Parasite clearance, cure rate, post-treatment prophylaxis and safety of standard 3-day versus an extended 6-day treatment of artemether-lumefantrine and a single low-dose primaquine for uncomplicated Plasmodium falciparum malaria in Bagamoyo district, Tanzania: a randomized controlled trial

Lwidiko E Mhamilawa, Billy Ngasala, Ulrika Morris, Eliford Ngaimisi Kitabi, Rory Barnes, Aung Paing Soe, Bruno P Mmbando, Anders Björkman, Andreas Mårtensson, Lwidiko E Mhamilawa, Billy Ngasala, Ulrika Morris, Eliford Ngaimisi Kitabi, Rory Barnes, Aung Paing Soe, Bruno P Mmbando, Anders Björkman, Andreas Mårtensson

Abstract

Background: Artemisinin-based combination therapy (ACT) resistant Plasmodium falciparum represents an increasing threat to Africa. Extended ACT regimens from standard 3 to 6 days may represent a means to prevent its development and potential spread in Africa.

Methods: Standard 3-day treatment with artemether-lumefantrine (control) was compared to extended 6-day treatment and single low-dose primaquine (intervention); in a randomized controlled, parallel group, superiority clinical trial of patients aged 1-65 years with microscopy confirmed uncomplicated P. falciparum malaria, enrolled in Bagamoyo district, Tanzania. The study evaluated parasite clearance, including proportion of PCR detectable P. falciparum on days 5 and 7 (primary endpoint), cure rate, post-treatment prophylaxis, safety and tolerability. Clinical, and laboratory assessments, including ECG were conducted during 42 days of follow-up. Blood samples were collected for parasite detection (by microscopy and PCR), molecular genotyping and pharmacokinetic analyses. Kaplan-Meier survival analyses were done for both parasite clearance and recurrence.

Results: A total of 280 patients were enrolled, 141 and 139 in the control and intervention arm, respectively, of whom 121 completed 42 days follow-up in each arm. There was no difference in proportion of PCR positivity across the arms at day 5 (80/130 (61.5%) vs 89/134 (66.4%), p = 0.44), or day 7 (71/129 (55.0%) vs 70/134 (52.2%), p = 0.71). Day 42 microscopy determined cure rates (PCR adjusted) were 97.4% (100/103) and 98.3% (110/112), p = 0.65, in the control and intervention arm, respectively. Microscopy determined crude recurrent parasitaemia during follow-up was 21/121 (17.4%) in the control and 14/121 (11.6%) in the intervention arm, p = 0.20, and it took 34 days and 42 days in the respective arms for 90% of the patients to remain without recurrent parasitaemia. Lumefantrine exposure was significantly higher in intervention arm from D3 to D42, but cardiac, biochemical and haematological safety was high and similar in both arms.

Conclusion: Extended 6-day artemether-lumefantrine treatment and a single low-dose of primaquine was not superior to standard 3-day treatment for ACT sensitive P. falciparum infections but, importantly, equally efficacious and safe. Thus, extended artemether-lumefantrine treatment may be considered as a future treatment regimen for ACT resistant P. falciparum, to prolong the therapeutic lifespan of ACT in Africa. Trial registration ClinicalTrials.gov, NCT03241901. Registered July 27, 2017 https://ichgcp.net/clinical-trials-registry/NCT03241901.

Keywords: Artemether–lumefantrine; Drug resistance; Malaria; Plasmodium falciparum; Tanzania.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Flow of patients through the study
Fig. 2
Fig. 2
Parasite clearance by microscopy and fever clearance within treatment arms. a Percent of patients in each treatment arm positive by microscopy during first 72 h of treatment. b Percent of patients in each arm that remained febrile after initiation of artemether–lumefantrine during the first 72 h
Fig. 3
Fig. 3
Kaplan–Meier cumulative survival curve; time to microscopy determined recurrent parasitaemia (crude cure rates)
Fig. 4
Fig. 4
Proportion of P. falciparum detected by PCR among 81 patients for 20 sampling time points. Out of the 81 patients, 35 had recurrent parasitaemia by microscopy and 46 did not have recurrent parasitaemia during the 42-day follow-up period
Fig. 5
Fig. 5
Dot plot of qPCR determined parasite densities for 81 patients for 20 sampling time points. Out of the 81 patients, 35 had recurrent parasitaemia by microscopy and 46 did not have recurrent parasitaemia during the 42-day follow-up period
Fig. 6
Fig. 6
Bar plot modelling distribution of lumefantrine concentration at days 2–42 after treatment across treatment arms

