Sporozoite immunization of human volunteers under mefloquine prophylaxis is safe, immunogenic and protective: a double-blind randomized controlled clinical trial

Else M Bijker, Remko Schats, Joshua M Obiero, Marije C Behet, Geert-Jan van Gemert, Marga van de Vegte-Bolmer, Wouter Graumans, Lisette van Lieshout, Guido J H Bastiaens, Karina Teelen, Cornelus C Hermsen, Anja Scholzen, Leo G Visser, Robert W Sauerwein, Else M Bijker, Remko Schats, Joshua M Obiero, Marije C Behet, Geert-Jan van Gemert, Marga van de Vegte-Bolmer, Wouter Graumans, Lisette van Lieshout, Guido J H Bastiaens, Karina Teelen, Cornelus C Hermsen, Anja Scholzen, Leo G Visser, Robert W Sauerwein

Abstract

Immunization of healthy volunteers with chloroquine ChemoProphylaxis and Sporozoites (CPS-CQ) efficiently and reproducibly induces dose-dependent and long-lasting protection against homologous Plasmodium falciparum challenge. Here, we studied whether chloroquine can be replaced by mefloquine, which is the only other licensed anti-malarial chemoprophylactic drug that does not affect pre-erythrocytic stages, exposure to which is considered essential for induction of protection by CPS immunization. In a double blind randomized controlled clinical trial, volunteers under either chloroquine prophylaxis (CPS-CQ, n = 5) or mefloquine prophylaxis (CPS-MQ, n = 10) received three sub-optimal CPS immunizations by bites from eight P. falciparum infected mosquitoes each, at monthly intervals. Four control volunteers received mefloquine prophylaxis and bites from uninfected mosquitoes. CPS-MQ immunization is safe and equally potent compared to CPS-CQ inducing protection in 7/10 (70%) versus 3/5 (60%) volunteers, respectively. Furthermore, specific antibody levels and cellular immune memory responses were comparable between both groups. We therefore conclude that mefloquine and chloroquine are equally effective in CPS-induced immune responses and protection. Trial registration: ClinicalTrials.gov NCT01422954.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Study flow diagram.
Figure 1. Study flow diagram.
Thirty-six subjects were screened for eligibility, of whom twenty were included in the trial and randomized over three groups. One control subject was excluded after initiation of chemoprophylaxis but before the first immunization because of an unexpected visit to a malaria-endemic area during the study period. In a double-blind fashion, fifteen subjects received either CPS-CQ or CPS-MQ immunization and four control subjects received bites from uninfected mosquitoes and mefloquine prophylaxis. Subjects received a challenge infection by bites of five infected mosquitoes sixteen weeks after discontinuation of prophylaxis.
Figure 2. Parasitemia during CPS immunization.
Figure 2. Parasitemia during CPS immunization.
Parasitemia was determined retrospectively, once daily from day 6 until day 10 after each immunization, by real-time quantitative PCR (qPCR). Each line represents an individual subject from the CPS-MQ (dashed blue lines) or CPS-CQ group (red lines). The number of subjects with a positive qPCR/total number of volunteers in the CPS-MQ (blue) and CPS-CQ (red) groups after each immunization are shown above the graph. Values shown as 17.5 on the log-scale were negative (i.e. half the detection limit of the qPCR: 35 parasites/ml).
Figure 3. Adverse events during CPS immunization.
Figure 3. Adverse events during CPS immunization.
Percentage of volunteers in each group experiencing possibly or probably related AE after the first (I), second (II) and third (III) immunization. AEs were evaluated at each visit and graded for severity as described in the methods paragraph: mild (light grey), moderate (dark grey) and severe (black). Only the highest intensity per subject is listed. No Serious Adverse Events occurred.
Figure 4. Parasitemia after challenge infection.
Figure 4. Parasitemia after challenge infection.
Parasitemia was assessed retrospectively by real-time quantitative PCR (qPCR) twice daily from day 5 until day 15 and once daily up until day 21 after challenge. Each line represents an individual subject. Red lines represent CPS-CQ immunized volunteers (n = 5), dashed blue lines CPS-MQ immunized subjects (n = 10) and dotted grey lines malaria-naive control subjects (n = 4). Values shown as 17.5 on the log-scale were negative (i.e. half the detection limit of the qPCR: 35 parasites/ml).
Figure 5. Antibody responses induced by CPS-CQ…
Figure 5. Antibody responses induced by CPS-CQ and CPS-MQ immunization.
Antibodies against CSP (A and B; in AU), LSA-1 (C and D), and MSP-1 (E and F) were analyzed at baseline (B), 28 days after the first (I1) and second (I2) immunization and one day before challenge (C-1; 20 weeks after the last immunization) for all CPS-CQ (A, C and E, n = 5) and CPS-MQ (B, D and F, n = 10) immunized volunteers. Data are shown as individual titers with medians. Open squares indicate protected subjects, filled circles indicate unprotected subjects. Differences between the time points were analyzed by Friedman test with Dunn’s multiple comparison post-hoc test. Significant differences are indicated by asterices with * (p

