1
|
White NFD, Whitton G, Wasakul V, Amenga-Etego L, Dara A, Andrianaranjaka V, Randrianarivelojosia M, Miotto O, D'Alessandro U, Djimdé A, Ariani CV, Pearson RD, Amambua-Ngwa A. Disparate co-evolution and prevalence of sulfadoxine and pyrimethamine resistance alleles and haplotypes at dhfr and dhps genes across Africa. Sci Rep 2025; 15:13222. [PMID: 40247054 PMCID: PMC12006366 DOI: 10.1038/s41598-025-98035-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Sulfadoxine-pyrimethamine (SP), despite emergence of mutations in dhfr and dhps genes associated with lower treatment efficacy, is still recommended for preventive malaria treatment. Therefore, it is important to understand the evolution of P. falciparum dhfr and dhps genes. We used the MalariaGEN Pf7 dataset to describe haplotype frequencies across 22 African countries, including changes over time in The Gambia, Mali, Ghana, and Kenya. We show that the triple mutant of dhfr, N51I/C59R/S108N, has remained the dominant haplotype across the continent with limited evidence of additional mutations. There is greater variation for dhps in terms of haplotype diversity and spatial heterogeneity of haplotypes found across Africa. Although samples from Madagascar have low genetic differentiation from samples from mainland East Africa at the whole genome level, we show that dhps K540E is highly differentiated between the two populations, being at very low frequency in Madagascar (4%). Whole genome data reveal 12 SNPs which are also highly differentiated between Madagascar and East Africa, including aat1 and a possible novel drug resistance locus approximately 20 kb 3' of mdr1. We highlight the value of longitudinal sampling and whole genome sequence data for understanding the heterogeneity and ongoing changes in anti-malarial drug resistance genetic markers.
Collapse
Affiliation(s)
- Nina F D White
- Genomic Surveillance Unit, Wellcome Sanger Institute, Hinxton, UK
| | - Georgia Whitton
- Genomic Surveillance Unit, Wellcome Sanger Institute, Hinxton, UK
| | - Varanya Wasakul
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Lucas Amenga-Etego
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali
| | - Antoine Dara
- Malaria Research and Training Centre, Faculty of Pharmacy, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali
| | - Voahangy Andrianaranjaka
- Institut Pasteur de Madagascar, Antananarivo, Madagascar
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali
| | - Milijaona Randrianarivelojosia
- Institut Pasteur de Madagascar, Antananarivo, Madagascar
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali
| | - Olivo Miotto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali
| | - Umberto D'Alessandro
- Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Abdoulaye Djimdé
- Malaria Research and Training Centre, Faculty of Pharmacy, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali
| | | | | | - Alfred Amambua-Ngwa
- Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia.
- Pathogens Genomic Diversity Network Africa (PDNA), Sotuba, Bamako, Mali.
| |
Collapse
|
2
|
Alruwaili M, Elderdery A, Manni E, Mills J. A Narrative Review on the Prevalence of Plasmodium falciparum Resistance Mutations to Antimalarial Drugs in Rwanda. Trop Med Infect Dis 2025; 10:89. [PMID: 40278762 PMCID: PMC12030788 DOI: 10.3390/tropicalmed10040089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/26/2025] Open
Abstract
Malaria has been and remains a significant challenge in Africa and other endemic settings. Roughly, 95% of global morbidity and mortality due to malaria occurs within African populations and affects millions of individuals, especially those living in sub-Saharan countries, predominantly due to disease complications. Cultural factors such as unawareness of and disinterest in using recommended preventive tools and combating the primary host (i.e., the female Anopheles mosquito) play a significant role. This host transmits the malaria-causing Plasmodium parasite by biting an infected individual and spreading it to humans. The current overview focuses on the molecular markers associated with antimalarial drug resistance in Plasmodium falciparum (P. falciparum) in Rwanda, considered an exemplar of sub-Saharan countries where malaria is prevalent and effective policies on the development of malaria treatment, approved recently by WHO in 2025, have been adopted. The prevalence of mutations in key resistance genes, including pfcrt, pfmdr1, and pfdhfr/pfdhps, are linked to resistance against common antimalarial drugs such as chloroquine and sulfadoxine-pyrimethamine (SP). In addition, the Plasmodium falciparum kelch13 (pfk13) gene is linked to resistance against artemisinin, as its mutations can cause delayed parasite clearance and treatment failure. Despite changes in therapeutic use policies owing to high prevalence of variant alleles, which reduce the drug's efficacy resistance to SP, the gene persists in Rwanda. Malaria parasites are becoming more resistant to chloroquine, leading to diminished effectiveness and slower recovery or treatment failure. Surveillance data reported from several studies provide crucial insights into the evolving trends of resistance markers and are vital for guiding treatment protocols and informing therapeutic use policy decisions. It is important that we continue to maintain and develop the effectiveness of malaria prevention strategies and treatments, due to the multiple types of resistance found in the population.
Collapse
Affiliation(s)
- Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Abozer Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Emad Manni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Jeremy Mills
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK;
| |
Collapse
|
3
|
Abebe W, Mekuanint A, Asmare Z, Woldesenbet D, Mihret Y, Setegn A, Emagneneh T. Prevalence of molecular markers of chloroquine resistance in malaria parasites in East Africa: A systematic review and meta-analysis. J Glob Antimicrob Resist 2025; 41:117-137. [PMID: 39725320 DOI: 10.1016/j.jgar.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 10/23/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Malaria is a serious global public health problem, which is caused by genus Plasmodium. Resistance of the human malaria parasite to antimalarial drugs is a public health concern in malaria endemic countries. Chloroquine is resistant for both P. vivax and P. falciparum. Chloroquine resistance is understood throughout all of Africa's P. falciparum endemic regions. Molecular markers play a crucial role in tracking and understanding the prevalence of antimalarial drug resistance. Currently, there is inadequate information on the prevalence of molecular markers of chloroquine resistance in malaria parasites. OBJECTIVE This systematic review and meta-analysis aimed to determine the pooled prevalence of molecular markers of chloroquine resistance in malaria parasites in East Africa. METHODS Systematic search was performed to retrieve articles from PubMed, Scopus, Science Direct, and the Google Scholar search engine. Twenty potential studies that provided important data on markers of chloroquine resistance in malaria parasites were systematically reviewed and analyzed. Five antimalarial drug resistance markers of chloroquine resistance were extracted separately into Microsoft Excel and analyzed using STATA 17.0. The Inverse of variance (I2) was done to evaluate heterogeneity across studies. The funnel plot and the Egger's test were used to determine the existence or absence of publication bias. A trim-and-fill meta-analysis was carried out to generate a bias-adjusted effect estimate. A random effect model was used to determine the pooled prevalence of molecular markers associated with chloroquine resistance in malaria parasites. Subgroup analysis was performed based on country and year of publication. RESULTS A total of 20 studies were included for this systematic review and meta-analysis. The molecular markers like K76T, 76T, N86Y, Y184F, and 86Y were selected for meta-analysis. From this meta-analysis, the pooled prevalence of K76T, 76T, N86Y, Y184F, and 86Y was 34.5%, 47.3%, 43.8%, 58.3%, and 29.2%, respectively. After adjusting for publication bias, the estimated pooled prevalence of K76T, 76T, N86Y, Y184F, and 86Y were 34.5%, 47.3%, 43.8%, 58.3%, and 29.2%, respectively. Meta-analysis showed a significant difference in all molecular marker prevalence like K76T and 86Y among studies on year of publication except 76T, N86Y, and Y184F. In addition, the meta-analysis showed a significant difference in all molecular marker prevalence like K76T, 76T, N86Y, Y184F, and 86Y among studies at the country level. CONCLUSIONS The findings of this systematic review and meta-analysis concerning the molecular markers of chloroquine resistance of malaria parasites in East Africa revealed a significant prevalence of antimalarial drug resistance markers of chloroquine. As a result, continued surveillance and monitoring of the prevalence of molecular markers of chloroquine resistance, identification and limitation of drug-resistant malaria parasite strains, and development of new antimalarial treatments are required to guide malaria treatment policies, interventions, control, and elimination of malaria worldwide.
Collapse
Affiliation(s)
- Wagaw Abebe
- Department of Medical Laboratory Sciences, College of Health Sciences, Woldia University, Woldia, Ethiopia.
| | - Amare Mekuanint
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Zelalem Asmare
- Department of Medical Laboratory Sciences, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Dagmawi Woldesenbet
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wachemo University, Hossana, Ethiopia
| | - Yenesew Mihret
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Setegn
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tadele Emagneneh
- Department of Midwifery, College of Health Sciences, Woldia University, Woldia, Ethiopia
| |
Collapse
|
4
|
Segovia X, Srivastava B, Serrato-Arroyo S, Guerrero A, Huijben S. Assessing fitness costs in malaria parasites: a comprehensive review and implications for drug resistance management. Malar J 2025; 24:65. [PMID: 40025552 PMCID: PMC11871665 DOI: 10.1186/s12936-025-05286-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 02/10/2025] [Indexed: 03/04/2025] Open
Abstract
Artemisinin-based combination therapy (ACT) remains a broadly effective anti-malarial drug combination, but the emergence of resistance is threatening its effectiveness. Limiting the spread of these drug-resistant parasites and delaying the emergence of resistance in new areas are of high priority. Understanding the evolution of resistance relies on discerning the fitness costs and benefits associated with resistance mutations. If the cost associated with resistance in an untreated host is sufficiently large relative to the benefit of resistance in a treated host, then the spread of resistance can be mitigated by ensuring sufficient hosts free from that active pharmaceutical ingredient. There is no straightforward way to measure these fitness costs, and each approach that has been used has its limitations. Here, the evidence of fitness costs as measured using field data, animal models, and in vitro models is reviewed for three of the main current or past first-line treatments for malaria: chloroquine (CQ), sulfadoxine-pyrimethamine (SP), and artemisinin derivatives (ART). Despite the difficulties of assessing fitness costs, there is a good amount of evidence of fitness costs in drug-resistant Plasmodium falciparum parasites. The most persuasive evidence comes from resistance reversal observed following the cessation of the use of chloroquine. Comparable evidence cannot be obtained for SP- and ART-resistant parasites, due to the absence of complete cessation of these drugs in the field. Data from in vitro and animal models are variable. While fitness costs are often observed, their presence is not universal across all resistant strains. The extent and nature of these fitness costs can vary greatly depending on the specific genetic factors involved and the ecological context in which the parasites evolve. As a result, it is essential to avoid making broad generalizations about the prevalence or impact of fitness costs in drug-resistant malaria parasites. Focusing on fitness costs as a vulnerability in resistant parasites can guide their evolutionary trajectory towards minimizing their fitness. By accurately predicting these costs, efforts to extend the effectiveness of anti-malarials can be enhanced, limiting resistance evolution and advancing malaria control and elimination goals.
Collapse
Affiliation(s)
- Xyonane Segovia
- The Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | | | - Sergio Serrato-Arroyo
- The Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Ashley Guerrero
- The Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Silvie Huijben
- The Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA.
- Computational and Modeling Sciences Center, Simon A. Levin Mathematical, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
5
|
Nuwa A, Baker K, Kajubi R, Nnaji CA, Theiss-Nyland K, Odongo M, Kyagulanyi T, Nabakooza J, Salandini D, Asua V, Nakirunda M, Rassi C, Rutazaana D, Achuma R, Sagaki P, Bwanika JB, Magumba G, Yeka A, Nsobya S, Kamya MR, Tibenderana J, Opigo J. Effectiveness of sulfadoxine-pyrimethamine plus amodiaquine and dihydroartemisinin-piperaquine for seasonal malaria chemoprevention in Uganda: a three-arm, open-label, non-inferiority and superiority, cluster-randomised, controlled trial. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00746-1. [PMID: 39826559 DOI: 10.1016/s1473-3099(24)00746-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/23/2024] [Accepted: 11/06/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Seasonal malaria chemoprevention (SMC) with sulfadoxine-pyrimethamine combined with amodiaquine (SPAQ) effectively protects eligible children from malaria in areas of high and seasonal transmission. However, concerns about parasite resistance to sulfadoxine-pyrimethamine in East and Southern Africa necessitate evaluating alternative drug regimens. This study assessed the effectiveness of SPAQ and dihydroartemisinin-piperaquine for SMC in Uganda. METHODS This three-arm, open-label, non-inferiority and superiority, cluster-randomised, controlled trial was conducted in Karamoja subregion, Uganda, among children aged 3-59 months and 6-59 months for SPAQ and dihydroartemisinin-piperaquine, respectively. Of 427 villages, 380 were randomly assigned (1:1) to the SPAQ group and dihydroartemisinin-piperaquine group, and 47 were assigned to the control group (no SMC). The superiority component compared the SPAQ and dihydroartemisinin-piperaquine groups with the control group, whereas the non-inferiority component compared the dihydroartemisinin-piperaquine group with the SPAQ group. The primary endpoint was confirmed malaria incidence using rapid diagnostic tests or microscopy. Survival analyses were done on an intention-to-treat basis (in all randomised participants), with adjustments made for covariate imbalances at baseline. Additionally, molecular markers associated with resistance to sulfadoxine-pyrimethamine and amodiaquine were analysed on 750 malaria-positive blood samples from children younger than 5 years before and after five SMC cycles. This trial was registered with ClinicalTrials.gov, NCT05323721, and has been completed. FINDINGS During June 18-30, 2022, 3881 children were enrolled; 1755 in SPAQ, 1736 in dihydroartemisinin-piperaquine, and 390 in control villages. Of these children, 3629 were analysed. Incidence rates were 0·90 cases per 100 person-months in the SPAQ group, 0·80 cases per 100 person-months in the dihydroartemisinin-piperaquine group, and 18·26 cases per 100 person-months in the control group. SPAQ and dihydroartemisinin-piperaquine reduced malaria risk by 94% (hazard ratio [HR] 0·06 [95% CI 0·04-0·08]; p<0·001) and 96% (0·04 [0·03-0·06]; p<0·001), respectively. Based on the prespecified non-inferiority margin of 1·4, there was non-inferiority between the protective effectiveness of dihydroartemisinin-piperaquine and that of SPAQ (HR 0·90 [95% CI 0·58-1·39]). Prevalence of mutations linked to moderate (Plasmodium falciparum dihydrofolate reductase [PfDHFR] and P falciparum dihydropteroate synthetase reductase [PfDHPS]) and high (PfDHFR Ile164Leu and PfDHPS Ala581Gly) sulfadoxine-pyrimethamine resistance were more than 88% and less than 5%, respectively. Mutations associated with 4-aminoquinolone resistance (P falciparum multidrug resistance protein-1 [PfMDR1] Asp1246Tyr and PfMDR1 Asn86Tyr) were less than 1%. There was no significant increase in the prevalence of antifolate and artemisinin partial resistance-associated mutations, but a decrease was observed for key aminoquinoline resistance-associated alleles: P falciparum chloroquine resistance transporter protein Lys76Thr, P falciparum multidrug resistance protein Asn86Tyr, and PfMDR1 Asp1246Tyr (p<0·001). No serious or fatal adverse events were reported. INTERPRETATION SPAQ and dihydroartemisinin-piperaquine effectively reduced malaria in children younger than 5 years, with no safety concerns. There was no evidence of resistance selection by SMC. Although these findings support SPAQ-based SMC in Eastern and Southern Africa, ongoing resistance surveillance and efficacy monitoring are essential for sustained impact. FUNDING GiveWell. TRANSLATION For the Swahili translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
| | - Kevin Baker
- Malaria Consortium, London, UK; Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | | | | - Victor Asua
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | | | | | | | | | | | - Adoke Yeka
- Makerere University College of Health Sciences, Kampala, Uganda
| | - Sam Nsobya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Moses R Kamya
- Makerere University College of Health Sciences, Kampala, Uganda; Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | |
Collapse
|
6
|
Alruwaili M, Uwimana A, Sethi R, Murindahabi M, Piercefield E, Umulisa N, Abram A, Eckert E, Munguti K, Mbituyumuremyi A, Gutman JR, Sullivan DJ. Peripheral and Placental Prevalence of Sulfadoxine-Pyrimethamine Resistance Markers in Plasmodium falciparum among Pregnant Women in Southern Province, Rwanda. Am J Trop Med Hyg 2023; 109:1057-1062. [PMID: 37783456 PMCID: PMC10622487 DOI: 10.4269/ajtmh.23-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/22/2023] [Indexed: 10/04/2023] Open
Abstract
Intermittent preventive therapy during pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is recommended in areas of moderate to high malaria transmission intensity. As a result of the increasing prevalence of SP resistance markers, IPTp-SP was withdrawn from Rwanda in 2008. Nonetheless, more recent findings suggest that SP may improve birthweight even in the face of parasite resistance, through alternative mechanisms that are independent of antimalarial effects. The prevalence of single nucleotide polymorphisms in Plasmodium falciparum dihydropteroate synthase (pfdhps) and dihydrofolate reductase (pfdhfr) genes associated with SP resistance among 148 pregnant women from 2016 to 2018 within Rwanda's Southern Province (Huye and Kamonyi districts) was measured using a ligase detection reaction-fluorescent microsphere assay. The frequency of pfdhps K540E, A581G, and the quintuple (pfdhfr N51I + C59R + S108N/pfdhps A437G + K540E) and sextuple (pfdhfr N51I + C59R + S108N/pfdhps A437G + K540E + A581G) mutant genotypes was 90%, 38%, 75%, and 28%, respectively. No significant genotype difference was seen between the two districts, which are approximately 50 km apart. Observed agreements for matched peripheral to placental blood were reported and found to be 207 of 208 (99%) for pfdhfr and 239 of 260 (92%) for pfdhps. The peripheral blood sample did not miss any pfdhfr drug-resistant mutants or pfdhps except at the S436 loci. At this level of the sextuple mutant, the antimalarial efficacy of SP for preventing low birthweight is reduced, although overall SP still exerts a nonmalarial benefit during pregnancy. This study further reveals the need to intensify preventive measures to sustain malaria control in Rwanda to keep the overall incidence of malaria during pregnancy low.
