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

      Impact of piperaquine resistance in Plasmodium falciparum on malaria treatment effectiveness in The Guianas: a descriptive epidemiological study

      research-article
      , PhD a , , MD b , c , , PhD d , e , , MSc a , , PhD a , , PhD a , f , , PhD g , h , , PharmD a , , PharmD i , , MD b , , PhD j , , MD g , k , , PhD l , , MD m , n , , PhD o , , Prof, MD p , q , , PhD r , s , , Prof, MD r , t , , MD u , , MSc v , , MD c , , Prof, MD w , , PhD d , e , , MD x , , PharmD a , *
      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

          Plasmodium falciparum is an apicomplexan parasite responsible for lethal cases of malaria. According to WHO recommendations, P falciparum cases are treated with artemisinin-based combination therapy including dihydroartemisinin–piperaquine. However, the emergence of resistant parasites against dihydroartemisinin–piperaquine was reported in southeast Asia in 2008 and, a few years later, suspected in South America.

          Methods

          To characterise resistance emergence, a treatment efficacy study was performed on the reported patients infected with P falciparum and treated with dihydroartemisinin–piperaquine in French Guiana (n=6, 2016–18). Contemporary isolates collected in French Guiana were genotyped for P falciparum chloroquine resistance transporter ( pfCRT; n=845) and pfpm2 and pfpm3 copy number (n=231), phenotyped using the in vitro piperaquine survival assay (n=86), and analysed through genomic studies (n=50). Additional samples from five Amazonian countries and one outside the region were genotyped (n=1440).

          Findings

          In field isolates, 40 (47%) of 86 (95% CI 35·9–57·1) were resistant to piperaquine in vitro; these phenotypes were more associated with pfCRT C350R (ie, Cys350Arg) and pfpm2 and pfpm3 amplifications (Dunn test, p<0·001). Those markers were also associated with dihydroartemisinin–piperaquine treatment failure (n=3 [50%] of 6). A high prevalence of piperaquine resistance markers was observed in Suriname in 19 (83%) of 35 isolates and in Guyana in 579 (73%) of 791 isolates. The pfCRT C350R mutation emerged before pfpm2 and pfpm3 amplification in a temporal sequence different from southeast Asia, and in the absence of artemisinin partial resistance, suggesting a geographically distinctive epistatic relationship between these genetic markers.

          Interpretation

          The high prevalence of piperaquine resistance markers in parasite populations of the Guianas, and the risk of associated therapeutic failures calls for caution on dihydroartemisinin–piperaquine use in the region. Furthermore, greater attention should be given to potential differences in genotype to phenotype mapping across genetically distinct parasite populations from different continents.

          Funding

          Pan American Health Organization and WHO, French Ministry for Research, European Commission, Santé publique France, Agence Nationale de la Recherche, Fundação de Amparo à Pesquisa do Estado do Amazonas, Ministry of Health of Brazil, Oswaldo Cruz Foundation, and National Institutes of Health.

          Translations

          For the French and Portuguese translations of the abstract see Supplementary Materials section.

