Direct venous inoculation of Plasmodium falciparum sporozoites for controlled human malaria infection: a dose-finding trial in two centres

Benjamin Mordmüller, Christian Supan, Kim Lee Sim, Gloria P Gómez-Pérez, Carmen Lucelly Ospina Salazar, Jana Held, Stefanie Bolte, Meral Esen, Serena Tschan, Fanny Joanny, Carlos Lamsfus Calle, Sascha J Z Löhr, Albert Lalremruata, Anusha Gunasekera, Eric R James, Peter F Billingsley, Adam Richman, Sumana Chakravarty, Almudena Legarda, Jose Muñoz, Rosa M Antonijoan, Maria Rosa Ballester, Stephen L Hoffman, Pedro L Alonso, Peter G Kremsner, Benjamin Mordmüller, Christian Supan, Kim Lee Sim, Gloria P Gómez-Pérez, Carmen Lucelly Ospina Salazar, Jana Held, Stefanie Bolte, Meral Esen, Serena Tschan, Fanny Joanny, Carlos Lamsfus Calle, Sascha J Z Löhr, Albert Lalremruata, Anusha Gunasekera, Eric R James, Peter F Billingsley, Adam Richman, Sumana Chakravarty, Almudena Legarda, Jose Muñoz, Rosa M Antonijoan, Maria Rosa Ballester, Stephen L Hoffman, Pedro L Alonso, Peter G Kremsner

Abstract

Background: Controlled human malaria infection (CHMI) accelerates development of anti-malarial interventions. So far, CHMI is done by exposure of volunteers to bites of five mosquitoes carrying Plasmodium falciparum sporozoites (PfSPZ), a technique available in only a few centres worldwide. Mosquito-mediated CHMI is logistically complex, exact PfSPZ dosage is impossible and live mosquito-based interventions are not suitable for further clinical development.

Methods: An open-labelled, randomized, dose-finding study in 18-45 year old, healthy, malaria-naïve volunteers was performed to assess if intravenous (IV) injection of 50 to 3,200 aseptic, purified, cryopreserved PfSPZ is safe and achieves infection kinetics comparable to published data of mosquito-mediated CHMI. An independent study site verified the fully infectious dose using direct venous inoculation of PfSPZ. Parasite kinetics were assessed by thick blood smear microscopy and quantitative real time PCR.

Results: IV inoculation with 50, 200, 800, or 3,200 PfSPZ led to parasitaemia in 1/3, 1/3, 7/9, and 9/9 volunteers, respectively. The geometric mean pre-patent period (GMPPP) was 11.2 days (range 10.5-12.5) in the 3,200 PfSPZ IV group. Subsequently, six volunteers received 3,200 PfSPZ by direct venous inoculation at an independent investigational site. All six developed parasitaemia (GMPPP: 11.4 days, range: 10.4-12.3). Inoculation of PfSPZ was safe. Infection rate and pre-patent period depended on dose, and injection of 3,200 PfSPZ led to a GMPPP similar to CHMI with five PfSPZ-infected mosquitoes. The infectious dose of PfSPZ predicted dosage of radiation-attenuated PfSPZ required for successful vaccination.

Conclusions: IV inoculation of PfSPZ is safe, well tolerated and highly reproducible. It shall further accelerate development of anti-malarial interventions through standardization and facilitation of CHMI. Beyond this, rational dose selection for whole PfSPZ-based immunization and complex study designs are now possible.

Trial registration: ClinicalTrials.gov NCT01624961 and NCT01771848 .

Figures

Figure 1
Figure 1
Trial profile. In Tübingen the IV dose of PfSPZ Challenge IV was increased sequentially in 4 steps from 50 (1), to 200 (2), to 800 (3) and to 3,200 PfSPZ (4). In steps 1 and 2, volunteers were randomly assigned to receive PfSPZ Challenge ID or IV. An independent verification group (3,200 PfSPZ DVI) in Barcelona was added after completion of the 3,200 PfSPZ IV group in Tübingen.
Figure 2
Figure 2
Effect of IV PfSPZ Challenge dose on probability of infection. Observed values are given as grey dots, with size representing weight. Model estimates are represented as the black line and the 95% confidence interval as grey ribbon.
Figure 3
Figure 3
Kaplan-Meier plot of time to infection.
Figure 4
Figure 4
Effect of number of mosquito bites on pre-patent period. Published data are from 44 volunteers bitten by 1–7 infected mosquitoes at one centre (Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands) [26]. Observed values are given as grey dots with size representing weight. A Loess smooth is displayed with the estimate (black line) and 95% confidence interval (grey ribbon).
Figure 5
Figure 5
Parasite quantification by qPCR. Blood was assessed every other day beginning on Day 5 and on the day of microscopically detected parasitaemia if no sampling was scheduled for that day in Tübingen. In Barcelona blood was sampled every day from Days 6 to 9 and twice daily from Day 9 onward. The dotted line indicates the limit of quantification of qPCR (30 parasites per mL).
Figure 6
Figure 6
Adverse event pattern. Adverse event (AE) episodes are given over time from Day 0 through Day 21. Shades of blue represent AE grading from Grade 1 (dark), to Grade 2 (blue), to Grade 3 (light blue). Volunteer IDs with a star indicate those volunteers who did not develop parasitaemia. The time of parasite detection by microscopy is given as the letter ‘M’ in red.

