IMRAS-A clinical trial of mosquito-bite immunization with live, radiation-attenuated P. falciparum sporozoites: Impact of immunization parameters on protective efficacy and generation of a repository of immunologic reagents

Bradley Hickey, Nimfa Teneza-Mora, Joanne Lumsden, Sharina Reyes, Martha Sedegah, Lindsey Garver, Michael R Hollingdale, Jo Glenna Banania, Harini Ganeshan, Megan Dowler, Anatalio Reyes, Cindy Tamminga, Alexandra Singer, Alicia Simmons, Maria Belmonte, Arnel Belmonte, Jun Huang, Sandra Inoue, Rachel Velasco, Steve Abot, Carlos S Vasquez, Ivelese Guzman, Mimi Wong, Patrick Twomey, Mariusz Wojnarski, James Moon, Yolanda Alcorta, Santina Maiolatesi, Michele Spring, Silas Davidson, Sidhartha Chaudhury, Eileen Villasante, Thomas L Richie, Judith E Epstein, Bradley Hickey, Nimfa Teneza-Mora, Joanne Lumsden, Sharina Reyes, Martha Sedegah, Lindsey Garver, Michael R Hollingdale, Jo Glenna Banania, Harini Ganeshan, Megan Dowler, Anatalio Reyes, Cindy Tamminga, Alexandra Singer, Alicia Simmons, Maria Belmonte, Arnel Belmonte, Jun Huang, Sandra Inoue, Rachel Velasco, Steve Abot, Carlos S Vasquez, Ivelese Guzman, Mimi Wong, Patrick Twomey, Mariusz Wojnarski, James Moon, Yolanda Alcorta, Santina Maiolatesi, Michele Spring, Silas Davidson, Sidhartha Chaudhury, Eileen Villasante, Thomas L Richie, Judith E Epstein

Abstract

Background: Immunization with radiation-attenuated sporozoites (RAS) by mosquito bite provides >90% sterile protection against Plasmodium falciparum (Pf) malaria in humans. RAS invade hepatocytes but do not replicate. CD8+ T cells recognizing parasite-derived peptides on the surface of infected hepatocytes are likely the primary protective mechanism. We conducted a randomized clinical trial of RAS immunization to assess safety, to achieve 50% vaccine efficacy (VE) against controlled human malaria infection (CHMI), and to generate reagents from protected and non-protected subjects for future identification of protective immune mechanisms and antigens.

Methods: Two cohorts (Cohort 1 and Cohort 2) of healthy, malaria-naïve, non-pregnant adults age 18-50 received five monthly immunizations with infected (true-immunized, n = 21) or non-infected (mock-immunized, n = 5) mosquito bites and underwent homologous CHMI at 3 weeks. Immunization parameters were selected for 50% protection based on prior clinical data. Leukapheresis was done to collect plasma and peripheral blood mononuclear cells.

Results: Adverse event rates were similar in true- and mock-immunized subjects. Two true- and two mock-immunized subjects developed large local reactions likely caused by mosquito salivary gland antigens. In Cohort 1, 11 subjects received 810-1235 infected bites; 6/11 (55%) were protected against CHMI vs. 0/3 mock-immunized and 0/6 infectivity controls (VE 55%). In Cohort 2, 10 subjects received 839-1131 infected bites with a higher first dose and a reduced fifth dose; 9/10 (90%) were protected vs. 0/2 mock-immunized and 0/6 controls (VE 90%). Three/3 (100%) protected subjects administered three booster immunizations were protected against repeat CHMI vs. 0/6 controls (VE 100%). Cohort 2 uniquely showed a significant rise in IFN-γ responses after the third and fifth immunizations and higher antibody responses to CSP.

Conclusions: PfRAS were generally safe and well tolerated. Cohort 2 had a higher first dose, reduced final dose, higher antibody responses to CSP and significant rise of IFN-γ responses after the third and fifth immunizations. Whether any of these factors contributed to increased protection in Cohort 2 requires further investigation. A cryobank of sera and cells from protected and non-protected individuals was generated for future immunological studies and antigen discovery.

