Identifying an optimal dihydroartemisinin-piperaquine dosing regimen for malaria prevention in young Ugandan children

Erika Wallender, Ali Mohamed Ali, Emma Hughes, Abel Kakuru, Prasanna Jagannathan, Mary Kakuru Muhindo, Bishop Opira, Meghan Whalen, Liusheng Huang, Marvin Duvalsaint, Jenny Legac, Moses R Kamya, Grant Dorsey, Francesca Aweeka, Philip J Rosenthal, Rada M Savic, Erika Wallender, Ali Mohamed Ali, Emma Hughes, Abel Kakuru, Prasanna Jagannathan, Mary Kakuru Muhindo, Bishop Opira, Meghan Whalen, Liusheng Huang, Marvin Duvalsaint, Jenny Legac, Moses R Kamya, Grant Dorsey, Francesca Aweeka, Philip J Rosenthal, Rada M Savic

Abstract

Intermittent preventive treatment (IPT) with dihydroartemisinin-piperaquine (DP) is highly protective against malaria in children, but is not standard in malaria-endemic countries. Optimal DP dosing regimens will maximize efficacy and reduce toxicity and resistance selection. We analyze piperaquine (PPQ) concentrations (n = 4573), malaria incidence data (n = 326), and P. falciparum drug resistance markers from a trial of children randomized to IPT with DP every 12 weeks (n = 184) or every 4 weeks (n = 96) from 2 to 24 months of age (NCT02163447). We use nonlinear mixed effects modeling to establish malaria protective PPQ levels and risk factors for suboptimal protection. Compared to DP every 12 weeks, DP every 4 weeks is associated with 95% protective efficacy (95% CI: 84-99%). A PPQ level of 15.4 ng/mL reduces the malaria hazard by 95%. Malnutrition reduces PPQ exposure. In simulations, we show that DP every 4 weeks is optimal across a range of transmission intensities, and age-based dosing improves malaria protection in young or malnourished children.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Summary of trial interventions and…
Fig. 1. Summary of trial interventions and sampling for piperaquine concentrations.
Blue dots indicate dihydroartemisinin-piperaquine (DP) courses dispensed, and dashes indicate episodes when a placebo was given. The red dots indicate sparse pharmacokinetic (PK) sampling for piperaquine concentrations (all participants contributed data) and red lines indicate typical pharmacokinetic profiles. Intensive sampling, in orange dots, occurred at 32 and 104 weeks (22 individuals in every 12 weeks and 10 in the every 4 weeks arm).
Fig. 2. Study participant follow-up and malaria…
Fig. 2. Study participant follow-up and malaria outcomes in the clinical trial.
DP indicates dihydroartemisinin-piperaquine.
Fig. 3. Raw pharmacokinetic data.
Fig. 3. Raw pharmacokinetic data.
A Piperaquine (PPQ) concentration from intensive sampling after the third daily dihydroartemisinin-piperaquine (DP) dose (day 2) for 32 children at 32 and 104 weeks of age. B PPQ concentrations from sparse sampling obtained from 280 children at 28-days intervals. Boxes indicate PPQ levels for 25% (minima), 50% (center), and 75% (maxima) of the population.
Fig. 4. Malaria outcomes data.
Fig. 4. Malaria outcomes data.
A Malaria incidence per person year, stratified by dihydroartemisinin-piperaquine (DP) regimen. The lines indicate malaria incidence for the DP every 12 weeks (n = 184) (blue) and every 4 weeks (n = 96) (red) arms. Gray bands indicate periods of indoor residual spraying, and horizontal bars indicate times when participants received DP (pink) or were monitored off DP (purple). As study enrollment occurred over several months, there was a period where part of the cohort was on DP and others had completed the intervention (overlap between pink and purple bars). Time to malaria after receiving DP in the every 12-week arm B and the every 4-week arm C stratified by malaria transmission period. The low transmission periods are combined and the high transmission periods are shown separately.
Fig. 5. Relationship between piperaquine (PPQ) concentration…
Fig. 5. Relationship between piperaquine (PPQ) concentration and protective efficacy.
A Measured PPQ concentrations by days after dihydroartemisinin-piperaquine (DP) and at time of malaria diagnosis (n = 280 children). Points indicate observed data, boxes indicate PPQ levels for 25% (minima), 50% (center), and 75% (maxima) of the population, and vertical bars represent PPQ levels for 95% of the population. B Relationship between PPQ concentration and protective efficacy. The solid line indicates the median estimate from the pharmacokinetic/pharmacodynamic model, and the shaded areas the 95% confidence interval. The indicated concentration (15.4 ng/mL) reduced the hazard of incident malaria by 95%.
Fig. 6. Simulation results stratified by weight…
Fig. 6. Simulation results stratified by weight for age z-score (WAZ).
A Predicted piperaquine (PPQ) trough concentrations for simulated dihydroartemisinin-piperaquine (DP) regimens, stratified by nutritional status. Data are derived from 1000 simulations of 856 children <2 years of age. The red line indicates the 15.4 ng/mL PPQ target. Points indicate observed data, boxes indicate PPQ levels for 25% (minima), 50% (center), and 75% (maxima) of the population, and vertical bars represent PPQ levels for 95% of the population. B Predicted malaria incidence by DP regimen with increasing baseline malaria transmission, stratified by nutritional status and adherence level (1/3 adherence indicates bioavailability observed for non-direct observed therapy in the study, 2/3 adherence indicates a bioavailability midpoint between the directly and non-directly observed population, and full adherence indicates the bioavailability observed in the directly observed therapy group). Data are derived from 10,000 simulations of 856 children <2 years of age. C Predicted peak PPQ concentrations by DP regimen, assuming full adherence. Data are derived from 1000 simulations of 280 children <2 years of age. Points indicate observed data, boxes indicate PPQ levels for 25% (minima), 50% (center), and 75% (maxima) of the population, and vertical bars represent PPQ levels for 95% of the population. In text is the median and 2.5–97.5% range of predicted population values for peak PPQ concentrations during chemoprevention. Age-based dosing indicates daily PPQ doses as follows: <6 months = 160 mg; 6–<18 mo = 240 mg; 18–26 mo = 320 mg.

