Comparison of artemether-lumefantrine and chloroquine with and without primaquine for the treatment of Plasmodium vivax infection in Ethiopia: A randomized controlled trial

Tesfay Abreha, Jimee Hwang, Kamala Thriemer, Yehualashet Tadesse, Samuel Girma, Zenebe Melaku, Ashenafi Assef, Moges Kassa, Mark D Chatfield, Keren Z Landman, Stella M Chenet, Naomi W Lucchi, Venkatachalam Udhayakumar, Zhiyong Zhou, Ya Ping Shi, S Patrick Kachur, Daddi Jima, Amha Kebede, Hiwot Solomon, Addis Mekasha, Bereket Hailegiorgis Alemayehu, Joseph L Malone, Gunewardena Dissanayake, Hiwot Teka, Sarah Auburn, Lorenz von Seidlein, Ric N Price, Tesfay Abreha, Jimee Hwang, Kamala Thriemer, Yehualashet Tadesse, Samuel Girma, Zenebe Melaku, Ashenafi Assef, Moges Kassa, Mark D Chatfield, Keren Z Landman, Stella M Chenet, Naomi W Lucchi, Venkatachalam Udhayakumar, Zhiyong Zhou, Ya Ping Shi, S Patrick Kachur, Daddi Jima, Amha Kebede, Hiwot Solomon, Addis Mekasha, Bereket Hailegiorgis Alemayehu, Joseph L Malone, Gunewardena Dissanayake, Hiwot Teka, Sarah Auburn, Lorenz von Seidlein, Ric N Price

Abstract

Background: Recent efforts in malaria control have resulted in great gains in reducing the burden of Plasmodium falciparum, but P. vivax has been more refractory. Its ability to form dormant liver stages confounds control and elimination efforts. To compare the efficacy and safety of primaquine regimens for radical cure, we undertook a randomized controlled trial in Ethiopia.

Methods and findings: Patients with normal glucose-6-phosphate dehydrogenase status with symptomatic P. vivax mono-infection were enrolled and randomly assigned to receive either chloroquine (CQ) or artemether-lumefantrine (AL), alone or in combination with 14 d of semi-supervised primaquine (PQ) (3.5 mg/kg total). A total of 398 patients (n = 104 in the CQ arm, n = 100 in the AL arm, n = 102 in the CQ+PQ arm, and n = 92 in the AL+PQ arm) were followed for 1 y, and recurrent episodes were treated with the same treatment allocated at enrolment. The primary endpoints were the risk of P. vivax recurrence at day 28 and at day 42. The risk of recurrent P. vivax infection at day 28 was 4.0% (95% CI 1.5%-10.4%) after CQ treatment and 0% (95% CI 0%-4.0%) after CQ+PQ. The corresponding risks were 12.0% (95% CI 6.8%-20.6%) following AL alone and 2.3% (95% CI 0.6%-9.0%) following AL+PQ. On day 42, the risk was 18.7% (95% CI 12.2%-28.0%) after CQ, 1.2% (95% CI 0.2%-8.0%) after CQ+PQ, 29.9% (95% CI 21.6%-40.5%) after AL, and 5.9% (95% CI 2.4%-13.5%) after AL+PQ (overall p < 0.001). In those not prescribed PQ, the risk of recurrence by day 42 appeared greater following AL treatment than CQ treatment (HR = 1.8 [95% CI 1.0-3.2]; p = 0.059). At the end of follow-up, the incidence rate of P. vivax was 2.2 episodes/person-year for patients treated with CQ compared to 0.4 for patients treated with CQ+PQ (rate ratio: 5.1 [95% CI 2.9-9.1]; p < 0.001) and 2.3 episodes/person-year for AL compared to 0.5 for AL+PQ (rate ratio: 6.4 [95% CI 3.6-11.3]; p < 0.001). There was no difference in the occurrence of adverse events between treatment arms. The main limitations of the study were the early termination of the trial and the omission of haemoglobin measurement after day 42, resulting in an inability to estimate the cumulative risk of anaemia.

Conclusions: Despite evidence of CQ-resistant P. vivax, the risk of recurrence in this study was greater following treatment with AL unless it was combined with a supervised course of PQ. PQ combined with either CQ or AL was well tolerated and reduced recurrence of vivax malaria by 5-fold at 1 y.

Trial registration: ClinicalTrials.gov NCT01680406.

Conflict of interest statement

LvS receives a stipend as a specialty consulting editor for PLOS Medicine and serves on the journal's editorial board.

