Severe Acute Malnutrition Results in Lower Lumefantrine Exposure in Children Treated With Artemether-Lumefantrine for Uncomplicated Malaria

Palang Chotsiri, Lise Denoeud-Ndam, Elisabeth Baudin, Ousmane Guindo, Halimatou Diawara, Oumar Attaher, Michiel Smit, Philippe J Guerin, Ogobara K Doumbo, Lubbe Wiesner, Karen I Barnes, Richard M Hoglund, Alassane Dicko, Jean-Francois Etard, Joel Tarning, Palang Chotsiri, Lise Denoeud-Ndam, Elisabeth Baudin, Ousmane Guindo, Halimatou Diawara, Oumar Attaher, Michiel Smit, Philippe J Guerin, Ogobara K Doumbo, Lubbe Wiesner, Karen I Barnes, Richard M Hoglund, Alassane Dicko, Jean-Francois Etard, Joel Tarning

Abstract

Severe acute malnutrition (SAM) has been reported to be associated with increased malaria morbidity in Sub-Saharan African children and may affect the pharmacology of antimalarial drugs. This population pharmacokinetic (PK)-pharmacodynamic study included 131 SAM and 266 non-SAM children administered artemether-lumefantrine twice daily for 3 days. Lumefantrine capillary plasma concentrations were adequately described by two transit-absorption compartments followed by two distribution compartments. Allometrically scaled body weight and an enzymatic maturation effect were included in the PK model. Mid-upper arm circumference was associated with decreased absorption of lumefantrine (25.4% decreased absorption per 1 cm reduction). Risk of recurrent malaria episodes (i.e., reinfection) were characterized by an interval-censored time-to-event model with a sigmoid maximum-effect model describing the effect of lumefantrine. SAM children were at risk of underexposure to lumefantrine and an increased risk of malaria reinfection compared with well-nourished children. Research on optimized regimens should be considered for malaria treatment in malnourished children.

Trial registration: ClinicalTrials.gov NCT01958905.

Conflict of interest statement

The authors declared no competing interests for this work.

© 2019 The Authors Clinical Pharmacology & Therapeutics published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Simulation‐based diagnostics for the final population pharmacokinetic model of lumefantrine. The top panel represents the prediction‐corrected visual predictive check. Open circles represent lumefantrine concentrations in severe acute malnutrition (SAM) children, and open triangles represent lumefantrine concentration in non‐SAM children. The solid line represents the 50th percentile of the observations, and the dashed lines represent the 5th and 95th percentiles of the observations. The gray areas represent the 95% confidence intervals of the simulated percentiles. The horizontal dashed line represents the lower limit of quantification (LLOQ) of lumefantrine (39.1 ng/mL). The bottom panel represents the visual predictive check of the data below the limit of quantification. The dashed line represents the observed proportion of LLOQ samples, and the shaded area represents the simulated 95% prediction interval of the proportion of LLOQ samples.
Figure 2
Figure 2
Effect of malnutrition descriptors on the pharmacokinetic (PK) parameters of lumefantrine. The graphs show the relative difference in PK parameter estimates in severe acute malnutrition (SAM) and non‐SAM children (a), and change in PK parameters estimates per 1‐cm mid‐upper arm circumferences reduction (b), and per 1 weight‐for‐age z‐score reduction (c). Y‐axes represent the change (%) in each PK parameter associated with altered malnutrition status, calculated from 1,000 bootstraps of the full covariate models. The shaded areas represent a covariate effect of ± 25%, assumed to be clinically insignificant. CL/F, oral clearance; F, relative bioavailability; MTT, mean transit time; QP/F, intercompartmental clearance; VC/F, apparent volume of distribution of the central compartment.
Figure 3
Figure 3
Simulation‐based diagnostics for the interval‐censoring time‐to‐event model of lumefantrine in severe acute malnutrition (SAM) and non‐SAM children. (a) Time‐to‐blood stage parasitemia and (b) time‐to‐malaria diagnostics. Black solid lines represent the observed Kaplan–Meier plots. Shaded areas represent the simulated 95% prediction intervals from the final pharmacodynamic model.
Figure 4
Figure 4
In silico lumefantrine dose optimization in severe acute malnutrition (SAM) and non‐SAM children. Predicted day 7 concentrations (a), area under the concentration‐time curve (AUC) (b), and maximum concentrations (Cmax) (c) of lumefantrine were plotted. Boxes and whiskers represent the median with interquartile range and the 95% prediction intervals, respectively. Horizontal dashed lines in panel a represent the median day 7 lumefantrine concentration after standard dosing regimen in nonpregnant adult patients (801 ng/mL).20 The dotted (200 ng/mL) and dashed‐dotted (175 ng/mL) lines in panel a represent the previously defined day 7 lumefantrine concentrations associated with therapeutic efficacy.21, 41 Gray areas in panel a represent the predicted clinical minimum inhibitory concentration value between 164 ng/mL and 182 ng/mL. Horizontal dashed lines in panels b and c represent the median lumefantrine exposure (AUC; 647,025 hour × ng/mL) and Cmax (6,731 ng/mL) after standard dosing in nonpregnant adult patients.20