Trial registration: ClinicalTrials.gov NCT01994525.

Conflict of interest statement

The authors have read the journal's policy and have the following competing interests: TLR is a full time salaried employee of Sanaria Inc. This affiliation does not alter our adherence to PLOS One Policies on sharing data and materials. There are no patents, products in development or marketed products associated with this research to declare.

Figures

Fig 1. Cohort 1: Flow diagram of…
Fig 1. Cohort 1: Flow diagram of immunized and control subjects.
Twenty-four subjects met all eligibility criteria and 14 were assigned to the true-immunization group, four were assigned to the mock-immunization group and the remaining six subjects were assigned as infectivity controls.
Fig 2. Cohort 2: Flow diagram of…
Fig 2. Cohort 2: Flow diagram of immunized and control subjects.
Twenty-two subjects met all eligibility criteria and 12 were assigned to the true-immunization group, four were assigned to the mock-immunization group and the remaining six subjects were assigned as infectivity controls.
Fig 3. Cumulative numbers of bites in…
Fig 3. Cumulative numbers of bites in Cohort 1 and Cohort 2.
The cumulative numbers of P. falciparum RAS-infected mosquitoes administered at immunizations 1–5 for each protected and non-protected subjects and the average cumulative total for all subjects. Panel A: Cohort 1. Panel B: Cohort 2.
Fig 4. Development of parasitemia in the…
Fig 4. Development of parasitemia in the true- and mock-immunized and infectivity control subjects.
Parasitemia-free survival curves (Kaplan-Meier) for true- and mock-immunized subjects and infectivity controls based on microscopic examination of peripheral blood smears. Panel A: Cohort 1 where 11 true-immunized, 3 mock-immunized and 6 infectivity controls received CHMI. Panel B: Cohort 1 where 10 true-immunized, 2 mock-immunized and 6 infectivity controls received CHMI.
Fig 5. Comparison of total number of…
Fig 5. Comparison of total number of infectious bites and time to CHMI in Cohort 1 and Cohort 2 with the target range of infectious bites.
The total number of infectious bites and days between final immunization and CHMI are compared for Cohort 1 (protected: blue circles; non-protected: open circles) and Cohort 2 (protected: green triangles; non-protected: open triangles). All Cohort 1 and Cohort 2 subjects fell within the target box except one protected subject and one non-protected subject in Cohort 1 that fell just outside the target box. The distribution of subjects in Cohort 1 and Cohort 2 showed no association of protection with total numbers of infectious bites or time (days) before CHMI.
Fig 6. Cohort 1 and Cohort 2:…
Fig 6. Cohort 1 and Cohort 2: FluoroSpot activities to whole sporozoites after each immunization and CHMI.
FluoroSpot IFN-γ, IL2 and IFN-γ+IL2 activities to whole sporozoites were measured pre-immunization (Pre), after each immunization (1, 2, 3, 4, 5) and after CHMI (Post-C); subjects in Cohort 1 only were also measured 7 days after the first immunization (1*). Cohort 1 and Cohort 2 protected and non-protected subjects are shown by indicated symbols. Geometric means are indicated by red bars. Panel A: IFN-γ: Geometric mean activities in both Cohorts significantly rose (p = <0.001) after the first immunization and were higher in Cohort 1 than Cohort 2 after the first and second immunizations (see text). In Cohort 2, activities significantly rose after the fifth immunization. Panel B: IL2: Geometric mean activities in both Cohorts significantly rose (p = <10−3) after the first and second (p = <0.01) immunizations and activities of Cohort 1 were significantly higher than Cohort 2 after the first and second immunizations (see text). In Cohort 2, activities significantly rose after the third and fifth immunizations. Panel C: IFN-γ+IL2: Geometric mean activities in both Cohorts 1 were significantly higher after the first immunization, and activities of Cohort 1 were higher than Cohort 2 after the first and second immunizations (see text). In Cohort 2, activities significantly rose after the third and fifth immunizations.
Fig 7. Cohort 1 and Cohort 2:…
Fig 7. Cohort 1 and Cohort 2: ELISA activities after third and fifth immunizations and CHMI, and IFA activities after fifth immunization and CHMI.
ELISA activities to CSP repeat, CSP full length (FL) and AMA1 of Cohort 1 (blue circles) and Cohort 2 (green triangles) were measured pre-immunization (Pre), 14 days after the third (3) and 22 days after fifth (5) immunizations and 28 days after CHMI (Post-CHMI). IFA activities to sporozoites were measured pre-immunization (Pre), and 22 days after fifth (5) immunizations / pre-CHMI. Protected subjects (closed symbols) and non-protected subjects (open symbols). The Geometric mean (red bar) is shown for all subjects in each Cohort. Panel A: CSP repeat: Geometric mean activities in both Cohorts significantly rose after the third and fifth immunizations (see text). Activities in Cohort 2 were significantly higher than Cohort 1 after the third and fifth immunizations. Panel B: CSP Full Length (FL): activities were similar to those with CSP repeat and activities of Cohort 2 were only significantly higher than Cohort 1 after the third and fifth immunizations. Panel C: AMA1: Geometric mean activities of Cohort 1 and Cohort 2 significantly rose after the third and fifth immunization (see text) but were not different between Cohort 1 and Cohort 2. Panel D IFA: Activities of Cohort 1 and Cohort 2 significantly rose after the fifth immunizations (see text) but were similar in Cohort 1 and Cohort 2.