          Related collections

          Most cited references29

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

          Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study

          Summary Background Artemisinin-resistant falciparum malaria has arisen in western Cambodia. A concerted international effort is underway to contain artemisinin-resistant Plasmodium falciparum, but containment strategies are dependent on whether resistance has emerged elsewhere. We aimed to establish whether artemisinin resistance has spread or emerged on the Thailand–Myanmar (Burma) border. Methods In malaria clinics located along the northwestern border of Thailand, we measured six hourly parasite counts in patients with uncomplicated hyperparasitaemic falciparum malaria (≥4% infected red blood cells) who had been given various oral artesunate-containing regimens since 2001. Parasite clearance half-lives were estimated and parasites were genotyped for 93 single nucleotide polymorphisms. Findings 3202 patients were studied between 2001 and 2010. Parasite clearance half-lives lengthened from a geometric mean of 2·6 h (95% CI 2·5–2·7) in 2001, to 3·7 h (3·6–3·8) in 2010, compared with a mean of 5·5 h (5·2–5·9) in 119 patients in western Cambodia measured between 2007 and 2010. The proportion of slow-clearing infections (half-life ≥6·2 h) increased from 0·6% in 2001, to 20% in 2010, compared with 42% in western Cambodia between 2007 and 2010. Of 1583 infections genotyped, 148 multilocus parasite genotypes were identified, each of which infected between two and 13 patients. The proportion of variation in parasite clearance attributable to parasite genetics increased from 30% between 2001 and 2004, to 66% between 2007 and 2010. Interpretation Genetically determined artemisinin resistance in P falciparum emerged along the Thailand–Myanmar border at least 8 years ago and has since increased substantially. At this rate of increase, resistance will reach rates reported in western Cambodia in 2–6 years. Funding The Wellcome Trust and National Institutes of Health.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Determinants of dihydroartemisinin-piperaquine treatment failure in Plasmodium falciparum malaria in Cambodia, Thailand, and Vietnam: a prospective clinical, pharmacological, and genetic study

            Summary Background The emergence and spread of resistance in Plasmodium falciparum malaria to artemisinin combination therapies in the Greater Mekong subregion poses a major threat to malaria control and elimination. The current study is part of a multi-country, open-label, randomised clinical trial (TRACII, 2015–18) evaluating the efficacy, safety, and tolerability of triple artemisinin combination therapies. A very high rate of treatment failure after treatment with dihydroartemisinin-piperaquine was observed in Thailand, Cambodia, and Vietnam. The immediate public health importance of our findings prompted us to report the efficacy data on dihydroartemisinin-piperaquine and its determinants ahead of the results of the overall trial, which will be published later this year. Methods Patients aged between 2 and 65 years presenting with uncomplicated P falciparum or mixed species malaria at seven sites in Thailand, Cambodia, and Vietnam were randomly assigned to receive dihydroartemisinin-piperaquine with or without mefloquine, as part of the TRACII trial. The primary outcome was the PCR-corrected efficacy at day 42. Next-generation sequencing was used to assess the prevalence of molecular markers associated with artemisinin resistance (kelch13 mutations, in particular Cys580Tyr) and piperaquine resistance (plasmepsin-2 and plasmepsin-3 amplifications and crt mutations). This study is registered with ClinicalTrials.gov, number NCT02453308. Findings Between Sept 28, 2015, and Jan 18, 2018, 539 patients with acute P falciparum malaria were screened for eligibility, 292 were enrolled, and 140 received dihydroartemisinin-piperaquine. The overall Kaplan-Meier estimate of PCR-corrected efficacy of dihydroartemisinin-piperaquine at day 42 was 50·0% (95% CI 41·1–58·3). PCR-corrected efficacies for individual sites were 12·7% (2·2–33·0) in northeastern Thailand, 38·2% (15·9–60·5) in western Cambodia, 73·4% (57·0–84·3) in Ratanakiri (northeastern Cambodia), and 47·1% (33·5–59·6) in Binh Phuoc (southwestern Vietnam). Treatment failure was associated independently with plasmepsin2/3 amplification status and four mutations in the crt gene (Thr93Ser, His97Tyr, Phe145Ile, and Ile218Phe). Compared with the results of our previous TRACI trial in 2011–13, the prevalence of molecular markers of artemisinin resistance (kelch13 Cys580Tyr mutations) and piperaquine resistance (plasmepsin2/3 amplifications and crt mutations) has increased substantially in the Greater Mekong subregion in the past decade. Interpretation Dihydroartemisinin-piperaquine is not treating malaria effectively across the eastern Greater Mekong subregion. A highly drug-resistant P falciparum co-lineage is evolving, acquiring new resistance mechanisms, and spreading. Accelerated elimination of P falciparum malaria in this region is needed urgently, to prevent further spread and avoid a potential global health emergency. Funding UK Department for International Development, Wellcome Trust, Bill & Melinda Gates Foundation, Medical Research Council, and National Institutes of Health.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Soft sweeps II--molecular population genetics of adaptation from recurrent mutation or migration.

