Optimal dosing of dihydroartemisinin-piperaquine for seasonal malaria chemoprevention in young children

Palang Chotsiri, Issaka Zongo, Paul Milligan, Yves Daniel Compaore, Anyirékun Fabrice Somé, Daniel Chandramohan, Warunee Hanpithakpong, François Nosten, Brian Greenwood, Philip J Rosenthal, Nicholas J White, Jean-Bosco Ouédraogo, Joel Tarning, Palang Chotsiri, Issaka Zongo, Paul Milligan, Yves Daniel Compaore, Anyirékun Fabrice Somé, Daniel Chandramohan, Warunee Hanpithakpong, François Nosten, Brian Greenwood, Philip J Rosenthal, Nicholas J White, Jean-Bosco Ouédraogo, Joel Tarning

Abstract

Young children are the population most severely affected by Plasmodium falciparum malaria. Seasonal malaria chemoprevention (SMC) with amodiaquine and sulfadoxine-pyrimethamine provides substantial benefit to this vulnerable population, but resistance to the drugs will develop. Here, we evaluate the use of dihydroartemisinin-piperaquine as an alternative regimen in 179 children (aged 2.33-58.1 months). Allometrically scaled body weight on pharmacokinetic parameters of piperaquine result in lower drug exposures in small children after a standard mg per kg dosage. A covariate-free sigmoidal EMAX-model describes the interval to malaria re-infections satisfactorily. Population-based simulations suggest that small children would benefit from a higher dosage according to the WHO 2015 guideline. Increasing the dihydroartemisinin-piperaquine dosage and extending the dose schedule to four monthly doses result in a predicted relative reduction in malaria incidence of up to 58% during the high transmission season. The higher and extended dosing schedule to cover the high transmission period for SMC could improve the preventive efficacy substantially.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Visual predictive checks of the final population pharmacokinetic and pharmacodynamic model of piperaquine. a The final population pharmacokinetic model, b the interval-censoring time-to-event model of the internal data, and c the final pharmacodynamic model predicting the external data. Open circles represent observed capillary plasma piperaquine concentrations and open triangles represent observed venous plasma concentrations. A solid line represents the median observed plasma concentrations and dashed lines represent the 5th and 95th percentiles of the observed plasma concentrations. Shaded areas represent the predicted 95% confidence intervals of each percentile. Solid lines in panel (b) and (c) represent observed Kaplan–Meier survival plots. Shaded areas represent the 95% prediction intervals
Fig. 2
Fig. 2
Simulated venous piperaquine concentrations. a Day-7 piperaquine concentrations and b peak piperaquine concentrations after different dosing regimens, stratified by body weight. The box-whisker plots represent the median with inter-quartile range and the 95% prediction interval of 1000 simulated individuals per body weight. The horizontal dashed line represents the previously defined 30 ng mL-1 cut-off concentration at day 7 associated with therapeutic success
Fig. 3
Fig. 3
Simulation of the expected pharmacodynamic outcome. a remaining malaria-free after a single treatment regimen, b remaining malaria-free after three rounds of monthly dose regimens (day 0, 30, and 60), and c remaining malaria-free after four rounds of monthly dose regimens (day 0, 30, 60, and 90). Red lines represent the standard WHO 2010 dosing regimen, and black lines the increased dosing regimens according to the revised WHO 2015 recommendation in children (4–20 kg; n = 200 individuals per body weight, 100 replications). Solid lines represent the predicted median survival estimate of the Kaplan–Meier plot and shaded areas represent the 95% prediction intervals. Upward red arrows represent the time of DHA-PQ administrations
Fig. 4
Fig. 4
Comparisons of the expected pharmacodynamic outcomes. a Children (4–20 kg) remaining malaria-free after a single treatment regimen by day 60, b children (4–20 kg) remaining malaria-free after three rounds of a monthly dose regimen by day 120, c children (4–20 kg) remaining malaria-free after three rounds of a monthly dose regimen by day 90, d children (4–20 kg) remaining malaria-free after four rounds of a monthly dose regimen by day 120. Red box-whisker plots represent the standard WHO 2010 dosing regimen, and blue box-whisker plots represent the increased dosing regimens according to the revised WHO 2015 recommendation, stratified by body weight. The simulations are based on 200 individuals per body weight for 100 replications. The box-whisker plots represent the median with inter-quartile range and the 95% prediction interval. Right panels represent the proportions of children remaining malaria-free at the end time point; the red filled density plots represent the standard WHO 2010 dosing regimen and the blue filled density plots represent the revised WHO 2015 dosing regimen

