Optimising Controlled Human Malaria Infection Studies Using Cryopreserved P. falciparum Parasites Administered by Needle and Syringe

Susanne H Sheehy, Alexandra J Spencer, Alexander D Douglas, B Kim Lee Sim, Rhea J Longley, Nick J Edwards, Ian D Poulton, Domtila Kimani, Andrew R Williams, Nicholas A Anagnostou, Rachel Roberts, Simon Kerridge, Merryn Voysey, Eric R James, Peter F Billingsley, Anusha Gunasekera, Alison M Lawrie, Stephen L Hoffman, Adrian V S Hill, Susanne H Sheehy, Alexandra J Spencer, Alexander D Douglas, B Kim Lee Sim, Rhea J Longley, Nick J Edwards, Ian D Poulton, Domtila Kimani, Andrew R Williams, Nicholas A Anagnostou, Rachel Roberts, Simon Kerridge, Merryn Voysey, Eric R James, Peter F Billingsley, Anusha Gunasekera, Alison M Lawrie, Stephen L Hoffman, Adrian V S Hill

Abstract

Background: Controlled human malaria infection (CHMI) studies have become a routine tool to evaluate efficacy of candidate anti-malarial drugs and vaccines. To date, CHMI trials have mostly been conducted using the bite of infected mosquitoes, restricting the number of trial sites that can perform CHMI studies. Aseptic, cryopreserved P. falciparum sporozoites (PfSPZ Challenge) provide a potentially more accurate, reproducible and practical alternative, allowing a known number of sporozoites to be administered simply by injection.

Methodology: We sought to assess the infectivity of PfSPZ Challenge administered in different dosing regimens to malaria-naive healthy adults (n = 18). Six participants received 2,500 sporozoites intradermally (ID), six received 2,500 sporozoites intramuscularly (IM) and six received 25,000 sporozoites IM.

Findings: Five out of six participants receiving 2,500 sporozoites ID, 3/6 participants receiving 2,500 sporozoites IM and 6/6 participants receiving 25,000 sporozoites IM were successfully infected. The median time to diagnosis was 13.2, 17.8 and 12.7 days for 2,500 sporozoites ID, 2,500 sporozoites IM and 25,000 sporozoites IM respectively (Kaplan Meier method; p = 0.024 log rank test).

Conclusions: 2,500 sporozoites ID and 25,000 sporozoites IM have similar infectivities. Given the dose response in infectivity seen with IM administration, further work should evaluate increasing doses of PfSPZ Challenge IM to identify a dosing regimen that reliably infects 100% of participants.

Trial registration: ClinicalTrials.gov NCT01465048.

