Outcomes of controlled human malaria infection after BCG vaccination

Jona Walk, L Charlotte J de Bree, Wouter Graumans, Rianne Stoter, Geert-Jan van Gemert, Marga van de Vegte-Bolmer, Karina Teelen, Cornelus C Hermsen, Rob J W Arts, Marije C Behet, Farid Keramati, Simone J C F M Moorlag, Annie S P Yang, Reinout van Crevel, Peter Aaby, Quirijn de Mast, André J A M van der Ven, Christine Stabell Benn, Mihai G Netea, Robert W Sauerwein, Jona Walk, L Charlotte J de Bree, Wouter Graumans, Rianne Stoter, Geert-Jan van Gemert, Marga van de Vegte-Bolmer, Karina Teelen, Cornelus C Hermsen, Rob J W Arts, Marije C Behet, Farid Keramati, Simone J C F M Moorlag, Annie S P Yang, Reinout van Crevel, Peter Aaby, Quirijn de Mast, André J A M van der Ven, Christine Stabell Benn, Mihai G Netea, Robert W Sauerwein

Abstract

Recent evidence suggests that certain vaccines, including Bacillus-Calmette Guérin (BCG), can induce changes in the innate immune system with non-specific memory characteristics, termed 'trained immunity'. Here we present the results of a randomised, controlled phase 1 clinical trial in 20 healthy male and female volunteers to evaluate the induction of immunity and protective efficacy of the anti-tuberculosis BCG vaccine against a controlled human malaria infection. After malaria challenge infection, BCG vaccinated volunteers present with earlier and more severe clinical adverse events, and have significantly earlier expression of NK cell activation markers and a trend towards earlier phenotypic monocyte activation. Furthermore, parasitemia in BCG vaccinated volunteers is inversely correlated with increased phenotypic NK cell and monocyte activation. The combined data demonstrate that BCG vaccination alters the clinical and immunological response to malaria, and form an impetus to further explore its potential in strategies for clinical malaria vaccine development.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Parasitemia, clinical symptoms and laboratory abnormalities after CHMI. Parasitemia was measured by daily qPCR from day 6 after CHMI until the third day after antimalarial treatment. a The Kaplan–Meier survival curve shows percent of volunteers remaining untreated. 8/9 BCG vaccinated (green) and 10/10 control volunteers (grey) surpassed the treatment threshold of 100 parasites per millilitre, and were treated on day 7 after challenge. 1/9 BCG vaccinated volunteers remained below 100 Pf/mL until day 9. b All volunteers did have parasitemia detectable by qPCR on day 7 after CHMI. The graph shows log parasites per millilitre on day 7 post CHMI for BCG vaccinated (green) and control (grey) volunteers. c Adverse events were collected daily. The Kaplan–Meier curve shows the percentage of volunteers experiencing one or more moderate or severe, solicited, symptoms during follow-up, BCG vaccinated volunteers (green) compared to controls (grey). df Absolute platelet, lymphocyte and neutrophil differentiation counts were determined by daily hemocytometry starting on day 6 post-challenge. Graphs show relative change in cell counts compared to pre-challenge values in both BCG vaccinated (n = 9, each coloured dot shows and individual volunteer, colours consistently represent the same volunteers across each graph) and non-BCG vaccinated controls (n = 10, grey dots)
Fig. 2
Fig. 2
In vivo activation of lymphocytes, monocytes and neutrophils after CHMI. In vivo leucocyte activation was determined by direct staining of fresh whole blood with fluorescent antibodies every 2 days post-challenge. Lymphocytes were defined based on forward scatter and sideward scatter characteristics, and duplet events were excluded. a NK cell activation was defined as the percentage of CD3-CD56dimCD16+ live cells expressing CD69. b γδT cell activation was defined as the percentage of CD3+γδTCR+ live cells expressing CD69. c NKT cell activation was defined as the percentage of CD3+γδTCR-CD56+ live cells expressing CD69. d αβT cell activation was defined the as percentage of CD3+γδTCR-CD56− live cells expressing CD69. e Monocytes were defined based on forward and side scatter characteristics, and the as HLA-DR+CD14+. Within the monocyte population, cells were then divided into CD16- and CD16+ monocytes. fg Within the CD16- monocyte population, the relative change in mean fluorescent intensity of HLA-DR and CD86 compared to pre-malaria challenge values was determined. h Neutrophils were defined based on forward and side scatter characteristics, and then defined as HLA-DR-CD14-CD16+CD11b+. Activated neutrophils were defined as CD62LdimCD11bhigh. ij IFN-γ and granzyme B were measured by Luminex assay in citrate plasma taken ever 2 days. Circulating cytokine levels are corrected for baseline levels (pre-BCG vaccination time point) at each time point. In all graphs the grey dots represent non-BCG vaccinated control group volunteers (n = 10), and each coloured dot shows an individual BCG vaccinated volunteer (n = 9). Statistical analysis are between BCG vaccinated and control volunteers at a single time point, and p-values are the results of Mann–Whitney U-test. *p < 0.05. k Circulating CRP levels were measured in citrate plasma are shown for each BCG vaccinated volunteer (colours consistently represent the same volunteers across each graph)
Fig. 3
Fig. 3
Early NK cell and monocyte activation correlates with decreased parasitemia. Correlations between a NK cell CD69 expression and log parasitemia on day 7 after challenge infection, and b increase in monocyte HLA-DR MFI and log parasitemia on day 7 are shown for BCG vaccinated (n = 9, green) and control (n = 10, grey) volunteers. Lines show the result of linear regression analysis for both groups
Fig. 4
Fig. 4
BCG vaccination increases NK cell responses against P. falciparum. a Percentage of total NK cells staining positive for IFN-γ after 24 h of stimulation with PfRBC before, and 37 days after malaria challenge infection as compared to baseline (pre-BCG vaccination time point), each dot represents an individual BCG vaccinated (n = 9, green) or control volunteers (n = 10, grey). b Percent NK cells staining positive for the degranulation marker CD107a. c Percent NK cells staining positive for granzyme B. Lines and error bars show median and interquartile range. p-values are the result of Mann–Whitney U-test. *p < 0.05. Stimulation was performed in duplo, with replicates combined for flow cytometry analysis

