Antiretroviral Choice for HIV Impacts Antimalarial Exposure and Treatment Outcomes in Ugandan Children

Sunil Parikh, Richard Kajubi, Liusheng Huang, Joshua Ssebuliba, Sylvia Kiconco, Qin Gao, Fangyong Li, Moses Were, Abel Kakuru, Jane Achan, Norah Mwebaza, Francesca T Aweeka, Sunil Parikh, Richard Kajubi, Liusheng Huang, Joshua Ssebuliba, Sylvia Kiconco, Qin Gao, Fangyong Li, Moses Were, Abel Kakuru, Jane Achan, Norah Mwebaza, Francesca T Aweeka

Abstract

Background: The optimal treatment of malaria in human immunodeficiency virus (HIV)-infected children requires consideration of critical drug-drug interactions in coinfected children, as these may significantly impact drug exposure and clinical outcomes.

Methods: We conducted an intensive and sparse pharmacokinetic/pharmacodynamic study in Uganda of the most widely adopted artemisinin-based combination therapy, artemether-lumefantrine. HIV-infected children on 3 different first-line antiretroviral therapy (ART) regimens were compared to HIV-uninfected children not on ART, all of whom required treatment for Plasmodium falciparum malaria. Pharmacokinetic sampling for artemether, dihydroartemisinin, and lumefantrine exposure was conducted through day 21, and associations between drug exposure and outcomes through day 42 were investigated.

Results: One hundred forty-five and 225 children were included in the intensive and sparse pharmacokinetic analyses, respectively. Compared with no ART, efavirenz (EFV) reduced exposure to all antimalarial components by 2.1- to 3.4-fold; lopinavir/ritonavir (LPV/r) increased lumefantrine exposure by 2.1-fold; and nevirapine reduced artemether exposure only. Day 7 concentrations of lumefantrine were 10-fold lower in children on EFV vs LPV/r-based ART, changes that were associated with an approximate 4-fold higher odds of recurrent malaria by day 28 in those on EFV vs LPV/r-based ART.

Conclusions: The choice of ART in children living in a malaria-endemic region has highly significant impacts on the pharmacokinetics and pharmacodynamics of artemether-lumefantrine treatment. EFV-based ART reduces all antimalarial components and is associated with the highest risk of recurrent malaria following treatment. For those on EFV, close clinical follow-up for recurrent malaria following artemether-lumefantrine treatment, along with the study of modified dosing regimens that provide higher exposure, is warranted.

Keywords: HIV; antimalarial; antiretroviral; malaria; pharmacokinetics.

© The Author 2016. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Plasma concentration-time profile of artemether (A), dihydroartemisinin (DHA) (B), and lumefantrine (C) in human immunodeficiency virus (HIV)-uninfected (no antiretroviral therapy [ART]) and HIV-infected children (stabilized on either an efavirenz [EFV]-, nevirapine [NVP]-, or lopinavair/ritonavir [LPV/r]–based regimen). Data are represented as median, and values below the limit of quantitation (BLQ) are shown.
Figure 2.
Figure 2.
Cumulative risk of recurrent malaria by day 42 following treatment with artemether-lumefantrine stratified by human immunodeficiency virus (HIV) status and antiretroviral therapy regimen. Includes children from intensive and sparse cohorts. Risk-adjusted for repeated measures, age, parasite density, and hemoglobin on day 0. Abbreviations: EFV, efavirenz; LPV/r, lopinavir/ritonavir; NVP, nevirapine.
Figure 3.
Figure 3.
Cumulative risk of recurrent malaria by day 42 following treatment with artemether-lumefantrine stratified by human immunodeficiency virus (HIV) status and a day 7 lumefantrine concentration of 200 ng/mL. Includes children from intensive and sparse cohorts. Risk-adjusted for repeated measures, age, parasite density, and hemoglobin on day 0.

