-boost (day 63). Group 1—16 weeks, group 2—8 weeks, and group 3—1 week old at first vaccination. All comparisons use Kruskal–Wallis with Dunn’s post-test, comparing responses at day 0 with post-vaccination responses for groups 1–3 combined. No significant differences between groups were observed at any time point. (B) Comparison of peak IgG responses with data from previous trials (14). Responses in all groups combined from the present study were compared to those from a study in the same population that received the same dose and vaccine regimen in the absence of EPI coadministration. (C,D) Responses to EPI vaccination at 24 weeks, 8 weeks after last primary EPI, among ME-TRAP vaccinees and controls for diphtheria, tetanus, pertussis, Haemophilus influenza B, Hepatitis B, and polio serotypes 1–3 (C) and pneumococcal immunization (PCV 13) by serotype (D). No significant differences between groups were observed (*p < 0.05, **p < 0.01, ****p < 0.001).

References

    1. WHO. World Malaria Report 2016. Geneva: World Health Organization; (2016).
    1. Crompton PD, Pierce SK, Miller LH. Advances and challenges in malaria vaccine development. J Clin Invest (2010) 120(12):4168–78.10.1172/JCI44423
    1. Greenwood B, Targett G. Do we still need a malaria vaccine? Parasite Immunol (2009) 31(9):582–6.10.1111/j.1365-3024.2009.01140.x
    1. Greenwood B, Doumbo OK. Implementation of the malaria candidate vaccine RTS,S/AS01. Lancet (2016) 387(10016):318–9.10.1016/S0140-6736(15)00807-7
    1. Radosevic K, Rodriguez A, Lemckert A, Goudsmit J. Heterologous prime-boost vaccinations for poverty-related diseases: advantages and future prospects. Expert Rev Vaccines (2009) 8(5):577–92.10.1586/erv.09.14
    1. Rodriguez A, Mintardjo R, Tax D, Gillissen G, Custers J, Pau MG, et al. Evaluation of a prime-boost vaccine schedule with distinct adenovirus vectors against malaria in rhesus monkeys. Vaccine (2009) 27(44):6226–33.10.1016/j.vaccine.2009.07.106
    1. Ratto-Kim S, Currier JR, Cox JH, Excler JL, Valencia-Micolta A, Thelian D, et al. Heterologous prime-boost regimens using rAd35 and rMVA vectors elicit stronger cellular immune responses to HIV proteins than homologous regimens. PLoS One (2012) 7(9):e45840.10.1371/journal.pone.0045840
    1. O’Hara GA, Duncan CJ, Ewer KJ, Collins KA, Elias SC, Halstead FD, et al. Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J Infect Dis (2012) 205(5):772–81.10.1093/infdis/jir850
    1. Ewer KJ, O’Hara GA, Duncan CJ, Collins KA, Sheehy SH, Reyes-Sandoval A, et al. Protective CD8+ T-cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation. Nat Commun (2013) 4:2836.10.1038/ncomms3836
    1. Ogwang C, Afolabi M, Kimani D, Jagne YJ, Sheehy SH, Bliss CM, et al. Safety and immunogenicity of heterologous prime-boost immunisation with Plasmodium falciparum malaria candidate vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in healthy Gambian and Kenyan adults. PLoS One (2013) 8(3):e57726.10.1371/journal.pone.0057726
    1. Kimani D, Jagne YJ, Cox M, Kimani E, Bliss CM, Gitau E, et al. Translating the immunogenicity of prime-boost immunization with ChAd63 and MVA ME-TRAP from malaria naive to malaria-endemic populations. Mol Ther (2014) 22(11):1992–2003.10.1038/mt.2014.109
    1. Ogwang C, Kimani D, Edwards NJ, Roberts R, Mwacharo J, Bowyer G, et al. Prime-boost vaccination with chimpanzee adenovirus and modified vaccinia Ankara encoding TRAP provides partial protection against Plasmodium falciparum infection in Kenyan adults. Sci Transl Med (2015) 7(286):286re5.10.1126/scitranslmed.aaa2373
    1. Afolabi MO, Tiono AB, Adetifa UJ, Yaro JB, Drammeh A, Nebie I, et al. Safety and immunogenicity of ChAd63 and MVA ME-TRAP in West African children and infants. Mol Ther (2016) 24(8):1470–7.10.1038/mt.2016.83
    1. Bliss CM, Drammeh A, Bowyer G, Sanou GS, Jagne YJ, Ouedraogo O, et al. Viral vector malaria vaccines induce high-level T cell and antibody responses in West African children and infants. Mol Ther (2017) 25(2):547–59.10.1016/j.ymthe.2016.11.003
    1. Ota MO, Odutola AA, Owiafe PK, Donkor S, Owolabi OA, Brittain NJ, et al. Immunogenicity of the tuberculosis vaccine MVA85A is reduced by coadministration with EPI vaccines in a randomized controlled trial in Gambian infants. Sci Transl Med (2011) 3(88):88ra56.10.1126/scitranslmed.3002461
    1. McMurray DN. Do new TB vaccines have a place in the expanded program on immunization? Expert Rev Vaccines (2011) 10(12):1675–7.10.1586/erv.11.144
