1
|
Juliano JJ, Giesbrecht DJ, Simkin A, Fola AA, Lyimo BM, Pereus D, Bakari C, Madebe RA, Seth MD, Mandara CI, Popkin-Hall ZR, Moshi R, Mbwambo RB, Niaré K, MacInnis B, Francis F, Mbwambo D, Garimo I, Chacky F, Aaron S, Lusasi A, Molteni F, Njau RJA, Nhiga SL, Mohamed A, Bailey JA, Ishengoma DS. Prevalence of mutations associated with artemisinin partial resistance and sulfadoxine-pyrimethamine resistance in 13 regions in Tanzania in 2021: a cross-sectional survey. THE LANCET. MICROBE 2024; 5:100920. [PMID: 39159629 PMCID: PMC11464622 DOI: 10.1016/s2666-5247(24)00160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND The emergence of the artemisinin partial resistance (ART-R) mutation in the Plasmodium falciparum kelch13 gene (k13), Arg561His, in Rwanda and the regional presence of polymorphisms affecting sulfadoxine-pyrimethamine have raised concern in neighbouring Tanzania. The goal of this study was to assess the status of antimalarial resistance in Tanzania, with a focus on the border with Rwanda, to understand the distribution of the Arg561His mutation, partner drug resistance, and resistance to chemoprevention drugs. METHODS In this cross-sectional survey, capillary dried blood spots were collected from malaria positive asymptomatic individuals in the community and symptomatic individuals in health facilities aged 6 months and older, in 13 regions of mainland Tanzania from Jan 31 to June 26, 2021. Exclusion criteria included residence of the areas other than the target sites, presenting to the health facility for care and treatment of conditions other than malaria, and not providing informed consent. Samples were assessed for antimalarial resistance polymorphisms and genetic relatedness using molecular inversion probes targeting P falciparum and short-read whole-genome sequencing. The primary outcome was the prevalence of molecular markers of antimalarial resistance at the region level, as well as at the district level in Kagera, a region in the northwest of the country at the border with Rwanda. FINDINGS 6855 (88·1%) of 7782 capillary dried blood spot samples collected were successfully genotyped. The overall prevalence of k13 Arg561His in Kagera was 7·7% (90% CI 6·0-9·4; 50 of 649), with the highest prevalence in the districts near the Rwandan border (22·8% [31 of 136] in Karagwe, 14·4% [17 of 118]) in Kyerwa, and 1·4% [two of 144] in Ngara). k13 Arg561His was uncommon in the other regions. Haplotype analysis suggested that some of these parasites are related to isolates collected in Rwanda in 2015, supporting regional spread of Arg561His. However, a novel k13 Arg561His haplotype was observed, potentially indicating a second origin in the region. Other validated k13 resistance polymorphisms (one Arg622Ile and two Ala675Val isolates) were also identified. A region of prevalent dihydrofolate reductase Ile164Leu mutation, associated with sulfadoxine-pyrimethamine resistance, was also identified in Kagera (15·2% [12·6-17·8%]; 80 of 526). The mutant crt Lys76Thr mutation, associated with chloroquine and amodiaquine resistance, was uncommon, occurring only in 75 of 2861 genotyped isolates, whereases the wild-type mdr1 Asn86Tyr allele, associated with reduced sensitivity to lumefantrine, was found in 99·7% (3819 of 3830) of samples countrywide. INTERPRETATION These findings show that the k13 Arg561His mutation is common in northwest Tanzania and that multiple emergences of ART-R, similar as to what was seen in southeast Asia, have occurred. Mutations associated with high levels of sulfadoxine-pyrimethamine resistance are common. These results raise concerns about the long-term efficacy of artemisinin and chemoprevention antimalarials in the region. Understanding how multiple emergences interact with drivers of regional spread is essential for combating ART-R in Africa. FUNDING This study was funded by the Bill & Melinda Gates Foundation and the National Institutes of Health.
Collapse
Affiliation(s)
- Jonathan J Juliano
- Division of Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum of Genetics and Molecular Biology, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Institute for Global Health and Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David J Giesbrecht
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Alfred Simkin
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Abebe A Fola
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Beatus M Lyimo
- National Institute for Medical Research, Dar es Salaam, Tanzania; School of Life Sciences and Bioengineering, Nelson Mandela African Institute of Science and Technology, Arusha, Tanzania
| | - Dativa Pereus
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Catherine Bakari
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Rashid A Madebe
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Misago D Seth
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Celine I Mandara
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Zachary R Popkin-Hall
- Institute for Global Health and Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ramadhan Moshi
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Ruth B Mbwambo
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Karamoko Niaré
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Bronwyn MacInnis
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA; Infectious Disease and Microbiome Program, Broad Institute, Boston, MA, USA
| | - Filbert Francis
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | | | - Issa Garimo
- National Malaria Control Programme, Dodoma, Tanzania
| | - Frank Chacky
- National Malaria Control Programme, Dodoma, Tanzania
| | | | | | | | - Ritha J A Njau
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Ally Mohamed
- National Malaria Control Programme, Dodoma, Tanzania
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Deus S Ishengoma
- National Institute for Medical Research, Dar es Salaam, Tanzania; Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA; Department of Biochemistry, Kampala International University in Tanzania, Dar es Salaam, Tanzania.
| |
Collapse
|
2
|
Ouédraogo A, Pouplin JNN, Mukaka M, Kaendiao T, Ruecker A, Millet P, Vallet T, Ruiz F, Sirima SB, Taylor WR. Anti-infectivity efficacy and pharmacokinetics of WHO recommended single low-dose primaquine in children with acute Plasmodium falciparum in Burkina Faso: study protocol. Trials 2024; 25:583. [PMID: 39227956 PMCID: PMC11373093 DOI: 10.1186/s13063-024-08428-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Primaquine (PQ) has activity against mature P. falciparum gametocytes and proven transmission blocking efficacy (TBE) between humans and mosquitoes. WHO formerly recommended a single transmission blocking dose of 0.75 mg/kg but this was little used. Then in 2012, faced with the emergence of artemisinin-resistant P. falciparum (ARPf) in SE Asia, the WHO recommended a lower dose of 0.25 mg/kg to be added to artemisinin-based combination therapy in falciparum-infected patients in low transmission areas. This dose was considered safe in glucose-6-phosphate dehydrogenase deficiency (G6PDd) and not requiring G6PD testing. Subsequent single low-dose primaquine (SLDPQ) studies have demonstrated safety in different G6PD variants. Dosing remains challenging in children under the age of 5 because of the paucity of PQ pharmacokinetic (PK) data. We plan to assess the anti-infectivity efficacy of SLDPQ using an allometrically scaled, weight-based regimen, with a target dose of 0.25 mg/kg, in children with acute uncomplicated falciparum malaria. METHODS This study is an open label, randomised 1:1, phase IIb study to assess TBE, tolerability, pharmacokinetics and acceptability of artesunate pyronaridine (ASPYR) administered alone or combined with SLDPQ in 56 Burkinabe children aged ≥ 6 months- < 5 years, with uncomplicated P. falciparum and a haemoglobin (Hb) concentration of ≥ 5 g/dL. We will assess TBE, using direct membrane feeding assays (DMFA), and further investigate PQ pharmacokinetics, adverse events, Hb dynamics, G6PD, sickle cells, thalassaemia and cytochrome 2D6 (CYP2D6) status, acceptability of flavoured PQ [CAST-ClinSearch Acceptability Score Test®], and the population's knowledge, attitude and practices on malaria. EXPECTED RESULTS AND DISCUSSION We expect children to accept tablets, confirm the TBE and gametocytocidal effects of SLDPQ and then construct a PK infectivity model (including age, sex, baseline Hb, G6PD and CYP2D6 status) to define the dose response TBE relationship that may lead to fine tuning our SLDPQ regimen. Our study will complement others that have examined factors associated with Hb dynamics and PQ PK. It will provide much needed, high-quality evidence of SLDPQ in sick African children and provide reassurance that SLDPQ should be used as a strategy against emerging ARPf in Africa. TRIAL REGISTRATION ISRCTN16297951. Registered on September 26, 2021.
Collapse
Affiliation(s)
- Alphonse Ouédraogo
- Groupe de Recherche Action en Santé (GRAS), 06 BP 10248, Ouagadougou 06, Burkina Faso.
| | | | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thoopmanee Kaendiao
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
| | - Andrea Ruecker
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pascal Millet
- ReMeD, 21bis Avenue du Commandant de L'Herminier, Saint-Nazaire, 44 600, France
| | - Thibaut Vallet
- ClinSearch, 110 Avenue Pierre Brossolette, Malakoff, 92240, France
| | - Fabrice Ruiz
- ClinSearch, 110 Avenue Pierre Brossolette, Malakoff, 92240, France
| | - Sodiomon B Sirima
- Groupe de Recherche Action en Santé (GRAS), 06 BP 10248, Ouagadougou 06, Burkina Faso
| | - Walter R Taylor
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Von Wowern F, Makenga G, Wellmann Thomsen S, Wellmann Thomsen L, Filtenborg Hocke E, Baraka V, Opot BH, Minja DTR, Lusingu JPA, Van-Geertruyden JP, Hansson H, Alifrangis M. Lack of selection of antimalarial drug resistance markers after intermittent preventive treatment of schoolchildren (IPTsc) against malaria in northeastern Tanzania. Int J Infect Dis 2024; 146:107102. [PMID: 38876161 DOI: 10.1016/j.ijid.2024.107102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/07/2024] [Accepted: 05/16/2024] [Indexed: 06/16/2024] Open
Abstract
OBJECTIVE Intermittent Preventive Treatment of schoolchildren (IPTsc) is recommended by WHO as a strategy to protect against malaria; to explore whether IPTsc with dihydroartemisinin-piperaquine (DP) or artesunate-amodiaquine (ASAQ) cause a selection of molecular markers in Plasmodium falciparum genes associated with resistance in children in seven schools in Tanga region, Tanzania. METHODS SNPs in P. falciparum genes Pfmdr1, Pfexo, Pfkelch13, and Pfcrt and copy number variations in Pfplasmepsin-2 and Pfmdr1 were assessed in samples collected at 12 months (visit 4, n=74) and 20 months (visit 6, n=364) after initiation of IPTsc and compared with the baseline prevalence (n=379). RESULTS The prevalence of Pfmdr1 N86 and Pfexo 415G was >99% and 0%, respectively without any temporal differences observed. The prevalence of Pfmdr1 184F changed significantly from baseline (52.2%) to visit 6 (64.6%) (χ2=6.11, P=0.013), but no differences were observed between the treatment arms (χ2=0.05, P=0.98). Finally, only minor differences in the amplification of Pfmdr1 were observed; from 10.2% at baseline to 16.7% at visit 6 (χ2=0.98, P=0.32). CONCLUSIONS The IPTsc strategy does not seem to pose a risk for the selection of markers associated with DP or ASAQ resistance. Continuously and timely surveillance of markers of antimalarial drug resistance is recommended.
Collapse
Affiliation(s)
- Frederik Von Wowern
- Centre for translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Geofrey Makenga
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania; Global Health Institute, University of Antwerp, Antwerp, Belgium
| | - Sarah Wellmann Thomsen
- Centre for translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Louise Wellmann Thomsen
- Centre for translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emma Filtenborg Hocke
- Centre for translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vito Baraka
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Benjamin H Opot
- Kenya Medical Research Institute (KEMRI)/Walter Reed Project, United States Army Medical Research Directorate-Africa (USAMRD-A), Kisumu, Kenya
| | - Daniel T R Minja
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - John P A Lusingu
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | | | - Helle Hansson
- Centre for translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michael Alifrangis
- Centre for translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
4
|
Brown N, da Silva C, Webb C, Matias D, Dias B, Cancio B, Silva M, Viegas R, Salvador C, Chivale N, Luis S, Arnaldo P, Zulawinska J, Moore CC, Nogueira F, Guler JL. Antimalarial resistance risk in Mozambique detected by a novel quadruplex droplet digital PCR assay. Antimicrob Agents Chemother 2024; 68:e0034624. [PMID: 38771031 PMCID: PMC11232384 DOI: 10.1128/aac.00346-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
While the Plasmodium falciparum malaria parasite continues to cause severe disease globally, Mozambique is disproportionally represented in malaria case totals. Acquisition of copy number variations (CNVs) in the parasite genome contributes to antimalarial drug resistance through overexpression of drug targets. Of interest, piperaquine resistance is associated with plasmepsin 2 and 3 CNVs (pfpmp2 and pfpmp3, respectively), while CNVs in the multidrug efflux pump, multidrug resistance-1 (pfmdr1), increase resistance to amodiaquine and lumefantrine. These antimalarials are partner drugs in artemisinin combination therapies (ACTs) and therefore, CNV detection with accurate and efficient tools is necessary to track ACT resistance risk. Here, we evaluated ~300 clinically derived samples collected from three sites in Mozambique for resistance-associated CNVs. We developed a novel, medium-throughput, quadruplex droplet digital PCR (ddPCR) assay to simultaneously quantify the copy number of pfpmp3, pfpmp2, and pfmdr1 loci in these clinical samples. By using DNA from laboratory parasite lines, we show that this nanodroplet-based method is capable of detecting picogram levels of parasite DNA, which facilitates its application for low yield and human host-contaminated clinical surveillance samples. Following ddPCR and the application of quality control standards, we detected CNVs in 13 of 229 high-quality samples (prevalence of 5.7%). Overall, our study revealed a low number of resistance CNVs present in the parasite population across all three collection sites, including various combinations of pfmdr1, pfpmp2, and pfpmp3 CNVs. The potential for future ACT resistance across Mozambique emphasizes the need for continued molecular surveillance across the region.
Collapse
Affiliation(s)
- Noah Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Clemente da Silva
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | - Caroline Webb
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniela Matias
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | - Brigite Dias
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | - Beatriz Cancio
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | - Miguel Silva
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | - Ruben Viegas
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | | | - Nordino Chivale
- Instituto Nacional de Saúde, Maputo (INS), Maputo, Mozambique
| | - Sonia Luis
- Hospital Provincial de Matola, Matola, Mozambique
| | - Paulo Arnaldo
- Instituto Nacional de Saúde, Maputo (INS), Maputo, Mozambique
| | - Julia Zulawinska
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Christopher C. Moore
- Division of Infectious Disease and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Fatima Nogueira
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Lisbon, Portugal
| | - Jennifer L. Guler
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Division of Infectious Disease and International Health, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Sima-Biyang YV, Ontoua SS, Longo-Pendy NM, Mbou-Boutambe C, Makouloutou-Nzassi P, Moussadji CK, Lekana-Douki JB, Boundenga L. Epidemiology of malaria in Gabon: A systematic review and meta-analysis from 1980 to 2023. J Infect Public Health 2024; 17:102459. [PMID: 38870682 DOI: 10.1016/j.jiph.2024.05.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024] Open
Abstract
The objective of this were conducted to elucidate spatiotemporal variations in malaria epidemiology in Gabon since 1980. For that, five databases, were used to collect and identify all studies published between 1980 and 2023 on malaria prevalence, antimalarial drug resistance, markers of antimalarial drug resistance and insecticide resistance marker. The findings suggest that Gabon continues to face malaria as an urgent public health problem, with persistently high prevalence rates. Markers of resistance to CQ persist despite its withdrawal, and markers of resistance to SP have emerged with a high frequency, reaching 100 %, while ACTs remain effective. Also, recent studies have identified markers of resistance to the insecticides Kdr-w and Kdr-e at frequencies ranging from 25 % to 100 %. Ace1R mutation was reported with a frequency of 0.4 %. In conclusion, the efficacy of ACTs remains above the threshold recommended by the WHO. Organo-phosphates and carbamates could provide an alternative for vector control.
