Safety and comparability of controlled human Plasmodium falciparum infection by mosquito bite in malaria-naïve subjects at a new facility for sporozoite challenge

Angela K Talley, Sara A Healy, Olivia C Finney, Sean C Murphy, James Kublin, Carola J Salas, Susan Lundebjerg, Peter Gilbert, Wesley C Van Voorhis, John Whisler, Ruobing Wang, Chris F Ockenhouse, D Gray Heppner, Stefan H Kappe, Patrick E Duffy, Angela K Talley, Sara A Healy, Olivia C Finney, Sean C Murphy, James Kublin, Carola J Salas, Susan Lundebjerg, Peter Gilbert, Wesley C Van Voorhis, John Whisler, Ruobing Wang, Chris F Ockenhouse, D Gray Heppner, Stefan H Kappe, Patrick E Duffy

Abstract

Background: Controlled human malaria infection (CHMI) studies which recapitulate mosquito-borne infection are a critical tool to identify protective vaccine and drug candidates for advancement to field trials. In partnership with the Walter Reed Army Institute of Research, the CHMI model was established at the Seattle Biomedical Research Institute's Malaria Clinical Trials Center (MCTC). Activities and reagents at both centers were aligned to ensure comparability and continued safety of the model. To demonstrate successful implementation, CHMI was performed in six healthy malaria-naïve volunteers.

Methods: All volunteers received NF54 strain Plasmodium falciparum by the bite of five infected Anopheles stephensi mosquitoes under controlled conditions and were monitored for signs and symptoms of malaria and for parasitemia by peripheral blood smear. Subjects were treated upon diagnosis with chloroquine by directly observed therapy. Immunological (T cell and antibody) and molecular diagnostic (real-time quantitative reverse transcriptase polymerase chain reaction [qRT-PCR]) assessments were also performed.

Results: All six volunteers developed patent parasitemia and clinical malaria. No serious adverse events occurred during the study period or for six months post-infection. The mean prepatent period was 11.2 days (range 9-14 days), and geometric mean parasitemia upon diagnosis was 10.8 parasites/µL (range 2-69) by microscopy. qRT-PCR detected parasites an average of 3.7 days (range 2-4 days) earlier than blood smears. All volunteers developed antibodies to the blood-stage antigen merozoite surface protein 1 (MSP-1), which persisted up to six months. Humoral and cellular responses to pre-erythrocytic antigens circumsporozoite protein (CSP) and liver-stage antigen 1 (LSA-1) were limited.

Conclusion: The CHMI model was safe, well tolerated and characterized by consistent prepatent periods, pre-symptomatic diagnosis in 3/6 subjects and adverse event profiles as reported at established centers. The MCTC can now evaluate candidates in the increasingly diverse vaccine and drug pipeline using the CHMI model.

Trial registration: ClinicalTrials.gov NCT01058226.

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.
Eighteen subjects were screened for eligibility to participate in the trial and 7 healthy volunteers were considered eligible and willing to participate. On the day of enrollment, 6 subjects were enrolled and one backup subject was discharged from the study. The six subjects underwent CHMI and completed the 56 day study. Five subjects returned for optional long term safety and immunology follow up assessments at 3 and 6 months post-challenge.
Figure 2. Comparison of prepatent and incubation…
Figure 2. Comparison of prepatent and incubation periods.
Kaplan-Meier survival curve showing the percentage of subjects without patent parasitemia by blood smear (blue line) or without symptoms (dashed red line) following challenge.
Figure 3. qRT-PCR-based course of parasitemia.
Figure 3. qRT-PCR-based course of parasitemia.
Parasite density based on qRT-PCR measurements are presented individually for each participant.
Figure 4. Humoral immune responses to P.…
Figure 4. Humoral immune responses to P. falciparum antigens.
ELISAs were performed on the indicated days post-CHMI to test for responses against the indicated P. falciparum antigens. The positivity cut-off (dotted line) was calculated per ELISA plate as three standard deviations above the mean of the two negative control wells. All samples with an OD higher than the calculated cut-off were deemed positive. Data are presented for the five subjects completing the follow up at 3 and 6 months. Data to Day 56 for the sixth subject did not differ considerably from the five subjects in the graph.
Figure 5. Cellular immune response to CSP…
Figure 5. Cellular immune response to CSP and LSA-1.
IFNγ ELISpot assays were performed using the indicated CSP and LSA-1 peptide pools on the indicated days post-CHMI. Spot forming units (SFU) per million PBMC are shown per individual subject for all six subjects.