References

    1. World Health Organization. World malaria report. Geneva, Switzerland: WHO, 2014.
    1. Yeka A, Gasasira A, Mpimbaza A et al. . Malaria in Uganda: challenges to control on the long road to elimination: I. Epidemiology and current control efforts. Acta Trop 2012; 121:184–95.
    1. Jagannathan P, Muhindo MK, Kakuru A et al. . Increasing incidence of malaria in children despite insecticide-treated bed nets and prompt anti-malarial therapy in Tororo, Uganda. Malar J 2012; 11:435.
    1. Achan J, Kakuru A, Ikilezi G et al. . Antiretroviral agents and prevention of malaria in HIV-infected Ugandan children. N Engl J Med 2012; 367:2110–8.
    1. World Health Organization. HIV/AIDS fact sheet. Geneva, Switzerland: WHO, 2014.
    1. Joint United Nations Programme on HIV/AIDS. The gap report. Geneva, Switzerland: WHO, 2014.
    1. World Health Organization. Guideline on when to start antiretroviral therapy and on pre-exposure prophylaxis for HIV. Geneva, Switzerland: WHO, 2015.
    1. World Health Organization. Guidelines on post-exposure prophylaxis for HIV and the use of co-trimoxazole prophylaxis for HIV-related infections among adults, adolescents and children: recommendations for a public health approach. Geneva, Switzerland: WHO, 2014.
    1. World Health Organization. Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: recommendations for a public health approach. Geneva, Switzerland: WHO, 2013.
    1. Flateau C, Le Loup G, Pialoux G. Consequences of HIV infection on malaria and therapeutic implications: a systematic review. Lancet Infect Dis 2011; 11:541–56.
    1. Church JA, Fitzgerald F, Walker AS, Gibb DM, Prendergast AJ. The expanding role of co-trimoxazole in developing countries. Lancet Infect Dis 2015; 15:327–39.
    1. Khoo S, Back D, Winstanley P. The potential for interactions between antimalarial and antiretroviral drugs. AIDS 2005; 19:995–1005.
    1. Barnes KI, Watkins WM, White NJ. Antimalarial dosing regimens and drug resistance. Trends Parasitol 2008; 24:127–34.
    1. Ilett KF, Ethell BT, Maggs JL et al. . Glucuronidation of dihydroartemisinin in vivo and by human liver microsomes and expressed UDP-glucuronosyltransferases. Drug Metab Dispos 2002; 30:1005–12.
    1. Wong RP, Salman S, Ilett KF, Siba PM, Mueller I, Davis TM. Desbutyl-lumefantrine is a metabolite of lumefantrine with potent in vitro antimalarial activity that may influence artemether-lumefantrine treatment outcome. Antimicrob Agents Chemother 2011; 55:1194–8.
    1. Greenhouse B, Myrick A, Dokomajilar C et al. . Validation of microsatellite markers for use in genotyping polyclonal Plasmodium falciparum infections. Am J Trop Med Hyg 2006; 75:836–42.
    1. Ashley EA, Stepniewska K, Lindegardh N et al. . How much fat is necessary to optimize lumefantrine oral bioavailability? Trop Med Int Health 2007; 12:195–200.
    1. Huang L, Olson A, Gingrich D, Aweeka FT. Determination of artemether and dihydroartemisinin in human plasma with a new hydrogen peroxide stabilization method. Bioanalysis 2013; 5:1501–6.
    1. Huang L, Li X, Marzan F, Lizak PS, Aweeka FT. Determination of lumefantrine in small-volume human plasma by LC-MS/MS: using a deuterated lumefantrine to overcome matrix effect and ionization saturation. Bioanalysis 2012; 4:157–66.
    1. National Institute of Allergy and Infectious Diseases—Division of AIDS. Division of AIDS table for grading the severity of adult and pediatric adverse events, 2004. Available at: Accessed 23 October 2015.
    1. World Health Organization. Methods for surveillance of antimalarial drug efficacy, 2009. Available at: Accessed 23 October 2015.
    1. MacKinnon DP, Dwyer JH. Estimating mediated effects in prevention studies. Eval Rev 1993; 17:144–58.
    1. Price RN, Uhlemann AC, van Vugt M et al. . Molecular and pharmacological determinants of the therapeutic response to artemether-lumefantrine in multidrug-resistant Plasmodium falciparum malaria. Clin Infect Dis 2006; 42:1570–7.
    1. White NJ, van Vugt M, Ezzet F. Clinical pharmacokinetics and pharmacodynamics and pharmacodynamics of artemether-lumefantrine. Clin Pharmacokinet 1999; 37:105–25.
    1. WorldWide Antimalarial Resistance Network Lumefantrine PKPDSG. 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. Riska P, Lamson M, MacGregor T et al. . Disposition and biotransformation of the antiretroviral drug nevirapine in humans. Drug Metab Dispos 1999; 27:895–901.
    1. Back D, Gibbons S, Khoo S. Pharmacokinetic drug interactions with nevirapine. J Acquir Immune Defic Syndr 2003; 34(suppl 1):S8–14.
    1. Barnes KI, Little F, Smith PJ, Evans A, Watkins WM, White NJ. Sulfadoxine-pyrimethamine pharmacokinetics in malaria: pediatric dosing implications. Clin Pharmacol Ther 2006; 80:582–96.
    1. Takala-Harrison S, Jacob CG, Arze C et al. . Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis 2015; 211:670–9.
    1. Ashley EA, Dhorda M, Fairhurst RM et al. . Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 2014; 371:411–23.
