A PfSPZ vaccine immunization regimen equally protective against homologous and heterologous controlled human malaria infection

Benjamin Mordmüller, Zita Sulyok, Mihály Sulyok, Zsofia Molnar, Albert Lalremruata, Carlos Lamsfus Calle, Patricia Granados Bayon, Meral Esen, Markus Gmeiner, Jana Held, Henri-Lynn Heimann, Tamirat Gebru Woldearegai, Javier Ibáñez, Judith Flügge, Rolf Fendel, Andrea Kreidenweiss, Natasha Kc, Tooba Murshedkar, Sumana Chakravarty, Pouria Riyahi, Peter F Billingsley, L W Preston Church, Thomas L Richie, B Kim Lee Sim, Stephen L Hoffman, Peter G Kremsner, Benjamin Mordmüller, Zita Sulyok, Mihály Sulyok, Zsofia Molnar, Albert Lalremruata, Carlos Lamsfus Calle, Patricia Granados Bayon, Meral Esen, Markus Gmeiner, Jana Held, Henri-Lynn Heimann, Tamirat Gebru Woldearegai, Javier Ibáñez, Judith Flügge, Rolf Fendel, Andrea Kreidenweiss, Natasha Kc, Tooba Murshedkar, Sumana Chakravarty, Pouria Riyahi, Peter F Billingsley, L W Preston Church, Thomas L Richie, B Kim Lee Sim, Stephen L Hoffman, Peter G Kremsner

Abstract

Immunization with radiation-attenuated Plasmodium falciparum (Pf) sporozoites (SPZ) in PfSPZ Vaccine, has provided better vaccine efficacy (VE) against controlled human malaria infection (CHMI) with the same parasites as in the vaccine (homologous) than with genetically distant parasites (heterologous). We sought to identify an immunization regimen that provided similar VE against CHMI with homologous and heterologous Pf for at least 9 weeks in malaria-naïve adults. Such a regimen was identified in part 1 (optimization), an open label study, and confirmed in part 2 (verification), a randomized, double-blind, placebo-controlled study in which VE was assessed by cross-over repeat CHMI with homologous (PfNF54) and heterologous (Pf7G8) PfSPZ at 3 and 9-10 weeks. VE was calculated using Bayesian generalized linear regression. In part 1, vaccination with 9 × 105 PfSPZ on days 1, 8, and 29 protected 5/5 (100%) subjects against homologous CHMI at 3 weeks after the last immunization. In part 2, the same 3-dose regimen protected 5/6 subjects (83%) against heterologous CHMI at both 3 and 9-10 weeks after the last immunization. Overall VE was 78% (95% predictive interval: 57-92%), and against heterologous and homologous was 79% (95% PI: 54-95%) and 77% (95% PI: 50-95%) respectively. PfSPZ Vaccine was safe and well tolerated. A 4-week, 3-dose regimen of PfSPZ Vaccine provided similar VE for 9-10 weeks against homologous and heterologous CHMI. The trial is registered with ClinicalTrials.gov, NCT02704533.