Collapse
Affiliation(s)
- Yann Vital Sima-Biyang
- Unit of Research in Ecology of Health (URES), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon; Central African Regional Doctoral School in Tropical Infectiology (EDR), BP 876 Franceville, Gabon
| | - Steede Seinnat Ontoua
- Central African Regional Doctoral School in Tropical Infectiology (EDR), BP 876 Franceville, Gabon; Unit of Evolution, Epidemiology and Parasite Resistance (UNEEREP), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon
| | - Neil Michel Longo-Pendy
- Unit of Research in Ecology of Health (URES), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon
| | - Clark Mbou-Boutambe
- Unit of Research in Ecology of Health (URES), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon; Central African Regional Doctoral School in Tropical Infectiology (EDR), BP 876 Franceville, Gabon
| | - Patrice Makouloutou-Nzassi
- Unit of Research in Ecology of Health (URES), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon; Department of Animal Biology and Ecology, Tropical Ecology Research Institute (IRET/CENAREST), Libreville BP 13354, Gabon
| | - Cyr Kinga Moussadji
- Primatology Center, Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon
| | - Jean-Bernard Lekana-Douki
- Unit of Evolution, Epidemiology and Parasite Resistance (UNEEREP), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon; Department of Parasitology-Mycology-Tropical Medicine, University of Health Sciences, Faculty of Medicine, BP 4009 Libreville, Gabon
| | - Larson Boundenga
- Unit of Research in Ecology of Health (URES), Franceville Interdisciplinary Center for Medical Research (CIRMF), BP 769 Franceville, Gabon; Department of Anthropology, Durham University, South Road, Durham DH1 3LE, UK.
| |
Collapse
|
6
|
Nguyen Ngoc Pouplin J, Kaendiao T, Rahimi BA, Soni M, Basopia H, Shah D, Patil J, Dholakia V, Suthar Y, Tarning J, Mukaka M, Taylor WR. Bioequivalence of a new coated 15 mg primaquine formulation for malaria elimination. Malar J 2024; 23:176. [PMID: 38840151 PMCID: PMC11155120 DOI: 10.1186/s12936-024-04947-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/12/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND With only one 15 mg primaquine tablet registered by a stringent regulatory authority and marketed, more quality-assured primaquine is needed to meet the demands of malaria elimination. METHODS A classic, two sequence, crossover study, with a 10-day wash out period, of 15 mg of IPCA-produced test primaquine tablets and 15 mg of Sanofi reference primaquine tablets was conducted. Healthy volunteers, aged 18-45 years, without glucose-6-phosphate dehydrogenase deficiency, a baseline haemoglobin ≥ 11 g/dL, creatinine clearance ≥ 70 mL/min/1.73 ms, and body mass index of 18.5-30 kg/m2 were randomized to either test or reference primaquine, administered on an empty stomach with 240 mL of water. Plasma primaquine and carboxyprimaquine concentrations were measured at baseline, then 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 1.75, 2.0, 2.333, 2.667, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 8.0, 10.0, 12.0, 16.0, 24.0, 36.0, 48.0 and 72.0 h by liquid chromatography coupled to tandem mass spectrometry. Primaquine pharmacokinetic profiles were evaluated by non-compartmental analysis and bioequivalence concluded if the 90% confidence intervals (CI) of geometric mean (GM) ratios of test vs. reference formulation for the peak concentrations (Cmax) and area under the drug concentration-time (AUC0-t) were within 80.00 to 125.00%. RESULTS 47 of 50 volunteers, median age 33 years, completed both dosing rounds and were included in the bioequivalence analysis. For primaquine, GM Cmax values for test and reference formulations were 62.12 vs. 59.63 ng/mL, resulting in a GM ratio (90% CI) of 104.17% (96.92-111.96%); the corresponding GM AUC0-t values were 596.56 vs. 564.09 ngxh/mL, for a GM ratio of 105.76% (99.76-112.08%). Intra-subject coefficient of variation was 20.99% for Cmax and 16.83% for AUC0-t. Median clearances and volumes of distribution were similar between the test and reference products: 24.6 vs. 25.2 L/h, 189.4 vs. 191.0 L, whilst the median half-lives were the same, 5.2 h. CONCLUSION IPCA primaquine was bioequivalent to the Sanofi primaquine. This opens the door to prequalification, registration in malaria endemic countries, and programmatic use for malaria elimination. Trial registration The trial registration reference is ISRCTN 54640699.
Collapse
Affiliation(s)
- Julie Nguyen Ngoc Pouplin
- Réseau Médicaments et Développement, 21Bis Avenue du Commandant l'Herminier, 44600, Saint-Nazaire, France.
| | - Thoopmanee Kaendiao
- Mahidol Oxford Tropical Medicine Clinical Research Unit, Mahidol University, Bangkok, Thailand
| | - Bilal Ahmad Rahimi
- Department of Paediatrics, Faculty of Medicine, Kandahar University, Kandahar, Afghanistan
| | - Mayur Soni
- Cliantha Research Limited, Cliantha Corporate, Ahmedabad, Gujarat, India
| | - Hensi Basopia
- Cliantha Research Limited, Cliantha Corporate, Ahmedabad, Gujarat, India
| | - Darshana Shah
- Cliantha Research Limited, Cliantha Corporate, Ahmedabad, Gujarat, India
| | - Jitendra Patil
- Cliantha Research Limited, Cliantha Corporate, Ahmedabad, Gujarat, India
| | - Vyom Dholakia
- Cliantha Research Limited, Cliantha Corporate, Ahmedabad, Gujarat, India
| | - Yash Suthar
- Cliantha Research Limited, Cliantha Corporate, Ahmedabad, Gujarat, India
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Clinical Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Clinical Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Walter R Taylor
- Mahidol Oxford Tropical Medicine Clinical Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Rosenthal PJ, Asua V, Conrad MD. Emergence, transmission dynamics and mechanisms of artemisinin partial resistance in malaria parasites in Africa. Nat Rev Microbiol 2024; 22:373-384. [PMID: 38321292 DOI: 10.1038/s41579-024-01008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 02/08/2024]
Abstract
Malaria, mostly due to Plasmodium falciparum infection in Africa, remains one of the most important infectious diseases in the world. Standard treatment for uncomplicated P. falciparum malaria is artemisinin-based combination therapy (ACT), which includes a rapid-acting artemisinin derivative plus a longer-acting partner drug, and standard therapy for severe P. falciparum malaria is intravenous artesunate. The efficacy of artemisinins and ACT has been threatened by the emergence of artemisinin partial resistance in Southeast Asia, mediated principally by mutations in the P. falciparum Kelch 13 (K13) protein. High ACT treatment failure rates have occurred when resistance to partner drugs is also seen. Recently, artemisinin partial resistance has emerged in Rwanda, Uganda and the Horn of Africa, with independent emergences of different K13 mutants in each region. In this Review, we summarize our current knowledge of artemisinin partial resistance and focus on the emergence of resistance in Africa, including its epidemiology, transmission dynamics and mechanisms. At present, the clinical impact of emerging resistance in Africa is unclear and most available evidence suggests that the efficacies of leading ACTs remain excellent, but there is an urgent need to better appreciate the extent of the problem and its consequences for the treatment and control of malaria.
Collapse
Affiliation(s)
| | - Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda
- University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
8
|
Amador LA, Colón-Lorenzo EE, Rodríguez AD, Serrano AE. Probing the Antiplasmodial Properties of Plakortinic Acids C and D: An Uncommon Pair of Marine Peroxide-Polyketides Isolated from a Two-Sponge Association of Plakortis symbiotica and Xetospongia deweerdtae Collected near Puerto Rico. Life (Basel) 2024; 14:684. [PMID: 38929667 PMCID: PMC11204963 DOI: 10.3390/life14060684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Plakortinic acids C (1) and D (2), an unseparable pair of endoperoxide polyketides isolated and purified from the symbiotic association of Caribbean Sea sponges Plakortis symbiotica-Xestospongia deweerdtae, underwent in vitro evaluation for antiplasmodial activity against the malaria parasite Plasmodium berghei using a drug luminescence assay. Initial screening at 10 µM revealed 50% in vitro parasite growth inhibition. The title compounds displayed antiplasmodial activity with an EC50 of 5.3 µM toward P. berghei parasites. The lytic activity against erythrocytes was assessed through an erythrocyte cell lysis assay, which showed non-lytic activity at lower concentrations ranging from 1.95 to 3.91 µM. The antiplasmodial activity and the absence of hemolytic activity support the potential of plakortinic acids C (1) and D (2) as promising lead compounds. Moreover, drug-likeness (ADMET) properties assessed through the pkCSM server predicted high intestinal absorption, hepatic metabolism, and volume of distribution, indicating favorable pharmacokinetic profiles for oral administration. These findings suggest the potential suitability of these metabolites for further investigations of antiplasmodial activity in multiple parasitic stages in the mosquito and Plasmodium falciparum. Notably, this study represents the first report of a marine natural product exhibiting the unique 7,8-dioxatricyclo[4.2.2.02,5]dec-9-ene motif being evaluated against malaria.
Collapse
Affiliation(s)
- Luis A. Amador
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan 00926, Puerto Rico;
| | - Emilee E. Colón-Lorenzo
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan 00921, Puerto Rico;
| | - Abimael D. Rodríguez
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan 00926, Puerto Rico;
| | - Adelfa E. Serrano
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan 00921, Puerto Rico;
| |
Collapse
|
9
|
Ngasala BE, Chiduo MG, Mmbando BP, Francis FT, Bushukatale S, Makene T, Mandara CI, Ishengoma DS, Kamugisha E, Ahmed M, Mahende MK, Kavishe RA, Muro F, Molteni F, Reaves E, Kitojo C, Greer G, Nyinondi S, Kabula B, Lalji S, Chacky F, Njau RJ, Warsame M, Mohamed A. Efficacy and safety of artemether-lumefantrine for the treatment of uncomplicated falciparum malaria in mainland Tanzania, 2019. Malar J 2024; 23:101. [PMID: 38594679 PMCID: PMC11005286 DOI: 10.1186/s12936-024-04931-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Artemisinin-based combination therapy (ACT) has been a major contributor to the substantial reductions in global malaria morbidity and mortality over the last decade. In Tanzania, artemether-lumefantrine (AL) was introduced as the first-line treatment for uncomplicated Plasmodium falciparum malaria in 2006. The World Health Organization (WHO) recommends regular assessment and monitoring of the efficacy of the first-line treatment, specifically considering that artemisinin resistance has been confirmed in the Greater Mekong sub-region. This study's main aim was to assess the efficacy and safety of AL for treating uncomplicated P. falciparum malaria in Tanzania. METHODS This was a single-arm prospective antimalarial drug efficacy trial conducted in four of the eight National Malaria Control Programme (NMCP) sentinel sites in 2019. The trial was carried out in outpatient health facilities in Karume-Mwanza region, Ipinda-Mbeya region, Simbo-Tabora region, and Nagaga-Mtwara region. Children aged six months to 10 years with microscopy confirmed uncomplicated P. falciparum malaria who met the inclusion criteria were recruited based on the WHO protocol. The children received AL (a 6-dose regimen of AL twice daily for three days). Clinical and parasitological parameters were monitored during follow-up over 28 days to evaluate drug efficacy. RESULTS A total of 628 children were screened for uncomplicated malaria, and 349 (55.6%) were enrolled between May and September 2019. Of the enrolled children, 343 (98.3%) completed the 28-day follow-up or attained the treatment outcomes. There were no early treatment failures; recurrent infections during follow-up were common at two sites (Karume 29.5%; Simbo 18.2%). PCR-corrected adequate clinical and parasitological response (ACPR) by survival analysis to AL on day 28 of follow-up varied from 97.7% at Karume to 100% at Ipinda and Nagaga sites. The commonly reported adverse events were cough, skin pallor, and abdominal pain. The drug was well tolerated, and no serious adverse event was reported. CONCLUSION This study showed that AL had adequate efficacy and safety for the treatment of uncomplicated falciparum malaria in Tanzania in 2019. The high recurrent infections were mainly due to new infections, highlighting the potential role of introducing alternative artemisinin-based combinations that offer improved post-treatment prophylaxis, such as artesunate-amodiaquine (ASAQ).
Collapse
Affiliation(s)
- Billy E Ngasala
- Department of Parasitology, School of Public Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania.
| | - Mercy G Chiduo
- National Institute for Medical Research, Tanga Research Centre, Tanga, Tanzania
| | - Bruno P Mmbando
- National Institute for Medical Research, Tanga Research Centre, Tanga, Tanzania
| | - Filbert T Francis
- National Institute for Medical Research, Tanga Research Centre, Tanga, Tanzania
| | - Samwel Bushukatale
- Department of Parasitology, School of Public Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Twilumba Makene
- Department of Parasitology, School of Public Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Celine I Mandara
- National Institute for Medical Research, Headquarters, Dar es Salaam, Tanzania
| | - Deus S Ishengoma
- National Institute for Medical Research, Headquarters, Dar es Salaam, Tanzania
| | - Erasmus Kamugisha
- Catholic University of Health and Allied Sciences, Bugando Medical Centre, Mwanza, Tanzania
| | - Maimuna Ahmed
- Catholic University of Health and Allied Sciences, Bugando Medical Centre, Mwanza, Tanzania
| | | | - Reginald A Kavishe
- Kilimanjaro Christian Medical Centre, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Florida Muro
- Kilimanjaro Christian Medical Centre, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | | | - Erik Reaves
- U.S. President's Malaria Initiative, U.S. Centers for Disease Control and Prevention, Dar es Salaam, Tanzania
| | - Chonge Kitojo
- U.S. President's Malaria Initiative, U.S. Agency for International Development, Dar es Salaam, Tanzania
| | - George Greer
- U.S. President's Malaria Initiative, U.S. Agency for International Development, Dar es Salaam, Tanzania
| | | | | | | | - Frank Chacky
- National Malaria Control Program, Dodoma, Tanzania
| | - Ritha J Njau
- Muhimbili University of Health and Allied Science, Dar es Salaam, Tanzania
| | | | - Ally Mohamed
- National Malaria Control Program, Dodoma, Tanzania
| |
Collapse
|
10
|
Dorkenoo AM, Warsame M, Ataba E, Hemou M, Yakpa K, Sossou E, Mitigmsagou M, Teou CD, Caspar E, Ma L, Djadou KE, Atcha-Oubou T, Rasmussen C, Menard D. Efficacy of artemether-lumefantrine and dihydroartemisinin-piperaquine and prevalence of molecular markers of anti-malarial drug resistance in children in Togo in 2021. Malar J 2024; 23:92. [PMID: 38570791 PMCID: PMC10988893 DOI: 10.1186/s12936-024-04922-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Artemether-lumefantrine (AL) and dihydroartemisinin-piperaquine (DP) are the currently recommended first- and second-line therapies for uncomplicated Plasmodium falciparum infections in Togo. This study assessed the efficacy of these combinations, the proportion of Day3-positive patients (D3 +), the proportion of molecular markers associated with P. falciparum resistance to anti-malarial drugs, and the variable performance of HRP2-based malaria rapid diagnostic tests (RDTs). METHODS A single arm prospective study evaluating the efficacy of AL and DP was conducted at two sites (Kouvé and Anié) from September 2021 to January 2022. Eligible children were enrolled, randomly assigned to treatment at each site and followed up for 42 days after treatment initiation. The primary endpoint was polymerase chain reaction (PCR) adjusted adequate clinical and parasitological response (ACPR). At day 0, samples were analysed for mutations in the Pfkelch13, Pfcrt, Pfmdr-1, dhfr, dhps, and deletions in the hrp2/hrp3 genes. RESULTS A total of 179 and 178 children were included in the AL and DP groups, respectively. After PCR correction, cure rates of patients treated with AL were 97.5% (91.4-99.7) at day 28 in Kouvé and 98.6% (92.4-100) in Anié, whereas 96.4% (CI 95%: 89.1-98.8) and 97.3% (CI 95%: 89.5-99.3) were observed at day 42 in Kouvé and Anié, respectively. The cure rates of patients treated with DP at day 42 were 98.9% (CI 95%: 92.1-99.8) in Kouvé and 100% in Anié. The proportion of patients with parasites on day 3 (D3 +) was 8.5% in AL and 2.6% in DP groups in Anié and 4.3% in AL and 2.1% DP groups in Kouvé. Of the 357 day 0 samples, 99.2% carried the Pfkelch13 wild-type allele. Two isolates carried nonsynonymous mutations not known to be associated with artemisinin partial resistance (ART-R) (A578S and A557S). Most samples carried the Pfcrt wild-type allele (97.2%). The most common Pfmdr-1 allele was the single mutant 184F (75.6%). Among dhfr/dhps mutations, the quintuple mutant haplotype N51I/C59R/S108N + 437G/540E, which is responsible for SP treatment failure in adults and children, was not detected. Single deletions in hrp2 and hrp3 genes were detected in 1/357 (0.3%) and 1/357 (0.3%), respectively. Dual hrp2/hrp3 deletions, which could affect the performances of HRP2-based RDTs, were not observed. CONCLUSION The results of this study confirm that the AL and DP treatments are highly effective. The absence of the validated Pfkelch13 mutants in the study areas suggests the absence of ART -R, although a significant proportion of D3 + cases were found. The absence of dhfr/dhps quintuple or sextuple mutants (quintuple + 581G) supports the continued use of SP for IPTp during pregnancy and in combination with amodiaquine for seasonal malaria chemoprevention. TRIAL REGISTRATION ACTRN12623000344695.