References

    1. WHO . World Malaria Report 2014. Geneva: World Health Organization; 2014.
    1. Mordmüller B, Kremsner PG. Malarial parasites vs. antimalarials: never-ending rumble in the jungle. Curr Mol Med. 2006;6:247–51. doi: 10.2174/156652406776055122.
    1. Sauerwein RW, Roestenberg M, Moorthy VS. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat Rev Immunol. 2011;11:57–64. doi: 10.1038/nri2902.
    1. Fairley H. Chemotherapeutic suppression and prophylaxis in malaria. Trans R Soc Trop Med Hyg. 1945;38:311–55. doi: 10.1016/0035-9203(45)90038-1.
    1. Verhage DF, Telgt DSC, Bousema JT, Hermsen CC, van Gemert GJA, van der Meer JWM, et al. Clinical outcome of experimental human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J Med. 2005;63:52–8.
    1. Rickman LS, Jones TR, Long GW, Paparello S, Schneider I, Paul CF, et al. Plasmodium falciparum-infected Anopheles stephensi inconsistently transmit malaria to humans. Am J Trop Med Hyg. 1990;43:441–5.
    1. Lyke KE, Laurens M, Adams M, Billingsley PF, Richman A, Loyevsky M, et al. Plasmodium falciparum malaria challenge by the bite of aseptic Anopheles stephensi mosquitoes: results of a randomized infectivity trial. PLoS One. 2010;5:e13490. doi: 10.1371/journal.pone.0013490.
    1. Epstein JE, Rao S, Williams F, Freilich D, Luke T, Sedegah M, et al. Safety and clinical outcome of experimental challenge of human volunteers with Plasmodium falciparum-infected mosquitoes: an update. J Infect Dis. 2007;196:145–54. doi: 10.1086/518510.
    1. Laurens MB, Billingsley P, Richman A, Eappen AG, Adams M, Li T, et al. Successful human infection with P. falciparum using three aseptic Anopheles stephensi mosquitoes: a new model for controlled human malaria infection. PLoS One. 2013;8:e68969. doi: 10.1371/journal.pone.0068969.
    1. Roestenberg M, O’Hara GA, Duncan CJA, Epstein JE, Edwards NJ, Scholzen A, et al. Comparison of clinical and parasitological data from controlled human malaria infection trials. PLoS One. 2012;7:e38434. doi: 10.1371/journal.pone.0038434.
    1. Hoffman SL, Goh LML, Luke TC, Schneider I, Le TP, Doolan DL, et al. Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis. 2002;185:1155–64. doi: 10.1086/339409.
    1. Clyde DF, Most H, McCarthy VC, Vanderberg JP. Immunization of man against sporozite-induced falciparum malaria. Am J Med Sci. 1973;266:169–77. doi: 10.1097/00000441-197309000-00002.
    1. Seder RA, Chang L-J, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science. 2013;341:1359–65. doi: 10.1126/science.1241800.
    1. Epstein JE, Tewari K, Lyke KE, Sim BKL, Billingsley PF, Laurens MB, et al. Live attenuated malaria vaccine designed to protect through hepatic CD8+ T cell immunity. Science. 2011;334:475–80. doi: 10.1126/science.1211548.
    1. Sheehy SH, Spencer AJ, Douglas AD, Sim BKL, Longley RJ, Edwards NJ, et al. Optimising controlled human malaria infection studies using cryopreserved P. falciparum parasites administered by needle and syringe. PLoS One. 2013;8:e65960. doi: 10.1371/journal.pone.0065960.
    1. Roestenberg M, Bijker EM, Sim BKL, Billingsley PF, James ER, Bastiaens GJH, 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. Shekalaghe S, Rutaihwa M, Billingsley PF, Chemba M, Daubenberger CA, James ER, et al. Controlled human malaria infection of Tanzanians by intradermal injection of aseptic, purified, cryopreserved Plasmodium falciparum sporozoites. Am J Trop Med Hyg. 2014;91:471–80. doi: 10.4269/ajtmh.14-0119.
