1
|
Abdi AI, Achcar F, Sollelis L, Silva-Filho JL, Mwikali K, Muthui M, Mwangi S, Kimingi HW, Orindi B, Andisi Kivisi C, Alkema M, Chandrasekar A, Bull PC, Bejon P, Modrzynska K, Bousema T, Marti M. Plasmodium falciparum adapts its investment into replication versus transmission according to the host environment. eLife 2023; 12:e85140. [PMID: 36916164 PMCID: PMC10059685 DOI: 10.7554/elife.85140] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/01/2023] [Indexed: 03/14/2023] Open
Abstract
The malaria parasite life cycle includes asexual replication in human blood, with a proportion of parasites differentiating to gametocytes required for transmission to mosquitoes. Commitment to differentiate into gametocytes, which is marked by activation of the parasite transcription factor ap2-g, is known to be influenced by host factors but a comprehensive model remains uncertain. Here, we analyze data from 828 children in Kilifi, Kenya with severe, uncomplicated, and asymptomatic malaria infection over 18 years of falling malaria transmission. We examine markers of host immunity and metabolism, and markers of parasite growth and transmission investment. We find that inflammatory responses associated with reduced plasma lysophosphatidylcholine levels are associated with markers of increased investment in parasite sexual reproduction (i.e. transmission investment) and reduced growth (i.e. asexual replication). This association becomes stronger with falling transmission and suggests that parasites can rapidly respond to the within-host environment, which in turn is subject to changing transmission.
Collapse
Affiliation(s)
- Abdirahman I Abdi
- KEMRI-Wellcome Trust Research ProgrammeKilifiKenya
- Pwani University Biosciences Research Centre, Pwani UniversityKilifiKenya
| | - Fiona Achcar
- Wellcome Center for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of ZurichZurichSwitzerland
| | - Lauriane Sollelis
- Wellcome Center for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of ZurichZurichSwitzerland
| | - João Luiz Silva-Filho
- Wellcome Center for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of ZurichZurichSwitzerland
| | | | | | | | | | | | - Cheryl Andisi Kivisi
- KEMRI-Wellcome Trust Research ProgrammeKilifiKenya
- Pwani University Biosciences Research Centre, Pwani UniversityKilifiKenya
| | - Manon Alkema
- Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Amrita Chandrasekar
- Wellcome Center for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
| | - Peter C Bull
- KEMRI-Wellcome Trust Research ProgrammeKilifiKenya
| | - Philip Bejon
- KEMRI-Wellcome Trust Research ProgrammeKilifiKenya
| | - Katarzyna Modrzynska
- Wellcome Center for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
| | - Teun Bousema
- Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Matthias Marti
- Wellcome Center for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of ZurichZurichSwitzerland
| |
Collapse
|
2
|
Waweru H, Kanoi BN, Kuja JO, Maranga M, Kongere J, Maina M, Kinyua J, Gitaka J. Limited genetic variations of the Rh5-CyRPA-Ripr invasion complex in Plasmodium falciparum parasite population in selected malaria-endemic regions, Kenya. FRONTIERS IN TROPICAL DISEASES 2023; 4:1102265. [PMID: 38406638 PMCID: PMC7615667 DOI: 10.3389/fitd.2023.1102265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Abstract
The invasion of human erythrocytes by Plasmodium falciparum merozoites requires interaction between parasite ligands and host receptors. Interaction of PfRh5-CyRPA-Ripr protein complex with basigin, an erythrocyte surface receptor, via PfRh5 is essential for erythrocyte invasion. Antibodies raised against each antigen component of the complex have demonstrated erythrocyte invasion inhibition, making these proteins potential blood-stage vaccine candidates. Genetic polymorphisms present a significant challenge in developing efficacious vaccines, leading to variant-specific immune responses. This study investigated the genetic variations of the PfRh5 complex proteins in P. falciparum isolates from Lake Victoria islands, Western Kenya. Here, twenty-nine microscopically confirmed P. falciparum field samples collected from islands in Lake Victoria between July 2014 and July 2016 were genotyped by whole genome sequencing, and results compared to sequences mined from the GenBank database, from a study conducted in Kilifi, as well as other sequences from the MalariaGEN repository. We analyzed the frequency of polymorphisms in the PfRh5 protein complex proteins, PfRh5, PfCyRPA, PfRipr, and PfP113, and their location mapped on the 3D protein complex structure. We identified a total of 58 variants in the PfRh5 protein complex. PfRh5 protein was the most polymorphic with 30 SNPs, while PfCyRPA was relatively conserved with 3 SNPs. The minor allele frequency of the SNPs ranged between 1.9% and 21.2%. Ten high-frequency alleles (>5%) were observed in PfRh5 at codons 147, 148, 277, 410, and 429 and in PfRipr at codons 190, 255, 259, and 1003. A SNP was located in protein-protein interaction region C203Y and F292V of PfRh5 and PfCyRPA, respectively. Put together, this study revealed low polymorphisms in the PfRh5 invasion complex in the Lake Victoria parasite population. However, the two mutations identified on the protein interaction regions prompts for investigation on their impacts on parasite invasion process to support the consideration of PfRh5 components as potential malaria vaccine candidates.
Collapse
Affiliation(s)
- Harrison Waweru
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Bernard N. Kanoi
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
- Centre for Research in Tropical Medicine and Community Development, Nairobi, Kenya
| | - Josiah O. Kuja
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mary Maranga
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - James Kongere
- Centre for Research in Tropical Medicine and Community Development, Nairobi, Kenya
| | - Michael Maina
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Johnson Kinyua
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Jesse Gitaka
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
- Centre for Research in Tropical Medicine and Community Development, Nairobi, Kenya
| |
Collapse
|
3
|
Temporal distribution of Plasmodium falciparum recrudescence following artemisinin-based combination therapy: an individual participant data meta-analysis. Malar J 2022; 21:106. [PMID: 35331243 PMCID: PMC8943927 DOI: 10.1186/s12936-021-03980-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/12/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The duration of trial follow-up affects the ability to detect recrudescent infections following anti-malarial treatment. The aim of this study was to explore the proportions of recrudescent parasitaemia as ascribed by genotyping captured at various follow-up time-points in treatment efficacy trials for uncomplicated Plasmodium falciparum malaria. METHODS Individual patient data from 83 anti-malarial efficacy studies collated in the WorldWide Antimalarial Resistance Network (WWARN) repository with at least 28 days follow-up were available. The temporal and cumulative distributions of recrudescence were characterized using a Cox regression model with shared frailty on study-sites. Fractional polynomials were used to capture non-linear instantaneous hazard. The area under the density curve (AUC) of the constructed distribution was used to estimate the optimal follow-up period for capturing a P. falciparum malaria recrudescence. Simulation studies were conducted based on the constructed distributions to quantify the absolute overestimation in efficacy due to sub-optimal follow-up. RESULTS Overall, 3703 recurrent infections were detected in 60 studies conducted in Africa (15,512 children aged < 5 years) and 23 studies conducted in Asia and South America (5272 patients of all ages). Using molecular genotyping, 519 (14.0%) recurrences were ascribed as recrudescent infections. A 28 day artemether-lumefantrine (AL) efficacy trial would not have detected 58% [95% confidence interval (CI) 47-74%] of recrudescences in African children and 32% [95% CI 15-45%] in patients of all ages in Asia/South America. The corresponding estimate following a 42 day dihydroartemisinin-piperaquine (DP) efficacy trial in Africa was 47% [95% CI 19-90%] in children under 5 years old treated with > 48 mg/kg total piperaquine (PIP) dose and 9% [95% CI 0-22%] in those treated with ≤ 48 mg/kg PIP dose. In absolute terms, the simulation study found that trials limited to 28 days follow-up following AL underestimated the risk of recrudescence by a median of 2.8 percentage points compared to day 63 estimates and those limited to 42 days following DP underestimated the risk of recrudescence by a median of 2.0 percentage points compared to day 42 estimates. The analysis was limited by few clinical trials following patients for longer than 42 days (9 out of 83 trials) and the imprecision of PCR genotyping which overcalls recrudescence in areas of higher transmission biasing the later distribution. CONCLUSIONS Restricting follow-up of clinical efficacy trials to day 28 for AL and day 42 for DP will miss a proportion of late recrudescent treatment failures but will have a modest impact in derived efficacy. The results highlight that as genotyping methods improve consideration should be given for trials with longer duration of follow-up to detect early indications of emerging drug resistance.
Collapse
|
4
|
Marwa K, Kapesa A, Baraka V, Konje E, Kidenya B, Mukonzo J, Kamugisha E, Swedberg G. Therapeutic efficacy of artemether-lumefantrine, artesunate-amodiaquine and dihydroartemisinin-piperaquine in the treatment of uncomplicated Plasmodium falciparum malaria in Sub-Saharan Africa: A systematic review and meta-analysis. PLoS One 2022; 17:e0264339. [PMID: 35271592 PMCID: PMC8912261 DOI: 10.1371/journal.pone.0264339] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Sub-Saharan Africa has the highest burden of malaria in the world. Artemisinin-based combination therapies (ACTs) have been the cornerstone in the efforts to reduce the global burden of malaria. In the effort to facilitate early detection of resistance for artemisinin derivatives and partner drugs, WHO recommends monitoring of ACT's efficacy in the malaria endemic countries. The present systematic meta-analysis study summarises the evidence of therapeutic efficacy of the commonly used artemisinin-based combinations for the treatment of uncomplicated P. falciparum malaria in Sub-Saharan Africa after more than a decade since the introduction of the drugs. METHODS Fifty two studies carried out from 2010 to 2020 on the efficacy of artemether-lumefantrine or dihydro-artemisinin piperaquine or artesunate amodiaquine in patients with uncomplicated P. falciparum malaria in Sub-Saharan Africa were searched for using the Google Scholar, Cochrane Central Register of controlled trials (CENTRAL), PubMed, Medline, LILACS, and EMBASE online data bases. Data was extracted by two independent reviewers. Random analysis effect was performed in STATA 13. Heterogeneity was established using I2 statistics. RESULTS Based on per protocol analysis, unadjusted cure rates in malaria infected patients treated with artemether-lumefantrine (ALU), artesunate-amodiaquine (ASAQ) and dihydroartemisinin-piperaquine (DHP) were 89%, 94% and 91% respectively. However, the cure rates after PCR correction were 98% for ALU, 99% for ASAQ and 99% for DHP. CONCLUSION The present meta-analysis reports the overall high malaria treatment success for artemether-lumefantrine, artesunate-amodiaquine and dihydroartemisinin-piperaquine above the WHO threshold value in Sub-Saharan Africa.
Collapse
Affiliation(s)
- Karol Marwa
- Department of Pharmacology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Anthony Kapesa
- Department of Community Medicine, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Vito Baraka
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Evelyne Konje
- Department of Epidemiology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Benson Kidenya
- Department of Biochemistry, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Jackson Mukonzo
- Department of Pharmacology and Therapeutics, Makerere University, Kampala, Uganda
| | - Erasmus Kamugisha
- Department of Biochemistry, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Gote Swedberg
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Assefa DG, Zeleke ED, Molla W, Mengistu N, Sefa A, Mebratu A, Bate AF, Bekele E, Yesmaw G, Makonnen E. Safety of dihydroartemisinin-piperaquine versus artemether-lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria among children in Africa: a systematic review and meta-analysis of randomized control trials. Malar J 2022; 21:4. [PMID: 34983552 PMCID: PMC8725395 DOI: 10.1186/s12936-021-04032-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 12/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The efficacies of artemisinin based combinations have been excellent in Africa, but also comprehensive evidence regarding their safety would be important. The aim of this review was to synthesize available evidence on the safety of dihydroartemisinin-piperaquine (DHA-PQ) compared to artemether-lumefantrine (AL) for the treatment of uncomplicated Plasmodium falciparum malaria among children in Africa. METHODS A systematic literature search was done to identify relevant articles from online databases PubMed/ MEDLINE, Embase, and Cochrane Center for Clinical Trial database (CENTRAL) for retrieving randomized control trials comparing safety of DHA-PQ and AL for treatment of uncomplicated P. falciparum malaria among children in Africa. The search was performed from August 2020 to 30 April 2021. Using Rev-Man software (V5.4.1), the extracted data from eligible studies were pooled as risk ratio (RR) with 95% confidence interval (CI). RESULTS In this review, 18 studies were included, which involved 10,498 participants were included. Compared to AL, DHA-PQ was associated with a slightly higher frequency of early vomiting (RR 2.26, 95% CI 1.46 to 3.50; participants = 7796; studies = 10; I2 = 0%, high quality of evidence), cough (RR 1.06, 95% CI 1.01 to 1.11; participants = 8013; studies = 13; I2 = 0%, high quality of evidence), and diarrhoea (RR 1.16, 95% CI 1.03 to 1.31; participants = 6841; studies = 11; I2 = 8%, high quality of evidence) were more frequent in DHA-PQ treatment arm. CONCLUSION From this review, it can be concluded that early vomiting, diarrhoea, and cough were common were significantly more frequent in patients who were treated with the DHA-PQ than that of AL, and both drugs are well tolerated. More studies comparing AL with DHA-PQ are needed to determine the comparative safety of these drugs.
Collapse
Affiliation(s)
- Dawit Getachew Assefa
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.
- School of Public Health, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia.
| | - Eden Dagnachew Zeleke
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Midwifery, College of Health Science, Bule Hora University, Bule Hora, Ethiopia
| | - Wondwosen Molla
- Department of Midwifery, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia
| | - Nebiyu Mengistu
- Department of Psychiatry, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia
| | - Ahmedin Sefa
- Department of Nursing, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia
| | - Andualem Mebratu
- Department of Midwifery, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia
| | - Asresu Feleke Bate
- Department of Nursing, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia
| | - Etaferaw Bekele
- Department of Nursing, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia
| | - Gizachew Yesmaw
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Eyasu Makonnen
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
6
|
Assefa DG, Yismaw G, Makonnen E. Efficacy of dihydroartemisinin-piperaquine versus artemether-lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria among children in Africa: a systematic review and meta-analysis of randomized control trials. Malar J 2021; 20:340. [PMID: 34384431 PMCID: PMC8359548 DOI: 10.1186/s12936-021-03873-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Emergence of Plasmodium falciparum resistance to artemisinin and its derivatives poses a threat to the global effort to control malaria. The emergence of anti-malarial resistance has become a great public health challenge and continues to be a leading threat to ongoing malaria control efforts. The aim of this review was to synthesize available evidence on the efficacy of dihydroartemisinin-piperaquine (DHA-PQ) compared to artemether-lumefantrine (AL) for the treatment of uncomplicated falciparum malaria among children in Africa. METHODS A systematic literature search was done to identify relevant articles from online databases PubMed/ MEDLINE, Embase, and Cochrane Central Register of Controlled Trials' database (CENTRAL) for retrieving randomized control trials comparing efficacy of DHA-PQ and AL for treatment of uncomplicated falciparum malaria in African children. The search was performed from August 2020 to April 2021. Using Rev-Man software (V5.4.1), R-studio and Comprehensive Meta-analysis software version 3, the extracted data from eligible studies were pooled as risk ratio (RR) with 95% confidence interval (CI). RESULTS In this review, 25 studies which involved a total of 13,198 participants were included. PCR-unadjusted treatment failure in children aged between 6 months and 15 years was significantly lower in the DHA-PQ treatment arm on day 28 than that of AL (RR 0.14, 95% CI 0.08-0.26; participants = 1302; studies = 4; I2 = 0%, high quality of evidence). Consistently, the PCR-adjusted treatment failure was significantly lower with DHA-PQ treatment group on day 28 (RR 0.45, 95% CI 0.29-0.68; participants = 8508; studies = 16; I2 = 51%, high quality of evidence) and on day 42 (RR 0.60, 95% CI 0.47-0.78; participants = 5959; studies = 17; I2 = 0%, high quality of evidence). However, the efficacy was ≥ 95% in both treatment groups on day 28. CONCLUSION From this review, it can be concluded that DHA-PQ reduces new infection and recrudescence on days 28 and 42 more than AL. This may trigger DHA-PQ to become a first-line treatment option.