References

    1. WHO. World Malaria Report 2015. (World Health Organization, Geneva, 2015).
    1. Olotu A, et al. Seven-year efficacy of RTS,S/AS01 malaria vaccine among young african children. N. Engl. J. Med. 2016;374:2519–2529. doi: 10.1056/NEJMoa1515257.
    1. Netea MG, Latz E, Mills KH, O’Neill LA. Innate immune memory: a paradigm shift in understanding host defense. Nat. Immunol. 2015;16:675–679. doi: 10.1038/ni.3178.
    1. Kleinnijenhuis J, et al. Long-lasting effects of BCG vaccination on both heterologous Th1/Th17 responses and innate trained immunity. J. Innate Immun. 2014;6:152–158. doi: 10.1159/000355628.
    1. Kleinnijenhuis J, et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc. Natl Acad. Sci. U.S.A. 2012;109:17537–17542. doi: 10.1073/pnas.1202870109.
    1. Cheng SC, et al. mTOR− and HIF-1alpha-mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345:1250684. doi: 10.1126/science.1250684.
    1. Saeed S, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science. 2014;345:1251086. doi: 10.1126/science.1251086.
    1. Quintin J, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe. 2012;12:223–232. doi: 10.1016/j.chom.2012.06.006.
    1. Clark IA, Allison AC, Cox FE. Protection of mice against Babesia and Plasmodium with BCG. Nature. 1976;259:309–311. doi: 10.1038/259309a0.
    1. Matsumoto S, et al. Mycobacterium bovis bacillus calmette-guerin induces protective immunity against infection by Plasmodium yoelii at blood-stage depending on shifting immunity toward Th1 type and inducing protective IgG2a after the parasite infection. Vaccine. 2000;19:779–787. doi: 10.1016/S0264-410X(00)00257-7.
    1. Murphy JR. Host defenses in murine malaria: nonspecific resistance to Plasmodium berghei generated in response to Mycobacterium bovis infection or Corynebacterium parvum stimulation. Infect. Immun. 1981;33:199–211.
    1. Parra M, et al. Molecular analysis of non-specific protection against murine malaria induced by BCG vaccination. PLoS ONE. 2013;8:e66115. doi: 10.1371/journal.pone.0066115.
    1. Roth A, et al. BCG vaccination scar associated with better childhood survival in Guinea-Bissau. Int. J. Epidemiol. 2005;34:540–547. doi: 10.1093/ije/dyh392.
    1. Teirlinck AC, et al. Plasmodium falciparum infection of human volunteers activates monocytes and CD16 + dendritic cells and induces upregulation of CD16 and CD1c expression. Infect. Immun. 2015;83:3732–3739. doi: 10.1128/IAI.00473-15.
    1. Kleinnijenhuis J, et al. BCG-induced trained immunity in NK cells: Role for non-specific protection to infection. Clin. Immunol. 2014;155:213–219. doi: 10.1016/j.clim.2014.10.005.
    1. Arts RJW, et al. BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe. 2018;23:89–100 e105. doi: 10.1016/j.chom.2017.12.010.
    1. Harpaz R, et al. Serum cytokine profiles in experimental human malaria. Relationship to protection and disease course after challenge. J. Clin. Investig. 1992;90:515–523. doi: 10.1172/JCI115889.
    1. Hermsen CC, et al. Circulating concentrations of soluble granzyme A and B increase during natural and experimental Plasmodium falciparum infections. Clin. Exp. Immunol. 2003;132:467–472. doi: 10.1046/j.1365-2249.2003.02160.x.
    1. Walther M, et al. Innate immune responses to human malaria: heterogeneous cytokine responses to blood-stage Plasmodium falciparum correlate with parasitological and clinical outcomes. J. Immunol. 2006;177:5736–5745. doi: 10.4049/jimmunol.177.8.5736.
    1. Church LW, et al. Clinical manifestations of Plasmodium falciparum malaria experimentally induced by mosquito challenge. J. Infect. Dis. 1997;175:915–920. doi: 10.1086/513990.
    1. Roestenberg M, 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. Walk J, et al. Diagnosis and treatment based on quantitative PCR after controlled human malaria infection. Malar. J. 2016;15:398. doi: 10.1186/s12936-016-1434-z.