Collapse
Affiliation(s)
- Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Aline Uwimana
- Malaria and Other Parasitic Diseases Division, Rwanda Biomedical Center, Kigali, Rwanda
| | - Reena Sethi
- Maternal and Child Survival Program/Jhpiego, Washington, District of Columbia
| | - Monique Murindahabi
- Malaria and Other Parasitic Diseases Division, Rwanda Biomedical Center, Kigali, Rwanda
- Roll Back Malaria, Ouagadougou, Burkina Faso
| | - Emily Piercefield
- U.S. President’s Malaria Initiative, Malaria Branch, U.S. Centers for Disease Control and Prevention, Kigali, Rwanda
| | - Noella Umulisa
- Maternal and Child Survival Program/Jhpiego, Kigali, Rwanda
| | | | - Erin Eckert
- RTI International, Washington, District of Columbia
| | - Kaendi Munguti
- U.S. President’s Malaria Initiative, U.S. Agency for International Development, Kigali, Rwanda
| | | | - Julie R. Gutman
- Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
7
|
Shelus V, Mumbere N, Mulogo EM, Barrington C, Baguma E, Muhindo R, Herrington JE, Emch M, Maman S, Boyce RM. Private sector antimalarial sales a decade after "test and treat": A cross-sectional study of drug shop clients in rural Uganda. Front Public Health 2023; 11:1140405. [PMID: 37056663 PMCID: PMC10089286 DOI: 10.3389/fpubh.2023.1140405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Background The World Health Organization has promoted "test and treat" guidelines for malaria since 2010, recommending all suspected malaria cases be confirmed with a parasitological test, typically a rapid diagnostic test (RDT), prior to treatment with antimalarial medications. However, many fevers at private drug shops in Uganda continue to be treated presumptively as malaria without diagnostic testing. Methods The purpose of this study was to document private sector malaria case management in rural Uganda through a cross-sectional survey of drug shop clients in Bugoye sub-county. Drug shop vendors (n = 46) recorded information about sales interactions with clients reporting fever or requesting antimalarials and collected capillary blood samples from clients who purchased medications without an RDT. We estimated the proportion of clients who purchased an RDT, adhered to the RDT result, and received antimalarials without having laboratory-confirmed malaria. Results Most drug shops were unlicensed (96%) and sold RDTs (98%). Of 934 clients with suspected malaria who visited study drug shops during the data collection period, only 25% bought an RDT. Since some clients reported previous RDTs from the public sector, 40% of clients were aware of their malaria status at the drug shop. Among those with negative tests, 36% still purchased antimalarials. Sixty-five percent of clients who purchased an antimalarial without an RDT subsequently tested negative. Conclusions Despite national guidelines, drug shop clients who purchase antimalarials from drug shops in Bugoye are often not tested to confirm a malaria diagnosis prior to treatment. Most clients treated presumptively with antimalarials did not have malaria. Interventions are needed to improve malaria case management and rational drug use in the private sector.
Collapse
Affiliation(s)
- Victoria Shelus
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nobert Mumbere
- Department of Community Health, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Edgar M. Mulogo
- Department of Community Health, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Clare Barrington
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Emmanuel Baguma
- Department of Community Health, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Rabbison Muhindo
- Department of Community Health, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - James E. Herrington
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael Emch
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Geography, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Suzanne Maman
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ross M. Boyce
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
8
|
Moyeh MN, Fankem SN, Ali IM, Sofeu D, Sandie SM, Njimoh DL, Ghogomu SM, Kimbi HK, Mbacham WF. Current status of 4-aminoquinoline resistance markers 18 years after cessation of chloroquine use for the treatment of uncomplicated falciparum malaria in the littoral coastline region of Cameroon. Pathog Glob Health 2022; 116:509-514. [PMID: 35357271 PMCID: PMC9639544 DOI: 10.1080/20477724.2022.2056674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The onset and rapid spread of chloroquine resistance and the introduction of amodiaquine for the treatment of uncomplicated malaria in Cameroon have influenced the proportion of Plasmodium falciparum sensitive and resistant alleles related to 4-aminoquinoline drugs. This study was undertaken to determine the prevalence of resistance markers to antimalarial 4-aminoquinolines in Douala in the Littoral Region, and Buea in the South West Region in June 2020. Dry blood spots were prepared from malaria microscopy positive cases and used for parasite DNA extraction by chelex-100 method. Plasmodium species identification was carried out by PCR amplification/agarose gel electrophoresis of 18srRNA. The Pfcrt and Pfmdr1 genes were amplified by PCR followed by restriction digestion. The prevalence of single nucleotide polymorphisms (SNPs) was compared between study sites and with previous studies carried out between 2003-2005 and 2009-2011 using the Chi square test. The results showed that Plasmodium falciparum was the dominant species occurring as mono-infections (84.6%). The wild type K76 allele of the Pfcrt gene was found in 74.9% of isolates while the wild N86, Y184 and D1246 alleles of the Pfmdr1 gene were found respectively in 87.2%, 89.6% and 100% of field isolates. The results showed a significant reduction in the mutant alleles compared to results obtained in 2003-2005 and 2009-2013. The KNYD haplotype was observed to be the most prevalent. The results indicated that there is a gradual erosion of the mutant Pfcrt and Pfmdr1 genotype and a gradual return to the sensitive P. falciparum genotype in Cameroon.
Collapse
Affiliation(s)
- Marcel Nyuylam Moyeh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea South West Region, Cameroon
- MARCAD Program, The Biotechnology Centre, University of Yaoundé 1, BP 8094, Yaoundé, Centre Region, Cameroon
- Department of Chemical and Biological Engineering, School of Engineering, University of Bamenda, Bambili, North West Region
| | - Sandra Noukimi Fankem
- Department of Biochemistry and Molecular Biology, University of Buea, Buea South West Region, Cameroon
| | - Innocent Mbulli Ali
- MARCAD Program, The Biotechnology Centre, University of Yaoundé 1, BP 8094, Yaoundé, Centre Region, Cameroon
- Department of Biochemistry, Faculty of Science, BP 67, University of Dschang. West Region, Cameroon
| | - Denis Sofeu
- Department of Biochemistry and Molecular Biology, University of Buea, Buea South West Region, Cameroon
| | - Sorelle Mekachie Sandie
- Department of Biochemistry and Molecular Biology, University of Buea, Buea South West Region, Cameroon
| | - Dieudonne Lemuh Njimoh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea South West Region, Cameroon
| | - Stephen Mbigha Ghogomu
- Department of Biochemistry and Molecular Biology, University of Buea, Buea South West Region, Cameroon
| | - Helen Kuokuo Kimbi
- Department of Zoology and Animal Physiology, University of Buea, Buea South West Region, Cameroon
- Department of Biomedical Sciences, The University of Bamenda, Bambili North West Region, Cameroon
| | - Wilfred Fon Mbacham
- MARCAD Program, The Biotechnology Centre, University of Yaoundé 1, BP 8094, Yaoundé, Centre Region, Cameroon
| |
Collapse
|
9
|
Njiro BJ, Mutagonda RF, Chamani AT, Mwakyandile T, Sabas D, Bwire GM. Molecular surveillance of chloroquine-resistant Plasmodium falciparum in sub-Saharan African countries after withdrawal of chloroquine for treatment of uncomplicated malaria: a systematic review. J Infect Public Health 2022; 15:550-557. [DOI: 10.1016/j.jiph.2022.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 03/14/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022] Open
|
10
|
Omedo I, Bartilol B, Kimani D, Gonçalves S, Drury E, Rono MK, Abdi AI, Almagro-Garcia J, Amato R, Pearson RD, Ochola-Oyier LI, Kwiatkowski D, Bejon P. Spatio-temporal distribution of antimalarial drug resistant gene mutations in a Plasmodium falciparum parasite population from Kilifi, Kenya: A 25-year retrospective study. Wellcome Open Res 2022. [DOI: 10.12688/wellcomeopenres.17656.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Antimalarial drug resistance is a major obstacle to sustainable malaria control. Here we use amplicon sequencing to describe molecular markers of drug resistance in Plasmodium falciparum parasites from Kilifi county in the coastal region of Kenya over a 25-year period. Methods: We performed P. falciparum amplicon sequencing on 1162 malaria-infected blood samples collected between 1994 and 2018 to identify markers of antimalarial drug resistance in the Pfcrt, Pfdhfr, Pfdhps, Pfmdr1, Pfexo, Pfkelch13, plasmepsin 2/3, Pfarps10, Pffd, and Pfmdr2 genes. We further interrogated parasite population structure using a genetic barcode of 101 drug resistance-unrelated single nucleotide polymorphisms (SNPs) distributed across the genomes of 1245 P. falciparum parasites. Results: Two major changes occurred in the parasite population over the 25 years studied. In 1994, approximately 75% of parasites carried the marker of chloroquine resistance, CVIET. This increased to 100% in 1999 and then declined steadily, reaching 6.7% in 2018. Conversely, the quintuple mutation form of sulfadoxine-pyrimethamine resistance increased from 16.7% in 1994 to 83.6% in 2018. Several non-synonymous mutations were identified in the Kelch13 gene, although none of them are currently associated with artemisinin resistance. We observed a temporal increase in the Pfmdr1 NFD haplotype associated with lumefantrine resistance, but observed no evidence of piperaquine resistance. SNPs in other parts of the genome showed no significant temporal changes despite the marked changes in drug resistance loci over this period. Conclusions: We identified substantial changes in molecular markers of P. falciparum drug resistance over 25 years in coastal Kenya, but no associated changes in the parasite population structure.
Collapse
|
11
|
Tuedom AGB, Sarah-Matio EM, Moukoko CEE, Feufack-Donfack BL, Maffo CN, Bayibeki AN, Awono-Ambene HP, Ayong L, Berry A, Abate L, Morlais I, Nsango SE. Antimalarial drug resistance in the Central and Adamawa regions of Cameroon: Prevalence of mutations in P. falciparum crt, Pfmdr1, Pfdhfr and Pfdhps genes. PLoS One 2021; 16:e0256343. [PMID: 34411157 PMCID: PMC8376100 DOI: 10.1371/journal.pone.0256343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/05/2021] [Indexed: 11/19/2022] Open
Abstract
The spread of Plasmodium falciparum resistant parasites remains one of the major challenges for malaria control and elimination in Sub Saharan Africa. Monitoring of molecular markers conferring resistance to different antimalarials is important to track the spread of resistant parasites and to optimize the therapeutic lifespan of current drugs. This study aimed to evaluate the prevalence of known mutations in the drug resistance genes Pfcrt, Pfmdr1, Pfdhfr and Pfdhps in two different epidemiological settings in Cameroon. Dried blood spots collected in 2018 and 2019 from asymptomatic individuals were used for DNA extraction and then the Plasmodium infection status was determined byPCR. Detection of SNPs was performed by nested PCR followed by allele-specific restriction analysis (ASRA). The prevalence of each genotype was compared between sites using the Chi square and Fisher's exact tests. A high prevalence of the Pfcrt K76 wild type allele was found in both sites (88.5 and 62.29% respectively; P< 0,0001). The prevalence of Pfmdr1 mutations 86Y and 1246Y was respectively 55.83 and 1.45% in Mfou and 45.87 and 5.97% in Tibati, with significant difference between the studied areas (P<0.0001). Overall, the Pfdhfr triple-mutant genotype (51I/59R/108N) was highly prevalent (> 96%), however no SNP was detected at codon 164. In Pfdhps, the prevalence of the 437G mutation reached (90%) and was at higher frequency in Mfou (P< 0.0001). Overall, the Pfdhps mutations 540E and 581G were less common (0.33 and 3.26%, respectively). The quadruple resistant genotype (Pfdhfr 51I/59R/108N+Pfdhp437G) was found almost 90% of the samples. The wild-type genotype (Pfdhfr N51/C59/S108/164I+Pfdhps A437/K540/A581) was never identified and the sextuple mutant (Pfdhfr 51I/59R/108N+Pfdhp437G/540E/581G), kwon as super resistant appeared in two samples from Tibati. These findings demonstrate declining trends in the prevalence of mutations conferring resistance to 4-aminoquinolines, especially to chloroquine. However, a high level of mutations in P. falciparum genes related to SP resistance was detected and this raises concerns about the future efficacy of IPTp-SP and SMC in Cameroon.
Collapse
Affiliation(s)
- Aline Gaelle Bouopda Tuedom
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
| | - Elangwe Milo Sarah-Matio
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
- UMR MIVEGEC, IRD, CNRS, Institut de Recherche pour le Développement, Université Montpellier, Montpellier Cedex, France
| | - Carole Else Eboumbou Moukoko
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
| | - Brice Lionel Feufack-Donfack
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
- CNRS UPR9022, INSERM U963, Strasbourg, France
| | - Christelle Ngou Maffo
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
- UMR MIVEGEC, IRD, CNRS, Institut de Recherche pour le Développement, Université Montpellier, Montpellier Cedex, France
| | - Albert Ngano Bayibeki
- Université Catholique d’Afrique Centrale, Yaoundé-Campus Messa Cameroun, Yaoundé, Cameroun
| | - Hermann Parfait Awono-Ambene
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale (OCEAC), Yaoundé, Cameroun
| | - Lawrence Ayong
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
| | - Antoine Berry
- Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire de Toulouse et UMR152 UPS-IRD, Université de Toulouse, Toulouse, France
| | - Luc Abate
- UMR MIVEGEC, IRD, CNRS, Institut de Recherche pour le Développement, Université Montpellier, Montpellier Cedex, France
| | - Isabelle Morlais
- UMR MIVEGEC, IRD, CNRS, Institut de Recherche pour le Développement, Université Montpellier, Montpellier Cedex, France
| | - Sandrine Eveline Nsango
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroun
| |
Collapse
|
12
|
Da Veiga Leal S, Ward D, Campino S, Benavente ED, Ibrahim A, Claret T, Isaías V, Monteiro D, Clark TG, Gonçalves L, Valdez T, da Luz Lima Mendonça M, Silveira H, Nogueira F. Drug resistance profile and clonality of Plasmodium falciparum parasites in Cape Verde: the 2017 malaria outbreak. Malar J 2021; 20:172. [PMID: 33789667 PMCID: PMC8011132 DOI: 10.1186/s12936-021-03708-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cape Verde is an archipelago located off the West African coast and is in a pre-elimination phase of malaria control. Since 2010, fewer than 20 Plasmodium falciparum malaria cases have been reported annually, except in 2017, when an outbreak in Praia before the rainy season led to 423 autochthonous cases. It is important to understand the genetic diversity of circulating P. falciparum to inform on drug resistance, potential transmission networks and sources of infection, including parasite importation. METHODS Enrolled subjects involved malaria patients admitted to Dr Agostinho Neto Hospital at Praia city, Santiago island, Cape Verde, between July and October 2017. Neighbours and family members of enrolled cases were assessed for the presence of anti-P. falciparum antibodies. Sanger sequencing and real-time PCR was used to identify SNPs in genes associated with drug resistance (e.g., pfdhfr, pfdhps, pfmdr1, pfk13, pfcrt), and whole genome sequencing data were generated to investigate the population structure of P. falciparum parasites. RESULTS The study analysed 190 parasite samples, 187 indigenous and 3 from imported infections. Malaria cases were distributed throughout Praia city. There were no cases of severe malaria and all patients had an adequate clinical and parasitological response after treatment. Anti-P. falciparum antibodies were not detected in the 137 neighbours and family members tested. No mutations were detected in pfdhps. The triple mutation S108N/N51I/C59R in pfdhfr and the chloroquine-resistant CVIET haplotype in the pfcrt gene were detected in almost all samples. Variations in pfk13 were identified in only one sample (R645T, E668K). The haplotype NFD for pfmdr1 was detected in the majority of samples (89.7%). CONCLUSIONS Polymorphisms in pfk13 associated with artemisinin-based combination therapy (ACT) tolerance in Southeast Asia were not detected, but the majority of the tested samples carried the pfmdr1 haplotype NFD and anti-malarial-associated mutations in the the pfcrt and pfdhfr genes. The first whole genome sequencing (WGS) was performed for Cape Verdean parasites that showed that the samples cluster together, have a very high level of similarity and are close to other parasites populations from West Africa.