References

    1. World Health Organization. World Malaria Report 2019 (WHO, Geneva, Switzerland, 2019).
    1. Kakuru A, et al. Impact of intermittent preventive treatment of malaria in pregnancy with dihydroartemisinin-piperaquine versus sulfadoxine-pyrimethamine on the incidence of malaria in infancy: a randomized controlled trial. BMC Med. 2020;18:207.
    1. Bigira V, et al. Protective efficacy and safety of three antimalarial regimens for the prevention of malaria in young Ugandan children: a randomized controlled trial. PLoS Med. 2014;11:e1001689.
    1. Zongo I, et al. Randomized noninferiority trial of dihydroartemisinin-piperaquine compared with sulfadoxine-pyrimethamine plus amodiaquine for seasonal malaria chemoprevention in Burkina Faso. Antimicrob. Agents Chemother. 2015;59:4387–4396.
    1. Access-Smc Partnership. Effectiveness of seasonal malaria chemoprevention at scale in west and central Africa: an observational study. Lancet. 2020;396:1829–1840.
    1. Kamya MR, et al. Efficacy and safety of three regimens for the prevention of malaria in young HIV-exposed Ugandan children: a randomized controlled trial. AIDS. 2014;28:2701–2709.
    1. German PI, Aweeka FT. Clinical pharmacology of artemisinin-based combination therapies. Clin. Pharmacokinet. 2008;47:91–102.
    1. Muhindo MK, et al. Intermittent preventive treatment with dihydroartemisinin-piperaquine and risk of malaria following cessation in young Ugandan children: a double-blind, randomised, controlled trial. Lancet Infect. Dis. 2019;19:962–972.
    1. Staedke SG, et al. Assessment of community-level effects of intermittent preventive treatment for malaria in schoolchildren in Jinja, Uganda (START-IPT trial): a cluster-randomised trial. Lancet Glob. Health. 2018;6:e668–e679.
    1. Chan XHS, et al. Risk of sudden unexplained death after use of dihydroartemisinin-piperaquine for malaria: a systematic review and Bayesian meta-analysis. Lancet Infect. Dis. 2018;18:913–923.
    1. Chotsiri P, et al. Population pharmacokinetics and electrocardiographic effects of dihydroartemisinin-piperaquine in healthy volunteers. Br. J. Clin. Pharm. 2017;83:2752–2766.
    1. Bergstrand M, et al. Characterization of an in vivo concentration-effect relationship for piperaquine in malaria chemoprevention. Sci. Transl. Med. 2014;6:260ra147.
    1. Chotsiri P, et al. Optimal dosing of dihydroartemisinin-piperaquine for seasonal malaria chemoprevention in young children. Nat. Commun. 2019;10:480.
    1. Nsobya SL, Dokomajilar C, Joloba M, Dorsey G, Rosenthal PJ. Resistance-mediating Plasmodium falciparum pfcrt and pfmdr1 alleles after treatment with artesunate-amodiaquine in Uganda. Antimicrob. Agents Chemother. 2007;51:3023–3025.
    1. Savic RM, et al. Intermittent preventive treatment for malaria in pregnancy: optimization of target concentrations of of dihydroartemisinin-piperaquine. Clin. Infect. Dis. 2018;67:1079–1088.
    1. Cairns ME, et al. Evaluation of seasonal malaria chemoprevention in two areas of intense seasonal malaria transmission: secondary analysis of a household-randomised, placebo-controlled trial in Hounde District, Burkina Faso and Bougouni District, Mali. PLoS Med. 2020;17:e1003214.
    1. Chotsiri P, et al. Severe acute malnutrition results in lower lumefantrine exposure in children treated with artemether-lumefantrine for uncomplicated malaria. Clin. Pharm. Ther. 2019;106:1299–1309.
