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

Richard Mwaiswelo, Billy Ngasala, Irina Jovel, Weiping Xu, Erik Larsson, Maja Malmberg, Jose Pedro Gil, Zul Premji, Bruno P Mmbando, Andreas Mårtensson, Richard Mwaiswelo, Billy Ngasala, Irina Jovel, Weiping Xu, Erik Larsson, Maja Malmberg, Jose Pedro Gil, Zul Premji, Bruno P Mmbando, Andreas Mårtensson

Abstract

Prevalence of and risk factors associated with polymerase chain reaction (PCR)-determined Plasmodium falciparum positivity were assessed on day 3 after initiation of treatment, pre-implementation and up to 8 years post-deployment of artemether-lumefantrine as first-line treatment for uncomplicated malaria in Bagamoyo district, Tanzania. Samples originated from previously reported trials conducted between 2006 and 2014. Cytochrome b-nested PCR was used to detect malaria parasites from blood samples collected on a filter paper on day 3. Chi-square and McNemar chi-squared tests, logistic regression models, and analysis of variance were used as appropriate. Primary outcome was based on the proportion of patients with day 3 PCR-determined P. falciparum positivity. Overall, 256/584 (43.8%) of screened patients had day 3 PCR-determined positivity, whereas only 2/584 (0.3%) had microscopy-determined asexual parasitemia. Day 3 PCR-determined positivity increased from 28.0% (14/50) in 2006 to 74.2% (132/178) in 2007-2008 and declined, thereafter, to 36.0% (50/139) in 2012-2013 and 27.6% (60/217) in 2014. When data were pooled, pretreatment microscopy-determined asexual parasitemia ≥ 100,000/µL, hemoglobin < 10 g/dL, age < 5 years, temperature ≥ 37.5°C, and year of study 2007-2008 and 2012-2013 were significantly associated with PCR-determined positivity on day 3. Significant increases in P. falciparum multidrug resistance gene 1 N86 and P. falciparum chloroquine resistant transporter K76 across years were not associated with PCR-determined positivity on day 3. No statistically significant association was observed between day 3 PCR-determined positivity and PCR-adjusted recrudescence. Day 3 PCR-determined P. falciparum positivity remained common in patients treated before and after implementation of artemether-lumefantrine in Bagamoyo district, Tanzania. However, its presence was associated with pretreatment characteristics. Trials registration numbers: NCT00336375, ISRCTN69189899, NCT01998295, and NCT02090036.

Figures

Figure 1.
Figure 1.
Distribution of patients with PCR-determined parasitemia on day 3 from 2006 to 2014. (A) All ages, (B) Patients < 5 years, (C) Patients > 5 years.
Figure 2.
Figure 2.
Distribution of Pfmdr1 N86Y (A) and Pfcrt K76T (B) on day 3 across years of study.