    1. Liehl P, et al. Host-cell sensors for Plasmodium activate innate immunity against liver-stage infection. Nat. Med. 2014;20:47–53. doi: 10.1038/nm.3424.
    1. Miller JL, Sack BK, Baldwin M, Vaughan AM, Kappe SH. Interferon-mediated innate immune responses against malaria parasite liver stages. Cell Rep. 2014;7:436–447. doi: 10.1016/j.celrep.2014.03.018.
    1. McCall MB, et al. Memory-like IFN-gamma response by NK cells following malaria infection reveals the crucial role of T cells in NK cell activation by P. falciparum. Eur. J. Immunol. 2010;40:3472–3477. doi: 10.1002/eji.201040587.
    1. Leentjens J, et al. BCG vaccination enhances the immunogenicity of subsequent influenza vaccination in healthy volunteers: A randomized, placebo-controlled pilot study. J. Infect. Dis. 2015;212:1930–1938. doi: 10.1093/infdis/jiv332.
    1. Artavanis-Tsakonas K, et al. Activation of a subset of human NK cells upon contact with Plasmodium falciparum-infected erythrocytes. J. Immunol. 2003;171:5396–5405. doi: 10.4049/jimmunol.171.10.5396.
    1. Buffen K, et al. Autophagy controls BCG-induced trained immunity and the response to intravesical BCG therapy for bladder cancer. PLoS Pathog. 2014;10:e1004485. doi: 10.1371/journal.ppat.1004485.
    1. Roth A, Garly ML, Jensen H, Nielsen J, Aaby P. Bacillus Calmette-Guerin vaccination and infant mortality. Expert. Rev. Vaccin. 2006;5:277–293. doi: 10.1586/14760584.5.2.277.
    1. Aaby P, et al. Randomized trial of BCG vaccination at birth to low-birth-weight children: beneficial nonspecific effects in the neonatal period? J. Infect. Dis. 2011;204:245–252. doi: 10.1093/infdis/jir240.
    1. Biering-Sorensen S, et al. Early BCG-denmark and neonatal mortality among infants weighing < 2500 g: A randomized controlled trial. Clin. Infect. Dis. 2017;65:1183–1190. doi: 10.1093/cid/cix525.
    1. de Bree, L. C. J., et al. Non-specific effects of vaccines: Current evidence and potential implications. Semin. Immunol.39, 35–43 (2018).
    1. Netea MG, van Crevel R. BCG-induced protection: effects on innate immune memory. Semin. Immunol. 2014;26:512–517. doi: 10.1016/j.smim.2014.09.006.
    1. Benn CS, Fisker AB, Whittle HC, Aaby P. Revaccination with live attenuated vaccines confer additional beneficial nonspecific effects on overall survival: A review. EBioMedicine. 2016;10:312–317. doi: 10.1016/j.ebiom.2016.07.016.
    1. Rodrigues A, et al. Revaccination with Bacillus Calmette-Guerin (BCG) vaccine does not reduce morbidity from malaria in African children. Trop. Med. Int. Health.: TM & IH. 2007;12:224–229.
    1. Roth AE, et al. Effect of revaccination with BCG in early childhood on mortality: randomised trial in Guinea-Bissau. BMJ. 2010;340:c671. doi: 10.1136/bmj.c671.
    1. Bijker EM, 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;110:7862–7867. doi: 10.1073/pnas.1220360110.
    1. Hermsen CC, et al. Detection of Plasmodium falciparum malaria parasites in vivo by real-time quantitative PCR. Mol. Biochem. Parasitol. 2001;118:247–251. doi: 10.1016/S0166-6851(01)00379-6.
    1. Schats R, et al. Heterologous protection against malaria after immunization with plasmodium falciparum sporozoites. PLoS ONE. 2015;10:e0124243. doi: 10.1371/journal.pone.0124243.
    1. Roestenberg M, et al. Protection against a malaria challenge by sporozoite inoculation. N. Engl. J. Med. 2009;361:468–477. doi: 10.1056/NEJMoa0805832.
    1. Nahrendorf W, et al. Memory B-cell and antibody responses induced by Plasmodium falciparum sporozoite immunization. J. Infect. Dis. 2014;210:1981–1990. doi: 10.1093/infdis/jiu354.
    1. Verhave JP, Leeuwenberg AD, Ponnudurai T, Meuwissen JH, van Druten JA. The biotin-streptavidin system in a two-site ELISA for the detection of plasmodial sporozoite antigen in mosquitoes. Parasite Immunol. 1988;10:17–31. doi: 10.1111/j.1365-3024.1988.tb00200.x.

Source: PubMed

3
Abonnieren