References

    1. World Health Organization (WHO). World Malaria Report 2018 (2018).
    1. Gosling RD, Cairns ME, Chico RM, Chandramohan D. Intermittent preventive treatment against malaria: an update. Expert. Rev. Anti. Infect. Ther. 2010;8:589–606. doi: 10.1586/eri.10.36.
    1. Bojang K, et al. A randomised trial to compare the safety, tolerability and efficacy of three drug combinations for intermittent preventive treatment in children. PLoS. One. 2010;5:e11225. doi: 10.1371/journal.pone.0011225.
    1. Cisse B, et al. Randomized trial of piperaquine with sulfadoxine-pyrimethamine or dihydroartemisinin for malaria intermittent preventive treatment in children. PLoS. One. 2009;4:e7164. doi: 10.1371/journal.pone.0007164.
    1. Some AF, et al. Selection of known Plasmodium falciparum resistance-mediating polymorphisms by artemether-lumefantrine and amodiaquine-sulfadoxine-pyrimethamine but not dihydroartemisinin-piperaquine in Burkina Faso. Antimicrob. Agents Chemother. 2010;54:1949–1954. doi: 10.1128/AAC.01413-09.
    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. doi: 10.1128/AAC.04923-14.
    1. Zwang J, et al. Safety and efficacy of dihydroartemisinin-piperaquine in falciparum malaria: a prospective multi-centre individual patient data analysis. PLoS. One. 2009;4:e6358. doi: 10.1371/journal.pone.0006358.
    1. Tarning J, et al. Population pharmacokinetics of dihydroartemisinin and piperaquine in pregnant and nonpregnant women with uncomplicated malaria. Antimicrob. Agents Chemother. 2012;56:1997–2007. doi: 10.1128/AAC.05756-11.
    1. Moore BR, et al. Effect of coadministered fat on the tolerability, safety, and pharmacokinetic properties of dihydroartemisinin-piperaquine in Papua New Guinean children with uncomplicated malaria. Antimicrob. Agents Chemother. 2014;58:5784–5794. doi: 10.1128/AAC.03314-14.
    1. Tarning J, et al. Population pharmacokinetics and pharmacodynamics of piperaquine in children with uncomplicated falciparum malaria. Clin. Pharmacol. Ther. 2012;91:497–505. doi: 10.1038/clpt.2011.254.
    1. Lee TM, et al. In vitro metabolism of piperaquine is primarily mediated by CYP3A4. Xenobiotica; fate Foreign Compd. Biol. Syst. 2012;42:1088–1095. doi: 10.3109/00498254.2012.693972.
    1. Tarning J, et al. Characterization of human urinary metabolites of the antimalarial piperaquine. Drug Metab. Dispos.: Biol. fate Chem. 2006;34:2011–2019. doi: 10.1124/dmd.106.011494.
    1. Sim IK, Davis TM, Ilett KF. Effects of a high-fat meal on the relative oral bioavailability of piperaquine. Antimicrob. Agents Chemother. 2005;49:2407–2411. doi: 10.1128/AAC.49.6.2407-2411.2005.
    1. Reuter SE, et al. Effect of food on the pharmacokinetics of piperaquine and dihydroartemisinin. Clin. Drug. Investig. 2015;35:559–567. doi: 10.1007/s40261-015-0312-8.
    1. Annerberg A, et al. A small amount of fat does not affect piperaquine exposure in patients with malaria. Antimicrob. Agents Chemother. 2011;55:3971–3976. doi: 10.1128/AAC.00279-11.
    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. doi: 10.1128/AAC.02318-13.
    1. Sundell K, et al. Variable piperaquine exposure significantly impacts protective efficacy of monthly dihydroartemisinin-piperaquine for the prevention of malaria in Ugandan children. Malar. J. 2015;14:368. doi: 10.1186/s12936-015-0908-8.
    1. Lwin KM, et al. Randomized, double-blind, placebo-controlled trial of monthly versus bimonthly dihydroartemisinin-piperaquine chemoprevention in adults at high risk of malaria. Antimicrob. Agents Chemother. 2012;56:1571–1577. doi: 10.1128/AAC.05877-11.