Collapse
Affiliation(s)
- Dawit Getachew Assefa
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia. .,Department of Nursing, College of Health Science and Medicine, Dilla University, Dilla, Ethiopia.
| | - Gizachew Yismaw
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Eyasu Makonnen
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.,Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
7
|
Kaur R, Gorki V, Katare OP, Dhingra N, Chauhan M, Kaur R, Nirmalan N, Singh B. Improved biopharmaceutical attributes of lumefantrine using choline mimicking drug delivery system: preclinical investigation on NK-65 P.berghei murine model. Expert Opin Drug Deliv 2021; 18:1533-1552. [PMID: 34176411 DOI: 10.1080/17425247.2021.1946512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Lumefantrine (LMF) is first-line antimalarial drug, possesses activity against almost all human malarial parasites, but the in vivo activity of this molecule gets thwarted due to its low and inconsistent oral bioavailability (i.e. 4-12%) owing to poor biopharmaceutical attributes. METHODS Lumefantrine phospholipid complex (LMF-PC) was prepared by rota-evaporation method following job's plot technique for the selection of apt stoichiometric ratios. Docking studies were carried out to determine the possible interaction(s) of LMF with phosphatidylcholine analogue. Comparative in vitro physiochemical, solid-state characterization, MTT assay, dose-response on P. falciparum, in vivo efficacy studies including pharmacokinetic and chemosuppression on NK-65 P. berghei infected mice were carried out. RESULTS Aqueous solubility was distinctly improved (i.e. 345 times) with phospholipid complex of LMF. Cytotoxicity studies on Hela and fibroblast cell lines demonstrated safety of LMF-PC with selectivity indices of 4395 and 5139, respectively. IC50 value was reduced almost 2.5 folds. Significant enhancement in Cmax (3.3-folds) and AUC (2.7-folds) of rat plasma levels indicated notable pharmacokinetic superiority of LMF-PC over LMF suspension. Differential leukocytic count and cytokine assay delineated plausible immunoregulatory role of LMF-PC with nearly 98% chemosuppression and over 30 days of post-survival. CONCLUSION Superior antimalarial efficacy and survival time with full recovery of infected mice revealed through histopathological studies.
Collapse
Affiliation(s)
- Ripandeep Kaur
- UGC Centre of Advanced Studies, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.,School of Science, Engineering & Environment, University of Salford, Manchester, UK.,UGC-Centre of Excellence in Nano Applications (Biomedical Sciences), Panjab University, Chandigarh, India
| | - Varun Gorki
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India
| | - O P Katare
- UGC Centre of Advanced Studies, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Neelima Dhingra
- UGC Centre of Advanced Studies, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Monika Chauhan
- UGC Centre of Advanced Studies, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranjot Kaur
- UGC Centre of Advanced Studies, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Niroshini Nirmalan
- School of Science, Engineering & Environment, University of Salford, Manchester, UK
| | - Bhupinder Singh
- UGC Centre of Advanced Studies, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.,UGC-Centre of Excellence in Nano Applications (Biomedical Sciences), Panjab University, Chandigarh, India
| |
Collapse
|
8
|
Ndwiga L, Osoti V, Ochwedo KO, Wamae K, Bejon P, Rayner JC, Githinji G, Ochola-Oyier LI. The Plasmodium falciparum Rh5 invasion protein complex reveals an excess of rare variant mutations. Malar J 2021; 20:278. [PMID: 34162366 PMCID: PMC8220363 DOI: 10.1186/s12936-021-03815-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 06/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The invasion of the red blood cells by Plasmodium falciparum merozoites involves the interplay of several proteins that are also targets for vaccine development. The proteins PfRh5-PfRipr-PfCyRPA-Pfp113 assemble into a complex at the apical end of the merozoite and are together essential for erythrocyte invasion. They have also been shown to induce neutralizing antibodies and appear to be less polymorphic than other invasion-associated proteins, making them high priority blood-stage vaccine candidates. Using available whole genome sequencing data (WGS) and new capillary sequencing data (CS), this study describes the genetic polymorphism in the Rh5 complex in P. falciparum isolates obtained from Kilifi, Kenya. METHODS 162 samples collected in 2013 and 2014 were genotyped by capillary sequencing (CS) and re-analysed WGS from 68 culture-adapted P. falciparum samples obtained from a drug trial conducted from 2005 to 2007. The frequency of polymorphisms in the merozoite invasion proteins, PfRh5, PfRipr, PfCyRPA and PfP113 were examined and where possible polymorphisms co-occurring in the same isolates. RESULTS From a total 70 variants, including 2 indels, 19 SNPs [27.1%] were identified by both CS and WGS, while an additional 15 [21.4%] and 36 [51.4%] SNPs were identified only by either CS or WGS, respectively. All the SNPs identified by CS were non-synonymous, whereas WGS identified 8 synonymous and 47 non-synonymous SNPs. CS identified indels in repeat regions in the p113 gene in codons 275 and 859 that were not identified in the WGS data. The minor allele frequencies of the SNPs ranged between 0.7 and 34.9% for WGS and 1.1-29.6% for CS. Collectively, 12 high frequency SNPs (> 5%) were identified: four in Rh5 codon 147, 148, 203 and 429, two in p113 at codons 7 and 267 and six in Ripr codons 190, 259, 524, 985, 1003 and 1039. CONCLUSION This study reveals that the majority of the polymorphisms are rare variants and confirms a low level of genetic polymorphisms in all proteins within the Rh5 complex.
Collapse
Affiliation(s)
- Leonard Ndwiga
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Victor Osoti
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Kevin Omondi Ochwedo
- Centre for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya
| | - Kevin Wamae
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
- Nuffield Department of Medicine, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | - Julian C Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - George Githinji
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | | |
Collapse
|
9
|
Kayode AT, Akano K, Ajogbasile FV, Uwanibe JN, Oluniyi PE, Bankole BE, Eromon PJ, Sowunmi A, Folarin OA, Volkman SK, McInnis B, Sabeti P, Wirth DF, Happi CT. Polymorphisms in Plasmodium falciparum chloroquine resistance transporter (Pfcrt) and multidrug-resistant gene 1 (Pfmdr-1) in Nigerian children 10 years post-adoption of artemisinin-based combination treatments. Int J Parasitol 2021; 51:301-310. [PMID: 33359205 PMCID: PMC7940560 DOI: 10.1016/j.ijpara.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/08/2020] [Accepted: 10/03/2020] [Indexed: 12/13/2022]
Abstract
The emergence and spread of Plasmodium falciparum parasites resistant to artemisinin derivatives and their partners in southeastern Asia threatens malaria control and elimination efforts, and heightens the need for an alternative therapy. We have explored the distribution of P. falciparum chloroquine resistance transporter (Pfcrt) and multidrug-resistant gene 1 (Pfmdr-1) haplotypes 10 years following adoption of artemisinin-based combination therapies in a bid to investigate the possible re-emergence of Chloroquine-sensitive parasites in Nigeria, and investigated the effect of these P. falciparum haplotypes on treatment outcomes of patients treated with artemisinin-based combination therapies. A total of 271 children aged <5 years with uncomplicated falciparum malaria were included in this study. Polymorphisms on codons 72-76 of the Pfcrt gene and codon 86 and 184 of Pfmdr-1 were determined using the high resolution melting assay. Of 240 (88.6%) samples successfully genotyped with HRM for Pfcrt, wildtype C72M74N75K76 (42.9%) and mutant C72I74E75T76 (53.8%) were observed. Also, wildtype N86Y184 (62.9%) and mutant N86F184 (21.1%), Y86Y184 (6.4%), and Y86F184 (0.4%) haplotypes of Pfmdr-1 were observed. Measures of responsiveness to ACTs were similar in children infected with P. falciparum crt haplotypes (C72I74E75T76 and C72M74N75K76) and major mdr-1 haplotypes (N86Y184, N86F184 and Y86Y184). Despite a 10 year gap since the malaria treatment policy changed to ACTs, over 50% of the P. falciparum parasites investigated in this study harboured the Chloroquine-resistant C72I74E75T76 haplotype, however this did not compromise the efficacy of artemisinin-based combination therapies. Should complete artemisinin resistance emerge from or spread to Nigeria, chloroquine might not be a good alternative therapy.
Collapse
Affiliation(s)
- Adeyemi T Kayode
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Kazeem Akano
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Fehintola V Ajogbasile
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Jessica N Uwanibe
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Paul E Oluniyi
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Bolajoko E Bankole
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Philomena J Eromon
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria
| | - Akintunde Sowunmi
- Institute of Medical Research and Training, College of Medicine, University of Ibadan; Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Onikepe A Folarin
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Sarah K Volkman
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Pardis Sabeti
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Christian T Happi
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
10
|
Mahamar A, Lanke K, Graumans W, Diawara H, Sanogo K, Diarra K, Niambele SM, Gosling R, Drakeley C, Chen I, Dicko A, Bousema T, Roh ME. Persistence of mRNA indicative of Plasmodium falciparum ring-stage parasites 42 days after artemisinin and non-artemisinin combination therapy in naturally infected Malians. Malar J 2021; 20:34. [PMID: 33422068 PMCID: PMC7797096 DOI: 10.1186/s12936-020-03576-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/31/2020] [Indexed: 11/10/2022] Open
Abstract
Background Malaria control in sub-Saharan Africa relies upon prompt case management with artemisinin-based combination therapy (ACT). Ring-stage parasite mRNA, measured by sbp1 quantitative reverse-transcriptase PCR (qRT-PCR), was previously reported to persist after ACT treatment and hypothesized to reflect temporary arrest of the growth of ring-stage parasites (dormancy) following exposure to artemisinins. Here, the persistence of ring-stage parasitaemia following ACT and non-ACT treatment was examined. Methods Samples were used from naturally infected Malian gametocyte carriers who received dihydroartemisinin–piperaquine (DP) or sulfadoxine–pyrimethamine (SP–AQ) with or without gametocytocidal drugs. Gametocytes and ring-stage parasites were quantified by qRT-PCR during 42 days of follow-up. Results At baseline, 89% (64/73) of participants had measurable ring-stage parasite mRNA. Following treatment, the proportion of ring-stage parasite-positive individuals and estimated densities declined for all four treatment groups. Ring-stage parasite prevalence and density was generally lower in arms that received DP compared to SP–AQ. This finding was most apparent days 1, 2, and 42 of follow-up (p < 0.01). Gametocytocidal drugs did not influence ring-stage parasite persistence. Ring-stage parasite density estimates on days 14 and 28 after initiation of treatment were higher among individuals who subsequently experienced recurrent parasitaemia compared to those who remained free of parasites until day 42 after initiation of treatment (pday 14 = 0.011 and pday 28 = 0.068). No association of ring-stage persistence with gametocyte carriage was observed. Conclusions The current findings of lower ring-stage persistence after ACT without an effect of gametocytocidal partner drugs affirms the use of sbp1 as ring-stage marker. Lower persistence of ring-stage mRNA after ACT treatment suggests the marker may not reflect dormant parasites whilst it was predictive of re-appearance of parasitaemia.
Collapse
Affiliation(s)
- Almahamoudou Mahamar
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, PO Box 9101, 6525GA, Nijmegen, The Netherlands
| | - Wouter Graumans
- Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, PO Box 9101, 6525GA, Nijmegen, The Netherlands
| | - Halimatou Diawara
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Koualy Sanogo
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Kalifa Diarra
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Sidi Mohamed Niambele
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Roly Gosling
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, CA, USA
| | - Chris Drakeley
- Department of Infection & Immunity, London School of Hygiene & Tropical Medicine, London, UK
| | - Ingrid Chen
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, CA, USA
| | - Alassane Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, PO Box 9101, 6525GA, Nijmegen, The Netherlands. .,Department of Infection & Immunity, London School of Hygiene & Tropical Medicine, London, UK.
| | - Michelle E Roh
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, CA, USA
| |
Collapse
|
11
|
Tola M, Ajibola O, Idowu ET, Omidiji O, Awolola ST, Amambua-Ngwa A. Molecular detection of drug resistant polymorphisms in Plasmodium falciparum isolates from Southwest, Nigeria. BMC Res Notes 2020; 13:497. [PMID: 33109270 PMCID: PMC7588951 DOI: 10.1186/s13104-020-05334-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/14/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Nigeria bears 25% of global malaria burden despite concerted efforts towards its control and elimination. The emergence of drug resistance to first line drugs, artemisinin combination therapies (ACTs), indicates an urgent need for continuous molecular surveillance of drug resistance especially in high burden countries where drug interventions are heavily relied on. This study describes mutations in Plasmodium falciparum genes associated with drug resistance in malaria; Pfk13, Pfmdr1, PfATPase6 and Pfcrt in isolates obtained from 83 symptomatic malaria patients collected in August 2014, aged 1-61 years old from South-west Nigeria. RESULTS Two Pfmdr1, N86 and Y184 variants were present at a prevalence of 56% and 13.25% of isolates respectively. There was one synonymous (S679S) and two non-synonymous (M699V, S769M) mutations in the PATPase6 gene, while Pfcrt genotype (CVIET), had a prevalence of 45%. The Pfk13 C580Y mutant allele was suspected by allelic discrimination in two samples with mixed genotypes although this could not be validated with independent isolation or additional methods. Our findings call for robust molecular surveillance of antimalarial drug resistance markers in west Africa especially with increased use of antimalarial drugs as prophylaxis for Covid-19.
Collapse
Affiliation(s)
- Monday Tola
- Public Health Division, Nigerian Institute of Medical Research, Lagos, Nigeria.,Department of Cell Biology and Genetics, University of Lagos, Lagos, Nigeria
| | - Olumide Ajibola
- First Technical University, Ibadan, Oyo State, Nigeria.,Medical Research Council Unit The Gambia At London, School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | | | - Olusesan Omidiji
- Department of Cell Biology and Genetics, University of Lagos, Lagos, Nigeria
| | | | - Alfred Amambua-Ngwa
- Medical Research Council Unit The Gambia At London, School of Hygiene and Tropical Medicine, Banjul, The Gambia.
| |
Collapse
|
12
|
Different Plasmodium falciparum clearance times in two Malian villages following artesunate monotherapy. Int J Infect Dis 2020; 95:399-405. [PMID: 32320811 PMCID: PMC7294218 DOI: 10.1016/j.ijid.2020.03.082] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 11/23/2022] Open
Abstract
High prevalence of residual parasitemia at day 3 post-artesunate monotherapy treatment using qPCR while no parasites were detected by microscopy at the same timepoint. A longer parasite clearance time observed in a Malian village. Artesunate treatment is still efficacious on Plasmodium falciparum in Mali.
Background Artemisinin resistance described as increased parasite clearance time (PCT) is rare in Africa. More sensitive methods such as qPCR might better characterize the clearance phenotype in sub-Saharan Africa. Methods PCT is explored in Mali using light microscopy and qPCR after artesunate for uncomplicated malaria. In two villages, patients were followed for 28 days. Blood smears and spots were collected respectively for microscopy and qPCR. Parasitemia slope half-life was calculated after microscopy. Patient residual parasitemia were measured by qPCR. Results Uncorrected adequate clinical and parasitological responses (ACPR) observed in Faladje and Bougoula-Hameau were 78% and 92%, respectively (p = 0.01). This reached 100% for both after molecular correction. Proportions of 24H microscopy positive patients in Faladje and Bougoula-Hameau were 97.2% and 72%, respectively (p < 0.0001). Slope half-life was 2.8 h in Faladje vs 2H in Bougoula-Hameau (p < 0.001) and Proportions of 72H patients with residual parasitemia were 68.5% and 40% in Faladje and Bougoula-Hameau, respectively (p = 0.003). The mean residual parasitemia was 2.9 in Faladje vs. 0.008 in Bougoula-Hameau (p = 0.002). Although artesunate is efficacious in Mali, the longer parasite clearance time with submicroscopic parasitemia observed may represent early signs of developing P. falciparum resistance to artemisinins.
