Plasmodium falciparum clearance rates in response to artesunate in Malian children with malaria: effect of acquired immunity

Tatiana M Lopera-Mesa, Saibou Doumbia, Serena Chiang, Amir E Zeituni, Drissa S Konate, Mory Doumbouya, Abdoul S Keita, Kasia Stepniewska, Karim Traore, Seidina A S Diakite, Daouda Ndiaye, Juliana M Sa, Jennifer M Anderson, Michael P Fay, Carole A Long, Mahamadou Diakite, Rick M Fairhurst, Tatiana M Lopera-Mesa, Saibou Doumbia, Serena Chiang, Amir E Zeituni, Drissa S Konate, Mory Doumbouya, Abdoul S Keita, Kasia Stepniewska, Karim Traore, Seidina A S Diakite, Daouda Ndiaye, Juliana M Sa, Jennifer M Anderson, Michael P Fay, Carole A Long, Mahamadou Diakite, Rick M Fairhurst

Abstract

Background: Artemisinin resistance, a long parasite clearance half-life in response to artemisinin, has been described in patients with Plasmodium falciparum malaria in southeast Asia. Few baseline half-lives have been reported from Africa, where artemisinins were recently introduced.

Methods: We treated P. falciparum malaria in 215 Malian children aged 0.5-15 years with artesunate (0, 24, 48 hours) and amodiaquine (72, 96, 120 hours). We estimated half-life by measuring parasite density every 6 hours until undetectable and evaluated the effects of age, sex, ethnicity, and red blood cell (RBC) polymorphisms on half-life. We quantified the proportion of parasitized RBCs recognized by autologous immunoglobulin G (IgG).

Results: The geometric mean half-life was 1.9 hours (95% confidence interval, 1.8-2.0) and did not correlate with parasite ex vivo susceptibility to artemisinins. In a linear model accounting for host factors, half-life decreased by 4.1 minutes for every 1-year increase in age. The proportion of parasitized RBCs recognized by IgG correlated inversely with half-life (r = -0.475; P = .0006).

Conclusions: Parasite clearance in response to artesunate is faster in Mali than in southeast Asia. IgG responses to parasitized RBCs shorten half-life and may influence this parameter in areas where age is not an adequate surrogate of immunity and correlates of parasite-clearing immunity have not been identified.

Clinical trials registration: NCT00669084.