    1. Bergstrand M, Nosten F, Lwin KM, Karlsson MO, White NJ, Tarning J. Characterization of an in vivo concentration-effect relationship for piperaquine in malaria chemoprevention. Sci. Transl. Med. 2014;6:260ra147. doi: 10.1126/scitranslmed.3005311.
    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. doi: 10.1371/journal.pmed.1001689.
    1. Nankabirwa JI, et al. Impact of intermittent preventive treatment with dihydroartemisinin-piperaquine on malaria in Ugandan schoolchildren: a randomized, placebo-controlled trial. Clin. Infect. Dis. 2014;58:1404–1412. doi: 10.1093/cid/ciu150.
    1. Tarning J, et al. Population Pharmacokinetics and Antimalarial Pharmacodynamics of Piperaquine in Patients With Plasmodium vivax Malaria in Thailand. CPT Pharmacomet. Syst. Pharmacol. 2014;3:e132. doi: 10.1038/psp.2014.29.
    1. The WorldWide Antimalarial Resistance Network (WWARN) DP Study Group.. The effect of dosing regimens on the antimalarial efficacy of dihydroartemisinin-piperaquine: a pooled analysis of individual patient data. PLoS. Med. 2013;10:e1001564. doi: 10.1371/annotation/3db421e4-3e27-4442-8092-2ad1b778f371.
    1. Price RN, et al. Clinical and pharmacological determinants of the therapeutic response to dihydroartemisinin-piperaquine for drug-resistant malaria. Antimicrob. Agents Chemother. 2007;51:4090–4097. doi: 10.1128/AAC.00486-07.
    1. Denis MB, et al. Efficacy and safety of dihydroartemisinin-piperaquine (Artekin) in Cambodian children and adults with uncomplicated falciparum malaria. Clin. Infect. Dis. 2002;35:1469–1476. doi: 10.1086/344647.
    1. World Health Organization. Guidelines for the treatment of malaria, Third edn. (WHO, Geneva, 2015).
    1. Creek D, et al. Increased risk of early vomiting among infants and young children treated with dihydroartemisinin-piperaquine compared with artemether-lumefantrine for uncomplicated malaria. Am. J. Trop. Med. Hyg. 2010;83:873–875. doi: 10.4269/ajtmh.2010.10-0158.
    1. Karunajeewa HA, et al. Pharmacokinetics and efficacy of piperaquine and chloroquine in Melanesian children with uncomplicated malaria. Antimicrob. Agents Chemother. 2008;52:237–243. doi: 10.1128/AAC.00555-07.
    1. Hung TY, et al. Population pharmacokinetics of piperaquine in adults and children with uncomplicated falciparum or vivax malaria. Br. J. Clin. Pharmacol. 2004;57:253–262. doi: 10.1046/j.1365-2125.2003.02004.x.
    1. Hoglund RM, et al. A population pharmacokinetic model of piperaquine in pregnant and non-pregnant women with uncomplicated Plasmodium falciparum malaria in Sudan. Malar. J. 2012;11:398. doi: 10.1186/1475-2875-11-398.
    1. Ashley EA, et al. Comparison of plasma, venous and capillary blood levels of piperaquine in patients with uncomplicated falciparum malaria. Eur. J. Clin. Pharmacol. 2010;66:705–712. doi: 10.1007/s00228-010-0804-7.
    1. Ouedraogo A, et al. Malaria morbidity in high and seasonal malaria transmission area of Burkina Faso. PLoS ONE. 2013;8:e50036. doi: 10.1371/journal.pone.0050036.
    1. Simpson JA, Aarons L, Collins WE, Jeffery GM, White NJ. Population dynamics of untreated Plasmodium falciparum malaria within the adult human host during the expansion phase of the infection. Parasitology. 2002;124:247–263. doi: 10.1017/S0031182001001202.
    1. White NJ. Malaria parasite clearance. Malar. J. 2017;16:88. doi: 10.1186/s12936-017-1731-1.
    1. White NJ. The assessment of antimalarial drug efficacy. Trends Parasitol. 2002;18:458–464. doi: 10.1016/S1471-4922(02)02373-5.
    1. White NJ. Assessment of the pharmacodynamic properties of antimalarial drugs in vivo. Antimicrob. Agents Chemother. 1997;41:1413–1422. doi: 10.1128/AAC.41.7.1413.
