30
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Molecular epidemiology of resistance to antimalarial drugs in the Greater Mekong subregion: an observational study

      research-article
      , Prof, PhD a , b , * , , PhD b , d , , MD e , , MD f , , PhD f , g , , MSc f , , BSc b , , MSc b , , PhD b , , MSc b , , MSc d , , MSc d , , PhD h , , PhD i , , MSc h , , PhD j , , PhD k , , MSc l , , MMedSc m , , BS m , , MD m , , BS n , , Prof, DrPH o , , MD b , p , , MD q , r , , PhD s , , MD r , , MD b , p , , PhD b , p , , MD b , p , , MD t , , MD t , , MRCP b , p , , MD t , , PhD t , , MD t , , Prof, FRS u , v , , Prof, PhD b , u , v , , BS p , r , , PhD w , , Prof, DPhil b , c , x , , Prof, FMedSci b , p , , Prof, MD b , p , , MD g , p , y , , Prof, MD f , p , , Prof, FRS b , p
      The Lancet. Infectious Diseases
      Elsevier Science ;, The Lancet Pub. Group

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          Background

          The Greater Mekong subregion is a recurrent source of antimalarial drug resistance in Plasmodium falciparum malaria. This study aimed to characterise the extent and spread of resistance across this entire region between 2007 and 2018.

          Methods

          P falciparum isolates from Myanmar, Thailand, Laos, and Cambodia were obtained from clinical trials and epidemiological studies done between Jan 1, 2007, and Dec 31, 2018, and were genotyped for molecular markers ( pfkelch, pfcrt, pfplasmepsin2, and pfmdr1) of antimalarial drug resistance. Genetic relatedness was assessed using microsatellite and single nucleotide polymorphism typing of flanking sequences around target genes.

          Findings

          10 632 isolates were genotyped. A single long pfkelch Cys580Tyr haplotype (from −50 kb to +31·5 kb) conferring artemisinin resistance (PfPailin) now dominates across the eastern Greater Mekong subregion. Piperaquine resistance associated with pfplasmepsin2 gene amplification and mutations in pfcrt downstream of the Lys76Thr chloroquine resistance locus has also developed. On the Thailand–Myanmar border a different pfkelch Cys580Tyr lineage rose to high frequencies before it was eliminated. Elsewhere in Myanmar the Cys580Tyr allele remains widespread at low allele frequencies. Meanwhile a single artemisinin-resistant pfkelch Phe446Ile haplotype has spread across Myanmar. Despite intense use of dihydroartemisinin–piperaquine in Kayin state, eastern Myanmar, both in treatment and mass drug administrations, no selection of piperaquine resistance markers was observed. pfmdr1 amplification, a marker of resistance to mefloquine, remains at low prevalence across the entire region.

          Interpretation

          Artemisinin resistance in P falciparum is now prevalent across the Greater Mekong subregion. In the eastern Greater Mekong subregion a multidrug resistant P falciparum lineage (PfPailin) dominates. In Myanmar a long pfkelch Phe446Ile haplotype has spread widely but, by contrast with the eastern Greater Mekong subregion, there is no indication of artemisinin combination therapy (ACT) partner drug resistance from genotyping known markers, and no evidence of spread of ACT resistant P falciparum from the east to the west. There is still a window of opportunity to prevent global spread of ACT resistance.

          Funding

          Thailand Science Research and Innovation, Initiative 5%, Expertise France, Wellcome Trust.

          Related collections

          Most cited references35

          • Record: found
          • Abstract: found
          • Article: not found

          A molecular marker of artemisinin-resistant Plasmodium falciparum malaria.

          Plasmodium falciparum resistance to artemisinin derivatives in southeast Asia threatens malaria control and elimination activities worldwide. To monitor the spread of artemisinin resistance, a molecular marker is urgently needed. Here, using whole-genome sequencing of an artemisinin-resistant parasite line from Africa and clinical parasite isolates from Cambodia, we associate mutations in the PF3D7_1343700 kelch propeller domain ('K13-propeller') with artemisinin resistance in vitro and in vivo. Mutant K13-propeller alleles cluster in Cambodian provinces where resistance is prevalent, and the increasing frequency of a dominant mutant K13-propeller allele correlates with the recent spread of resistance in western Cambodia. Strong correlations between the presence of a mutant allele, in vitro parasite survival rates and in vivo parasite clearance rates indicate that K13-propeller mutations are important determinants of artemisinin resistance. K13-propeller polymorphism constitutes a useful molecular marker for large-scale surveillance efforts to contain artemisinin resistance in the Greater Mekong Subregion and prevent its global spread.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Artemisinin resistance in Plasmodium falciparum malaria.

