Triple artemisinin-based combination therapies versus artemisinin-based combination therapies for uncomplicated Plasmodium falciparum malaria: a multicentre, open-label, randomised clinical trial

Rob W van der Pluijm, Rupam Tripura, Richard M Hoglund, Aung Pyae Phyo, Dysoley Lek, Akhter Ul Islam, Anupkumar R Anvikar, Parthasarathi Satpathi, Sanghamitra Satpathi, Prativa Kumari Behera, Amar Tripura, Subrata Baidya, Marie Onyamboko, Nguyen Hoang Chau, Yok Sovann, Seila Suon, Sokunthea Sreng, Sivanna Mao, Savuth Oun, Sovannary Yen, Chanaki Amaratunga, Kitipumi Chutasmit, Chalermpon Saelow, Ratchadaporn Runcharern, Weerayuth Kaewmok, Nhu Thi Hoa, Ngo Viet Thanh, Borimas Hanboonkunupakarn, James J Callery, Akshaya Kumar Mohanty, James Heaton, Myo Thant, Kripasindhu Gantait, Tarapada Ghosh, Roberto Amato, Richard D Pearson, Christopher G Jacob, Sónia Gonçalves, Mavuto Mukaka, Naomi Waithira, Charles J Woodrow, Martin P Grobusch, Michele van Vugt, Rick M Fairhurst, Phaik Yeong Cheah, Thomas J Peto, Lorenz von Seidlein, Mehul Dhorda, Richard J Maude, Markus Winterberg, Nguyen Thanh Thuy-Nhien, Dominic P Kwiatkowski, Mallika Imwong, Podjanee Jittamala, Khin Lin, Tin Maung Hlaing, Kesinee Chotivanich, Rekol Huy, Caterina Fanello, Elizabeth Ashley, Mayfong Mayxay, Paul N Newton, Tran Tinh Hien, Neena Valecha, Frank Smithuis, Sasithon Pukrittayakamee, Abul Faiz, Olivo Miotto, Joel Tarning, Nicholas P J Day, Nicholas J White, Arjen M Dondorp, Tracking Resistance to Artemisinin Collaboration, Rob W van der Pluijm, Rupam Tripura, Richard M Hoglund, Aung Pyae Phyo, Dysoley Lek, Akhter Ul Islam, Anupkumar R Anvikar, Parthasarathi Satpathi, Sanghamitra Satpathi, Prativa Kumari Behera, Amar Tripura, Subrata Baidya, Marie Onyamboko, Nguyen Hoang Chau, Yok Sovann, Seila Suon, Sokunthea Sreng, Sivanna Mao, Savuth Oun, Sovannary Yen, Chanaki Amaratunga, Kitipumi Chutasmit, Chalermpon Saelow, Ratchadaporn Runcharern, Weerayuth Kaewmok, Nhu Thi Hoa, Ngo Viet Thanh, Borimas Hanboonkunupakarn, James J Callery, Akshaya Kumar Mohanty, James Heaton, Myo Thant, Kripasindhu Gantait, Tarapada Ghosh, Roberto Amato, Richard D Pearson, Christopher G Jacob, Sónia Gonçalves, Mavuto Mukaka, Naomi Waithira, Charles J Woodrow, Martin P Grobusch, Michele van Vugt, Rick M Fairhurst, Phaik Yeong Cheah, Thomas J Peto, Lorenz von Seidlein, Mehul Dhorda, Richard J Maude, Markus Winterberg, Nguyen T Thuy-Nhien, Dominic P Kwiatkowski, Mallika Imwong, Podjanee Jittamala, Khin Lin, Tin Maung Hlaing, Kesinee Chotivanich, Rekol Huy, Caterina Fanello, Elizabeth Ashley, Mayfong Mayxay, Paul N Newton, Tran Tinh Hien, Neena Valeche, Frank Smithuis, Sasithon