Collapse
Affiliation(s)
- Silvania Da Veiga Leal
- Laboratório de Entomologia Médica, Largo Do Desastre da Assistência, Instituto Nacional de Saúde Pública, Chã de Areia, Praia 719, Cape Verde.
| | - Daniel Ward
- Department of Infection and Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Susana Campino
- Department of Infection and Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Ernest Diez Benavente
- Department of Infection and Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Amy Ibrahim
- Department of Infection and Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Tânia Claret
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Varela Isaías
- Laboratório de Entomologia Médica, Largo Do Desastre da Assistência, Instituto Nacional de Saúde Pública, Chã de Areia, Praia 719, Cape Verde
| | - Davidson Monteiro
- Laboratório de Entomologia Médica, Largo Do Desastre da Assistência, Instituto Nacional de Saúde Pública, Chã de Areia, Praia 719, Cape Verde
| | - Taane G Clark
- Department of Infection and Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene E Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Luzia Gonçalves
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene E Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
- Centro de Estatística E Aplicações da Universidade de Lisboa (CEAUL), Campo Grande, Bloco C6, Piso 4, 1749-016, Lisbon, Portugal
| | - Tomas Valdez
- Ministério da Saúde E da Segurança, Palácio Do Governo, nº47, Praia, Cape Verde
| | - Maria da Luz Lima Mendonça
- Laboratório de Entomologia Médica, Largo Do Desastre da Assistência, Instituto Nacional de Saúde Pública, Chã de Areia, Praia 719, Cape Verde
| | - Henrique Silveira
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene E Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Fatima Nogueira
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene E Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| |
Collapse
|
13
|
Asua V, Conrad MD, Aydemir O, Duvalsaint M, Legac J, Duarte E, Tumwebaze P, Chin DM, Cooper RA, Yeka A, Kamya MR, Dorsey G, Nsobya SL, Bailey J, Rosenthal PJ. Changing Prevalence of Potential Mediators of Aminoquinoline, Antifolate, and Artemisinin Resistance Across Uganda. J Infect Dis 2021; 223:985-994. [PMID: 33146722 PMCID: PMC8006419 DOI: 10.1093/infdis/jiaa687] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/27/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND In Uganda, artemether-lumefantrine is recommended for malaria treatment and sulfadoxine-pyrimethamine for chemoprevention during pregnancy, but drug resistance may limit efficacies. METHODS Genetic polymorphisms associated with sensitivities to key drugs were characterized in samples collected from 16 sites across Uganda in 2018 and 2019 by ligase detection reaction fluorescent microsphere, molecular inversion probe, dideoxy sequencing, and quantitative polymerase chain reaction assays. RESULTS Considering transporter polymorphisms associated with resistance to aminoquinolines, the prevalence of Plasmodium falciparum chloroquine resistance transporter (PfCRT) 76T decreased, but varied markedly between sites (0-46% in 2018; 0-23% in 2019); additional PfCRT polymorphisms and plasmepsin-2/3 amplifications associated elsewhere with resistance to piperaquine were not seen. For P. falciparum multidrug resistance protein 1, in 2019 the 86Y mutation was absent at all sites, the 1246Y mutation had prevalence ≤20% at 14 of 16 sites, and gene amplification was not seen. Considering mutations associated with high-level sulfadoxine-pyrimethamine resistance, prevalences of P. falciparum dihydrofolate reductase 164L (up to 80%) and dihydropteroate synthase 581G (up to 67%) were high at multiple sites. Considering P. falciparum kelch protein propeller domain mutations associated with artemisinin delayed clearance, prevalence of the 469Y and 675V mutations has increased at multiple sites in northern Uganda (up to 23% and 41%, respectively). CONCLUSIONS We demonstrate concerning spread of mutations that may limit efficacies of key antimalarial drugs.
Collapse
Affiliation(s)
- Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Melissa D Conrad
- University of California, San Francisco, San Francisco, California, USA
| | | | - Marvin Duvalsaint
- University of California, San Francisco, San Francisco, California, USA
| | - Jennifer Legac
- University of California, San Francisco, San Francisco, California, USA
| | - Elias Duarte
- University of California, San Francisco, San Francisco, California, USA
| | | | | | - Roland A Cooper
- Dominican University of California, San Rafael, California, USA
| | - Adoke Yeka
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Grant Dorsey
- University of California, San Francisco, San Francisco, California, USA
| | - Sam L Nsobya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | |
Collapse
|
14
|
Kassaza K, Long AC, McDaniels JM, Andre M, Fredrickson W, Nyehangane D, Orikiriza P, Operario DJ, Bazira J, Mwanga-Amumpaire JA, Moore CC, Guler JL, Boum Y. Surveillance of Plasmodium falciparum pfcrt haplotypes in southwestern uganda by high-resolution melt analysis. Malar J 2021; 20:114. [PMID: 33632242 PMCID: PMC7908690 DOI: 10.1186/s12936-021-03657-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/18/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Chloroquine (CQ) resistance is conferred by mutations in the Plasmodium falciparum CQ resistance transporter (pfcrt). Following CQ withdrawal for anti-malarial treatment, studies across malaria-endemic countries have shown a range of responses. In some areas, CQ sensitive parasites re-emerge, and in others, mutant haplotypes persist. Active surveillance of resistance mutations in clinical parasites is essential to inform treatment regimens; this effort requires fast, reliable, and cost-effective methods that work on a variety of sample types with reagents accessible in malaria-endemic countries. METHODS Quantitative PCR followed by High-Resolution Melt (HRM) analysis was performed in a field setting to assess pfcrt mutations in two groups of clinical samples from Southwestern Uganda. Group 1 samples (119 in total) were collected in 2010 as predominantly Giemsa-stained slides; Group 2 samples (125 in total) were collected in 2015 as blood spots on filter paper. The Rotor-Gene Q instrument was utilized to assess the impact of different PCR-HRM reagent mixes and the detection of mixed haplotypes present in the clinical samples. Finally, the prevalence of the wild type (CVMNK) and resistant pfcrt haplotypes (CVIET and SVMNT) was evaluated in this understudied Southwestern region of Uganda. RESULTS The sample source (i.e. Giemsa-stained slides or blood spots) and type of LCGreen-based reagent mixes did not impact the success of PCR-HRM. The detection limit of 10- 5 ng and the ability to identify mixed haplotypes as low as 10 % was similar to other HRM platforms. The CVIET haplotype predominated in the clinical samples (66 %, 162/244); however, there was a large regional variation between the sample groups (94 % CVIET in Group 1 and 44 % CVIET in Group 2). CONCLUSIONS The HRM-based method exhibits the flexibility required to conduct reliable assessment of resistance alleles from various sample types generated during the clinical management of malaria. Large regional variations in CQ resistance haplotypes across Southwestern Uganda emphasizes the need for continued local parasite genotype assessment to inform anti-malarial treatment policies.
Collapse
Affiliation(s)
- Kennedy Kassaza
- Epicentre Mbarara Research Centre, Mbarara, Uganda
- Department of Microbiology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Anna C Long
- Department of Biology, University of Virginia, Box 400328, 22904, Charlottesville, VA, USA
| | - Jennifer M McDaniels
- Department of Biology, University of Virginia, Box 400328, 22904, Charlottesville, VA, USA
| | - Mharlove Andre
- Department of Biology, University of Virginia, Box 400328, 22904, Charlottesville, VA, USA
| | - Wasswa Fredrickson
- Department of Microbiology, Mbarara University of Science and Technology, Mbarara, Uganda
| | | | | | - Darwin J Operario
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, 22904, USA
| | - Joel Bazira
- Department of Microbiology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Juliet A Mwanga-Amumpaire
- Epicentre Mbarara Research Centre, Mbarara, Uganda
- Department of Pediatrics and Child Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Christopher C Moore
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, 22904, USA
| | - Jennifer L Guler
- Department of Biology, University of Virginia, Box 400328, 22904, Charlottesville, VA, USA.
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, 22904, USA.
| | - Yap Boum
- Epicentre Mbarara Research Centre, Mbarara, Uganda.
- Department of Microbiology, Mbarara University of Science and Technology, Mbarara, Uganda.
| |
Collapse
|
15
|
Adamu A, Jada MS, Haruna HMS, Yakubu BO, Ibrahim MA, Balogun EO, Sakura T, Inaoka DK, Kita K, Hirayama K, Culleton R, Shuaibu MN. Plasmodium falciparum multidrug resistance gene-1 polymorphisms in Northern Nigeria: implications for the continued use of artemether-lumefantrine in the region. Malar J 2020; 19:439. [PMID: 33256739 PMCID: PMC7708160 DOI: 10.1186/s12936-020-03506-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Background The analysis of single nucleotide polymorphism (SNPs) in drug-resistance associated genes is a commonly used strategy for the surveillance of anti-malarial drug resistance in populations of parasites. The present study was designed and performed to provide genetic epidemiological data of the prevalence of N86Y-Y184F-D1246Y SNPs in Plasmodium falciparum multidrug resistance 1 (pfmdr1) in the malaria hotspot of Northern Nigeria. Methods Plasmodium falciparum-positive blood samples on Whatman-3MM filter papers were collected from 750 symptomatic patients from four states (Kano, Kaduna, Yobe and Adamawa) in Northern Nigeria, and genotyped via BigDye (v3.1) terminator cycle sequencing for the presence of three SNPs in pfmdr1. SNPs in pfmdr1 were used to construct NYD, NYY, NFY, NFD, YYY, YYD, YFD and YFY haplotypes, and all data were analysed using Pearson Chi square and Fisher’s exact (FE) tests. Results The prevalence of the pfmdr1 86Y allele was highest in Kaduna (12.50%, 2 = 10.50, P = 0.02), whilst the 184F allele was highest in Kano (73.10%, 2 = 13.20, P = 0.00), and the pfmdr1 1246Y allele was highest in Yobe (5.26%, 2 = 9.20, P = 0.03). The NFD haplotype had the highest prevalence of 69.81% in Kano (2 = 36.10, P = 0.00), followed by NYD with a prevalence of 49.00% in Adamawa, then YFD with prevalence of 11.46% in Kaduna. The YYY haplotype was not observed in any of the studied states. Conclusion The present study suggests that strains of P. falciparum with reduced sensitivity to the lumefantrine component of AL exist in Northern Nigeria and predominate in the North-West region.
Collapse
Affiliation(s)
- Auwal Adamu
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Mahmoud Suleiman Jada
- Department of Biochemistry, Modibbo Adama University of Technology Yola, Yola, Nigeria
| | | | | | | | | | - Takaya Sakura
- Institute of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Daniel Ken Inaoka
- Institute of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Kiyoshi Kita
- Institute of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Richard Culleton
- Department of Molecular Parasitology, Proteo-Science Center, Ehime University, Ehime, Japan
| | | |
Collapse
|
16
|
Associations between Aminoquinoline Resistance Genotypes and Clinical Presentations of Plasmodium falciparum Infection in Uganda. Antimicrob Agents Chemother 2020; 64:AAC.00721-20. [PMID: 32660999 DOI: 10.1128/aac.00721-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/07/2020] [Indexed: 01/30/2023] Open
Abstract
Mutations that mediate resistance of Plasmodium falciparum to aminoquinoline antimalarials are selected by prior drug use and may alter parasite fitness, but associations with clinical presentations are uncertain. We evaluated genotypes in samples from a case-control study of determinants of severe malaria in Ugandan children 4 months to 10 years of age. We studied 274 cases with severe malaria matched by age and geography to 275 uncomplicated malaria controls and 179 asymptomatic parasitemic controls. The overall prevalence of mutations of interest (considering mixed results as mutant) was 67.0% for PfCRT K76T, 8.5% for PfMDR1 N86Y, 71.5% for PfMDR1 Y184F, and 14.7% for PfMDR1 D1246Y. Compared to asymptomatic controls, the odds of mutant PfCRT 76T were lower for uncomplicated (odds ratio, 0.42 [95% confidence interval, 0.24 to 0.72]; P < 0.001) or severe (0.56 [0.32 to 0.97]; P = 0.031) malaria; the odds of mutant PfMDR1 86Y were lower for uncomplicated (0.33 [0.16 to 0.65]; P < 0.001) or severe (0.21 [0.09 to 0.45]; P < 0.001) malaria; and the odds of mutant PfMDR1 1246Y were higher for uncomplicated (1.83 [0.90 to 3.98]; P = 0.076) or severe (2.06 [1.01 to 4.55]; P = 0.033) malaria. The odds of mutant PfMDR1 184F were lower in severe than asymptomatic (0.59 [0.37 to 0.92]; P = 0.016) or uncomplicated (0.61 [0.41 to 0.90]; P = 0.009) malaria. Overall, the PfCRT 76T and PfMDR1 86Y mutations were associated with decreased risk of symptomatic malaria, PfMDR1 1246Y was associated with increased risk of symptomatic malaria, and PfMDR1 184F was associated with decreased risk of severe malaria. These results offer insights into parasite genotypes in children with different presentations, although the basis for the identified associations is likely complex.
Collapse
|
17
|
Boyce RM, Brazeau N, Fulton T, Hathaway N, Matte M, Ntaro M, Mulogo E, Juliano JJ. Prevalence of Molecular Markers of Antimalarial Drug Resistance across Altitudinal Transmission Zones in Highland Western Uganda. Am J Trop Med Hyg 2020; 101:799-802. [PMID: 31436149 DOI: 10.4269/ajtmh.19-0081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We explored spatial variation in the prevalence of established molecular markers of antimalarial resistance across a geographically diverse, highland region of western Uganda. We identified Plasmodium falciparum CQ resistance transporter 76T mutations in all pools, but there was no evidence of spatial differences across village-based strata defined by either altitude or river valley. In contrast, we identified a significant inverse association between altitude and the prevalence of Plasmodium falciparum multidrug resistance 1 mutations with the largest proportion of Y184F mutations observed in the low-elevation, high-transmission villages. These results demonstrate the substantial heterogeneity in resistance markers observed across geographic settings, even at relatively small scales, but highlight the complex nature of these ecological relationships.
Collapse
Affiliation(s)
- Ross M Boyce
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda.,Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nicholas Brazeau
- Division of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Travis Fulton
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nick Hathaway
- Program in Bioinformatics and Integrative Biology, University of Massachusetts, Worcester, Massachusetts
| | - Michael Matte
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Moses Ntaro
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Edgar Mulogo
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Jonathan J Juliano
- Curriculum in Genetics and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Division of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
18
|
Bretscher MT, Dahal P, Griffin J, Stepniewska K, Bassat Q, Baudin E, D'Alessandro U, Djimde AA, Dorsey G, Espié E, Fofana B, González R, Juma E, Karema C, Lasry E, Lell B, Lima N, Menéndez C, Mombo-Ngoma G, Moreira C, Nikiema F, Ouédraogo JB, Staedke SG, Tinto H, Valea I, Yeka A, Ghani AC, Guerin PJ, Okell LC. The duration of chemoprophylaxis against malaria after treatment with artesunate-amodiaquine and artemether-lumefantrine and the effects of pfmdr1 86Y and pfcrt 76T: a meta-analysis of individual patient data. BMC Med 2020; 18:47. [PMID: 32098634 PMCID: PMC7043031 DOI: 10.1186/s12916-020-1494-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/09/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The majority of Plasmodium falciparum malaria cases in Africa are treated with the artemisinin combination therapies artemether-lumefantrine (AL) and artesunate-amodiaquine (AS-AQ), with amodiaquine being also widely used as part of seasonal malaria chemoprevention programs combined with sulfadoxine-pyrimethamine. While artemisinin derivatives have a short half-life, lumefantrine and amodiaquine may give rise to differing durations of post-treatment prophylaxis, an important additional benefit to patients in higher transmission areas. METHODS We analyzed individual patient data from 8 clinical trials of AL versus AS-AQ in 12 sites in Africa (n = 4214 individuals). The time to PCR-confirmed reinfection after treatment was used to estimate the duration of post-treatment protection, accounting for variation in transmission intensity between settings using hidden semi-Markov models. Accelerated failure-time models were used to identify potential effects of covariates on the time to reinfection. The estimated duration of chemoprophylaxis was then used in a mathematical model of malaria transmission to determine the potential public health impact of each drug when used for first-line treatment. RESULTS We estimated a mean duration of post-treatment protection of 13.0 days (95% CI 10.7-15.7) for AL and 15.2 days (95% CI 12.8-18.4) for AS-AQ overall. However, the duration varied significantly between trial sites, from 8.7-18.6 days for AL and 10.2-18.7 days for AS-AQ. Significant predictors of time to reinfection in multivariable models were transmission intensity, age, drug, and parasite genotype. Where wild type pfmdr1 and pfcrt parasite genotypes predominated (<=20% 86Y and 76T mutants, respectively), AS-AQ provided ~ 2-fold longer protection than AL. Conversely, at a higher prevalence of 86Y and 76T mutant parasites (> 80%), AL provided up to 1.5-fold longer protection than AS-AQ. Our simulations found that these differences in the duration of protection could alter population-level clinical incidence of malaria by up to 14% in under-5-year-old children when the drugs were used as first-line treatments in areas with high, seasonal transmission. CONCLUSION Choosing a first-line treatment which provides optimal post-treatment prophylaxis given the local prevalence of resistance-associated markers could make a significant contribution to reducing malaria morbidity.