    1. Rts SCTP, Agnandji ST, Lell B, Fernandes JF, Abossolo BP, Methogo BG, et al. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med (2012) 367(24):2284–95.10.1056/NEJMoa1208394
    1. Mwesigwa J, Okebe J, Affara M, Di Tanna GL, Nwakanma D, Janha O, et al. On-going malaria transmission in The Gambia despite high coverage of control interventions: a nationwide cross-sectional survey. Malar J (2015) 14:314.10.1186/s12936-015-0829-6
    1. Odutola AA, Afolabi MO, Jafali J, Baldeh I, Owolabi OA, Owiafe P, et al. Haematological and biochemical reference values of Gambian infants. Trop Med Int Health (2014) 19(3):275–83.10.1111/tmi.12245
    1. Hanekom WA, Hughes J, Mavinkurve M, Mendillo M, Watkins M, Gamieldien H, et al. Novel application of a whole blood intracellular cytokine detection assay to quantitate specific T-cell frequency in field studies. J Immunol Methods (2004) 291(1–2):185–95.10.1016/j.jim.2004.06.010
    1. de Voer RM, Schepp RM, Versteegh FG, van der Klis FR, Berbers GA. Simultaneous detection of Haemophilus influenzae type b polysaccharide-specific antibodies and Neisseria meningitidis serogroup A, C, Y, and W-135 polysaccharide-specific antibodies in a fluorescent-bead-based multiplex immunoassay. Clin Vaccine Immunol (2009) 16(3):433–6.10.1128/CVI.00364-08
    1. Elberse KE, Tcherniaeva I, Berbers GA, Schouls LM. Optimization and application of a multiplex bead-based assay to quantify serotype-specific IgG against Streptococcus pneumoniae polysaccharides: response to the booster vaccine after immunization with the pneumococcal 7-valent conjugate vaccine. Clin Vaccine Immunol (2010) 17(4):674–82.10.1128/CVI.00408-09
    1. van Gageldonk PG, van Schaijk FG, van der Klis FR, Berbers GA. Development and validation of a multiplex immunoassay for the simultaneous determination of serum antibodies to Bordetella pertussis, diphtheria and tetanus. J Immunol Methods (2008) 335(1–2):79–89.10.1016/j.jim.2008.02.018
    1. Roederer M, Nozzi JL, Nason MC. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A (2011) 79(2):167–74.10.1002/cyto.a.21015
    1. Shearer WT, Rosenblatt HM, Gelman RS, Oyomopito R, Plaeger S, Stiehm ER, et al. Lymphocyte subsets in healthy children from birth through 18 years of age: the Pediatric AIDS Clinical Trials Group P1009 study. J Allergy Clin Immunol (2003) 112(5):973–80.10.1016/j.jaci.2003.07.003
    1. Robson KJ, Dolo A, Hackford IR, Doumbo O, Richards MB, Keita MM, et al. Natural polymorphism in the thrombospondin-related adhesive protein of Plasmodium falciparum. Am J Trop Med Hyg (1998) 58(1):81–9.10.4269/ajtmh.1998.58.81
    1. Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Whittle H, Lambert PH, et al. Hepatitis B immunisation induces higher antibody and memory Th2 responses in new-borns than in adults. Vaccine (2004) 22(3–4):511–9.10.1016/j.vaccine.2003.07.020
    1. Vekemans J, Ota MO, Wang EC, Kidd M, Borysiewicz LK, Whittle H, et al. T cell responses to vaccines in infants: defective IFNgamma production after oral polio vaccination. Clin Exp Immunol (2002) 127(3):495–8.10.1046/j.1365-2249.2002.01788.x
    1. Martinez X, Brandt C, Saddallah F, Tougne C, Barrios C, Wild F, et al. DNA immunization circumvents deficient induction of T helper type 1 and cytotoxic T lymphocyte responses in neonates and during early life. Proc Natl Acad Sci U S A (1997) 94(16):8726–31.10.1073/pnas.94.16.8726
    1. Vekemans J, Amedei A, Ota MO, D’Elios MM, Goetghebuer T, Ismaili J, et al. Neonatal bacillus Calmette-Guerin vaccination induces adult-like IFN-gamma production by CD4+ T lymphocytes. Eur J Immunol (2001) 31(5):1531–5.10.1002/1521-4141(200105)31:5<1531::AID-IMMU1531>;2-1
    1. Murray RA, Mansoor N, Harbacheuski R, Soler J, Davids V, Soares A, et al. Bacillus Calmette Guerin vaccination of human newborns induces a specific, functional CD8+ T cell response. J Immunol (2006) 177(8):5647–51.10.4049/jimmunol.177.8.5647
    1. Hermann E, Truyens C, Alonso-Vega C, Even J, Rodriguez P, Berthe A, et al. Human fetuses are able to mount an adultlike CD8 T-cell response. Blood (2002) 100(6):2153–8.
    1. Marchant A, Appay V, Van Der Sande M, Dulphy N, Liesnard C, Kidd M, et al. Mature CD8(+) T lymphocyte response to viral infection during fetal life. J Clin Invest (2003) 111(11):1747–55.10.1172/JCI200317470
    1. Hodgson SH, Ewer KJ, Bliss CM, Edwards NJ, Rampling T, Anagnostou NA, et al. Evaluation of the efficacy of ChAd63-MVA vectored vaccines expressing circumsporozoite protein and ME-TRAP against controlled human malaria infection in malaria-naive individuals. J Infect Dis (2015) 211(7):1076–86.10.1093/infdis/jiu579

Source: PubMed

3
Suscribir