References

    1. Payne D. Spread of chloroquine resistance in Plasmodium falciparum. Parasitol Today. 1987;3:241–246.
    1. Trape JF. The public health impact of chloroquine resistance in Africa. Am J Trop Med Hyg. 2001;64:12–17.
    1. Zucker JR, Lackritz EM, Ruebush TK, 2nd, Hightower AW, Adungosi JE, Were JB, et al. Childhood mortality during and after hospitalization in western Kenya: effect of malaria treatment regimens. Am J Trop Med Hyg. 1996;55:655–660.
    1. WHO . World malaria report 2018. Geneva: World Health Organization; 2018.
    1. WHO . World malaria report 2019. Geneva: World Health Organization; 2019.
    1. Dondorp AM, Nosten F, Yi P, Das D, Phyo P, Tarning J, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–467.
    1. Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–423.
    1. Das S, Saha B, Hati AK, Roy S. Evidence of artemisinin-resistant Plasmodium falciparum malaria in Eastern India. N Engl J Med. 2018;379:1962–1964.
    1. WHO . Status report on artemisinin resistance and ACT efficacy (August 2018) Geneva: World Health Organization; 2018.
    1. Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois A-C, Khim N, et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505:50–55.
    1. Tun KM, Jeeyapant A, Imwong M, Thein M, Soe S, Aung M, et al. Parasite clearance rates in Upper Myanmar indicate a distinctive artemisinin resistance phenotype: a therapeutic efficacy study. Malar J. 2016;15:185.
    1. WWARN Group GS 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. Brieger B. Presence of k13 561H artemisinin resistance mutations in Plasmodium falciparum infections from Rwanda. Trop Health Matters. 2019. . Accessed 2 June 2020.
    1. Ouattara A, Kone A, Adams M, Fofana B, Maiga AW, Hampton S, et al. Polymorphisms in the K13-propeller gene in artemisinin-susceptible Plasmodium falciparum parasites from Bougoula-Hameau and Bandiagara, Mali. Am J Trop Med Hyg. 2015;92:1202–1206.
    1. Miotto O, Amato R, Ashley EA, MacInnis B, Almagro-Garcia J, Amaratunga C, et al. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet. 2015;47:226–234.
    1. Mwaiswelo R, Ngasala B, Jovel I, Xu W, Larson E, Malmberg M, et al. Prevalence of and risk factors associated with Polymerase Chain Reaction-determined Plasmodium falciparum positivity on Day 3 after initiation of artemether–lumefantrine treatment for uncomplicated malaria in Bagamoyo District, Tanzania. Am J Trop Med Hyg. 2019;100:1179–1186.
    1. Mhamilawa LE, Aydin-Schmidt B, Mmbando BP, Ngasala B, Morris U. Detection of Plasmodium falciparum by light microscopy, loop-mediated isothermal amplification, and polymerase chain reaction on day 3 after initiation of artemether–lumefantrine treatment for uncomplicated malaria in Bagamoyo District, Tanzania: a comparative trial. Am J Trop Med Hyg. 2019;101:1144–1147.
    1. Mideo N, Bailey JA, Hathaway NJ, Ngasala B, Saunders DL, Lon C, et al. A deep-sequencing tool for partitioning clearance rates following antimalarial treatment in polyclonal infections. Evol Med Public Health. 2016;2016:21–36.
    1. Malmberg M, Ngasala B, Ferreira PE, Larsson E, Jovel I, Hjalmarsson A, et al. Temporal trends of molecular markers associated with artemether–lumefantrine tolerance/resistance in Bagamoyo district, Tanzania. Malar J. 2013;12:103.