Collapse
Affiliation(s)
- Michael T Bretscher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.
| | - Prabin Dahal
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.,Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jamie Griffin
- School of Mathematical Sciences, Queen Mary University of London, London, UK
| | - Kasia Stepniewska
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.,Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.,Pediatric Infectious Diseases Unit, Pediatrics Department, Hospital Sant Joan de Déu (University of Barcelona), Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | | | - Umberto D'Alessandro
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Abdoulaye A Djimde
- Malaria Research and Training Center, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco, USA
| | - Emmanuelle Espié
- Epicentre, Paris, France.,Clinical and Epidemiology Department, GSK Vaccines, R&D Center, Wavre, Belgium
| | - Bakary Fofana
- Malaria Research and Training Center, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Raquel González
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Elizabeth Juma
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Corine Karema
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Bertrand Lell
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria.,Centre de Recherches Medicales de Lambarene, Lambarene, Gabon
| | - Nines Lima
- Department of Paediatrics, University of Calabar, Calabar, Nigeria
| | - Clara Menéndez
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Ghyslain Mombo-Ngoma
- Centre de Recherches Medicales de Lambarene, Lambarene, Gabon.,Institute for Tropical Medicine, University of Tubingen, Tubingen, Germany.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clarissa Moreira
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.,Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Frederic Nikiema
- Institut de Recherche en Science de la Sante, Bobo-Dioulasso, Burkina Faso
| | - Jean B Ouédraogo
- Institut de Recherche en Science de la Sante, Bobo-Dioulasso, Burkina Faso
| | - Sarah G Staedke
- Department of Clinical Research, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Halidou Tinto
- Institut de Recherche en Science de la Sante, Nanoro, Burkina Faso
| | - Innocent Valea
- Institut de Recherche en Science de la Sante, Nanoro, Burkina Faso
| | - Adoke Yeka
- Uganda Malaria Surveillance Project, Kampala, Uganda
| | - Azra C Ghani
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Philippe J Guerin
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.,Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lucy C Okell
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.
| |
Collapse
|
19
|
Balikagala B, Sakurai-Yatsushiro M, Tachibana SI, Ikeda M, Yamauchi M, Katuro OT, Ntege EH, Sekihara M, Fukuda N, Takahashi N, Yatsushiro S, Mori T, Hirai M, Opio W, Obwoya PS, Anywar DA, Auma MA, Palacpac NMQ, Tsuboi T, Odongo-Aginya EI, Kimura E, Ogwang M, Horii T, Mita T. Recovery and stable persistence of chloroquine sensitivity in Plasmodium falciparum parasites after its discontinued use in Northern Uganda. Malar J 2020; 19:76. [PMID: 32070358 PMCID: PMC7026951 DOI: 10.1186/s12936-020-03157-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/09/2020] [Indexed: 11/10/2022] Open
Abstract
Background Usage of chloroquine was discontinued from the treatment of Plasmodium falciparum infection in almost all endemic regions because of global spread of resistant parasites. Since the first report in Malawi, numerous epidemiological studies have demonstrated that the discontinuance led to re-emergence of chloroquine-susceptible P. falciparum, suggesting a possible role in future malaria control. However, most studies were cross-sectional, with few studies looking at the persistence of chloroquine recovery in long term. This study fills the gap by providing, for a period of at least 6 years, proof of persistent re-emergence/stable recovery of susceptible parasite populations using both molecular and phenotypic methods. Methods Ex vivo drug-susceptibility assays to chloroquine (n = 319) and lumefantrine (n = 335) were performed from 2013 to 2018 in Gulu, Northern Uganda, where chloroquine had been removed from the official malaria treatment regimen since 2006. Genotyping of pfcrt and pfmdr1 was also performed. Results Chloroquine resistance (≥ 100 nM) was observed in only 3 (1.3%) samples. Average IC50 values for chloroquine were persistently low throughout the study period (17.4–24.9 nM). Parasites harbouring pfcrt K76 alleles showed significantly lower IC50s to chloroquine than the parasites harbouring K76T alleles (21.4 nM vs. 43.1 nM, p-value = 3.9 × 10−8). Prevalence of K76 alleles gradually increased from 71% in 2013 to 100% in 2018. Conclusion This study found evidence of stable persistence of chloroquine susceptibility with the fixation of pfcrt K76 in Northern Uganda after discontinuation of chloroquine in the region. Accumulation of similar evidence in other endemic areas in Uganda could open channels for possible future re-use of chloroquine as an option for malaria treatment or prevention.
Collapse
Affiliation(s)
- Betty Balikagala
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Miki Sakurai-Yatsushiro
- Department of International Affairs and Tropical Medicine, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shin-Ichiro Tachibana
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mie Ikeda
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Masato Yamauchi
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Osbert T Katuro
- Mildmay Uganda, Nazibwa Hill, Lweza, P.O. Box 24985, Kampala, Uganda
| | - Edward H Ntege
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Ehime, 790-8577, Japan
| | - Makoto Sekihara
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Naoyuki Fukuda
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Nobuyuki Takahashi
- Department of International Affairs and Tropical Medicine, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shouki Yatsushiro
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14 Hayashi-cho, Takamatsu, Kagawa, 761-0395, Japan
| | - Toshiyuki Mori
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Makoto Hirai
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Walter Opio
- St. Mary's Hospital Lacor, P.O. Box 180, Gulu, Uganda
| | - Paul S Obwoya
- St. Mary's Hospital Lacor, P.O. Box 180, Gulu, Uganda
| | - Denis A Anywar
- Faculty of Science, Gulu University, P.O. Box 166, Gulu, Uganda
| | - Mary A Auma
- St. Mary's Hospital Lacor, P.O. Box 180, Gulu, Uganda
| | - Nirianne M Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Ehime, 790-8577, Japan
| | | | - Eisaku Kimura
- Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki, 852-8523, Japan
| | - Martin Ogwang
- St. Mary's Hospital Lacor, P.O. Box 180, Gulu, Uganda
| | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihiro Mita
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
20
|
Yeka A, Wallender E, Mulebeke R, Kibuuka A, Kigozi R, Bosco A, Kyambadde P, Opigo J, Kalyesubula S, Senzoga J, Vinden J, Conrad M, Rosenthal PJ. Comparative Efficacy of Artemether-Lumefantrine and Dihydroartemisinin-Piperaquine for the Treatment of Uncomplicated Malaria in Ugandan Children. J Infect Dis 2020; 219:1112-1120. [PMID: 30418593 DOI: 10.1093/infdis/jiy637] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/01/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND In Uganda, artemether-lumefantrine (AL) and dihydroartemisinin-piperaquine (DHA-PQ) showed excellent treatment efficacy for uncomplicated malaria in prior trials. Because the frequency of resistance to artemisinins and piperaquine is increasing in Southeast Asia and the prevalence of Plasmodium falciparum polymorphisms associated with resistance has changed, we reassessed treatment efficacies at 3 sites in Uganda. METHODS For this randomized, single-blinded clinical trial, children aged 6-59 months with uncomplicated falciparum malaria were assigned treatment with AL or DHA-PQ and followed for 42 days. Primary end points were risks of recurrent parasitemia, either unadjusted or adjusted to distinguish recrudescence from new infection. We assessed selection by study regimens of relevant P. falciparum genetic polymorphisms associated with drug resistance. RESULTS Of 599 patients enrolled, 578 completed follow-up. There were no early treatment failures. The risk of recurrent parasitemia was lower with DHA-PQ as compared to AL at all 3 sites at 42 days (26.0% vs 47.0%; P < .001). Recrudescent infections were uncommon in both the DHA-PQ and AL arms (1.1% and 2.2%, respectively; P = .25). Neither regimen selected for pfcrt or pfmdr1 polymorphisms associated with drug resistance. CONCLUSIONS AL and DHA-PQ remain effective for the treatment of malaria in Uganda. Neither regimen selected for genetic polymorphisms associated with drug resistance. CLINICAL TRIALS REGISTRATION ISRCTN15793046.
Collapse
Affiliation(s)
- Adoke Yeka
- School of Public Health, Makerere University College of Health Sciences
| | - Erika Wallender
- Department of Medicine, University of California, San Francisco
| | - Ronald Mulebeke
- School of Public Health, Makerere University College of Health Sciences
| | - Afizi Kibuuka
- School of Public Health, Makerere University College of Health Sciences
| | - Ruth Kigozi
- Malaria Action Programme for Districts, Malaria Consortium
| | - Agaba Bosco
- National Malaria Control Program, Ministry of Health, Uganda
| | - Paul Kyambadde
- National Malaria Control Program, Ministry of Health, Uganda
| | - Jimmy Opigo
- National Malaria Control Program, Ministry of Health, Uganda
| | - Simeon Kalyesubula
- East African Public Health Laboratories Networking Project, Kampala, Uganda
| | - Joseph Senzoga
- East African Public Health Laboratories Networking Project, Kampala, Uganda
| | - Joanna Vinden
- School of Public Health, University of California, Berkeley
| | - Melissa Conrad
- Department of Medicine, University of California, San Francisco
| | | |
Collapse
|
21
|
Conrad MD, Rosenthal PJ. Antimalarial drug resistance in Africa: the calm before the storm? THE LANCET. INFECTIOUS DISEASES 2019; 19:e338-e351. [DOI: 10.1016/s1473-3099(19)30261-0] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 11/26/2022]
|
22
|
Achol E, Ochaya S, Malinga GM, Edema H, Echodu R. High prevalence of Pfmdr-1 N86 and D1246 genotypes detected among febrile malaria outpatients attending Lira Regional Referral Hospital, Northern Uganda. BMC Res Notes 2019; 12:235. [PMID: 31014391 PMCID: PMC6480777 DOI: 10.1186/s13104-019-4269-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/15/2019] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To determine the prevalence of Plasmodium falciparum multi-drug resistant gene-1 (Pfmdr-1) N86Y and D1246Y genotypes among febrile malaria outpatients attending Lira Regional Referral Hospital, Uganda. RESULTS Overall, 92.3% (n = 48/52) and 90% (n = 45/50) of the parasites detected carried the wild type alleles 1246D and N86, respectively. Only 7.7% (n = 4/52) and 10% (n = 5/50) of these P. falciparum isolates carried the Pfmdr-1 mutant alleles 1246Y and 86Y, respectively. Our results show high prevalence of wild type alleles N86 and D1246 in P. falciparum isolates from Lira Regional Referral Hospital, which could translate to a decreased sensitivity to artemether-lumefantrine. Continued monitoring of prevalence of single nucleotide polymorphisms is warranted to timely inform malaria treatment policies and guidelines.
Collapse
Affiliation(s)
- Emmanuel Achol
- Department of Biology, Faculty of Science, Gulu University, P.O. Box 166, Gulu, Uganda.,Gulu University Bioscience Research Laboratories, P.O. Box 166, Gulu, Uganda
| | - Stephen Ochaya
- Department of Immunology and Microbiology, Gulu University, P.O. Box 166, Gulu, Uganda
| | - Geoffrey M Malinga
- Department of Biology, Faculty of Science, Gulu University, P.O. Box 166, Gulu, Uganda.,Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland
| | - Hillary Edema
- Gulu University Bioscience Research Laboratories, P.O. Box 166, Gulu, Uganda
| | - Richard Echodu
- Department of Biology, Faculty of Science, Gulu University, P.O. Box 166, Gulu, Uganda. .,Gulu University Bioscience Research Laboratories, P.O. Box 166, Gulu, Uganda.
| |
Collapse
|
23
|
Juma DW, Muiruri P, Yuhas K, John-Stewart G, Ottichilo R, Waitumbi J, Singa B, Polyak C, Kamau E. The prevalence and antifolate drug resistance profiles of Plasmodium falciparum in study participants randomized to discontinue or continue cotrimoxazole prophylaxis. PLoS Negl Trop Dis 2019; 13:e0007223. [PMID: 30897090 PMCID: PMC6445470 DOI: 10.1371/journal.pntd.0007223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 04/02/2019] [Accepted: 02/06/2019] [Indexed: 11/19/2022] Open
Abstract
Objective Cotrimoxazole prevents opportunistic infections including falciparum malaria in HIV-infected individuals but there are concerns of cross-resistance to other antifolate drugs such as sulphadoxine-pyrimethamine (SP). In this study, we investigated the prevalence of antifolate-resistance mutations in Plasmodium falciparum that are associated with SP resistance in HIV-infected individuals on antiretroviral treatment randomized to discontinue (STOP-CTX), or continue (CTX) cotrimoxazole in Western Kenya. Design Samples were obtained from an unblinded, non-inferiority randomized controlled trial where participants were recruited on a rolling basis for the first six months of the study, then followed-up for 12 months with samples collected at enrollment, quarterly, and during sick visits. Method Plasmodium DNA was extracted from blood specimens. Initial screening to determine the presence of Plasmodium spp. was performed by quantitative reverse transcriptase real-time PCR, followed by genotyping for the presence of SP-resistance associated mutations by Sanger sequencing. Results The prevalence of mutant haplotypes associated with SP-resistant parasites in pfdhfr (51I/59R/108N) and pfdhps (437G/540E) genes were significantly higher (P = 0.0006 and P = 0.027, respectively) in STOP-CTX compared to CTX arm. The prevalence of quintuple haplotype (51I/59R/108N/437G/540E) was 51.8% in STOP-CTX vs. 6.3% (P = 0.0007) in CTX arm. There was a steady increase in mutant haplotypes in both genes in STOP-CTX arm overtime through the study period, reaching statistical significance (P < 0.0001). Conclusion The frequencies of mutations in pfdhfr and pfdhps genes were higher in STOP-CTX arm compared to CTX arm, suggesting cotrimoxazole effectively controls and selects against SP-resistant parasites. Trial registration ClinicalTrials.gov NCT01425073 Cotrimoxazole, an antifolate, is a fixed-dose trimethoprim-sulfamethoxazole used to prevent opportunistic infections including malaria in HIV-infected individuals. There are concerns that widespread use of cotrimoxazole for prophylaxis may result in selection of P. falciparum parasites with cross-resistance to other antifolate drugs such as sulphadoxine-pyrimethamine (SP), which is used as intermittent preventive treatment of malaria in pregnancy (IPTp) and in infants (IPTi) in Africa. This sub-study used samples from a clinical trial in which HIV-infected individuals on antiretroviral treatment were randomized to discontinue (STOP-CTX) or continue (CTX) cotrimoxazole prophylaxis for 12 months. The sub-study was designed to assess whether taking cotrimoxazole increased the risk of selecting for parasites with SP-resistant mutations in HIV-infected individuals. Samples were genotyped by sequencing to assess the prevalence of mutations associated with SP-resistance. We found there was no risk of selecting for parasites with SP-resistance mutations while on cotrimoxazole. In fact, the opposite was true; cotrimoxazole controlled parasites carrying SP-resistance mutations as evident by the gradual increase in the prevalence of parasites with mutant alleles in the STOP-CTX arm and not in the CTX-arm. We concluded that cotrimoxazole remains effective in controlling malaria infection despite of the high prevalence of SP-resistant parasites, and its use does not select for SP mutations.
Collapse
Affiliation(s)
- Dennis W. Juma
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
| | - Peninah Muiruri
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Krista Yuhas
- Department of Global Health, University of Washington, Seattle, Washington United States of America
| | - Grace John-Stewart
- Departments of Global Health, Medicine, Epidemiology, and Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Ronald Ottichilo
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
| | - John Waitumbi
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
| | - Benson Singa
- Centre for Clinical Research, KEMRI, Nairobi, Kenya
| | - Christina Polyak
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Edwin Kamau
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- * E-mail:
| |
Collapse
|
24
|
Ocan M, Akena D, Nsobya S, Kamya MR, Senono R, Kinengyere AA, Obuku EA. Persistence of chloroquine resistance alleles in malaria endemic countries: a systematic review of burden and risk factors. Malar J 2019; 18:76. [PMID: 30871535 PMCID: PMC6419488 DOI: 10.1186/s12936-019-2716-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Background Chloroquine, a previous highly efficacious, easy to use and affordable anti-malarial agent was withdrawn from malaria endemic regions due to high levels of resistance. This review collated evidence from published-reviewed articles to establish prevalence of Pfcrt 76T and Pfmdr-1 86Y alleles in malaria affected countries following official discontinuation of chloroquine use. Methods A review protocol was developed, registered in PROSPERO (#CRD42018083957) and published in a peer-reviewed journal. Article search was done in PubMed, Scopus, Lilacs/Vhl and Embase databases by two experienced librarians (AK, RS) for the period 1990-to-Febuary 2018. Mesh terms and Boolean operators (AND, OR) were used. Data extraction form was designed in Excel spread sheet 2007. Data extraction was done by three reviewers (NL, BB and MO), discrepancies were resolved by discussion. Random effects analysis was done in Open Meta Analyst software. Heterogeneity was established using I2-statistic. Results A total of 4721 citations were retrieved from article search (Pubmed = 361, Lilac/vhl = 28, Science Direct = 944, Scopus = 3388). Additional targeted search resulted in three (03) eligible articles. After removal of duplicates (n = 523) and screening, 38 articles were included in the final review. Average genotyping success rate was 63.6% (18,343/28,820) for Pfcrt K76T and 93.5% (16,232/17,365) for Pfmdr-1 86Y mutations. Prevalence of Pfcrt 76T was as follows; East Africa 48.9% (2528/5242), Southern Africa 18.6% (373/2163), West Africa 58.3% (3321/6608), Asia 80.2% (1951/2436). Prevalence of Pfmdr-1 86Y was; East Africa 32.4% (1447/5722), Southern Africa 36.1% (544/1640), West Africa 52.2% (1986/4200), Asia 46.4% (1276/2217). Over half, 52.6% (20/38) of included studies reported continued unofficial chloroquine use following policy change. Studies done in Madagascar and Kenya reported re-emergence of chloroquine sensitive parasites (IC50 < 30.9 nM). The average time (years) since discontinuation of chloroquine use to data collection was 8.7 ± 7.4. There was high heterogeneity (I2 > 95%). Conclusion The prevalence of chloroquine resistance alleles among Plasmodium falciparum parasites have steadily declined since discontinuation of chloroquine use. However, Pfcrt K76T and Pfmdr-1 N86Y mutations still persist at moderate frequencies in most malaria affected countries.