Collapse
|
13
|
Lingani M, Bonkian LN, Yerbanga I, Kazienga A, Valéa I, Sorgho H, Ouédraogo JB, Mens PF, Schallig HDFH, Ravinetto R, d'Alessandro U, Tinto H. In vivo/ex vivo efficacy of artemether-lumefantrine and artesunate-amodiaquine as first-line treatment for uncomplicated falciparum malaria in children: an open label randomized controlled trial in Burkina Faso. Malar J 2020; 19:8. [PMID: 31906948 PMCID: PMC6945612 DOI: 10.1186/s12936-019-3089-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/25/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Artemisinin-based combination therapy (ACT) is recommended to improve malaria treatment efficacy and limit drug-resistant parasites selection in malaria endemic areas. 5 years after they were adopted, the efficacy and safety of artemether-lumefantrine (AL) and artesunate-amodiaquine (ASAQ), the first-line treatments for uncomplicated malaria were assessed in Burkina Faso. METHODS In total, 440 children with uncomplicated Plasmodium falciparum malaria were randomized to receive either AL or ASAQ for 3 days and were followed up weekly for 42 days. Blood samples were collected to investigate the ex vivo susceptibility of P. falciparum isolates to lumefantrine, dihydroartemisinin (the active metabolite of artemisinin derivatives) and monodesethylamodiaquine (the active metabolite of amodiaquine). The modified isotopic micro test technique was used to determine the 50% inhibitory concentration (IC50) values. Primary endpoints were the risks of treatment failure at days 42. RESULTS Out of the 440 patients enrolled, 420 (95.5%) completed the 42 days follow up. The results showed a significantly higher PCR unadjusted cure rate in ASAQ arm (71.0%) than that in the AL arm (49.8%) on day 42, and this trend was similar after correction by PCR, with ASAQ performing better (98.1%) than AL (91.1%). Overall adverse events incidence was low and not significantly different between the two treatment arms. Ex vivo results showed that 6.4% P. falciparum isolates were resistant to monodesthylamodiaquine. The coupled in vivo/ex vivo analysis showed increased IC50 values for lumefantrine and monodesethylamodiaquine at day of recurrent parasitaemia compared to baseline values while for artesunate, IC50 values remained stable at baseline and after treatment failure (p > 0.05). CONCLUSION These findings provide substantial evidence that AL and ASAQ are highly efficacious for the treatment of uncomplicated malaria in children in Burkina Faso. However, the result of P. falciparum susceptibility to the partner drugs advocates the need to regularly replicate such surveillance studies. This would be particularly indicated when amodiaquine is associated in seasonal malaria chemoprophylaxis (SMC) mass drug administration in children under 5 years in Burkina Faso. Trial registration clinicaltrials, NCT00808951. Registered 05 December 2008,https://clinicaltrials.gov/ct2/show/NCT00808951?cond=NCT00808951&rank=1.
Collapse
Affiliation(s)
- Moussa Lingani
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso. .,Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso. .,École de Santé Publique, Université Libre de Bruxelles, CP594, Route de Lennik 808, 1070, Bruxelles, Belgique.
| | - Léa Nadège Bonkian
- Unité de Recherche sur le Paludisme et Maladies Tropicales Négligées, Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Isidore Yerbanga
- Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso
| | - Adama Kazienga
- Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso
| | - Innocent Valéa
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso.,Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso
| | - Hermann Sorgho
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso.,Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso
| | - Jean Bosco Ouédraogo
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| | - Petronella Francisca Mens
- Department of Medical Microbiology, Experimental Parasitology Unit, Amsterdam University Medical Centres, Academic Medical Centre at the University of Amsterdam, Amsterdam, The Netherlands
| | - Henk D F H Schallig
- Department of Medical Microbiology, Experimental Parasitology Unit, Amsterdam University Medical Centres, Academic Medical Centre at the University of Amsterdam, Amsterdam, The Netherlands
| | | | - Umberto d'Alessandro
- Medical Research Council Unit, The Gambia, Disease Control & Elimination Theme, Fajara, The Gambia
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso.,Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso
| |
Collapse
|
14
|
Omondi P, Burugu M, Matoke-Muhia D, Too E, Nambati EA, Chege W, Musyoka KB, Thiongo K, Otinga M, Muregi F, Kimani F. Gametocyte clearance in children, from western Kenya, with uncomplicated Plasmodium falciparum malaria after artemether-lumefantrine or dihydroartemisinin-piperaquine treatment. Malar J 2019; 18:398. [PMID: 31801562 PMCID: PMC6891957 DOI: 10.1186/s12936-019-3032-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/24/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The efficacy and safety of artemether-lumefantrine (AL) and dihydroartemisinin-piperaquine (DP) against asexual parasites population has been documented. However, the effect of these anti-malarials on sexual parasites is still less clear. Gametocyte clearance following treatment is essential for malaria control and elimination efforts; therefore, the study sought to determine trends in gametocyte clearance after AL or DP treatment in children from a malaria-endemic site in Kenya. METHODS Children aged between 0.5 and 12 years from Busia, western Kenya with uncomplicated Plasmodium falciparum malaria were assigned randomly to AL or DP treatment. A total of 334 children were enrolled, and dried blood spot samples were collected for up to 6 weeks after treatment during the peak malaria transmission season in 2016 and preserved. Plasmodium falciparum gametocytes were detected by qRT-PCR and gametocyte prevalence, density and mean duration of gametocyte carriage were determined. RESULTS At baseline, all the 334 children had positive asexual parasites by microscopy, 12% (40/334) had detectable gametocyte by microscopy, and 83.7% (253/302) children had gametocytes by RT-qPCR. Gametocyte prevalence by RT-qPCR decreased from 85.1% (126/148) at day 0 to 7.04% (5/71) at day 42 in AL group and from 82.4% (127/154) at day 0 to 14.5% (11/74) at day 42 in DP group. The average duration of gametocyte carriage as estimated by qRT-PCR was slightly shorter in the AL group (4.5 days) than in the DP group (5.1 days) but not significantly different (p = 0.301). CONCLUSION The study identifies no significant difference between AL and DP in gametocyte clearance. Gametocytes persisted up to 42 days post treatment in minority of individuals in both treatment arms. A gametocytocidal drug, in combination with artemisinin-based combination therapy, will be useful in blocking malaria transmission more efficiently.
Collapse
Affiliation(s)
- Protus Omondi
- Department of Biochemistry, Microbiology, and Biotechnology, Kenyatta University, P.O Box 43884-00100, Nairobi, Kenya.,Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - Marion Burugu
- Department of Biochemistry, Microbiology, and Biotechnology, Kenyatta University, P.O Box 43884-00100, Nairobi, Kenya
| | - Damaris Matoke-Muhia
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - Edwin Too
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - Eva A Nambati
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - William Chege
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - Kelvin B Musyoka
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O Box 62000-00200, Nairobi, Kenya
| | - Kelvin Thiongo
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - Maureen Otinga
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya
| | - Francis Muregi
- Department of Biological Sciences, Mount Kenya University, P.O Box 342-00100, Nairobi, Kenya
| | - Francis Kimani
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O Box 54840-00200, Nairobi, Kenya.
| |
Collapse
|
15
|
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]
|
16
|
Roth JM, Sawa P, Omweri G, Makio N, Osoti V, de Jong MD, Schallig HDFH, Mens PF. Molecular Detection of Residual Parasitemia after Pyronaridine-Artesunate or Artemether-Lumefantrine Treatment of Uncomplicated Plasmodium falciparum Malaria in Kenyan Children. Am J Trop Med Hyg 2019; 99:970-977. [PMID: 30105967 DOI: 10.4269/ajtmh.18-0233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Artemisinin resistance is rapidly rising in Southeast Asia and may spread to African countries, where efficacy estimates are currently still excellent. Extensive monitoring of parasite clearance dynamics after treatment is needed to determine whether responsiveness to artemisinin-based combination therapies (ACT) is changing in Africa. In this study, Kenyan children with uncomplicated falciparum malaria were randomly assigned to pyronaridine-artesunate (PA) or artemether-lumefantrine (AL) treatment. Parasite clearance was evaluated over 7 days following the start of treatment by quantitative polymerase chain reaction (qPCR) and direct-on-blood PCR nucleic acid lateral flow immunoassay (db-PCR-NALFIA), a simplified molecular malaria diagnostic. Residual parasitemia at day 7 was detected by qPCR in 37.1% (26/70) of AL-treated children and in 46.1% (35/76) of PA-treated participants (P = 0.275). Direct-on-blood PCR nucleic acid lateral flow immunoassay detected residual parasites at day 7 in 33.3% (23/69) and 30.3% (23/76) of AL and PA-treated participants, respectively (P = 0.692). qPCR-determined parasitemia at day 7 was associated with increased prevalence and density of gametocytes at baseline (P = 0.014 and P = 0.003, for prevalence and density, respectively) and during follow-up (P = 0.007 and P = 0.011, respectively, at day 7). A positive db-PCR-NALFIA outcome at day 7 was associated with treatment failure (odds ratio [OR]: 3.410, 95% confidence interval [CI]: 1.513-7.689, P = 0.003), but this association was not found for qPCR (OR: 0.701, 95% CI: 0.312-1.578, P = 0.391). Both qPCR and db-PCR-NALFIA detected substantial residual submicroscopic parasitemia after microscopically successful PA and AL treatment and can be useful tools to monitor parasite clearance. To predict treatment outcome, db-PCR-NALFIA may be more suitable than qPCR.
Collapse
Affiliation(s)
- Johanna M Roth
- Department of Medical Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Patrick Sawa
- Human Health Division, International Centre of Insect Physiology and Ecology, Mbita Point, Kenya
| | - George Omweri
- Human Health Division, International Centre of Insect Physiology and Ecology, Mbita Point, Kenya
| | - Nicodemus Makio
- Human Health Division, International Centre of Insect Physiology and Ecology, Mbita Point, Kenya
| | - Victor Osoti
- Human Health Division, International Centre of Insect Physiology and Ecology, Mbita Point, Kenya
| | - Menno D de Jong
- Department of Medical Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Henk D F H Schallig
- Department of Medical Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Pètra F Mens
- Department of Medical Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Sowunmi A, Ntadom G, Akano K, Ibironke FO, Ayede AI, Agomo C, Folarin OA, Gbotosho GO, Happi C, Oguche S, Okafor HU, Meremikwu M, Agomo P, Ogala W, Watila I, Mokuolu O, Finomo F, Ebenebe JC, Jiya N, Ambe J, Wammanda R, Emechebe G, Oyibo W, Useh F, Aderoyeje T, Dokunmu TM, Alebiosu OT, Amoo S, Basorun OK, Wewe OA, Okafor C, Akpoborie O, Fatunmbi B, Adewoye EO, Ezeigwe NM, Oduola A. Declining responsiveness of childhood Plasmodium falciparum infections to artemisinin-based combination treatments ten years following deployment as first-line antimalarials in Nigeria. Infect Dis Poverty 2019; 8:69. [PMID: 31383040 PMCID: PMC6683392 DOI: 10.1186/s40249-019-0577-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
Background The development and spread of artemisinin-resistant Plasmodium falciparum malaria in Greater Mekong Subregion has created impetus for continuing global monitoring of efficacy of artemisinin-based combination therapies (ACTs). This post analyses is aimed to evaluate changes in early treatment response markers 10 years after the adoption of ACTs as first-line treatments of uncomplicated falciparum malaria in Nigeria. Methods At 14 sentinel sites in six geographical areas of Nigeria, we evaluated treatment responses in 1341 children under 5 years and in additional 360 children under 16 years with uncomplicated malaria enrolled in randomized trials of artemether-lumefantrine versus artesunate-amodiaquine at 5-year interval in 2009–2010 and 2014–2015 and at 2-year interval in 2009–2010 and 2012–2015, respectively after deployment in 2005. Results Asexual parasite positivity 1 day after treatment initiation (APPD1) rose from 54 to 62% and 2 days after treatment initiation from 5 to 26% in 2009–2010 to 2014–2015 (P = 0.002 and P < 0.0001, respectively). Parasite clearance time increased significantly from 1.6 days (95% confidence interval [CI]: 1.55–1.64) to 1.9 days (95% CI, 1.9–2.0) and geometric mean parasite reduction ratio 2 days after treatment initiation decreased significantly from 11 000 to 4700 within the same time period (P < 0.0001 for each). Enrolment parasitaemia > 75 000 μl− 1, haematocrit > 27% 1 day post-treatment initiation, treatment with artemether-lumefantrine and enrolment in 2014–2015 independently predicted APPD1. In parallel, Kaplan-Meier estimated risk of recurrent infections by day 28 rose from 8 to 14% (P = 0.005) and from 9 to 15% (P = 0.02) with artemether-lumefantrine and artesunate-amodiaquine, respectively. Mean asexual parasitaemia half-life increased significantly from 1.1 h to 1.3 h within 2 years (P < 0.0001). Conclusions These data indicate declining parasitological responses through time to the two ACTs may be due to emergence of parasites with reduced susceptibility or decrease in immunity to the infections in these children. Trial registration Pan African Clinical Trial Registration PACTR201508001188143, 3 July 2015; PACTR201508001191898, 7 July 2015 and PACTR201508001193368, 8 July 2015 PACTR201510001189370, 3 July 2015; PACTR201709002064150, 1 March 2017; https://www.pactr.samrca.ac.za Electronic supplementary material The online version of this article (10.1186/s40249-019-0577-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akintunde Sowunmi
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria. .,Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria. .,Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria. .,Department of Clinical Pharmacology, University College Hospital, Ibadan, Ibadan, Nigeria.