Collapse
Affiliation(s)
| | - Marian Warsame
- School of Public Health and Community Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Essoham Ataba
- Programme National de Lutte Contre le Paludisme, Lomé, Togo
| | - Manani Hemou
- Service de Pédiatrie, Centre Hospitalier Universitaire Campus, Lomé, Togo
| | - Kossi Yakpa
- Programme National de Lutte Contre le Paludisme, Lomé, Togo
| | - Efoe Sossou
- Service des Laboratoires, Centre Hospitalier Universitaire Sylvanus Olympio Lomé, Lomé, Togo
| | | | | | - Emmanuelle Caspar
- Institute of Parasitology and Tropical Diseases, Université de Strasbourg, UR7292 Dynamics of Host-Pathogen Interactions, 67000, Strasbourg, France
| | - Laurence Ma
- Biomics Platform, C2RT, Institut Pasteur, 75015, Paris, France
| | | | | | | | - Didier Menard
- Institute of Parasitology and Tropical Diseases, Université de Strasbourg, UR7292 Dynamics of Host-Pathogen Interactions, 67000, Strasbourg, France
- Malaria Genetics and Resistance Unit, Institut Pasteur, Université Paris Cité, INSERM U1201, 75015, Paris, France
- Malaria Parasite Biology and Vaccines, Institut Pasteur, Université Paris Cité, 75015, Paris, France
- Laboratory of Parasitology and Medical Mycology, CHU Strasbourg, 67000, Strasbourg, France
| |
Collapse
|
11
|
Ngasala B, Bushukatale S, Chiduo M, Makene T, Mkony L, Mohamed A, Molteni F, Chacky F, Njau RJA, Mwaiswelo R. Efficacy of artesunate-amodiaquine for treatment of uncomplicated Plasmodium falciparum malaria in mainland Tanzania. Malar J 2024; 23:90. [PMID: 38553737 PMCID: PMC10979577 DOI: 10.1186/s12936-024-04923-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Diversification of artemisinin-based combination therapy (ACT) is suggested as one of the strategies that can be used to contain artemisinin resistance. Artesunate-amodiaquine (ASAQ) is one of the artemisinin-based combinations that can be used in the diversification strategy as an alternative first-line treatment for uncomplicated malaria in mainland Tanzania. There is however limited data on the efficacy of ASAQ in mainland Tanzania. This study assessed the efficacy of ASAQ for treatment of uncomplicated Plasmodium falciparum malaria in selected sentinel sites for therapeutic efficacy studies in mainland Tanzania. METHODS Between December 2018 and March 2020, children aged between 6 months and 10 years, attending at Nagaga, Mkuzi, and Mlimba primary health facilities, and with suspected uncomplicated malaria infection were screened for eligibility to participate in the study. Malaria infection was screened using microscopy. Children with uncomplicated P. falciparum monoinfection and who fulfilled all other inclusion criteria, and had none of the exclusion criteria, according to the World Health Organization (WHO) guidelines, were treated with ASAQ. Follow-up visits were scheduled on days 0, 1, 2, 3, 7, 14, 21, and 28 or on any day of recurrent infection for clinical and laboratory assessment. Polymerase chain reaction (PCR)-corrected cure rate on day 28 was the primary outcome. RESULTS A total of 264 children, 88 in each of the three study sites (Mlimba, Mkuzi and Nagaga health facilities) were enrolled and treated with ASAQ. The ASAQ PCR-corrected cure rate was 100% at all the three study sites. None of the participants had early treatment failure or late clinical failure. Furthermore, none of the participants had a serious adverse event. CONCLUSION ASAQ was highly efficacious for the treatment of uncomplicated P. falciparum malaria in mainland Tanzania, therefore, it can be deployed as an alternative first-line treatment for uncomplicated malaria as part of diversification strategy to contain the spread of partial artemisinin resistance in the country.
Collapse
Affiliation(s)
- Billy Ngasala
- Department of Medical Parasitology and Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O. Box 65011, Dar es Salaam, Tanzania
| | - Samwel Bushukatale
- Department of Medical Parasitology and Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O. Box 65011, Dar es Salaam, Tanzania
| | - Mercy Chiduo
- National Institute for Medical Research, Tanga Research Centre, P.O Box 5004, Tanga, Tanzania
| | - Twilumba Makene
- Department of Medical Parasitology and Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O. Box 65011, Dar es Salaam, Tanzania
| | - Lilian Mkony
- Department of Medical Parasitology and Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O. Box 65011, Dar es Salaam, Tanzania
| | - Ally Mohamed
- National Malaria Control Program (NMCP), Ministry of Health, P.O. Box 743, Dar Es Salaam, Tanzania
| | - Fablizio Molteni
- National Malaria Control Program (NMCP), Ministry of Health, P.O. Box 743, Dar Es Salaam, Tanzania
| | - Frank Chacky
- National Malaria Control Program (NMCP), Ministry of Health, P.O. Box 743, Dar Es Salaam, Tanzania
| | - Ritha J A Njau
- Department of Medical Parasitology and Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O. Box 65011, Dar es Salaam, Tanzania
| | - Richard Mwaiswelo
- Department of Microbiology, Immunology, and Parasitology, Faculty of Medicine, Hubert Kairuki Memorial University, P.O Box 65300, Dar es Salaam, Tanzania.
| |
Collapse
|
12
|
Bakari C, Mandara CI, Madebe RA, Seth MD, Ngasala B, Kamugisha E, Ahmed M, Francis F, Bushukatale S, Chiduo M, Makene T, Kabanywanyi AM, Mahende MK, Kavishe RA, Muro F, Mkude S, Mandike R, Molteni F, Chacky F, Bishanga DR, Njau RJA, Warsame M, Kabula B, Nyinondi SS, Lucchi NW, Talundzic E, Venkatesan M, Moriarty LF, Serbantez N, Kitojo C, Reaves EJ, Halsey ES, Mohamed A, Udhayakumar V, Ishengoma DS. Trends of Plasmodium falciparum molecular markers associated with resistance to artemisinins and reduced susceptibility to lumefantrine in Mainland Tanzania from 2016 to 2021. Malar J 2024; 23:71. [PMID: 38461239 PMCID: PMC10924419 DOI: 10.1186/s12936-024-04896-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Therapeutic efficacy studies (TESs) and detection of molecular markers of drug resistance are recommended by the World Health Organization (WHO) to monitor the efficacy of artemisinin-based combination therapy (ACT). This study assessed the trends of molecular markers of artemisinin resistance and/or reduced susceptibility to lumefantrine using samples collected in TES conducted in Mainland Tanzania from 2016 to 2021. METHODS A total of 2,015 samples were collected during TES of artemether-lumefantrine at eight sentinel sites (in Kigoma, Mbeya, Morogoro, Mtwara, Mwanza, Pwani, Tabora, and Tanga regions) between 2016 and 2021. Photo-induced electron transfer polymerase chain reaction (PET-PCR) was used to confirm presence of malaria parasites before capillary sequencing, which targeted two genes: Plasmodium falciparum kelch 13 propeller domain (k13) and P. falciparum multidrug resistance 1 (pfmdr1). RESULTS Sequencing success was ≥ 87.8%, and 1,724/1,769 (97.5%) k13 wild-type samples were detected. Thirty-seven (2.1%) samples had synonymous mutations and only eight (0.4%) had non-synonymous mutations in the k13 gene; seven of these were not validated by the WHO as molecular markers of resistance. One sample from Morogoro in 2020 had a k13 R622I mutation, which is a validated marker of artemisinin partial resistance. For pfmdr1, all except two samples carried N86 (wild-type), while mutations at Y184F increased from 33.9% in 2016 to about 60.5% in 2021, and only four samples (0.2%) had D1246Y mutations. pfmdr1 haplotypes were reported in 1,711 samples, with 985 (57.6%) NYD, 720 (42.1%) NFD, and six (0.4%) carrying minor haplotypes (three with NYY, 0.2%; YFD in two, 0.1%; and NFY in one sample, 0.1%). Between 2016 and 2021, NYD decreased from 66.1% to 45.2%, while NFD increased from 38.5% to 54.7%. CONCLUSION This is the first report of the R622I (k13 validated mutation) in Tanzania. N86 and D1246 were nearly fixed, while increases in Y184F mutations and NFD haplotype were observed between 2016 and 2021. Despite the reports of artemisinin partial resistance in Rwanda and Uganda, this study did not report any other validated mutations in these study sites in Tanzania apart from R622I suggesting that intensified surveillance is urgently needed to monitor trends of drug resistance markers and their impact on the performance of ACT.
Collapse
Affiliation(s)
- Catherine Bakari
- National Institute for Medical Research, Dar Es Salaam, Tanzania
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Celine I Mandara
- National Institute for Medical Research, Dar Es Salaam, Tanzania
| | - Rashid A Madebe
- National Institute for Medical Research, Dar Es Salaam, Tanzania
| | - Misago D Seth
- National Institute for Medical Research, Dar Es Salaam, Tanzania
| | - Billy Ngasala
- Department of Parasitology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Erasmus Kamugisha
- Catholic University of Health and Allied Sciences, Bugando Medical Centre, Mwanza, Tanzania
| | - Maimuna Ahmed
- Catholic University of Health and Allied Sciences, Bugando Medical Centre, Mwanza, Tanzania
| | - Filbert Francis
- National Institute for Medical Research, Tanga Research Centre, Tanga, Tanzania
| | - Samwel Bushukatale
- Department of Parasitology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Mercy Chiduo
- National Institute for Medical Research, Tanga Research Centre, Tanga, Tanzania
| | - Twilumba Makene
- Department of Parasitology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | | | - Muhidin K Mahende
- Ifakara Health Institute, Dar Es Salaam Office, Dar Es Salaam, Tanzania
| | | | - Florida Muro
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | | | - Fabrizio Molteni
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- National Malaria Control Program, Dodoma, Tanzania
| | - Frank Chacky
- National Malaria Control Program, Dodoma, Tanzania
| | - Dunstan R Bishanga
- Ifakara Health Institute, Dar Es Salaam Office, Dar Es Salaam, Tanzania
- Maternal and Child Survival Program, Jhpiego, Dar Es Salaam, Tanzania
- School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Ritha J A Njau
- Malariologist and Public Health Specialist, Dar Es Salaam, Tanzania
| | | | - Bilali Kabula
- PMI/Okoa Maisha Dhibiti Malaria, RTI International, Dar Es Salaam, Tanzania
- National Institute for Medical Research, Amani Research Centre, Muheza, Tanga, Tanzania
| | - Ssanyu S Nyinondi
- PMI/Okoa Maisha Dhibiti Malaria, RTI International, Dar Es Salaam, Tanzania
| | - Naomi W Lucchi
- Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
- Division of Global Health Protection, U.S. Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Eldin Talundzic
- Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Leah F Moriarty
- Malaria Branch, U.S. President's Malaria Initiative, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Naomi Serbantez
- U.S. President's Malaria Initiative, USAID, Dar Es Salaam, Tanzania
| | - Chonge Kitojo
- U.S. President's Malaria Initiative, USAID, Dar Es Salaam, Tanzania
| | - Erik J Reaves
- U.S. President's Malaria Initiative, US Centers for Disease Control and Prevention, Dar Es Salaam, Tanzania
| | - Eric S Halsey
- Malaria Branch, U.S. President's Malaria Initiative, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ally Mohamed
- National Malaria Control Program, Dodoma, Tanzania
| | - Venkatachalam Udhayakumar
- Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
- Independenant Consultant, Decatur, Georgia
| | - Deus S Ishengoma
- National Institute for Medical Research, Dar Es Salaam, Tanzania.
- Faculty of Pharmaceutical Sciences, Monash University, Melbourne, Australia.
- Harvard T.H Chan School of Public Health, Harvard University, Boston, MA, USA.
- Department of Biochemistry, Kampala International University, Dar Es Salaam, Tanzania.
| |
Collapse
|
13
|
Konaté-Touré A, Gnagne AP, Bedia-Tanoh AV, Menan EIH, Yavo W. Increase of Plasmodium falciparum parasites carrying lumefantrine-tolerance molecular markers and lack of South East Asian pfk13 artemisinin-resistance mutations in samples collected from 2013 to 2016 in Côte d'Ivoire. J Parasit Dis 2024; 48:59-66. [PMID: 38440764 PMCID: PMC10908703 DOI: 10.1007/s12639-023-01640-4] [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] [Received: 04/27/2023] [Accepted: 12/21/2023] [Indexed: 03/06/2024] Open
Abstract
One of the major obstacles to malaria elimination in the world is the resistance in Plasmodium falciparum to most antimalarial drugs. This study aimed to estimate the prevalence of molecular markers of antimalarial drugs resistance in Côte d'Ivoire. Samples were collected from 2013 to 2016 from asymptomatic and symptomatic subjects in Abengourou, Abidjan, Grand Bassam, and San Pedro. A total of 704 participants aged between 1 year and 65 years (Mean age: 9 years ± 7.7) were enrolled. All the dried filter paper blood spots were genotyped by sequencing. Plasmodium falciparum kelch propeller domain 13 (pfk13) gene were analyzed for all the samples, while 344 samples were examined for Plasmodium falciparum multi-drug resistance 1 (pfmdr1). Overall, the success rate of molecular tests was 98.8% (340/344), 99.1% (341/344), and 94.3% (664/704) for pfmdr1 N86Y, pfmdr1 Y184F, and pfk13 genes respectively. Molecular analysis revealed twenty (5.9%; 20/340) and 219 (64.2%; 219/341) mutant alleles for pfmdr1 86Y and pfmdr1 184 F, respectively. Twenty-nine mutations in pfk13 gene (4.4%; 29/664) with 2.7% (18/664) of non-synonymous mutations was found. None of the mutations previously described in South East Asia (SEA) involved in P. falciparum resistance to artemisinin derivatives were observed in this study. According to year of collection, a decrease of the prevalence of pfk13 mutation (from 3.6 to 1.8%) and pfmdr1 N86Y mutation (from 8.5 to 4.5%) and an increase of mutant allele of pfmdr1 Y184F proportion (from 39.8 to 66.4%) were found. Comparing to previous studies in the country, this study showed an increase in lumefantrine tolerance of P. falciparum strains. This demonstrates the importance of establishing a strong system for molecular surveillance of malaria in Côte d'Ivoire.
Collapse
Affiliation(s)
- Abibatou Konaté-Touré
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34, Abidjan, Côte d’Ivoire
- Malaria Research and Control Centre, National Institute of Public Health, BPV 47, Abidjan, Côte d’Ivoire
| | - Akpa Paterne Gnagne
- Malaria Research and Control Centre, National Institute of Public Health, BPV 47, Abidjan, Côte d’Ivoire
| | - Akoua Valérie Bedia-Tanoh
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34, Abidjan, Côte d’Ivoire
- Malaria Research and Control Centre, National Institute of Public Health, BPV 47, Abidjan, Côte d’Ivoire
| | - Eby Ignace Hervé Menan
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34, Abidjan, Côte d’Ivoire
| | - William Yavo
- Department of Parasitology, Mycology, Animal Biology and, Zoology, Felix Houphouët-Boigny University, BPV 34, Abidjan, Côte d’Ivoire
- Malaria Research and Control Centre, National Institute of Public Health, BPV 47, Abidjan, Côte d’Ivoire
| |
Collapse
|
14
|
Matrevi SA, Adams T, Tandoh KZ, Opoku-Agyeman P, Bruku S, Ennuson NA, Apau-Danso PK, Fiagbedzi E, Avornyo M, Myers CJ, Futagbi J, Hagan OC, Abuaku B, Koram KA, Awandare G, Quashie NB, Duah-Quashie NO. Putative molecular markers of Plasmodium falciparum resistance to antimalarial drugs in malaria parasites from Ghana. FRONTIERS IN EPIDEMIOLOGY 2024; 4:1279835. [PMID: 38456076 PMCID: PMC10910922 DOI: 10.3389/fepid.2024.1279835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/29/2024] [Indexed: 03/09/2024]
Abstract
Introduction Antimalarial drugs including artemisinin-based combination therapy (ACT) regimens and sulphadoxine-pyrimethamine (SP) are used in Ghana for malaria therapeutics and prophylaxis respectively. The genetic basis of Plasmodium falciparum development of drug resistance involves single nucleotide polymorphisms in genes encoding proteins for multiple cellular and metabolic processes. The prevalence of single nucleotide polymorphisms in nine P. falciparum genes linked to ACT and SP resistance in the malaria parasite population was determined. Methods Archived filter paper blood blot samples from patients aged 9 years and below with uncomplicated malaria reporting at 10 sentinel sites located in three ecological zones for the Malaria Therapeutic Efficacy Studies were used. The samples used were collected from 2007-2018 malaria transmission seasons and mutations in the genes were detected using PCR and Sanger sequencing. Results In all 1,142 samples were used for the study. For falcipain-2 gene (pffp2), Sanger sequencing was successful for 872 samples and were further analysed. The prevalence of the mutants was 45% (392/872) with pffp2 markers V51I and S59F occurring in 15.0% (128/872) and 3.0% (26/872) of the samples respectively. Prevalence of other P. falciparum gene mutations: coronin (pfcoronin) was 44.8% (37/90); cysteine desulfurase (pfnfs) was 73.9% (68/92); apicoplast ribosomal protein S10 (pfarps10) was 36.8% (35/95); ferredoxin (pffd) was 8.8% (8/91); multidrug resistance protein-1 (pfmrp1) was 95.2.0% (80/84); multidrug resistance protein-2 (pfmrp2) was 91.4% (32/35); dihydrofolate reductase (pfdhfr) was 99.0% (84/85); dihydropteroate synthase (pfdhps) was 72% (68/95). Discussion The observation of numerous mutations in these genes of interest in the Ghanaian isolates, some of which have been implicated in delayed parasite clearance is of great interest. The presence of these genotypes may account for the decline in the efficacies of ACT regimens being used to treat uncomplicated malaria in the country. The need for continuous monitoring of these genetic markers to give first-hand information on parasite susceptibility to antimalarial drugs to inform policy makers and stakeholders in malaria elimination in the country is further discussed.
