Safety and Immunogenicity of an AS03B-Adjuvanted Inactivated Tetravalent Dengue Virus Vaccine Administered on Varying Schedules to Healthy U.S. Adults: A Phase 1/2 Randomized Study

Leyi Lin, Kirsten E Lyke, Michael Koren, Richard G Jarman, Kenneth H Eckels, Edith Lepine, Monica A McArthur, Jeffrey R Currier, Heather Friberg, Philippe Moris, Paul B Keiser, Rafael De La Barrera, David W Vaughn, Robert M Paris, Stephen J Thomas, Alexander C Schmidt, Leyi Lin, Kirsten E Lyke, Michael Koren, Richard G Jarman, Kenneth H Eckels, Edith Lepine, Monica A McArthur, Jeffrey R Currier, Heather Friberg, Philippe Moris, Paul B Keiser, Rafael De La Barrera, David W Vaughn, Robert M Paris, Stephen J Thomas, Alexander C Schmidt

Abstract

Dengue disease and its causative agents, the dengue viruses (DENV-1-4), cause high morbidity in tropical and subtropical regions. We evaluated three dosing regimens of the investigational tetravalent AS03B-adjuvanted dengue-purified inactivated vaccine (DPIV+AS03B). In this phase 1/2, observer-blind, placebo-controlled study (NCT02421367), 140 healthy adults were randomized 1:1:2 to receive DPIV+AS03B according to the following regimens: 0-1 month (M), 0-1-6 M, or 0-3 M. Participants received DPIV+AS03B or placebo at M0, M1, M3, and M6 according to their dosing schedule. Primary objectives were 1) to evaluate the safety of DPIV+AS03B for 28 days (D) after each dose; 2) to demonstrate the added value of a booster dose (0-1-6 M versus 0-1 M) based on neutralizing antibody titers to each DENV type (DENV-1-4) at 28 D after the last dose; and, if this objective was met, 3) to demonstrate the benefit of a longer interval between the first and second doses (0-1 M versus 0-3 M). Adverse events (AEs) within 7 D after vaccination tended to be more frequent after DPIV+AS03B doses than placebo; the number of grade 3 AEs was low (≤ 4.5% after DPIV+AS03B; ≤ 2.9% after placebo), with no obvious differences across groups. Within 28 D following each dose, the frequency of unsolicited AEs after DPIV+AS03B appeared higher for three-dose (0-1-6 M) than two-dose (0-1 M and 0-3 M) regimens. No serious AEs were considered related to vaccination, and no potential immune-mediated diseases were reported during the study. All three schedules were well tolerated. Both primary immunogenicity objectives were demonstrated. The 0-3 M and 0-1-6 M regimens were more immunogenic than the 0-1 M regimen.

Conflict of interest statement

Disclosures: P. M. and R. M. P. are currently employees of the GSK group of companies. E. L., D. W. V., and A. C. S. were employees of the GSK group of companies during the conduct of the study. P. M., R. M. P., and A. C. S. hold shares in the GSK group of companies. L. L., M. K., R. G. J., K. H. E., J. R. C., H. F., P. K., R. D. L. B., and S. J. T. received financial and other support without any personal financial benefit from the GSK group of companies as part of the CRADA between the U.S. Army and the GSK group of companies. M. A. M. received consulting fees from Takeda outside of the submitted work. K. E. reports having a licensed patent for which he received royalties. K. L. declares no financial and non-financial relationships and activities and no conflicts of interest. All authors had full access to the data and gave final approval before submission. GlaxoSmithKline Biologicals SA was involved in all stages of the study conduct and analysis, and also took charge of all costs associated with the development and publishing of this manuscript. Dengvaxia is a trademark of Sanofi Pasteur.

Disclaimer: The opinions or assertions contained herein are the private views of the authors and are not to be construed as reflecting the official views of the U.S. Army or the U.S. Department of Defense.

