Advancing Global Health through Development and Clinical Trials Partnerships: A Randomized, Placebo-Controlled, Double-Blind Assessment of Safety, Tolerability, and Immunogenicity of PfSPZ Vaccine for Malaria in Healthy Equatoguinean Men

Ally Olotu, Vicente Urbano, Ali Hamad, Martin Eka, Mwajuma Chemba, Elizabeth Nyakarungu, Jose Raso, Esther Eburi, Dolores O Mandumbi, Dianna Hergott, Carl D Maas, Mitoha O Ayekaba, Diosdado N Milang, Matilde R Rivas, Tobias Schindler, Oscar M Embon, Adam J Ruben, Elizabeth Saverino, Yonas Abebe, Natasha Kc, Eric R James, Tooba Murshedkar, Anita Manoj, Sumana Chakravarty, Minglin Li, Matthew Adams, Christopher Schwabe, J Luis Segura, Claudia Daubenberger, Marcel Tanner, Thomas L Richie, Peter F Billingsley, B Kim Lee Sim, Salim Abdulla, Stephen L Hoffman, Ally Olotu, Vicente Urbano, Ali Hamad, Martin Eka, Mwajuma Chemba, Elizabeth Nyakarungu, Jose Raso, Esther Eburi, Dolores O Mandumbi, Dianna Hergott, Carl D Maas, Mitoha O Ayekaba, Diosdado N Milang, Matilde R Rivas, Tobias Schindler, Oscar M Embon, Adam J Ruben, Elizabeth Saverino, Yonas Abebe, Natasha Kc, Eric R James, Tooba Murshedkar, Anita Manoj, Sumana Chakravarty, Minglin Li, Matthew Adams, Christopher Schwabe, J Luis Segura, Claudia Daubenberger, Marcel Tanner, Thomas L Richie, Peter F Billingsley, B Kim Lee Sim, Salim Abdulla, Stephen L Hoffman

Abstract

Equatorial Guinea (EG) has implemented a successful malaria control program on Bioko Island. A highly effective vaccine would be an ideal complement to this effort and could lead to halting transmission and eliminating malaria. Sanaria® PfSPZ Vaccine (Plasmodium falciparum sporozoite Vaccine) is being developed for this purpose. To begin the process of establishing the efficacy of and implementing a PfSPZ Vaccine mass vaccination program in EG, we decided to conduct a series of clinical trials of PfSPZ Vaccine on Bioko Island. Because no clinical trial had ever been conducted in EG, we first successfully established the ethical, regulatory, quality, and clinical foundation for conducting trials. We now report the safety, tolerability, and immunogenicity results of the first clinical trial in the history of the country. Thirty adult males were randomized in the ratio 2:1 to receive three doses of 2.7 × 105 PfSPZ of PfSPZ Vaccine (N = 20) or normal saline placebo (N = 10) by direct venous inoculation at 8-week intervals. The vaccine was safe and well tolerated. Seventy percent, 65%, and 45% of vaccinees developed antibodies to Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) by enzyme-linked immunosorbent assay, PfSPZ by automated immunofluorescence assay, and PfSPZ by inhibition of sporozoite invasion assay, respectively. Antibody responses were significantly lower than responses in U.S. adults who received the same dosage regimen, but not significantly different than responses in young adult Malians. Based on these results, a clinical trial enrolling 135 subjects aged 6 months to 65 years has been initiated in EG; it includes PfSPZ Vaccine and first assessment in Africa of PfSPZ-CVac. ClinicalTrials.gov identifier: NCT02418962.