Collapse
Affiliation(s)
- Sena Adzoa Matrevi
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Tryphena Adams
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kwesi Zandoh Tandoh
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Philip Opoku-Agyeman
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Selassie Bruku
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Nana Aba Ennuson
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Paa Kwesi Apau-Danso
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Emmanuel Fiagbedzi
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Mary Avornyo
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Charles James Myers
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Joy Futagbi
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Oheneba Charles Hagan
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Benjamin Abuaku
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kwadwo Ansah Koram
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Gordon Awandare
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Neils Ben Quashie
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Centre for Tropical Clinical Pharmacology and Therapeutics, University of Ghana Medical School, University of Ghana, Accra, Ghana
| | - Nancy Odurowah Duah-Quashie
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| |
Collapse
|
15
|
Schäfer TM, Pessanha de Carvalho L, Inoue J, Kreidenweiss A, Held J. The problem of antimalarial resistance and its implications for drug discovery. Expert Opin Drug Discov 2024; 19:209-224. [PMID: 38108082 DOI: 10.1080/17460441.2023.2284820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Malaria remains a devastating infectious disease with hundreds of thousands of casualties each year. Antimalarial drug resistance has been a threat to malaria control and elimination for many decades and is still of concern today. Despite the continued effectiveness of current first-line treatments, namely artemisinin-based combination therapies, the emergence of drug-resistant parasites in Southeast Asia and even more alarmingly the occurrence of resistance mutations in Africa is of great concern and requires immediate attention. AREAS COVERED A comprehensive overview of the mechanisms underlying the acquisition of drug resistance in Plasmodium falciparum is given. Understanding these processes provides valuable insights that can be harnessed for the development and selection of novel antimalarials with reduced resistance potential. Additionally, strategies to mitigate resistance to antimalarial compounds on the short term by using approved drugs are discussed. EXPERT OPINION While employing strategies that utilize already approved drugs may offer a prompt and cost-effective approach to counter antimalarial drug resistance, it is crucial to recognize that only continuous efforts into the development of novel antimalarial drugs can ensure the successful treatment of malaria in the future. Incorporating resistance propensity assessment during this developmental process will increase the likelihood of effective and enduring malaria treatments.
Collapse
Affiliation(s)
| | | | - Juliana Inoue
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
16
|
Holzschuh A, Lerch A, Fakih BS, Aliy SM, Ali MH, Ali MA, Bruzzese DJ, Yukich J, Hetzel MW, Koepfli C. Using a mobile nanopore sequencing lab for end-to-end genomic surveillance of Plasmodium falciparum: A feasibility study. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0002743. [PMID: 38300956 PMCID: PMC10833559 DOI: 10.1371/journal.pgph.0002743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/20/2023] [Indexed: 02/03/2024]
Abstract
Genomic epidemiology holds promise for malaria control and elimination efforts, for example by informing on Plasmodium falciparum genetic diversity and prevalence of mutations conferring anti-malarial drug resistance. Limited sequencing infrastructure in many malaria-endemic areas prevents the rapid generation of genomic data. To address these issues, we developed and validated assays for P. falciparum nanopore sequencing in endemic sites using a mobile laboratory, targeting key antimalarial drug resistance markers and microhaplotypes. Using two multiplexed PCR reactions, we amplified six highly polymorphic microhaplotypes and ten drug resistance markers. We developed a bioinformatics workflow that allows genotyping of polyclonal malaria infections, including minority clones. We validated the panels on mock dried blood spot (DBS) and rapid diagnostic test (RDT) samples and archived DBS, demonstrating even, high read coverage across amplicons (range: 580x to 3,212x median coverage), high haplotype calling accuracy, and the ability to explore within-sample diversity of polyclonal infections. We field-tested the feasibility of rapid genotyping in Zanzibar in close collaboration with the local malaria elimination program using DBS and routinely collected RDTs as sample inputs. Our assay identified haplotypes known to confer resistance to known antimalarials in the dhfr, dhps and mdr1 genes, but no evidence of artemisinin partial resistance. Most infections (60%) were polyclonal, with high microhaplotype diversity (median HE = 0.94). In conclusion, our assays generated actionable data within a few days, and we identified current challenges for implementing nanopore sequencing in endemic countries to accelerate malaria control and elimination.
Collapse
Affiliation(s)
- Aurel Holzschuh
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
| | - Anita Lerch
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Bakar S. Fakih
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Ifakara Health Institute, Dar es Salaam, United Republic of Tanzania
| | - Safia Mohammed Aliy
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, United Republic of Tanzania
| | - Mohamed Haji Ali
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, United Republic of Tanzania
| | - Mohamed Ali Ali
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, United Republic of Tanzania
| | - Daniel J. Bruzzese
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Joshua Yukich
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, United States of America
| | - Manuel W. Hetzel
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
| | - Cristian Koepfli
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
17
|
Mesia Kahunu G, Wellmann Thomsen S, Wellmann Thomsen L, Muhindo Mavoko H, Mitashi Mulopo P, Filtenborg Hocke E, Mandoko Nkoli P, Baraka V, Minja DTR, Mousa A, Roper C, Mbongi Moke D, Mumba Ngoyi D, Mukomena Sompwe E, Muyembe Tanfum JJ, Hansson H, Alifrangis M. Identification of the PfK13 mutations R561H and P441L in the Democratic Republic of Congo. Int J Infect Dis 2024; 139:41-49. [PMID: 38016502 DOI: 10.1016/j.ijid.2023.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVES Partial artemisinin resistance, mediated by Plasmodium falciparum K13 (PfK13) mutations, has been confirmed in certain areas of East Africa that are historically associated with high-level antimalarial resistance. The Democratic Republic of Congo (DRC) borders these areas in the East. This study aimed to determine the prevalence of resistance markers in six National Malaria Control Program surveillance sites; Boende, Kabondo, Kapolowe, Kimpese, Mikalayi, and Rutshuru. METHODS The single nucleotide polymorphisms (SNPs) in P. falciparum genes PfK13, Pfdhfr, Pfdhps, Pfmdr1, and Pfcrt were assessed using targeted next-generation sequencing of isolates collected at enrollment in therapeutic efficacy studies. RESULTS PfK13 SNPs were detected in two samples: in Kabondo (R561H) and in Rutshuru (P441L), both areas near Uganda and Rwanda. The Pfdhps ISGEGA haplotype, associated with reduced sulfadoxine-pyrimethamine chemoprevention efficacy, ranged from 0.8% in Mikalayi (central DRC) to 42.2% in Rutshuru (East DRC). CONCLUSIONS R561H and P441L observed in eastern DRC are a concern, as they are associated with delayed artemisinin-based combination therapies-clearance and candidate marker of resistance, respectively. This is consistent with previous observations of shared drug resistance profiles in parasites of that region with bordering areas of Rwanda and Uganda. The likely circulation of parasites has important implications for the ongoing surveillance of partial artemisinin-resistant P. falciparum and for future efforts to mitigate its dispersal.
Collapse
Affiliation(s)
- Gauthier Mesia Kahunu
- Department of Pharmacology and Therapeutics, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Sarah Wellmann Thomsen
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Louise Wellmann Thomsen
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Emma Filtenborg Hocke
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Papy Mandoko Nkoli
- National Institute of Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Vito Baraka
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Daniel T R Minja
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Andria Mousa
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Cally Roper
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Dieudonné Mumba Ngoyi
- Department of Tropical Medicine, University of Kinshasa, Democratic Republic of Congo; National Institute of Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Eric Mukomena Sompwe
- Faculty of Medicine, University of Lubumbashi, Democratic Republic of the Congo, National Malaria Control Program, Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | - Jean Jacques Muyembe Tanfum
- Department of Tropical Medicine, University of Kinshasa, Democratic Republic of Congo; National Institute of Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Helle Hansson
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michael Alifrangis
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
18
|
Jeang B, Zhong D, Lee MC, Atieli H, Yewhalaw D, Yan G. Molecular surveillance of Kelch 13 polymorphisms in Plasmodium falciparum isolates from Kenya and Ethiopia. Malar J 2024; 23:36. [PMID: 38287365 PMCID: PMC10823687 DOI: 10.1186/s12936-023-04812-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/30/2023] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Timely molecular surveillance of Plasmodium falciparum kelch 13 (k13) gene mutations is essential for monitoring the emergence and stemming the spread of artemisinin resistance. Widespread artemisinin resistance, as observed in Southeast Asia, would reverse significant gains that have been made against the malaria burden in Africa. The purpose of this study was to assess the prevalence of k13 polymorphisms in western Kenya and Ethiopia at sites representing varying transmission intensities between 2018 and 2022. METHODS Dried blood spot samples collected through ongoing passive surveillance and malaria epidemiological studies, respectively, were investigated. The k13 gene was genotyped in P. falciparum isolates with high parasitaemia: 775 isolates from four sites in western Kenya (Homa Bay, Kakamega, Kisii, and Kombewa) and 319 isolates from five sites across Ethiopia (Arjo, Awash, Gambella, Dire Dawa, and Semera). DNA sequence variation and neutrality were analysed within each study site where mutant alleles were detected. RESULTS Sixteen Kelch13 haplotypes were detected in this study. Prevalence of nonsynonymous k13 mutations was low in both western Kenya (25/783, 3.19%) and Ethiopia (5/319, 1.57%) across the study period. Two WHO-validated mutations were detected: A675V in three isolates from Kenya and R622I in four isolates from Ethiopia. Seventeen samples from Kenya carried synonymous mutations (2.17%). No synonymous mutations were detected in Ethiopia. Genetic variation analyses and tests of neutrality further suggest an excess of low frequency polymorphisms in each study site. Fu and Li's F test statistic in Semera was 0.48 (P > 0.05), suggesting potential population selection of R622I, which appeared at a relatively high frequency (3/22, 13.04%). CONCLUSIONS This study presents an updated report on the low frequency of k13 mutations in western Kenya and Ethiopia. The WHO-validated R622I mutation, which has previously only been reported along the north-west border of Ethiopia, appeared in four isolates collected from eastern Ethiopia. The rapid expansion of R622I across Ethiopia signals the need for enhanced monitoring of the spread of drug-resistant P. falciparum parasites in East Africa. Although ACT remains currently efficacious in the study areas, continued surveillance is necessary to detect early indicators of artemisinin partial resistance.
Collapse
Affiliation(s)
- Brook Jeang
- Program in Public Health, University of California Irvine, Irvine, CA, USA
| | - Daibin Zhong
- Program in Public Health, University of California Irvine, Irvine, CA, USA
| | - Ming-Chieh Lee
- Program in Public Health, University of California Irvine, Irvine, CA, USA
| | - Harrysone Atieli
- School of Public Health and Community Development, Maseno University, Kisumu, Kenya
- International Center of Excellence for Malaria Research, Tom Mboya University College, Homa Bay, Kenya
| | - Delenasaw Yewhalaw
- School of Medical Laboratory Sciences, Faculty of Health Sciences, Jimma University, Jimma, Ethiopia
- Tropical and Infectious Diseases Research Center, Jimma University, Jimma, Ethiopia
| | - Guiyun Yan
- Program in Public Health, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
19
|
Gnondjui AA, Toure OA, Ako BA, Koui TS, Assohoun SE, Gbessi EA, N'Guessan LT, Tuo K, Beourou S, Assi SB, Yapo FA, Sanogo I, Jambou R. In vitro delayed response to dihydroartemisinin of malaria parasites infecting sickle cell erythocytes. Malar J 2024; 23:9. [PMID: 38178227 PMCID: PMC10768257 DOI: 10.1186/s12936-023-04819-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 12/09/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Decreased efficacy of artemisinin-based combination therapy (ACT) for Plasmodium falciparum malaria has been previously reported in patients with sickle cell disease (SCD). The main purpose of this study was to investigate the in vitro susceptibility of isolates to dihydro-artemisinin (DHA) to provide a hypothesis to explain this treatment failure. METHODS Isolates were collected from patients attending health centres in Abidjan with uncomplicated P. falciparum malaria. The haemoglobin type has been identified and in vitro drug sensitivity tests were conducted with the ring stage assay and maturation inhibition assay. RESULTS 134 isolates were obtained. Parasitaemia and haemoglobin levels at inclusion were lower in patients with haemoglobin HbSS and HbSC than in patients with normal HbAA. After ex vivo RSA and drug inhibition assays, the lowest rate of parasitic growth was found with isolates from HbAS red cells. Conversely, a significantly higher survival rate of parasites ranging from 15 to 34% were observed in isolates from HbSS. Isolates with in vitro reduced DHA sensitivity correlate with lower RBC count and haematocrit and higher parasitaemia at inclusion compared to those with isolates with normal DHA sensitivity. However, this decrease of in vitro sensitivity to DHA was not associated with Kelch 13-Propeller gene polymorphism. CONCLUSION This study highlights an in vitro decreased sensitivity to DHA, for isolates collected from HbSS patients, not related to the Pfkelch13 gene mutations. These results are in line with recent studies pointing out the role of the redox context in the efficacy of the drug. Indeed, SCD red cells harbour a highly different ionic and redox context in comparison with normal red cells. This study offers new insights into the understanding of artemisinin selective pressure on the malaria parasite in the context of haemoglobinopathies in Africa.
Collapse
Affiliation(s)
- Albert A Gnondjui
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
- Laboratoire Biologie et Santé, Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Offianan A Toure
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
| | - Berenger A Ako
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
| | - Tossea S Koui
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
- Laboratoire Biologie et Santé, Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Stanislas E Assohoun
- Laboratoire de Mécanique et Informatique, Université Felix Houphouët BoignyCôte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Eric A Gbessi
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
- Laboratoire Biologie et Santé, Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Landry T N'Guessan
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
| | - Karim Tuo
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
| | - Sylvain Beourou
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire
| | - Serge-Brice Assi
- Institut Pierre Richet/Programme National de Lutte contre le Paludisme, Bouaké, Côte d'Ivoire
| | - Francis A Yapo
- Laboratoire Biologie et Santé, Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | | | - Ronan Jambou
- Unité de Paludologie, Institut Pasteur Côte d'Ivoire, 01 BP 490, Abidjan 01, Côte d'Ivoire.
- Global Health Department, Institut Pasteur Paris, 25 rue du Dr Roux, 75015, Paris, France.
| |
Collapse
|
20
|
Rai S, Shukla S, Scotti L, Mani A. Drug Repurposing against Novel Therapeutic Targets in Plasmodium falciparum for Malaria: The Computational Perspective. Curr Med Chem 2024; 31:6272-6287. [PMID: 37550911 DOI: 10.2174/0929867331666230807151708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/28/2023] [Accepted: 07/01/2023] [Indexed: 08/09/2023]
Abstract
Malaria remains one of the most challenging tropical diseases. Since malaria cases are reportedly alarming in terms of infections and mortality, urgent attention is needed for addressing the issues of drug resistance in falciparum malaria. High throughput screening methods have paved way for rapid identification of anti-malarial. Furthermore, drug repurposing helps in shortening the time required for drug safety approvals. Hence, discovery of new antimalarials by drug repurposing is a promising approach for combating the disease. This article summarizes the recent computational approaches used for identifying novel antimalarials by using drug target interaction tools followed by pharmacokinetic studies.
Collapse
Affiliation(s)
- Shweta Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, 211004 India
| | - Shruti Shukla
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, 211004 India
| | - Luciana Scotti
- Postgraduate Programa in Natural and Synthetic Bioactive Compounds, University Hospital, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| | - Ashutosh Mani
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, 211004 India
| |
Collapse
|
21
|
Parikesit AA, Hermantara R, Gregorius K, Siddharta E. Designing hybrid CRISPR-Cas12 and LAMP detection systems for treatment-resistant Plasmodium falciparum with in silico method. NARRA J 2023; 3:e301. [PMID: 38455618 PMCID: PMC10919703 DOI: 10.52225/narra.v3i3.301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/21/2023] [Indexed: 03/09/2024]
Abstract
Genes associated with drug resistance of first line drugs for Plasmodium falciparum have been identified and characterized of which three genes most commonly associated with drug resistance are P. falciparum chloroquine resistance transporter gene (PfCRT), P. falciparum multidrug drug resistance gene 1 (PfMDR1), and P. falciparum Kelch protein K13 gene (PfKelch13). Polymorphism in these genes could be used as molecular markers for identifying drug resistant strains. Nucleic acid amplification test (NAAT) along with DNA sequencing is a powerful diagnostic tool that could identify these polymorphisms. However, current NAAT and DNA sequencing technologies require specific instruments which might limit its application in rural areas. More recently, a combination of isothermal amplification and CRISPR detection system showed promising results in detecting mutations at a nucleic acid level. Moreover, the Loop-mediated isothermal amplification (LAMP)-CRISPR systems offer robust and straightforward detection, enabling it to be deployed in rural and remote areas. The aim of this study was to develop a novel diagnostic method, based on LAMP of targeted genes, that would enable the identification of drug-resistant P. falciparum strains. The methods were centered on sequence analysis of P. falciparum genome, LAMP primers design, and CRISPR target prediction. Our designed primers are satisfactory for identifying polymorphism associated with drug resistant in PfCRT, PfMDR1, and PfKelch13. Overall, the developed system is promising to be used as a detection method for P. falciparum treatment-resistant strains. However, optimization and further validation the developed CRISPR-LAMP assay are needed to ensure its accuracy, reliability, and feasibility.