Conflict of interest statement

NKC, SC, PR, LWPC, TLR, BKLS, and SLH are salaried, full-time employees of Sanaria, the developer and sponsor of Sanaria PfSPZ Vaccine and PfSPZ Challenge. SLH and BKLS also have financial interests in Sanaria. BKLS and SLH are inventors on patents and patent applications that have been assigned to Sanaria. BM and PGK received funding from DZIF to conduct the trial. All other authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Study flow Chart.
Fig. 1. Study flow Chart.
aOptimization of PfSPZ Vaccine regimen and (b) Verification of PfSPZ Vaccine regimen. *9 of the 45 subjects were allocated to a 7G8 safety analysis that will be published elsewhere. $One volunteer received only the first vaccination. #One volunteer received the 2nd vaccination on day 15 instead of day 8.
Fig. 2. Results of CHMIs to assess…
Fig. 2. Results of CHMIs to assess VE during Verification.
a First CHMI at 3 weeks (top row) and second CHMI at 9–10 weeks (bottom row) of the individual volunteers. The black indicates no parasitemia (protected); the white represents parasitaemia (not protected). b Kaplan-Meier plots for heterologous CHMIs with Pf7G8. Five of 6 vaccinees were protected at 3 weeks and at 9–10 weeks. Six of 6 CHMIs with PfSPZ Challenge (7G8) in the controls resulted in parasitemia (infected). c Kaplan-Meier plots for homologous CHMIs with PfNF54. Four of 6 vaccinees were protected at 3 weeks and 5/6 were protected at 9–10 weeks. Five of 6 CHMIs with PfSPZ Challenge (NF54) in the controls resulted in parasitemia (infected).
Fig. 3. Antibodies to PfCSP and PfSPZ…
Fig. 3. Antibodies to PfCSP and PfSPZ and functional activity of sera against PfSPZ.
a, b Median and interquartile range of net OD 1.0 for IgG and IgM antibodies to PfCSP by ELISA one day prior to CHMI in malaria-naïve adults who were uninfected (protected) and infected during CHMI administered 3 weeks after the 3rd dose (CHMI 1). c, d Median and interquartile range of net IgG antibodies to PfSPZ by aIFA and net inhibition of PfSPZ invasion of hepatocytes by aISI one day prior to CHMI in malaria-naïve adults who were uninfected (protected) and infected during CHMI administered 3 weeks after the 3rd dose (CHMI 1). e, f Median and interquartile range of net OD 1.0 for IgG and IgM antibodies to PfCSP by ELISA the day prior to CHMI in adults who were uninfected (protected) and infected during CHMI administered 9–10 weeks after the 3rd dose (CHMI 2). g, h Median and interquartile range of net IgG antibodies to PfSPZ by aIFA and net inhibition of PfSPZ invasion of hepatocytes by aISI one day prior to CHMI in adults who were uninfected (protected) and infected during CHMI administered 9–10 weeks after the 3rd dose (CHMI 2). P values were calculated by Wilcoxon-Mann-Whitney test. For each panel, filled triangles are uninfected subjects and open triangles are infected subjects who received heterologous CHMI with PfSPZ Challenge (7G8) and filled circles are uninfected subjects and open circles are infected subjects who received homologous CHMI with PfSPZ Challenge (NF54).