    1. Dondorp AM, Nosten F, Yi P et al. . Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 2009; 361:455–67.
    1. Conrad MD, LeClair N, Arinaitwe E et al. . Comparative impacts over 5 years of artemisinin-based combination therapies on Plasmodium falciparum polymorphisms that modulate drug sensitivity in Ugandan children. J Infect Dis 2014; 210:344–53.
    1. Hoglund RM, Byakika-Kibwika P, Lamorde M et al. . Artemether-lumefantrine co-administration with antiretrovirals: population pharmacokinetics and dosing implications. Br J Clin Pharmacol 2015; 79:636–49.
    1. Maganda BA, Ngaimisi E, Kamuhabwa AA, Aklillu E, Minzi OM. The influence of nevirapine and efavirenz-based anti-retroviral therapy on the pharmacokinetics of lumefantrine and anti-malarial dose recommendation in HIV-malaria co-treatment. Malar J 2015; 14:179.
    1. Hobbs CV, Neal J, Conteh S et al. . HIV treatments reduce malaria liver stage burden in a non-human primate model of malaria infection at clinically relevant concentrations in vivo. PLoS One 2014; 9:e100138.
    1. Hobbs CV, Voza T, Coppi A et al. . HIV protease inhibitors inhibit the development of preerythrocytic-stage plasmodium parasites. J Infect Dis 2009; 199:134–41.
    1. Redmond AM, Skinner-Adams T, Andrews KT et al. . Antimalarial activity of sera from subjects taking HIV protease inhibitors. AIDS 2007; 21:763–5.
    1. Porter KA, Cole SR, Eron JJ Jr et al. . HIV-1 protease inhibitors and clinical malaria: a secondary analysis of the AIDS Clinical Trials Group A5208 study. Antimicrob Agents Chemother 2012; 56:995–1000.
    1. White NJ, Stepniewska K, Barnes K, Price RN, Simpson J. Simplified antimalarial therapeutic monitoring: using the day-7 drug level? Trends Parasitol 2008; 24:159–63.
    1. Parikh S, Gut J, Istvan E, Goldberg DE, Havlir DV, Rosenthal PJ. Antimalarial activity of human immunodeficiency virus type 1 protease inhibitors. Antimicrob Agents Chemother 2005; 49:2983–5.
    1. Violari A, Cotton MF, Gibb DM et al. . Early antiretroviral therapy and mortality among HIV-infected infants. N Engl J Med 2008; 359:2233–44.
    1. Ruel TD, Kakuru A, Ikilezi G et al. . Virologic and immunologic outcomes of HIV-infected Ugandan children randomized to lopinavir/ritonavir or nonnucleoside reverse transcriptase inhibitor therapy. J Acquir Immune Defic Syndr 2014; 65:535–41.
    1. Greenhalgh S, Ndeffo M, Galvani AP, Parikh S. The epidemiological impact of HIV antiretroviral therapy on malaria in children. AIDS 2015; 29:473–82.
    1. Gasasira AF, Kamya MR, Ochong EO et al. . Effect of trimethoprim-sulphamethoxazole on the risk of malaria in HIV-infected Ugandan children living in an area of widespread antifolate resistance. Malar J 2010; 9:177.
    1. Maganda BA, Minzi OM, Kamuhabwa AA, Ngasala B, Sasi PG. Outcome of artemether-lumefantrine treatment for uncomplicated malaria in HIV-infected adult patients on anti-retroviral therapy. Malar J 2014; 13:205.
    1. Byakika-Kibwika P, Lamorde M, Mayito J et al. . Significant pharmacokinetic interactions between artemether/lumefantrine and efavirenz or nevirapine in HIV-infected Ugandan adults. J Antimicrob Chemother 2012; 67:2213–21.
    1. Byakika-Kibwika P, Lamorde M, Okaba-Kayom V et al. . Lopinavir/ritonavir significantly influences pharmacokinetic exposure of artemether/lumefantrine in HIV-infected Ugandan adults. J Antimicrob Chemother 2012; 67:1217–23.
    1. Kredo T, Mauff K, Workman L et al. . The interaction between artemether-lumefantrine and lopinavir/ritonavir-based antiretroviral therapy in HIV-1 infected patients. BMC Infect Dis 2016; 16:30.
    1. German P, Greenhouse B, Coates C et al. . Hepatotoxicity due to a drug interaction between amodiaquine plus artesunate and efavirenz. Clin Infect Dis 2007; 44:889–91.
    1. German P, Parikh S, Lawrence J et al. . Lopinavir/ritonavir affects pharmacokinetic exposure of artemether/lumefantrine in HIV-uninfected healthy volunteers. J Acquir Immune Defic Syndr 2009; 51:424–9.
    1. Huang L, Parikh S, Rosenthal PJ et al. . Concomitant efavirenz reduces pharmacokinetic exposure to the antimalarial drug artemether-lumefantrine in healthy volunteers. J Acquir Immune Defic Syndr 2012; 61:310–6.
    1. Sambol NC, Yan L, Creek DJ et al. . Population pharmacokinetics of piperaquine in young Ugandan children treated with dihydroartemisinin-piperaquine for uncomplicated malaria. Clin Pharmacol Ther 2015; 98:87–95.
    1. Mwesigwa J, Parikh S, McGee B et al. . Pharmacokinetics of artemether-lumefantrine and artesunate-amodiaquine in children in Kampala, Uganda. Antimicrob Agents Chemother 2010; 54:52–9.
    1. Parikh S, Fehintola F, Huang L et al. . Artemether-lumefantrine exposure in HIV-infected Nigerian subjects on nevirapine-containing antiretroviral therapy. Antimicrob Agents Chemother 2015; 59:7852–6.
    1. Doherty M, Beusenberg M, Asamoah-Odei E et al. . Rapid uptake and adoption of the WHO 2013 consolidated ARV guideline recommendations: paving the way to achieving the 90/90/90 global target. J Int AIDS Soc 2015; 18:26–7.

Source: PubMed

3
Abonner