Figures

Figure 1.
Figure 1.
Clearance of ring-stage Plasmodium falciparum parasites from peripheral blood during a parasite clearance rate study. Dihydroartemisinin, the active metabolite of all artemisinins, causes ring-stage parasites to undergo pyknosis (A). These circulating pyknotic forms are eventually “pitted” from red blood cells (RBCs) as they pass through endothelial slits in the spleen, which returns the previously infected, intact RBCs to the peripheral blood. This process occurs in all patients treated with artesunate and is likely the predominant mechanism of parasite clearance in most cases. Depending on the maturity of ring-stage parasites, however, 2 additional processes may also work to reduce parasite density in an artemisinin-independent manner. Ring-stage parasites that are sufficiently mature (indicated by a thicker ring of light blue cytoplasm in the figure) may place PfEMP1-laden knobs on the surface of their host RBCs at the same time artesunate is consumed and begins to kill parasites (B and C). If these PfEMP1-expressing parasitized RBCs are traveling through a microvessel at this time, they may successfully sequester there and disappear from the peripheral blood (C). On the other hand, if these parasitized RBCs are traveling through other blood vessels where sequestration does not occur, they become targets for antibodies against PfEMP1 and other surface antigens (B). By preventing parasitized RBCs from sequestering and opsonizing them for phagocytosis in the spleen, these antibodies may contribute to the clearance of ring-stage parasites. Abbreviation: DHA, dihydroartemisinin.
Figure 2.
Figure 2.
In vivo and ex vivo responses of parasites to artemisinin derivatives. A, Distribution of parasite clearance half-lives in 215 children with uncomplicated Plasmodium falciparum malaria in Kenieroba, Mali, compared with that of 168 (mostly adult) patients in Pursat, western Cambodia. The geometric mean half-life in Pursat (5.9 hours) is 3.0 times longer (95% confidence interval [CI], 2.8–3.3; P < .0001) than in Kenieroba (1.9 hours). The geometric mean parasite clearance half-life 95% CIs are indicated by horizontal lines and whiskers. B, Ex vivo responses of P. falciparum isolates to antimalarial drugs. The geometric mean (GM) IC50 values for chloroquine (CQ; n = 87), amodiaquine (AQ; n = 41), monodesethylamodiaquine (MDAQ; n = 57), quinine (QN; n = 27), artesunate (AS; n = 96), and dihydroartemisinin (DHA; n = 87) were 119 nM, 7.79 nM, 16.7 nM, 49.9 nM, 1.17 nM, and 1.50 nM, respectively. The GM IC50 and 95% CIs are indicated by horizontal lines and whiskers. This assay showed that approximately 50% of parasite isolates have reduced susceptibility to CQ (IC50 > 100 nM) but remain susceptible to the other antimalarial drugs tested. These data are consistent with the only other in vitro responses of parasite isolates reported from Mali [36]. The IC50 values for CQ correlated with those for AQ (r = 0.608; P = .0008) and MDAQ (r = 0.707; P < .0001). The only other IC50 correlation among the 6 drugs was that observed for QN and AS (r = 0.585; P = .002). Abbreviations: AQ, amodiaquine; AS, artesunate; CQ, chloroquine; DHA, dihydroartemisinin; IC50, median inhibitory concentration; MDAQ, monodesethylamodiaquine; QN, quinine.
Figure 3.
Figure 3.
Influence of host factors on parasite clearance half-life. A, Results from a linear model in which the response is half-life in hours and the independent variables are age, sex (female reference), ethnicity (non-Fulani reference), hemoglobin (Hb) AC, HbAS, G6PD deficiency, α-thalassemia, and loge-transformed initial parasite density. Each effect is given as the predicted change in half-life while holding all other variables constant. Hemoglobinopathy effects compare heterozygous with wild type (with homozygous and hemizygous having double effects), whereas age and ln (Pf day 0) are compared with adding 1 unit to that variable. Without correcting for multiple comparisons, we see that only the age effect is significant. The age parameter states that for every 1-year increase in age, the half-life changes by −0.0686 hours (95% confidence interval [CI], −.0946 to −.0425). The parameters with 95% CIs (uncorrected for multiple comparisons) are plotted. B, Half-life is inversely correlated with age (r = −0.312; P < .0001), a surrogate of naturally acquired immunity in our study population. The linear model estimates that there is a 4.1-minute (95% CI, 2.6–5.7; P < .0001) shortening of half-life for every 1-year increase in age. Abbreviations: Hb, hemoglobin; HE, heterozygote; Pf day 0, initial P. falciparum density.
Figure 4.
Figure 4.
Autologous immunoglobulin G (IgG) responses against the surface of Plasmodium falciparum–infected red blood cells (RBCs) obtained directly from Malian children with malaria and cultured to the trophozoite-stage. A, Correlation between screening and initial parasite densities. Although screening and initial parasite densities correlated (r = 0.736; P < .0001), the parasite densities of many children dropped before the first artesunate dose was taken (data points below the line of equality). This finding may be due to the often-synchronous sequestration of late ring-stage parasitized RBCs in microvessels [26] and opsonic removal of parasitized RBCs, both of which may decrease half-life. B, Representative flow cytometry scatterplot showing that 20.6% of parasitized RBCs in this child were recognized by autologous IgG. C, The median proportions of IgG-positive parasitized RBCs were 21.0% (interquartile range [IQR], 11.7–37.2), 10.8% (IQR, 6.92–23.7), and 5.94% (IQR, 4.04–17.6) for plasma tested at 1:10 (n = 48), 1:20 (n = 48), and 1:40 (n = 22) dilutions, respectively. For each dilution doubling, the proportion of IgG-positive parasitized RBCs changed on average by a factor of 0.61 (95% CI, .54–.69; P < .0001). D, The plot shows an inverse correlation between IgG responses and half-life (r = −0.475; P = .0006), suggesting they are involved in clearing ring-stage parasites from peripheral blood. Abbreviations: IgG, immunoglobulin; FL1-H, fluorescence channel 1-height; FL2-H, fluorescence channel 2-height.