Figure 6. Cellular immune responses: CD107a expression…

Figure 6. Cellular immune responses: CD107a expression by CD4 T cells and granzyme B production…

Figure 6. Cellular immune responses: CD107a expression by CD4 T cells and granzyme B production by CD8 T cells.
CD107a expression by CD4 T cells after PfRBC re-stimulation, corrected for uRBC background in CPS-CQ (A) and CPS-MQ (B) groups; granzyme B production by CD8 T cells after PfRBC re-stimulation, corrected for uRBC background in CPS-CQ (C) and CPS-MQ (D) groups. Symbols and lines represent individual subjects before immunization (B) and one day before challenge (C-1). Open squares indicate protected subjects, filled circles indicate unprotected subjects. Differences between B and C-1 for all subjects were tested by Wilcoxon matched-pairs signed rank test.
Figure 6. Cellular immune responses: CD107a expression…
Figure 6. Cellular immune responses: CD107a expression by CD4 T cells and granzyme B production by CD8 T cells.
CD107a expression by CD4 T cells after PfRBC re-stimulation, corrected for uRBC background in CPS-CQ (A) and CPS-MQ (B) groups; granzyme B production by CD8 T cells after PfRBC re-stimulation, corrected for uRBC background in CPS-CQ (C) and CPS-MQ (D) groups. Symbols and lines represent individual subjects before immunization (B) and one day before challenge (C-1). Open squares indicate protected subjects, filled circles indicate unprotected subjects. Differences between B and C-1 for all subjects were tested by Wilcoxon matched-pairs signed rank test.