            Artemisinin-based combination therapies are the recommended first-line treatments of falciparum malaria in all countries with endemic disease. There are recent concerns that the efficacy of such therapies has declined on the Thai-Cambodian border, historically a site of emerging antimalarial-drug resistance. In two open-label, randomized trials, we compared the efficacies of two treatments for uncomplicated falciparum malaria in Pailin, western Cambodia, and Wang Pha, northwestern Thailand: oral artesunate given at a dose of 2 mg per kilogram of body weight per day, for 7 days, and artesunate given at a dose of 4 mg per kilogram per day, for 3 days, followed by mefloquine at two doses totaling 25 mg per kilogram. We assessed in vitro and in vivo Plasmodium falciparum susceptibility, artesunate pharmacokinetics, and molecular markers of resistance. We studied 40 patients in each of the two locations. The overall median parasite clearance times were 84 hours (interquartile range, 60 to 96) in Pailin and 48 hours (interquartile range, 36 to 66) in Wang Pha (P<0.001). Recrudescence confirmed by means of polymerase-chain-reaction assay occurred in 6 of 20 patients (30%) receiving artesunate monotherapy and 1 of 20 (5%) receiving artesunate-mefloquine therapy in Pailin, as compared with 2 of 20 (10%) and 1 of 20 (5%), respectively, in Wang Pha (P=0.31). These markedly different parasitologic responses were not explained by differences in age, artesunate or dihydroartemisinin pharmacokinetics, results of isotopic in vitro sensitivity tests, or putative molecular correlates of P. falciparum drug resistance (mutations or amplifications of the gene encoding a multidrug resistance protein [PfMDR1] or mutations in the gene encoding sarco-endoplasmic reticulum calcium ATPase6 [PfSERCA]). Adverse events were mild and did not differ significantly between the two treatment groups. P. falciparum has reduced in vivo susceptibility to artesunate in western Cambodia as compared with northwestern Thailand. Resistance is characterized by slow parasite clearance in vivo without corresponding reductions on conventional in vitro susceptibility testing. Containment measures are urgently needed. (ClinicalTrials.gov number, NCT00493363, and Current Controlled Trials number, ISRCTN64835265.) 2009 Massachusetts Medical Society
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Spread of artemisinin resistance in Plasmodium falciparum malaria.

              Artemisinin resistance in Plasmodium falciparum has emerged in Southeast Asia and now poses a threat to the control and elimination of malaria. Mapping the geographic extent of resistance is essential for planning containment and elimination strategies. Between May 2011 and April 2013, we enrolled 1241 adults and children with acute, uncomplicated falciparum malaria in an open-label trial at 15 sites in 10 countries (7 in Asia and 3 in Africa). Patients received artesunate, administered orally at a daily dose of either 2 mg per kilogram of body weight per day or 4 mg per kilogram, for 3 days, followed by a standard 3-day course of artemisinin-based combination therapy. Parasite counts in peripheral-blood samples were measured every 6 hours, and the parasite clearance half-lives were determined. The median parasite clearance half-lives ranged from 1.9 hours in the Democratic Republic of Congo to 7.0 hours at the Thailand-Cambodia border. Slowly clearing infections (parasite clearance half-life >5 hours), strongly associated with single point mutations in the "propeller" region of the P. falciparum kelch protein gene on chromosome 13 (kelch13), were detected throughout mainland Southeast Asia from southern Vietnam to central Myanmar. The incidence of pretreatment and post-treatment gametocytemia was higher among patients with slow parasite clearance, suggesting greater potential for transmission. In western Cambodia, where artemisinin-based combination therapies are failing, the 6-day course of antimalarial therapy was associated with a cure rate of 97.7% (95% confidence interval, 90.9 to 99.4) at 42 days. Artemisinin resistance to P. falciparum, which is now prevalent across mainland Southeast Asia, is associated with mutations in kelch13. Prolonged courses of artemisinin-based combination therapies are currently efficacious in areas where standard 3-day treatments are failing. (Funded by the U.K. Department of International Development and others; ClinicalTrials.gov number, NCT01350856.).
                Bookmark

                Author and article information

                Contributors
                Journal
                Lancet Infect Dis
                Lancet Infect Dis
                The Lancet. Infectious Diseases
                Elsevier Science ;, The Lancet Pub. Group
                1473-3099
                1474-4457
                1 December 2020
                December 2020
                : 20
                : 12
                : 1470-1480
                Affiliations
                [a ]Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
                [b ]Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
                [c ]Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
                [d ]Worldwide Antimalarial Resistance Network, Bangkok, Thailand
                [e ]Department of Preventive and Social Medicine, Defence Services Medical Academy, Yangon, Myanmar
                [f ]Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
                [g ]Myanmar Oxford Clinical Research Unit, Yangon, Myanmar
                [h ]Bureau of Vector-borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
                [i ]Office of Disease Prevention and Control Region 1, Chiang Mai, Thailand
                [j ]Faculty of Medical Technology, Prince of Songkla University, Songkhla, Thailand
                [k ]Department of Microbiology, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
                [l ]Buntharik Hospital, Amphoe Buntharik, Ubon Ratchathani, Thailand
                [m ]Department of Medical Research, Ministry of Health and Sports, Yangon, Myanmar
                [n ]Department of Tropical and Infectious Diseases, University of Medicine 1, Yangon, Myanmar
                [o ]Defence Services Medical Research Centre, Naypyitaw, Myanmar
                [p ]Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
                [q ]Institute of Research and Education Development, University of Health Sciences, Ministry of Health, Vientiane, Laos
                [r ]Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Vientiane, Laos
                [s ]Savannakhet Provincial Health Department, Phonsavangnuea village, Kaysone-Phomvihan district, Savannakhet, Laos
                [t ]National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
                [u ]Wellcome Sanger Institute, Hinxton, UK
                [v ]Medical Research Council Centre for Genomics and Global Health, Big Data Institute, University of Oxford, Oxford, UK
                [w ]Department of Medical Research, Myanmar Health Network Organization, Yangon, Myanmar
                [x ]The Royal Society of Thailand, Dusit, Bangkok, Thailand
                [y ]Medical Action Myanmar, Yangon, Myanmar
                Author notes
                [* ]Correspondence to: Prof Mallika Imwong, Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand mallika.imw@ 123456mahidol.ac.th
                Article
                S1473-3099(20)30228-0
                10.1016/S1473-3099(20)30228-0
                7689289
                32679084
                85198f60-68bf-4acf-9066-02d154b2b30a
                © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                Categories
                Articles

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article