| | - Godwin Ntadom
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Kazeem Akano
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria.,Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Folasade O Ibironke
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Ibadan, Nigeria
| | | | - Chimere Agomo
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Medical Laboratory Science, University of Lagos, Lagos, Nigeria
| | - Onikepe A Folarin
- Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Grace O Gbotosho
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria.,Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria.,Department of Pharmacology and Toxicology, University of Ibadan, Ibadan, Nigeria
| | - Christian Happi
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Stephen Oguche
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, University of Jos, Jos, Nigeria
| | - Henrietta U Okafor
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Pediatrics, Institute of Child Health, University of Nigeria Teaching Hospital, Enugu, Nigeria
| | - Martin Meremikwu
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, University of Calabar, Calabar, Cross River State, Nigeria
| | - Philip Agomo
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Nigeria Institute of Medical Research, Lagos, Nigeria
| | - William Ogala
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Ahmadu Bello University, Zaria, Nigeria
| | - Ismaila Watila
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Specialist Hospital, Maiduguri, Nigeria
| | - Olugbenga Mokuolu
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics and Child Health, University of Ilorin, Ilorin, Nigeria
| | - Finomo Finomo
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Federal Medical Centre, Yenagoa, Nigeria
| | - Joy C Ebenebe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Nnamdi Azikiwe University, Awka, Nigeria
| | - Nma Jiya
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Uthman Dan Fodio University, Sokoto, Nigeria
| | - Jose Ambe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, University of Maiduguri, Maiduguri, Nigeria
| | - Robinson Wammanda
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Ahmadu Bello University, Zaria, Nigeria
| | - George Emechebe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Paediatrics, Imo State University Teaching Hospital, Orlu, Nigeria
| | - Wellington Oyibo
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Medical Microbiology and Parasitology, University of Lagos, Lagos, Nigeria
| | - Francis Useh
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,Department of Medical Laboratory Science, University of Calabar, Calabar, Nigeria
| | - Temitope Aderoyeje
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Ibadan, Nigeria
| | | | - Omobolaji T Alebiosu
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Sikiru Amoo
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Oluwabunmi K Basorun
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Olubunmi A Wewe
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Chukwuebuka Okafor
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Odafe Akpoborie
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Bayo Fatunmbi
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria.,World Health Organization, Country Office, Kampala, Uganda
| | - Elsie O Adewoye
- Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - Nnenna M Ezeigwe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
| | - Ayoade Oduola
- University of Ibadan Research Foundation, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
18
|
Talisuna AO, Zurovac D, Githinji S, Oburu A, Malinga J, Nyandigisi A, Jones CO, Snow RW. Efficacy of Mobile Phone Short Message Service (SMS) Reminders on Malaria Treatment Adherence and Day 3 Post-Treatment Reviews (SMS-RES-MAL) in Kenya: A Study Protocol. ACTA ACUST UNITED AC 2019; 5:217. [PMID: 31285980 PMCID: PMC6614027 DOI: 10.4172/2167-0870.1000217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background Mobile phone short messaging services (SMS) have been investigated in health information reporting, provider performance, drug and diagnostic stock management and patient adherence to treatment for chronic diseases. However, their potential role in improving patients’ adherence to malaria treatment and day 3 post treatment reviews remains unclear. Methods/Design A “proof of concept” open label randomised controlled trial will be conducted at four sites in Western Kenya. Principal research questions are: 1) Can mobile phone SMS reminders improve patient adherence to malaria treatment? 2) Can mobile phone SMS reminders improve day 3 post treatment reviews? Eligible caregivers (n=1000 per arm) of children under five years old with uncomplicated malaria will be randomly assigned (one to one) to: a) the current standard of care (provider counselling and health education); and b) the current standard of care plus SMS reminders. Within each arm, caregivers will be further randomized to three different categories. In categories 1 and 2, 300 caregivers per arm per category will be visited at home on day 1 and 2 of follow up respectively, to measure appropriate timing and adherence of the second Artemether-Lumefantrine (AL) dose and doses 3 and 4. Further, caregivers in categories 1 and 2 will be required to come to the health facility for the day 3 post treatment reviews. Finally, in category 3, 400 caregivers per arm will be visited at home on day 3 to measure adherence for the full AL course. Each category will be visited at home only once to avoid biases in the measures of adherence as a result of home consultations. Primary outcomes will be adherence to the full AL course (category 3), as well as, the proportion of patients reporting back for day 3 post treatment reviews (categories 1 and 2). The primary analysis will be intention-to-treat. Costs of the intervention will be measured over the period of the intervention, and a cost-effectiveness ratio will be estimated. Discussion If successful, evidence from this trial could improve malaria treatment adherence and offer pragmatic approaches for antimalarial drug resistance surveillance and risk mitigation in Africa. Current Controlled Trials ISRCTN39512726
Collapse
Affiliation(s)
- Ambrose O Talisuna
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya.,Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, UK
| | - Dejan Zurovac
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya.,Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, UK
| | - Sophie Githinji
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya
| | - Amos Oburu
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya
| | - Josephine Malinga
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya
| | | | - Caroline Oh Jones
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya.,Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, UK
| | - Robert W Snow
- Department of Public Health Research, KEMRI-Welcome Trust Research Program, Kenya.,Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, UK
| |
Collapse
|
19
|
Akano K, Ntadom G, Agomo C, Happi CT, Folarin OA, Gbotosho GO, Mokuolu O, Finomo F, Ebenebe JC, Jiya N, Ambe J, Wammanda R, Emechebe G, Basorun OK, Wewe OA, Amoo S, Ezeigwe N, Oguche S, Fatunmbi B, Sowunmi A. Parasite reduction ratio one day after initiation of artemisinin-based combination therapies and its relationship with parasite clearance time in acutely malarious children. Infect Dis Poverty 2018; 7:122. [PMID: 30522524 PMCID: PMC6284283 DOI: 10.1186/s40249-018-0503-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In acute falciparum malaria, asexual parasite reduction ratio two days post-treatment initiation (PRRD2) ≥ 10 000 per cycle has been used as a measure of the rapid clearance of parasitaemia and efficacy of artemisinin derivatives. However, there is little evaluation of alternative measures; for example, parasite reduction ratio one day after treatment initiation (PRRD1) and its relationship with parasite clearance time (PCT) or PRRD2. This study evaluated the use of PRRD1 as a measure of responsiveness to antimalarial drugs. METHODS In acutely malarious children treated with artesunate-amodiaquine (AA), artemether-lumefantrine (AL) or dihydroartemisinin-piperaquine (DHP), the relationships between PRRD1 or PRRD2 and PCT, and between PRRD1 and PRRD2 were evaluated using linear regression. Agreement between estimates of PCT using PRRD1 and PRRD2 linear regression equations was evaluated using the Bland-Altman analysis. Predictors of PRRD1 > 5000 per half cycle and PRRD2 ≥ 10 000 per cycle were evaluated using stepwise multiple logistic regression models. Using the linear regression equation of the relationship between PRRD1 and PCT previously generated in half of the DHP-treated children during the early study phase, PCT estimates were compared in a prospective blinded manner with PCTs determined by microscopy during the later study phase in the remaining half. RESULTS In 919 malarious children, PRRD1 was significantly higher in DHP- and AA-treated compared with AL-treated children (P < 0.0001). PRRD1 or PRRD2 values correlated significantly negatively with PCT values (P < 0.0001 for each) and significantly positively with each other (P < 0.0001). PCT estimates from linear regression equations for PRRD1 and PRRD2 showed insignificant bias on the Bland-Altman plot (P = 0.7) indicating the estimates can be used interchangeably. At presentation, age > 15 months, parasitaemia > 10 000/μl and DHP treatment independently predicted PRRD1 > 5000 per half cycle, while age > 30 months, haematocrit ≥31%, body temperature > 37.4 °C, parasitaemia > 100 000/μl, PRRD1 value > 1000 and no gametocytaemia independently predicted PRRD2 ≥ 10 000 per cycle. Using the linear regression equation generated during the early phase in 166 DHP-treated children, PCT estimates and PCTs determined by microscopy in the 155 children in the later phase were similar in the same patients. CONCLUSIONS PRRD1 and estimates of PCT using PRRD1 linear regression equation of PRRD1 and PCT can be used in therapeutic efficacy studies. TRIAL REGISTRATION Pan African Clinical Trial Registration PACTR201709002064150, 1 March 2017, http://www.pactr.org.
Collapse
Affiliation(s)
- Kazeem Akano
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
| | - Godwin Ntadom
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
| | - Chimere Agomo
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Medical Laboratory Science, University of Lagos, Lagos, Nigeria
| | - Christian T. Happi
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Onikepe A. Folarin
- Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Grace O. Gbotosho
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria
| | - Olugbenga Mokuolu
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, University of Ilorin, Ilorin, Nigeria
| | - Finomo Finomo
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, Federal Medical Centre, Yenagoa, Nigeria
| | - Joy C. Ebenebe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, Nnamdi Azikiwe University, Awka, Nigeria
| | - Nma Jiya
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, Uthman Dan Fodio University, Sokoto, Nigeria
| | - Jose Ambe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, University of Maiduguri, Maiduguri, Nigeria
| | - Robinson Wammanda
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, Ahmadu Bello University, Zaria, Nigeria
| | - George Emechebe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, Imo State University Teaching Hospital, Orlu, Nigeria
| | | | - Olubunmi A. Wewe
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Sikiru Amoo
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Nnenna Ezeigwe
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
| | - Stephen Oguche
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, University of Jos, Jos, Nigeria
| | - Bayo Fatunmbi
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- World Health Organization, Country Office, Kampala, Uganda
| | - Akintunde Sowunmi
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
20
|
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
|
21
|
Okell LC, Reiter LM, Ebbe LS, Baraka V, Bisanzio D, Watson OJ, Bennett A, Verity R, Gething P, Roper C, Alifrangis M. Emerging implications of policies on malaria treatment: genetic changes in the Pfmdr-1 gene affecting susceptibility to artemether-lumefantrine and artesunate-amodiaquine in Africa. BMJ Glob Health 2018; 3:e000999. [PMID: 30397515 PMCID: PMC6202998 DOI: 10.1136/bmjgh-2018-000999] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/21/2018] [Accepted: 08/31/2018] [Indexed: 11/04/2022] Open
Abstract
Artemether–lumefantrine (AL) and artesunate–amodiaquine (AS-AQ) are the most commonly used artemisinin-based combination therapies (ACT) for treatment of Plasmodium falciparum in Africa. Both treatments remain efficacious, but single nucleotide polymorphisms (SNPs) in the Plasmodium falciparum multidrug resistance 1 (Pfmdr1) gene may compromise sensitivity. AL and AS-AQ exert opposing selective pressures: parasites with genotype 86Y, Y184 and 1246Y are partially resistant to AS-AQ treatment, while N86, 184 F and D1246 are favoured by AL treatment. Through a systematic review, we identified 397 surveys measuring the prevalence of Pfmdr1 polymorphisms at positions 86 184 or 1246 in 30 countries in Africa. Temporal trends in SNP frequencies after introduction of AL or AS-AQ as first-line treatment were analysed in 32 locations, and selection coefficients estimated. We examined associations between antimalarial policies, consumption, transmission intensity and rate of SNP selection. 1246Y frequency decreased on average more rapidly in locations where national policy recommended AL (median selection coefficient(s) of −0.083), compared with policies of AS-AQ or both AL and AS-AQ (median s=−0.035 and 0.021, p<0.001 respectively). 86Y frequency declined markedly after ACT policy introduction, with a borderline significant trend for a more rapid decline in countries with AL policies (p=0.055). However, these trends could also be explained by a difference in initial SNP frequencies at the time of ACT introduction. There were non-significant trends for faster selection of N86 and D1246 in areas with higher AL consumption and no trend with transmission intensity. Recorded consumption of AS-AQ was low in the locations and times Pfmdr1 data were collected. SNP trends in countries with AL policies suggest a broad increase in sensitivity of parasites to AS-AQ, by 7–10 years after AL introduction. Observed rates of selection have implications for planning strategies to cycle drugs or use multiple first-line therapies to maintain drug efficacy.
Collapse
Affiliation(s)
- Lucy C Okell
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Lisa Malene Reiter
- Global Health Section, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Lene Sandø Ebbe
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Disease, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vito Baraka
- Department of Biomedical Sciences, National Institute for Medical Research, Tanga, United Republic of Tanzania
| | - Donal Bisanzio
- RTI International, Washington, District of Columbia, USA
| | - Oliver J Watson
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Adam Bennett
- Malaria Elimination Initiative, Global Health Group, University of San FranciscO, San Francisco, California, USA
| | - Robert Verity
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Peter Gething
- Malaria Atlas Project, Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Cally Roper
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Ebenebe JC, Ntadom G, Ambe J, Wammanda R, Jiya N, Finomo F, Emechebe G, Mokuolu O, Akano K, Agomo C, Folarin OA, Oguche S, Useh F, Oyibo W, Aderoyeje T, Abdulkadir M, Ezeigwe NM, Happi C, Sowunmi A. Efficacy of Artemisinin-Based Combination Treatments of Uncomplicated Falciparum Malaria in Under-Five-Year-Old Nigerian Children Ten Years Following Adoption as First-Line Antimalarials. Am J Trop Med Hyg 2018; 99:649-664. [PMID: 29943725 PMCID: PMC6169162 DOI: 10.4269/ajtmh.18-0115] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/08/2018] [Indexed: 11/07/2022] Open
Abstract
The efficacies of 3-day regimens of artemether-lumefantrine (AL), artesunate-amodiaquine (AA), and dihydroartemisinin-piperaquine (DHP) were evaluated in 910 children < 5 years old with uncomplicated malaria from six geographical areas of Nigeria. Parasite positivity 1 day and Kaplan-Meier estimated risk of persistent parasitemia 3 days after therapy initiation were both significantly higher, and geometric mean parasite reduction ratio 1 day after treatment initiation (PRRD1) was significantly lower in AL-treated children than in AA- and DHP-treated children. No history of fever, temperature > 38°C, enrollment parasitemia > 75,000 μL-1, and PRRD1 < 5,000 independently predicted persistent parasitemia 1 day after treatment initiation. Parasite clearance was significantly faster and risk of reappearance of asexual parasitemia after initial clearance was significantly lower in DHP-treated children. Overall, day 42 polymerase chain reaction-corrected efficacy was 98.3% (95% confidence interval [CI]: 96.1-100) and was similar for all treatments. In a non-compartment model, declines of parasitemias were monoexponential with mean terminal elimination half-life of 1.3 hours and unimodal frequency distribution of half-lives. All treatments were well tolerated. In summary, all three treatments evaluated remain efficacious treatments of uncomplicated malaria in young Nigerian children, but DHP appears more efficacious than AL or AA.
Collapse
Affiliation(s)
- Joy C. Ebenebe
- Department of Paediatrics, Nnamdi Azikiwe University, Awka, Nigeria
| | - Godwin Ntadom
- National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
| | - Jose Ambe
- Department of Paediatrics, University of Maiduguri, Maiduguri, Nigeria
| | | | - Nma Jiya
- Department of Paediatrics, Uthman Dan Fodio University, Sokoto, Nigeria
| | - Finomo Finomo
- Department of Paediatrics, Federal Medical Centre, Yenagoa, Nigeria
| | - George Emechebe
- Department of Paediatrics, Imo State University Teaching Hospital, Orlu, Nigeria
| | - Olugbenga Mokuolu
- Department of Paediatrics and Child Health, University of Ilorin, Ilorin, Nigeria
| | - Kazeem Akano
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Chimere Agomo
- Department of Medical Laboratory Science, University of Lagos, Lagos, Nigeria
| | - Onikepe A. Folarin
- Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Stephen Oguche
- Department of Paediatrics, University of Jos, Jos, Nigeria
| | - Francis Useh
- Department of Medical Laboratory Science, University of Calabar, Calabar, Nigeria
| | - Wellington Oyibo
- Department of Medical Microbiology and Parasitology, University of Lagos, Lagos, Nigeria
| | - Temitope Aderoyeje
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
- Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria
| | - Mohammed Abdulkadir
- Department of Paediatrics and Child Health, University of Ilorin, Ilorin, Nigeria
| | - Nnenna M. Ezeigwe
- National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
| | - Christian Happi
- Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
| | - Akintunde Sowunmi
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
- Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria
| | - for the Antimalarial Therapeutic Efficacy Monitoring Group, National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, Nnamdi Azikiwe University, Awka, Nigeria
- National Malaria Elimination Programme, The Federal Ministry of Health, Abuja, Nigeria
- Department of Paediatrics, University of Maiduguri, Maiduguri, Nigeria
- Department of Paediatrics, Ahmadu Bello University, Zaria, Nigeria
- Department of Paediatrics, Uthman Dan Fodio University, Sokoto, Nigeria
- Department of Paediatrics, Federal Medical Centre, Yenagoa, Nigeria
- Department of Paediatrics, Imo State University Teaching Hospital, Orlu, Nigeria
- Department of Paediatrics and Child Health, University of Ilorin, Ilorin, Nigeria
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Department of Medical Laboratory Science, University of Lagos, Lagos, Nigeria
- Department of Biological Sciences and African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer University, Ede, Nigeria
- Department of Paediatrics, University of Jos, Jos, Nigeria
- Department of Medical Laboratory Science, University of Calabar, Calabar, Nigeria
- Department of Medical Microbiology and Parasitology, University of Lagos, Lagos, Nigeria
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
- Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
23
|
Chan XHS, Win YN, Mawer LJ, Tan JY, Brugada J, White NJ. Risk of sudden unexplained death after use of dihydroartemisinin-piperaquine for malaria: a systematic review and Bayesian meta-analysis. THE LANCET. INFECTIOUS DISEASES 2018; 18:913-923. [PMID: 29887371 PMCID: PMC6060085 DOI: 10.1016/s1473-3099(18)30297-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dihydroartemisinin-piperaquine is an effective and well tolerated artemisinin-based combination therapy that has been assessed extensively for the prevention and treatment of malaria. Piperaquine, similar to several structurally related antimalarials currently used, can prolong cardiac ventricular repolarisation duration and the electrocardiographic QT interval, leading to concerns about its proarrhythmic potential. We aimed to assess the risk of potentially lethal iatrogenic ventricular arrhythmias in individuals receiving dihydroartemisinin-piperaquine. METHODS We did a systematic review and Bayesian meta-analysis. We searched clinical bibliographic databases (last on May 24, 2017) for studies of dihydroartemisinin-piperaquine in human beings. Further unpublished studies were identified with the WHO Evidence Review Group on the Cardiotoxicity of Antimalarials. We searched for articles containing "dihydroartemisinin-piperaquine" as title, abstract, or subject heading keywords, with synonyms and variant spellings as additional search terms. We excluded animal studies, but did not apply limits on language or publication date. Eligible studies were prospective, randomised, controlled trials or cohort studies in which individuals received at least one 3-day treatment course of dihydroartemisinin-piperaquine for mass drug administration, preventive therapy, or case management of uncomplicated malaria, with follow-up over at least 3 days. At least two independent reviewers screened titles, abstracts, and full texts, agreed study eligibility, and extracted information about study and participant characteristics, adverse event surveillance methodology, dihydroartemisinin-piperaquine exposures, loss-to-follow up, and any deaths after dihydroartemisinin-piperaquine treatment into a standardised database. The risk of sudden unexplained death after dihydroartemisinin-piperaquine with 95% credible intervals (CI) generated by Bayesian meta-analysis was compared with the baseline rate of sudden cardiac death. FINDINGS Our search identified 94 eligible primary studies including data for 197 867 individuals who had received dihydroartemisinin-piperaquine: 154 505 in mass drug administration programmes; 15 188 in 14 studies of repeated courses in preventive therapies and case management of uncomplicated malaria; and 28 174 as single-course treatments of uncomplicated malaria in 76 case-management studies. There was one potentially drug-related sudden unexplained death: a healthy woman aged 16 in Mozambique who developed heart palpitations several hours after the second dose of dihydroartemisinin-piperaquine and collapsed and died on the way to hospital (no autopsy or ECG was done). The median pooled risk estimate of sudden unexplained death after dihydroartemisinin-piperaquine was 1 in 757 950 (95% CI 1 in 2 854 490 to 1 in 209 114). This risk estimate was not higher than the baseline rate of sudden cardiac death (0·7-11·9 per 100 000 person-years or 1 in 1 714 280 to 1 in 100 835 over a 30-day risk period). The risk of bias was low in most studies and unclear in a few. INTERPRETATION Dihydroartemisinin-piperaquine was associated with a low risk of sudden unexplained death that was not higher than the baseline rate of sudden cardiac death. Concerns about repolarisation-related cardiotoxicity need not limit its current use for the prevention and treatment of malaria. FUNDING Wellcome Trust, UK Medical Research Council, WHO, Bill & Melinda Gates Foundation, and University of Oxford.
