Whole sporozoite immunization with Plasmodium falciparum strain NF135 in a randomized trial

Saskia C van der Boor, Manon Alkema, Geert-Jan van Gemert, Karina Teelen, Marga van de Vegte-Bolmer, Jona Walk, Reinout van Crevel, Quirijn de Mast, Christian F Ockenhouse, Robert W Sauerwein, Matthew B B McCall, Saskia C van der Boor, Manon Alkema, Geert-Jan van Gemert, Karina Teelen, Marga van de Vegte-Bolmer, Jona Walk, Reinout van Crevel, Quirijn de Mast, Christian F Ockenhouse, Robert W Sauerwein, Matthew B B McCall

Abstract

Background: Whole sporozoite immunization under chemoprophylaxis (CPS regime) induces long-lasting sterile homologous protection in the controlled human malaria infection model using Plasmodium falciparum strain NF54. The relative proficiency of liver-stage parasite development may be an important factor determining immunization efficacy. Previous studies show that Plasmodium falciparum strain NF135 produces relatively high numbers of large liver-stage schizonts in vitro. Here, we evaluate this strain for use in CPS immunization regimes.

Methods: In a partially randomized, open-label study conducted at the Radboudumc, Nijmegen, the Netherlands, healthy, malaria-naïve adults were immunized by three rounds of fifteen or five NF135-infected mosquito bites under mefloquine prophylaxis (cohort A) or fifteen NF135-infected mosquito bites and presumptive treatment with artemether/lumefantrine (cohort B). Cohort A participants were exposed to a homologous challenge 19 weeks after immunization. The primary objective of the study was to evaluate the safety and tolerability of CPS immunizations with NF135.

Results: Relatively high liver-to-blood inocula were observed during immunization with NF135 in both cohorts. Eighteen of 30 (60%) high-dose participants and 3/10 (30%) low-dose participants experienced grade 3 adverse events 7 to 21 days following their first immunization. All cohort A participants and two participants in cohort B developed breakthrough blood-stage malaria infections during immunizations requiring rescue treatment. The resulting compromised immunizations induced modest sterile protection against homologous challenge in cohort A (5/17; 29%).

Conclusions: These CPS regimes using NF135 were relatively poorly tolerated and frequently required rescue treatment, thereby compromising immunization efficiency and protective efficacy. Consequently, the full potential of NF135 sporozoites for induction of immune protection remains inconclusive. Nonetheless, the high liver-stage burden achieved by this strain highlights it as an interesting potential candidate for novel whole sporozoite immunization approaches.

Trial registration: The trial was registered at ClinicalTrials.gov under identifier NCT03813108.

Keywords: Malaria; NF135; Plasmodium falciparum; Vaccine-induced protection; Whole sporozoite immunization.

Conflict of interest statement

The authors declare they have no competing interests.

© 2023. The Author(s).