    1. Venkatesan M, Gadalla NB, Stepniewska K, Dahal P, Nsanzabana C, Moriera C, et al. 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. Sisowath C, Petersen I, Veiga MI, Mårtensson A, Premji Z, Björkman A, et al. In vivo selection of Plasmodium falciparum parasites carrying the chloroquine-susceptible pfcrt K76 allele after treatment with artemether–lumefantrine in Africa. J Infect Dis. 2009;199:750–757.
    1. Mårtensson A, Strömberg J, Sisowath C, Msellem MI, Gil JP, Montgomery SM, et al. Efficacy of artesunate plus amodiaquine versus that of artemether–lumefantrine for the treatment of uncomplicated childhood Plasmodium falciparum malaria in Zanzibar, Tanzania. Clin Infect Dis. 2005;41:1079–1086.
    1. Malmberg M, Ferreira PE, Tarning J, Ursing J, Ngasala B, Björkman A, et al. Plasmodium falciparum drug resistance phenotype as assessed by patient antimalarial drug levels and its association with pfmdr1 polymorphisms. J Infect Dis. 2013;207:842–847.
    1. von Seidlein L, Bojang K, Jones P, Jaffar S, Pinder M, Obaro S, et al. A randomized controlled trial of artemether/benflumetol, a new antimalarial and pyrimethamine/sulfadoxine in the treatment of uncomplicated falciparum malaria in African children. Am J Trop Med Hyg. 1998;58:638–644.
    1. Omari AAA, Gamble C, Garner P. Artemether–lumefantrine (four-dose regimen) for treating uncomplicated falciparum malaria. Cochrane Datab Syst Rev. 2006;2:CD005965.
    1. Tarning J. Treatment of malaria in pregnancy. N Engl J Med. 2016;10:981–982.
    1. Anstey NM, Price RN, Davis TME, Karunajeewa HA, Mueller I, D’Alessandro U, et al. The effect of dose on the antimalarial efficacy of artemether–lumefantrine: a systematic review and pooled analysis of individual patient data. Lancet Infect Dis. 2015;15:692–702.
    1. Kloprogge F, Workman L, Borrmann S, Tekete M, Lefevre G, Hamed K, et al. Artemether–lumefantrine dosing for malaria treatment in young children and pregnant women: a pharmacokinetic-pharmacodynamic meta-analysis. PLoS Med. 2018;15:e1002579.
    1. Tun KM, Jeeyapant A, Myint AH, Kyaw ZT, Dhorda M, Mukaka M, et al. Effectiveness and safety of 3 and 5 day courses of artemether–lumefantrine for the treatment of uncomplicated falciparum malaria in an area of emerging artemisinin resistance in Myanmar NCT02020330 NCT. Malar J. 2018;17:1–8.
    1. Onyamboko MA, Hoglund RM, Lee SJ, Kabedi C, Kayembe D, Badjanga BB, et al. A randomised controlled trial of 3 versus 5 days artemether–lumefantrine regimen for uncomplicated Plasmodium falciparum treatment in pregnancy in Africa. Antimicrob Agents Chemother. 2019 doi: 10.1128/AAC.01140-19.
    1. Drakeley CJ, Eling W, Teelen K, Bousema JT, Sauerwein R, Greenwood BM, et al. Parasite infectivity and immunity to Plasmodium falciparum gametocytes in Gambian children. Parasite Immunol. 2004;26:159–165.
    1. Bousema JT, Gouagna LC, Meutstege AM, Okech BE, Akim NIJ, Githure JI, et al. Treatment failure of pyrimethamine–sulphadoxine and induction of Plasmodium falciparum gametocytaemia in children in western Kenya. Trop Med Int Health. 2003;8:427–430.
    1. Barnes KI, White NJ. Population biology and antimalarial resistance: the transmission of antimalarial drug resistance in Plasmodium falciparum. Acta Trop. 2005;94:230–240.