Collapse
Affiliation(s)
- Xin Hui S Chan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Yan Naung Win
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Defence Services Medical Research Centre & Health and Disease Control Unit, Naypyidaw, Myanmar
| | - Laura J Mawer
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Royal Free London NHS Foundation Trust, London, UK
| | - Jireh Y Tan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Josep Brugada
- Arrhythmia Section, Cardiology Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Spain
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
24
|
Ghosh S, Pathak S, Sonawat HM, Sharma S, Sengupta A. Metabolomic changes in vertebrate host during malaria disease progression. Cytokine 2018; 112:32-43. [PMID: 30057363 DOI: 10.1016/j.cyto.2018.07.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022]
Abstract
Metabolomics refers to top-down systems biological analysis of metabolites in biological specimens. Phenotypic proximity of metabolites makes them interesting candidates for studying biomarkers of environmental stressors such as parasitic infections. Moreover, the host-parasite interaction directly impinges upon metabolic pathways since the parasite uses the host metabolite pool as a biosynthetic resource. Malarial infection, although not recognized as a classic metabolic disorder, often leads to severe metabolic changes such as hypoglycemia and lactic acidosis. Thus, metabolomic analysis of the infection has become an invaluable tool for promoting a better understanding of the host-parasite interaction and for the development of novel therapeutics. In this review, we summarize the current knowledge obtained from metabolomic studies of malarial infection in rodent models and human patients. Metabolomic analysis of experimental rodent malaria has provided significant insights into the mechanisms of disease progression including utilization of host resources by the parasite, sexual dimorphism in metabolic phenotypes, and cellular changes in host metabolism. Moreover, these studies also provide proof of concept for prediction of cerebral malaria. On the other hand, metabolite analysis of patient biofluids generates extensive data that could be of use in identifying biomarkers of infection severity and in monitoring disease progression. Through the use of metabolomic datasets one hopes to assess crucial infection-specific issues such as clinical severity, drug resistance, therapeutic targets, and biomarkers. Also discussed are nascent or newly emerging areas of metabolomics such as pre-erythrocytic stages of the infection and the host immune response. This review is organized in four broad sections-methodologies for metabolomic analysis, rodent infection models, studies of human clinical specimens, and potential of immunometabolomics. Data summarized in this review should serve as a springboard for novel hypothesis testing and lead to a better understanding of malarial infection and parasite biology.
Collapse
Affiliation(s)
- Soumita Ghosh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Sulabha Pathak
- Department of Biological Sciences, Tata Institute of Fundamental Research, 1, Homi Bhabha Road, Mumbai 400005, India
| | - Haripalsingh M Sonawat
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1, Homi Bhabha Road, Mumbai 400005, India
| | - Shobhona Sharma
- Department of Biological Sciences, Tata Institute of Fundamental Research, 1, Homi Bhabha Road, Mumbai 400005, India
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Kloprogge F, Workman L, Borrmann S, Tékété M, Lefèvre G, Hamed K, Piola P, Ursing J, Kofoed PE, Mårtensson A, Ngasala B, Björkman A, Ashton M, Friberg Hietala S, Aweeka F, Parikh S, Mwai L, Davis TME, Karunajeewa H, Salman S, Checchi F, Fogg C, Newton PN, Mayxay M, Deloron P, Faucher JF, Nosten F, Ashley EA, McGready R, van Vugt M, Proux S, Price RN, Karbwang J, Ezzet F, Bakshi R, Stepniewska K, White NJ, Guerin PJ, Barnes KI, Tarning J. Artemether-lumefantrine dosing for malaria treatment in young children and pregnant women: A pharmacokinetic-pharmacodynamic meta-analysis. PLoS Med 2018; 15:e1002579. [PMID: 29894518 PMCID: PMC5997317 DOI: 10.1371/journal.pmed.1002579] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 05/04/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The fixed dose combination of artemether-lumefantrine (AL) is the most widely used treatment for uncomplicated Plasmodium falciparum malaria. Relatively lower cure rates and lumefantrine levels have been reported in young children and in pregnant women during their second and third trimester. The aim of this study was to investigate the pharmacokinetic and pharmacodynamic properties of lumefantrine and the pharmacokinetic properties of its metabolite, desbutyl-lumefantrine, in order to inform optimal dosing regimens in all patient populations. METHODS AND FINDINGS A search in PubMed, Embase, ClinicalTrials.gov, Google Scholar, conference proceedings, and the WorldWide Antimalarial Resistance Network (WWARN) pharmacology database identified 31 relevant clinical studies published between 1 January 1990 and 31 December 2012, with 4,546 patients in whom lumefantrine concentrations were measured. Under the auspices of WWARN, relevant individual concentration-time data, clinical covariates, and outcome data from 4,122 patients were made available and pooled for the meta-analysis. The developed lumefantrine population pharmacokinetic model was used for dose optimisation through in silico simulations. Venous plasma lumefantrine concentrations 7 days after starting standard AL treatment were 24.2% and 13.4% lower in children weighing <15 kg and 15-25 kg, respectively, and 20.2% lower in pregnant women compared with non-pregnant adults. Lumefantrine exposure decreased with increasing pre-treatment parasitaemia, and the dose limitation on absorption of lumefantrine was substantial. Simulations using the lumefantrine pharmacokinetic model suggest that, in young children and pregnant women beyond the first trimester, lengthening the dose regimen (twice daily for 5 days) and, to a lesser extent, intensifying the frequency of dosing (3 times daily for 3 days) would be more efficacious than using higher individual doses in the current standard treatment regimen (twice daily for 3 days). The model was developed using venous plasma data from patients receiving intact tablets with fat, and evaluations of alternative dosing regimens were consequently only representative for venous plasma after administration of intact tablets with fat. The absence of artemether-dihydroartemisinin data limited the prediction of parasite killing rates and recrudescent infections. Thus, the suggested optimised dosing schedule was based on the pharmacokinetic endpoint of lumefantrine plasma exposure at day 7. CONCLUSIONS Our findings suggest that revised AL dosing regimens for young children and pregnant women would improve drug exposure but would require longer or more complex schedules. These dosing regimens should be evaluated in prospective clinical studies to determine whether they would improve cure rates, demonstrate adequate safety, and thereby prolong the useful therapeutic life of this valuable antimalarial treatment.
Collapse
Affiliation(s)
- Frank Kloprogge
- WorldWide Antimalarial Resistance Network, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Institute for Global Health, University College London, London, United Kingdom
| | - Lesley Workman
- WorldWide Antimalarial Resistance Network, Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Steffen Borrmann
- Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
- Institute for Tropical Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Mamadou Tékété
- Institute for Tropical Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | | | - Kamal Hamed
- Novartis Pharmaceuticals, East Hanover, New Jersey, United States of America
| | | | - Johan Ursing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Danderyds Hospital, Stockholm, Sweden
- Bandim Health Project, Bissau, Guinea-Bissau
| | - Poul Erik Kofoed
- Bandim Health Project, Bissau, Guinea-Bissau
- Department of Paediatrics, Kolding Hospital, Kolding, Denmark
| | - Andreas Mårtensson
- Department of Women’s and Children’s Health, International Maternal and Child Health, Uppsala University, Uppsala, Sweden
| | - Billy Ngasala
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Michael Ashton
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden
| | - Sofia Friberg Hietala
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden
- Pharmetheus, Uppsala, Sweden
| | - Francesca Aweeka
- UCSF School of Pharmacy, San Francisco, California, United States of America
| | - Sunil Parikh
- Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Leah Mwai
- Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
- Institute for Tropical Medicine and Joanna Briggs Institute Affiliate Centre for Evidence Based Health Care Evidence Synthesis and Translation Unit, Afya Research Africa, Nairobi, Kenya
- International Development Research Centre, Ottawa, Ontario, Canada
| | - Timothy M. E. Davis
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Harin Karunajeewa
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Sam Salman
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Francesco Checchi
- Epicentre, Paris, France
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Carole Fogg
- Epicentre, Paris, France
- Faculty of Science, University of Portsmouth, Portsmouth, United Kingdom
| | - Paul N. Newton
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Vientiane, Laos
| | - Mayfong Mayxay
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Vientiane, Laos
- Faculty of Postgraduate Studies, University of Health Sciences, Vientiane, Laos
| | - Philippe Deloron
- UMR216 Institut de Recherche pour le Développement, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | | | - François Nosten
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mae Sot, Thailand
| | - Elizabeth A. Ashley
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Myanmar Oxford Clinical Research Unit, Yangon, Myanmar
| | - Rose McGready
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mae Sot, Thailand
| | - Michele van Vugt
- Shoklo Malaria Research Unit, Mae Sot, Thailand
- Amsterdam Medical Centre, Amsterdam, The Netherlands
| | - Stephane Proux
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mae Sot, Thailand
| | - Ric N. Price
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- WorldWide Antimalarial Resistance Network, Darwin, Northern Territory, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Charles Darwin University, Darwin, Northern Territory, Australia
| | - Juntra Karbwang
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Farkad Ezzet
- Novartis Pharmaceuticals, East Hanover, New Jersey, United States of America
| | | | - Kasia Stepniewska
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
| | - Nicholas J. White
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Philippe J. Guerin
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
| | - Karen I. Barnes
- WorldWide Antimalarial Resistance Network, Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Joel Tarning
- WorldWide Antimalarial Resistance Network, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
26
|
Baraka V, Mavoko HM, Nabasumba C, Francis F, Lutumba P, Alifrangis M, Van geertruyden JP. Impact of treatment and re-treatment with artemether-lumefantrine and artesunate-amodiaquine on selection of Plasmodium falciparum multidrug resistance gene-1 polymorphisms in the Democratic Republic of Congo and Uganda. PLoS One 2018; 13:e0191922. [PMID: 29390014 PMCID: PMC5794077 DOI: 10.1371/journal.pone.0191922] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The emergence of resistance against artemisinin combination treatment is a major concern for malaria control. ACTs are recommended as the rescue treatment, however, there is limited evidence as to whether treatment and re-treatment with ACTs select for drug-resistant P. falciparum parasites. Thus, the purpose of the present study is to investigate the impact of (re-)treatment using artesunate-amodiaquine (ASAQ) and artemether-lumefantrine (AL) on the selection of P. falciparum multidrug resistance-1 (Pfmdr1) alleles in clinical settings. METHODS P. falciparum positive samples were collected from children aged 12-59 months in a clinical trial in DR Congo and Uganda. Pfmdr1 single nucleotide polymorphisms (SNPs) analysis at codons N86Y, Y184F, and D1246Y were performed at baseline and post-treatment with either AL or ASAQ as a rescue treatment using nested PCR followed by restriction fragment length polymorphism (RFLP) assays. RESULTS The pre-treatment prevalence of Pfmdr1 N86 and D1246Y varied significantly between the sites, (p>0.001) and (p = 0.013), respectively. There was borderline significant directional selection for Pfmdr1 184F in recurrent malaria infections after treatment with AL in Uganda site (p = 0.05). Pfmdr1 NFD haplotype did not significantly change in post-treatment infections after re-treatment with either AL or ASAQ. Comparison between pre-treatment and post-treatment recurrences did not indicate directional selection of Pfmdr1 N86, D1246 alleles in the pre-RCT, RCT and post-RCT phases in both AL and ASAQ treatment arms. Pfmdr1 86Y was significantly associated with reduced risk of AL treatment failure (RR = 0.34, 95% CI:0.11-1.05, p = 0.04) while no evidence for D1246 allele (RR = 1.02; 95% CI: 0.42-2.47, p = 1.0). Survival estimates showed that the Pfmdr1 alleles had comparable mean-time to PCR-corrected recrudescence and new infections in both AL and ASAQ treatment arms. CONCLUSION We found limited impact of (re-)treatment with AL or ASAQ on selection for Pfmdr1 variants and haplotypes associated with resistance to partner drugs. These findings further supplement the evidence use of same or alternative ACTs as a rescue therapy for recurrent P.falciparum infections. Continued monitoring of genetic signatures of resistance is warranted to timely inform malaria (re-)treatment policies and guidelines.
Collapse
Affiliation(s)
- Vito Baraka
- National Institute for Medical Research, Tanga Centre, Tanga, United Republic of Tanzania
- Global Health Institute, University of Antwerp, Antwerp, Belgium
| | - Hypolite Muhindo Mavoko
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Carolyn Nabasumba
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Epicentre Mbarara Research Base, Mbarara, Uganda
| | - Filbert Francis
- National Institute for Medical Research, Tanga Centre, Tanga, United Republic of Tanzania
| | - Pascal Lutumba
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Michael Alifrangis
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, National University Hospital (Rigshospitalet), Copenhagen, Denmark
| | | |
Collapse
|
27
|
Ayalew MB. Therapeutic efficacy of artemether-lumefantrine in the treatment of uncomplicated Plasmodium falciparum malaria in Ethiopia: a systematic review and meta-analysis. Infect Dis Poverty 2017; 6:157. [PMID: 29137664 PMCID: PMC5686809 DOI: 10.1186/s40249-017-0372-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/26/2017] [Indexed: 12/11/2022] Open
Abstract
Background As Ethiopia is one of the sub-Saharan countries with a great burden of malaria the effectiveness of first line anti-malarial drugs is the major concern. The aim of this study was to synthesize the available evidence on the efficacy of artemether-lumefantrine in the treatment of uncomplicated Plasmodium falciparum malaria in Ethiopia. This was done by performing a meta-analysis of recent studies conducted in the country on this topic. Methods Studies published between January 2010 and January 2017 that reported on the efficacy of artemether-lumefantrine in the treatment of P. falciparum malaria in Ethiopian patients were searched for using the PubMed and Google Scholar databases. Ten prospective single-arm cohort studies that followed patients for 28–42 days were included in this analysis. All of the included studies were deemed to be of high quality. Results Ten studies involving 1179 patients that were eligible for meta-analysis were identified. At recruitment, the average parasite count per patient was 1 2981/μl of blood. On the third day of treatment, 96.7% and 98.5% of the study subjects become fever-free and parasite-free, respectively. Based on the per protocol analysis, the cure rate after use of artemether-lumefantrine was 98.2% (polymerase chain reaction corrected) and 97.01% (polymerase chain reaction uncorrected) after 28 days of follow-up. The reinfection rate within 28 days was 1.1% and the recrudescence rate was 1.9%. Conclusions This review found that the cure rate for uncomplicated P. falciparum malaria using artemether-lumefantrine in Ethiopia is still high enough to recommend the drug as a first-line agent. There should be careful periodic monitoring of the efficacy of this drug, as treatment failure may occur due to resistance, sub-therapeutic levels that may occur due to non-adherence, or inadequate absorption. Electronic supplementary material The online version of this article (10.1186/s40249-017-0372-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohammed Biset Ayalew
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, Gondar University, Gondar, Ethiopia.
| |
Collapse
|
28
|
Schieck E, Poole EJ, Rippert A, Peshu J, Sasi P, Borrmann S, Bull PC. Plasmodium falciparum variant erythrocyte surface antigens: a pilot study of antibody acquisition in recurrent natural infections. Malar J 2017; 16:450. [PMID: 29115961 PMCID: PMC5678811 DOI: 10.1186/s12936-017-2097-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During intra-erythrocytic replication Plasmodium falciparum escapes the human host immune system by switching expression of variant surface antigens (VSA). Piecemeal acquisition of variant specific antibody responses to these antigens as a result of exposure to multiple re-infections has been proposed to play a role in acquisition of naturally acquired immunity. METHODS Immunofluorescence was used to explore the dynamics of anti-VSA IgG responses generated by children to (i) primary malaria episodes and (ii) recurrent P. falciparum infections. RESULTS Consistent with previous studies on anti-VSA responses, sera from each child taken at the time of recovery from their respective primary infection tended to recognize their own secondary parasites poorly. Additionally, compared to patients with reinfections by parasites of new merozoite surface protein 2 (MSP2) genotypes, baseline sera sampled from patients with persistent infections (recrudescence) tended to have higher recognition of heterologous parasites. This is consistent with the prediction that anti-VSA IgG responses may play a role in promoting chronic asymptomatic infections. CONCLUSIONS This pilot study validates the utility of recurrent natural malaria infections as a functional readout for examining the incremental acquisition of immunity to malaria.