    1. Permala J., Tarning J., Nosten F., White N. J., Karlsson M. O., Bergstrand M. Prediction of improved antimalarial chemoprevention with weekly dosing of dihydroartemisinin-piperaquine. Antimicrob. Agents Chemother. 61, e02491-16 (2017).
    1. Basco LK, Ringwald P. In vitro activities of piperaquine and other 4-aminoquinolines against clinical isolates of Plasmodium falciparum in Cameroon. Antimicrob. Agents Chemother. 2003;47:1391–1394. doi: 10.1128/AAC.47.4.1391-1394.2003.
    1. Tumwebaze P, et al. Impact of antimalarial treatment and chemoprevention on the drug sensitivity of malaria parasites isolated from ugandan children. Antimicrob. Agents Chemother. 2015;59:3018–3030. doi: 10.1128/AAC.05141-14.
    1. Rasmussen S. A., et al. Changing antimalarial drug sensitivities in Uganda. Antimicrob. Agents Chemother. 61, e01516–17 (2017).
    1. Ahmed T, et al. Safety, tolerability, and single- and multiple-dose pharmacokinetics of piperaquine phosphate in healthy subjects. J. Clin. Pharmacol. 2008;48:166–175. doi: 10.1177/0091270007310384.
    1. Nebie I, et al. Humoral responses to Plasmodium falciparum blood-stage antigens and association with incidence of clinical malaria in children living in an area of seasonal malaria transmission in Burkina Faso, West Africa. Infect. Immun. 2008;76:759–766. doi: 10.1128/IAI.01147-07.
    1. World Health Organization. Guidelines for the treatment of malaria. (WHO, Geneva, 2010).
    1. Lindegardh N, Annerberg A, White NJ, Day NP. Development and validation of a liquid chromatographic-tandem mass spectrometric method for determination of piperaquine in plasma stable isotope labeled internal standard does not always compensate for matrix effects. J. Chromatogr. 2008;862:227–236.
    1. Lourens C, et al. Benefits of a pharmacology antimalarial reference standard and proficiency testing program provided by the Worldwide Antimalarial Resistance Network (WWARN) Antimicrob. Agents Chemother. 2014;58:3889–3894. doi: 10.1128/AAC.02362-14.
    1. Keizer RJ, van Benten M, Beijnen JH, Schellens JH, Huitema AD. Pirana and PCluster: a modeling environment and cluster infrastructure for NONMEM. Comput. Methods Prog. Biomed. 2011;101:72–79. doi: 10.1016/j.cmpb.2010.04.018.
    1. Lindbom L, Ribbing J, Jonsson EN. Perl-speaks-NONMEM (PsN)–a Perl module for NONMEM related programming. Comput. Methods Prog. Biomed. 2004;75:85–94. doi: 10.1016/j.cmpb.2003.11.003.
    1. Gisleskog PO, Karlsson MO, Beal SL. Use of prior information to stabilize a population data analysis. J. Pharmacokinet. Pharmacodyn. 2002;29:473–505. doi: 10.1023/A:1022972420004.
    1. Anderson BJ, Holford NH. Mechanistic basis of using body size and maturation to predict clearance in humans. Drug. Metab. Pharmacokinet. 2009;24:25–36. doi: 10.2133/dmpk.24.25.
    1. Bejon P, et al. Calculation of liver-to-blood inocula, parasite growth rates, and preerythrocytic vaccine efficacy, from serial quantitative polymerase chain reaction studies of volunteers challenged with malaria sporozoites. J. Infect. Dis. 2005;191:619–626. doi: 10.1086/427243.
    1. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction-corrected visual predictive checks for diagnosing nonlinear mixed-effects models. AAPS J. 2011;13:143–151. doi: 10.1208/s12248-011-9255-z.
    1. World Health Organization. Sigma-tau: Application for Inclusion of Dihydroartemisinin Plus Piperaquine (DHA/PPQ) Fixed Dose Combination Tablets in the 17th edition of WHO Model Lists of Essential Medicines. (WHO, Geneva, 2010).

Source: PubMed

3
Abonnere