Conflict of interest statement

Competing Interests: BKLS, ERJ, PFB, AG, and SLH are employees of Sanaria Inc., which manufactures PfSPZ Challenge. Sanaria also manufactures other products in development based on aseptic, purified, cryopreserved PfSPZ, including PfSPZ Vaccine and PfSPZ-CVac. Sanaria sells parasites and mosquitoes to the research community, but none of its major products in development have been commercialized. BKLS and SLH are inventors on patents relevant to PfSPZ Challenge. None of the authors have had any consultancies relevant to this paper. This conflict of interest does not alter these authors’ adherence to all PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Figure 1. Flow chart of study design…
Figure 1. Flow chart of study design and volunteer recruitment.
20 participants were excluded following screening for the following reasons: significant psychiatric history (six individuals), consent withdrawn (four individuals), recruitment complete (2 individuals), lack of response from General Practitioner to medical screening letter (2 individuals), unexplained systolic murmur, elevated alanine transaminase, unexplained tachycardia and significant prior malaria exposure. In each group, the total dose of sporozoites was split between two injection sites and administered as two 50 µL injections, one in each deltoid.
Figure 2. PfSPZ Challenge Infectivity Data.
Figure 2. PfSPZ Challenge Infectivity Data.
Kaplan-Meier analysis of time to patent parasitemia in days between injection and diagnosis (p = 0.024 logrank test). 2,500 ID = 2,500 sporozoites administered intradermally (ID). 2,500 IM = 2,500 sporozoites administered intramuscularly. 25,000 = 25,000 sporozoites administered intramuscularly. Median pre-patent period = 13.19 days for 2,500 sporozoites ID, 17.8 days for 2,500 sporozoites IM, 12.72 days for 25,000 sporozoites IM.
Figure 3. qPCR-measured parasite density for each…
Figure 3. qPCR-measured parasite density for each individual subject grouped by dosing regimen.
Y axis = qPCR. X axis = days post injection of PfSPZ Challenge. Black subtitles indicate subject identification numbers; red subtitles indicate time to diagnosis by thick film microscopy in days. NI = not infected.
Figure 4. Estimation of Burden of Liver…
Figure 4. Estimation of Burden of Liver Infection and Liver-to-Blood Inoculum.
Data from participants successfully infected with malaria in groups 1, 2 and 3 compared to historical data from mosquito bite CHMI trials undertaken at our centre. SpZ = sporozoites. LLQ = lower limit of quantification by qPCR. ID = intradermal administration. IM = intramuscular administration. Mosquito bite = malaria naïve participants infected with P.falciparum by mosquito bite as infectivity control participants in vaccine efficacy studies undertaken recently at our centre (Ewer et al. submitted). (A) Peak qPCR-measured parasitaemia in first asexual cycle for each regimen. (B) Matrix scatterplot illustrating close correlation of different LBI measures with each other and with time to microscopic patency.
Figure 5. Parasite Multiplication Rate following PfSPZ…
Figure 5. Parasite Multiplication Rate following PfSPZ Challenge is comparable to mosquito-bite subjects with similar LBIs.
Figure shows relationship Between PMR and LBI for participants successfully infected with malaria in groups 1–3 (green crosses), compared to historical data from mosquito bite CHMI trials undertaken at our centre involving malaria naïve unvaccinated subjects (blue dots), and malaria naïve ChAd63-MVA ME-TRAP vaccinated subjects (red dots). These data were obtained using the linear model; results were similar with the sine model (data not shown).
Figure 6. IFN-γ ELISPOT responses to individual…
Figure 6. IFN-γ ELISPOT responses to individual malaria antigens post CHMI.
Median ex-vivo IFN-γ ELISPOT responses from the day before CHMI (C-1) and days 7, 11, 14, 35 and 90 post CHMI with cells stimulated against peptide pools to numerous malaria antigens. Each volunteer is represented as a single point. Lines represent the median response at each time point. (A) Individual responses to numerous malaria antigens over time (B) Sum responses to all peptides over time.