Collapse
Affiliation(s)
- Arli A. Parikesit
- Department of Bioinformatics, School of Life Sciences, Indonesia International Institute for Life Sciences (I3L), Jakarta, Indonesia
| | - Rio Hermantara
- Department of Biomedicine, School of Life Sciences, Indonesia International Institute for Life Sciences (I3L), Jakarta, Indonesia
| | - Kevin Gregorius
- Department of Biomedicine, School of Life Sciences, Indonesia International Institute for Life Sciences (I3L), Jakarta, Indonesia
| | - Elizabeth Siddharta
- Department of Biomedicine, School of Life Sciences, Indonesia International Institute for Life Sciences (I3L), Jakarta, Indonesia
| |
Collapse
|
22
|
Kayiba NK, Tshibangu-Kabamba E, Rosas-Aguirre A, Kaku N, Nakagama Y, Kaneko A, Makaba DM, Malekita DY, Devleesschauwer B, Likwela JL, Zakayi PK, DeMol P, Lelo GM, Hayette MP, Dikassa PL, Kido Y, Speybroeck N. The landscape of drug resistance in Plasmodium falciparum malaria in the Democratic Republic of Congo: a mapping systematic review. Trop Med Health 2023; 51:64. [PMID: 37968745 PMCID: PMC10647042 DOI: 10.1186/s41182-023-00551-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023] Open
Abstract
CONTEXT The Democratic Republic of Congo (DRC), one of the most malaria-affected countries worldwide, is a potential hub for global drug-resistant malaria. This study aimed at summarizing and mapping surveys of malaria parasites carrying molecular markers of drug-resistance across the country. METHODS A systematic mapping review was carried out before July 2023 by searching for relevant articles through seven databases (PubMed, Embase, Scopus, African Journal Online, African Index Medicus, Bioline and Web of Science). RESULTS We identified 1541 primary studies of which 29 fulfilled inclusion criteria and provided information related to 6385 Plasmodium falciparum clinical isolates (collected from 2000 to 2020). We noted the PfCRT K76T mutation encoding for chloroquine-resistance in median 32.1% [interquartile interval, IQR: 45.2] of analyzed malaria parasites. The proportion of parasites carrying this mutation decreased overtime, but wide geographic variations persisted. A single isolate had encoded the PfK13 R561H substitution that is invoked in artemisinin-resistance emergence in the Great Lakes region of Africa. Parasites carrying various mutations linked to resistance to the sulfadoxine-pyrimethamine combination were widespread and reflected a moderate resistance profile (PfDHPS A437G: 99.5% [IQR: 3.9]; PfDHPS K540E: 38.9% [IQR: 47.7]) with median 13.1% [IQR: 10.3] of them being quintuple IRN-GE mutants (i.e., parasites carrying the PfDHFR N51I-C59R-S108N and PfDHPS A437G-K540E mutations). These quintuple mutants tended to prevail in eastern regions of the country. Among circulating parasites, we did not record any parasites harboring mutations related to mefloquine-resistance, but we could suspect those with decreased susceptibility to quinine, amodiaquine, and lumefantrine based on corresponding molecular surrogates. CONCLUSIONS Drug resistance poses a serious threat to existing malaria therapies and chemoprevention options in the DRC. This review provides a baseline for monitoring public health efforts as well as evidence for decision-making in support of national malaria policies and for implementing regionally tailored control measures across the country.
Collapse
Affiliation(s)
- Nadine Kalenda Kayiba
- Research Institute of Health and Society, Université Catholique de Louvain, Brussels, Belgium
- Department of Public Health, Faculty of Medicine, University of Mbujimayi, Mbujimayi, Democratic Republic of Congo
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Evariste Tshibangu-Kabamba
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Internal Medicine, Faculty of Medicine, University of Mbujimayi, Mbujimayi, Democratic Republic of Congo
| | - Angel Rosas-Aguirre
- Research Institute of Health and Society, Université Catholique de Louvain, Brussels, Belgium
| | - Natsuko Kaku
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yu Nakagama
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akira Kaneko
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Dieudonné Mvumbi Makaba
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
- Department of Quality of Laboratories, Sciensano, Brussels, Belgium
| | - Doudou Yobi Malekita
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Brecht Devleesschauwer
- Department of Epidemiology and Public Health, Sciensano, Brussels, Belgium
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Joris Losimba Likwela
- Department of Public Health, Faculty of Medicine, University of Kisangani, Kisangani, Democratic Republic of Congo
| | - Pius Kabututu Zakayi
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Patrick DeMol
- Laboratory of Clinical Microbiology, Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | - Georges Mvumbi Lelo
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Marie-Pierre Hayette
- Laboratory of Clinical Microbiology, Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | - Paul Lusamba Dikassa
- School of Public Health, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Yasutoshi Kido
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan.
| | - Niko Speybroeck
- Research Institute of Health and Society, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
23
|
Issa MS, Warsame M, Mahamat MHT, Saleh IDM, Boulotigam K, Djimrassengar H, Issa AH, Abdelkader O, Hassoumi M, Djimadoum M, Doderer-Lang C, Ndihiokubwayo JB, Rasmussen C, Menard D. Therapeutic efficacy of artesunate-amodiaquine and artemether-lumefantrine for the treatment of uncomplicated falciparum malaria in Chad: clinical and genetic surveillance. Malar J 2023; 22:240. [PMID: 37612601 PMCID: PMC10464190 DOI: 10.1186/s12936-023-04644-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/10/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Artesunate-amodiaquine (AS-AQ) and artemether-lumefantrine (AL) are the currently recommended first-and second-line therapies for uncomplicated Plasmodium falciparum infections in Chad. This study assessed the efficacy of these artemisinin-based combinations, proportion of day 3 positive patients, proportions of molecular markers associated with P. falciparum resistance to anti-malarial drugs and variable performance of HRP2-based malaria rapid diagnostic tests (RDTs). METHODS A single-arm prospective study assessing the efficacy of AS-AQ and AL at three sites (Doba, Kelo and Koyom) was conducted between November 2020 to January 2021. Febrile children aged 6 to 59 months with confirmed uncomplicated P. falciparum infection were enrolled sequentially first to AS-AQ and then AL at each site and followed up for 28 days. The primary endpoint was PCR-adjusted adequate clinical and parasitological response (ACPR). Samples collected on day 0 were analysed for mutations in pfkelch13, pfcrt, pfmdr-1, pfdhfr, pfdhps genes and deletions in pfhrp2/pfhrp3 genes. RESULTS By the end of 28-day follow-up, per-protocol PCR corrected ACPR of 97.8% (CI 95% 88.2-100) in Kelo and 100% in Doba and Kayoma were observed among AL treated patients. For ASAQ, 100% ACPR was found in all sites. All, but one patient, did not have parasites detected on day 3. Out of the 215 day 0 samples, 96.7% showed pfkelch13 wild type allele. Seven isolates carried nonsynonymous mutations not known to be associated artemisinin partial resistance (ART-R). Most of samples had a pfcrt wild type allele (79% to 89%). The most prevalent pfmdr-1 allele detected was the single mutant 184F (51.2%). For pfdhfr and pfdhps mutations, the quintuple mutant allele N51I/C59R/S108N + G437A/540E responsible for SP treatment failures in adults and children was not detected. Single deletion in the pfhrp2 and pfhrp3 gene were detected in 10/215 (4.7%) and 2/215 (0.9%), respectively. Dual pfhrp2/pfhrp3 deletions, potentially threatening the efficacy of HRP2-based RDTs, were observed in 5/215 (2.3%) isolates. CONCLUSION The results of this study confirm that AS-AQ and AL treatments are highly efficacious in study areas in Chad. The absence of known pfkelch13 mutations in the study sites and the high parasite clearance rate at day 3 suggest the absence of ART-R. The absence of pfdhfr/pfdhps quintuple or sextuple (quintuple + 581G) mutant supports the continued use of SP for IPTp during pregnancy. The presence of parasites with dual pfhrp2/pfhrp3 deletions, potentially threatening the efficacy of HRP2-based RDTs, warrants the continued surveillance. Trial registration ACTRN12622001476729.
Collapse
Affiliation(s)
| | - Marian Warsame
- School of Public Health and Community Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | - Ali Haggar Issa
- Ecole Nationale des Agents Sanitaires et Sociaux (ENASS), N'Djamena, Chad
| | | | | | - Mbanga Djimadoum
- Faculty of Science and Human Health, University of N'Djamena, N'Djamena, Chad
| | - Cécile Doderer-Lang
- Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Université de Strasbourg, 67000, Strasbourg, France
| | | | | | - Didier Menard
- Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Université de Strasbourg, 67000, Strasbourg, France
- Malaria Genetics and Resistance Unit, INSERM U1201, Institut Pasteur, Université Paris Cité, 75015, Paris, France
- Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Université Paris Cité, 75015, Paris, France
- Laboratory of Parasitology and Medical Mycology, CHU Strasbourg, 67000, Strasbourg, France
| |
Collapse
|
24
|
Kojom Foko LP, Narang G, Jakhan J, Tamang S, Moun A, Singh V. Nationwide spatiotemporal drug resistance genetic profiling from over three decades in Indian Plasmodium falciparum and Plasmodium vivax isolates. Malar J 2023; 22:236. [PMID: 37582796 PMCID: PMC10428610 DOI: 10.1186/s12936-023-04651-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/18/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Drug resistance is a serious impediment to efficient control and elimination of malaria in endemic areas. METHODS This study aimed at analysing the genetic profile of molecular drug resistance in Plasmodium falciparum and Plasmodium vivax parasites from India over a ~ 30-year period (1993-2019). Blood samples of P. falciparum and/or P. vivax-infected patients were collected from 14 regions across India. Plasmodial genome was extracted and used for PCR amplification and sequencing of drug resistance genes in P. falciparum (crt, dhps, dhfr, mdr1, k13) and P. vivax (crt-o, dhps, dhfr, mdr1, k12) field isolates. RESULTS The double mutant pfcrt SVMNT was highly predominant across the country over three decades, with restricted presence of triple mutant CVIET from Maharashtra in 2012. High rates of pfdhfr-pfdhps quadruple mutants were observed with marginal presence of "fully resistant" quintuple mutant ACIRNI-ISGEAA. Also, resistant pfdhfr and pfdhps haplotype has significantly increased in Delhi between 1994 and 2010. For pfmdr1, only 86Y and 184F mutations were present while no pfk13 mutations associated with artemisinin resistance were observed. Regarding P. vivax isolates, the pvcrt-o K10 "AAG" insertion was absent in all samples collected from Delhi in 2017. Pvdhps double mutant SGNAV was found only in Goa samples of year 2008 for the first time. The pvmdr1 908L, 958M and 1076L mutations were highly prevalent in Delhi and Haryana between 2015 and 2019 at complete fixation. One nonsynonymous novel pvk12 polymorphism was identified (K264R) in Goa. CONCLUSIONS These findings support continuous surveillance and characterization of P. falciparum and P. vivax populations as proxy for effectiveness of anti-malarial drugs in India, especially for independent emergence of artemisinin drug resistance as recently seen in Africa.
Collapse
Affiliation(s)
- Loick P Kojom Foko
- Parasite & Host Biology Group, ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi, 110077, India
| | - Geetika Narang
- Parasite & Host Biology Group, ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi, 110077, India
| | - Jahnvi Jakhan
- Parasite & Host Biology Group, ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi, 110077, India
| | - Suman Tamang
- Parasite & Host Biology Group, ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi, 110077, India
| | - Amit Moun
- Parasite & Host Biology Group, ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi, 110077, India
| | - Vineeta Singh
- Parasite & Host Biology Group, ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi, 110077, India.
| |
Collapse
|
25
|
Holzschuh A, Lerch A, Gerlovina I, Fakih BS, Al-Mafazy AWH, Reaves EJ, Ali A, Abbas F, Ali MH, Ali MA, Hetzel MW, Yukich J, Koepfli C. Multiplexed ddPCR-amplicon sequencing reveals isolated Plasmodium falciparum populations amenable to local elimination in Zanzibar, Tanzania. Nat Commun 2023; 14:3699. [PMID: 37349311 PMCID: PMC10287761 DOI: 10.1038/s41467-023-39417-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
Zanzibar has made significant progress toward malaria elimination, but recent stagnation requires novel approaches. We developed a highly multiplexed droplet digital PCR (ddPCR)-based amplicon sequencing method targeting 35 microhaplotypes and drug-resistance loci, and successfully sequenced 290 samples from five districts covering both main islands. Here, we elucidate fine-scale Plasmodium falciparum population structure and infer relatedness and connectivity of infections using an identity-by-descent (IBD) approach. Despite high genetic diversity, we observe pronounced fine-scale spatial and temporal parasite genetic structure. Clusters of near-clonal infections on Pemba indicate persistent local transmission with limited parasite importation, presenting an opportunity for local elimination efforts. Furthermore, we observe an admixed parasite population on Unguja and detect a substantial fraction (2.9%) of significantly related infection pairs between Zanzibar and the mainland, suggesting recent importation. Our study provides a high-resolution view of parasite genetic structure across the Zanzibar archipelago and provides actionable insights for prioritizing malaria elimination efforts.
Collapse
Affiliation(s)
- Aurel Holzschuh
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Indiana, IN, USA.
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland.
| | - Anita Lerch
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Indiana, IN, USA
| | - Inna Gerlovina
- EPPIcenter Research Program, Division of HIV, ID and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Bakar S Fakih
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Ifakara Health Institute, Dar es Salaam, United Republic of Tanzania
| | | | - Erik J Reaves
- U.S. Centers for Disease Control and Prevention, President's Malaria Initiative, Dar es Salaam, United Republic of Tanzania
| | - Abdullah Ali
- Zanzibar Malaria Elimination Programme, Zanzibar, United Republic of Tanzania
| | - Faiza Abbas
- Zanzibar Malaria Elimination Programme, Zanzibar, United Republic of Tanzania
| | - Mohamed Haji Ali
- Zanzibar Malaria Elimination Programme, Zanzibar, United Republic of Tanzania
| | - Mohamed Ali Ali
- Zanzibar Malaria Elimination Programme, Zanzibar, United Republic of Tanzania
| | - Manuel W Hetzel
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Joshua Yukich
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Cristian Koepfli
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Indiana, IN, USA.
| |
Collapse
|
26
|
da Silva C, Boene S, Datta D, Rovira-Vallbona E, Aranda-Díaz A, Cisteró P, Hathaway N, Tessema S, Chidimatembue A, Matambisso G, Nhama A, Macete E, Pujol A, Nhamussua L, Galatas B, Guinovart C, Enosse S, De Carvalho E, Rogier E, Plucinski MM, Colborn J, Zulliger R, Saifodine A, Alonso PL, Candrinho B, Greenhouse B, Aide P, Saute F, Mayor A. Targeted and whole-genome sequencing reveal a north-south divide in P. falciparum drug resistance markers and genetic structure in Mozambique. Commun Biol 2023; 6:619. [PMID: 37291425 PMCID: PMC10250372 DOI: 10.1038/s42003-023-04997-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Mozambique is one of the four African countries which account for over half of all malaria deaths worldwide, yet little is known about the parasite genetic structure in that country. We performed P. falciparum amplicon and whole genome sequencing on 2251 malaria-infected blood samples collected in 2015 and 2018 in seven provinces of Mozambique to genotype antimalarial resistance markers and interrogate parasite population structure using genome-wide microhaplotyes. Here we show that the only resistance-associated markers observed at frequencies above 5% were pfmdr1-184F (59%), pfdhfr-51I/59 R/108 N (99%) and pfdhps-437G/540E (89%). The frequency of pfdhfr/pfdhps quintuple mutants associated with sulfadoxine-pyrimethamine resistance increased from 80% in 2015 to 89% in 2018 (p < 0.001), with a lower expected heterozygosity and higher relatedness of microhaplotypes surrounding pfdhps mutants than wild-type parasites suggestive of recent selection. pfdhfr/pfdhps quintuple mutants also increased from 72% in the north to 95% in the south (2018; p < 0.001). This resistance gradient was accompanied by a concentration of mutations at pfdhps-436 (17%) in the north, a south-to-north increase in the genetic complexity of P. falciparum infections (p = 0.001) and a microhaplotype signature of regional differentiation. The parasite population structure identified here offers insights to guide antimalarial interventions and epidemiological surveys.
Collapse
Affiliation(s)
- Clemente da Silva
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Simone Boene
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Debayan Datta
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | | | - Andrés Aranda-Díaz
- EPPIcenter Research Program, Division of HIV, ID, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Pau Cisteró
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | | | - Sofonias Tessema
- EPPIcenter Research Program, Division of HIV, ID, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | | | - Glória Matambisso
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Abel Nhama
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Eusebio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Arnau Pujol
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Lidia Nhamussua
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Beatriz Galatas
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | | | - Sónia Enosse
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Eva De Carvalho
- World Health Organization, WHO Country Office Maputo, Maputo, Mozambique
| | - Eric Rogier
- Malaria Branch, Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mateusz M Plucinski
- United States President's Malaria Initiative, Malaria Branch, Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - James Colborn
- Clinton Health Access Initiative, Maputo, Mozambique
| | - Rose Zulliger
- U.S. President's Malaria Initiative, USAID, Washington, DC, USA
| | | | - Pedro L Alonso
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Hospital Clinic-Universitat de Barcelona, Barcelona, Spain
| | - Baltazar Candrinho
- National Malaria Control Programme, Ministry of Health, Maputo, Mozambique
| | - Bryan Greenhouse
- EPPIcenter Research Program, Division of HIV, ID, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Francisco Saute
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Alfredo Mayor
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain.