References

    1. World Health Organization , 2014. World Malaria Report 2014. Geneva, Switzerland: WHO.
    1. World Health Organization , 2015. World Malaria Report 2015. Geneva, Switzerland: WHO.
    1. White NJ, 2008. Qinghaosu (artemisinin): the price of success. Science 320: 330–334.
    1. Dondorp AM, et al. 2009. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 361: 455–467.
    1. World Health Organization , 2016. Artemisinin and Artemisinin-Based Combination Therapy Resistance. Geneva, Switzerland: WHO.
    1. Ariey F, et al. 2014. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 505: 50–55.
    1. Noedl H, et al. 2010. Artemisinin resistance in Cambodia: a clinical trial designed to address an emerging problem in southeast Asia. Clin Infect Dis 51: e82–e89.
    1. Saunders DL, Vanachayangkul P, Lon C, 2014. Dihydroartemisinin-piperaquine failure in Cambodia. N Engl J Med 371: 484–485.
    1. Lu F, et al. 2017. Emergence of indigenous artemisinin-resistant Plasmodium falciparum in Africa. N Engl J Med 376: 991–993.
    1. Ajayi NA, Ukwaja KN, 2013. Possible artemisinin-based combination therapy-resistant malaria in Nigeria: a report of three cases. Rev da Soc Bras Med Trop 46: 525–527.
    1. Taylor SM, et al. 2014. Absence of putative artemisinin resistance mutations among Plasmodium falciparum in sub-Saharan Africa: a molecular epidemiologic study. J Infect Dis 211: 680–688.
    1. Kamau E, et al. 2015. K13-propeller polymorphisms in Plasmodium falciparum parasites from sub-Saharan Africa. J Infect Dis 211: 1352–1355.
    1. Ashley E, et al. 2014. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 371: 411–423.
    1. Maiga AW, et al. 2012. No evidence of delayed parasite clearance after oral artesunate treatment of uncomplicated falciparum malaria in Mali. Am J Trop Med Hyg 87: 23–28.
    1. Mwaiswelo R, Ngasala B, Gil JP, Malmberg M, Jovel I, Xu W, Premji Z, Mmbando BP, Bjorkman A, Mårtensson A, 2017. 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 97: 526–532.
    1. Beshir KB, et al. 2013. Residual Plasmodium falciparum parasitemia in Kenyan children after artemisinin-combination therapy is associated with increased transmission to mosquitoes and parasite recurrence. J Infect Dis 208: 2017–2024.
    1. Kiaco K, Teixeira J, Machado M, do Rosário V, Lopes D, 2015. Evaluation of artemether-lumefantrine efficacy in the treatment of uncomplicated malaria and its association with pfmdr1, pfatpase6 and K13-propeller polymorphisms in Luanda, Angola. Malar J 14: 504.
    1. Djimdé A, et al. 2003. Clearance of drug-resistant parasites as a model for protective immunity in Plasmodium falciparum malaria. Am J Trop Med Hyg 69: 558–563.
    1. Zwang J, Dorsey G, Mårtensson A, d’Alessandro U, Ndiaye JL, Karema C, Djimde A, Brasseur P, Olliaro P, 2014. Plasmodium falciparum clearance in clinical studies of artesunate-amodiaquine and comparator treatments in sub-Saharan Africa, 1999–2009. Malar J 13: 114.
    1. WWARN Artemisinin based Combination Therapy (ACT) Africa Baseline Study Group Dahal P, d’Alessandro U, Dorsey G, Guerin PJ, Nsanzabana C, Price RN, Sibley CH, Stepniewska K, Talisuna AO, 2015. Clinical determinants of early parasitological response to ACTs in African patients with uncomplicated falciparum malaria: a literature review and meta-analysis of individual patient data. BMC Med 13: 212.
    1. White NJ, 2011. The parasite clearance curve. Malar J 10: 278.
    1. Carlsson AM, et al. 2011. Plasmodium falciparum population dynamics during the early phase of anti-malarial drug treatment in Tanzanian children with acute uncomplicated malaria. Malar J 10: 380.
    1. Ngasala BE, et al. 2011. Efficacy and effectiveness of artemether-lumefantrine after initial and repeated treatment in children < 5 years of age with acute uncomplicated Plasmodium falciparum malaria in rural Tanzania: a randomized trial. Clin Infect Dis 52: 873–882.
    1. Mwaiswelo R, Ngasala BE, Jovel I, Gosling R, Premji Z, Poirot E, Mmbando BP, Björkman A, Mårtensson A, 2016. 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 15: 316.