Pukrittayakamee, Abul Faiz, Olivo Miotto, Joel Tarning, Nicholas Pj Day, Nicholas J White, Arjen M Dondorp, Rob W van der Pluijm, Rupam Tripura, Richard M Hoglund, Aung Pyae Phyo, Dysoley Lek, Akhter Ul Islam, Anupkumar R Anvikar, Parthasarathi Satpathi, Sanghamitra Satpathi, Prativa Kumari Behera, Amar Tripura, Subrata Baidya, Marie Onyamboko, Nguyen Hoang Chau, Yok Sovann, Seila Suon, Sokunthea Sreng, Sivanna Mao, Savuth Oun, Sovannary Yen, Chanaki Amaratunga, Kitipumi Chutasmit, Chalermpon Saelow, Ratchadaporn Runcharern, Weerayuth Kaewmok, Nhu Thi Hoa, Ngo Viet Thanh, Borimas Hanboonkunupakarn, James J Callery, Akshaya Kumar Mohanty, James Heaton, Myo Thant, Kripasindhu Gantait, Tarapada Ghosh, Roberto Amato, Richard D Pearson, Christopher G Jacob, Sónia Gonçalves, Mavuto Mukaka, Naomi Waithira, Charles J Woodrow, Martin P Grobusch, Michele van Vugt, Rick M Fairhurst, Phaik Yeong Cheah, Thomas J Peto, Lorenz von Seidlein, Mehul Dhorda, Richard J Maude, Markus Winterberg, Nguyen Thanh Thuy-Nhien, Dominic P Kwiatkowski, Mallika Imwong, Podjanee Jittamala, Khin Lin, Tin Maung Hlaing, Kesinee Chotivanich, Rekol Huy, Caterina Fanello, Elizabeth Ashley, Mayfong Mayxay, Paul N Newton, Tran Tinh Hien, Neena Valecha, Frank Smithuis, Sasithon Pukrittayakamee, Abul Faiz, Olivo Miotto, Joel Tarning, Nicholas P J Day, Nicholas J White, Arjen M Dondorp, Tracking Resistance to Artemisinin Collaboration, Rob W van der Pluijm, Rupam Tripura, Richard M Hoglund, Aung Pyae Phyo, Dysoley Lek, Akhter Ul Islam, Anupkumar R Anvikar, Parthasarathi Satpathi, Sanghamitra Satpathi, Prativa Kumari Behera, Amar Tripura, Subrata Baidya, Marie Onyamboko, Nguyen Hoang Chau, Yok Sovann, Seila Suon, Sokunthea Sreng, Sivanna Mao, Savuth Oun, Sovannary Yen, Chanaki Amaratunga, Kitipumi Chutasmit, Chalermpon Saelow, Ratchadaporn Runcharern, Weerayuth Kaewmok, Nhu Thi Hoa, Ngo Viet Thanh, Borimas Hanboonkunupakarn, James J Callery, Akshaya Kumar Mohanty, James Heaton, Myo Thant, Kripasindhu Gantait, Tarapada Ghosh, Roberto Amato, Richard D Pearson, Christopher G Jacob, Sónia Gonçalves, Mavuto Mukaka, Naomi Waithira, Charles J Woodrow, Martin P Grobusch, Michele van Vugt, Rick M Fairhurst, Phaik Yeong Cheah, Thomas J Peto, Lorenz von Seidlein, Mehul Dhorda, Richard J Maude, Markus Winterberg, Nguyen T Thuy-Nhien, Dominic P Kwiatkowski, Mallika Imwong, Podjanee Jittamala, Khin Lin, Tin Maung Hlaing, Kesinee Chotivanich, Rekol Huy, Caterina Fanello, Elizabeth Ashley, Mayfong Mayxay, Paul N Newton, Tran Tinh Hien, Neena Valeche, Frank Smithuis, Sasithon Pukrittayakamee, Abul Faiz, Olivo Miotto, Joel Tarning, Nicholas Pj Day, Nicholas J White, Arjen M Dondorp