References

    1. World Health Organization (WHO) . World Malaria Report 2018 (World Health Organization, Geneva, Switzerland, 2018).
    1. Rice, A.L. , Sacco, L. , Hyder, A. & Black, R.E. Malnutrition as an underlying cause of childhood deaths associated with infectious diseases in developing countries. Bull. World Health Organ. 78, 1207–1221 (2000).
    1. World Health Organization (WHO) & United Nations Children's Fund . WHO Child Growth Standards and the Identification of Severe Acute Malnutrition in Infants and Children (World Health Organization, Geneva, Switzerland, 2009).
    1. Pussard, E. et al Quinine disposition in globally malnourished children with cerebral malaria. Clin. Pharmacol. Ther. 65, 500–510 (1999).
    1. Krishnaswamy, K. Drug metabolism and pharmacokinetics in malutrition. Clin. Pharmacokinet. 3, 216–240 (1978).
    1. Krishnaswamy, K. Drug metabolism and pharmacokinetics in malnourished children. Clin. Pharmacokinet. 17 (suppl. 1), 68–88 (1989).
    1. Das, D. et al Complex interactions between malaria and malnutrition: a systematic literature review. BMC Med. 16, 186 (2018).
    1. Treluyer, J.M. , Roux, A. , Mugnier, C. , Flouvat, B. & Lagardere, B. Metabolism of quinine in children with global malnutrition. Pediatr. Res. 40, 558–563 (1996).
    1. Salako, L.A. , Sowunmi, A. & Akinbami, F.O. Pharmacokinetics of quinine in African children suffering from kwashiorkor. Br. J. Clin. Pharmacol. 28, 197–201 (1989).
    1. Tulpule, A. & Krishnaswamy, K. Chloroquine kinetics in the undernourished. Eur. J. Clin. Pharmacol. 24, 273–276 (1983).
    1. Walker, O. , Dawodu, A.H. , Salako, L.A. , Alvan, G. & Johnson, A.O. Single dose disposition of chloroquine in kwashiorkor and normal children – evidence for decreased absorption in kwashiorkor. Br. J. Clin. Pharmacol. 23, 467–472 (1987).
    1. World Health Organization (WHO) . Guidelines for the Treatment of Malaria, 3rd edn (World Health Organization, Geneva, Switzerland, 2015).
    1. Karbwang, J. , Na‐Bangchang, K. , Congpuong, K. , Thanavibul, A. , Wattanakoon, Y. & Molunto, P. Pharmacokinetics of oral artemether in Thai patients with uncomplicated falciparum malaria. Fundam. Clin. Pharmacol. 12, 242–244 (1998).
    1. Wong, R.P. , Salman, S. , Ilett, K.F. , Siba, P.M. , Mueller, I. & Davis, T.M. Desbutyl‐lumefantrine is a metabolite of lumefantrine with potent in vitro antimalarial activity that may influence artemether‐lumefantrine treatment outcome. Antimicrob. Agents Chemother. 55, 1194–1198 (2011).
    1. van Vugt, M. , Ezzet, F. , Phaipun, L. , Nosten, F. & White, N.J. The relationship between capillary and venous concentrations of the antimalarial drug lumefantrine (benflumetol). Trans. R Soc. Trop. Med. Hyg. 92, 564–565 (1998).
    1. Kloprogge, F. et al Population pharmacokinetics of lumefantrine in pregnant and nonpregnant women with uncomplicated Plasmodium falciparum malaria in Uganda. CPT Pharmacometrics Syst. Pharmacol. 2, e83 (2013).
    1. Huang, L. et al Strong correlation of lumefantrine concentrations in capillary and venous plasma from malaria patients. PLoS One 13, e0202082 (2018).
    1. Borrmann, S. et al The effect of food consumption on lumefantrine bioavailability in African children receiving artemether‐lumefantrine crushed or dispersible tablets (Coartem) for acute uncomplicated Plasmodium falciparum malaria. Trop. Med. Int. Health 15, 434–441 (2010).
    1. Ashley, E.A. et al How much fat is necessary to optimize lumefantrine oral bioavailability? Trop. Med. Int. Health 12, 195–200 (2007).
    1. Kloprogge, F. et al Artemether‐lumefantrine dosing for malaria treatment in young children and pregnant women: a pharmacokinetic‐pharmacodynamic meta‐analysis. PLoS Med. 15, e1002579 (2018).
    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. 13, 227 (2015).