    1. Mwaiswelo R, Ngasala BE, Jovel I, Gosling R, Premji Z, Poirot E, et al. Safety of a single low-dose of primaquine in addition to standard artemether–lumefantrine regimen for treatment of acute uncomplicated Plasmodium falciparum malaria in Tanzania. Malar J. 2016;15:316.
    1. Mwaiswelo R, Ngasala B, Jovel I, Aydin-Schmidt B, Gosling RR, Premji Z, et al. Adding a single low-dose of primaquine (0.25 mg/kg) to artemether–lumefantrine did not compromise treatment outcome of uncomplicated Plasmodium falciparum malaria in Tanzania: a randomized, single-blinded clinical trial. Malar J. 2016;15:435.
    1. Lawpoolsri S, Klein EY, Singhasivanon P, Yimsamran S, Thanyavanich N, Maneeboonyang W, et al. Optimally timing primaquine treatment to reduce Plasmodium falciparum transmission in low endemicity Thai-Myanmar border populations. Malar J. 2009;8:159.
    1. National Malaria Control Programme, Tanzania. Supplementary malaria midterm strategic plan (2018–2020). 2018. Report No.: 43.
    1. Mwaiswelo R, Ngasala B, Gil JP, Malmberg M, Jovel I, Xu W, et al. Sustained high cure rate of artemether–lumefantrine against uncomplicated Plasmodium falciparum malaria after 8 years of its wide-scale use in Bagamoyo District, Tanzania. Am J Trop Med Hyg. 2017;97:526–532.
    1. United Republic of Tanzania. TANZANIA malaria indicator survey 2017. United Republic of Tanzania. 2017.
    1. Ministry of Health, Community Development, Gender, Elderly and Children, Tanzania. Standard treatment guidelines & national essential medicines list Tanzania mainland. 2017. .
    1. Premji ZG, Abdulla S, Ogutu B, Ndong A, Falade CO, Sagara I, et al. The content of African diets is adequate to achieve optimal efficacy with fixed-dose artemether–lumefantrine: a review of the evidence. Malar J. 2008;7:244.
    1. Borrmann S, Sallas WM, Machevo S, González R, Björkman A, Mårtensson A, 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. 2010;15:434–441.
    1. Buchanan AM, Fiorillo SP, Omondi MW, Cunningham CK, Crump JA. Establishment of biochemistry reference values for healthy Tanzanian infants, children and adolescents in Kilimanjaro Region. Trop Med Int Health. 2015;20:1569–1577.
    1. Xu W, Morris U, Aydin-Schmidt B, Msellem MI, Shakely D, Petzold M, et al. SYBR Green real-time PCR-RFLP assay targeting the Plasmodium cytochrome B gene—a highly sensitive molecular tool for malaria parasite detection and species determination. PLoS ONE. 2015;10:e0120210.
    1. Kamau E, Alemayehu S, Feghali KC, Saunders D, Ockenhouse CF. Multiplex qPCR for detection and absolute quantification of malaria. PLoS ONE. 2013;8:e71539.
    1. Greenhouse B, Dokomajilar C, Hubbard AE, Rosenthal PJ, Dorsey G, Gatton LM, et al. Methods and techniques for clinical trials on antimalarial drug efficacy: genotyping to identify parasite populations. Malar J. 2006;5:68.
    1. Martensson A, Ngasala B, Ursing J, Isabel Veiga M, Wiklund L, Membi C, et al. Influence of consecutive-day blood sampling on polymerase chain reaction-adjusted parasitological cure rates in an antimalarial-drug trial conducted in Tanzania. J Infect Dis. 2007;195:597–601.
    1. Snounou G, Zhu X, Siripoon N, Jarra W, Thaithong S, Brown KN, et al. Biased distribution of msp1 and msp2 allelic variants in Plasmodium falciparum populations in Thailand. Trans R Soc Trop Med Hyg. 1999;93:369–374.