    1. de Kock M, et al. Population pharmacokinetic properties of sulfadoxine and pyrimethamine: a pooled analysis to inform optimal dosing in African children with uncomplicated malaria. Antimicrob. Agents Chemother. 2018;62:e01370–17.
    1. Denoeud-Ndam L, et al. Efficacy of artemether-lumefantrine in relation to drug exposure in children with and without severe acute malnutrition: an open comparative intervention study in Mali and Niger. BMC Med. 2016;14:167.
    1. WorldWide Antimalarial Resistance Network (WWARN) Lumefantrine PK/PD Study Group. Artemether-lumefantrine treatment of uncomplicated Plasmodium falciparum malaria: a systematic review and meta-analysis of day 7 lumefantrine concentrations and therapeutic response using individual patient data. BMC Med. 2015;13:227.
    1. Oshikoya KA, Senbanjo IO. Pathophysiological changes that affect drug disposition in protein-energy malnourished children. Nutr. Metab. 2009;6:50.
    1. Radtke KK, et al. Alternative dosing guidelines to improve outcomes in childhood tuberculosis: a mathematical modelling study. Lancet Child Adolesc. Health. 2019;3:636–645.
    1. Ding J, et al. Adherence and population pharmacokinetic properties of amodiaquine when used for seasonal malaria chemoprevention in African Children. Clin. Pharm. Ther. 2020;107:1179–1188.
    1. Mace KE, et al. Adherence to treatment with artemether-lumefantrine for uncomplicated malaria in rural Malawi. Clin. Infect. Dis. 2011;53:772–779.
    1. Gerstl S, et al. High adherence to malaria treatment: promising results of an adherence study in South Kivu, Democratic Republic of the Congo. Malar. J. 2015;14:414.
    1. Tarning J, et al. Population pharmacokinetic assessment of the effect of food on piperaquine bioavailability in patients with uncomplicated malaria. Antimicrob. Agents Chemother. 2014;58:2052–2058.
    1. Reuter SE, et al. Effect of food on the pharmacokinetics of piperaquine and dihydroartemisinin. Clin. Drug Investig. 2015;35:559–567.
    1. Rodriguez-Barraquer I, et al. Quantifying heterogeneous malaria exposure and clinical protection in a cohort of Ugandan children. J. Infect. Dis. 2016;214:1072–1080.
    1. D’Alessandro U, et al. Malaria in infants aged less than six months - is it an area of unmet medical need? Malar. J. 2012;11:400.
    1. Wallender E, et al. Modeling prevention of malaria and selection of drug resistance with different dosing schedules of dihydroartemisinin-piperaquine preventive therapy during pregnancy in Uganda. Antimicrob. Agents Chemother. 2019;63:e01393–18.
    1. Stepniewska K, White NJ. Pharmacokinetic determinants of the window of selection for antimalarial drug resistance. Antimicrob. Agents Chemother. 2008;52:1589–1596.
    1. Asua, V. et al. Changing prevalence of potential mediators of aminoquinoline, antifolate, and artemisinin resistance across Uganda. J. Infect. Dis. 223, 985–994 (2020).
    1. Uwimana A, et al. Emergence and clonal expansion of in vitro artemisinin-resistant Plasmodium falciparum kelch13 R561H mutant parasites in Rwanda. Nat. Med. 2020;26:1602–1608.
    1. Uwimana, A. et al. Association of Plasmodium falciparum kelch13 R561H genotypes with delayed parasite clearance in Rwanda: an open-label, single-arm, multicentre, therapeutic efficacy study. Lancet Infect. Dis. 21, 1120–1128 (2021).
