Safety and immunogenicity of the two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in children in Sierra Leone: a randomised, double-blind, controlled trial

Muhammed O Afolabi, David Ishola, Daniela Manno, Babajide Keshinro, Viki Bockstal, Baimba Rogers, Kwabena Owusu-Kyei, Alimamy Serry-Bangura, Ibrahim Swaray, Brett Lowe, Dickens Kowuor, Frank Baiden, Thomas Mooney, Elizabeth Smout, Brian Köhn, Godfrey T Otieno, Morrison Jusu, Julie Foster, Mohamed Samai, Gibrilla Fadlu Deen, Heidi Larson, Shelley Lees, Neil Goldstein, Katherine E Gallagher, Auguste Gaddah, Dirk Heerwegh, Benoit Callendret, Kerstin Luhn, Cynthia Robinson, Brian Greenwood, Maarten Leyssen, Macaya Douoguih, Bailah Leigh, Deborah Watson-Jones, EBL3001 study group

Abstract

Background: Children account for a substantial proportion of cases and deaths from Ebola virus disease. We aimed to assess the safety and immunogenicity of a two-dose heterologous vaccine regimen, comprising the adenovirus type 26 vector-based vaccine encoding the Ebola virus glycoprotein (Ad26.ZEBOV) and the modified vaccinia Ankara vector-based vaccine, encoding glycoproteins from the Ebola virus, Sudan virus, and Marburg virus, and the nucleoprotein from the Tai Forest virus (MVA-BN-Filo), in a paediatric population in Sierra Leone.

Methods: This randomised, double-blind, controlled trial was done at three clinics in Kambia district, Sierra Leone. Healthy children and adolescents aged 1-17 years were enrolled in three age cohorts (12-17 years, 4-11 years, and 1-3 years) and randomly assigned (3:1), via computer-generated block randomisation (block size of eight), to receive an intramuscular injection of either Ad26.ZEBOV (5 × 1010 viral particles; first dose) followed by MVA-BN-Filo (1 × 108 infectious units; second dose) on day 57 (Ebola vaccine group), or a single dose of meningococcal quadrivalent (serogroups A, C, W135, and Y) conjugate vaccine (MenACWY; first dose) followed by placebo (second dose) on day 57 (control group). Study team personnel (except for those with primary responsibility for study vaccine preparation), participants, and their parents or guardians were masked to study vaccine allocation. The primary outcome was safety, measured as the occurrence of solicited local and systemic adverse symptoms during 7 days after each vaccination, unsolicited systemic adverse events during 28 days after each vaccination, abnormal laboratory results during the study period, and serious adverse events or immediate reportable events throughout the study period. The secondary outcome was immunogenicity (humoral immune response), measured as the concentration of Ebola virus glycoprotein-specific binding antibodies at 21 days after the second dose. The primary outcome was assessed in all participants who had received at least one dose of study vaccine and had available reactogenicity data, and immunogenicity was assessed in all participants who had received both vaccinations within the protocol-defined time window, had at least one evaluable post-vaccination sample, and had no major protocol deviations that could have influenced the immune response. This study is registered at ClinicalTrials.gov, NCT02509494.

Findings: From April 4, 2017, to July 5, 2018, 576 eligible children or adolescents (192 in each of the three age cohorts) were enrolled and randomly assigned. The most common solicited local adverse event during the 7 days after the first and second dose was injection-site pain in all age groups, with frequencies ranging from 0% (none of 48) of children aged 1-3 years after placebo injection to 21% (30 of 144) of children aged 4-11 years after Ad26.ZEBOV vaccination. The most frequently observed solicited systemic adverse event during the 7 days was headache in the 12-17 years and 4-11 years age cohorts after the first and second dose, and pyrexia in the 1-3 years age cohort after the first and second dose. The most frequent unsolicited adverse event after the first and second dose vaccinations was malaria in all age cohorts, irrespective of the vaccine types. Following vaccination with MenACWY, severe thrombocytopaenia was observed in one participant aged 3 years. No other clinically significant laboratory abnormalities were observed in other study participants, and no serious adverse events related to the Ebola vaccine regimen were reported. There were no treatment-related deaths. Ebola virus glycoprotein-specific binding antibody responses at 21 days after the second dose of the Ebola virus vaccine regimen were observed in 131 (98%) of 134 children aged 12-17 years (9929 ELISA units [EU]/mL [95% CI 8172-12 064]), in 119 (99%) of 120 aged 4-11 years (10 212 EU/mL [8419-12 388]), and in 118 (98%) of 121 aged 1-3 years (22 568 EU/mL [18 426-27 642]).