References

    1. World Health Organization. World Malaria Report 2019. 232 (World Health Organization, 2019).
    1. World Health Organization. WHO calls for reinvigorated action to fight malaria (News release). Retrieved from (30 November 2020).
    1. Khuu D, et al. Malaria-Related Hospitalizations in the United States, 2000–2014. Am. J. Trop. Med Hyg. 2017;97:213–221. doi: 10.4269/ajtmh.17-0101.
    1. European Centre for Disease Prevention and Control. Malaria. In ECDC. Annual epidemiological report for 2018. Retrieved from (Stockholm, 2020).
    1. Tatem AJ, et al. The geography of imported malaria to non-endemic countries: a meta-analysis of nationally reported statistics. Lancet Infect. Dis. 2017;17:98–107. doi: 10.1016/S1473-3099(16)30326-7.
    1. Ahluwalia J, Brooks SK, Weinman J, Rubin GJ. A systematic review of factors affecting adherence to malaria chemoprophylaxis amongst travellers from non-endemic countries. Malar. J. 2020;19:16. doi: 10.1186/s12936-020-3104-4.
    1. RTS, S Clinical Trials Partnership. 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. 2015;386:31–45. doi: 10.1016/S0140-6736(15)60721-8.
    1. World Health Organization. First malaria vaccine in Africa: A potential new tool for child health and improved malaria control. 1–4 (2018).
    1. World Health Organaization. WHO recommends groundbreaking malaria vaccine for children at risk [News release]. Retrieved from (6 October 2021).
    1. Datoo, M. S., et al. Efficacy of a low-dose candidate malaria vaccine, R21 in adjuvant Matrix-M, with seasonal administration to children in Burkina Faso: a randomised controlled trial. Lancet.10.1016/S0140-6736(21)00943-0 (2021).
    1. Hoffman SL, et al. Development of a metabolically active,non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Hum. Vaccines. 2010;6:97–106. doi: 10.4161/hv.6.1.10396.
    1. Epstein JE, et al. Live attenuated malaria vaccine designed to protect through hepatic CD8+T cell immunity. Science. 2011;334:475–480. doi: 10.1126/science.1211548.
    1. Seder RA, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science. 2013;341:1359–1365. doi: 10.1126/science.1241800.
    1. Epstein JE, et al. Protection against Plasmodium falciparum malaria by PfSPZ Vaccine. JCI Insight. 2017;2:e89154. doi: 10.1172/jci.insight.89154.
    1. Lyke KE, et al. Attenuated PfSPZ Vaccine induces strain-transcending T cells and durable protection against heterologous controlled human malaria infection. Proc. Natl Acad. Sci. USA. 2017;114:2711–2716. doi: 10.1073/pnas.1615324114.
    1. Sissoko MS, et al. 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. 2017;17:498–509. doi: 10.1016/S1473-3099(17)30104-4.
    1. Jongo SA, et al. Increase of dose associated with decrease in protection against controlled human malaria infection by PfSPZ Vaccine in Tanzanian adults. Clin. Infect. Dis. 2020;71:2849–2857. doi: 10.1093/cid/ciz1152.
    1. Sissoko MS, et al. Safety and efficacy of a three-dose regimen of Plasmodium falciparum sporozoite vaccine in adults during an intense malaria transmission season in Mali: a randomised, controlled phase 1 trial. Lancet Infect. Dis. 2022;22:377–389. doi: 10.1016/S1473-3099(21)00332-7.
    1. Silva JC, et al. Plasmodium falciparum 7G8 challenge provides conservative prediction of efficacy of PfNF54-based PfSPZ Vaccine in Africa. Nat. Commun. 2022;13:3390. doi: 10.1038/s41467-022-30882-8.
    1. Olotu A, et al. Advancing Global Health through Development and Clinical Trials Partnerships: A Randomized, Placebo-Controlled, Double-Blind Assessment of Safety, Tolerability, and Immunogenicity of Plasmodium falciparum Sporozoites Vaccine for Malaria in Healthy Equatoguinean Men. Am. J. Trop. Med. Hyg. 2018;98:308–318. doi: 10.4269/ajtmh.17-0449.
    1. Jongo SA, et al. Safety and Differential Antibody and T-Cell Responses to the Plasmodium falciparum Sporozoite Malaria Vaccine, PfSPZ Vaccine, by Age in Tanzanian Adults, Adolescents, Children, and Infants. Am. J. Trop. Med. Hyg. 2019;100:1433–1444. doi: 10.4269/ajtmh.18-0835.
    1. Lyke KE, et al. Multidose Priming and Delayed Boosting Improve PfSPZ Vaccine Efficacy against Heterologous P. falciparum Controlled Human Malaria Infection. Clin. Infect. Dis. 2021;73:e2424–e2435. doi: 10.1093/cid/ciaa1294.
    1. CIC Research Inc. Survey of international air travelers, U.S. travelers to overseas, January - March 2012. (ed. 1Q12_US_to_Overseas_Banner1_30Sep13.pdf) (National Travel and Tourism Office, 2013).
    1. Moser KA, et al. Strains used in whole organism Plasmodium falciparum vaccine trials differ in genome structure, sequence, and immunogenic potential. Genome Med. 2020;12:6. doi: 10.1186/s13073-019-0708-9.
    1. Ishizuka AS, et al. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat. Med. 2016;22:614–623. doi: 10.1038/nm.4110.
    1. Jongo SA, et al. Safety, Immunogenicity, and Protective Efficacy against Controlled Human Malaria Infection of Plasmodium falciparum Sporozoite Vaccine in Tanzanian Adults. Am. J. Trop. Med. Hyg. 2018;99:338–349. doi: 10.4269/ajtmh.17-1014.