- Department of Physiologic Sciences, Faculty of Medicine, Universidade Eduardo Mondlane, Maputo, Mozambique.
| |
Collapse
|
27
|
Namayanja C, Eregu EEI, Ongodia P, Okalebo CB, Okiror W, Okello F, Okibure A, Paasi G, Kakungulu H, Grace A, Muhindo R, Banks D, Martin C, Taylor-Robinson S, Olupot-Olupot P. Unusual clinical spectra of childhood severe malaria during malaria epidemic in eastern Uganda: a prospective study. Malar J 2023; 22:169. [PMID: 37259110 DOI: 10.1186/s12936-023-04586-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND In sub-Saharan Africa (SSA), malaria remains a public health problem despite recent reports of declining incidence. Severe malaria is a multiorgan disease with wide-ranging clinical spectra and outcomes that have been reported to vary by age, geographical location, transmission intensity over time. There are reports of recent malaria epidemics or resurgences, but few data, if any, focus on the clinical spectrum of severe malaria during epidemics. This describes the clinical spectrum and outcomes of childhood severe malaria during the disease epidemic in Eastern Uganda. METHODS This prospective cohort study from October 1, 2021, to September 7, 2022, was nested within the 'Malaria Epidemiological, Pathophysiological and Intervention studies in Highly Endemic Eastern Uganda' (TMA2016SF-1514-MEPIE Study) at Mbale Regional Referral Hospital, Uganda. Children aged 60 days to 12 years who at admission tested positive for malaria and fulfilled the clinical WHO criteria for surveillance of severe malaria were enrolled on the study. Follow-up was performed until day 28. Data were collected using a customized proforma on social demographic characteristics, clinical presentation, treatment, and outcomes. Laboratory analyses included complete blood counts, malaria RDT (SD BIOLINE Malaria Ag P.f/Pan, Ref. 05FK60-40-1) and blood slide, lactate, glucose, blood gases and electrolytes. In addition, urinalysis using dipsticks (Multistix® 10 SG, SIEMENS, Ref.2300) at the bedside was done. Data were analysed using STATA V15.0. The study had prior ethical approval. RESULTS A total of 300 participants were recruited. The median age was 4.6 years, mean of 57.2 months and IQR of 44.5 months. Many children, 164/300 (54.7%) were under 5 years, and 171/300 (57.0%) were males. The common clinical features were prostration 236/300 (78.7%), jaundice in 205/300 (68.3%), severe malarial anaemia in 158/300 (52.7%), black water fever 158/300 (52.7%) and multiple convulsions 51/300 (17.0%), impaired consciousness 50/300(16.0%), acidosis 41/300(13.7%), respiratory distress 26/300(6.7%) and coma in 18/300(6.0%). Prolonged hospitalization was found in 56/251 (22.3%) and was associated with acidosis, P = 0.041. The overall mortality was 19/300 (6.3%). Day 28 follow-up was achieved in 247/300 (82.3%). CONCLUSION During the malaria epidemic in Eastern Uganda, severe malaria affected much older children and the spectrum had more of prostration, jaundice severe malarial anaemia, black water fever and multiple convulsions with less of earlier reported respiratory distress and cerebral malaria.
Collapse
Affiliation(s)
- Cate Namayanja
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda.
- Department of Pediatrics and Child Health, Busitema University Faculty of Health Sciences, Mbale, Uganda.
| | - Egiru Emma Isaiah Eregu
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
- Department of Pediatrics and Child Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
| | - Paul Ongodia
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
| | - Charles Benard Okalebo
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
| | - William Okiror
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
| | - Francis Okello
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
- Varimetrics Group Limited, Mbale, Uganda
| | | | - George Paasi
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
| | - Hellen Kakungulu
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
- Varimetrics Group Limited, Mbale, Uganda
| | - Abongo Grace
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
| | - Rita Muhindo
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
| | - Duncan Banks
- Busitema University, TORORO, Uganda
- The Open University, Milton Keynes, UK
| | - Chebet Martin
- Department of Pediatrics and Child Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
| | - Simon Taylor-Robinson
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
- Imperial College London, London, UK
| | - Peter Olupot-Olupot
- Mbale Clinical Research Institute, P.O. Box 1966, Mbale, Uganda
- Department of Community and Public Health, Busitema University Faculty of Health Sciences, Mbale, Uganda
| |
Collapse
|
28
|
da Silva C, Matias D, Dias B, Cancio B, Silva M, Viegas R, Chivale N, Luis S, Salvador C, Duarte D, Arnaldo P, Enosse S, Nogueira F. Anti-malarial resistance in Mozambique: Absence of Plasmodium falciparum Kelch 13 (K13) propeller domain polymorphisms associated with resistance to artemisinins. Malar J 2023; 22:160. [PMID: 37208708 DOI: 10.1186/s12936-023-04589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Malaria remains one of the most serious public health problems in sub-Saharan Africa and Mozambique is the world's fourth largest contributor, with 4.7% of disease cases and 3.6% of total deaths due to malaria. Its control relies on the fight against the vector and treatment of confirmed cases with anti-malarial drugs. Molecular surveillance is an important tool for monitoring the spread of anti-malarial drug resistance. METHODS A cross-sectional study recruited 450 participants with malaria infection detected by Rapid Diagnostic Tests, from three different study sites (Niassa, Manica and Maputo) between April and August 2021. Correspondent blood samples were collected on filter paper (Whatman® FTA® cards), parasite DNA extracted and pfk13 gene sequenced using Sanger method. SIFT software (Sorting Intolerant From Tolerant) was used, predict whether an amino acid substitution affects protein function. RESULTS No pfkelch13-mediated artemisinin resistance gene mutation was detected in this study settings. However, non-synonymous mutations were detected at prevalence of 10.2%, 6% and 5% in Niassa, Manica and Maputo, respectively. Most (56.3%) of the reported non-synonymous mutations were due to substitution at the first base of the codon, 25% at the second base and 18.8% at the third base. Additionally, 50% of non-synonymous mutations showed a SIFTscore bellow cut off value of 0.05, therefore, they were predicted to be deleterious. CONCLUSION These results do not show an emergence of artemisinin resistance cases in Mozambique. However, the increased number of novel non-synonymous mutations highlights the relevance of increasing the number of studies focused on the molecular surveillance of artemisinin resistance markers, for its early detection.
Collapse
Affiliation(s)
- Clemente da Silva
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal.
| | - Daniela Matias
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| | - Brigite Dias
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| | - Beatriz Cancio
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| | - Miguel Silva
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| | - Ruben Viegas
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| | - Nordino Chivale
- Instituto Nacional de Saúde (INS), Av. Eduardo Mondlane Nº 1008, Caixa Postal 264, Maputo, Mozambique
| | - Sonia Luis
- Hospital Provincial de Matola, 2CPV+55, Matola, Mozambique
| | - Crizolgo Salvador
- Instituto Nacional de Saúde (INS), Av. Eduardo Mondlane Nº 1008, Caixa Postal 264, Maputo, Mozambique
| | - Denise Duarte
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| | - Paulo Arnaldo
- Instituto Nacional de Saúde (INS), Av. Eduardo Mondlane Nº 1008, Caixa Postal 264, Maputo, Mozambique
| | - Sonia Enosse
- Malaria Consortium, Av. Lucas Elias Kumato, Nº 118. Bairro da Sommershield, Maputo, Mozambique
| | - Fatima Nogueira
- Global Health and Tropical Medicine (GHTM), Department of Medical Parasitology, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira Nº100, 1349-008, Lisbon, Portugal
| |
Collapse
|
29
|
Ali IM, Kom Tchuenkam PV, Tagomo SS, Hornela M, Moyeh MN, Nfor EN, Nji AM, Fomboh CT, Nana WD, Chedjou Kengne JP, Ngwa Niba PT, Ekoyol GE, Achu DF, Bigoga JD, Mbacham WF. Allelic Frequencies of Mutants of the Plasmodium falciparum, Quinoline and Folate Metabolizing genes in the West Region of Cameroon. Heliyon 2022; 8:e11861. [DOI: 10.1016/j.heliyon.2022.e11861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/03/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
|
30
|
Hassen J, Alemayehu GS, Dinka H, Golassa L. High prevalence of Pfcrt 76T and Pfmdr1 N86 genotypes in malaria infected patients attending health facilities in East Shewa zone, Oromia Regional State, Ethiopia. Malar J 2022; 21:286. [PMID: 36207750 PMCID: PMC9547420 DOI: 10.1186/s12936-022-04304-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
Background Plasmodium falciparum resistance to series of anti-malarial drugs is a major challenge in efforts to control and/or eliminate malaria globally. In 1998, following the widespread of chloroquine (CQ) resistant P. falciparum, Ethiopia switched from CQ to sulfadoxine–pyrimethamine (SP) and subsequently in 2004 from SP to artemether–lumefantrine (AL) for the treatment of uncomplicated falciparum malaria. Data on the prevalence of CQ resistance markers after more than two decades of its removal is important to map the selection pressure behind the targets codons of interest. The present study was conducted to determine the prevalence of mutations in Pfcrt K76T and Pfmdr1 N86Y codons among malaria-infected patients from Adama, Olenchiti and Metehara sites of East Shewa zone, Oromia Regional State, Ethiopia. Methods Finger-prick whole blood samples were collected on 3MM Whatman ® filter papers from a total of 121 microscopically confirmed P. falciparum infected patients. Extraction of parasite DNA was done by Chelex-100 method from dried blood spot (DBS). Genomic DNA template was used to amplify Pfcrt K76T and Pfmdr1 N86Y codons by nested PCR. Nested PCR products were subjected to Artherobacter protophormiae-I (APoI) restriction enzyme digestion to determine mutations at codons 76 and 86 of Pfcrt and Pfmdr1 genes, respectively. Results Of 83 P. falciparum isolates successfully genotyped for Pfcrt K76T, 91.6% carried the mutant genotypes (76T). The prevalence of Pfcrt 76T was 95.7%, 92.5% and 84.5% in Adama, Metehara and Olenchiti, respectively. The prevalence of Pfcrt 76T mutations in three of the study sites showed no statistical significance difference (χ2 = 1.895; P = 0.388). On the other hand, of the 80 P. falciparum samples successfully amplified for Pfmdr1, all carried the wild-type genotypes (Pfmdr1 N86). Conclusion Although CQ officially has been ceased for the treatment of falciparum malaria for more than two decades in Ethiopia, greater proportions of P. falciparum clinical isolates circulating in the study areas carry the mutant 76T genotypes indicating the presence of indirect CQ pressure in the country. However, the return of Pfmdr1 N86 wild-type allele may be favoured by the use of AL for the treatment of uncomplicated falciparum malaria. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04304-5.
Collapse
Affiliation(s)
- Jifar Hassen
- Department of Applied Biology, School of Applied Natural Science, Adama Science and Technology University, P. O. Box 1888, Adama, Ethiopia.
| | - Gezahegn Solomon Alemayehu
- Research and Community Service Center, College of Health Science, Defense University, P. O. Box 1419, Bishoftu, Ethiopia
| | - Hunduma Dinka
- Department of Applied Biology, School of Applied Natural Science, Adama Science and Technology University, P. O. Box 1888, Adama, Ethiopia
| | - Lemu Golassa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, P. O. Box 1176, Addis Ababa, Ethiopia
| |
Collapse
|
31
|
Edgar RCS, Siddiqui G, Hjerrild K, Malcolm TR, Vinh NB, Webb CT, Holmes C, MacRaild CA, Chernih HC, Suen WW, Counihan NA, Creek DJ, Scammells PJ, McGowan S, de Koning-Ward TF. Genetic and chemical validation of Plasmodium falciparum aminopeptidase PfA-M17 as a drug target in the hemoglobin digestion pathway. eLife 2022; 11:e80813. [PMID: 36097817 PMCID: PMC9470162 DOI: 10.7554/elife.80813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
Plasmodium falciparum, the causative agent of malaria, remains a global health threat as parasites continue to develop resistance to antimalarial drugs used throughout the world. Accordingly, drugs with novel modes of action are desperately required to combat malaria. P. falciparum parasites infect human red blood cells where they digest the host's main protein constituent, hemoglobin. Leucine aminopeptidase PfA-M17 is one of several aminopeptidases that have been implicated in the last step of this digestive pathway. Here, we use both reverse genetics and a compound specifically designed to inhibit the activity of PfA-M17 to show that PfA-M17 is essential for P. falciparum survival as it provides parasites with free amino acids for growth, many of which are highly likely to originate from hemoglobin. We further show that loss of PfA-M17 results in parasites exhibiting multiple digestive vacuoles at the trophozoite stage. In contrast to other hemoglobin-degrading proteases that have overlapping redundant functions, we validate PfA-M17 as a potential novel drug target.
Collapse
Affiliation(s)
- Rebecca CS Edgar
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | | | - Tess R Malcolm
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Natalie B Vinh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Chaille T Webb
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Clare Holmes
- CSIRO Australian Centre for Disease PreparednessGeelongAustralia
| | - Christopher A MacRaild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Hope C Chernih
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Willy W Suen
- CSIRO Australian Centre for Disease PreparednessGeelongAustralia
| | - Natalie A Counihan
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Sheena McGowan
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| |
Collapse
|
32
|
Temporal trends in molecular markers of drug resistance in Plasmodium falciparum in human blood and profiles of corresponding resistant markers in mosquito oocysts in Asembo, western Kenya. Malar J 2022; 21:265. [PMID: 36100912 PMCID: PMC9472345 DOI: 10.1186/s12936-022-04284-6] [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: 06/13/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Over the last two decades, the scale-up of vector control and changes in the first-line anti-malarial, from chloroquine (CQ) to sulfadoxine-pyrimethamine (SP) and then to artemether-lumefantrine (AL), have resulted in significant decreases in malaria burden in western Kenya. This study evaluated the long-term effects of control interventions on molecular markers of Plasmodium falciparum drug resistance using parasites obtained from humans and mosquitoes at discrete time points. METHODS Dried blood spot samples collected in 2012 and 2017 community surveys in Asembo, Kenya were genotyped by Sanger sequencing for markers associated with resistance to SP (Pfdhfr, Pfdhps), CQ, AQ, lumefantrine (Pfcrt, Pfmdr1) and artemisinin (Pfk13). Temporal trends in the prevalence of these markers, including data from 2012 to 2017 as well as published data from 1996, 2001, 2007 from same area, were analysed. The same markers from mosquito oocysts collected in 2012 were compared with results from human blood samples. RESULTS The prevalence of SP dhfr/dhps quintuple mutant haplotype C50I51R59N108I164/S436G437E540A581A613 increased from 19.7% in 1996 to 86.0% in 2012, while an increase in the sextuple mutant haplotype C50I51R59N108I164/H436G437E540A581A613 containing Pfdhps-436H was found from 10.5% in 2012 to 34.6% in 2017. Resistant Pfcrt-76 T declined from 94.6% in 2007 to 18.3% in 2012 and 0.9% in 2017. Mutant Pfmdr1-86Y decreased across years from 74.8% in 1996 to zero in 2017, mutant Pfmdr1-184F and wild Pfmdr1-D1246 increased from 17.9% to 58.9% in 2007 to 55.9% and 90.1% in 2017, respectively. Pfmdr1 haplotype N86F184S1034N1042D1246 increased from 11.0% in 2007 to 49.6% in 2017. No resistant mutations in Pfk13 were found. Prevalence of Pfdhps-436H was lower while prevalence of Pfcrt-76 T was higher in mosquitoes than in human blood samples. CONCLUSION This study showed an increased prevalence of dhfr/dhps resistant markers over 20 years with the emergence of Pfdhps-436H mutant a decade ago in Asembo. The reversal of Pfcrt from CQ-resistant to CQ-sensitive genotype occurred following 19 years of CQ withdrawal. No Pfk13 markers associated with artemisinin resistance were detected, but the increased haplotype of Pfmdr1 N86F184S1034N1042D1246 was observed. The differences in prevalence of Pfdhps-436H and Pfcrt-76 T SNPs between two hosts and the role of mosquitoes in the transmission of drug resistant parasites require further investigation.
Collapse
|
33
|
Kagoro FM, Allen E, Mabuza A, Workman L, Magagula R, Kok G, Davies C, Malatje G, Guérin PJ, Dhorda M, Maude RJ, Raman J, Barnes KI. Making data map-worthy-enhancing routine malaria data to support surveillance and mapping of Plasmodium falciparum anti-malarial resistance in a pre-elimination sub-Saharan African setting: a molecular and spatiotemporal epidemiology study. Malar J 2022; 21:207. [PMID: 35768869 PMCID: PMC9244181 DOI: 10.1186/s12936-022-04224-4] [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: 02/02/2022] [Accepted: 05/29/2022] [Indexed: 11/15/2022] Open
Abstract
Background Independent emergence and spread of artemisinin-resistant Plasmodium falciparum malaria have recently been confirmed in Africa, with molecular markers associated with artemisinin resistance increasingly detected. Surveillance to promptly detect and effectively respond to anti-malarial resistance is generally suboptimal in Africa, especially in low transmission settings where therapeutic efficacy studies are often not feasible due to recruitment challenges. However, these communities may be at higher risk of anti-malarial resistance. Methods From March 2018 to February 2020, a sequential mixed-methods study was conducted to evaluate the feasibility of the near-real-time linkage of individual patient anti-malarial resistance profiles with their case notifications and treatment response reports, and map these to fine scales in Nkomazi sub-district, Mpumalanga, a pre-elimination area in South Africa. Results Plasmodium falciparum molecular marker resistance profiles were linked to 55.1% (2636/4787) of notified malaria cases, 85% (2240/2636) of which were mapped to healthcare facility, ward and locality levels. Over time, linkage of individual malaria case demographic and molecular data increased to 75.1%. No artemisinin resistant validated/associated Kelch-13 mutations were detected in the 2385 PCR positive samples. Almost all 2812 samples assessed for lumefantrine susceptibility carried the wildtype mdr86ASN and crt76LYS alleles, potentially associated with decreased lumefantrine susceptibility. Conclusion Routine near-real-time mapping of molecular markers associated with anti-malarial drug resistance on a fine spatial scale provides a rapid and efficient early warning system for emerging resistance. The lessons learnt here could inform scale-up to provincial, national and regional malaria elimination programmes, and may be relevant for other antimicrobial resistance surveillance. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04224-4.