References

    1. Moxon R (2005) Microbial Challenge Studies of Human Volunteers. The Academy of Medical Sciences.
    1. Sauerwein RW, Roestenberg M, Moorthy VS (2011) Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat Rev Immunol 11: 57–64.
    1. Moorthy VS, Diggs C, Ferro S, Good MF, Herrera S, et al. (2009) Report of a consultation on the optimization of clinical challenge trials for evaluation of candidate blood stage malaria vaccines, 18–19 March 2009, Bethesda, MD, USA. Vaccine 27: 5719–5725.
    1. Ponnudurai T, Leeuwenberg AD, Meuwissen JH (1981) Chloroquine sensitivity of isolates of Plasmodium falciparum adapted to in vitro culture. Trop Geogr Med 33: 50–54.
    1. Ponnudurai T, Meuwissen JH, Leeuwenberg AD, Verhave JP, Lensen AH (1982) The production of mature gametocytes of Plasmodium falciparum in continuous cultures of different isolates infective to mosquitoes. Trans R Soc Trop Med Hyg 76: 242–250.
    1. Walliker D, Quakyi IA, Wellems TE, McCutchan TF, Szarfman A, et al. (1987) Genetic analysis of the human malaria parasite Plasmodium falciparum. Science 236: 1661–1666.
    1. Rosenberg R, Wirtz RA, Schneider I, Burge R (1990) An estimation of the number of malaria sporozoites ejected by a feeding mosquito. Trans R Soc Trop Med Hyg 84: 209–212.
    1. Jin Y, Kebaier C, Vanderberg J (2007) Direct microscopic quantification of dynamics of Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infect Immun 75: 5532–5539.
    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. Beier JC, Onyango FK, Koros JK, Ramadhan M, Ogwang R, et al. (1991) Quantitation of malaria sporozoites transmitted in vitro during salivation by wild Afrotropical Anopheles. Med Vet Entomol 5: 71–79.
    1. Frischknecht F, Baldacci P, Martin B, Zimmer C, Thiberge S, et al. (2004) Imaging movement of malaria parasites during transmission by Anopheles mosquitoes. Cell Microbiol 6: 687–694.
    1. Roestenberg M, Bijker EM, Sim BK, Billingsley PF, James ER, et al... (2012) Controlled Human Malaria Infections by Intradermal Injection of Cryopreserved Plasmodium falciparum Sporozoites. Am J Trop Med Hyg.
    1. Ploemen IH, Chakravarty S, van Gemert GJ, Annoura T, Khan SM, et al... (2012) Plasmodium liver load following parenteral sporozoite administration in rodents. Vaccine.
    1. Schwenk RJ, Richie TL (2011) Protective immunity to pre-erythrocytic stage malaria. Trends Parasitol 27: 306–314.
    1. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, et al. (2011) Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 334: 475–480.
    1. Hoffman SL, Isenbarger D, Long GW, Sedegah M, Szarfman A, et al. (1989) Sporozoite vaccine induces genetically restricted T cell elimination of malaria from hepatocytes. Science 244: 1078–1081.
    1. Khusmith S, Sedegah M, Hoffman SL (1994) Complete protection against Plasmodium yoelii by adoptive transfer of a CD8+ cytotoxic T-cell clone recognizing sporozoite surface protein 2. Infect Immun 62: 2979–2983.
    1. Romero P, Maryanski JL, Corradin G, Nussenzweig RS, Nussenzweig V, et al. (1989) Cloned cytotoxic T cells recognize an epitope in the circumsporozoite protein and protect against malaria. Nature 341: 323–326.
    1. Schofield L, Villaquiran J, Ferreira A, Schellekens H, Nussenzweig R, et al. (1987) Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature 330: 664–666.
    1. Weiss WR, Mellouk S, Houghten RA, Sedegah M, Kumar S, et al. (1990) Cytotoxic T cells recognize a peptide from the circumsporozoite protein on malaria-infected hepatocytes. J Exp Med 171: 763–773.
    1. Dodoo D, Hollingdale MR, Anum D, Koram KA, Gyan B, et al. (2011) Measuring naturally acquired immune responses to candidate malaria vaccine antigens in Ghanaian adults. Malar J 10: 168.
    1. Malik A, Egan JE, Houghten RA, Sadoff JC, Hoffman SL (1991) Human cytotoxic T lymphocytes against the Plasmodium falciparum circumsporozoite protein. Proc Natl Acad Sci U S A 88: 3300–3304.
    1. Wizel B, Houghten R, Church P, Tine JA, Lanar DE, et al. (1995) HLA-A2-restricted cytotoxic T lymphocyte responses to multiple Plasmodium falciparum sporozoite surface protein 2 epitopes in sporozoite-immunized volunteers. J Immunol 155: 766–775.
    1. Doolan DL, Hoffman SL, Southwood S, Wentworth PA, Sidney J, et al. (1997) Degenerate cytotoxic T cell epitopes from P. falciparum restricted by multiple HLA-A and HLA-B supertype alleles. Immunity 7: 97–112.
    1. Flanagan KL, Mwangi T, Plebanski M, Odhiambo K, Ross A, et al. (2003) Ex vivo interferon-gamma immune response to thrombospondin-related adhesive protein in coastal Kenyans: longevity and risk of Plasmodium falciparum infection. Am J Trop Med Hyg 68: 421–430.
    1. Doolan DL, Southwood S, Freilich DA, Sidney J, Graber NL, et al. (2003) Identification of Plasmodium falciparum antigens by antigenic analysis of genomic and proteomic data. Proc Natl Acad Sci U S A 100: 9952–9957.