    1. Asua V, et al. Changing prevalence of potential mediators of aminoquinoline, antifolate, and artemisinin resistance across Uganda. J. Infect. Dis. 2021;223:985–994.
    1. Asua, V. et al. Changing molecular markers of antimalarial drug sensitivity across Uganda. Antimicrob. Agents Chemother. 63, e01818-18 (2019).
    1. Conrad MD, Rosenthal PJ. Antimalarial drug resistance in Africa: the calm before the storm? Lancet Infect. Dis. 2019;19:e338–e351.
    1. Wattanakul T, et al. Pooled multicenter analysis of cardiovascular safety and population pharmacokinetic properties of piperaquine in african patients with uncomplicated Falciparum malaria. Antimicrob. Agents Chemother. 2020;64:e01848–19.
    1. Savic RM, et al. Intermittent preventive treatment for malaria in pregnancy: optimization of target concentrations of dihydroartemisinin-piperaquine. Clin. Infect. Dis. 2018;67:1079–1088.
    1. Wallender E, et al. Predicting optimal dihydroartemisinin-piperaquine regimens to prevent malaria during pregnancy for human immunodeficiency virus-infected women receiving efavirenz. J. Infect. Dis. 2018;217:964–972.
    1. Pitt C, et al. Large-scale delivery of seasonal malaria chemoprevention to children under 10 in Senegal: an economic analysis. Health Policy Plan. 2017;32:1256–1266.
    1. Cairns M, et al. Estimating the potential public health impact of seasonal malaria chemoprevention in African children. Nat. Commun. 2012;3:881.
    1. Kakuru A, et al. Dihydroartemisinin-piperaquine for the prevention of malaria in pregnancy. N. Engl. J. Med. 2016;374:928–939.
    1. Whalen ME, et al. Reduced exposure to piperaquine, compared to adults, in young children receiving dihydroartemisinin-piperaquine as malaria chemoprevention. Clin. Pharm. Ther. 2019;106:1310–1318.
    1. Mwebaza N, et al. Determination of piperaquine concentration in human plasma and the correlation of capillary versus venous plasma concentrations. PLoS ONE. 2020;15:e0233893.
    1. LeClair NP, et al. Optimization of a ligase detection reaction-fluorescent microsphere assay for characterization of resistance-mediating polymorphisms in African samples of Plasmodium falciparum. J. Clin. Microbiol. 2013;51:2564–2570.
    1. Beal SL. Ways to fit a PK model with some data below the quantification limit. J. Pharmacokinet. Pharmacodyn. 2001;28:481–504.
    1. Nguyen TH, et al. Model evaluation of continuous data pharmacometric models: metrics and graphics. CPT Pharmacomet. Syst. Pharm. 2017;6:87–109.
    1. Hoglund RM, et al. Population pharmacokinetic properties of piperaquine in Falciparum malaria: an individual participant data meta-analysis. PLoS Med. 2017;14:e1002212.
    1. World Health Organization. (2012). WHO Policy Recommendation: Seasonal Malaria Chemoprevention (SMC) for Plasmodium Falciparum Malaria Control in High Seasonal Transmission Areas of the Sahel Sub-Region in Africa (WHO, Geneva, Switzerland, 2012).
    1. Hughes, E. et al. Piperaquine exposure is altered by pregnancy, hiv, and nutritional status in Ugandan women. Antimicrob. Agents Chemother.64, e01013-20 (2020).
    1. Conrad MD, et al. Impact of intermittent preventive treatment during pregnancy on Plasmodium falciparum drug resistance-mediating polymorphisms in Uganda. J. Infect. Dis. 2017;16:1008–1017.

Source: PubMed

3
Abonnere