Collapse
Affiliation(s)
- Frank M Kagoro
- Collaborating Centre for Optimising Antimalarial Therapy (CCOAT), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town (UCT), Cape Town, South Africa.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,WorldWide Antimalarial Resistance Network (WWARN), Southern African Regional Hub, Division of Clinical Pharmacology, Department of Medicine, UCT, Mbombela, South Africa.,Infectious Diseases Data Observatory (IDDO), Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth Allen
- Collaborating Centre for Optimising Antimalarial Therapy (CCOAT), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town (UCT), Cape Town, South Africa.,WorldWide Antimalarial Resistance Network (WWARN), Southern African Regional Hub, Division of Clinical Pharmacology, Department of Medicine, UCT, Mbombela, South Africa.,Infectious Diseases Data Observatory (IDDO), Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aaron Mabuza
- Collaborating Centre for Optimising Antimalarial Therapy (CCOAT), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town (UCT), Cape Town, South Africa.,WorldWide Antimalarial Resistance Network (WWARN), Southern African Regional Hub, Division of Clinical Pharmacology, Department of Medicine, UCT, Mbombela, South Africa
| | - Lesley Workman
- Collaborating Centre for Optimising Antimalarial Therapy (CCOAT), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town (UCT), Cape Town, South Africa.,WorldWide Antimalarial Resistance Network (WWARN), Southern African Regional Hub, Division of Clinical Pharmacology, Department of Medicine, UCT, Mbombela, South Africa.,Infectious Diseases Data Observatory (IDDO), Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ray Magagula
- Mpumalanga Provincial Malaria Elimination Programme, Mbombela, Mpumalanga, South Africa
| | - Gerdalize Kok
- Mpumalanga Provincial Malaria Elimination Programme, Mbombela, Mpumalanga, South Africa
| | - Craig Davies
- Malaria Programme, Clinton Health Access Initiative, Pretoria, South Africa
| | - Gillian Malatje
- Mpumalanga Provincial Malaria Elimination Programme, Mbombela, Mpumalanga, South Africa
| | - Philippe J Guérin
- WorldWide Antimalarial Resistance Network (WWARN), Southern African Regional Hub, Division of Clinical Pharmacology, Department of Medicine, UCT, Mbombela, South Africa.,Infectious Diseases Data Observatory (IDDO), Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Infectious Diseases Data Observatory (IDDO), Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Richard J Maude
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA.,The Open University, Milton Keynes, UK
| | - Jaishree Raman
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Disease, Johannesburg, Gauteng, South Africa.,Wits Research Institute for Malaria, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.,UP Institute for Sustainable Malaria Control, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Karen I Barnes
- Collaborating Centre for Optimising Antimalarial Therapy (CCOAT), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town (UCT), Cape Town, South Africa. .,WorldWide Antimalarial Resistance Network (WWARN), Southern African Regional Hub, Division of Clinical Pharmacology, Department of Medicine, UCT, Mbombela, South Africa. .,Infectious Diseases Data Observatory (IDDO), Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
34
|
Cohen JM, Okumu F, Moonen B. The fight against malaria: Diminishing gains and growing challenges. Sci Transl Med 2022; 14:eabn3256. [PMID: 35767649 DOI: 10.1126/scitranslmed.abn3256] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Since the year 2000, historic reductions in malaria incidence and mortality have been driven by the widespread distribution of bed nets, drugs, and insecticides for the prevention and treatment of malaria. Scale-up of these tools has been enabled by an increase in malaria financing compounded by price reductions, yet these trends are unlikely to continue at the same rate. Rapid population growth in high-endemic areas requires procurement of more of these tools just to maintain current coverage, even as prices are likely to increase as resistance to drugs and insecticides forces shifts to newer products. Further progress toward the long-term goal of malaria eradication requires a combination of greater funding, more cost-effective resource allocation, and fundamental changes to the global malaria control strategy.
Collapse
Affiliation(s)
| | - Fredros Okumu
- Environmental Health and Ecological Science Department, Ifakara Health Institute, Ifakara, Tanzania.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK.,School of Life Science and Bioengineering, Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania
| | - Bruno Moonen
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| |
Collapse
|
35
|
Gupta Y, Sharma N, Singh S, Romero JG, Rajendran V, Mogire RM, Kashif M, Beach J, Jeske W, Poonam, Ogutu BR, Kanzok SM, Akala HM, Legac J, Rosenthal PJ, Rademacher DJ, Durvasula R, Singh AP, Rathi B, Kempaiah P. The Multistage Antimalarial Compound Calxinin Perturbates P. falciparum Ca 2+ Homeostasis by Targeting a Unique Ion Channel. Pharmaceutics 2022; 14:1371. [PMID: 35890267 PMCID: PMC9319510 DOI: 10.3390/pharmaceutics14071371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 12/22/2022] Open
Abstract
Malaria elimination urgently needs novel antimalarial therapies that transcend resistance, toxicity, and high costs. Our multicentric international collaborative team focuses on developing multistage antimalarials that exhibit novel mechanisms of action. Here, we describe the design, synthesis, and evaluation of a novel multistage antimalarial compound, 'Calxinin'. A compound that consists of hydroxyethylamine (HEA) and trifluoromethyl-benzyl-piperazine. Calxinin exhibits potent inhibitory activity in the nanomolar range against the asexual blood stages of drug-sensitive (3D7), multidrug-resistant (Dd2), artemisinin-resistant (IPC4912), and fresh Kenyan field isolated Plasmodium falciparum strains. Calxinin treatment resulted in diminished maturation of parasite sexual precursor cells (gametocytes) accompanied by distorted parasite morphology. Further, in vitro liver-stage testing with a mouse model showed reduced parasite load at an IC50 of 79 nM. A single dose (10 mg/kg) of Calxinin resulted in a 30% reduction in parasitemia in mice infected with a chloroquine-resistant strain of the rodent parasite P. berghei. The ex vivo ookinete inhibitory concentration within mosquito gut IC50 was 150 nM. Cellular in vitro toxicity assays in the primary and immortalized human cell lines did not show cytotoxicity. A computational protein target identification pipeline identified a putative P. falciparum membrane protein (Pf3D7_1313500) involved in parasite calcium (Ca2+) homeostasis as a potential Calxinin target. This highly conserved protein is related to the family of transient receptor potential cation channels (TRP-ML). Target validation experiments showed that exposure of parasitized RBCs (pRBCs) to Calxinin induces a rapid release of intracellular Ca2+ from pRBCs; leaving de-calcinated parasites trapped in RBCs. Overall, we demonstrated that Calxinin is a promising antimalarial lead compound with a novel mechanism of action and with potential therapeutic, prophylactic, and transmission-blocking properties against parasites resistant to current antimalarials.
Collapse
Affiliation(s)
- Yash Gupta
- Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (Y.G.); (R.D.)
| | - Neha Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, New Delhi 110021, India; (N.S.); (S.S.)
| | - Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, New Delhi 110021, India; (N.S.); (S.S.)
| | - Jesus G. Romero
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
- School of Biology, Institute of Experimental Biology, Central University of Venezuela, Caracas 1040, Venezuela
| | - Vinoth Rajendran
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India;
| | - Reagan M. Mogire
- Centre Clinical Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya; (R.M.M.); (B.R.O.); (H.M.A.)
| | - Mohammad Kashif
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi 110067, India; (M.K.); (A.P.S.)
| | - Jordan Beach
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
| | - Walter Jeske
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi, New Delhi 110021, India;
- Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi 110007, India
| | - Bernhards R. Ogutu
- Centre Clinical Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya; (R.M.M.); (B.R.O.); (H.M.A.)
| | - Stefan M. Kanzok
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA;
| | - Hoseah M. Akala
- Centre Clinical Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya; (R.M.M.); (B.R.O.); (H.M.A.)
| | - Jennifer Legac
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (J.L.); (P.J.R.)
| | - Philip J. Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (J.L.); (P.J.R.)
| | - David J. Rademacher
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
- Core Imaging Facility and Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Ravi Durvasula
- Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (Y.G.); (R.D.)
| | - Agam P. Singh
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi 110067, India; (M.K.); (A.P.S.)
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, New Delhi 110021, India; (N.S.); (S.S.)
- Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi 110007, India
| | - Prakasha Kempaiah
- Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (Y.G.); (R.D.)
| |
Collapse
|
36
|
Paton DG, Probst AS, Ma E, Adams KL, Shaw WR, Singh N, Bopp S, Volkman SK, Hien DFS, Paré PSL, Yerbanga RS, Diabaté A, Dabiré RK, Lefèvre T, Wirth DF, Catteruccia F. Using an antimalarial in mosquitoes overcomes Anopheles and Plasmodium resistance to malaria control strategies. PLoS Pathog 2022; 18:e1010609. [PMID: 35687594 PMCID: PMC9223321 DOI: 10.1371/journal.ppat.1010609] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/23/2022] [Accepted: 05/20/2022] [Indexed: 11/18/2022] Open
Abstract
The spread of insecticide resistance in Anopheles mosquitoes and drug resistance in Plasmodium parasites is contributing to a global resurgence of malaria, making the generation of control tools that can overcome these roadblocks an urgent public health priority. We recently showed that the transmission of Plasmodium falciparum parasites can be efficiently blocked when exposing Anopheles gambiae females to antimalarials deposited on a treated surface, with no negative consequences on major components of mosquito fitness. Here, we demonstrate this approach can overcome the hurdles of insecticide resistance in mosquitoes and drug resistant in parasites. We show that the transmission-blocking efficacy of mosquito-targeted antimalarials is maintained when field-derived, insecticide resistant Anopheles are exposed to the potent cytochrome b inhibitor atovaquone, demonstrating that this drug escapes insecticide resistance mechanisms that could potentially interfere with its function. Moreover, this approach prevents transmission of field-derived, artemisinin resistant P. falciparum parasites (Kelch13 C580Y mutant), proving that this strategy could be used to prevent the spread of parasite mutations that induce resistance to front-line antimalarials. Atovaquone is also highly effective at limiting parasite development when ingested by mosquitoes in sugar solutions, including in ongoing infections. These data support the use of mosquito-targeted antimalarials as a promising tool to complement and extend the efficacy of current malaria control interventions. Effective control of malaria is hampered by resistance to vector-targeted insecticides and parasite-targeted drugs. This situation is exacerbated by a critical lack of chemical diversity in both interventions and, as such, new interventions are urgently needed. Recent laboratory studies have shown that an alternative approach based on treating Anopheles mosquitoes directly with antimalarial compounds can make mosquitoes incapable of transmitting the Plasmodium parasites that cause malaria. While promising, showing that mosquito-targeted antimalarials remain effective against wild parasites and mosquitoes, including drug- and insecticide-resistant populations in malaria-endemic countries, is crucial to the future viability of this approach. In this study, carried out in the US and Burkina Faso, we show that insecticide-resistance mechanisms found in highly resistant, natural Anopheles mosquito populations do not interfere with the transmission blocking activity of tarsal exposure to the antimalarial atovaquone, and that mosquito-targeted antimalarial exposure can block transmission of parasites resistant to the main therapeutic antimalarial drug artemisinin. By combining lab, and field-based studies in this way we have demonstrated that this novel approach can be effective in areas where conventional control measures are no longer as effective.
Collapse
Affiliation(s)
- Douglas G. Paton
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
- * E-mail: (DGP); (FC)
| | - Alexandra S. Probst
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Erica Ma
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Kelsey L. Adams
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - W. Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Naresh Singh
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Selina Bopp
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Sarah K. Volkman
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Domombele F. S. Hien
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Prislaure S. L. Paré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Rakiswendé S. Yerbanga
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Abdoullaye Diabaté
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Roch K. Dabiré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Thierry Lefèvre
- MIVEGEC, IRD, CNRS, University of Montpellier, Montpellier, France
- Laboratoire mixte international sur les vecteurs (LAMIVECT), Bobo Dioulasso, Burkina Faso
- Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
| | - Dyann F. Wirth
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
- * E-mail: (DGP); (FC)
| |
Collapse
|
37
|
Targeted Amplicon Deep Sequencing for Monitoring Antimalarial Resistance Markers in Western Kenya. Antimicrob Agents Chemother 2022; 66:e0194521. [PMID: 35266823 PMCID: PMC9017353 DOI: 10.1128/aac.01945-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Molecular surveillance of Plasmodium falciparum parasites is important to track emerging and new mutations and trends in established mutations and should serve as an early warning system for antimalarial resistance. Dried blood spots were obtained from a Plasmodium falciparum malaria survey in school children conducted across eight counties in western Kenya in 2019. Real-time PCR identified 500 P. falciparum-positive samples that were amplified at five drug resistance loci for targeted amplicon deep sequencing (TADS). The absence of important kelch 13 mutations was similar to previous findings in Kenya pre-2019, and low-frequency mutations were observed in codons 569 and 578. The chloroquine resistance transporter gene codons 76 and 145 were wild type, indicating that the parasites were chloroquine and piperaquine sensitive, respectively. The multidrug resistance gene 1 haplotypes based on codons 86, 184, and 199 were predominantly present in mixed infections with haplotypes NYT and NFT, driven by the absence of chloroquine pressure and the use of lumefantrine, respectively. The sulfadoxine-pyrimethamine resistance profile was a “superresistant” combination of triple mutations in both Pfdhfr (51I 59R 108N) and Pfdhps (436H 437G 540E), rendering sulfadoxine-pyrimethamine ineffective. TADS highlighted the low-frequency variants, allowing the early identification of new mutations, Pfmdr1 codon 199S and Pfdhfr codon 85I and emerging 164L mutations. The added value of TADS is its accuracy in identifying mixed-genotype infections and for high-throughput monitoring of antimalarial resistance markers.
Collapse
|
38
|
Murmu LK, Barik TK. An analysis of Plasmodium falciparum-K13 mutations in India. J Parasit Dis 2022; 46:296-303. [PMID: 35299922 PMCID: PMC8901923 DOI: 10.1007/s12639-021-01425-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/23/2021] [Indexed: 10/20/2022] Open
Abstract
Malaria is one of the deadliest parasitic diseases in human. Currently, Artemisinin-based combination therapy is considered as the gold standard and most common treatment option. However, the origin and transmission of Plasmodium falciparum from the Greater Mekong Subregion, which has decreased artemisinin (ART) sensitivity, has sparked global concern. The reduced ART sensitivity has been associated with mutations in the Atpase6 and Kelch13 propeller domain of Plasmodium falciparum. A molecular marker is critically needed to monitor the spread of artemisinin resistance. In this article, we reviewed the k13 mutations and potential marker for ART resistance in India. There have been fourteen mutations identified, three of which have been validated by the World Health Organization (WHO) as artemisinin resistance mutations (F446I, R561H/C, and R539T). Among them, the role of F446I and R561H/C in ART resistance is conflicting. R539T and G625R mutation has been identified as an ART- resistance marker in India.
Collapse
Affiliation(s)
- Laxman Kumar Murmu
- P.G. Department of Zoology, Berhampur University, Berhampur, Ganjam, 760007 Odisha India
| | - Tapan Kumar Barik
- P.G. Department of Zoology, Berhampur University, Berhampur, Ganjam, 760007 Odisha India
| |
Collapse
|
39
|
Njiro BJ, Mutagonda RF, Chamani AT, Mwakyandile T, Sabas D, Bwire GM. Molecular surveillance of chloroquine-resistant Plasmodium falciparum in sub-Saharan African countries after withdrawal of chloroquine for treatment of uncomplicated malaria: a systematic review. J Infect Public Health 2022; 15:550-557. [DOI: 10.1016/j.jiph.2022.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 03/14/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022] Open
|
40
|
Niba PTN, Nji AM, Ali IM, Akam LF, Dongmo CH, Chedjou JPK, Fomboh CT, Nana WD, Oben OLA, Selly-Ngaloumo AA, Moyeh MN, Ngu JA, Ludovic AJ, Aboh PM, Ambani MCE, Omgba PAM, Kotcholi GB, Adzemye LM, Nna DRA, Douanla A, Ango Z, Ewane MS, Ticha JT, Tatah FM, Dinza G, Ndikum VN, Fosah DA, Bigoga JD, Alifrangis M, Mbacham WF. Effectiveness and safety of artesunate-amodiaquine versus artemether-lumefantrine for home-based treatment of uncomplicated Plasmodium falciparum malaria among children 6-120 months in Yaoundé, Cameroon: a randomized trial. BMC Infect Dis 2022; 22:166. [PMID: 35189818 PMCID: PMC8862275 DOI: 10.1186/s12879-022-07101-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 01/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background Many studies have reported high efficacy and safety of artesunate-amodiaquine (AS-AQ) and artemether-lumefantrine (AL) when administered under direct observation in Cameroon. There is paucity of data to support their continuous use in home-based treatment of uncomplicated Plasmodium falciparum malaria in Cameroon. Hence, this study aimed to assess the effectiveness and safety of AS-AQ versus AL for home-based treatment of uncomplicated P. falciparum malaria among children 6–120 months in Yaoundé, Cameroon. Methods A two-arm, open-label, randomized, controlled trial comparing the equivalence of AS-AQ (experimental group) and AL (control group) was carried out from May 2019 to April 2020 at two secondary hospitals in Yaoundé. Participants were randomized to receive either AS-AQ or AL. After the first dose, antimalarial drugs were given at home, rather than under direct observation by a study staff. The conventional on-treatment and post-treatment laboratory and clinical evaluations were not done until day 3 of the full antimalarial treatment course. The evaluation of effectiveness was mainly based on per protocol polymerase chain reaction adjusted adequate clinical and parasitological response (PP PCR adjusted ACPR) on day 28 post-treatment. Safety was based on assessment of adverse events (AEs) and severe adverse events (SAEs) from day 1 to day 28. Results A total of 242 children were randomized to receive AS-AQ (n = 114) and AL (n = 128). The PP PCR adjusted day 28 cure rates were [AS-AQ = 96.9% (95% CI, 91.2–99.4) versus AL = 95.5% (95% CI, 89.9–98.5), P = 0.797]. Expected mild to moderate adverse events were reported in both arms [AS-AQ = 83 (84.7%) versus AL = 99 (86.1%), P = 0.774]. The most common adverse events included: transient changes of hematologic indices and fever. Conclusions This study demonstrated that AS-AQ and AL are effective and safe for home management of malaria in Yaoundé. The evidence from this study supports the parallel use of the two drugs in routine practice. However, the findings from this study do not describe the likely duration of antimalarial effectiveness in holoendemic areas where multiple courses of treatment might be required. Trial registration: This study is a randomized controlled trial and it was retrospectively registered on 23/09/2020 at ClinicalTrials.gov with registration number NCT04565184. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07101-2.