References

    1. Hoffman SL, Goh LM, 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(8):1155–64. Epub 2002/04/04. JID010922 [pii] 10.1086/339409 .
    1. Weiss WR, Mellouk S, Houghten RA, Sedegah M, Kumar S, Good MF, et al. Cytotoxic T cells recognize a peptide from the circumsporozoite protein on malaria-infected hepatocytes. The Journal of experimental medicine. 1990;171(3):763–73. 10.1084/jem.171.3.763
    1. Olotu A, Fegan G, Wambua J, Nyangweso G, Awuondo KO, Leach A, et al. Four-year efficacy of RTS,S/AS01E and its interaction with malaria exposure. N Engl J Med. 2013;368(12):1111–20. 10.1056/NEJMoa1207564 .
    1. Polhemus ME, Remich SA, Ogutu BR, Waitumbi JN, Otieno L, Apollo S, et al. Evaluation of RTS,S/AS02A and RTS,S/AS01B in adults in a high malaria transmission area. PloS one. 2009;4(7):e6465 10.1371/journal.pone.0006465
    1. Doolan DL, Southwood S, Freilich DA, Sidney J, Graber NL, Shatney L, et al. Identification of Plasmodium falciparum antigens by antigenic analysis of genomic and proteomic data. Proc Natl Acad Sci U S A. 2003;100(17):9952–7. Epub 2003/07/30. 10.1073/pnas.1633254100 [pii]. .
    1. Gruner AC, Mauduit M, Tewari R, Romero JF, Depinay N, Kayibanda M, et al. Sterile protection against malaria is independent of immune responses to the circumsporozoite protein. PloS one. 2007;2(12):e1371 10.1371/journal.pone.0001371
    1. Trieu A, Kayala MA, Burk C, Molina DM, Freilich DA, Richie TL, et al. Sterile protective immunity to malaria is associated with a panel of novel P. falciparum antigens. Molecular & cellular proteomics: MCP. 2011;10(9):M111 007948. Epub 2011/06/02. 10.1074/mcp.M111.007948
    1. Limbach K, Aguiar J, Gowda K, Patterson N, Abot E, Sedegah M, et al. Identification of two new protective pre-erythrocytic malaria vaccine antigen candidates. Malar J. 2011;10:65 Epub 2011/03/18. 10.1186/1475-2875-10-65
    1. Aguiar JC, Bolton J, Wanga J, Sacci JB, Iriko H, Mazeika JK, et al. Discovery of Novel Plasmodium falciparum Pre-Erythrocytic Antigens for Vaccine Development. PloS one. 2015;10(8):e0136109 10.1371/journal.pone.0136109
    1. Kariu T, Ishino T, Yano K, Chinzei Y, Yuda M. CelTOS, a novel malarial protein that mediates transmission to mosquito and vertebrate hosts. Molecular microbiology. 2006;59(5):1369–79. Epub 2006/02/14. 10.1111/j.1365-2958.2005.05024.x .
    1. Bergmann-Leitner ES, Mease RM, De La Vega P, Savranskaya T, Polhemus M, Ockenhouse C, et al. Immunization with pre-erythrocytic antigen CelTOS from Plasmodium falciparum elicits cross-species protection against heterologous challenge with Plasmodium berghei. PloS one. 2010;5(8):e12294 10.1371/journal.pone.0012294
    1. Hickey BW, Lumsden JM, Reyes S, Sedegah M, Hollingdale MR, Freilich DA, et al. Mosquito bite immunization with radiation-attenuated Plasmodium falciparum sporozoites: safety, tolerability, protective efficacy and humoral immunogenicity. Malar J. 2016;15(1):377 10.1186/s12936-016-1435-y