Figures

Figure 1.
Figure 1.
Study design. AEs = adverse events; D = day(s); DPIV+AS03B = adjuvant system 03B-adjuvanted inactivated tetravalent dengue virus vaccine; M = month; MAEs = medically attended AEs; N = number of participants in each group included in the total vaccinated cohort; pIMDs = potential immune-mediated diseases; SAEs = serious AEs; 0–1 M = participants receiving two doses of DPIV+AS03B administered 1 M apart, at M0 and M1; 0–3 M = participants receiving two doses of DPIV+AS03B administered 3 M apart, at M3 and M6; 0–1–6 M = participants receiving three doses of DPIV+AS03B with the first two given 1 M apart and the third given 6 M after the first, at M0, M1, and M6. Only immunogenicity blood samples for analyses described here are shown in this figure.
Figure 2.
Figure 2.
Flow diagram of study participants. AE = adverse event; ATP = according-to-protocol; DPIV+AS03B = adjuvant system 03B-adjuvanted inactivated tetravalent dengue virus vaccine; M = month; N = number of participants in each group; 0–1 M = participants receiving two doses of DPIV+AS03B administered 1 M apart, at M0 and M1; 0–3 M = participants receiving two doses of DPIV+AS03B administered 3 M apart, at M3 and M6; 0–1–6 M = participants receiving three doses of DPIV+AS03B with the first two given 1 M apart and the third given 6 M after the first, at M0, M1, and M6. *Non-compliance by participant and outside of window for visit (participation discontinued).
Figure 3.
Figure 3.
Overall per dose incidence of solicited injection-site (A) and general (B) adverse events during the 7-D post-vaccination period (TVC). DPIV+AS03B = adjuvant system 03B-adjuvanted inactivated tetravalent dengue virus vaccine; GI symtoms = gastrointestinal symptoms (nausea, vomiting, diarrhea, and/or abdominal pain); M = month; TVC = total vaccinated cohort; 0–1 M = participants receiving two doses of DPIV+AS03B administered 1 M apart, at M0 and M1; 0–3 M = participants receiving two doses of DPIV+AS03B administered 3 M apart, at M3 and M6; 0–1–6 M = participants receiving three doses of DPIV+AS03B with the first two given 1 M apart and the third given 6 M after the first, at M0, M1, and M6. Error bars indicate 95% CIs.
Figure 4.
Figure 4.
Kinetics of neutralizing antibody responses against DENV-1–4 (ATP cohort for immunogenicity). ATP = according-to-protocol; DENV = dengue virus; GMT = geometric mean titer; N = number of participants in each group; M = month; 0–1 M = participants receiving two doses of adjuvant system 03B-adjuvanted inactivated tetravalent dengue virus vaccine (DPIV+AS03B) administered one M apart, at M0 and M1; 0–3 M = participants receiving two doses of DPIV+AS03B administered 3 M apart, at M3 and M6; 0–1–6 M = participants receiving three doses of DPIV+AS03B with the first two given 1 M apart and the third given 6 M after the first, at M0, M1, and M6. Error bars depict 95% CIs.
Figure 5.
Figure 5.
Seroconversion rates to DENV types at peak response and at study end (ATP cohort for immunogenicity). ATP = according-to-protocol; DENV = dengue virus; N = number of participants in each group; 0–1M = participants receiving two doses of adjuvant system 03B-adjuvanted inactivated tetravalent dengue virus vaccine (DPIV+AS03B) administered 1 M apart, at M0 and M1; 0–3 M = participants receiving two doses of DPIV+AS03B administered 3 M apart, at M3 and M6; 0–1–6 M = participants receiving three doses of DPIV+AS03B with the first two given 1 M apart and the third given 6 M after the first, at M0, M1, and M6. *Peak, 1 M after dose two for groups 0–1 M and 0–3 M and 1 M after dose 3 for group 0–1–6 M.
Figure 6.
Figure 6.
Kinetics for the median (Q1/Q3) frequency (%) of DENV-type–specific memory B cells (by ELISpot) up to study end (ATP cohort for immunogenicity). ATP = according-to-protocol; DENV = dengue virus; ELISpot = enzyme-linked immunospot assay; N = number of participants in each group; M = month; Q1/Q3 = first and third quantiles; 0–1 M = participants receiving two doses of adjuvant system 03B-adjuvanted inactivated tetravalent dengue virus vaccine (DPIV+AS03B) administered 1 M apart, at M0 and M1; 0–3 M = participants receiving two doses of DPIV+AS03B administered 3 M apart, at M3 and M6; 0–1–6 M = participants receiving three doses of DPIV+AS03B with the first two given 1 M apart and the third given 6 M after the first, at M0, M1, and M6. Error bars depict interquartile ranges.