    1. Fröberg G, Jörnhagen L, Morris U, Shakely D, Msellem MI, Gil JP, et al. Decreased prevalence of Plasmodium falciparum resistance markers to amodiaquine despite its wide scale use as ACT partner drug in Zanzibar. Malar J. 2012;11:321.
    1. Veiga MI, Ferreira PE, Björkman A, Gil JP. Multiplex PCR–RFLP methods for pfcrt, pfmdr1 and pfdhfr mutations in Plasmodium falciparum. Mol Cell Probes. 2006;20:100–104.
    1. Kamau E, Campino S, Amenga-Etego L, Drury E, Ishengoma D, Johnson K, et al. K13-propeller polymorphisms in Plasmodium falciparum parasites from sub-saharan Africa. J Infect Dis. 2015;211:1352–1355.
    1. Price RN, Uhlemann A-C, Brockman A, McGready R, Ashley E, Phaipun L, et al. Mefloquine resistance in Plasmodium falciparum and increased pfmdr1 gene copy number. Lancet. 2004;364:438–447.
    1. Bretscher MT, Griffin JT, Hugo P, Baker M, Ghani A, Okell L. A comparison of the duration of post-treatment protection of artemether–lumefantrine, dihydroartemisinin–piperaquine and artesunate–amodiaquine for the treatment of uncomplicated malaria. Malar J. 2014;13:P19.
    1. Roth JM, Omweri G, de Jong MD, Osoti V, Mens PF, Sawa P, et al. Molecular detection of residual parasitemia after pyronaridine–artesunate or artemether–lumefantrine treatment of uncomplicated Plasmodium falciparum malaria in Kenyan children. Am J Trop Med Hyg. 2018;99:970–977.
    1. Beshir KB, Sutherland CJ, Sawa P, Drakeley CJ, Okell L, Mweresa CK, et al. Residual Plasmodium falciparum parasitemia in Kenyan children after artemisinin-combination therapy is associated with increased transmission to mosquitoes and parasite recurrence. J Infect Dis. 2013;208:2017–2024.
    1. Chang H-H, Meibalan E, Zelin J, Daniels R, Eziefula AC, Meyer EC, et al. Persistence of Plasmodium falciparum parasitemia after artemisinin combination therapy: evidence from a randomized trial in Uganda. Sci Rep. 2016;6:26330.
    1. Tadesse FG, Lanke K, Nebie I, Schildkraut JA, Gonçalves BP, Tiono AB, et al. Molecular markers for sensitive detection of Plasmodium falciparum asexual stage parasites and their application in a malaria clinical trial. Am J Trop Med Hyg. 2017;97:188–198.
    1. Angus BJ, Chotivanich K, Udomsangpetch R, White NJ. In vivo removal of malaria parasites from red blood cells without their destruction in acute falciparum malaria. Blood. 1997;90:2037–2040.
    1. White NJ. The parasite clearance curve. Malar J. 2011;10:278.
    1. Ghani AC, Sutherland CJ, Riley EM, Drakeley CJ, Griffin JT, Gosling RD, et al. Loss of population levels of immunity to malaria as a result of exposure-reducing interventions: consequences for interpretation of disease trends. PLoS ONE. 2009;4:e4383.
    1. Rutta ASM, Francis F, Mmbando BP, Ishengoma DS, Sembuche SH, Malecela EK, et al. Using community-owned resource persons to provide early diagnosis and treatment and estimate malaria burden at community level in north-eastern Tanzania. Malar J. 2012;11:152.
    1. Duraisingh MT, Cowman AF. Contribution of the pfmdr1 gene to antimalarial drug-resistance. Acta Trop. 2005;94:181–190.
    1. Dini S, Zaloumis S, Cao P, Price RN, Fowkes FJI, van der Pluijm RW, et al. Investigating the efficacy of triple artemisinin-based combination therapies for treating Plasmodium falciparum malaria patients using mathematical model. Antimicrob Agents Chemother. 2018;62:e01068-18.

Source: PubMed

3
Prenumerera