Figures

Figure 1.
Figure 1.
Consort diagram for EGSPZV1 clinical trial.
Figure 2.
Figure 2.
Comparison of adverse events (AEs) in volunteers receiving normal saline (NS) and PfSPZ Vaccine. The percent of volunteers with a specific AE is depicted. Only a single volunteer had any individual AE after each immunization (maximum of 1/10 [10%] for NS and 1/20 [5%] for PfSPZ Vaccine). ¥, €, £, β are individual volunteers. This figure appears in color at www.ajtmh.org.
Figure 3.
Figure 3.
Antibody responses 2 weeks after the last vaccination. Responses in all vaccinees and controls who received three doses of PfSPZ Vaccine or normal saline were measured by (A) Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) enzyme-linked immunosorbent assay (ELISA), (B) Automated immunofluorescence assay (aIFA), and (C) Inhibition of sporozoite invasion assay (ISI) assay. In the ELISA, antibody responses are reported as the net OD 1.0. OD 1.0 is the serum dilution at which the optical density is 1.0, and net OD 1.0 is the difference between the post- and pre-immunization OD 1.0. In the aIFA assay, antibody responses are reported as the reciprocal serum dilution at which the arbitrary fluorescence units (AFU) were 2 × 105. In the ISI assay, antibody responses are reported as the reciprocal serum dilution that gave 80% inhibition of PfSPZ invasion. Medians with interquartile ranges are shown.
Figure 4.
Figure 4.
Comparison of anti-Plasmodium falciparum circumsporozoite protein (PfCSP) antibody responses in four different clinical trials in which the same dose of PfSPZ Vaccine was administered. Antibody responses measured by enzyme-linked immunosorbent assay (ELISA) are reported as the net OD 1.0. The OD 1.0 is the serum dilution at which the optical density is 1.0, and net OD 1.0 is the difference between the post- and pre-immunization OD 1.0. In the USA (VRC314/WRAIR 2080) (most subjects), Mali and Tanzania vaccinations were at 0, 4, and 8 weeks. In Equatorial Guinea (EG), vaccinations were at 0, 8, and 16 weeks. In the United States, Mali, and EG, sera were drawn at 2 weeks after the last vaccination. In Tanzania, sera were drawn at 4 weeks after the last vaccination. Bars with asterisks indicate the statistical significance, as determined by a Kruskal–Wallis test followed by a Dunn’s multiple comparisons test (P = * < 0.0001; ** < 0.0001; *** 0.0008).