    1. Chelimo K, Embury PB, Sumba PO, Vulule J, Ofulla AV, et al. (2011) Age-related differences in naturally acquired T cell memory to Plasmodium falciparum merozoite surface protein 1. PLoS One 6: e24852.
    1. Hoffman SL, Billingsley PF, James E, Richman A, Loyevsky M, et al. (2010) Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Hum Vaccin 6: 97–106.
    1. Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, et al... (2012) ChAd63-MVA-vectored Blood-stage Malaria Vaccines Targeting MSP1 and AMA1: Assessment of Efficacy Against Mosquito Bite Challenge in Humans. Mol Ther.
    1. Douglas AD, Edwards NJ, Duncan CJ, Thompson FM, Sheehy SH, et al... (2013) Comparison of modelling methods to determine liver-to-blood inocula and parasite multiplication rates during controlled human malaria infection. J Infect Dis.
    1. Sheehy SH, Duncan CJ, Elias SC, Collins KA, Ewer KJ, et al. (2011) Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol Ther 19: 2269–2276.
    1. Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, et al. (2012) ChAd63-MVA-vectored Blood-stage Malaria Vaccines Targeting MSP1 and AMA1: Assessment of Efficacy Against Mosquito Bite Challenge in Humans. Mol Ther 20: 2355–2368.
    1. Sheehy SH, Duncan CJ, Elias SC, Biswas S, Collins KA, et al. (2012) Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors. PLoS One 7: e31208.
    1. O'Hara GA, Duncan CJ, Ewer KJ, Collins KA, Elias SC, et al. (2012) Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J Infect Dis 205: 772–781.
    1. Porter DW, Thompson FM, Berthoud TK, Hutchings CL, Andrews L, et al. (2011) A human Phase I/IIa malaria challenge trial of a polyprotein malaria vaccine. Vaccine 29: 7514–7522.
    1. Perlaza BL, Valencia AZ, Zapata C, Castellanos A, Sauzet JP, et al. (2008) Protection against Plasmodium falciparum challenge induced in Aotus monkeys by liver-stage antigen-3-derived long synthetic peptides. Eur J Immunol 38: 2610–2615.
    1. Hope IA, Hall R, Simmons DL, Hyde JE, Scaife JG (1984) Evidence for immunological cross-reaction between sporozoites and blood stages of a human malaria parasite. Nature 308: 191–194.
    1. Hill AV, Elvin J, Willis AC, Aidoo M, Allsopp CE, et al. (1992) Molecular analysis of the association of HLA-B53 and resistance to severe malaria. Nature 360: 434–439.
    1. Aidoo M, Lalvani A, Gilbert SC, Hu JT, Daubersies P, et al. (2000) Cytotoxic T-lymphocyte epitopes for HLA-B53 and other HLA types in the malaria vaccine candidate liver-stage antigen 3. Infect Immun 68: 227–232.
    1. Fidock DA, Bottius E, Brahimi K, Moelans, II, Aikawa M, et al. (1994) Cloning and characterization of a novel Plasmodium falciparum sporozoite surface antigen, STARP. Mol Biochem Parasitol 64: 219–232.
    1. Moelans, II, Meis JF, Kocken C, Konings RN, Schoenmakers JG (1991) A novel protein antigen of the malaria parasite Plasmodium falciparum, located on the surface of gametes and sporozoites. Mol Biochem Parasitol 45: 193–204.
    1. Bergmann-Leitner ES, Mease RM, De La Vega P, Savranskaya T, Polhemus M, et al. (2010) Immunization with pre-erythrocytic antigen CelTOS from Plasmodium falciparum elicits cross-species protection against heterologous challenge with Plasmodium berghei. PLoS One 5: e12294.
    1. Caspers P, Etlinger H, Matile H, Pink JR, Stuber D, et al. (1991) A Plasmodium falciparum malaria vaccine candidate which contains epitopes from the circumsporozoite protein and a blood stage antigen, 5.1. Mol Biochem Parasitol 47: 143–150.
    1. Sauzet JP, Perlaza BL, Brahimi K, Daubersies P, Druilhe P (2001) DNA immunization by Plasmodium falciparum liver-stage antigen 3 induces protection against Plasmodium yoelii sporozoite challenge. Infect Immun 69: 1202–1206.
    1. Rennenberg A, Lehmann C, Heitmann A, Witt T, Hansen G, et al. (2010) Exoerythrocytic Plasmodium parasites secrete a cysteine protease inhibitor involved in sporozoite invasion and capable of blocking cell death of host hepatocytes. PLoS Pathog 6: e1000825.
    1. Siau A, Silvie O, Franetich JF, Yalaoui S, Marinach C, et al. (2008) Temperature shift and host cell contact up-regulate sporozoite expression of Plasmodium falciparum genes involved in hepatocyte infection. PLoS Pathog 4: e1000121.
    1. Vignali M, McKinlay A, LaCount DJ, Chettier R, Bell R, et al. (2008) Interaction of an atypical Plasmodium falciparum ETRAMP with human apolipoproteins. Malar J 7: 211.
    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. Simpson JA, Aarons L, Collins WE, Jeffery GM, White NJ (2002) Population dynamics of untreated Plasmodium falciparum malaria within the adult human host during the expansion phase of the infection. Parasitology 124: 247–263.
    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. 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. Roestenberg M, Teirlinck AC, McCall MB, 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.

Source: PubMed

3
Tilaa