Interpretation: The Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen was well tolerated with no safety concerns in children aged 1-17 years, and induced robust humoral immune responses, suggesting suitability of this regimen for Ebola virus disease prophylaxis in children.

Funding: Innovative Medicines Initiative 2 Joint Undertaking and Janssen Vaccines & Prevention BV.

Conflict of interest statement

Declaration of interests BKe was a full-time employee of Janssen Pharmaceutical Companies of Johnson & Johnson at the time of the study. NG, ML, AG, DH, VB, KL, BC, CR, and MD were full-time employees of Janssen Pharmaceutical Companies of Johnson & Johnson at the time of the study, and declare ownership of shares in Janssen Pharmaceutical Companies of Johnson & Johnson. DW-J reports grants from the Innovative Medicines Initiative and non-financial support from Janssen Vaccines & Prevention BV during the conduct of the study; and grants from the Coalition for Epidemic Preparedness Innovations and non-financial support from Janssen Vaccines & Prevention BV outside the submitted work. HL reports grants from GSK and Merck outside the submitted work. All other authors declare no competing interests.

Copyright © 2022 Elsevier Ltd. All rights reserved.

Figures

Figure 1. Trial profiles for the 12–17…
Figure 1. Trial profiles for the 12–17 years (A), 4–11 years (B), and 1–3 years (C) age cohorts
Ad26.ZEBOV=adenovirus type 26 vector-based vaccine encoding the Ebola virus glycoprotein. MenACWY=meningococcal quadrivalent (serogroups A, C, W135, and Y) conjugate vaccine. MVA-BN-Filo=modified vaccinia Ankara vector-based vaccine, encoding glycoproteins from the Ebola virus, Sudan virus, and Marburg virus, and the nucleoprotein from the Tai Forest virus. *One participant received Ad26.ZEBOV followed by placebo and was therefore excluded from further analyses.
Figure 2. Solicited local and systemic AEs…
Figure 2. Solicited local and systemic AEs in the paediatric cohorts
Solicited local (A) and systemic (B) AEs during 7 days after the first dose, and solicited local (C) and systemic (D) AEs during 7 days after the second dose in the 12–17 years age cohort. Solicited local (E) and systemic (F) AEs during 7 days after the first dose, and solicited local (G) and systemic (H) AEs during 7 days after the second dose in the 4–11 years age cohort. Solicited local (I) and systemic (J) AEs during 7 days after the first dose, and solicited local (K) and systemic (L) AEs during 7 days after the second dose in the 1–3 years age cohort. Ad26.ZEBOV=adenovirus type 26 vector-based vaccine encoding the Ebola virus glycoprotein. AE=adverse event. MenACWY=meningococcal quadrivalent (serogroups A, C, W135, and Y) conjugate vaccine. MVA-BN-Filo=modified vaccinia Ankara vector-based vaccine, encoding glycoproteins from the Ebola virus, Sudan virus, and Marburg virus, and the nucleoprotein from the Tai Forest virus.
Figure 3. Geometric mean concentrations of Ebola…
Figure 3. Geometric mean concentrations of Ebola virus glycoprotein-specific binding antibodies before and after each vaccination
Figure 4. Ebola virus glycoprotein-specific neutralising antibody…
Figure 4. Ebola virus glycoprotein-specific neutralising antibody responses before and after each vaccination
The response profile for each group is shown as geometric mean titres, measured by use of the pseudovirion neutralisation assay. The error bars show the 95% CIs. The black dotted line represents the LLOQ. Day 1 is baseline, day 57 is 56 days after the first dose, day 78 is 21 days after the second dose, day 240 is 179 days after the second dose, and day 360 is 359 days after the first dose. Ad26.ZEBOV=adenovirus type 26 vector-based vaccine encoding the Ebola virus glycoprotein. IC50=half maximal inhibitory concentration. LLOQ=lower limit of quantification. MenACWY=meningococcal quadrivalent (serogroups A, C, W135, and Y) conjugate vaccine. MVA-BN-Filo=modified vaccinia Ankara vector-based vaccine, encoding glycoproteins from the Ebola virus, Sudan virus, and Marburg virus, and the nucleoprotein from the Tai Forest virus.