References

    1. Jiang JB, Li GQ, Guo XB, Kong YC, Arnold K. Antimalarial activity of mefloquine and qinghaosu. Lancet. 1982;2:285–8.
    1. Dondorp A, Nosten F, Stepniewska K, Day N, White N. Artesunate versus quinine for treatment of severe falciparum malaria: a randomised trial. Lancet. 2005;366:717–25.
    1. Dondorp AM, Fanello CI, Hendriksen IC, et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010;376:1647–57.
    1. World Health Organization. Guidelines for the treatment of malaria. 2nd ed. Geneva, Switzerland: World Health Organization; 2010.
    1. Nosten F, White NJ. Artemisinin-based combination treatment of falciparum malaria. Am J Trop Med Hyg. 2007;77:181–92.
    1. Eastman RT, Fidock DA. Artemisinin-based combination therapies: a vital tool in efforts to eliminate malaria. Nat Rev Microbiol. 2009;7:864–74.
    1. World Health Organization. Geneva, Switzerland: World Health Organization; 2010. Global report on antimalarial efficacy and drug resistance: 2000–2010.
    1. Dondorp AM, Nosten F, Yi P, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–67.
    1. Amaratunga C, Sreng S, Suon S, et al. Artemisinin-resistant Plasmodium falciparum in Pursat province, western Cambodia: a parasite clearance rate study. Lancet Infect Dis. 2012;12:851–8.
    1. Phyo AP, Nkhoma S, Stepniewska K, et al. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–6.
    1. Toshihiro M, Venkatesan M, Ohashi J, et al. Limited geographical origin and global spread of sulfadoxine-resistant dhps alleles in Plasmodium falciparum populations. J Infect Dis. 2011;204:1980–8.
    1. Roper C, Pearce R, Nair S, Sharp B, Nosten F, Anderson T. Intercontinental spread of pyrimethamine-resistant malaria. Science. 2004;305:1124.
    1. Ariey F, Fandeur T, Durand R, et al. Invasion of Africa by a single pfcrt allele of South East Asian type. Malar J. 2006;5:34.
    1. Fairhurst RM, Nayyar GM, Breman JG, et al. Artemisinin-resistant malaria: research challenges, opportunities, and public health implications. Am J Trop Med Hyg. 2012;87:231–41.
    1. Lim P, Alker AP, Khim N, et al. Pfmdr1 copy number and arteminisin derivatives combination therapy failure in falciparum malaria in Cambodia. Malar J. 2009;8:11.
    1. Anderson TJ, Nair S, Nkhoma S, et al. High heritability of malaria parasite clearance rate indicates a genetic basis for artemisinin resistance in western Cambodia. J Infect Dis. 2010;201:1326–30.
    1. Cheeseman IH, Miller BA, Nair S, et al. A major genome region underlying artemisinin resistance in malaria. Science. 2012;336:79–82.
    1. Stepniewska K, Ashley E, Lee SJ, et al. In vivo parasitological measures of artemisinin susceptibility. J Infect Dis. 2010;201:570–9.
    1. Maiga AW, Fofana B, Sagara I, et al. No evidence of delayed parasite clearance after oral artesunate treatment of uncomplicated falciparum malaria in Mali. Am J Trop Med Hyg. 2012;87:23–8.
    1. Newton PN, Green MD, Mildenhall DC, et al. Poor quality vital anti-malarials in Africa—an urgent neglected public health priority. Malar J. 2011;10:352.
    1. O'Neill PM, Barton VE, Ward SA. The molecular mechanism of action of artemisinin—the debate continues. Molecules. 2010;15:1705–21.
    1. Chotivanich K, Udomsangpetch R, Dondorp A, et al. The mechanisms of parasite clearance after antimalarial treatment of Plasmodium falciparum malaria. J Infect Dis. 2000;182:629–33.
    1. Newton PN, Chotivanich K, Chierakul W, et al. A comparison of the in vivo kinetics of Plasmodium falciparum ring-infected erythrocyte surface antigen-positive and -negative erythrocytes. Blood. 2001;98:450–7.