Collapse
Affiliation(s)
- Moses Ocan
- Department of Pharmacology & Therapeutics, Makerere University, P.O. Box 7072, Kampala, Uganda. .,Africa Centre for Systematic Reviews and Knowledge Translation, Makerere University College of Health Sciences, P.O. Box 7072, Kampala, Uganda.
| | - Dickens Akena
- Department of Psychiatry, Makerere University, P.O. Box 7072, Kampala, Uganda.,Infectious Disease Institute, Makerere University, P. O. Box 22418, Kampala, Uganda
| | - Sam Nsobya
- Department of Medical Microbiology, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Moses R Kamya
- Department of Medicine, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Richard Senono
- Infectious Disease Institute, Makerere University, P. O. Box 22418, Kampala, Uganda.,Africa Centre for Systematic Reviews and Knowledge Translation, Makerere University College of Health Sciences, P.O. Box 7072, Kampala, Uganda
| | - Alison Annet Kinengyere
- Albert Cook Library, Makerere University, P.O. Box 7072, Kampala, Uganda.,Africa Centre for Systematic Reviews and Knowledge Translation, Makerere University College of Health Sciences, P.O. Box 7072, Kampala, Uganda
| | - Ekwaro A Obuku
- Clinical Epidemiology Unit, Department of Medicine, Makerere University, P.O. Box 7072, Kampala, Uganda.,Africa Centre for Systematic Reviews and Knowledge Translation, Makerere University College of Health Sciences, P.O. Box 7072, Kampala, Uganda.,Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
25
|
Changing Molecular Markers of Antimalarial Drug Sensitivity across Uganda. Antimicrob Agents Chemother 2019; 63:AAC.01818-18. [PMID: 30559133 DOI: 10.1128/aac.01818-18] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/06/2018] [Indexed: 11/20/2022] Open
Abstract
The potential spread of antimalarial drug resistance to Africa, in particular for artemisinins and key partner drugs, is a major concern. We surveyed Plasmodium falciparum genetic markers associated with drug sensitivity on 3 occasions at ∼6-month intervals in 2016 and 2017 at 10 sites representing a range of epidemiological settings in Uganda. For putative drug transporters, we found continued evolution toward wild-type sequences associated with increased sensitivity to chloroquine. For pfcrt K76T, by 2017 the prevalence of the wild type was >60% at all sites and >90% at 6 sites. For the pfmdr1 N86Y and D1246Y alleles, wild type prevalence ranged from 80 to 100%. We found low prevalence of K13 propeller domain mutations, which are associated with artemisinin resistance in Asia, but one mutation previously identified in northern Uganda, 675V, was seen in 2.0% of samples, including 5.5% of those from the 3 northernmost sites. Amplification of the pfmdr1 and plasmepsin2 genes, associated elsewhere with decreased sensitivity to lumefantrine and piperaquine, respectively, was seen in <1% of samples. For the antifolate targets pfdhfr and pfdhps, 5 mutations previously associated with resistance were very common, and the pfdhfr 164L and pfdhps 581G mutations associated with higher-level resistance were seen at multiple sites, although prevalence did not clearly increase over time. Overall, changes were consistent with the selective pressure of the national treatment regimen, artemether-lumefantrine, with increased sensitivity to chloroquine, and with poor efficacy of antifolates. Strong evidence for resistance to artemisinins was not seen. Continued surveillance of markers that predict antimalarial drug sensitivity is warranted.
Collapse
|
26
|
Otienoburu SD, Suay I, Garcia S, Thomas NV, Srisutham S, Björkman A, Humphreys GS. An online mapping database of molecular markers of drug resistance in Plasmodium falciparum: the ACT Partner Drug Molecular Surveyor. Malar J 2019; 18:12. [PMID: 30658642 PMCID: PMC6339428 DOI: 10.1186/s12936-019-2645-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/10/2019] [Indexed: 01/14/2023] Open
Abstract
Background Prior to this project, only a handful of online visualizations existed for exploring the published literature on molecular markers of antimalarial drug resistance, and none specifically for the markers associated with Plasmodium falciparum resistance to the partner drugs in artemisinin-based combination therapy (ACT). Molecular information is collected in studies with different designs, using a variety of molecular methodologies and data analysis strategies, making it difficult to compare across studies. The purpose of this project was to develop a free online tool, which visualizes the widely published data on molecular markers of antimalarial drug resistance, starting with the two genes pfcrt and pfmdr-1, associated with resistance to the three most common partner drugs; amodiaquine, lumefantrine and mefloquine. Methods A literature review was conducted, and a standardized method was used to extract data from publications, and critical decisions on visualization were made. A global geospatial database was developed of specific pfmdr1 and pfcrt single nucleotide polymorphisms and pfmdr1 copy number variation. An informatics framework was developed that allowed flexibility in development of the tool over time and efficient adaptation to different source data. Results The database discussed in this paper has pfmdr1 and pfcrt marker prevalence information, from 579 geographic sites in 76 different countries, including results from over 86,000 samples from 456 articles published January 2001–May 2017. The ACT Partner Drugs Molecular Surveyor was launched by the WorldWide Antimalarial Resistance Network (WWARN) in March 2015 and it has attracted over 3000 unique visitors since then. Presented here is a demonstration of how the Surveyor database can be explored to monitor local, temporal changes in the prevalence of molecular markers. Here publications up to May 2017 were included, however the online ACT partner drug Molecular Surveyor is continuously updated with new data and relevant markers. Conclusions The WWARN ACT Partner Drugs Molecular Surveyor summarizes data on resistance markers in the pfmdr1 and pfcrt genes. The database is fully accessible, providing users with a rich resource to explore and analyze, and thus utilize a centralized, standardized database for different purposes. This open-source software framework can be adapted to other data, as demonstrated by the subsequent launch of the Artemisinin Molecular Surveyor and the Vivax Surveyor. Electronic supplementary material The online version of this article (10.1186/s12936-019-2645-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sabina Dahlström Otienoburu
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.,Department of Computer Science and Engineering, Johnson C. Smith University, Charlotte, NC, USA
| | - Ignacio Suay
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Steven Garcia
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Nigel V Thomas
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Suttipat Srisutham
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Anders Björkman
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
27
|
Konaté A, Gnagne PA, Bédia-Tanoh VA, Amiah-Droh M, Tano DK, Ignace Eby Menan H, Yavo W. Low rates of Plasmodium falciparum Pfcrt K76T mutation in three sentinel sites of malaria monitoring in Côte d'Ivoire. Acta Parasitol 2018; 63:795-801. [PMID: 30367773 DOI: 10.1515/ap-2018-0096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/21/2018] [Indexed: 11/15/2022]
Abstract
Despite efforts to eliminate it, malaria remains a major public health concern, particularly in Côte d'Ivoire. Chloroquine (CQ) was one of the first drugs used for its treatment, but was officially withdrawn from the market in 2007 following reports of high levels of chloroquine resistance. The present study was carried out after the withdrawal of CQ and provides an update on the rates of CQ resistance in Côte d'Ivoire. Samples were collected between September 2013 and March 2014 in Abidjan and from January to May 2016 in Abengourou and San Pedro through cross-sectional studies. Parasitemia was assessed by microscopy, and single nucleotide polymorphism in the Pfcrt (codon 76) gene was analyzed by nested PCR and restriction fragment length polymorphism. A total of 343 samples were analyzed: 119, 106 and 118 were from Abidjan, Abengourou, and San Pedro, respectively. The sex ratio of patients was 0.92. The mean age of patients enrolled was 9.6 years (SD = 10.8). The geometric mean of parasite density was 21,337 parasites/μL (SD = 49,508; range, 2,000-200,000). Molecular analysis revealed 57 K76T mutants (16.6%): 33, 9, and 15 in Abidjan, Abengourou and in San Pedro, respectively. Most of these were found in patients aged ≤15 years (42/57) who had parasitemia greater than 10,000 parasites/μL (40/57). This is the first study conducted in Côte d'Ivoire reporting a decline in Pfcrt K76T mutation rate. Thus, our results indicate the importance of following up on the observed trend also at a national level.
Collapse
Affiliation(s)
- Abibatou Konaté
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34,Abidjan, Côte d'Ivoire
- Malaria Research and Control Centre/National Institute of Public Health, Abidjan, Côte d'Ivoire, BPV 47,Abidjan, Côte d'Ivoire
| | - Paterne Akpa Gnagne
- Malaria Research and Control Centre/National Institute of Public Health, Abidjan, Côte d'Ivoire, BPV 47,Abidjan, Côte d'Ivoire
| | - Valérie Akoua Bédia-Tanoh
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34,Abidjan, Côte d'Ivoire
- Malaria Research and Control Centre/National Institute of Public Health, Abidjan, Côte d'Ivoire, BPV 47,Abidjan, Côte d'Ivoire
| | - Mireille Amiah-Droh
- Malaria Research and Control Centre/National Institute of Public Health, Abidjan, Côte d'Ivoire, BPV 47,Abidjan, Côte d'Ivoire
| | - Dominique Konan Tano
- Malaria Research and Control Centre/National Institute of Public Health, Abidjan, Côte d'Ivoire, BPV 47,Abidjan, Côte d'Ivoire
| | - Hervé Ignace Eby Menan
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34,Abidjan, Côte d'Ivoire
- Parasitology and Mycology Laboratory of the Diagnosis and Research Centre on AIDS and the others infectious diseases, 01 BPV 13,Abidjan, Côte d'Ivoire
| | - William Yavo
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34,Abidjan, Côte d'Ivoire
- Malaria Research and Control Centre/National Institute of Public Health, Abidjan, Côte d'Ivoire, BPV 47,Abidjan, Côte d'Ivoire
| |
Collapse
|
28
|
Muiruri P, Juma DW, Ingasia LA, Chebon LJ, Opot B, Ngalah BS, Cheruiyot J, Andagalu B, Akala HM, Nyambati VCS, Ng'ang'a JK, Kamau E. Selective sweeps and genetic lineages of Plasmodium falciparum multi-drug resistance (pfmdr1) gene in Kenya. Malar J 2018; 17:398. [PMID: 30376843 PMCID: PMC6208105 DOI: 10.1186/s12936-018-2534-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 10/20/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND There are concerns that resistance to artemisinin-based combination therapy might emerge in Kenya and sub-Saharan Africa (SSA) in the same pattern as was with chloroquine and sulfadoxine-pyrimethamine. Single nucleotide polymorphisms (SNPs) in critical alleles of pfmdr1 gene have been associated with resistance to artemisinin and its partner drugs. Microsatellite analysis of loci flanking genes associated with anti-malarial drug resistance has been used in defining the geographic origins, dissemination of resistant parasites and identifying regions in the genome that have been under selection. METHODS This study set out to investigate evidence of selective sweep and genetic lineages in pfmdr1 genotypes associated with the use of artemether-lumefantrine (AL), as the first-line treatment in Kenya. Parasites (n = 252) from different regions in Kenya were assayed for SNPs at codons 86, 184 and 1246 and typed for 7 neutral microsatellites and 13 microsatellites loci flanking (± 99 kb) pfmdr1 in Plasmodium falciparum infections. RESULTS The data showed differential site and region specific prevalence of SNPs associated with drug resistance in the pfmdr1 gene. The prevalence of pfmdr1 N86, 184F, and D1246 in western Kenya (Kisumu, Kericho and Kisii) compared to the coast of Kenya (Malindi) was 92.9% vs. 66.7%, 53.5% vs. to 24.2% and 96% vs. to 87.9%, respectively. The NFD haplotype which is consistent with AL selection was at 51% in western Kenya compared to 25% in coastal Kenya. CONCLUSION Selection pressures were observed to be different in different regions of Kenya, especially the western region compared to the coastal region. The data showed independent genetic lineages for all the pfmdr1 alleles. The evidence of soft sweeps in pfmdr1 observed varied in direction from one region to another. This is challenging for malaria control programs in SSA which clearly indicate effective malaria control policies should be based on the region and not at a country wide level.
Collapse
Affiliation(s)
- Peninah Muiruri
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
- Department of Biochemistry, School of Biomedical Sciences, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200, Nairobi, Kenya
| | - Denis W Juma
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Luicer A Ingasia
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Lorna J Chebon
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Benjamin Opot
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Bidii S Ngalah
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Jelagat Cheruiyot
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Ben Andagalu
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Hoseah M Akala
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya
| | - Venny C S Nyambati
- Department of Biochemistry, School of Biomedical Sciences, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200, Nairobi, Kenya
| | - Joseph K Ng'ang'a
- Department of Biochemistry, School of Biomedical Sciences, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200, Nairobi, Kenya
| | - Edwin Kamau
- Global Emerging Infections Surveillance Program, United States Army Medical Research Directorate-Africa, Kenya Medical Research Institute, P.O. Box 54, 40100, Kisumu, Kenya.
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA.
| |
Collapse
|
29
|
Osei M, Ansah F, Matrevi SA, Asante KP, Awandare GA, Quashie NB, Duah NO. Amplification of GTP-cyclohydrolase 1 gene in Plasmodium falciparum isolates with the quadruple mutant of dihydrofolate reductase and dihydropteroate synthase genes in Ghana. PLoS One 2018; 13:e0204871. [PMID: 30265714 PMCID: PMC6162080 DOI: 10.1371/journal.pone.0204871] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/14/2018] [Indexed: 11/19/2022] Open
Abstract
Sulfadoxine-pyrimethamine (SP) is used as malaria chemoprophylaxis for pregnant women and children in Ghana. Plasmodium falciparum resistance to SP is linked to mutations in the dihydropteroate synthase gene (pfdhps), dihydrofolate reductase gene (pfdhfr) and amplification of GTP cyclohydrolase 1 (pfgch1) gene. The pfgch1 duplication is associated with pfdhfr L164, a crucial mutant for high level pyrimethamine resistance which is rare in Ghana. The presence of amplified pfgch1 in Ghanaian isolates could be an indicator of the evolution of the L164 mutant. This study therefore determined the pfgch1 copy number variations and SP resistance mutations in clinical isolates from Ghana. One hundred and ninety-two (192) blood samples collected from children aged ≤14 years with uncomplicated malaria in 2013-14 and 2015-16 were used. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the pfgch1 copy number and nested PCR-Sanger sequencing used to detect mutations in pfdhps and pfdhfr genes. Twelve parasites (6.3%) harbored double copies of the pfgch1 gene out of the 192 samples. Of the 12, 75% had the pfdhfr I51-R59-N108, 92% had the pfdhps G437 mutant, 8% had the pfdhps E540 and 67% had the SP resistance haplotype IRNG. No L164 was detected in samples with amplified pfgch1. The rare T108 mutant associated with cycloguanil resistance showed predominance (60%) over N108 in the 2015-16 isolates. The observation of parasites with increased copy number of pfgch1 gene is indicative of the future evolution of the rare quadruple pfdhfr mutant, I51-R59-N108-L164, in Ghanaian parasites. Mutant pfdhps isolates also had increased gch1 copy number suggestive that it may also facilitate sulphadoxine resistance. The selection of parasites with pfgch1 gene amplification will enhance the sustenance and persistence of parasites with SP resistance in the country. Policy makers need to begin the search for a replacement chemoprophylaxis drug for malaria vulnerable groups in Ghana.