Abstract

Background: Artemisinin and partner-drug resistance in Plasmodium falciparum are major threats to malaria control and elimination. Triple artemisinin-based combination therapies (TACTs), which combine existing co-formulated ACTs with a second partner drug that is slowly eliminated, might provide effective treatment and delay emergence of antimalarial drug resistance.

Methods: In this multicentre, open-label, randomised trial, we recruited patients with uncomplicated P falciparum malaria at 18 hospitals and health clinics in eight countries. Eligible patients were aged 2-65 years, with acute, uncomplicated P falciparum malaria alone or mixed with non-falciparum species, and a temperature of 37·5°C or higher, or a history of fever in the past 24 h. Patients were randomly assigned (1:1) to one of two treatments using block randomisation, depending on their location: in Thailand, Cambodia, Vietnam, and Myanmar patients were assigned to either dihydroartemisinin-piperaquine or dihydroartemisinin-piperaquine plus mefloquine; at three sites in Cambodia they were assigned to either artesunate-mefloquine or dihydroartemisinin-piperaquine plus mefloquine; and in Laos, Myanmar, Bangladesh, India, and the Democratic Republic of the Congo they were assigned to either artemether-lumefantrine or artemether-lumefantrine plus amodiaquine. All drugs were administered orally and doses varied by drug combination and site. Patients were followed-up weekly for 42 days. The primary endpoint was efficacy, defined by 42-day PCR-corrected adequate clinical and parasitological response. Primary analysis was by intention to treat. A detailed assessment of safety and tolerability of the study drugs was done in all patients randomly assigned to treatment. This study is registered at ClinicalTrials.gov, NCT02453308, and is complete.

Findings: Between Aug 7, 2015, and Feb 8, 2018, 1100 patients were given either dihydroartemisinin-piperaquine (183 [17%]), dihydroartemisinin-piperaquine plus mefloquine (269 [24%]), artesunate-mefloquine (73 [7%]), artemether-lumefantrine (289 [26%]), or artemether-lumefantrine plus amodiaquine (286 [26%]). The median age was 23 years (IQR 13 to 34) and 854 (78%) of 1100 patients were male. In Cambodia, Thailand, and Vietnam the 42-day PCR-corrected efficacy after dihydroartemisinin-piperaquine plus mefloquine was 98% (149 of 152; 95% CI 94 to 100) and after dihydroartemisinin-piperaquine was 48% (67 of 141; 95% CI 39 to 56; risk difference 51%, 95% CI 42 to 59; p<0·0001). Efficacy of dihydroartemisinin-piperaquine plus mefloquine in the three sites in Myanmar was 91% (42 of 46; 95% CI 79 to 98) versus 100% (42 of 42; 95% CI 92 to 100) after dihydroartemisinin-piperaquine (risk difference 9%, 95% CI 1 to 17; p=0·12). The 42-day PCR corrected efficacy of dihydroartemisinin-piperaquine plus mefloquine (96% [68 of 71; 95% CI 88 to 99]) was non-inferior to that of artesunate-mefloquine (95% [69 of 73; 95% CI 87 to 99]) in three sites in Cambodia (risk difference 1%; 95% CI -6 to 8; p=1·00). The overall 42-day PCR-corrected efficacy of artemether-lumefantrine plus amodiaquine (98% [281 of 286; 95% CI 97 to 99]) was similar to that of artemether-lumefantrine (97% [279 of 289; 95% CI 94 to 98]; risk difference 2%, 95% CI -1 to 4; p=0·30). Both TACTs were well tolerated, although early vomiting (within 1 h) was more frequent after dihydroartemisinin-piperaquine plus mefloquine (30 [3·8%] of 794) than after dihydroartemisinin-piperaquine (eight [1·5%] of 543; p=0·012). Vomiting after artemether-lumefantrine plus amodiaquine (22 [1·3%] of 1703) and artemether-lumefantrine (11 [0·6%] of 1721) was infrequent. Adding amodiaquine to artemether-lumefantrine extended the electrocardiogram corrected QT interval (mean increase at 52 h compared with baseline of 8·8 ms [SD 18·6] vs 0·9 ms [16·1]; p<0·01) but adding mefloquine to dihydroartemisinin-piperaquine did not (mean increase of 22·1 ms [SD 19·2] for dihydroartemisinin-piperaquine vs 20·8 ms [SD 17·8] for dihydroartemisinin-piperaquine plus mefloquine; p=0·50).

Interpretation: Dihydroartemisinin-piperaquine plus mefloquine and artemether-lumefantrine plus amodiaquine TACTs are efficacious, well tolerated, and safe treatments of uncomplicated P falciparum malaria, including in areas with artemisinin and ACT partner-drug resistance.