Figures

Fig. 1
Fig. 1
Flow chart. Eighty-nine candidates were tested for eligibility and fourty-six were included in the study after screening: twenty-three in cohort A (three additional participants started mefloquine prophylaxis in case of dropouts prior to the first immunization), twenty in cohort B, and three in the control groups. Participants in cohort A were randomized to receive immunizations with either three times fifteen NF135-infected mosquitoes (high-dose) or three times five NF135-infected mosquitoes (low-dose). One participant dropped out after the first immunization and two participants after the third immunization, all three because of personal or logistical reasons. Participants in cohort B received one immunization with fifteen NF135-infected mosquitoes (high-dose). The control group was not immunized. Immunized participants from cohort A, and three malaria-naïve control participants were challenged by bites of five NF135-infected mosquitoes. In cohort B, the trial was prematurely ended after the first immunization and participants were not subsequently challenged
Fig. 2
Fig. 2
Parasitemia on day seven after immunization 1. Each dot represents one participant, the line represents the median value. Participants in the high-dose group were immunized with fifteen NF135-infected mosquitoes. Participants in the low-dose group were immunized with five NF135-infected mosquitoes. The statistical analysis performed was Welch’s t-test. ****p < 0001
Fig. 3
Fig. 3
Parasitemia after immunization in cohort A. Individual parasite curves are presented in gray. The median parasite density for each group is presented in green. Dashed lines indicate an immunization. A Participants immunized with the low-dose of three times five NF135-infected mosquitoes. B Participants immunized with the high dose of three times fifteen NF135-infected mosquitoes. C Number of participants with a positive qPCR and/or thick smear after each immunization
Fig. 4
Fig. 4
Sterile protection against PfNF135 mosquito bite challenge. A Kaplan–Meier curve depicting percentage of participants that remained sterilely protected after homologous challenge infection with PfNF135. Participants in the high-dose arm received three immunizations by bites of fifteen PfNF135-infected mosquitoes. Participants in the low-dose arm received three immunizations by bites of five infected mosquitoes with the same parasite strain. The control group received no immunizations. B Percentage of sterilely protected partially protected and unprotected participants after controlled human malaria infection with the homologous NF135 strain in each study arm. Participants in the high-dose arm received three immunizations by bites of fifteen P. falciparum NF135-infected mosquitoes and participants in the low-dose arm received three immunizations by bites of five infected mosquitoes with the same parasite clone. Mosq, mosquitoes
Fig. 5
Fig. 5
Anti-sporozoite IgG titers. IgG antibody titers against sporozoite extract in sera of cohort A participants immunized with three times fifteen PfNF135-infected mosquitoes (purple, n = 10) and of participants in a historical comparator trial immunized with three times fifteen PfNF54-infected mosquitoes (blue, n = 12), at baseline, after the first immunization, and after completion of the course of three immunizations. Thick and thin horizontal lines represent median and interquartile range, respectively. The Mann–Whitney U test was used for differences between studies. Friedman test with post hoc paired comparison using Bonferroni-corrected Wilcoxon signed rank test was used for comparisons of time points within studies
Fig. 6
Fig. 6
Adverse events after immunization 1. A Number of grade 1, 2, 3, and 4 adverse events per participant per cohort. B Percentage of participants who experienced a grade 1, 2, 3, or 4 adverse event per cohort. AE, Adverse event