Collapse
Affiliation(s)
- Musah Osei
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Felix Ansah
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Sena A. Matrevi
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | | | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Neils B. Quashie
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
- Centre for Tropical Clinical Pharmacology and Therapeutics, School of Medicine and Dentistry, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Nancy O. Duah
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
30
|
Moyeh MN, Njimoh DL, Evehe MS, Ali IM, Nji AM, Nkafu DN, Masumbe PN, Barbara AT, Ndikum VN, Mbacham WF. Effects of Drug Policy Changes on Evolution of Molecular Markers of Plasmodium falciparum Resistance to Chloroquine, Amodiaquine, and Sulphadoxine-Pyrimethamine in the South West Region of Cameroon. Malar Res Treat 2018; 2018:7071383. [PMID: 29854394 PMCID: PMC5954917 DOI: 10.1155/2018/7071383] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/15/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND As a result of the spread of parasites resistant to antimalarial drugs, Malaria treatment guidelines in Cameroon evolved from nonartemisinin monotherapy to artemisinin-based combination therapy. The aim of this study was to assess the effect of these therapy changes on the prevalence of molecular markers of resistance from 2003 to 2013 in Mutengene, Cameroon. METHODOLOGY Dry blood samples (collected in 2003-2005 and 2009-2013) were used for parasite DNA extraction. Drug resistance genes were amplified by PCR and hybridized with oligonucleotide probes or subjected to restriction digestion. The prevalence of individual marker polymorphisms and haplotypes was compared in these two study periods using the Chi square test. RESULTS Alleles conferring resistance to 4-aminoquinolines in the Pfcrt 76T and Pfmdr1 86Y, 184F, and 1246Y genotypes showed a significant reduction of 97.0% to 66.9%, 83.6% to 45.2%, 97.3% to 56.0%, and 3.1% to 0.0%, respectively (P < 0.05). No difference was observed in SNPs associated with antifolate drugs resistance 51I, 59R, 108N, or 540E (P > 0.05). Haplotype analysis in the Pfmdr1 gene showed a reduction in the YFD from 75.90% to 42.2%, P < 0.0001, and an increase in the NYD (2.9% to 30.1%; P < 0.0001). CONCLUSIONS The results indicated a gradual return of the 4-aminoquinoline sensitive genotype while the antifolate resistant genotypes increased to saturation.
Collapse
Affiliation(s)
- Marcel N. Moyeh
- Department of Biochemistry & Molecular Biology, University of Buea, PB 63, Buea, Cameroon
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
| | - Dieudonne L. Njimoh
- Department of Biochemistry & Molecular Biology, University of Buea, PB 63, Buea, Cameroon
| | - Marie Solange Evehe
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
- Department of Biochemistry, University of Yaoundé I, PB 812, Yaoundé, Cameroon
| | - Innocent M. Ali
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
- Department of Biochemistry, University of Dschang, BP 67, Dschang, Cameroon
| | - Akindeh M. Nji
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
- Department of Biochemistry, University of Yaoundé I, PB 812, Yaoundé, Cameroon
| | - Dominique N. Nkafu
- Department of Biochemistry & Molecular Biology, University of Buea, PB 63, Buea, Cameroon
| | - Palmer N. Masumbe
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
- Department of Biochemistry, University of Yaoundé I, PB 812, Yaoundé, Cameroon
| | - Atogho-Tiedeu Barbara
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
- Department of Biochemistry, University of Yaoundé I, PB 812, Yaoundé, Cameroon
| | - Valentine N. Ndikum
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
| | - Wilfred F. Mbacham
- Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, BP 8094, Yaoundé, Cameroon
- Department of Biochemistry, University of Yaoundé I, PB 812, Yaoundé, Cameroon
| |
Collapse
|
31
|
Kasule J, Gabriel EE, Anok A, Neal J, Eastman RT, Penzak S, Newell K, Serwadda D, Duffy PE, Reynolds SJ, Hobbs CV. Sulfamethoxazole Levels in HIV-Exposed Uninfected Ugandan Children. Am J Trop Med Hyg 2018; 98:1718-1721. [PMID: 29692311 PMCID: PMC6086194 DOI: 10.4269/ajtmh.17-0933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Trimethoprim–sulfamethoxazole (TMP–SMX) prophylaxis in HIV-uninfected, exposed (HUE) children variably reduces clinical malaria burden despite antifolate resistance, but data regarding achieved serum levels and adherence are lacking. Serum samples from 70 HUE children aged 3–12 months from Rakai, Uganda, enrolled in an observational study were assayed for random SMX levels using a colorimetric assay. Adherence with TMP–SMX prophylaxis data (yes/no) was also collected. Of 148 visits with concurrent SMX levels available, 56% had self-reported adherence with TMP–SMX therapy. Among these 82 visits, mean (standard deviation) level was 19.78 (19.22) µg/mL, but 33% had SMX levels below half maximal inhibitory concentrations (IC50) for Plasmodium falciparum with some, but not all, of the reported antifolate resistance mutations reported in Uganda. With TMP–SMX prophylaxis, suboptimal adherence is concerning. Sulfamethoxazole levels below IC50s required to overcome malaria parasites with multiple antifolate resistance mutations may be significant. Further study of TMP–SMX in this context is needed.
Collapse
Affiliation(s)
- Jingo Kasule
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Rakai Health Sciences Program, Kalisizo, Uganda
| | - Erin E Gabriel
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Aggrey Anok
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Rakai Health Sciences Program, Kalisizo, Uganda
| | - Jillian Neal
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Richard T Eastman
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Scott Penzak
- College of Pharmacy, University of North Texas, Fort Worth, Texas
| | - Kevin Newell
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., National Cancer Institute Campus at Frederick, Frederick, Maryland
| | - David Serwadda
- School of Public Health, Makerere College of Health Sciences, Kampala, Uganda.,Rakai Health Sciences Program, Kalisizo, Uganda
| | - Patrick E Duffy
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Steven J Reynolds
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Rakai Health Sciences Program, Kalisizo, Uganda
| | - Charlotte V Hobbs
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,University of Mississippi Medical Center, Batson Children's Hospital, Jackson, Mississippi
| |
Collapse
|
32
|
Grais RF, Laminou IM, Woi-Messe L, Makarimi R, Bouriema SH, Langendorf C, Amambua-Ngwa A, D'Alessandro U, Guérin PJ, Fandeur T, Sibley CH. Molecular markers of resistance to amodiaquine plus sulfadoxine-pyrimethamine in an area with seasonal malaria chemoprevention in south central Niger. Malar J 2018; 17:98. [PMID: 29486766 PMCID: PMC5830055 DOI: 10.1186/s12936-018-2242-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/17/2018] [Indexed: 04/04/2023] Open
Abstract
Background In Niger, malaria transmission is markedly seasonal with most of the disease burden occurring in children during the rainy season. Seasonal malaria chemoprevention (SMC) with amodiaquine plus sulfadoxine–pyrimethamine (AQ + SP) is recommended in the country to be administered monthly just before and during the rainy season. Moreover, clinical decisions on use of SP for intermittent preventive treatment in pregnancy (IPTp) now depend upon the validated molecular markers for SP resistance in Plasmodium falciparum observed in the local parasite population. However, little is known about molecular markers of resistance for either SP or AQ in the south of Niger. To address this question, clinical samples which met clinical and biological criteria, were collected in Gabi, Madarounfa district, Maradi region, Niger in 2011–2012 (before SMC implementation). Molecular markers of resistance to pyrimethamine (pfdhfr), sulfadoxine (pfdhps) and amodiaquine (pfmdr1) were assessed by DNA sequencing. Results Prior to SMC implementation, the samples showed a high proportion of clinical samples that carried the pfdhfr 51I/59R/108N haplotype associated with resistance to pyrimethamine and pfdhps 436A/F/H and 437G mutations associated with reduced susceptibility to sulfadoxine. In contrast mutations in codons 581G, and 613S in the pfdhps gene, and in pfmdr1, 86Y, 184Y, 1042D and 1246Y associated with resistance to amodiaquine, were less frequently observed. Importantly, pfdhfr I164L and pfdhps K540E mutations shown to be the most clinically relevant markers for high level clinical resistance to SP were not detected in Gabi. Conclusions Although parasites with genotypes associated with the highest levels of resistance to AQ + SP are not yet common in this setting, their importance for deployment of SMC and IPTp dictates that monitoring of these markers of resistance should accompany these interventions. This study also highlights the parasite heterogeneity within a small spatial area and the need to use caution when extrapolating results from surveys of molecular markers of resistance in a single site to inform regional policy decisions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Philippe J Guérin
- WorldWide Antimalarial Resistance Network, University of Oxford, Oxford, UK
| | - Thierry Fandeur
- CERMES, BP 11887, Niamey, Niger.,Division Internationale, Institut Pasteur, 28 rue du Dr Roux, 75725, Paris Cedex 15, France
| | - Carol H Sibley
- WorldWide Antimalarial Resistance Network, University of Washington, Seattle, USA
| |
Collapse
|
33
|
Changing Antimalarial Drug Sensitivities in Uganda. Antimicrob Agents Chemother 2017; 61:AAC.01516-17. [PMID: 28923866 DOI: 10.1128/aac.01516-17] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/03/2017] [Indexed: 11/20/2022] Open
Abstract
Dihydroartemisinin-piperaquine (DP) has demonstrated excellent efficacy for the treatment and prevention of malaria in Uganda. However, resistance to both components of this regimen has emerged in Southeast Asia. The efficacy of artemether-lumefantrine, the first-line regimen to treat malaria in Uganda, has also been excellent, but continued pressure may select for parasites with decreased sensitivity to lumefantrine. To gain insight into current drug sensitivity patterns, ex vivo sensitivities were assessed and genotypes previously associated with altered drug sensitivity were characterized for 58 isolates collected in Tororo, Uganda, from subjects presenting in 2016 with malaria from the community or as part of a clinical trial comparing DP chemoprevention regimens. Compared to community isolates, those from trial subjects had lower sensitivities to the aminoquinolines chloroquine, monodesethyl amodiaquine, and piperaquine and greater sensitivities to lumefantrine and mefloquine, an observation consistent with DP selection pressure. Compared to results for isolates from 2010 to 2013, the sensitivities of 2016 community isolates to chloroquine, amodiaquine, and piperaquine improved (geometric mean 50% inhibitory concentrations [IC50] = 248, 76.9, and 19.1 nM in 2010 to 2013 and 33.4, 14.9, and 7.5 nM in 2016, respectively [P < 0.001 for all comparisons]), the sensitivity to lumefantrine decreased (IC50 = 3.0 nM in 2010 to 2013 and 5.4 nM in 2016 [P < 0.001]), and the sensitivity to dihydroartemisinin was unchanged (IC50 = 1.4 nM). These changes were accompanied by decreased prevalence of transporter mutations associated with aminoquinoline resistance and low prevalence of polymorphisms recently associated with resistance to artemisinins or piperaquine. Antimalarial drug sensitivities are changing in Uganda, but novel genotypes associated with DP treatment failure in Asia are not prevalent.
Collapse
|
34
|
Lynch CA, Pearce R, Pota H, Egwang C, Egwang T, Bhasin A, Cox J, Abeku TA, Roper C. Travel and the emergence of high-level drug resistance in Plasmodium falciparum in southwest Uganda: results from a population-based study. Malar J 2017; 16:150. [PMID: 28415996 PMCID: PMC5392983 DOI: 10.1186/s12936-017-1812-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/08/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The I164L mutation on the dhfr gene confers high level resistance to sulfadoxine-pyrimethamine (SP) but it is rare in Africa except in a cluster of reports where prevalence >10% in highland areas of southwest Uganda and eastern Rwanda. The occurrence of the dhfr I164L mutation was investigated in community surveys in this area and examined the relationship to migration. METHODS A cross-sectional prevalence survey was undertaken in among villages within the catchment areas of two health facilities in a highland site (Kabale) and a highland fringe site (Rukungiri) in 2007. Sociodemographic details, including recent migration, were collected for each person included in the study. A total of 206 Plasmodium falciparum positive subjects were detected by rapid diagnostic test; 203 in Rukungiri and 3 in Kabale. Bloodspot samples were taken and were screened for dhfr I164L. RESULTS Sequence analysis confirmed the presence of the I164L mutations in twelve P. falciparum positive samples giving an estimated prevalence of 8.6% in Rukungiri. Of the three parasite positive samples in Kabale, none had I164L mutations. Among the twelve I164L positives three were male, ages ranged from 5 to 90 years of age. None of those with the I164L mutation had travelled in the 8 weeks prior to the survey, although three were from households from which at least one household member had travelled during that period. Haplotypes were determined in non-mixed infections and showed the dhfr I164L mutation occurs in both as a N51I + S108N + I164L haplotype (n = 2) and N51I + C59R + S108N + I164L haplotype (n = 5). Genotyping of flanking microsatellite markers showed that the I164L occurred independently on the triple mutant (N51I, C59R + S108N) and double mutant (N51I + S108N) background. CONCLUSIONS There is sustained local transmission of parasites with the dhfr I164L mutation in Rukungiri and no evidence to indicate its occurrence is associated with recent travel to highly resistant neighbouring areas. The emergence of a regional cluster of I164L in SW Uganda and Rwanda indicates that transmission of I164L is facilitated by strong drug pressure in low transmission areas potentially catalysed in those areas by travel and the importation of parasites from relatively higher transmission settings.
Collapse
Affiliation(s)
- Caroline A Lynch
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| | - Richard Pearce
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Hirva Pota
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | | | | | - Amit Bhasin
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Jonathan Cox
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Cally Roper
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
35
|
Gil JP, Krishna S. pfmdr1 (Plasmodium falciparum multidrug drug resistance gene 1): a pivotal factor in malaria resistance to artemisinin combination therapies. Expert Rev Anti Infect Ther 2017; 15:527-543. [DOI: 10.1080/14787210.2017.1313703] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- J. Pedro Gil
- Physiology and Pharmacology Department, Karolinska Institutet, Stockholm, Sweden
| | - S. Krishna
- St George’s University Hospital, Institute for Infection and Immunity, London, United Kingdom
| |
Collapse
|
36
|
Tukwasibwe S, Tumwebaze P, Conrad M, Arinaitwe E, Kamya MR, Dorsey G, Nsobya SL, Greenhouse B, Rosenthal PJ. Drug resistance mediating Plasmodium falciparum polymorphisms and clinical presentations of parasitaemic children in Uganda. Malar J 2017; 16:125. [PMID: 28327148 PMCID: PMC5361791 DOI: 10.1186/s12936-017-1777-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/15/2017] [Indexed: 11/19/2022] Open
Abstract
Background Plasmodium falciparum genetic polymorphisms that mediate altered drug sensitivity may impact upon virulence. In a cross-sectional study, Ugandan children with infections mutant at pfcrt K76T, pfmdr1 N86Y, or pfmdr1 D1246Y had about one-fourth the odds of symptomatic malaria compared to those with infections with wild type (WT) sequences. However, results may have been confounded by greater likelihood in those with symptomatic disease of higher density mixed infections and/or recent prior treatment that selected for WT alleles. Methods Polymorphisms in samples from paired episodes of asymptomatic and symptomatic parasitaemia in 114 subjects aged 4–11 years were followed longitudinally in Tororo District, Uganda. Paired episodes occurred within 3–12 months of each other and had no treatment for malaria in the prior 60 days. The prevalence of WT, mixed, and mutant alleles was determined using multiplex ligase detection reaction-fluorescent microsphere assays. Results Considering paired episodes in the same subject, the odds of symptomatic malaria were lower for infections with mutant compared to WT or mixed sequence at N86Y (OR 0.26, 95% CI 0.09–0.79, p = 0.018), but not K76T or D1246Y. However, symptomatic episodes (which had higher densities) were more likely than asymptomatic to be mixed (for N86Y OR 2.0, 95% CI 1.04–4.0, p = 0.036). Excluding mixed infections, the odds of symptomatic malaria were lower for infections with mutant compared to WT sequence at N86Y (OR 0.33, 95% CI 0.11–0.98, p = 0.046), but not the other alleles. However, if mixed genotypes were grouped with mutants in this analysis or assuming that mixed infections consisted of 50% WT and 50% mutant genotypes, the odds of symptomatic infection did not differ between infections that were mutant or WT at the studied alleles. Conclusions Although infections with only the mutant pfmdr1 86Y genotype were associated with symptomatic infection, this association could primarily be explained by greater parasite densities and therefore greater prevalence of mixed infections in symptomatic children. These results indicate limited association between the tested polymorphisms and risk of symptomatic disease and highlight the value of longitudinal studies for assessing associations between parasite factors and clinical outcomes.
Collapse
Affiliation(s)
| | | | - Melissa Conrad
- Department of Medicine, University of California, Box 0811, San Francisco, CA, 94143, USA
| | | | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California, Box 0811, San Francisco, CA, 94143, USA
| | | | - Bryan Greenhouse
- Department of Medicine, University of California, Box 0811, San Francisco, CA, 94143, USA
| | - Philip J Rosenthal
- Department of Medicine, University of California, Box 0811, San Francisco, CA, 94143, USA.
| |
Collapse
|
37
|
Tumwebaze P, Tukwasibwe S, Taylor A, Conrad M, Ruhamyankaka E, Asua V, Walakira A, Nankabirwa J, Yeka A, Staedke SG, Greenhouse B, Nsobya SL, Kamya MR, Dorsey G, Rosenthal PJ. Changing Antimalarial Drug Resistance Patterns Identified by Surveillance at Three Sites in Uganda. J Infect Dis 2017; 215:631-635. [PMID: 28039354 PMCID: PMC5853976 DOI: 10.1093/infdis/jiw614] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/07/2016] [Indexed: 11/12/2022] Open
Abstract
We assessed Plasmodium falciparum drug resistance markers in parasites collected in 2012, 2013, and 2015 at 3 sites in Uganda. The prevalence and frequency of parasites with mutations in putative transporters previously associated with resistance to aminoquinolines, but increased sensitivity to lumefantrine (pfcrt 76T; pfmdr1 86Y and 1246Y), decreased markedly at all sites. Antifolate resistance mutations were common, with apparent emergence of mutations (pfdhfr 164L; pfdhps 581G) associated with high-level resistance. K13 mutations linked to artemisinin resistance were uncommon and did not increase over time. Changing malaria treatment practices have been accompanied by profound changes in markers of resistance.