Funding: UK Department for International Development, Wellcome Trust, Bill & Melinda Gates Foundation, UK Medical Research Council, and US National Institutes of Health.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study profile ITT=intention-to-treat. QTc-interval=corrected QT interval. *Reasons are non-exclusive.
Figure 2
Figure 2
Parasite clearance half-lives and the presence of the Pfkelch13 mutations by study site (A) Parasite clearance half-lives for each individual participant by study site, with the dotted line showing the 5 h cutoff point; participants with polyclonal infections were excluded from this graph. (B) Location of the study sites and pie charts show the proportions of participants with a parasite clearance half-life of more than 5 h and less than 5 h and which drugs were trialled at each site. AL=artemether–lumefantrine. AQ=amodiaquine. AS-MQ=artesunate–mefloquine. DHA-PHQ=dihydroartemisinin–piperaquine. MQ=mefloquine.
Figure 3
Figure 3
Pharmacokinetic drug–drug interactions Effect of mefloquine on dihydroartemisinin (A) and on piperaquine (B) when treatment is dihydroartemisinin–piperaquine with or without mefloquine. Effect of amodiaquine on artemether (C), active metabolite dihydroartemisinin (D), lumefantrine (E), and desbutyl-lumefantrine when treatment is artemether–lumefantrine with or without amodiaquine. (G) Effect of mefloquine on day-7 piperaquine plasma concentrations when the treatment is dihydroartemisinin–piperaquine with or without mefloquine. Effect of amodiaquine on day 7 lumefantrine (H) and desbutyl-lumefantrine (I) plasma concentrations when the treatment is artemether–lumefantrine with or without amodiaquine. The plots in panels A–F show the geometric mean ratios and 90% CIs of drug–drug interactions related to the specific pharmacokinetic parameters. The dashed line represents zero effect, and the dotted lines show plus or minus 20% effect. In the scatter plots in panels G–I, the red bars show the median and IQR of day 7 plasma concentrations. Cmax=maximum plasma concentration divided with mg/kg dose. Tmax=time to reach maximum concentration. AUCT=area under the concentration-time curve to time T after administration of the first dose, divided by the mg/kg dose. AUCT_lastdose=area under the concentration-time curve to time T after administration of the last dose, divided by the mg/kg dose.