    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(9616):923–31. Epub 2008/03/18. S0140-6736(08)60418-3 [pii]. 10.1016/S0140-6736(08)60418-3 .
    1. Egan JE, Hoffman SL, Haynes JD, Sadoff JC, Schneider I, Grau GE, et al. Humoral immune responses in volunteers immunized with irradiated Plasmodium falciparum sporozoites. Am J Trop Med Hyg. 1993;49(2):166–73. Epub 1993/08/01. 10.4269/ajtmh.1993.49.166 .
    1. Herrington D, Davis J, Nardin E, Beier M, Cortese J, Eddy H, et al. Successful immunization of humans with irradiated malaria sporozoites: humoral and cellular responses of the protected individuals. Am J Trop Med Hyg. 1991;45(5):539–47. 10.4269/ajtmh.1991.45.539 .
    1. Chulay JD, Schneider I, Cosgriff TM, Hoffman SL, Ballou WR, Quakyi IA, et al. Malaria transmitted to humans by mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg. 1986;35(1):66–8. Epub 1986/01/01. 10.4269/ajtmh.1986.35.66 .
    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. Epub 2013/08/10. 10.1126/science.1241800 .
    1. Sedegah M, Hollingdale MR, Farooq F, Ganeshan H, Belmonte M, Kim Y, et al. Sterile Immunity to Malaria after DNA Prime/Adenovirus Boost Immunization Is Associated with Effector Memory CD8+T Cells Targeting AMA1 Class I Epitopes. PloS one. 2014;9(9):e106241 10.1371/journal.pone.0106241 .
    1. Walliker D, Quakyi IA, Wellems TE, McCutchan TF, Szarfman A, London WT, et al. Genetic analysis of the human malaria parasite Plasmodium falciparum. Science. 1987;236(4809):1661–6. 10.1126/science.3299700 .
    1. Wirtz RA, Zavala F, Charoenvit Y, Campbell GH, Burkot TR, Schneider I, et al. Comparative testing of monoclonal antibodies against Plasmodium falciparum sporozoites for ELISA development. Bull World Health Organ. 1987;65(1):39–45. .
    1. Schwenk R, DeBot M, Porter M, Nikki J, Rein L, Spaccapelo R, et al. IgG2 antibodies against a clinical grade Plasmodium falciparum CSP vaccine antigen associate with protection against transgenic sporozoite challenge in mice. PloS one. 2014;9(10):e111020 10.1371/journal.pone.0111020
    1. Hickey BW, Lumsden JM, Reyes S, Elbeshbishi Y, Mix A, Spurgeon J, et al. A Retrospective Analysis of the Adverse Event Data from the Phase 1 trial "Plasmodium Sp. Sporozoites Immunization of Human Volunteers. Am Soc Trop Med Hyg Annual Mtg 2013. 2013:Abstract 1161.
    1. Spring M, Murphy J, Nielsen R, Dowler M, Bennett JW, Zarling S, et al. First-in-human evaluation of genetically attenuated Plasmodium falciparum sporozoites administered by bite of Anopheles mosquitoes to adult volunteers. Vaccine. 2013;31(43):4975–83. 10.1016/j.vaccine.2013.08.007 .
    1. Churcher TS, Sinden RE, Edwards NJ, Poulton ID, Rampling TW, Brock PM, et al. Probability of Transmission of Malaria from Mosquito to Human Is Regulated by Mosquito Parasite Density in Naive and Vaccinated Hosts. PLoS Pathog. 2017;13(1):e1006108 10.1371/journal.ppat.1006108 .
    1. Ishizuka AS, Lyke KE, DeZure A, Berry AA, Richie TL, Mendoza FH, et al. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat Med. 2016;22(6):614–23. 10.1038/nm.4110 .