              In the classical model of molecular adaptation, a favored allele derives from a single mutational origin. This ignores that beneficial alleles can enter a population recurrently, either by mutation or migration, during the selective phase. In this case, descendants of several of these independent origins may contribute to the fixation. As a consequence, all ancestral haplotypes that are linked to any of these copies will be retained in the population, affecting the pattern of a selective sweep on linked neutral variation. In this study, we use analytical calculations based on coalescent theory and computer simulations to analyze molecular adaptation from recurrent mutation or migration. Under the assumption of complete linkage, we derive a robust analytical approximation for the number of ancestral haplotypes and their distribution in a sample from the population. We find that so-called "soft sweeps," where multiple ancestral haplotypes appear in a sample, are likely for biologically realistic values of mutation or migration rates.
                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 February 2024
                February 2024
                : 24
                : 2
                : 161-171
                Affiliations
                [a ]Laboratoire de parasitologie, World Health Organization Collaborating Center for Surveillance of Antimalarial Drug Resistance, Center Nationale de Référence du Paludisme, Institut Pasteur de la Guyane, Cayenne, French Guiana
                [b ]Service de Santé des Armées (SSA), Centre d'Epidémiologie et de Santé Publique des Armées (CESPA), Marseille, France
                [c ]Sciences Economiques Sociales de la Santé & Traitement de l'Information Médicale (SESSTIM), Aix Marseille University, INSERM, IRD, Marseille, France
                [d ]Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
                [e ]Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
                [f ]Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
                [g ]Diretoria de Ensino e Pesquisa, Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, Brazil
                [h ]Escola de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil
                [i ]Unité de développements analytiques et bioanalyse, Institut de recherche biomédicale des armées, Brétigny-sur-Orge, France
                [j ]Amazonic Center for Research and Control of Tropical Diseases “Simón Bolívar”, Puerto Ayacucho, Venezuela
                [k ]Instituto Leônidas & Maria Deane, Fiocruz, Manaus, Brazil
                [l ]Laboratory of Basic Research in Malaria, Evandro Chagas Institute, Brazil Ministry of Health, Ananindeua, Brazil
                [m ]Malaria Vaccine and Drug Development Center, Cali, Colombia
                [n ]Caucaseco Scientific Research Center, Cali, Colombia
                [o ]Department of Biochemistry Kernkampweg 5, Faculty of Medical Sciences, Anton de Kom Universiteit van Suriname, Paramaribo, Suriname
                [p ]Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
                [q ]Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, Nova University of Lisbon, Lisbon, Portugal
                [r ]Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
                [s ]Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
                [t ]Ministry of Public Health, Kabul, Afghanistan
                [u ]National Malaria Program, Ministry of Health, Georgetown, Guyana
                [v ]Department of Communicable Diseases and Environmental Determinants of Health, Pan American Health Organization/World Health Organization, Washington DC, USA
                [w ]Infectious and Tropical Diseases Unit, Cayenne General Hospital, Cayenne, French Guiana
                [x ]Global Malaria Programme, World Health Organization, Geneva, Switzerland
                Author notes
                [* ]Correspondence to: Dr L Musset, Laboratoire de parasitologie, World Health Organization Collaborating Center for Surveillance of Antimalarial Drug Resistance, Center Nationale de Référence du Paludisme, Institut Pasteur de la Guyane, Cayenne 97306, French Guiana lisemusset@ 123456gmail.com
                Article
                S1473-3099(23)00502-9
                10.1016/S1473-3099(23)00502-9
                10808503
                37858325
                cf4080e8-721a-43a7-8cd3-8d5ce767778a
                © 2024 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license

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

                History
                Categories
                Articles

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article