    1. Gething PW, Patil AP, Smith DL, Guerra C, Elyazar IRF, Johnston GL, Tatem AJ, Hay SI, 2011. A new world malaria map: Plasmodium falciparum endemicity in 2010. Malar J 10: 378.
    1. National Malaria Control Programme , 2006. National Guidelines for Diagnosis and Treatment of Malaria. Dar es Salaam, United Republic of Tanzania NMCP.
    1. National Malaria Control Programme , 2010. Tanzania Malaria Programme Review 2010. Programme Review Proposal. Dar es Salaam, United Republic of Tanzania NMCP.
    1. Mwaiswelo R, Ngasala BE, Jovel IT, Aydin-schmidt B, Gosling R, Premji Z, Mmbando B, Björkman A, Mårtensson A, 2016. 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 15: 435.
    1. World Health Organization , 2009. Methods for Surveillance of Antimalarial Drug Efficacy. Geneva, Switzerland: WHO.
    1. Hsiang MS, Lin MS, Dokomajilar M, Kemere J, Pilcher CD, Greenhouse B, 2010. PCR-based pooling of dried blood spots for detection of malaria parasites: optimization and application to a cohort. J Clin Microbiol 48: 3539–3543.
    1. Morris U, et al. 2013. Rapid diagnostic tests for molecular surveillance of Plasmodium falciparum malaria -assessment of DNA extraction methods and field applicability. Malar J 12: 106.
    1. Steenkeste N, Incardona S, Chy S, Duval L, Ekala M-T, Lim P, Hewitt S, Sochantha T, Socheat D, Rogier C, 2009. Towards high-throughput molecular detection of Plasmodium: new approaches and molecular markers. Malar J 8: 86.
    1. Djimdé A, et al. 2001. A molecular marker for chloroquine-resistant falciparum malaria. N Engl J Med 344: 257–263.
    1. Snounou G, Zhu X, Siripoon N, Jarra W, Thaithong S, Brown K, Viriyakosol S, 1999. Biased distribution of mspl populations in Thailand and msp2 allelic variants in Plasmodium falciparum. Trans R Soc Trop Med Hyg 2: 369–374.
    1. White NJ, 2017. Malaria parasite clearance. Malar J 16: 88.
    1. Le Manach C, et al. 2013. Fast in vitro methods to determine the speed of action and the stage-specificity of anti-malarials in Plasmodium falciparum. Malar J 12: 424.
    1. White NJ, Van Vugt M, Ezzet F, 1999. Clinical pharmacokinetics and pharmacodynamics of artemether-lumefantrine. Clin Pharmacokinet 37: 105–125.
    1. Lopera-Mesa TM, et al. 2013. Plasmodium falciparum clearance rates in response to artesunate in Malian children with malaria: effect of acquired immunity. J Infect Dis 207: 1655–1663.
    1. Muhindo MK, et al. 2014. Early parasite clearance following artemisinin- based combination therapy among Ugandan children with uncomplicated Plasmodium falciparum malaria. Malar J 13: 32.
    1. Malmberg M, et al. 2013. Temporal trends of molecular markers associated with artemether-lumefantrine tolerance/resistance in Bagamoyo district, Tanzania. Malar J 12: 103.
    1. Okombo J, Kamau AW, Marsh K, Sutherland CJ, Ochola-oyier LI, 2014. Temporal trends in prevalence of Plasmodium falciparum drug resistance alleles over two decades of changing antimalarial policy in coastal Kenya. Int J Parasitol Drug Resist 4: 152–163.
    1. Sutherland CJ, Alloueche A, Curtis J, Drakeley CJ, Ord R, Duraisingh M, Greenwood BM, Pinder M, Warhurst D, Targett GA, 2002. Gambian children successfully treated with chloroquine can harbor and transmit Plasmodium falciparum gametocytes carrying resistance genes. Am J Trop Med Hyg 67: 578–585.
    1. Ochong EO, van den Broek IV, Keus K, Nzila A, 2003. Short report: association between chloroquine and amodiaquine resistance and allelic variation in the Plasmodium falciparum multiple drug resistance 1 gene and the chloroquine resistance transporter gene in isolates from the upper Nile in southern Sudan. Am J Trop Med Hyg 69: 184–187.
    1. Humphreys GS, Merinopoulos I, Ahmed J, Whitty CJM, Mutabingwa TK, Sutherland CJ, Hallett RL, 2007. Amodiaquine and artemether-lumefantrine select distinct alleles of the Plasmodium falciparum mdr1 gene in Tanzanian children treated for uncomplicated malaria. Antimicrob Agents Chemother 51: 991–997.
    1. Chang H, et al. 2016. Persistence of Plasmodium falciparum parasitemia after artemisinin combination therapy: evidence from a randomized trial in Uganda. Sci Rep 6: 26330.

Source: PubMed

3
購読する