References

    1. Centres for Disease Control and Prevention. 2014–2016 Ebola outbreak in west Africa. 2019. [accessed June 23, 2020]. .
    1. Lo TQ, Marston BJ, Dahl BA, De Cock KM. Ebola: anatomy of an epidemic. Annu Rev Med. 2017;68:359–70.
    1. Agua-Agum J, Ariyarajah A, Blake IM, et al. Ebola virus disease among children in west Africa. N Engl J Med. 2015;372:1274–77.
    1. Centres for Disease Control and Prevention. Cost of the Ebola epidemic. 2019. [accessed June 23, 2020]. .
    1. WHO. Ebola virus disease Democratic Republic of the Congo: external siguation report 97/2020. 2020. [accessed Aug 12, 2021]. .
    1. Matz KM, Marzi A, Feldmann H. Ebola vaccine trials: progress in vaccine safety and immunogenicity. Expert Rev Vaccines. 2019;18:1229–42.
    1. Henao-Restrepo AM, Camacho A, Longini IM, et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ca Suffit!) Lancet. 2017;389:505–18.
    1. Shears P, Garavan C. The 2018/19 Ebola epidemic the Democratic Republic of the Congo (DRC): epidemiology, outbreak control, and conflict. Infect Prev Pract. 2020;2:10038
    1. European Commission. Vaccine against Ebola: commission grants first-ever market authorisation. 2019. [accessed June 23, 2020]. .
    1. US Food and Drug Administration. First FDA-approved vaccine for the prevention of Ebola virus disease, marking a critical milestone in public health preparedness and response. 2019. [accessed Dec 15, 2020]. .
    1. WHO. Strategic Advisory Group of Experts (SAGE) on Immunization interim recommendations on vaccination against Ebola virus disease (EVD) 2019. [accessed June 23, 2020]. .
    1. European Commission. Vaccine against Ebola: commission grants new market authorisations. 2020. [accessed July 9, 2020]. .
    1. Ishola D, Manno D, Afolabi MO, et al. Safety and long-term immunogenicity of the two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in adults in Sierra Leone: a combined open-label, non-randomised stage 1 trial, and a randomised, double-blind, controlled stage 2 trial. Lancet Infect Dis. 2021 doi: 10.1016/S1473-3099(21)00125-0. published online Sept 13.
    1. Mutua G, Anzala O, Luhn K, et al. Safety and immunogenicity of a 2-dose heterologous vaccine regimen with Ad26.ZEBOV and MVA-BN-Filo Ebola vaccines: 12-month data from a phase 1 randomized clinical trial in Nairobi, Kenya. J Infect Dis. 2019;220:57–67.
    1. Anywaine Z, Whitworth H, Kaleebu P, et al. Safety and immunogenicity of a 2-dose heterologous vaccination regimen with Ad26.ZEBOV and MVA-BN-Filo Ebola vaccines: 12-month data from a phase 1 randomized clinical trial in Uganda and Tanzania. J Infect Dis. 2019;220:46–56.
    1. US Food and Drug Administration. Guidance for industry: toxicity grading scale for healthy adult and adolescent volunteers enrolled in preventive vaccine clinical trials. 2007. [accessed June 23, 2020]. .
    1. Dosoo DK, Asante KP, Kayan K, et al. Biochemical and hematologic parameters for children in the middle belt of Ghana. Am J Trop Med Hyg. 2014;90:767–73.
    1. Pollard AJ, Launay O, Lelievre JD, et al. Safety and immunogenicity of a two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in adults in Europe (EBOVAC2): a randomised, observer-blind, participant-blind, placebo-controlled, phase 2 trial. Lancet Infect Dis. 2020;21:493–506.