Collapse
Affiliation(s)
- Elise Schieck
- Kenya Medical Research Institute/Wellcome Trust Research Programme, Center for Geographic Medicine Research-Coast, P. O. Box 428, Kilifi, 80108, Kenya. .,Institute of Hygiene, University of Heidelberg School of Medicine, 69120, Heidelberg, Germany. .,International Livestock Research Institute, Old Naivasha Road, P.O. Box 30709, Nairobi, 00100, Kenya.
| | - E Jane Poole
- International Livestock Research Institute, Old Naivasha Road, Nairobi, Kenya
| | - Anja Rippert
- Institute of Hygiene, University of Heidelberg School of Medicine, 69120, Heidelberg, Germany.,Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Judy Peshu
- Kenya Medical Research Institute/Wellcome Trust Research Programme, Center for Geographic Medicine Research-Coast, P. O. Box 428, Kilifi, 80108, Kenya
| | - Philip Sasi
- Department of Clinical Pharmacology, School of Medicine, Muhimbili University of Health and Allied Sciences, P.O. Box 65010, Dar es Salaam, Tanzania
| | - Steffen Borrmann
- Kenya Medical Research Institute/Wellcome Trust Research Programme, Center for Geographic Medicine Research-Coast, P. O. Box 428, Kilifi, 80108, Kenya.,German Center for Infection Research (DZIF), Wilhelmstraße 27, 72074, Tübingen, Germany.,Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Peter C Bull
- Kenya Medical Research Institute/Wellcome Trust Research Programme, Center for Geographic Medicine Research-Coast, P. O. Box 428, Kilifi, 80108, Kenya.,Nuffield Department of Medicine, Centre for Tropical Medicine, Oxford University, Oxford, OX3 7LJ, UK.,Department of Pathology, Cambridge University, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
29
|
Schallig HD, Tinto H, Sawa P, Kaur H, Duparc S, Ishengoma DS, Magnussen P, Alifrangis M, Sutherland CJ. Randomised controlled trial of two sequential artemisinin-based combination therapy regimens to treat uncomplicated falciparum malaria in African children: a protocol to investigate safety, efficacy and adherence. BMJ Glob Health 2017; 2:e000371. [PMID: 29082016 PMCID: PMC5656137 DOI: 10.1136/bmjgh-2017-000371] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Management of uncomplicated Plasmodium falciparum malaria relies on artemisinin-based combination therapies (ACTs). These highly effective regimens have contributed to reductions in malaria morbidity and mortality. However, artemisinin resistance in Asia and changing parasite susceptibility to ACT in Africa have now been well documented. Strategies that retain current ACT as efficacious treatments are urgently needed. METHODS We present an open-label, randomised three-arm clinical trial protocol in three African settings representative of varying malaria epidemiology to investigate whether prolonged ACT-based regimens using currently available formulations can eliminate potentially resistant parasites. The protocol investigates whether a sequential course of two licensed ACT in 1080 children aged 6-120 months exhibits superior efficacy against acute P. falciparum malaria and non-inferior safety compared with standard single-course ACT given to 540 children. The primary endpoint is PCR-corrected clinical and parasitological response at day 42 or day 63 of follow-up. Persistence of PCR-detectable parasitaemia at day 3 is analysed as a key covariate. Secondary endpoints include gametocytaemia, occurrence of treatment-related adverse events in the double-ACT versus single-ACT arms, carriage of molecular markers of drug resistance, drug kinetics and patient adherence to treatment. DISCUSSION This protocol addresses efficacy and safety of sequential ACT regimens in P. falciparum malaria in Africa. The approach is designed to extend the useful life of this class of antimalarials with maximal impact and minimal delay, by deploying licensed medicines that could be swiftly implemented as sequential double ACT by National Malaria Control Programmes, before emerging drug resistance in Africa becomes a major threat to public health.
Collapse
Affiliation(s)
- Henk Dfh Schallig
- Department of Medical Microbiology-Parasitology Unit, Academic Medical Centre, Amsterdam, The Netherlands
| | - Halidou Tinto
- Institute for Health Sciences Research-Clinical Research Unit (IRSS-CRUN), Nanoro, Burkina Faso
| | - Patrick Sawa
- International Centre for Insect Physiology and Ecology, Mbita Point, Kenya
| | - Harparkash Kaur
- Clinical Research Department, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Immunology & Infection Department, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | | | | | - Pascal Magnussen
- Department of International Health, Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Michael Alifrangis
- Department of International Health, Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Colin J Sutherland
- Immunology & Infection Department, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Public Health England Malaria Reference Laboratory, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
30
|
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
|
31
|
Host immunity to Plasmodium falciparum and the assessment of emerging artemisinin resistance in a multinational cohort. Proc Natl Acad Sci U S A 2017; 114:3515-3520. [PMID: 28289193 DOI: 10.1073/pnas.1615875114] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Artemisinin-resistant falciparum malaria, defined by a slow-clearance phenotype and the presence of kelch13 mutants, has emerged in the Greater Mekong Subregion. Naturally acquired immunity to malaria clears parasites independent of antimalarial drugs. We hypothesized that between- and within-population variations in host immunity influence parasite clearance after artemisinin treatment and the interpretation of emerging artemisinin resistance. Antibodies specific to 12 Plasmodium falciparum sporozoite and blood-stage antigens were determined in 959 patients (from 11 sites in Southeast Asia) participating in a multinational cohort study assessing parasite clearance half-life (PCt1/2) after artesunate treatment and kelch13 mutations. Linear mixed-effects modeling of pooled individual patient data assessed the association between antibody responses and PCt1/2.P. falciparum antibodies were lowest in areas where the prevalence of kelch13 mutations and slow PCt1/2 were highest [Spearman ρ = -0.90 (95% confidence interval, -0.97, -0.65), and Spearman ρ = -0.94 (95% confidence interval, -0.98, -0.77), respectively]. P. falciparum antibodies were associated with faster PCt1/2 (mean difference in PCt1/2 according to seropositivity, -0.16 to -0.65 h, depending on antigen); antibodies have a greater effect on the clearance of kelch13 mutant compared with wild-type parasites (mean difference in PCt1/2 according to seropositivity, -0.22 to -0.61 h faster in kelch13 mutants compared with wild-type parasites). Naturally acquired immunity accelerates the clearance of artemisinin-resistant parasites in patients with falciparum malaria and may confound the current working definition of artemisinin resistance. Immunity may also play an important role in the emergence and transmission potential of artemisinin-resistant parasites.
Collapse
|
32
|
Sowunmi A, Akano K, Ntadom G, Ayede AI, Ibironke FO, Aderoyeje T, Adewoye EO, Fatunmbi B, Oguche S, Okafor HU, Watila I, Meremikwu M, Agomo P, Ogala W, Agomo C, Folarin OA, Gbotosho GO, Happi CT. Therapeutic efficacy and effects of artemisinin-based combination treatments on uncomplicated Plasmodium falciparum malaria -associated anaemia in Nigerian children during seven years of adoption as first-line treatments. Infect Dis Poverty 2017; 6:36. [PMID: 28173853 PMCID: PMC5294876 DOI: 10.1186/s40249-016-0217-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 12/14/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Artemisinin-based combination treatments (ACTs) are the first-line treatments of uncomplicated Plasmodium falciparum malaria in many endemic areas but there are few evaluation of their efficacy in anaemic malarious children. METHODS Therapeutic efficacy of 3-day regimens of artesunate-amodiaquine and artemether-lumefantrine was evaluated in 437 anaemic and 909 non-anaemic malarious children following treatment during a seven-year period (2008-2014). Patterns of temporal changes in haematocrit were classified based on haematocrit values <30% and ≥30%. Kinetics of the disposition of the deficit in haematocrit from 30% following treatment were evaluated using a non-compartment model. RESULTS PCR-corrected parasitological efficacy 28 days after start of treatment was significantly higher in artesunate-amodiaquine- compared to artemether-lumefantrine-treated children [97% (95%CI: 92.8-100) versus 96.4% (95%CI: 91.3-99.4), P = 0.02], but it was similar in non-anaemic and anaemic children. Fall in haematocrit/1 000 asexual parasites cleared from peripheral blood was significantly greater at lower compared to higher parasitaemias (P < 0.0001), and in non-anaemic compared to anaemic children (P = 0.007). In anaemic children at presentation, mean anaemia recovery time (AnRT) was 15.4 days (95%CI: 13.3-17.4) and it did not change over the years. Declines in haematocrit deficits from 30% were monoexponential with mean estimated half-time of 1.4 days (95%CI: 1.2-1.6). Anaemia half-time (t½anaemia) correlated positively with AnRT in the same patients (r = 0.69, P < 0.0001). Bland-Altman analysis of 10 multiples of t½anaemia and AnRT showed narrow limit of agreement with insignificant bias (P = 0.07) suggesting both can be used interchangeably in the same patients. CONCLUSIONS Artesunate-amodiaquine and artemether-lumefantrine remain efficacious treatments of uncomplicated P. falciparum infections in non-anaemic and anaemic Nigerian children in the last 7 years of adoption as first-line treatments. These ACTs may also conserve haematocrit at high parasitaemias and in anaemic children. TRIALS REGISTRATION Pan African Clinical Trial Registry PACTR201508001188143 , 3 July 2015; PACTR201510001189370 , 3 July 2015; PACTR201508001191898 , 7 July 2015 and PACTR201508001193368 , 8 July 2015.
Collapse
Affiliation(s)
- Akintunde Sowunmi
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Institute for Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
| | - Kazeem Akano
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Godwin Ntadom
- National Malaria Elimination Programme, Federal Ministry of Health, Abuja, Nigeria
| | | | - Folasade O. Ibironke
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
| | - Temitope Aderoyeje
- Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
| | | | - Bayo Fatunmbi
- World Health Organization, Regional Office for the Western Pacific, Khan Daun Penh, Phnom Penh Cambodia
| | - Stephen Oguche
- Department of Paediatrics, University of Jos, Jos, Nigeria
| | - Henrietta U. Okafor
- Department of Paediatrics, Institute of Child Health, University of Nigeria Teaching Hospital, Enugu, Nigeria
| | - Ismaila Watila
- Department of Paediatrics, Specialist Hospital, Maiduguri, Borno Sate Nigeria
| | - Martin Meremikwu
- Department of Paediatrics, University of Calabar, Calabar, Cross Rivers State Nigeria
| | - Philip Agomo
- Nigeria Institute of Medical Research, Yaba, Lagos, Nigeria
| | - William Ogala
- Department of Paediatrics, Ahmadu Bello University, Zaria, Nigeria
| | - Chimere Agomo
- Nigeria Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Onikepe A. Folarin
- Department of Biological Sciences, Redeemer’s University, Ede, Osun State Nigeria
| | - Grace O. Gbotosho
- Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- Institute for Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Christian T. Happi
- Department of Biological Sciences, Redeemer’s University, Ede, Osun State Nigeria
| |
Collapse
|
33
|
Hoglund RM, Workman L, Edstein MD, Thanh NX, Quang NN, Zongo I, Ouedraogo JB, Borrmann S, Mwai L, Nsanzabana C, Price RN, Dahal P, Sambol NC, Parikh S, Nosten F, Ashley EA, Phyo AP, Lwin KM, McGready R, Day NPJ, Guerin PJ, White NJ, Barnes KI, Tarning J. Population Pharmacokinetic Properties of Piperaquine in Falciparum Malaria: An Individual Participant Data Meta-Analysis. PLoS Med 2017; 14:e1002212. [PMID: 28072872 PMCID: PMC5224788 DOI: 10.1371/journal.pmed.1002212] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/29/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Artemisinin-based combination therapies (ACTs) are the mainstay of the current treatment of uncomplicated Plasmodium falciparum malaria, but ACT resistance is spreading across Southeast Asia. Dihydroartemisinin-piperaquine is one of the five ACTs currently recommended by the World Health Organization. Previous studies suggest that young children (<5 y) with malaria are under-dosed. This study utilised a population-based pharmacokinetic approach to optimise the antimalarial treatment regimen for piperaquine. METHODS AND FINDINGS Published pharmacokinetic studies on piperaquine were identified through a systematic literature review of articles published between 1 January 1960 and 15 February 2013. Individual plasma piperaquine concentration-time data from 11 clinical studies (8,776 samples from 728 individuals) in adults and children with uncomplicated malaria and healthy volunteers were collated and standardised by the WorldWide Antimalarial Resistance Network. Data were pooled and analysed using nonlinear mixed-effects modelling. Piperaquine pharmacokinetics were described successfully by a three-compartment disposition model with flexible absorption. Body weight influenced clearance and volume parameters significantly, resulting in lower piperaquine exposures in small children (<25 kg) compared to larger children and adults (≥25 kg) after administration of the manufacturers' currently recommended dose regimens. Simulated median (interquartile range) day 7 plasma concentration was 29.4 (19.3-44.3) ng/ml in small children compared to 38.1 (25.8-56.3) ng/ml in larger children and adults, with the recommended dose regimen. The final model identified a mean (95% confidence interval) increase of 23.7% (15.8%-32.5%) in piperaquine bioavailability between each piperaquine dose occasion. The model also described an enzyme maturation function in very young children, resulting in 50% maturation at 0.575 (0.413-0.711) y of age. An evidence-based optimised dose regimen was constructed that would provide piperaquine exposures across all ages comparable to the exposure currently seen in a typical adult with standard treatment, without exceeding the concentration range observed with the manufacturers' recommended regimen. Limited data were available in infants and pregnant women with malaria as well as in healthy individuals. CONCLUSIONS The derived population pharmacokinetic model was used to develop a revised dose regimen of dihydroartemisinin-piperaquine that is expected to provide equivalent piperaquine exposures safely in all patients, including in small children with malaria. Use of this dose regimen is expected to prolong the useful therapeutic life of dihydroartemisinin-piperaquine by increasing cure rates and thereby slowing resistance development. This work was part of the evidence that informed the World Health Organization technical guidelines development group in the development of the recently published treatment guidelines (2015).