    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 Epub 2017/01/18. 10.1172/jci.insight.89154 [pii]. .
    1. Cardoso FC, Roddick JS, Groves P, Doolan DL. Evaluation of approaches to identify the targets of cellular immunity on a proteome-wide scale. PloS one. 2011;6(11):e27666 Epub 2011/11/19. 10.1371/journal.pone.0027666
    1. Aguiar JC, Bolton J, Wanga J, Sacci JB, Hideyuki I, Mazeika JK, et al. Discovery of novel Plasmodium falciparum pre-erythrocytic antigens for vaccine development. PloS one. 2015;10(8):e0136109 10.1371/journal.pone.0136109
    1. Doolan DL, Mu Y, Unal B, Sundaresh S, Hirst S, Valdez C, et al. Profiling humoral immune responses to P. falciparum infection with protein microarrays. Proteomics. 2008;8(22):4680–94. Epub 2008/10/22. 10.1002/pmic.200800194
    1. Sundaresh S, Doolan DL, Hirst S, Mu Y, Unal B, Davies DH, et al. Identification of humoral immune responses in protein microarrays using DNA microarray data analysis techniques. Bioinformatics. 2006;22(14):1760–6. Epub April 27, 2006. 10.1093/bioinformatics/btl162
    1. Donovan MJ, Messmore AS, Scrafford DA, Sacks DL, Kamhawi S, McDowell MA. Uninfected mosquito bites confer protection against infection with malaria parasites. Infect Immun. 2007;75(5):2523–30. 10.1128/IAI.01928-06
    1. Depinay N, Hacini F, Beghdadi W, Peronet R, Mecheri S. Mast cell-dependent down-regulation of antigen-specific immune responses by mosquito bites. J Immunol. 2006;176(7):4141–6. 10.4049/jimmunol.176.7.4141 .
    1. da Silva HB, Caetano SS, Monteiro I, Gomez-Conde I, Hanson K, Penha-Goncalves C, et al. Early skin immunological disturbance after Plasmodium-infected mosquito bites. Cellular immunology. 2012;277(1–2):22–32. 10.1016/j.cellimm.2012.06.003 .
    1. Kebaier C, Voza T, Vanderberg J. Kinetics of mosquito-injected Plasmodium sporozoites in mice: fewer sporozoites are injected into sporozoite-immunized mice. PLoS Pathog. 2009;5(4):e1000399 10.1371/journal.ppat.1000399
    1. Hopp CS, Sinnis P. The innate and adaptive response to mosquito saliva and Plasmodium sporozoites in the skin. Annals of the New York Academy of Sciences. 2015;1342:37–43. 10.1111/nyas.12661
    1. Resik S, Tejeda A, Sutter RW, Diaz M, Sarmiento L, Alemani N, et al. Priming after a fractional dose of inactivated poliovirus vaccine. N Engl J Med. 2013;368(5):416–24. 10.1056/NEJMoa1202541 .
    1. Vanderberg JP. Plasmodium berghei: quantitation of sporozoites injected by mosquitoes feeding on a rodent host. Exp Parasitol. 1977;42(1):169–81. Epub 1977/06/01. 10.1016/0014-4894(77)90075-3 .
    1. Vanderberg JP. Imaging mosquito transmission of Plasmodium sporozoites into the mammalian host: immunological implications. Parasitol Int. 2014;63(1):150–64. 10.1016/j.parint.2013.09.010 .
    1. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, Desmons P, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med. 1997;336(2):86–91. Epub 1997/01/09. 10.1056/NEJM199701093360202 .