    1. Chotivanich K, Udomsangpetch R, McGready R, et al. Central role of the spleen in malaria parasite clearance. J Infect Dis. 2002;185:1538–41.
    1. Buffet PA, Milon G, Brousse V, et al. Ex vivo perfusion of human spleens maintains clearing and processing functions. Blood. 2006;107:3745–52.
    1. Beaudry JT, Fairhurst RM. Microvascular sequestration of Plasmodium falciparum. Blood. 2011;117:6410.
    1. Guindo A, Fairhurst RM, Doumbo OK, Wellems TE, Diallo DA. X-linked G6PD deficiency protects hemizygous males but not heterozygous females against severe malaria. PLoS Med. 2007;4:e66.
    1. Crompton PD, Traore B, Kayentao K, et al. Sickle cell trait is associated with a delayed onset of malaria: implications for time-to-event analysis in clinical studies of malaria. J Infect Dis. 2008;198:1265–75.
    1. White NJ. The parasite clearance curve. Malar J. 2011;10:278.
    1. Flegg JA, Guerin PJ, White NJ, Stepniewska K. Standardizing the measurement of parasite clearance in falciparum malaria: the parasite clearance estimator. Malar J. 2011;10:339.
    1. Ndiaye D, Patel V, Demas A, et al. A non-radioactive DAPI-based high-throughput in vitro assay to assess Plasmodium falciparum responsiveness to antimalarials—increased sensitivity of P. falciparum to chloroquine in Senegal. Am J Trop Med Hyg. 2010;82:228–30.
    1. Wellems TE, Panton LJ, Gluzman IY, et al. Chloroquine resistance not linked to mdr-like genes in a Plasmodium falciparum cross. Nature. 1990;345:253–5.
    1. Walliker D, Quakyi IA, Wellems TE, et al. Genetic analysis of the human malaria parasite Plasmodium falciparum. Science. 1987;236:1661–6.
    1. Silamut K, Phu NH, Whitty C, et al. A quantitative analysis of the microvascular sequestration of malaria parasites in the human brain. Am J Pathol. 1999;155:395–410.
    1. Modiano D, Petrarca V, Sirima BS, et al. Different response to Plasmodium falciparum malaria in west African sympatric ethnic groups. Proc Natl Acad Sci U S A. 1996;93:13206–11.
    1. Kaddouri H, Djimde A, Dama S, et al. Baseline in vitro efficacy of ACT component drugs on Plasmodium falciparum clinical isolates from Mali. Int J Parasitol. 2008;38:791–8.
    1. Staalsoe T, Giha HA, Dodoo D, Theander TG, Hviid L. Detection of antibodies to variant antigens on Plasmodium falciparum–infected erythrocytes by flow cytometry. Cytometry. 1999;35:329–36.
    1. Warimwe GM, Keane TM, Fegan G, et al. Plasmodium falciparum var gene expression is modified by host immunity. Proc Natl Acad Sci U S A. 2009;106:21801–6.
    1. Diatta AM, Marrama L, Tall A, et al. Relationship of binding of immunoglobulin G to Plasmodium falciparum–infected erythrocytes with parasite endemicity and antibody responses to conserved antigen in immune individuals. Clin Diagn Lab Immunol. 2004;11:6–11.
    1. Bull PC, Lowe BS, Kortok M, Molyneux CS, Newbold CI, Marsh K. Parasite antigens on the infected red cell surface are targets for naturally acquired immunity to malaria. Nat Med. 1998;4:358–60.
    1. Mackintosh CL, Mwangi T, Kinyanjui SM, et al. Failure to respond to the surface of Plasmodium falciparum–infected erythrocytes predicts susceptibility to clinical malaria amongst African children. Int J Parasitol. 2008;38:1445–54.
    1. Djimde AA, Doumbo OK, Traore O, et al. Clearance of drug-resistant parasites as a model for protective immunity in Plasmodium falciparum malaria. Am J Trop Med Hyg. 2003;69:558–63.
    1. Cohen S, Mc GI, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192:733–7.

Source: PubMed

3
Abonneren