Figures

Fig 1. CONSORT flowchart of patient allocation…
Fig 1. CONSORT flowchart of patient allocation for the day 42 outcome.
AL, artemether-lumefantrine; AL+PQ, artemether-lumefantrine + primaquine; CQ, chloroquine; CQ+PQ, chloroquine + primaquine; G6PD, glucose-6-phosphate dehydrogenase; Lost, lost to follow-up.
Fig 2. Cumulative risk of P .…
Fig 2. Cumulative risk of P. vivax recurrence in all four treatment arms over the entire follow-up time.
AL, artemether-lumefantrine; AL+PQ, artemether-lumefantrine + primaquine; CQ, chloroquine; CQ+PQ, chloroquine + primaquine.
Fig 3. Histogram of the distribution of…
Fig 3. Histogram of the distribution of the number of recurrent P. vivax infections occurring during the 1-y follow-up period.
AL, artemether-lumefantrine; AL+PQ, artemether-lumefantrine + primaquine; CQ, chloroquine; CQ+PQ, chloroquine + primaquine.

References

    1. Guerra CA, Howes RE, Patil AP, Gething PW, Van Boeckel TP, Temperley WH, et al. The international limits and population at risk of Plasmodium vivax transmission in 2009. PLoS Negl Trop Dis. 2010;4(8):e774 10.1371/journal.pntd.0000774
    1. Price RN, Tjitra E, Guerra CA, Yeung S, White NJ, Anstey NM. Vivax malaria: neglected and not benign. Am J Trop Med Hyg. 2007;77:79–87.
    1. World Health Organization. World malaria report 2014. Geneva: World Health Organization; 2014.
    1. Price RN, von Seidlein L, Valecha N, Nosten F, Baird JK, White NJ. Global extent of chloroquine-resistant Plasmodium vivax: a systematic review and meta-analysis. Lancet Infect Dis. 2014;14(10):982–91. 10.1016/S1473-3099(14)70855-2
    1. Douglas NM, Pontororing GJ, Lampah DA, Yeo TW, Kenangalem E, Poespoprodjo JR, et al. Mortality attributable to Plasmodium vivax malaria: a clinical audit from Papua, Indonesia. BMC Med. 2014;12:217 10.1186/s12916-014-0217-z
    1. Price RN, Douglas NM, Anstey NM. New developments in Plasmodium vivax malaria: severe disease and the rise of chloroquine resistance. Curr Opin Infect Dis. 2009;22(5):430–5. 10.1097/QCO.0b013e32832f14c1
    1. Nigatu W, Abebe M, Dejene A. Plasmodium vivax and P. falciparum epidemiology in Gambella, south-west Ethiopia. Trop Med Parasitol. 1992;43(3):181–5.
    1. Olana D, Chibsa S, Teshome D, Mekasha A, Graves PM, Reithinger R. Malaria, Oromia regional state, Ethiopia, 2001–2006. Emerg Infect Dis. 2011;17(7):1336–7. 10.3201/eid1707.100942
    1. Manuel Ramos J, Reyes F, Tesfamariam A. Change in epidemiology of malaria infections in a rural area in Ethiopia. J Travel Med. 2005;12(3):155–6.
    1. Ethiopian Health and Nutrition Research Institute, Federal Ministry of Health. Ethiopia national malaria indicator survey 2011. Addis Ababa: Federal Ministry of Health; 2012.
    1. Lucchi NW, Karell MA, Journel I, Rogier E, Goldman I, Ljolje D, et al. PET-PCR method for the molecular detection of malaria parasites in a national malaria surveillance study in Haiti, 2011. Malar J. 2014;13:462 10.1186/1475-2875-13-462
    1. Imwong M, Nair S, Pukrittayakamee S, Sudimack D, Williams JT, Mayxay M, et al. Contrasting genetic structure in Plasmodium vivax populations from Asia and South America. Int J Parasitol. 2007;37(8–9):1013–22. 10.1016/j.ijpara.2007.02.010
    1. Joy DA, Gonzalez-Ceron L, Carlton JM, Gueye A, Fay M, McCutchan TF, et al. Local adaptation and vector-mediated population structure in Plasmodium vivax malaria. Mol Biol Evol. 2008;25(6):1245–52. 10.1093/molbev/msn073
    1. Patchen LC, Mount DL, Schwartz IK, Churchill FC. Analysis of filter-paper-absorbed, finger-stick blood samples for chloroquine and its major metabolite using high-performance liquid chromatography with fluorescence detection. J Chromatogr. 1983;278(1):81–9.
    1. Pukrittayakamee S, Tarning J, Jittamala P, Charunwatthana P, Lawpoolsri S, Lee SJ, et al. Pharmacokinetic interactions between primaquine and chloroquine. Antimicrob Agents Chemother. 2014;58(6):3354–9. 10.1128/AAC.02794-13
    1. Cabrera M, Cui L. In vitro activities of primaquine-schizonticide combinations on asexual blood stages and gametocytes of Plasmodium falciparum. Antimicrob Agents Chemother. 2015;59(12):7650–6. 10.1128/AAC.01948-15