References

    1. World Health Organization World malaria report 2021. Geneva. Licence: CC BY-NC-SA 3.0 IGO.
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361(5):468–77.
    1. Roestenberg M, Teirlinck AC, McCall MB, Teelen K, Makamdop KN, Wiersma J, et al. Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet. 2011;377(9779):1770–6.
    1. Mordmüller B, Surat G, Lagler H, Chakravarty S, Ishizuka AS, Lalremruata A, et al. Sterile protection against human malaria by chemoattenuated PfSPZ vaccine. Nature. 2017;542(7642):445–449. doi: 10.1038/nature21060.
    1. Sulyok Z, Fendel R, Eder B, Lorenz FR, Kc N, Karnahl M, et al. Heterologous protection against malaria by a simple chemoattenuated PfSPZ vaccine regimen in a randomized trial. Nat Commun. 2021;12(1):2518. doi: 10.1038/s41467-021-22740-w.
    1. Mwakingwe-Omari A, Healy SA, Lane J, Cook DM, Kalhori S, Wyatt C, et al. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature. 2021;595(7866):289–294. doi: 10.1038/s41586-021-03684-z.
    1. Jongo SA, Urbano V, Church LWP, Olotu A, Manock SR, Schindler T, et al. Immunogenicity and protective efficacy of radiation-attenuated and chemo-attenuated PfSPZ vaccines in Equatoguinean adults. Am J Trop Med Hyg. 2021;104(1):283–293. doi: 10.4269/ajtmh.20-0435.
    1. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science. 2013;341(6152):1359–65.
    1. Roestenberg M, Walk J, van der Boor SC, Langenberg MCC, Hoogerwerf MA, Janse JJ, et al. A double-blind, placebo-controlled phase 1/2a trial of the genetically attenuated malaria vaccine PfSPZ-GA1. Science translational medicine. 2020;12(544):eaaz5629.
    1. Epstein JE, Paolino KM, Richie TL, Sedegah M, Singer A, Ruben AJ, et al. Protection against Plasmodium falciparum malaria by PfSPZ vaccine. JCI Insight. 2017;2(1):e89154.
    1. McCall MBB, Wammes LJ, Langenberg MCC, van Gemert GJ, Walk J, Hermsen CC, et al. Infectivity of Plasmodium falciparum sporozoites determines emerging parasitemia in infected volunteers. Science translational medicine. 2017;9(395):eaag2490.
    1. Yang ASP, van Waardenburg YM, van de Vegte-Bolmer M, van Gemert GA, Graumans W, de Wilt JHW, et al. Zonal human hepatocytes are differentially permissive to Plasmodium falciparum malaria parasites. EMBO J. 2021;40(6):e106583. doi: 10.15252/embj.2020106583.
    1. Langenberg MCC, Wammes LJ, McCall MBB, Bijker EM, van Gemert GJ, Graumans W, et al. Controlled human malaria infection with graded numbers of Plasmodium falciparum NF135.C10- or NF166.C8-infected mosquitoes. Am J Trop Med Hyg. 2018;99(3):709–12. doi: 10.4269/ajtmh.18-0194.
    1. Teirlinck AC, Roestenberg M, van de Vegte-Bolmer M, Scholzen A, Heinrichs MJ, Siebelink-Stoter R, et al. NF135.C10: a new Plasmodium falciparum clone for controlled human malaria infections. J Infect Dis. 2013;207(4):656–60. doi: 10.1093/infdis/jis725.
    1. Mylan. Atovaquone/Proguanil Hydrochloride 250 mg/100 mg Film-coated tablets [updated 05 Jul 2017. Available from: .
    1. Ponnudurai T, Lensen AH, Gemert GJ, Bensink MP, Bolmer M, Meuwissen JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes. Parasitology. 1989;98:165–73. doi: 10.1017/S0031182000062065.
    1. Hermsen CC, Telgt DS, Linders EH, Locht LA, Eling WM, Mensink EJ, et al. Detection of Plasmodium falciparum malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol. 2001;118(2):247–51. doi: 10.1016/S0166-6851(01)00379-6.
    1. Gallay J, Prod'hom S, Mercier T, Bardinet C, Spaggiari D, Pothin E, et al. LC-MS/MS method for the simultaneous analysis of seven antimalarials and two active metabolites in dried blood spots for applications in field trials: Analytical and clinical validation. J Pharm Biomed Anal. 2018;154:263–77. doi: 10.1016/j.jpba.2018.01.017.
    1. Nixon GL, Moss DM, Shone AE, Lalloo DG, Fisher N, O’Neill PM, et al. Antimalarial pharmacology and therapeutics of atovaquone. J Antimicrob Chemother. 2013;68(5):977–985. doi: 10.1093/jac/dks504.
    1. Walk J, Reuling IJ, Behet MC, Meerstein-Kessel L, Graumans W, van Gemert GJ, et al. Modest heterologous protection after Plasmodium falciparum sporozoite immunization: a double-blind randomized controlled clinical trial. BMC Med. 2017;15(1):168. doi: 10.1186/s12916-017-0923-4.
    1. Edstein MD, Kotecka BM, Anderson KL, Pombo DJ, Kyle DE, Rieckmann KH, et al. Lengthy antimalarial activity of atovaquone in human plasma following atovaquone-proguanil administration. Antimicrob Agents Chemother. 2005;49(10):4421–4422. doi: 10.1128/AAC.49.10.4421-4422.2005.