    1. Steinhardt LC, et al. Safety, tolerability, and immunogenicity of PfSPZ Vaccine administered by direct venous inoculation to infants and young children: findings from an age de-escalation, dose-escalation double-blinded randomized, controlled study in western Kenya. Clin. Infect. Dis. 2020;71:1063–1071. doi: 10.1093/cid/ciz925.
    1. Oneko M, et al. Safety, immunogenicity and efficacy of PfSPZ Vaccine against malaria in infants in western Kenya: a double-blind, randomized, placebo-controlled phase 2 trial. Nat. Med. 2021;27:1636–1645. doi: 10.1038/s41591-021-01470-y.
    1. Jongo SA, et al. Multi-Dose Priming Regimens of PfSPZ Vaccine: Safety and Efficacy against Controlled Human Malaria Infection in Equatoguinean Adults. Am. J. Trop. Med. Hyg. 2022;106:1215–1226. doi: 10.4269/ajtmh.21-0942.
    1. Blom K, et al. Temporal dynamics of the primary human T cell response to yellow fever virus 17D as it matures from an effector- to a memory-type response. J. Immunol. 2013;190:2150–2158. doi: 10.4049/jimmunol.1202234.
    1. Mwakingwe-Omari A, et al. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature. 2021;595:289–294. doi: 10.1038/s41586-021-03684-z.
    1. Stavnezer J, Amemiya CT. Evolution of isotype switching. Semin. Immunol. 2004;16:257–275. doi: 10.1016/j.smim.2004.08.005.
    1. Duarte JH. Functional switching. Nat. Immunol. 2016;17:S12–S12. doi: 10.1038/ni.3607.
    1. Schofield L, et al. Gamma-interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature. 1987;330:664–666. doi: 10.1038/330664a0.
    1. Weiss WR, Sedegah M, Beaudoin RL, Miller LH, Good MF. CD8+ T cells (cytotoxic/suppressors) are required for protection in mice immunized with malaria sporozoites. Proc. Natl Acad. Sci. U. S. A. 1988;85:573–576. doi: 10.1073/pnas.85.2.573.
    1. Hoffman SL, et al. Sporozoite vaccine induces genetically restricted T cell elimination of malaria from hepatocytes. Science. 1989;244:1078–1081. doi: 10.1126/science.2524877.
    1. Doolan DL, Hoffman SL. The complexity of protective immunity against liver-stage malaria. J. Immunol. 2000;165:1453–1462. doi: 10.4049/jimmunol.165.3.1453.
    1. Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine. 2015;33(Suppl 4):D13–23. doi: 10.1016/j.vaccine.2015.07.091.
    1. Diawara, H. “Malaria Vaccination in Pregnancy: PfSPZ Vaccine in Pregnant Women”, Annual Meeting of the American Society of Tropical Medicine & Hygiene, (Virtual presentation) Nov 19 2021.
    1. Roestenberg M, et al. Controlled Human Malaria Infections by Intradermal Injection of Cryopreserved Plasmodium falciparum Sporozoites. Am. J. Trop. Med. Hyg. 2013;88:5–13. doi: 10.4269/ajtmh.2012.12-0613.
    1. Gomez-Perez GP, et al. 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. 2015;14:306. doi: 10.1186/s12936-015-0817-x.
    1. Mordmüller B, et al. Direct venous inoculation of Plasmodium falciparum sporozoites for controlled human malaria infection: a dose-finding trial in two centres. Malar. J. 2015;14:117. doi: 10.1186/s12936-015-0628-0.
    1. Mordmüller B, et al. Sterile protection against human malaria by chemoattenuated PfSPZ vaccine. Nature. 2017;542:445–449. doi: 10.1038/nature21060.
    1. Laurens MB, et al. Dose dependent infectivity of aseptic, purified, cryopreserved Plasmodium falciparum 7G8 sporozoites in malaria-naive adults. J. Infect. Dis. 2019;220:1962–1966. doi: 10.1093/infdis/jiz410.
    1. Sulyok Z, et al. Heterologous protection against malaria by a simple chemoattenuated PfSPZ vaccine regimen in a randomized trial. Nat. Commun. 2021;12:2518. doi: 10.1038/s41467-021-22740-w.
    1. Sulyok M, et al. DSM265 for Plasmodium falciparum chemoprophylaxis: a randomised, double blinded, phase 1 trial with controlled human malaria infection. Lancet Infect. Dis. 2017;17:636–644. doi: 10.1016/S1473-3099(17)30139-1.
    1. Lell B, et al. Impact of Sickle Cell Trait and Naturally Acquired Immunity on Uncomplicated Malaria after Controlled Human Malaria Infection in Adults in Gabon. Am. J. Trop. Med. Hyg. 2018;98:508–515. doi: 10.4269/ajtmh.17-0343.
    1. Metzger WG, et al. Ivermectin for causal malaria prophylaxis: a randomised controlled human infection trial. Trop. Med. Int. Health. 2020;25:380–386. doi: 10.1111/tmi.13357.
    1. Murphy SC, et al. PfSPZ-CVac efficacy against malaria increases from 0 to 75% when administered in the absence of erythrocyte stage parasitemia: A randomized, placebo-controlled trial with controlled human malaria infection. PLoS Pathog. 2021;17:e1009594. doi: 10.1371/journal.ppat.1009594.
    1. Achan J, et al. Serologic markers of previous malaria exposure and functional antibodies inhibiting parasite growth are associated with parasite kinetics following a Plasmodium falciparum controlled human infection. Clin. Infect. Dis. 2019;70:2544–2552. doi: 10.1093/cid/ciz740.
    1. Kapulu MC, Njuguna P, Hamaluba MM. Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA): a study protocol to investigate in vivo Plasmodium falciparum malaria parasite growth in the context of pre-existing immunity. Wellcome Open Res. 2019;3:155. doi: 10.12688/wellcomeopenres.14909.2.
    1. Joanny F, Lohr SJ, Engleitner T, Lell B, Mordmuller B. Limit of blank and limit of detection of Plasmodium falciparum thick blood smear microscopy in a routine setting in Central Africa. Malar. J. 2014;13:234. doi: 10.1186/1475-2875-13-234.

Source: PubMed

3
Abonnieren