References

    1. World Health Organization , 2016. World Malaria Report 2016. Geneva, Switzerland: World Health Organization.
    1. World Health Organization , 2015. Global Technical Strategy for Malaria 2016–2030. Geneva, Switzerland: World Health Organization.
    1. RTSS Clinical Trials Partnership , 2015. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet 386: 31–45.
    1. RTS,S Clinical Trials Partnership , 2014. Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLoS Med 11: e1001685.
    1. Neafsey DE, et al. 2015. Genetic diversity and protective efficacy of the RTS,S/AS01 malaria vaccine. N Engl J Med 373: 2025–2037.
    1. Seder RA, et al. VRC 312 Study Team , 2013. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341: 1359–1365.
    1. Sissoko MS, et al. 2017. Safety and efficacy of PfSPZ Vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial. Lancet Infect Dis 17: 498–509.
    1. Ishizuka AS, et al. 2016. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat Med 22: 614–623.
    1. Epstein JE, et al. 2017. Protection against Plasmodium falciparum malaria by PfSPZ vaccine. JCI Insight 2: e89154.
    1. Lyke KE, et al. 2017. Attenuated PfSPZ Vaccine induces strain-transcending T cells and durable protection against heterologous controlled human malaria infection. Proc Natl Acad Sci USA 114: 2711–2716.
    1. Kleinschmidt I, Sharp B, Benavente LE, Schwabe C, Torrez M, Kuklinski J, Morris N, Raman J, Carter J, 2006. Reduction in infection with Plasmodium falciparum one year after the introduction of malaria control interventions on Bioko Island, Equatorial Guinea. Am J Trop Med Hyg 74: 972–978.
    1. Hoffman SL, et al. 2010. Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Hum Vaccin 6: 97–106.
    1. Epstein JE, et al. 2011. Live attenuated malaria vaccine designed to protect through hepatic CD8+ T cell immunity. Science 334: 475–480.
    1. Guidance for Industry , Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials. Available at: . Accessed May 2, 2017.
    1. Cohee LM, Kalilani-Phiri L, Mawindo P, Joshi S, Adams M, Kenefic L, Jacob CG, Taylor TE, Laufer MK, 2016. Parasite dynamics in the peripheral blood and the placenta during pregnancy-associated malaria infection. Malar J 15: 483.
    1. Roestenberg M, et al. 2013. Controlled human malaria infections by intradermal injection of cryopreserved Plasmodium falciparum sporozoites. Am J Trop Med Hyg 88: 5–13.
    1. Shekalaghe S, et al. 2014. Controlled human malaria infection of Tanzanians by intradermal injection of aseptic, purified, cryopreserved Plasmodium falciparum sporozoites. Am J Trop Med Hyg 91: 471–480.
    1. Gómez-Pérez GP, et al. 2015. Controlled human malaria infection by intramuscular and direct venous inoculation of cryopreserved Plasmodium falciparum sporozoites in malaria-naive volunteers: effect of injection volume and dose on infectivity rates. Malar J 14: 306.
    1. Mordmüller B, et al. 2015. Direct venous inoculation of Plasmodium falciparum sporozoites for controlled human malaria infection: a dose-finding trial in two centres. Malar J 14: 117.
    1. Bastiaens GJ, et al. 2016. Safety, immunogenicity, and protective efficacy of intradermal immunization with aseptic, purified, cryopreserved Plasmodium falciparum sporozoites in volunteers under chloroquine prophylaxis: a randomized controlled trial. Am J Trop Med Hyg 94: 663–673.
    1. Mordmüller B, et al. 2017. Sterile protection against human malaria by chemoattenuated PfSPZ vaccine. Nature 542: 445–449.
    1. Hoffman SL, Vekemans J, Richie TL, Duffy PE, 2015. The march toward malaria vaccines. Vaccine 33 (Suppl 4): D13–D23.
    1. Richie TL, et al. 2015. Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine 33: 7452–7461.
    1. Muyanja E, et al. 2014. Immune activation alters cellular and humoral responses to yellow fever 17D vaccine. J Clin Invest 124: 3147–3158.
    1. Kilian HD, Nielsen G, 1989. Cell-mediated and humoral immune responses to BCG and rubella vaccinations and to recall antigens in onchocerciasis patients. Trop Med Parasitol 40: 445–453.
    1. Haseeb MA, Craig JP, 1997. Suppression of the immune response to diphtheria toxoid in murine schistosomiasis. Vaccine 15: 45–50.
    1. Da’Dara AA, Lautsch N, Dudek T, Novitsky V, Lee TH, Essex M, Harn DA, 2006. Helminth infection suppresses T-cell immune response to HIV-DNA-based vaccine in mice. Vaccine 24: 5211–5219.
    1. Esen M, et al. 2012. Reduced antibody responses against Plasmodium falciparum vaccine candidate antigens in the presence of Trichuris trichiura. Vaccine 30: 7621–7624.
    1. Kolbaum J, Tartz S, Hartmann W, Helm S, Nagel A, Heussler V, Sebo P, Fleischer B, Jacobs T, Breloer M, 2012. Nematode-induced interference with the anti-Plasmodium CD8+ T-cell response can be overcome by optimizing antigen administration. Eur J Immunol 42: 890–900.
    1. Apiwattanakul N, Thomas PG, Iverson AR, McCullers JA, 2014. Chronic helminth infections impair pneumococcal vaccine responses. Vaccine 32: 5405–5410.
    1. Noland GS, Chowdhury DR, Urban JF, Jr, Zavala F, Kumar N, 2010. Helminth infection impairs the immunogenicity of a Plasmodium falciparum DNA vaccine, but not irradiated sporozoites, in mice. Vaccine 28: 2917–2923.

Source: PubMed

3
Abonner