    1. Butcher GA, Sinden RE. Persistence of atovaquone in human sera following treatment: inhibition of Plasmodium falciparum development in vivo and in vitro. Am J Trop Med Hyg. 2003;68(1):111–114. doi: 10.4269/ajtmh.2003.68.111.
    1. Murphy SC, Deye GA, Sim BKL, Galbiati S, Kennedy JK, Cohen KW, 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(5):e1009594. doi: 10.1371/journal.ppat.1009594.
    1. Bejon P, Mwacharo J, Kai O, Todryk S, Keating S, Lowe B, et al. The induction and persistence of T cell IFN-gamma responses after vaccination or natural exposure is suppressed by Plasmodium falciparum. J Immunol. 2007;179(6):4193–4201. doi: 10.4049/jimmunol.179.6.4193.
    1. Ubillos I, Campo JJ, Requena P, Ome-Kaius M, Hanieh S, Rose H, et al. Chronic exposure to malaria is associated with inhibitory and activation markers on atypical memory B Cells and marginal zone-like B cells. Front Immunol. 2017;8:966-.
    1. Bijker EM, Schats R, Obiero JM, Behet MC, Gemert GJ, Vegte-Bolmer M, et al. Sporozoite immunization of human volunteers under mefloquine prophylaxis is safe, immunogenic and protective: a double-blind randomized controlled clinical trial. PLoS One. 2014;9(11):e112910.
    1. Moser KA, Drábek EF, Dwivedi A, Stucke EM, Crabtree J, Dara A, et al. Strains used in whole organism Plasmodium falciparum vaccine trials differ in genome structure, sequence, and immunogenic potential. Genome Med. 2020;12(1):6. doi: 10.1186/s13073-019-0708-9.
    1. Price RN, Uhlemann AC, Brockman A, McGready R, Ashley E, Phaipun L, et al. Mefloquine resistance in Plasmodium falciparum and increased pfmdr1 gene copy number. Lancet. 2004;364(9432):438–447. doi: 10.1016/S0140-6736(04)16767-6.
    1. Sondén K, Wyss K, Jovel I, Vieira da Silva A, Pohanka A, Asghar M, et al. High rate of treatment failures in nonimmune travelers treated with artemether-lumefantrine for uncomplicated Plasmodium falciparum malaria in Sweden: retrospective comparative analysis of effectiveness and case series. Clin Infect Dis. 2017;64(2):199–206. doi: 10.1093/cid/ciw710.
    1. Mwai L, Kiara SM, Abdirahman A, Pole L, Rippert A, Diriye A, et al. In vitro activities of piperaquine, lumefantrine, and dihydroartemisinin in Kenyan Plasmodium falciparum isolates and polymorphisms in pfcrt and pfmdr1. Antimicrob Agents Chemother. 2009;53(12):5069–5073. doi: 10.1128/AAC.00638-09.
    1. Ngo T, Duraisingh M, Reed M, Hipgrave D, Biggs B, Cowman AF. Analysis of pfcrt, pfmdr1, dhfr, and dhps mutations and drug sensitivities in Plasmodium falciparum isolates from patients in Vietnam before and after treatment with artemisinin. Am J Trop Med Hyg. 2003;68(3):350–356. doi: 10.4269/ajtmh.2003.68.350.
    1. Eyase FL, Akala HM, Ingasia L, Cheruiyot A, Omondi A, Okudo C, et al. The role of Pfmdr1 and Pfcrt in changing chloroquine, amodiaquine, mefloquine and lumefantrine susceptibility in western-Kenya P. falciparum samples during 2008–2011. PLoS One. 2013;8(5):e64299. doi: 10.1371/journal.pone.0064299.
    1. Bijker EM, Bastiaens GJ, Teirlinck AC, Gemert GJ, Graumans W, Vegte-Bolmer M, et al. Protection against malaria after immunization by chloroquine prophylaxis and sporozoites is mediated by preerythrocytic immunity. Proc Natl Acad Sci U S A. 2013;110.
    1. Alkema M, Yap XZ, de Jong GM, Reuling IJ, de Mast Q, van Crevel R, et al. Controlled human malaria infections by mosquito bites induce more severe clinical symptoms than asexual blood-stage challenge infections. EBioMedicine. 2022;77:103919. doi: 10.1016/j.ebiom.2022.103919.
    1. Nieman AE, de Mast Q, Roestenberg M, Wiersma J, Pop G, Stalenhoef A, et al. Cardiac complication after experimental human malaria infection: a case report. Malar J. 2009;8:277. doi: 10.1186/1475-2875-8-277.
    1. van Meer MP, Bastiaens GJ, Boulaksil M, de Mast Q, Gunasekera A, Hoffman SL, et al. Idiopathic acute myocarditis during treatment for controlled human malaria infection: a case report. Malar J. 2014;13:38. doi: 10.1186/1475-2875-13-38.
    1. Franke-Fayard B, Marin-Mogollon C, Geurten FJA, Chevalley-Maurel S, Ramesar J, Kroeze H, et al. Creation and preclinical evaluation of genetically attenuated malaria parasites arresting growth late in the liver. NPJ Vaccines. 2020;7:139.

Source: PubMed

3
Suscribir