References

    1. WHO World malaria report 2019. Geneva: World Health Organization, 2019. (accessed Feb 24, 2019).
    1. Ashley EA, Dhorda M, Fairhurst RM. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–423.
    1. Woodrow CJ, White NJ. The clinical impact of artemisinin resistance in southeast Asia and the potential for future spread. FEMS Microbiol Rev. 2017;41:34–48.
    1. Phyo AP, Ashley EA, Anderson TJC. Declining efficacy of artemisinin combination therapy against P. falciparum malaria on the Thai-Myanmar border (2003–2013): the role of parasite genetic factors. Clin Infect Dis. 2016;63:784–791.
    1. van der Pluijm RW, Imwong M, Chau NH. Determinants of dihydroartemisinin-piperaquine treatment failure in Plasmodium falciparum malaria in Cambodia, Thailand, and Vietnam: a prospective clinical, pharmacological, and genetic study. Lancet Infect Dis. 2019;19:952–961.
    1. Imwong M, Suwannasin K, Kunasol C. The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study. Lancet Infect Dis. 2017;17:491–497.
    1. Tse EG, Korsik M, Todd MH. The past, present and future of anti-malarial medicines. Malar J. 2019;18:93.
    1. Wootton JC, Feng X, Ferdig MT. Genetic diversity and chloroquine selective sweeps in Plasmodium falciparum. Nature. 2002;418:320–323.
    1. Roper C, Pearce R, Nair S, Sharp B, Nosten F, Anderson T. Intercontinental spread of pyrimethamine-resistant malaria. Science. 2004;305:1124.
    1. Trape JF. The public health impact of chloroquine resistance in Africa. Am J Trop Med Hyg. 2001;64(suppl 1–2):12–17.
    1. Worthington RJ, Melander C. Combination approaches to combat multidrug-resistant bacteria. Trends Biotechnol. 2013;31:177–184.
    1. Dini S, Zaloumis S, Cao P. Investigating the efficacy of triple artemisinin-based combination therapies for treating Plasmodium falciparum malaria patients using mathematical modeling. Antimicrob Agents Chemother. 2018;62:e01068–e01118.
    1. Amaratunga C, Lim P, Suon S. Dihydroartemisinin-piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis. 2016;16:357–365.
    1. Amato R, Lim P, Miotto O. Genetic markers associated with dihydroartemisinin-piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis. 2017;17:164–173.
    1. Eastman RT, Dharia NV, Winzeler EA, Fidock DA. Piperaquine resistance is associated with a copy number variation on chromosome 5 in drug-pressured Plasmodium falciparum parasites. Antimicrob Agents Chemother. 2011;55:3908–3916.
    1. Bopp S, Magistrado P, Wong W. Plasmepsin II-III copy number accounts for bimodal piperaquine resistance among Cambodian Plasmodium falciparum. Nat Commun. 2018;9:1769.
    1. Venkatesan M, Gadalla NB, Stepniewska K. Polymorphisms in Plasmodium falciparum chloroquine resistance transporter and multidrug resistance 1 genes: parasite risk factors that affect treatment outcomes for P. falciparum malaria after artemether-lumefantrine and artesunate-amodiaquine. Am J Trop Med Hyg. 2014;91:833–843.
    1. Veiga MI, Dhingra SK, Henrich PP. Globally prevalent PfMDR1 mutations modulate Plasmodium falciparum susceptibility to artemisinin-based combination therapies. Nat Commun. 2016;7:11553.
    1. WHO . Guidelines for the treatment of malaria, 3rd edn. World Health Organization; Geneva: April, 2015.
    1. Division of AIDS (DAIDS) table for grading the severity of adult and pediatric adverse events Version 2.0. Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services. November, 2014. (accessed Feb 24, 2020).
    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. 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 B Analyt Technol Biomed Life Sci. 2008;862:227–236.
    1. Hanpithakpong W, Kamanikom B, Dondorp AM. A liquid chromatographic-tandem mass spectrometric method for determination of artesunate and its metabolite dihydroartemisinin in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2008;876:61–68.
    1. Hanpithakpong W, Kamanikom B, Singhasivanon P, White NJ, Day NP, Lindegardh N. A liquid chromatographic-tandem mass spectrometric method for determination of artemether and its metabolite dihydroartemisinin in human plasma. Bioanalysis. 2009;1:37–46.
    1. Phyo AP, Nkhoma S, Stepniewska K. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–1966.
    1. Hamilton WL, Amato R, van der Pluijm RW. Evolution and expansion of multidrug-resistant malaria in southeast Asia: a genomic epidemiology study. Lancet Infect Dis. 2019;19:943–951.
    1. WWARN K13 Genotype-Phenotype Study Group Association of mutations in the Plasmodium falciparum Kelch13 gene (Pf3D7_1343700) with parasite clearance rates after artemisinin-based treatments-a WWARN individual patient data meta-analysis. BMC Med. 2019;17:1.
    1. WHO Methods and techniques for clinical trials on antimalarial drug efficacy: genotyping to identify parasite populations. Geneva: World Health Organization, 2008. (accessed Feb 24, 2020).
    1. Nguyen TH, Day NP, Ly VC. Post-malaria neurological syndrome. Lancet. 1996;348:917–921.
    1. Chan XHS, Win YN, Mawer LJ, Tan JY, Brugada J, White NJ. 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. Witkowski B, Duru V, Khim N. A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: a phenotype-genotype association study. Lancet Infect Dis. 2017;17:174–183.
    1. Rossi G, De Smet M, Khim N, Kindermans JM, Menard D. Emergence of Plasmodium falciparum triple mutant in Cambodia. Lancet Infect Dis. 2017;17:1233.
    1. Hanboonkunupakarn B, van der Pluijm RW, Hoglund R. Sequential open-label study of the safety, tolerability, and pharmacokinetic interactions between dihydroartemisinin-piperaquine and mefloquine in healthy Thai adults. Antimicrob Agents Chemother. 2019;63:e00060–e00119.
    1. Onyamboko MA, Fanello CI, Wongsaen K. Randomized comparison of the efficacies and tolerabilities of three artemisinin-based combination treatments for children with acute Plasmodium falciparum malaria in the Democratic Republic of the Congo. Antimicrob Agents Chemother. 2014;58:5528–5536.
    1. Das S, Manna S, Saha B, Hati AK, Roy S. Novel pfkelch13 gene polymorphism associates with artemisinin resistance in eastern India. Clin Infect Dis. 2019;69:1144–1152.
    1. Miotto O, Amato R, Ashley EA. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet. 2015;47:226–234.

Source: PubMed

3
購読する