    1. Ahn, J.E. , Karlsson, M.O. , Dunne, A. & Ludden, T.M. Likelihood based approaches to handling data below the quantification limit using NONMEM VI. J. Pharmacokinet. Pharmacodyn. 35, 401–421 (2008).
    1. Staehli Hodel, E.M. et al Population pharmacokinetics of mefloquine, piperaquine and artemether‐lumefantrine in Cambodian and Tanzanian malaria patients. Malar. J. 12, 235 (2013).
    1. Tchaparian, E. et al Population pharmacokinetics and pharmacodynamics of lumefantrine in young Ugandan children treated with artemether‐lumefantrine for uncomplicated malaria. J. Infect. Dis. 214, 1243–1251 (2016).
    1. Kloprogge, F. et al Lumefantrine and desbutyl‐lumefantrine population pharmacokinetic‐pharmacodynamic relationships in pregnant women with uncomplicated Plasmodium falciparum malaria on the Thailand‐Myanmar border. Antimicrob. Agents Chemother. 59, 6375–6384 (2015).
    1. Tarning, J. et al Population pharmacokinetics of lumefantrine in pregnant women treated with artemether‐lumefantrine for uncomplicated Plasmodium falciparum malaria. Antimicrob. Agents Chemother. 53, 3837–3846 (2009).
    1. Hoglund, R.M. et al Artemether‐lumefantrine co‐administration with antiretrovirals: population pharmacokinetics and dosing implications. Br. J. Clin. Pharmacol. 79, 636–649 (2015).
    1. Salman, S. et al Population pharmacokinetics of artemether, lumefantrine, and their respective metabolites in Papua New Guinean children with uncomplicated malaria. Antimicrob. Agents Chemother. 55, 5306–5313 (2011).
    1. Beal, S.L. Ways to fit a PK model with some data below the quantification limit. J. Pharmacokinet. Pharmacodyn. 28, 481–504 (2001).
    1. Anderson, B.J. & Holford, N.H. Mechanism‐based concepts of size and maturity in pharmacokinetics. Annu. Rev. Pharmacol. Toxicol. 48, 303–332 (2008).
    1. Bergstrand, M. , Nosten, F. , Lwin, K.M. , Karlsson, M.O. , White, N.J. & Tarning, J. Characterization of an in vivo concentration‐effect relationship for piperaquine in malaria chemoprevention. Sci. Transl. Med. 6, 260ra147 (2014).
    1. Chotsiri, P. et al Optimal dosing of dihydroartemisinin‐piperaquine for seasonal malaria chemoprevention in young children. Nat. Commun. 10, 480 (2019).
    1. Simpson, J.A. , Aarons, L. , Collins, W.E. , Jeffery, G.M. & White, N.J. Population dynamics of untreated Plasmodium falciparum malaria within the adult human host during the expansion phase of the infection. Parasitology 124, 247–263 (2002).
    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. 191, 619–626 (2005).
    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. 14, 167 (2016).
    1. Denoeud‐Ndam, L. et al A multi‐center, open‐label trial to compare the efficacy and pharmacokinetics of Artemether‐Lumefantrine in children with severe acute malnutrition versus children without severe acute malnutrition: study protocol for the MAL‐NUT study. BMC Infect. Dis. 15, 228 (2015).
    1. World Health Organization . Methods and Techniques for Assessing Exposure to Antimalarial Drugs in Clinical Field Studies (World Health Organization, Geneva, Switzerland, 2010).
    1. Keizer, R.J. , van Benten, M. , Beijnen, J.H. , Schellens, J.H. & Huitema, A.D. Pirana and PCluster: a modeling environment and cluster infrastructure for NONMEM. Comput. Methods Programs Biomed. 101, 72–79 (2011).
    1. Lindbom, L. , Ribbing, J. & Jonsson, E.N. Perl‐speaks‐NONMEM (PsN) – a Perl module for NONMEM related programming. Comput. Methods Programs Biomed. 75, 85–94 (2004).
    1. Jonsson, E.N. & Karlsson, M.O. Xpose – an S‐PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput. Methods Programs Biomed. 58, 51–64 (1999).
    1. Price, R.N. et al Molecular and pharmacological determinants of the therapeutic response to artemether‐lumefantrine in multidrug‐resistant Plasmodium falciparum malaria. Clin. Infect. Dis. 42, 1570–1577 (2006).

Source: PubMed

3
Iratkozz fel