    1. Afolabi MO, Tiono AB, Adetifa UJ, et al. Safety and immunogenicity of ChAd63 and MVA ME-TRAP in west African children and infants. Mol Ther. 2016;24:1470–77.
    1. Tapia MD, Sow SO, Mbaye KD, et al. Safety, reactogenicity, and immunogenicity of a chimpanzee adenovirus vectored Ebola vaccine in children in Africa: a randomised, observer-blind, placebo-controlled, phase 2 trial. Lancet Infect Dis. 2020;20:719–30.
    1. US Food and Drug Administration. BLA clinical review memorandum. 2020. [accessed Aug 12, 2020]. .
    1. GlaxoSmithKline. Highlights of prescribing information: Menveo. 2020. [accessed May 18, 2020]. .
    1. Mendoza YG, Garric E, Leach A, et al. Safety profile of the RTS,S/ AS01 malaria vaccine in infants and children: additional data from a phase III randomized controlled trial in sub-Saharan Africa. Human Vacc Immunother. 2019;15:2386–98.
    1. Odusanya OO, Kuyinu YA, Kehinde OA, et al. Safety and immunogenicity of 10-valent pneumococcal non typeable Haemophilus influenzae protein D conjugate vaccine (PHiD-CV) in Nigerian children. Hum Vaccin Immunother. 2014;10:757–66.
    1. Thompson A, Gurtman A, Patterson S, et al. Safety of 13-valent pneumococcal conjugate vaccine in infants and children: metaanalysis of 13 clinical trials in 9 countries. Vaccine. 2013;31:5289–95.
    1. National Institute of Allergy and Infectious Diseases. Division of Microbiology and Infectious Diseases (DMID) pediatric toxicity tables November 2007 draft. 2007. [accessed June 23, 2020]. .
    1. Wirth JP, Rohner F, Woodruff BA, et al. Anemia, micronutrient deficiencies, and malaria in children and women in Sierra Leone prior to the Ebola outbreak—findings of a cross-sectional study. PLoS One. 2016;11:e0155031.
    1. Mantadakis E, Farmaki E, Buchanan GR. Thrombocytopenic purpura after measles-mumps-rubella vaccination: a systematic review of the literature and guidance for management. J Pediatr. 2010;156:623–28.
    1. Cecinati V, Principi N, Brescia L, Giordano P, Esposito S. Vaccine administration and the development of immune thrombocytopenic purpura in children. Hum Vaccin Immunother. 2013;9:1158–62.
    1. Bliss CM, Drammeh A, Bowyer G, et al. Viral vector malaria vaccines induce high-level T cell and antibody responses in west African children and infants. Mol Ther. 2017;25:547–59.
    1. Mwangi TW, Bethony JM, Brooker S. Malaria and helminth interactions in humans: an epidemiological viewpoint. Ann Trop Med Parasitol. 2006;100:551–70.
    1. Bockstal V, Roozendaal R, Effelterre TV, et al. Immunobridging approach to assess clinical benefit as the basis for licensure of the monovalent Ebola vaccine. 2018. [accessed June 23, 2020]. .
    1. Roozendaal R, Hendriks J, van Effelterre T, et al. Nonhuman primate to human immunobridging to infer the protective effect of an Ebola virus vaccine candidate. NPJ Vaccines. 2020;5:112.
    1. Schiffer JM, Chen L, Dalton S, Niemuth NA, Sabourin CL, Quinn CP. Bridging non-human primate correlates of protection to reassess the anthrax vaccine adsorbed booster schedule in humans. Vaccine. 2015;33:3709–16.

Source: PubMed

3
订阅