References

    1. Church LW, Le TP, Bryan JP, Gordon DM, Edelman R, et al. (1997) Clinical manifestations of Plasmodium falciparum malaria experimentally induced by mosquito challenge. J Infect Dis 175: 915–920.
    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. Lyke KE, Laurens M, Adams M, Billingsley PF, Richman A, et al. (2010) Plasmodium falciparum malaria challenge by the bite of aseptic Anopheles stephensi mosquitoes: results of a randomized infectivity trial. PLoS One 5: e13490.
    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. Chulay JD, Schneider I, Cosgriff TM, Hoffman SL, Ballou WR, et al. (1986) Malaria transmitted to humans by mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg 35: 66–68.
    1. Epstein JE, Rao S, Williams F, Freilich D, Luke T, et al. (2007) Safety and clinical outcome of experimental challenge of human volunteers with Plasmodium falciparum-infected mosquitoes: an update. J Infect Dis 196: 145–154.
    1. Hoffman SL (1997) Experimental challenge of volunteers with malaria. Ann Intern Med 127: 233–235.
    1. Spring M, Polhemus M, Ockenhouse C (2014) Controlled human malaria infection. J Infect Dis 209 Suppl 2S40–45.
    1. Herrera S, Solarte Y, Jordan-Villegas A, Echavarria JF, Rocha L, et al. (2011) Consistent safety and infectivity in sporozoite challenge model of Plasmodium vivax in malaria-naive human volunteers. Am J Trop Med Hyg 84: 4–11.
    1. Herrera S, Fernandez O, Manzano MR, Murrain B, Vergara J, et al. (2009) Successful sporozoite challenge model in human volunteers with Plasmodium vivax strain derived from human donors. Am J Trop Med Hyg 81: 740–746.
    1. Douglas AD, Edwards NJ, Duncan CJ, Thompson FM, Sheehy SH, et al. (2013) Comparison of modeling methods to determine liver-to-blood inocula and parasite multiplication rates during controlled human malaria infection. J Infect Dis 208: 340–345.
    1. Bejon P, Andrews L, Andersen RF, Dunachie S, Webster D, et al. (2005) Calculation of liver-to-blood inocula, parasite growth rates, and preerythrocytic vaccine efficacy, from serial quantitative polymerase chain reaction studies of volunteers challenged with malaria sporozoites. J Infect Dis 191: 619–626.
    1. Roestenberg M, O'Hara GA, Duncan CJ, Epstein JE, Edwards NJ, et al. (2012) Comparison of clinical and parasitological data from controlled human malaria infection trials. PLoS One 7: e38434.
    1. Ballou WR, Arevalo-Herrera M, Carucci D, Richie TL, Corradin G, et al. (2004) Update on the clinical development of candidate malaria vaccines. Am J Trop Med Hyg 71: 239–247.
    1. Gaziano TA, Young CR, Fitzmaurice G, Atwood S, Gaziano JM (2008) Laboratory-based versus non-laboratory-based method for assessment of cardiovascular disease risk: the NHANES I Follow-up Study cohort. Lancet 371: 923–931.
    1. Murphy SC, Prentice JL, Williamson K, Wallis CK, Fang FC, et al. (2012) Real-time quantitative reverse transcription PCR for monitoring of blood-stage Plasmodium falciparum infections in malaria human challenge trials. Am J Trop Med Hyg 86: 383–394.
    1. Miura K, Orcutt AC, Muratova OV, Miller LH, Saul A, et al. (2008) Development and characterization of a standardized ELISA including a reference serum on each plate to detect antibodies induced by experimental malaria vaccines. Vaccine 26: 193–200.
    1. McElrath MJ, De Rosa SC, Moodie Z, Dubey S, Kierstead L, et al. (2008) HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case-cohort analysis. Lancet 372: 1894–1905.
    1. Dubey S, Clair J, Fu TM, Guan L, Long R, et al. (2007) Detection of HIV vaccine-induced cell-mediated immunity in HIV-seronegative clinical trial participants using an optimized and validated enzyme-linked immunospot assay. J Acquir Immune Defic Syndr 45: 20–27.
    1. James SP, Shute PG (1926) Malaria Commission: report on the first results of laboratory work on malaria in England. Geneva: League of Nations Health Organization.
    1. Nicol WD (1927) The care and management of induced malaria. J Ment Sci 6: 1–9.
    1. James SP (1931) Some general results of a study of induced malaria in England. Trans R Soc Trop Med Hyg 24: 477–538.
    1. Laurens MB, Duncan CJ, Epstein JE, Hill AV, Komisar JL, et al. (2012) A consultation on the optimization of controlled human malaria infection by mosquito bite for evaluation of candidate malaria vaccines. Vaccine 30: 5302–5304.
    1. Murphy SC, Hermsen CC, Douglas AD, Edwards N, Petersen I, et al. (2014) External quality assurance of malaria nucleic acid testing for clinical trials and eradication surveillance. PLoS One 9: e97398.
    1. Murphy SC, Daza G, Chang M, Coombs R (2012) Laser cutting eliminates nucleic acid cross-contamination in dried-blood-spot processing. J Clin Microbiol 50: 4128–4130.
    1. Zevering Y, Amante F, Smillie A, Currier J, Smith G, et al. (1992) High frequency of malaria-specific T cells in non-exposed humans. Eur J Immunol 22: 689–696.
    1. Todryk SM, Walther M, Bejon P, Hutchings C, Thompson FM, et al. (2009) Multiple functions of human T cells generated by experimental malaria challenge. Eur J Immunol 39: 3042–3051.

Source: PubMed

3
Suscribir