    1. Hodgson SH, Juma E, Salim A, Magiri C, Kimani D, Njenga D, et al. Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection. Front Microbiol. 2014;5:686. doi: 10.3389/fmicb.2014.00686.
    1. Gaziano TA, Young CR, Fitzmaurice G, Atwood S, Gaziano JM. Laboratory-based versus non-laboratory-based method for assessment of cardiovascular disease risk: the NHANES I Follow-up Study cohort. Lancet. 2008;371:923–31. doi: 10.1016/S0140-6736(08)60418-3.
    1. Hoffman SL, Billingsley PF, James E, Richman A, Loyevsky M, Li T, et al. Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Hum Vaccin. 2010;6:97–106. doi: 10.4161/hv.6.1.10396.
    1. Planche T, Krishna S, Kombila M, Engel K, Faucher JF, Ngou-Milama E, et al. Comparison of methods for the rapid laboratory assessment of children with malaria. Am J Trop Med Hyg. 2001;65:599–602.
    1. Hermsen CC, Telgt DS, Linders EH, van de 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;118:247–51. doi: 10.1016/S0166-6851(01)00379-6.
    1. Haas CN. Estimation of risk due to low doses of microorganisms: a comparison of alternative methodologies. Am J Epidemiol. 1983;118:573–82.
    1. Bonhoeffer J, Bentsi-Enchill A, Chen RT, Fisher MC, Gold MS, Hartman K, et al. Guidelines for collection, analysis and presentation of vaccine safety data in surveillance systems. Vaccine. 2009;27:2289–97. doi: 10.1016/j.vaccine.2008.11.035.
    1. R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria; 2014. []
    1. Turner L, Wang CW, Lavstsen T, Mwakalinga SB, Sauerwein RW, Hermsen CC, et al. Antibodies against PfEMP1, RIFIN, MSP3 and GLURP are acquired during controlled Plasmodium falciparum malaria infections in naïve volunteers. PLoS One. 2011;6:e29025. doi: 10.1371/journal.pone.0029025.
    1. Richie TL, Charoenvit Y, Wang R, Epstein JE, Hedstrom RC, Kumar S, et al. Clinical trial in healthy malaria-naïve adults to evaluate the safety, tolerability, immunogenicity and efficacy of MuStDO5, a five-gene, sporozoite/hepatic stage Plasmodium falciparum DNA vaccine combined with escalating dose human GM-CSF DNA. Hum Vaccines Immunother. 2012;8:1564–84. doi: 10.4161/hv.22129.
    1. Vanderberg JP, Frevert U. Intravital microscopy demonstrating antibody-mediated immobilisation of Plasmodium berghei sporozoites injected into skin by mosquitoes. Int J Parasitol. 2004;34:991–6. doi: 10.1016/j.ijpara.2004.05.005.
    1. Sinnis P, Zavala F. The skin: where malaria infection and the host immune response begin. Semin Immunopathol. 2012;34:787–92. doi: 10.1007/s00281-012-0345-5.
    1. Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of Plasmodium berghei. Nature. 1967;216:160–2. doi: 10.1038/216160a0.
    1. Choumet V, Attout T, Chartier L, Khun H, Sautereau J, Robbe-Vincent A, et al. Visualizing non infectious and infectious Anopheles gambiae blood feedings in naive and saliva-immunized mice. PLoS One. 2012;7:e50464. doi: 10.1371/journal.pone.0050464.
    1. Rieckmann KH, Carson PE, Beaudoin RL, Cassells JS, Sell KW. Sporozoite induced immunity in man against an Ethiopian strain of Plasmodium falciparum. Trans R Soc Trop Med Hyg. 1974;68:258–9. doi: 10.1016/0035-9203(74)90129-1.
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJF, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361:468–77. doi: 10.1056/NEJMoa0805832.
    1. Bijker EM, Bastiaens GJH, Teirlinck AC, van Gemert G-J, Graumans W, van de 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;110:7862–7. doi: 10.1073/pnas.1220360110.
    1. Safety and protective efficacy of intravenous immunization with cryopreserved Plasmodium falciparum sporozoites under chemoprophylaxis []

Source: PubMed

3
Subscribe