Collapse
Affiliation(s)
| | | | - Aimee Taylor
- Center for Communicable Disease Dynamics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Melissa Conrad
- Department of Medicine, University of California, San Francisco, USA
| | | | - Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Adoke Yeka
- Makerere University School of Public Health, College of Health Sciences, Kampala, Uganda
| | - Sarah G Staedke
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, USA
| | | | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, USA
| | | |
Collapse
|
38
|
Mwanza S, Joshi S, Nambozi M, Chileshe J, Malunga P, Kabuya JBB, Hachizovu S, Manyando C, Mulenga M, Laufer M. The return of chloroquine-susceptible Plasmodium falciparum malaria in Zambia. Malar J 2016; 15:584. [PMID: 27919256 PMCID: PMC5139104 DOI: 10.1186/s12936-016-1637-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/24/2016] [Indexed: 11/22/2022] Open
Abstract
Background Plasmodium falciparum resistance to anti-malarial drugs remains a major obstacle to malaria control and elimination. The parasite has developed resistance to every anti-malarial drug introduced for wide-scale treatment. However, the spread of resistance may be reversible. Malawi was the first country to discontinue chloroquine use due to widespread resistance. Within a decade of the removal of drug pressure, the molecular marker of chloroquine-resistant malaria had disappeared and the drug was shown to have excellent clinical efficacy. Many countries have observed decreases in the prevalence of chloroquine resistance with the discontinuation of chloroquine use. In Zambia, chloroquine was used as first-line treatment for uncomplicated malaria until treatment failures led the Ministry of Health to replace it with artemether-lumefantrine in 2003. Specimens from a recent study were analysed to evaluate prevalence of chloroquine-resistant malaria in Nchelenge district a decade after chloroquine use was discontinued. Methods Parasite DNA was extracted from dried blood spots collected by finger-prick in pregnant women who were enrolling in a clinical trial. The specimens underwent pyrosequencing to determine the genotype of the P. falciparum chloroquine resistance transporter, the gene that is associated with CQ resistance. Results Three-hundred and two specimens were successfully analysed. No chloroquine-resistant genotypes were detected. Conclusion The study found the disappearance of chloroquine-resistant malaria after the removal of chloroquine drug pressure. Chloroquine may have a role for malaria prevention or treatment in Zambia and throughout the region in the future.
Collapse
Affiliation(s)
- Sydney Mwanza
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Sudhaunshu Joshi
- Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201-1509, USA
| | - Michael Nambozi
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Justin Chileshe
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Phidelis Malunga
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Jean-Bertin Bukasa Kabuya
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Sebastian Hachizovu
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Christine Manyando
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Modest Mulenga
- Tropical Diseases Research Centre (TDRC), 6th and 7th Floors, Ndola Central Hospital Building, P.O. Box 71769, Ndola, Zambia
| | - Miriam Laufer
- Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201-1509, USA.
| |
Collapse
|
39
|
Kateera F, Nsobya SL, Tukwasibwe S, Hakizimana E, Mutesa L, Mens PF, Grobusch MP, van Vugt M, Kumar N. Molecular surveillance of Plasmodium falciparum drug resistance markers reveals partial recovery of chloroquine susceptibility but sustained sulfadoxine-pyrimethamine resistance at two sites of different malaria transmission intensities in Rwanda. Acta Trop 2016; 164:329-336. [PMID: 27647575 PMCID: PMC10600949 DOI: 10.1016/j.actatropica.2016.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 12/23/2022]
Abstract
Faced with intense levels of chloroquine (CQ) resistance in Plasmodium falciparum malaria, Rwanda replaced CQ with amodiaquine (AQ)+sulfadoxine-pyrimethamine (SP) in 2001, and subsequently with artemether-lumefantrine (AL) in 2006, as first-line treatments for uncomplicated malaria. Following years of discontinuation of CQ use, re-emergence of CQ-susceptible parasites has been reported in countries including Malawi, Kenya and Tanzania. In contrast, high levels of SP resistant mutant parasites continue to be reported even in countries of presumed reduced SP drug selection pressure. The prevalence and distributions of genetic polymorphisms linked with CQ and SP resistance at two sites of different malaria transmission intensities are described here to better understand drug-related genomic adaptations over time and exposure to varying drug pressures in Rwanda. Using filter paper blood isolates collected from P. falciparum infected patients, DNA was extracted and a nested PCR performed to identify resistance-mediating polymorphisms in the pfcrt, pfmdr1, pfdhps and pfdhfr genes. Amplicons from a total of 399 genotyped samples were analysed by ligase detection reaction fluorescent microsphere assay. CQ susceptible pfcrt 76K and pfmdr1 86N wild-type parasites were found in about 50% and 81% of isolates, respectively. Concurrently, SP susceptible pfdhps double (437G-540E), pfdhfr triple (108N-51I-59R), quintuple pfdhps 437G-540E/pfdhfr 51I-59R-108N and sextuple haplotypes were found in about 84%, 85%, 74% and 18% of isolates, respectively. High-level SP resistance associated pfdhfr 164L and pfdhps 581G mutant prevalences were noted to decline. Mutations pfcrt 76T, pfdhfr 59R and pfdhfr 164L were found differentially distributed between the two study sites with the pfdhfr 164L mutants found only at Ruhuha site, eastern Rwanda. Overall, sustained intense levels of SP resistance mutations and a recovery of CQ susceptible parasites were found in this study following 7 years and 14 years of drug withdrawal from use, respectively. Most likely, the sustained high prevalence of resistant parasites is due to the use of DHFR/DHPS inhibitors like trimethoprim-sulfamethoxazole (TS) for the treatment of and prophylaxis against bacterial infections among HIV infected individuals as well as the continued use of IPTp-SP within the East and Central African regions for malaria prevention among pregnant women. With regard to CQ, the slow recovery of CQ susceptible parasites may have been caused partly by the continued use of CQ and/or CQ mimicking antimalarial drugs like AQ in spite of policies to withdraw it from Rwanda and the neighbouring countries of Uganda and Tanzania. Continued surveillance of P. falciparum CQ and SP associated polymorphisms is recommended for guiding future rational drug policy-making and mitigation of future risk of anti-malaria drug resistance development.
Collapse
Affiliation(s)
- Fredrick Kateera
- Medical Research Centre Division, Rwanda Biomedical Centre, PO Box 7162, Kigali, Rwanda; Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
| | - Sam L Nsobya
- Molecular Research Laboratory, Infectious Disease Research Collaboration (IDRC), New Mulago Hospital Complex, PO Box 7051, Kampala, Uganda; School of Biomedical Science, College of Medicine, Makerere University, Uganda.
| | - Steven Tukwasibwe
- Molecular Research Laboratory, Infectious Disease Research Collaboration (IDRC), New Mulago Hospital Complex, PO Box 7051, Kampala, Uganda.
| | - Emmanuel Hakizimana
- Medical Research Centre Division, Rwanda Biomedical Centre, PO Box 7162, Kigali, Rwanda; Malaria & Other Parasitic Diseases Division, Rwanda Biomedical Centre, Kigali, Rwanda.
| | - Leon Mutesa
- College of Medicine & Health Sciences, University of Rwanda, PO Box 3286, Kigali, Rwanda.
| | - Petra F Mens
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands; Royal Tropical Institute/Koninklijk Instituutvoor de Tropen, KIT Biomedical Research, Meibergdreef 39, 1105 AZ, Amsterdam, The Netherlands.
| | - Martin P Grobusch
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
| | - Michèle van Vugt
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
| | - Nirbhay Kumar
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Vector-Borne Infectious Disease Research Centre, Tulane University, 333S Liberty Street, Mail code 8317, New Orleans, LA 70112, United States.
| |
Collapse
|
40
|
Taylor AR, Flegg JA, Holmes CC, Guérin PJ, Sibley CH, Conrad MD, Dorsey G, Rosenthal PJ. Artemether-Lumefantrine and Dihydroartemisinin-Piperaquine Exert Inverse Selective Pressure on Plasmodium Falciparum Drug Sensitivity-Associated Haplotypes in Uganda. Open Forum Infect Dis 2016; 4:ofw229. [PMID: 28480232 PMCID: PMC5413987 DOI: 10.1093/ofid/ofw229] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/24/2016] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Altered sensitivity to multiple antimalarial drugs is mediated by polymorphisms in pfmdr1, which encodes the Plasmodium falciparum multidrug resistance transporter. In Africa the N86Y and D1246Y polymorphisms have been shown to be selected by treatment, with artemether-lumefantrine (AL) and dihydroartemisinin-piperaquine (DP) selecting for wild-type and mutant alleles, respectively. However, there has been little study of pfmdr1 haplotypes, in part because haplotype analyses are complicated by multiclonal infections. METHODS We fit a haplotype frequency estimation model, which accounts for multiclonal infections, to the polymorphic pfmdr1 N86Y, Y184F, and D1246Y alleles in samples from a longitudinal trial comparing AL and DP to treat uncomplicated P falciparum malaria in Tororo, Uganda from 2007 to 2012. We regressed estimates onto covariates of trial arm and selective drug pressure. RESULTS Yearly trends showed increasing frequency estimates for haplotypes with wild type pfmdr1 N86 and D1246 alleles and decreasing frequency estimates for haplotypes with the mutant pfmdr1 86Y allele. Considering days since prior therapy, we saw evidence suggestive of selection by AL for haplotypes with N86 combined with 184F, D1246, or both, and against all haplotypes with 86Y, and evidence suggestive of selection by DP for 86Y only when combined with Y184 and 1246Y (haplotype YYY) and against haplotypes NFD and NYY. CONCLUSIONS Based on our model, AL selected several haplotypes containing N86, whereas DP selection was haplotype specific, demonstrating the importance of haplotype analyses. Inverse selective pressure of AL and DP on the complementary haplotypes NFD and YYY suggests that rotating artemisinin-based antimalarial combination regimens may be the best treatment option to prevent resistance selection.
Collapse
Affiliation(s)
- Aimee R Taylor
- WorldWide Antimalarial Resistance Network, University of Oxford, United Kingdom.,Department of Statistics, University of Oxford, United Kingdom.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Jennifer A Flegg
- WorldWide Antimalarial Resistance Network, University of Oxford, United Kingdom.,School of Mathematical Sciences and Monash Academy for Cross and Interdisciplinary Mathematical Applications, Monash University, Melbourne, Australia
| | - Chris C Holmes
- Department of Statistics, University of Oxford, United Kingdom
| | - Philippe J Guérin
- WorldWide Antimalarial Resistance Network, University of Oxford, United Kingdom.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Carol H Sibley
- WorldWide Antimalarial Resistance Network, University of Oxford, United Kingdom.,Department of Genome Sciences, University of Washington, Seattle
| | | | - Grant Dorsey
- Department of Medicine, University of California, San Francisco
| | | |
Collapse
|
41
|
Intermittent Preventive Treatment with Dihydroartemisinin-Piperaquine in Ugandan Schoolchildren Selects for Plasmodium falciparum Transporter Polymorphisms That Modify Drug Sensitivity. Antimicrob Agents Chemother 2016; 60:5649-54. [PMID: 27401569 DOI: 10.1128/aac.00920-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/02/2016] [Indexed: 01/12/2023] Open
Abstract
Dihydroartemisinin-piperaquine (DP) offers prolonged protection against malaria, but its impact on Plasmodium falciparum drug sensitivity is uncertain. In a trial of intermittent preventive treatment in schoolchildren in Tororo, Uganda, in 2011 to 2012, monthly DP for 1 year decreased the incidence of malaria by 96% compared to placebo; DP once per school term offered protection primarily during the first month after therapy. To assess the impact of DP on selection of drug resistance, we compared the prevalence of key polymorphisms in isolates that emerged at different intervals after treatment with DP. Blood obtained monthly and at each episode of fever was assessed for P. falciparum parasitemia by microscopy. Samples from 160 symptomatic and 650 asymptomatic episodes of parasitemia were assessed at 4 loci (N86Y, Y184F, and D1246Y in pfmdr1 and K76T in pfcrt) that modulate sensitivity to aminoquinoline antimalarials, utilizing a ligase detection reaction-fluorescent microsphere assay. For pfmdr1 N86Y and pfcrt K76T, but not the other studied polymorphisms, the prevalences of mutant genotypes were significantly greater in children who had received DP within the past 30 days than in those not treated within 60 days (86Y, 18.0% versus 8.3% [P = 0.03]; 76T, 96.0% versus 86.1% [P = 0.05]), suggesting selective pressure of DP. Full sequencing of pfcrt in a subset of samples did not identify additional polymorphisms selected by DP. In summary, parasites that emerged soon after treatment with DP were more likely than parasites not under drug pressure to harbor pfmdr1 and pfcrt polymorphisms associated with decreased sensitivity to aminoquinoline antimalarials. (This study has been registered at ClinicalTrials.gov under no. NCT01231880.).
Collapse
|
42
|
Genetically Determined Response to Artemisinin Treatment in Western Kenyan Plasmodium falciparum Parasites. PLoS One 2016; 11:e0162524. [PMID: 27611315 PMCID: PMC5017781 DOI: 10.1371/journal.pone.0162524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/24/2016] [Indexed: 12/16/2022] Open
Abstract
Genetically determined artemisinin resistance in Plasmodium falciparum has been described in Southeast Asia. The relevance of recently described Kelch 13-propeller mutations for artemisinin resistance in Sub-Saharan Africa parasites is still unknown. Southeast Asia parasites have low genetic diversity compared to Sub-Saharan Africa, where parasites are highly genetically diverse. This study attempted to elucidate whether genetics provides a basis for discovering molecular markers in response to artemisinin drug treatment in P. falciparum in Kenya. The genetic diversity of parasites collected pre- and post- introduction of artemisinin combination therapy (ACT) in western Kenya was determined. A panel of 12 microsatellites and 91 single nucleotide polymorphisms (SNPs) distributed across the P. falciparum genome were genotyped. Parasite clearance rates were obtained for the post-ACT parasites. The 12 microsatellites were highly polymorphic with post-ACT parasites being significantly more diverse compared to pre-ACT (p < 0.0001). The median clearance half-life was 2.55 hours for the post-ACT parasites. Based on SNP analysis, 15 of 90 post-ACT parasites were single-clone infections. Analysis revealed 3 SNPs that might have some causal association with parasite clearance rates. Further, genetic analysis using Bayesian tree revealed parasites with similar clearance phenotypes were more closely genetically related. With further studies, SNPs described here and genetically determined response to artemisinin treatment might be useful in tracking artemisinin resistance in Kenya.