Collapse
Affiliation(s)
- Richard M. Hoglund
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lesley Workman
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Michael D. Edstein
- Department of Drug Evaluation, Australian Army Malaria Institute, Brisbane, Queensland, Australia
| | - Nguyen Xuan Thanh
- Department of Malaria, Military Institute of Hygiene and Epidemiology, Hanoi, Viet Nam
| | - Nguyen Ngoc Quang
- Department of Infectious Diseases, Military Hospital 108, Hanoi, Viet Nam
| | - Issaka Zongo
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l’Ouest, Bobo-Dioulasso, Burkina Faso
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jean Bosco Ouedraogo
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l’Ouest, Bobo-Dioulasso, Burkina Faso
| | - Steffen Borrmann
- Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Leah Mwai
- Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
- Joanna Briggs Affiliate Centre for Evidence-Based Health Care, Evidence Synthesis and Translation Unit, Afya Research Africa, Nairobi, Kenya
| | - Christian Nsanzabana
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ric N. Price
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| | - Prabin Dahal
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nancy C. Sambol
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Sunil Parikh
- Yale School of Public Health and Medicine, New Haven, Connecticut, United States of America
| | - Francois Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Elizabeth A. Ashley
- Shoklo Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Aung Pyae Phyo
- Shoklo Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Khin Maung Lwin
- Shoklo Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Rose McGready
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Nicholas P. J. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Philippe J. Guerin
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Karen I. Barnes
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Joel Tarning
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
34
|
Sutherland CJ. Rescuing artemisinin combination therapy in Africa. LANCET GLOBAL HEALTH 2016; 5:e8-e9. [PMID: 27840068 DOI: 10.1016/s2214-109x(16)30291-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Colin J Sutherland
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel St, London, WC1E 7HT, UK.
| |
Collapse
|
35
|
Magistrado PA, Corey VC, Lukens AK, LaMonte G, Sasaki E, Meister S, Wree M, Winzeler E, Wirth DF. Plasmodium falciparum Cyclic Amine Resistance Locus (PfCARL), a Resistance Mechanism for Two Distinct Compound Classes. ACS Infect Dis 2016; 2:816-826. [PMID: 27933786 PMCID: PMC5109296 DOI: 10.1021/acsinfecdis.6b00025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
![]()
MMV007564
is a novel antimalarial benzimidazolyl piperidine chemotype
identified in cellular screens. To identify the genetic determinant
of MMV007564 resistance, parasites were cultured in the presence of
the compound to generate resistant lines. Whole genome sequencing
revealed distinct mutations in the gene named Plasmodium
falciparum cyclic amine resistance locus (pfcarl), encoding a conserved protein of unknown function.
Mutations in pfcarl are strongly associated with
resistance to a structurally unrelated class of compounds, the imidazolopiperazines,
including KAF156, currently in clinical trials. Our data demonstrate
that pfcarl mutations confer resistance to two distinct
compound classes, benzimidazolyl piperidines and imidazolopiperazines.
However, MMV007564 and the imidazolopiperazines, KAF156 and GNF179,
have different timings of action in the asexual blood stage and different
potencies against the liver and sexual blood stages. These data suggest
that pfcarl is a multidrug-resistance gene rather
than a common target for benzimidazolyl piperidines and imidazolopiperazines.
Collapse
Affiliation(s)
- Pamela A. Magistrado
- Department
of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Victoria C. Corey
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Amanda K. Lukens
- Department
of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, United States
- Infectious
Disease Program, The Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, United States
| | - Greg LaMonte
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Erika Sasaki
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Stephan Meister
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Melanie Wree
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Elizabeth Winzeler
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Dyann F. Wirth
- Department
of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, United States
- Infectious
Disease Program, The Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
36
|
Sondén K, Wyss K, Jovel I, Vieira da Silva A, Pohanka A, Asghar M, Homann MV, Gustafsson LL, Hellgren U, Färnert A. High Rate of Treatment Failures in Nonimmune Travelers Treated With Artemether-Lumefantrine for Uncomplicated Plasmodium falciparum Malaria in Sweden: Retrospective Comparative Analysis of Effectiveness and Case Series. Clin Infect Dis 2016; 64:199-206. [PMID: 27986683 DOI: 10.1093/cid/ciw710] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 10/22/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Artemisinin-based combination therapy (ACT) is the first-line treatment of Plasmodium falciparum malaria. Since the introduction of artemether-lumefantrine (AL) for treatment of uncomplicated malaria in Sweden, treatment failures have been reported in adults. METHODS A retrospective comparative analysis of treatment regimen for P. falciparum malaria in adults in Stockholm during 2000-2015 was performed to evaluate the effectiveness of AL. Parasite genotyping and drug concentrations were investigated in the AL treatment failures. RESULTS Among the total 397 P. falciparum episodes, 310 were treated with oral regimen only (95 AL, 162 mefloquine, 36 atovaquone-proguanil [AP], and 17 others), and 87 were administered initial intravenous therapy (38 artesunate and 49 quinine) followed by oral treatments. Five late treatment failures were detected after AL and one slow response to AP. The effectiveness of AL alone was 94.7% (95% confidence interval [CI], 88.1%-98.3%), compared with 99.5% for other oral regimens (P = .003). All AL failures occurred in European men and the effectiveness in this group was only 73.7% (95% CI, 48.8%-90.0%). Genotyping confirmed recrudescence of the initial parasite populations and drug resistance markers revealed no clinically significant resistance patterns. Lumefantrine concentrations suggested subtherapeutic concentrations in at least 2 cases. CONCLUSIONS Our findings indicate a high rate of symptomatic late treatment failures after 6-dose AL regime in nonimmune adults, especially in men. Our report warrants the need to establish optimal dosing of AL in adults and to alert clinicians about the importance of informing patients regarding the risk of parasites reappearing weeks after AL treatment.
Collapse
Affiliation(s)
- Klara Sondén
- Unit of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet;
| | - Katja Wyss
- Unit of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet.,Department of Emergency Medicine, Karolinska University Hospital Solna
| | - Irina Jovel
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet
| | | | - Anton Pohanka
- Department of Clinical Pharmacology, Karolinska University Hospital Huddinge.,Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet
| | - Muhammad Asghar
- Unit of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet
| | - Manijeh Vafa Homann
- Unit of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet
| | - Lars L Gustafsson
- Department of Clinical Pharmacology, Karolinska University Hospital Huddinge.,Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet
| | - Urban Hellgren
- Department of Infectious Diseases, Karolinska University Hospital; and.,Unit of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Anna Färnert
- Unit of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet.,Department of Infectious Diseases, Karolinska University Hospital; and
| |
Collapse
|
37
|
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
|
38
|
Woodrow CJ, White NJ. The clinical impact of artemisinin resistance in Southeast Asia and the potential for future spread. FEMS Microbiol Rev 2016; 41:34-48. [PMID: 27613271 PMCID: PMC5424521 DOI: 10.1093/femsre/fuw037] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/11/2016] [Accepted: 07/31/2016] [Indexed: 11/25/2022] Open
Abstract
Artemisinins are the most rapidly acting of currently available antimalarial drugs. Artesunate has become the treatment of choice for severe malaria, and artemisinin-based combination therapies (ACTs) are the foundation of modern falciparum malaria treatment globally. Their safety and tolerability profile is excellent. Unfortunately, Plasmodium falciparum infections with mutations in the ‘K13’ gene, with reduced ring-stage susceptibility to artemisinins, and slow parasite clearance in patients treated with ACTs, are now widespread in Southeast Asia. We review clinical efficacy data from the region (2000–2015) that provides strong evidence that the loss of first-line ACTs in western Cambodia, first artesunate-mefloquine and then DHA-piperaquine, can be attributed primarily to K13 mutated parasites. The ring-stage activity of artemisinins is therefore critical for the sustained efficacy of ACTs; once it is lost, rapid selection of partner drug resistance and ACT failure are inevitable consequences. Consensus methods for monitoring artemisinin resistance are now available. Despite increased investment in regional control activities, ACTs are failing across an expanding area of the Greater Mekong subregion. Although multiple K13 mutations have arisen independently, successful multidrug-resistant parasite genotypes are taking over and threaten to spread to India and Africa. Stronger containment efforts and new approaches to sustaining long-term efficacy of antimalarial regimens are needed to prevent a global malaria emergency. Artemisinin resistance in Plasmodium falciparum malaria is causing failure of artemisinin-based combination therapies across an expanding area of Southeast Asia, undermining control and elimination efforts. The potential global consequences can only be avoided by new approaches that ensure sustained efficacy for antimalarial regimens in malaria affected populations.
Collapse
Affiliation(s)
- Charles J Woodrow
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 420/6, Rajvithi Road, Bangkok 10400, Thailand
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 420/6, Rajvithi Road, Bangkok 10400, Thailand
| |
Collapse
|
39
|
Sowunmi A, Akano K, Ayede AI, Ntadom G, Adewoye EO, Fatunmbi B, Aderoyeje T. Therapeutic efficacy and effects of artesunate-amodiaquine and artemether-lumefantrine on malaria-associated anaemia in Nigerian children aged two years and under. Infect Dis Poverty 2016; 5:70. [PMID: 27384596 PMCID: PMC4933999 DOI: 10.1186/s40249-016-0165-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Artemisinin-based combination therapies are recommended as first-line treatments for uncomplicated falciparum malaria, but there is little evaluation of their efficacy and effects on uncomplicated malaria-associated anaemia in children aged 2 years and under. METHODS Parasitological efficacy and effects on malaria-associated anaemia were evaluated in 250 malarious children aged 2 years and under, and efficacy was evaluated in 603 malarious children older than two but younger than 5 years of age following treatment with artesunate-amodiaquine (AA) or artemether-lumefantrine (AL). Kinetics of the disposition of parasitaemia following treatment were evaluated using a non-compartment model. Late-appearing anaemia (LAA) was diagnosed using the following criteria: clearance of parasitaemia, fever and other symptoms occurring within 7 days of starting treatment, adequate clinical and parasitological response on days 28-42, haematocrit (HCT) ≥ 30 % at 1 and/or 2 weeks, a fall in HCT to < 30 % occurring at 3-6 weeks, absence of concomitant illness at 1-6 weeks, and absence of asexual parasitaemia detected using both microscopy and polymerase chain reaction (PCR) at 1-6 weeks. RESULTS Overall, in children aged 2 years and under, the PCR-corrected parasitological efficacy was 97.2 % (95 % CI 92.8-101.6), which was similar for both treatments. In children older than 2 years, parasitological efficacy was also similar for both treatments, but parasite prevalence 1 day after treatment began was significantly higher, and fever and parasite clearance times were significantly faster in the AA-treated children compared with the AL-treated children. Declines in parasitaemia were monoexponential with an estimated elimination half-time of 1 h. Elimination half-times were similar for both treatments. In children aged 2 years and under who were anaemic at presentation, the mean anaemia recovery time was 12.1 days (95 % CI 10.6-13.6, n = 127), which was similar for both treatments. Relatively asymptomatic LAA occurred in 11 children (4.4 %) aged 2 years and under, the recovery from which was uneventful. CONCLUSION This study showed that AA and AL are efficacious treatments for uncomplicated falciparum malaria in Nigerian children aged 2 years and under, and that AA clears parasitaemia and fever significantly faster than AL in children older than 2 years. Both treatments may cause a relatively asymptomatic LAA with uneventful recovery in a small proportion of children aged 2 years and under. TRIALS REGISTRATION Pan African Clinical Trial Registry PACTR201508001188143, 3 July 2015; PACTR201510001189370, 3 July 2015; PACTR201508001191898, 7 July 2015 and PACTR201508001193368, 8 July 2015 http://www.pactr.org .
Collapse
Affiliation(s)
- Akintunde Sowunmi
- />Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
- />Institute for Medical Research and Training, University of Ibadan, Ibadan, Nigeria
| | - Kazeem Akano
- />Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | | | - Godwin Ntadom
- />Malaria Elimination Programme, Federal Ministry of Health, Abuja, Nigeria
| | - Elsie O. Adewoye
- />Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - Bayo Fatunmbi
- />World Health Organization, Regional Office for the Western Pacific, Phnom Penh, Cambodia
| | - Temitope Aderoyeje
- />Department of Clinical Pharmacology, University College Hospital, Ibadan, Nigeria
| |
Collapse
|
40
|
Ménard S, Ben Haddou T, Ramadani AP, Ariey F, Iriart X, Beghain J, Bouchier C, Witkowski B, Berry A, Mercereau-Puijalon O, Benoit-Vical F. Induction of Multidrug Tolerance in Plasmodium falciparum by Extended Artemisinin Pressure. Emerg Infect Dis 2016; 21:1733-41. [PMID: 26401601 PMCID: PMC4593447 DOI: 10.3201/eid2110.150682] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Tolerance is not detected by current assays and represents a major threat to antimalarial drug policy. Plasmodium falciparum resistance to artemisinin derivatives in Southeast Asia threatens global malaria control strategies. Whether delayed parasite clearance, which exposes larger parasite numbers to artemisinins for longer times, selects higher-grade resistance remains unexplored. We investigated whether long-lasting artemisinin pressure selects a novel multidrug-tolerance profile. Although 50% inhibitory concentrations for 10 antimalarial drugs tested were unchanged, drug-tolerant parasites showed higher recrudescence rates for endoperoxides, quinolones, and an antifolate, including partner drugs of recommended combination therapies, but remained susceptible to atovaquone. Moreover, the age range of intraerythrocytic stages able to resist artemisinin was extended to older ring forms and trophozoites. Multidrug tolerance results from drug-induced quiescence, which enables parasites to survive exposure to unrelated antimalarial drugs that inhibit a variety of metabolic pathways. This novel resistance pattern should be urgently monitored in the field because this pattern is not detected by current assays and represents a major threat to antimalarial drug policy.
Collapse
|
41
|
Ochola-Oyier LI, Okombo J, Wagatua N, Ochieng J, Tetteh KK, Fegan G, Bejon P, Marsh K. Comparison of allele frequencies of Plasmodium falciparum merozoite antigens in malaria infections sampled in different years in a Kenyan population. Malar J 2016; 15:261. [PMID: 27154310 PMCID: PMC4858837 DOI: 10.1186/s12936-016-1304-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/20/2016] [Indexed: 11/24/2022] Open
Abstract
Background Plasmodium falciparum merozoite antigens elicit antibody responses in malaria-endemic populations, some of which are clinically protective, which is one of the reasons why merozoite antigens are the focus of malaria vaccine development efforts. Polymorphisms in several merozoite antigen-encoding genes are thought to arise as a result of selection by the human immune system. Methods The allele frequency distribution of 15 merozoite antigens over a two-year period, 2007 and 2008, was examined in parasites obtained from children with uncomplicated malaria. In the same population, allele frequency changes pre- and post-anti-malarial treatment were also examined. Any gene which showed a significant shift in allele frequencies was also assessed longitudinally in asymptomatic and complicated malaria infections. Results Fluctuating allele frequencies were identified in codons 147 and 148 of reticulocyte-binding homologue (Rh) 5, with a shift from HD to YH haplotypes over the two-year period in uncomplicated malaria infections. However, in both the asymptomatic and complicated malaria infections YH was the dominant and stable haplotype over the two-year and ten-year periods, respectively. A logistic regression analysis of all three malaria infection populations between 2007 and 2009 revealed, that the chance of being infected with the HD haplotype decreased with time from 2007 to 2009 and increased in the uncomplicated and asymptomatic infections. Conclusion Rh5 codons 147 and 148 showed heterogeneity at both an individual and population level and may be under some degree of immune selection. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1304-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - John Okombo
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Njoroge Wagatua
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Jacob Ochieng
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Kevin K Tetteh
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Greg Fegan
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Philip Bejon
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Kevin Marsh
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| |
Collapse
|
42
|
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
|
43
|
Sondo P, Derra K, Diallo Nakanabo S, Tarnagda Z, Kazienga A, Zampa O, Valéa I, Sorgho H, Owusu-Dabo E, Ouédraogo JB, Guiguemdé TR, Tinto H. Artesunate-Amodiaquine and Artemether-Lumefantrine Therapies and Selection of Pfcrt and Pfmdr1 Alleles in Nanoro, Burkina Faso. PLoS One 2016; 11:e0151565. [PMID: 27031231 PMCID: PMC4816516 DOI: 10.1371/journal.pone.0151565] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/01/2016] [Indexed: 11/18/2022] Open
Abstract
The adoption of Artemisinin based combination therapies (ACT) constitutes a basic strategy for malaria control in sub-Saharan Africa. Moreover, since cases of ACT resistance have been reported in South-East Asia, the need to understand P. falciparum resistance mechanism to ACT has become a global research goal. The selective pressure of ACT and the possibility that some specific Pfcrt and Pfmdr1 alleles are associated with treatment failures was assessed in a clinical trial comparing ASAQ to AL in Nanoro. Dried blood spots collected on Day 0 and on the day of recurrent parasitaemia during the 28-day follow-up were analyzed using the restriction fragments length polymorphism (PCR-RFLP) method to detect single nucleotide polymorphisms (SNPs) in Pfcrt (codon76) and Pfmdr1 (codons 86, 184, 1034, 1042, and 1246) genes. Multivariate analysis of the relationship between the presence of Pfcrt and Pfmdr1 alleles and treatment outcome was performed. AL and ASAQ exerted opposite trends in selecting Pfcrt K76T and Pfmdr1-N86Y alleles, raising the potential beneficial effect of using diverse ACT at the same time as first line treatments to reduce the selective pressure by each treatment regimen. No clear association between the presence of Pfcrt and Pfmdr1 alleles carried at baseline and treatment failure was observed.