    1. Stoute JA, Kester KE, Krzych U, Wellde BT, Hall T, White K, et al. Long-term efficacy and immune responses following immunization with the RTS,S malaria vaccine. J Infect Dis. 1998;178(4):1139–44. 10.1086/515657 .
    1. Regules JA, Cicatelli SB, Bennett JW, Paolino KM, Twomey PS, Moon JE, et al. Fractional third and fourth dose of RTS,S/AS01 malaria candidate vaccine: a phase 2a controlled human malaria infection and immunogenicity study. J Infect Dis. 2016. 10.1093/infdis/jiw237 .
    1. Medica DL, Sinnis P. Quantitative dynamics of Plasmodium yoelii sporozoite transmission by infected anopheline mosquitoes. Infect Immun. 2005;73(7):4363–9. 10.1128/IAI.73.7.4363-4369.2005
    1. Beier JC, Davis JR, Vaughan JA, Noden BH, Beier MS. Quantitation of Plasmodium falciparum sporozoites transmitted in vitro by experimentally infected Anopheles gambiae and Anopheles stephensi. Am J Trop Med Hyg. 1991;44(5):564–70. 10.4269/ajtmh.1991.44.564 .
    1. Ponnudurai T, Lensen AH, van Gemert GJ, Bolmer MG, Meuwissen JH. Feeding behaviour and sporozoite ejection by infected Anopheles stephensi. Trans R Soc Trop Med Hyg. 1991;85(2):175–80. 10.1016/0035-9203(91)90012-n .
    1. Rosenberg R, Wirtz RA, Schneider I, Burge R. An estimation of the number of malaria sporozoites ejected by a feeding mosquito. Trans R Soc Trop Med Hyg. 1990;84(2):209–12. 10.1016/0035-9203(90)90258-g .
    1. Jin Y, Kebaier C, Vanderberg J. Direct microscopic quantification of dynamics of Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infect Immun. 2007;75(11):5532–9. 10.1128/IAI.00600-07
    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(1):97–106. 10.4161/hv.6.1.10396 .
    1. Lyke KE, Ishizuka AS, Berry AA, Chakravarty S, DeZure A, Enama ME, et al. Attenuated PfSPZ Vaccine induces strain-transcending T cells and durable protection against heterologous controlled human malaria infection. Proc Natl Acad Sci U S A. 2017;114(10):2711–6. 1615324114 [pii]; 10.1073/pnas.1615324114
    1. Sissoko M, Healy SA, Duffy PE. Safety and efficacy of PfSPZ Vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial. The Lancet Infectious diseases. 2017;17(5):498–509. 10.1016/S1473-3099(17)30104-4
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361(5):468–77. Epub 2009/07/31. 361/5/468 [pii]. 10.1056/NEJMoa0805832 .
    1. Mordmuller 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):445–9. Epub 2017/02/16. 10.1038/nature21060 .
    1. Walk J, Stok JE, Sauerwein RW. Can Patrolling Liver-Resident T Cells Control Human Malaria Parasite Development? Trends in immunology. 2019;40(3):186–96. Epub 2019/02/05. 10.1016/j.it.2019.01.002 .
    1. Gbedande K, Stephens R. Initiating the T Cell Response to Liver-Stage Malaria. Trends Parasitol. 2019;35(7):489–90. Epub 2019/05/28. 10.1016/j.pt.2019.04.011 .
    1. Kurup SP, Anthony SM, Hancox LS, Vijay R, Pewe LL, Moioffer SJ, et al. Monocyte-Derived CD11c(+) Cells Acquire Plasmodium from Hepatocytes to Prime CD8 T Cell Immunity to Liver-Stage Malaria. Cell Host Microbe. 2019;25(4):565–77 e6. Epub 2019/03/25. 10.1016/j.chom.2019.02.014

Source: PubMed

3
Se inscrever