Collapse
|
43
|
Otienoburu SD, Maïga-Ascofaré O, Schramm B, Jullien V, Jones JJ, Zolia YM, Houzé P, Ashley EA, Kiechel JR, Guérin PJ, Le Bras J, Houzé S. Selection of Plasmodium falciparum pfcrt and pfmdr1 polymorphisms after treatment with artesunate-amodiaquine fixed dose combination or artemether-lumefantrine in Liberia. Malar J 2016; 15:452. [PMID: 27596849 PMCID: PMC5011943 DOI: 10.1186/s12936-016-1503-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 08/26/2016] [Indexed: 12/22/2022] Open
Abstract
Background Plasmodium falciparum uncomplicated malaria can successfully be treated with an artemisinin-based combination therapy (ACT). However resistance is spreading to the different ACT compounds; the artemisinin derivative and the partner drug. Studies of P. falciparum polymorphisms associated with drug resistance can provide a useful tool to track resistance and guide treatment policy as well as an in-depth understanding of the development and spread of resistance. Methods The role of P. falciparum molecular markers in selection of reinfections was assessed in an efficacy trial comparing artesunate–amodiaquine fixed-dose combination with artemether–lumefantrine to treat malaria in Nimba County, Liberia 2008–2009. P. falciparum polymorphisms in pfcrt 76, pfmdr1 86, 184 and 1246, and pfmrp1 876 and 1466 were analysed by PCR-RFLP and pyrosequencing. Results High baseline prevalence of pfmdr1 1246Y was found in Nimba county (38 %). Pfmdr1 1246Y and pfmdr1 86+184+1246 haplotypes NYY and YYY were selected in reinfections in the artesunate–amodiaquine arm and pfcrt K76, pfmdr1 N86 and pfmdr1 haplotype NFD were selected in artemether–lumefantrine reinfections. Parasites harbouring pfmdr1 1246Y could reinfect earlier after treatment with artesunate–amodiaquine and parasites carrying pfmdr1 N86 could reinfect at higher lumefantrine concentrations in patients treated with artemether–lumefantrine. Conclusions Although treatment is highly efficacious, selection of molecular markers in reinfections could indicate a decreased sensitivity or tolerance of parasites to the current treatments and the baseline prevalence of molecular markers should be closely monitored. Since individual drug levels and the day of reinfection were demonstrated to be key determinants for selection of reinfections, this data needs to be collected and taken into account for accurate evaluation of molecular markers for anti-malarial treatments. The protocols for the clinical trial was registered with Current Controlled Trials, under the Identifier Number ISRCTN51688713 on 9 October 2008
Collapse
Affiliation(s)
- Sabina Dahlström Otienoburu
- Institut de Médecine et d'Epidémiologie Appliquée, Bichat-C. Bernard Hospital, Paris, France. .,WorldWide Antimalarial Resistance Network, Oxford, UK. .,Johnson C. Smith University, Charlotte, NC, 28216, USA.
| | - Oumou Maïga-Ascofaré
- Institut de Médecine et d'Epidémiologie Appliquée, Bichat-C. Bernard Hospital, Paris, France.,Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | | | - Vincent Jullien
- INSERM U1129, Service de Pharmacologie, Hôpital Européen Georges Pompidou, Université Paris Descartes, 75015, Paris, France
| | - Joel J Jones
- National Malaria Control Programme, Ministry of Health and Social Welfare, Monrovia, Liberia
| | - Yah M Zolia
- National Malaria Control Programme, Ministry of Health and Social Welfare, Monrovia, Liberia
| | - Pascal Houzé
- AP-HP, Saint-Louis Hospital Biochemistry Laboratory, Paris, France
| | - Elizabeth A Ashley
- Epicentre, 75012, Paris, France.,Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | | | - Philippe J Guérin
- WorldWide Antimalarial Resistance Network, Oxford, UK.,Epicentre, 75012, Paris, France.,Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Jacques Le Bras
- Institut de Médecine et d'Epidémiologie Appliquée, Bichat-C. Bernard Hospital, Paris, France.,IRD UMR216, Paris-Descartes University, Paris, France.,Parasitology Laboratory-French National Malaria Reference Centre, AP-HP, Bichat-C. Bernard Hospital, Paris, France
| | - Sandrine Houzé
- IRD UMR216, Paris-Descartes University, Paris, France.,Parasitology Laboratory-French National Malaria Reference Centre, AP-HP, Bichat-C. Bernard Hospital, Paris, France
| |
Collapse
|
44
|
Atroosh WM, Al-Mekhlafi HM, Al-Jasari A, Sady H, Dawaki SS, Elyana FN, Al-Areeqi MA, Nasr NA, Abdulsalam AM, Subramaniam LR, Azzani M, Ithoi I, Lau YL, Surin J. Different patterns of pfcrt and pfmdr1 polymorphism in Plasmodium falciparum isolates from Tehama region, Yemen. PeerJ 2016; 4:e2191. [PMID: 27478699 PMCID: PMC4950566 DOI: 10.7717/peerj.2191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 06/08/2016] [Indexed: 11/20/2022] Open
Abstract
Introduction. Despite the efforts of the malaria control programme, malaria morbidity is still a common health problem in Yemen, with 60% of the population at risk. Plasmodium falciparum is responsible for 99% of malaria cases. The emergence in Yemen of parasite resistance to chloroquine (CQ) prompted the adoption of artemisinin combination therapy (ACT) in 2009, which involves the use of artesunate plus sulphadoxine-pyrimethamine (AS + SP). However, CQ was retained as the drug of choice for vivax malaria. To assess the impact of the change in the malaria treatment policy five years after its introduction, the present study investigated the mutations in the CQ resistance transporter (pfcrt) and multidrug resistance 1 (pfmdr1) genes. Method. A molecular investigation of 10 codons of pfcrt (72–76, 220, 271, 326, 356, and 371) and five codons of pfmdr1 (86, 184, 1034, 1042, and 1246) was conducted on P. falciparum isolates from districts with the highest malaria endemicity in the Hodeidah and Al-Mahwit governorates in Tehama region, Yemen. A total of 86 positive cases of falciparum monoinfection were investigated for the presence of mutations related to CQ and other antimalarials using a PCR-RFLP assay. Results. There was a wide prevalence of pfcrt gene mutations with the pfcrt 76T CQ resistance marker being predominant (97.7%). The prevalence of other pfcrt mutations varied from high (75E: 88%) to moderate (74I: 79.1%, 220S: 69.8%, 271E and 371I: 53.5%) or low (326S: 36%, 72S: 10.5%). Mutated pfcrt 72–76 amino acids haplotypes were highly prevalent (98.8%). Among these, the CVIET classic, old-world African/Southeast Asian haplotype was the most predominant, and was mostly found in the isolates from the Khamis Bani Saad district of Al-Mahwit (93.1%) and the AdDahi district of Hodeidah (88.9%). However, it was only found in 26.3% of the isolates from the Bajil district of Hodeidah. Surprisingly, the SVMNT new-world South American haplotype was exclusively detected in 9.3% of the isolates from the Bajil district of Hodeidah. Mutations at Y184F of pfmdr1 were found in all isolates (100%) from all districts. The mutation for codons 1034C and 86Y were found only in the isolates from the AdDahi and Khamis Bani Saad districts. Overall, the AdDahi and Khamis Bani Saad districts were similar in terms of carrying most of the mutations in the pfcrt and pfmdr1 genes, while there was a lower prevalence of mutation in the isolates from the Bajil district. Conclusion. The high prevalence of mutations in pfcrt 5 years after the official cessation of CQ use against P. falciparum suggests that there is sustained CQ pressure on P. falciparum isolates in the study area. Moreover, the low prevalence of mutations in the pfmdr1 gene could be a good indicator of the high susceptibility of P. falciparum isolates to antimalarials other than CQ. A new strategy to ensure the complete nationwide withdrawal of CQ from the private drug market is recommended.
Collapse
Affiliation(s)
- Wahib M Atroosh
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Unit of Microbiology and Parasitology, Department of Para-Clinic, Faculty of Medicine and Health Sciences, University of Aden, Khormaksar, Aden, Yemen
| | - Hesham M Al-Mekhlafi
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Endemic and Tropical Diseases Unit, Medical Research Centre, Jazan University, Jazan, Kingdom of Saudi Arabia; Department of Parasitology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Yemen
| | - Adel Al-Jasari
- National Malaria Control Programme, Ministry of Health and Population , Sana'a , Yemen
| | - Hany Sady
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Salwa S Dawaki
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Fatin N Elyana
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Mona A Al-Areeqi
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Nabil A Nasr
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Awatif M Abdulsalam
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Lahvanya R Subramaniam
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Meram Azzani
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Init Ithoi
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Johari Surin
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Centre for Research and Innovation, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
45
|
Eastman RT, Khine P, Huang R, Thomas CJ, Su XZ. PfCRT and PfMDR1 modulate interactions of artemisinin derivatives and ion channel blockers. Sci Rep 2016; 6:25379. [PMID: 27147113 PMCID: PMC4857081 DOI: 10.1038/srep25379] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/15/2016] [Indexed: 01/01/2023] Open
Abstract
Treatment of the symptomatic asexual stage of Plasmodium falciparum relies almost exclusively on artemisinin (ART) combination therapies (ACTs) in endemic regions. ACTs combine ART or its derivative with a long-acting partner drug to maximize efficacy during the typical three-day regimen. Both laboratory and clinical studies have previously demonstrated that the common drug resistance determinants P. falciparum chloroquine resistance transporter (PfCRT) and multidrug resistance transporter (PfMDR1) can modulate the susceptibility to many current antimalarial drugs and chemical compounds. Here we investigated the parasite responses to dihydroartemisinin (DHA) and various Ca2+ and Na+ channel blockers and showed positively correlated responses between DHA and several channel blockers, suggesting potential shared transport pathways or mode of action. Additionally, we demonstrated that PfCRT and PfMDR1 could also significantly modulate the pharmacodynamic interactions of the compounds and that the interactions were influenced by the parasite genetic backgrounds. These results provide important information for better understanding of drug resistance and for assessing the overall impact of drug resistance markers on parasite response to ACTs.
Collapse
Affiliation(s)
- Richard T Eastman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,Division of Preclinical Development, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Pwint Khine
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ruili Huang
- Division of Preclinical Development, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Craig J Thomas
- Division of Preclinical Development, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Yeka A, Kigozi R, Conrad MD, Lugemwa M, Okui P, Katureebe C, Belay K, Kapella BK, Chang MA, Kamya MR, Staedke SG, Dorsey G, Rosenthal PJ. Artesunate/Amodiaquine Versus Artemether/Lumefantrine for the Treatment of Uncomplicated Malaria in Uganda: A Randomized Trial. J Infect Dis 2015; 213:1134-42. [PMID: 26597254 DOI: 10.1093/infdis/jiv551] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/11/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In treating malaria in Uganda, artemether-lumefantrine (AL) has been associated with a lower risk of recurrent parasitemia, compared with artesunate-amodiaquine (AS/AQ), but changing treatment practices may have altered parasite susceptibility. METHODS We enrolled 602 children aged 6-59 months with uncomplicated falciparum malaria from 3 health centers in 2013-2014 and randomly assigned them to receive treatment with AS/AQ or AL. Primary outcomes were risks of recurrent parasitemia within 28 days, with or without adjustment to distinguish recrudescence from new infection. Drug safety and tolerability and Plasmodium falciparum resistance-mediating polymorphisms were assessed. RESULTS Of enrolled patients, 594 (98.7%) completed the 28-day study. Risks of recurrent parasitemia were lower with AS/AQ at all 3 sites (overall, 28.6% vs 44.6%; P < .001). Recrudescences were uncommon, and all occurred after AL treatment (0% vs 2.5%; P = .006). Recovery of the hemoglobin level was greater with AS/AQ (1.73 vs 1.39 g/dL; P = .04). Both regimens were well tolerated; serious adverse events were uncommon (1.7% in the AS/AQ group and 1.0% in the AL group). AS/AQ selected for mutant pfcrt/pfmdr1 polymorphisms and AL for wild-type pfcrt/pfmdr1 polymorphisms associated with altered drug susceptibility. CONCLUSIONS AS/AQ treatment was followed by fewer recurrences than AL treatment, contrasting with older data. Each regimen selected for polymorphisms associated with decreased treatment response. Research should consider multiple or rotating regimens to maintain treatment efficacies.
Collapse
Affiliation(s)
- Adoke Yeka
- School of Public Health, Makerere University College of Health Sciences Infectious Diseases Research Collaboration, President's Malaria Initiative, Kampala, Uganda
| | - Ruth Kigozi
- Infectious Diseases Research Collaboration, President's Malaria Initiative, Kampala, Uganda
| | | | - Myers Lugemwa
- National Malaria Control Program, Ministry of Health, President's Malaria Initiative, Kampala, Uganda
| | - Peter Okui
- National Malaria Control Program, Ministry of Health, President's Malaria Initiative, Kampala, Uganda
| | - Charles Katureebe
- World Health Organization, President's Malaria Initiative, Kampala, Uganda
| | - Kassahun Belay
- US Agency for International Development, President's Malaria Initiative, Kampala, Uganda
| | - Bryan K Kapella
- Centers for Disease Control and Prevention (CDC), President's Malaria Initiative, Kampala, Uganda
| | - Michelle A Chang
- Malaria Branch, Division of Parasitic Diseases and Malaria, CDC, Atlanta, Georgia
| | - Moses R Kamya
- Department of Medicine, Makerere University College of Health Sciences Infectious Diseases Research Collaboration, President's Malaria Initiative, Kampala, Uganda
| | - Sarah G Staedke
- Infectious Diseases Research Collaboration, President's Malaria Initiative, Kampala, Uganda London School of Hygiene and Tropical Medicine, United Kingdom
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco
| | | |
Collapse
|
47
|
Cui L, Mharakurwa S, Ndiaye D, Rathod PK, Rosenthal PJ. Antimalarial Drug Resistance: Literature Review and Activities and Findings of the ICEMR Network. Am J Trop Med Hyg 2015; 93:57-68. [PMID: 26259943 PMCID: PMC4574275 DOI: 10.4269/ajtmh.15-0007] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 04/27/2015] [Indexed: 11/07/2022] Open
Abstract
Antimalarial drugs are key tools for the control and elimination of malaria. Recent decreases in the global malaria burden are likely due, in part, to the deployment of artemisinin-based combination therapies. Therefore, the emergence and potential spread of artemisinin-resistant parasites in southeast Asia and changes in sensitivities to artemisinin partner drugs have raised concerns. In recognition of this urgent threat, the International Centers of Excellence for Malaria Research (ICEMRs) are closely monitoring antimalarial drug efficacy and studying the mechanisms underlying drug resistance. At multiple sentinel sites of the global ICEMR network, research activities include clinical studies to track the efficacies of antimalarial drugs, ex vivo/in vitro assays to measure drug susceptibilities of parasite isolates, and characterization of resistance-mediating parasite polymorphisms. Taken together, these efforts offer an increasingly comprehensive assessment of the efficacies of antimalarial therapies, and enable us to predict the emergence of drug resistance and to guide local antimalarial drug policies. Here we briefly review worldwide antimalarial drug resistance concerns, summarize research activities of the ICEMRs related to drug resistance, and assess the global impacts of the ICEMR programs.
Collapse
Affiliation(s)
- Liwang Cui
- *Address correspondence to Liwang Cui, Department of Entomology, Pennsylvania State University, 501 ASI Building, University Park, PA 16802, E-mail: or Philip J. Rosenthal, Department of Medicine, Box 0811, University of California, San Francisco, CA 94110. E-mail:
| | | | | | | | - Philip J. Rosenthal
- *Address correspondence to Liwang Cui, Department of Entomology, Pennsylvania State University, 501 ASI Building, University Park, PA 16802, E-mail: or Philip J. Rosenthal, Department of Medicine, Box 0811, University of California, San Francisco, CA 94110. E-mail:
| |
Collapse
|
48
|
Adaptive evolution of malaria parasites in French Guiana: Reversal of chloroquine resistance by acquisition of a mutation in pfcrt. Proc Natl Acad Sci U S A 2015; 112:11672-7. [PMID: 26261345 DOI: 10.1073/pnas.1507142112] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In regions with high malaria endemicity, the withdrawal of chloroquine (CQ) as first-line treatment of Plasmodium falciparum infections has typically led to the restoration of CQ susceptibility through the reexpansion of the wild-type (WT) allele K76 of the chloroquine resistance transporter gene (pfcrt) at the expense of less fit mutant alleles carrying the CQ resistance (CQR) marker K76T. In low-transmission settings, such as South America, drug resistance mutations can attain 100% prevalence, thereby precluding the return of WT parasites after the complete removal of drug pressure. In French Guiana, despite the fixation of the K76T allele, the prevalence of CQR isolates progressively dropped from >90% to <30% during 17 y after CQ withdrawal in 1995. Using a genome-wide association study with CQ-sensitive (CQS) and CQR isolates, we have identified a single mutation in pfcrt encoding a C350R substitution that is associated with the restoration of CQ susceptibility. Genome editing of the CQR reference strain 7G8 to incorporate PfCRT C350R caused a complete loss of CQR. A retrospective molecular survey on 580 isolates collected from 1997 to 2012 identified all C350R mutant parasites as being CQS. This mutation emerged in 2002 and rapidly spread throughout the P. falciparum population. The C350R allele is also associated with a significant decrease in piperaquine susceptibility in vitro, suggesting that piperaquine pressure in addition to potential fitness costs associated with the 7G8-type CQR pfcrt allele may have selected for this mutation. These findings have important implications for understanding the evolutionary dynamics of antimalarial drug resistance.
Collapse
|
49
|
Takala-Harrison S, Laufer MK. Antimalarial drug resistance in Africa: key lessons for the future. Ann N Y Acad Sci 2015; 1342:62-7. [PMID: 25891142 DOI: 10.1111/nyas.12766] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Drug-resistant parasites repeatedly arise as a result of widespread use of antimalarial drugs and have contributed significantly to the failure to control and eradicate malaria throughout the world. In this review, we describe the spread of resistance to chloroquine and sulfadoxine-pyrimethamine, two old drugs that are no longer used owing to high rates of resistance, and examine the effect of the removal of drug pressure on the survival of resistant parasites. Artemisinin-resistant malaria is now emerging in Southeast Asia in a unique and unexpected pattern. We will review the most recent genomic and clinical data to help predict the behavior of resistance to new antimalarial medications and inform strategies to prevent the spread of drug-resistant malaria in Africa in the future.
Collapse
Affiliation(s)
- Shannon Takala-Harrison
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
50
|
Djimde AA, Makanga M, Kuhen K, Hamed K. The emerging threat of artemisinin resistance in malaria: focus on artemether-lumefantrine. Expert Rev Anti Infect Ther 2015; 13:1031-45. [PMID: 26081265 DOI: 10.1586/14787210.2015.1052793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The development of artemisinin resistance in the Greater Mekong Subregion poses a significant threat to malaria elimination. Artemisinin-based combination therapies including artemether-lumefantrine (AL) are recommended by WHO as first-line treatment for uncomplicated Plasmodium falciparum malaria. This article provides a comprehensive review of the existing and latest data as a basis for interpretation of observed variability in parasite sensitivity to AL over the last 5 years. Clinical efficacy and preclinical data from a range of endemic countries are summarized, including potential molecular markers of resistance. Overall, AL remains effective in the treatment of uncomplicated P. falciparum malaria in most regions. Establishing validated molecular markers for resistance and strict efficacy monitoring will reinforce timely updates of treatment policies.
Collapse
Affiliation(s)
- Abdoulaye A Djimde
- Malaria Research and Training Center, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | | | | | | |
Collapse
|