Collapse
Affiliation(s)
- Paul Sondo
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
- * E-mail:
| | - Karim Derra
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Seydou Diallo Nakanabo
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
| | - Zekiba Tarnagda
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Adama Kazienga
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Odile Zampa
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
| | - Innocent Valéa
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
| | - Hermann Sorgho
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Ellis Owusu-Dabo
- Kumasi Center for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | | | | | - Halidou Tinto
- IRSS / Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
| |
Collapse
|
44
|
Abdulla S, Ashley EA, Bassat Q, Bethell D, Björkman A, Borrmann S, D'Alessandro U, Dahal P, Day NP, Diakite M, Djimde AA, Dondorp AM, Duong S, Edstein MD, Fairhurst RM, Faiz MA, Falade C, Flegg JA, Fogg C, Gonzalez R, Greenwood B, Guérin PJ, Guthmann JP, Hamed K, Hien TT, Htut Y, Juma E, Lim P, Mårtensson A, Mayxay M, Mokuolu OA, Moreira C, Newton P, Noedl H, Nosten F, Ogutu BR, Onyamboko MA, Owusu-Agyei S, Phyo AP, Premji Z, Price RN, Pukrittayakamee S, Ramharter M, Sagara I, Se Y, Suon S, Stepniewska K, Ward SA, White NJ, Winstanley PA. Baseline data of parasite clearance in patients with falciparum malaria treated with an artemisinin derivative: an individual patient data meta-analysis. Malar J 2015; 14:359. [PMID: 26390866 PMCID: PMC4578675 DOI: 10.1186/s12936-015-0874-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/26/2015] [Indexed: 11/15/2022] Open
Abstract
Background Artemisinin resistance in Plasmodium falciparum manifests as slow parasite clearance but this measure is also influenced by host immunity, initial parasite biomass and partner drug efficacy. This study collated data from clinical trials of artemisinin derivatives in falciparum malaria with frequent
parasite counts to provide reference parasite clearance estimates stratified by location, treatment and time, to examine host factors affecting parasite clearance, and to assess the relationships between parasite clearance and risk of recrudescence during follow-up. Methods Data from 24 studies, conducted from 1996 to 2013, with frequent parasite counts were pooled. Parasite clearance half-life (PC1/2) was estimated using the WWARN Parasite Clearance Estimator. Random effects regression models accounting for study and site heterogeneity were used to explore factors affecting PC1/2 and risk of recrudescence within areas with reported delayed parasite clearance (western Cambodia, western Thailand after 2000, southern Vietnam, southern Myanmar) and in all other areas where parasite populations are artemisinin sensitive. Results PC1/2 was estimated in 6975 patients, 3288 of whom also had treatment outcomes evaluate d during 28–63 days follow-up, with 93 (2.8 %) PCR-confirmed recrudescences. In areas with artemisinin-sensitive parasites, the median PC1/2 following three-day artesunate treatment (4 mg/kg/day) ranged from 1.8 to 3.0 h and the proportion of patients with PC1/2 >5 h from 0 to 10 %. Artesunate doses of 4 mg/kg/day decreased PC1/2 by 8.1 % (95 % CI 3.2–12.6) compared to 2 mg/kg/day, except in populations with delayed parasite clearance. PC1/2 was longer in children and in patients with fever or anaemia at enrolment. Long PC1/2 (HR = 2.91, 95 % CI 1.95–4.34 for twofold increase, p < 0.001) and high initial parasitaemia (HR = 2.23, 95 % CI 1.44–3.45 for tenfold increase, p < 0.001) were associated independently with an increased risk of recrudescence. In western Cambodia, the region with the highest prevalence of artemisinin resistance, there was no evidence for increasing PC1/2 since 2007. Conclusions Several factors affect PC1/2. As substantial heterogeneity in parasite clearance exists between locations, early detection of artemisinin resistance requires reference PC1/2 data. Studies with frequent parasite count measurements to characterize PC1/2 should be encouraged. In western Cambodia, where PC1/2 values are longest, there is no evidence for recent emergence of higher levels of artemisinin resistance. Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0874-1) contains supplementary material, which is available to authorized users.
Collapse
|
45
|
Artemether-lumefantrine treatment of uncomplicated Plasmodium falciparum malaria: a systematic review and meta-analysis of day 7 lumefantrine concentrations and therapeutic response using individual patient data. BMC Med 2015; 13:227. [PMID: 26381375 PMCID: PMC4574542 DOI: 10.1186/s12916-015-0456-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/18/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Achieving adequate antimalarial drug exposure is essential for curing malaria. Day 7 blood or plasma lumefantrine concentrations provide a simple measure of drug exposure that correlates well with artemether-lumefantrine efficacy. However, the 'therapeutic' day 7 lumefantrine concentration threshold needs to be defined better, particularly for important patient and parasite sub-populations. METHODS The WorldWide Antimalarial Resistance Network (WWARN) conducted a large pooled analysis of individual pharmacokinetic-pharmacodynamic data from patients treated with artemether-lumefantrine for uncomplicated Plasmodium falciparum malaria, to define therapeutic day 7 lumefantrine concentrations and identify patient factors that substantially alter these concentrations. A systematic review of PubMed, Embase, Google Scholar, ClinicalTrials.gov and conference proceedings identified all relevant studies. Risk of bias in individual studies was evaluated based on study design, methodology and missing data. RESULTS Of 31 studies identified through a systematic review, 26 studies were shared with WWARN and 21 studies with 2,787 patients were included. Recrudescence was associated with low day 7 lumefantrine concentrations (HR 1.59 (95% CI 1.36 to 1.85) per halving of day 7 concentrations) and high baseline parasitemia (HR 1.87 (95% CI 1.22 to 2.87) per 10-fold increase). Adjusted for mg/kg dose, day 7 concentrations were lowest in very young children (<3 years), among whom underweight-for-age children had 23% (95% CI -1 to 41%) lower concentrations than adequately nourished children of the same age and 53% (95% CI 37 to 65%) lower concentrations than adults. Day 7 lumefantrine concentrations were 44% (95% CI 38 to 49%) lower following unsupervised treatment. The highest risk of recrudescence was observed in areas of emerging artemisinin resistance and very low transmission intensity. For all other populations studied, day 7 concentrations ≥200 ng/ml were associated with >98% cure rates (if parasitemia <135,000/μL). CONCLUSIONS Current artemether-lumefantrine dosing recommendations achieve day 7 lumefantrine concentrations ≥200 ng/ml and high cure rates in most uncomplicated malaria patients. Three groups are at increased risk of treatment failure: very young children (particularly those underweight-for-age); patients with high parasitemias; and patients in very low transmission intensity areas with emerging parasite resistance. In these groups, adherence and treatment response should be monitored closely. Higher, more frequent, or prolonged dosage regimens should now be evaluated in very young children, particularly if malnourished, and in patients with hyperparasitemia.
Collapse
|
46
|
Dahal P, d'Alessandro U, Dorsey G, Guerin PJ, Nsanzabana C, Price RN, Sibley CH, Stepniewska K, Talisuna AO. Clinical determinants of early parasitological response to ACTs in African patients with uncomplicated falciparum malaria: a literature review and meta-analysis of individual patient data. BMC Med 2015; 13:212. [PMID: 26343145 PMCID: PMC4561425 DOI: 10.1186/s12916-015-0445-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Artemisinin-resistant Plasmodium falciparum has emerged in the Greater Mekong sub-region and poses a major global public health threat. Slow parasite clearance is a key clinical manifestation of reduced susceptibility to artemisinin. This study was designed to establish the baseline values for clearance in patients from Sub-Saharan African countries with uncomplicated malaria treated with artemisinin-based combination therapies (ACTs). METHODS A literature review in PubMed was conducted in March 2013 to identify all prospective clinical trials (uncontrolled trials, controlled trials and randomized controlled trials), including ACTs conducted in Sub-Saharan Africa, between 1960 and 2012. Individual patient data from these studies were shared with the WorldWide Antimalarial Resistance Network (WWARN) and pooled using an a priori statistical analytical plan. Factors affecting early parasitological response were investigated using logistic regression with study sites fitted as a random effect. The risk of bias in included studies was evaluated based on study design, methodology and missing data. RESULTS In total, 29,493 patients from 84 clinical trials were included in the analysis, treated with artemether-lumefantrine (n = 13,664), artesunate-amodiaquine (n = 11,337) and dihydroartemisinin-piperaquine (n = 4,492). The overall parasite clearance rate was rapid. The parasite positivity rate (PPR) decreased from 59.7 % (95 % CI: 54.5-64.9) on day 1 to 6.7 % (95 % CI: 4.8-8.7) on day 2 and 0.9 % (95 % CI: 0.5-1.2) on day 3. The 95th percentile of observed day 3 PPR was 5.3 %. Independent risk factors predictive of day 3 positivity were: high baseline parasitaemia (adjusted odds ratio (AOR) = 1.16 (95 % CI: 1.08-1.25); per 2-fold increase in parasite density, P <0.001); fever (>37.5 °C) (AOR = 1.50 (95 % CI: 1.06-2.13), P = 0.022); severe anaemia (AOR = 2.04 (95 % CI: 1.21-3.44), P = 0.008); areas of low/moderate transmission setting (AOR = 2.71 (95 % CI: 1.38-5.36), P = 0.004); and treatment with the loose formulation of artesunate-amodiaquine (AOR = 2.27 (95 % CI: 1.14-4.51), P = 0.020, compared to dihydroartemisinin-piperaquine). CONCLUSIONS The three ACTs assessed in this analysis continue to achieve rapid early parasitological clearance across the sites assessed in Sub-Saharan Africa. A threshold of 5 % day 3 parasite positivity from a minimum sample size of 50 patients provides a more sensitive benchmark in Sub-Saharan Africa compared to the current recommended threshold of 10 % to trigger further investigation of artemisinin susceptibility.
Collapse
Affiliation(s)
| | - Prabin Dahal
- WorldWide Antimalarial Resistance Network (WWARN); Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sondo P, Derra K, Diallo-Nakanabo S, Tarnagda Z, Zampa O, Kazienga A, Valea I, Sorgho H, Owusu-Dabo E, Ouedraogo JB, Guiguemde TR, Tinto H. Effectiveness and safety of artemether-lumefantrine versus artesunate-amodiaquine for unsupervised treatment of uncomplicated falciparum malaria in patients of all age groups in Nanoro, Burkina Faso: a randomized open label trial. Malar J 2015; 14:325. [PMID: 26289949 PMCID: PMC4545998 DOI: 10.1186/s12936-015-0843-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 08/10/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Several studies have reported high efficacy and safety of artemisinin-based combination therapy (ACT) mostly under strict supervision of drug intake and limited to children less than 5 years of age. Patients over 5 years of age are usually not involved in such studies. Thus, the findings do not fully reflect the reality in the field. This study aimed to assess the effectiveness and safety of ACT in routine treatment of uncomplicated malaria among patients of all age groups in Nanoro, Burkina Faso. METHODS A randomized open label trial comparing artesunate-amodiaquine (ASAQ) and artemether-lumefantrine (AL) was carried out from September 2010 to October 2012 at two primary health centres (Nanoro and Nazoanga) of Nanoro health district. A total of 680 patients were randomized to receive either ASAQ or AL without any distinction by age. Drug intake was not supervised as pertains in routine practice in the field. Patients or their parents/guardians were advised on the time and mode of administration for the 3 days treatment unobserved at home. Follow-up visits were performed on days 3, 7, 14, 21, and 28 to evaluate clinical and parasitological resolution of their malaria episode as well as adverse events. PCR genotyping of merozoite surface proteins 1 and 2 (msp-1, msp-2) was used to differentiate recrudescence and new infection. RESULTS By day 28, the PCR corrected adequate clinical and parasitological response was 84.1 and 77.8 % respectively for ASAQ and AL. The cure rate was higher in older patients than in children under 5 years old. The risk of re-infection by day 28 was higher in AL treated patients compared with those receiving ASAQ (p < 0.00001). Both AL and ASAQ treatments were well tolerated. CONCLUSION This study shows a lowering of the efficacy when drug intake is not directly supervised. This is worrying as both rates are lower than the critical threshold of 90 % required by the WHO to recommend the use of an anti-malarial drug in a treatment policy. TRIAL REGISTRATION NCT01232530.
Collapse
Affiliation(s)
- Paul Sondo
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | - Karim Derra
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | - Seydou Diallo-Nakanabo
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | - Zekiba Tarnagda
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | - Odile Zampa
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso.
| | - Adama Kazienga
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | - Innocent Valea
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso.
| | - Hermann Sorgho
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | - Ellis Owusu-Dabo
- Kumasi Center for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana.
| | - Jean-Bosco Ouedraogo
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
| | | | - Halidou Tinto
- IRSS, Clinical Research Unit of Nanoro (CRUN), CMA Saint Camille of Nanoro, BP 218 Ouagadougou CMS 11, Nanoro, Burkina Faso.
- Centre Muraz of Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso.
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Vreden SGS, Jitan JK, Bansie RD, Adhin MR. Evidence of an increased incidence of day 3 parasitaemia in Suriname: an indicator of the emerging resistance of Plasmodium falciparum to artemether. Mem Inst Oswaldo Cruz 2015; 108:968-73. [PMID: 24402149 PMCID: PMC4005544 DOI: 10.1590/0074-0276130167] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 11/11/2013] [Indexed: 11/25/2022] Open
Abstract
The emerging resistance to artemisinin derivatives that has been reported in
South-East Asia led us to assess the efficacy of artemether-lumefantrine as the first
line therapy for uncomplicated Plasmodium falciparum infections in
Suriname. This drug assessment was performed according to the recommendations of the
World Health Organization in 2011. The decreasing number of malaria cases in
Suriname, which are currently limited to migrating populations and gold miners,
precludes any conclusions on artemether efficacy because adequate numbers of patients
with 28-day follow-up data are difficult to obtain. Therefore, a comparison of day 3
parasitaemia in a 2011 study and in a 2005/2006 study was used to detect the
emergence of resistance to artemether. The prevalence of day 3 parasitaemia was
assessed in a study in 2011 and was compared to that in a study in 2005/2006. The
same protocol was used in both studies and artemether-lumefantrine was the study
drug. Of 48 evaluable patients in 2011, 15 (31%) still had parasitaemia on day 3
compared to one (2%) out of 45 evaluable patients in 2005/2006. Overall, 11 evaluable
patients in the 2011 study who were followed up until day 28 had negative slides and
similar findings were obtained in all 38 evaluable patients in the 2005/2006 study.
The significantly increased incidence of parasite persistence on day 3 may be an
indication of emerging resistance to artemether.
Collapse
Affiliation(s)
| | | | - Rakesh D Bansie
- Department of Internal Medicine, Academic Hospital Paramaribo, Suriname, Paramaribo
| | - Malti R Adhin
- Department of Biochemistry, Anton de Kom University of Suriname, Suriname, Paramaribo
| |
Collapse
|