References

    1. World Health Organization (2013) World malaria report 2013.
    1. Doolan DL, Dobano C, Baird JK (2009) Acquired immunity to malaria. Clin Microbiol Rev 22: 13–36.
    1. Tran TM, Li S, Doumbo S, Doumtabe D, Huang CY, et al. (2013) An Intensive Longitudinal Cohort Study of Malian Children and Adults Reveals No Evidence of Acquired Immunity to Plasmodium falciparum Infection. Clin Infect Dis 57: 40–47.
    1. RTS'S Clinical Trials Partnership, Agnandji ST, Lell B, Fernandes JF, Abossolo BP, et al. (2012) A phase 3 trial of RTS, S/AS01 malaria vaccine in African infants. N Engl J Med 367: 2284–2295.
    1. Crompton PD, Pierce SK, Miller LH (2010) Advances and challenges in malaria vaccine development. J Clin Invest 120: 4168–4178.
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, et al. (2009) Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 361: 468–477.
    1. Bijker EM, Teirlinck AC, Schats R, van Gemert G-J, van de Vegte-Bolmer M, et al.. (2014) Cytotoxic Markers Associate with Protection against Malaria in Human Volunteers Immunized with Plasmodium falciparum Sporozoites. Journal of Infectious Diseases: jiu293.
    1. Roestenberg M, Teirlinck AC, McCall MBB, Teelen K, Makamdop KN, et al. (2011) Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 377: 1770–1776.
    1. Bijker EM, Bastiaens GJ, Teirlinck AC, van Gemert GJ, Graumans W, et al. (2013) Protection against malaria after immunization by chloroquine prophylaxis and sporozoites is mediated by preerythrocytic immunity. Proc Natl Acad Sci U S A 110: 7862–7867.
    1. Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, et al. (2002) Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis 185: 1155–1164.
    1. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, et al.. (2013) Protection Against Malaria by Intravenous Immunization with a Nonreplicating Sporozoite Vaccine. Science.
    1. Sauerwein RW, Bijker EM, Richie TL (2010) Empowering malaria vaccination by drug administration. Curr Opin Immunol 22: 367–373.
    1. Accapezzato D, Visco V, Francavilla V, Molette C, Donato T, et al. (2005) Chloroquine enhances human CD8+ T cell responses against soluble antigens in vivo. J Exp Med 202: 817–828.
    1. Garulli B, Di Mario G, Sciaraffia E, Accapezzato D, Barnaba V, et al. (2013) Enhancement of T cell-mediated immune responses to whole inactivated influenza virus by chloroquine treatment in vivo. Vaccine 31: 1717–1724.
    1. Behet MC, Foquet L, van Gemert GJ, Bijker EM, Meuleman P, et al. (2014) Sporozoite immunization of human volunteers under chemoprophylaxis induces functional antibodies against pre-erythrocytic stages of Plasmodium falciparum. Malar J 13: 136.
    1. Felgner PL, Roestenberg M, Liang L, Hung C, Jain A, et al. (2013) Pre-erythrocytic antibody profiles induced by controlled human malaria infections in healthy volunteers under chloroquine prophylaxis. Sci Rep 3: 3549.
    1. Nahrendorf W, Scholzen A, Bijker EM, Teirlinck AC, Bastiaens GJ, et al.. (2014) Memory B-cell and antibody responses induced by Plasmodium falciparum sporozoite immunization. Journal of Infectious Diseases: jiu354.
    1. CDC (2014) Accessed 27-OCT-2014.
    1. Nederlandsche Internisten Vereeniging, Hartstichting N, voor Cardiologie NV, voor Heelkunde NV, voor Neurologie NV, et al.. (2006) Multidisciplinaire Richtlijn Cardiovasculair risicomanagement.
    1. Verhage DF, Telgt DS, Bousema JT, Hermsen CC, van Gemert GJ, et al. (2005) Clinical outcome of experimental human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J Med 63: 52–58.
    1. Lejeune D, Souletie I, Houze S, Le bricon T, Le bras J, et al. (2007) Simultaneous determination of monodesethylchloroquine, chloroquine, cycloguanil and proguanil on dried blood spots by reverse-phase liquid chromatography. J Pharm Biomed Anal 43: 1106–1115.
    1. Ponnudurai T, Lensen AH, Van Gemert GJ, Bensink MP, Bolmer M, et al. (1989) Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes. Parasitology 98 Pt 2: 165–173.
    1. Hermsen CC, Telgt DS, Linders EH, van de Locht LA, Eling WM, et al. (2001) Detection of Plasmodium falciparum malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol 118: 247–251.
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, et al. (2009) Protection against a malaria challenge by sporozoite inoculation. The New England journal of medicine 361: 468–477.
    1. GraphPad (Last accessed: 4 Dec 2013) QuickCalcs. Accessed.
    1. Lobel HO, Miani M, Eng T, Bernard KW, Hightower AW, et al. (1993) Long-term malaria prophylaxis with weekly mefloquine. Lancet 341: 848–851.
    1. Steffen R, Fuchs E, Schildknecht J, Naef U, Funk M, et al. (1993) Mefloquine compared with other malaria chemoprophylactic regimens in tourists visiting east Africa. Lancet 341: 1299–1303.
    1. Croft AM, Clayton TC, World MJ (1997) Side effects of mefloquine prophylaxis for malaria: an independent randomized controlled trial. Trans R Soc Trop Med Hyg 91: 199–203.
    1. Schlagenhauf P, Tschopp A, Johnson R, Nothdurft HD, Beck B, et al. (2003) Tolerability of malaria chemoprophylaxis in non-immune travellers to sub-Saharan Africa: multicentre, randomised, double blind, four arm study. BMJ 327: 1078.
    1. Barrett PJ, Emmins PD, Clarke PD, Bradley DJ (1996) Comparison of adverse events associated with use of mefloquine and combination of chloroquine and proguanil as antimalarial prophylaxis: postal and telephone survey of travellers. BMJ 313: 525–528.
    1. Centers for Disease Control and Prevention . Accessed 27 October 2014.
    1. Ponnudurai T, Lensen AH, van Gemert GJ, Bolmer MG, Meuwissen JH (1991) Feeding behaviour and sporozoite ejection by infected Anopheles stephensi. Trans R Soc Trop Med Hyg 85: 175–180.
    1. Duffy PE, Sahu T, Akue A, Milman N, Anderson C (2012) Pre-erythrocytic malaria vaccines: identifying the targets. Expert review of vaccines 11: 1261–1280.
    1. Roestenberg M, de Vlas SJ, Nieman A-E, Sauerwein RW, Hermsen CC (2012) Efficacy of preerythrocytic and blood-stage malaria vaccines can be assessed in small sporozoite challenge trials in human volunteers. J Infect Dis 206: 319–323.
    1. Roestenberg M, O'Hara GA, Duncan CJA, Epstein JE, Edwards NJ, et al. (2012) Comparison of clinical and parasitological data from controlled human malaria infection trials. PLoS One 7: e38434.
    1. Rombo L, Bergqvist Y, Hellgren U (1987) Chloroquine and desethylchloroquine concentrations during regular long-term malaria prophylaxis. Bull World Health Organ 65: 879–883.
    1. Palmer KJ, Holliday SM, Brogden RN (1993) Mefloquine. Drugs 45: 430–475.
    1. Nganou-Makamdop K, Sauerwein RW (2013) Liver or blood-stage arrest during malaria sporozoite immunization: the later the better? Trends Parasitol 29: 304–310.

Source: PubMed

3
購読する