    1. Price RN, Auburn S, Marfurt J, Cheng Q. Phenotypic and genotypic characterisation of drug-resistant Plasmodium vivax. Trends Parasitol. 2012;28(11):522–9. 10.1016/j.pt.2012.08.005
    1. Mekonnen SK, Aseffa A, Berhe N, Teklehaymanot T, Clouse RM, Gebru T, et al. Return of chloroquine-sensitive Plasmodium falciparum parasites and emergence of chloroquine-resistant Plasmodium vivax in Ethiopia. Malar J. 2014;13:244 10.1186/1475-2875-13-244
    1. Schunk M, Kumma WP, Miranda IB, Osman ME, Roewer S, Alano A, et al. High prevalence of drug-resistance mutations in Plasmodium falciparum and Plasmodium vivax in southern Ethiopia. Malar J. 2006;5:54 10.1186/1475-2875-5-54
    1. Getachew S, Thriemer K, Auburn S, Abera A, Gadisa E, Aseffa A, et al. Chloroquine efficacy for Plasmodium vivax malaria treatment in southern Ethiopia. Malar J. 2015;14(1):525.
    1. Ketema T, Getahun K, Bacha K. Therapeutic efficacy of chloroquine for treatment of Plasmodium vivax malaria cases in Halaba district, South Ethiopia. Parasit Vectors. 2011;4:46 10.1186/1756-3305-4-46
    1. Baird JK. Chloroquine resistance in Plasmodium vivax. Antimicrob Agents Chemother. 2004;48(11):4075–83. 10.1128/AAC.48.11.4075-4083.2004
    1. White NJ. Determinants of relapse periodicity in Plasmodium vivax malaria. Malar J. 2011;10:297 10.1186/1475-2875-10-297
    1. Karunajeewa HA, Mueller I, Senn M, Lin E, Law I, Gomorrai PS, et al. A trial of combination antimalarial therapies in children from Papua New Guinea. N Engl J Med. 2008;359(24):2545–57. 10.1056/NEJMoa0804915
    1. Baird JK, Rieckmann KH. Can primaquine therapy for vivax malaria be improved? Trends Parasitol. 2003;19(3):115–20.
    1. John GK, Douglas NM, von Seidlein L, Nosten F, Baird JK, White NJ, et al. Primaquine radical cure of Plasmodium vivax: a critical review of the literature. Malar J. 2012;11:280 10.1186/1475-2875-11-280
    1. Grietens KP, Soto V, Erhart A, Ribera JM, Toomer E, Tenorio A, et al. Adherence to 7-day primaquine treatment for the radical cure of P. vivax in the Peruvian Amazon. Am J Trop Med Hyg. 2010;82(6):1017–23. 10.4269/ajtmh.2010.09-0521
    1. Khantikul N, Butraporn P, Kim HS, Leemingsawat S, Tempongko MA, Suwonkerd W. Adherence to antimalarial drug therapy among vivax malaria patients in northern Thailand. J Health Popul Nutr. 2009;27(1):4–13.
    1. Pereira EA, Ishikawa EA, Fontes CJ. Adherence to Plasmodium vivax malaria treatment in the Brazilian Amazon Region. Malar J. 2011;10:355 10.1186/1475-2875-10-355
    1. Takeuchi R, Lawpoolsri S, Imwong M, Kobayashi J, Kaewkungwal J, Pukrittayakamee S, et al. Directly-observed therapy (DOT) for the radical 14-day primaquine treatment of Plasmodium vivax malaria on the Thai-Myanmar border. Malar J. 2010;9:308 10.1186/1475-2875-9-308
    1. Leslie T, Rab MA, Ahmadzai H, Durrani N, Fayaz M, Kolaczinski J, et al. Compliance with 14-day primaquine therapy for radical cure of vivax malaria—a randomized placebo-controlled trial comparing unsupervised with supervised treatment. Trans R Soc Trop Med Hyg. 2004;98(3):168–73.
    1. Bruxvoort K, Goodman C, Kachur SP, Schellenberg D. How patients take malaria treatment: a systematic review of the literature on adherence to antimalarial drugs. PLoS ONE. 2014;9(1):e84555 10.1371/journal.pone.0084555
    1. von Seidlein L, Auburn S, Espino F, Shanks D, Cheng Q, McCarthy J, et al. Review of key knowledge gaps in glucose-6-phosphate dehydrogenase deficiency detection with regard to the safe clinical deployment of 8-aminoquinoline treatment regimens: a workshop report. Malar J. 2013;12:112 10.1186/1475-2875-12-112
    1. World Health Organization. Guidelines for the treatment of malaria. 3rd edition Geneva: World Health Organization; 2015. [cited 2017 Apr 11]. .
    1. Douglas NM, Lampah DA, Kenangalem E, Simpson JA, Poespoprodjo JR, Sugiarto P, et al. Major burden of severe anemia from non-falciparum malaria species in Southern Papua: a hospital-based surveillance study. PLoS Med. 2013;10(12):e1001575 10.1371/journal.pmed.1001575

Source: PubMed

3
Subscribe