References

    1. World Health Organization , 2018. Dengue vaccine: WHO position paper–September 2018. Wkly Epidemiol Rec 93: 457–476.
    1. Stanaway JD, et al. 2016. The global burden of dengue: an analysis from the Global Burden of Disease Study 2013. Lancet Infect Dis 16: 712–723.
    1. Dayan G, et al. 2015. Prospective cohort study with active surveillance for fever in four dengue endemic countries in Latin America. Am J Trop Med Hyg 93: 18–23.
    1. Holbrook MR, 2017. Historical perspectives on flavivirus research. Viruses 9: 97.
    1. WHO , 2017. Updated Questions and Answers Related to Information Presented in the Sanofi Pasteur Press Release on 30 November 2017 with Regards to the Dengue Vaccine Dengvaxia®. Available at: . Accessed June 7, 2018.
    1. Bowman LR, Donegan S, McCall PJ, 2016. Is dengue vector control deficient in effectiveness or evidence? Systematic review and meta-analysis. PLoS Negl Trop Dis 10: e0004551.
    1. Torresi J, Ebert G, Pellegrini M, 2017. Vaccines licensed and in clinical trials for the prevention of dengue. Hum Vaccin Immunother 13: 1059–1072.
    1. Elong Ngono A, Shresta S, 2019. Cross-reactive T cell immunity to dengue and zika viruses: new insights into vaccine development. Front Immunol 10: 1316.
    1. Hadinegoro SR, et al. 2015. Efficacy and long-term safety of a dengue vaccine in regions of endemic disease. N Engl J Med 373: 1195–1206.
    1. Cohet C, van der Most R, Bauchau V, Bekkat-Berkani R, Doherty TM, Schuind A, Tavares Da Silva F, Rappuoli R, Garcon N, Innis BL, 2019. Safety of AS03-adjuvanted influenza vaccines: a review of the evidence. Vaccine 37: 3006–3021.
    1. Barban V, Mantel N, De Montfort A, Pagnon A, Pradezynski F, Lang J, Boudet F, 2018. Improvement of the dengue virus (DENV) nonhuman primate model via a reverse translational approach based on dengue vaccine clinical efficacy data against DENV-2 and -4. J Virol 92: e00440‒18.
    1. Borges MB, et al. 2019. Detection of post-vaccination enhanced dengue virus infection in macaques: an improved model for early assessment of dengue vaccines. PLoS Pathog 15: e1007721.
    1. Schmidt AC, et al. 2017. Phase 1 randomized study of a tetravalent dengue purified inactivated vaccine in healthy adults in the United States. Am J Trop Med Hyg 96: 1325–1337.
    1. Diaz C, et al. 2018. Phase I randomized study of a tetravalent dengue purified inactivated vaccine in healthy adults from Puerto Rico. Am J Trop Med Hyg 98: 1435–1443.
    1. International Conference on Harmonisation , 1994. Topic E2A: Clinical Safety Data Management: Definitions and Standards for Expedited Reporting. Available at: . Accessed May 1, 2019.
    1. FDA , 2007. Guidance for Industry. Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials. Available at: . Accessed May 1, 2019.
    1. Weiskopf D, et al. 2015. The human CD8+ T cell responses induced by a live attenuated tetravalent dengue vaccine are directed against highly conserved epitopes. J Virol 89: 120–128.
    1. Tran NH, et al. 2019. Long-term immunogenicity and safety of tetravalent dengue vaccine (CYD-TDV) in healthy populations in Singapore and Vietnam: 4-year follow-up of randomized, controlled, phase II trials. Hum Vaccin Immunother 6: 1–13.
    1. Ahmad Z, Poh CL, 2019. The conserved molecular determinants of virulence in dengue virus. Int J Med Sci 16: 355–365.
    1. Eder S, et al. 2011. Long term immunity following a booster dose of the inactivated Japanese Encephalitis vaccine IXIARO(R), IC51. Vaccine 29: 2607–2612.
    1. Grifoni A, et al. 2017. Patterns of cellular immunity associated with experimental infection with rDEN2Δ30 (Tonga/74) support its suitability as a human dengue virus challenge strain. J Virol 91: e02133–16.
    1. Mongkolsapaya J, et al. 2003. Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever. Nat Med 9: 921–927.

Source: PubMed

3
Subscribe