Collapse
Affiliation(s)
- Peter Thelma Ngwa Niba
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Akindeh Mbuh Nji
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Innocent Mbulli Ali
- The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Lawrence Fonyonga Akam
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Cedric Hermann Dongmo
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean Paul Kengne Chedjou
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Calvino Tah Fomboh
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - William Dorian Nana
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Ornella Laetitia Ayem Oben
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Abdel Aziz Selly-Ngaloumo
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Marcel N Moyeh
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, Buea, Cameroon
| | - Jude Achidi Ngu
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Ambassa Jean Ludovic
- District Medical Center, Minkoa-Meyos, Yaoundé, Cameroon.,District Hospital, Cité Verte, Yaoundé, Cameroon
| | | | | | | | | | | | | | - Adèle Douanla
- District Medical Center, Minkoa-Meyos, Yaoundé, Cameroon
| | - Ze Ango
- District Medical Center, Minkoa-Meyos, Yaoundé, Cameroon
| | | | - Joel Tewara Ticha
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Fritz Mbuh Tatah
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Golwa Dinza
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Valentine Nchafor Ndikum
- Department of Pharmacology and African Traditional Medicine, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Dorothy A Fosah
- National Malaria Control Program, Ministry of Public Health, Yaoundé, Cameroon
| | - Jude D Bigoga
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon.,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon.,Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Wilfred F Mbacham
- MARCAD-DELTAS Program, Laboratory for Public Health Research Biotechnologies, University of Yaoundé I, Yaoundé, Cameroon. .,The Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon. .,Cameroon Coalition Against Malaria, P.O. Box 8094, Yaoundé, Cameroon.
| |
Collapse
|
41
|
Omedo I, Bartilol B, Kimani D, Gonçalves S, Drury E, Rono MK, Abdi AI, Almagro-Garcia J, Amato R, Pearson RD, Ochola-Oyier LI, Kwiatkowski D, Bejon P. Spatio-temporal distribution of antimalarial drug resistant gene mutations in a Plasmodium falciparum parasite population from Kilifi, Kenya: A 25-year retrospective study. Wellcome Open Res 2022. [DOI: 10.12688/wellcomeopenres.17656.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Antimalarial drug resistance is a major obstacle to sustainable malaria control. Here we use amplicon sequencing to describe molecular markers of drug resistance in Plasmodium falciparum parasites from Kilifi county in the coastal region of Kenya over a 25-year period. Methods: We performed P. falciparum amplicon sequencing on 1162 malaria-infected blood samples collected between 1994 and 2018 to identify markers of antimalarial drug resistance in the Pfcrt, Pfdhfr, Pfdhps, Pfmdr1, Pfexo, Pfkelch13, plasmepsin 2/3, Pfarps10, Pffd, and Pfmdr2 genes. We further interrogated parasite population structure using a genetic barcode of 101 drug resistance-unrelated single nucleotide polymorphisms (SNPs) distributed across the genomes of 1245 P. falciparum parasites. Results: Two major changes occurred in the parasite population over the 25 years studied. In 1994, approximately 75% of parasites carried the marker of chloroquine resistance, CVIET. This increased to 100% in 1999 and then declined steadily, reaching 6.7% in 2018. Conversely, the quintuple mutation form of sulfadoxine-pyrimethamine resistance increased from 16.7% in 1994 to 83.6% in 2018. Several non-synonymous mutations were identified in the Kelch13 gene, although none of them are currently associated with artemisinin resistance. We observed a temporal increase in the Pfmdr1 NFD haplotype associated with lumefantrine resistance, but observed no evidence of piperaquine resistance. SNPs in other parts of the genome showed no significant temporal changes despite the marked changes in drug resistance loci over this period. Conclusions: We identified substantial changes in molecular markers of P. falciparum drug resistance over 25 years in coastal Kenya, but no associated changes in the parasite population structure.
Collapse
|
42
|
Edgar RCS, Counihan NA, McGowan S, de Koning-Ward TF. Methods Used to Investigate the Plasmodium falciparum Digestive Vacuole. Front Cell Infect Microbiol 2022; 11:829823. [PMID: 35096663 PMCID: PMC8794586 DOI: 10.3389/fcimb.2021.829823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum malaria remains a global health problem as parasites continue to develop resistance to all antimalarials in use. Infection causes clinical symptoms during the intra-erythrocytic stage of the lifecycle where the parasite infects and replicates within red blood cells (RBC). During this stage, P. falciparum digests the main constituent of the RBC, hemoglobin, in a specialized acidic compartment termed the digestive vacuole (DV), a process essential for survival. Many therapeutics in use target one or multiple aspects of the DV, with chloroquine and its derivatives, as well as artemisinin, having mechanisms of action within this organelle. In order to better understand how current therapeutics and those under development target DV processes, techniques used to investigate the DV are paramount. This review outlines the involvement of the DV in therapeutics currently in use and focuses on the range of techniques that are currently utilized to study this organelle including microscopy, biochemical analysis, genetic approaches and metabolomic studies. Importantly, continued development and application of these techniques will aid in our understanding of the DV and in the development of new therapeutics or therapeutic partners for the future.
Collapse
Affiliation(s)
- Rebecca C. S. Edgar
- School of Medicine, Deakin University, Geelong, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Natalie A. Counihan
- School of Medicine, Deakin University, Geelong, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Sheena McGowan
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
- Centre to Impact AMR, Monash University, Monash University, Clayton, VIC, Australia
| | - Tania F. de Koning-Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
43
|
Al-Mekhlafi HM, Madkhali AM, Abdulhaq AA, Atroosh WM, Ghzwani AH, Zain KA, Ghailan KY, Hamali HA, Mobarki AA, Alharazi TH, Eisa ZM, Lau YL. Polymorphism analysis of pfmdr1 gene in Plasmodium falciparum isolates 11 years post-adoption of artemisinin-based combination therapy in Saudi Arabia. Sci Rep 2022; 12:517. [PMID: 35017593 PMCID: PMC8752599 DOI: 10.1038/s41598-021-04450-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
A total of 227 Plasmodium falciparum isolates from Jazan region, southwestern Saudi Arabia were amplified for the P. falciparum multi-drug resistance 1 (pfmdr1) gene to detect point mutations 11 years after the introduction of artemisinin-based combination therapy (ACT) in Saudi Arabia. The pfmdr1 86Y mutation was found in 11.5% (26/227) of the isolates while the N86 wild allele was detected in 88.5%. Moreover, 184F point mutations dominated (86.3%) the instances of pfmdr1 polymorphism while no mutation was observed at codons 1034, 1042 and 1246. Three pfmdr1 haplotypes were identified, NFSND (74.9%), NYSND (13.7%) and YFSND (11.4%). Associations of the prevalence of 86Y mutation and YFSND haplotype with participants' nationality, residency and parasitaemia level were found to be significant (P < 0.05). The findings revealed significant decline in the prevalence of the pfmdr1 86Y mutation in P. falciparum isolates from Jazan region over a decade after the implementation of ACT treatment. Moreover, the high prevalence of the NFSND haplotype might be indicative of the potential emergence of CQ-sensitive but artemether-lumefantrine-resistant P. falciparum strains since the adoption of ACT. Therefore, continuous monitoring of the molecular markers of antimalarial drug resistance in Jazan region is highly recommended.
Collapse
Affiliation(s)
- Hesham M Al-Mekhlafi
- Medical Research Centre, Jazan University, Jazan, Kingdom of Saudi Arabia.
- Vector-Borne Diseases Research Group, Jazan University, Jazan, Kingdom of Saudi Arabia.
- Department of Parasitology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Yemen.
| | - Aymen M Madkhali
- Medical Research Centre, Jazan University, Jazan, Kingdom of Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | - Ahmed A Abdulhaq
- Vector-Borne Diseases Research Group, Jazan University, Jazan, Kingdom of Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Wahib M Atroosh
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Department of Microbiology and Parasitology, Faculty of Medicine and Health Sciences, University of Aden, Aden, Yemen
| | | | - Khalid Ammash Zain
- Medical Research Centre, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Khalid Y Ghailan
- Vector-Borne Diseases Research Group, Jazan University, Jazan, Kingdom of Saudi Arabia
- Faculty of Public Health and Tropical Medicine, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Hassan A Hamali
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Abdullah A Mobarki
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Talal H Alharazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail, Kingdom of Saudi Arabia
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, Taiz University, Taiz, Yemen
| | - Zaki M Eisa
- Saudi Centre for Disease Prevention and Control, Ministry of Health, Jazan, Kingdom of Saudi Arabia
| | - Yee-Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Afolabi R, Chinedu S, Ajamma Y, Adam Y, Koenig R, Adebiyi E. Computational identification of Plasmodium falciparum RNA pseudouridylate synthase as a viable drug target, its physicochemical properties, 3D structure prediction and prediction of potential inhibitors. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 97:105194. [PMID: 34968763 DOI: 10.1016/j.meegid.2021.105194] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
The increased resistance to the currently effective antimalarial drugs against Plasmodium falciparum has necessitated the development of new drugs for malaria treatment. Many proteins have been predicted using various means as potential drug targets for the treatment of the P. falciparum malaria infection. Meanwhile, only a few studies went on to predict the 3-dimensional (3D) structure of potential target. Therefore, this study aimed to predict potential antimalarial drug targets against the deadliest malaria parasite P. falciparum as well as to determine the 3D structure and possible inhibitors of one of the targets. We employed machine learning approach to predict suitable drug targets in P. falciparum. Five of the predicted protein targets were considered as potential drug targets as they were non-homologous to their human counterparts. Out of these, we determined the physicochemical properties, predicted the 3D structure and carried out docking-based virtual screening of P. falciparum RNA pseudouridylate synthase, putative (PfRPuSP). The PfRPuSP was one of the potential five target proteins. Homology modelling and the ab initio methods were used to predict the 3D structure of PfRPuSP. Then, a compound library of 5621 molecules was constructed from PubChem and ChEMBL databases using 5-fluorouridine as the control inhibitor. Docking-based virtual screening was performed using Autodock 4.2 and Autodock Vina to select compounds with high binding affinity. A total of 11 compounds were selected based on their binding energies from 881 compounds which were manually examined after docking. Seven of the 11 compounds that exhibited remarkable interactions with the residues in the active sites of PfRPuSP were analysed. These compounds performed favourably when compared to the control inhibitor and predicted to bind better than 5-fluorouridine. These seven compounds are suggested as new potential lead structures for antimalarial treatment.
Collapse
Affiliation(s)
- Rufus Afolabi
- Covenant University Bioinformatics Research, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria; Department of Biochemistry, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria.
| | - Shalom Chinedu
- Department of Biochemistry, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria.
| | - Yvonne Ajamma
- Covenant University Bioinformatics Research, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria.
| | - Yagoub Adam
- Covenant University Bioinformatics Research, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria.
| | - Rainer Koenig
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Systems Biology of Sepsis, Kollegiengasse 10, 07743 Jena, Germany.
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria; Department of Computer and Information Sciences, Covenant University, Km 10 Idiroko Road, P.M.B., 1023 Ota, Ogun State, Nigeria; Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), G200, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
45
|
Historical experiences on mass drug administration for malaria control and elimination, its challenges and China's experience: a narrative review. Acta Trop 2022; 225:106209. [PMID: 34687651 DOI: 10.1016/j.actatropica.2021.106209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 11/23/2022]
Abstract
After decades of efforts, malaria is still a major public health problem in many countries, especially in sub-Saharan Africa. Mass drug administration (MDA) has been one of the interventions used to control malaria. China, the largest and most populous country in the world, has recently achieved malaria elimination with MDA within its strategy. Therefore, knowing the history of the use of MDA, and its advantages and challenges are crucial to better implement MDA as a component of malaria control and elimination strategy. This narrative review focused on the use of MDA from the past to present, the experiences of successful and failed MDA interventions to control malaria in Africa, Asia, and South Pacific region, the challenges faced, as well as China's experience in malaria control and elimination. A direct search using key words and phrases was conducted using the web search engines Google and Google Scholar, peer-reviewed journal websites and PubMed database to mainly screen articles on MDA studies with positive and negative results, the World Health Organization guidelines, and other MDA-related reports. We also summarized our historical field experiences on MDA in malaria control to provide informed perspective on the challenges of MDA. Following the spirit of innovation, a comprehensive strategy with MDA at its core and each additional measure compensating one another's shortage based on different malaria transmission settings and stratification could be the highlight of future malaria control and elimination strategy to help achieve the vision of a malaria-free world.
Collapse
|
46
|
Owoloye A, Olufemi M, Idowu ET, Oyebola KM. Prevalence of potential mediators of artemisinin resistance in African isolates of Plasmodium falciparum. Malar J 2021; 20:451. [PMID: 34856982 PMCID: PMC8638531 DOI: 10.1186/s12936-021-03987-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022] Open
Abstract
Background The devastating public health impact of malaria has prompted the need for effective interventions. Malaria control gained traction after the introduction of artemisinin-based combination therapy (ACT). However, the emergence of artemisinin (ART) partial resistance in Southeast Asia and emerging reports of delayed parasite sensitivity to ACT in African parasites signal a gradual trend towards treatment failure. Monitoring the prevalence of mutations associated with artemisinin resistance in African populations is necessary to stop resistance in its tracks. Mutations in Plasmodium falciparum genes pfk13, pfcoronin and pfatpase6 have been linked with ART partial resistance. Methods Findings from published research articles on the prevalence of pfk13, pfcoronin and pfatpase6 polymorphisms in Africa were collated. PubMed, Embase and Google Scholar were searched for relevant articles reporting polymorphisms in these genes across Africa from 2014 to August 2021, for pfk13 and pfcoronin. For pfatpase6, relevant articles between 2003 and August 2021 were retrieved. Results Eighty-seven studies passed the inclusion criteria for this analysis and reported 742 single nucleotide polymorphisms in 37,864 P. falciparum isolates from 29 African countries. Five validated-pfk13 partial resistance markers were identified in Africa: R561H in Rwanda and Tanzania, M476I in Tanzania, F446I in Mali, C580Y in Ghana, and P553L in an Angolan isolate. In Tanzania, three (L263E, E431K, S769N) of the four mutations (L263E, E431K, A623E, S769N) in pfatpase6 gene associated with high in vitro IC50 were reported. pfcoronin polymorphisms were reported in Senegal, Gabon, Ghana, Kenya, and Congo, with P76S being the most prevalent mutation. Conclusions This meta-analysis provides an overview of the prevalence and widespread distribution of pfk13, pfcoronin and pfatpase6 mutations in Africa. Understanding the phenotypic consequences of these mutations can provide information on the efficacy status of artemisinin-based treatment of malaria across the continent. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03987-6.
Collapse
Affiliation(s)
- Afolabi Owoloye
- Genomic Research in Biomedicine Laboratory, Biochemistry and Nutrition Department, Nigerian Institute of Medical Research, Lagos, Nigeria.,Parasitology and Bioinformatics Unit, Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
| | - Michael Olufemi
- Genomic Research in Biomedicine Laboratory, Biochemistry and Nutrition Department, Nigerian Institute of Medical Research, Lagos, Nigeria.,Parasitology and Bioinformatics Unit, Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
| | - Emmanuel T Idowu
- Parasitology and Bioinformatics Unit, Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
| | - Kolapo M Oyebola
- Genomic Research in Biomedicine Laboratory, Biochemistry and Nutrition Department, Nigerian Institute of Medical Research, Lagos, Nigeria. .,Parasitology and Bioinformatics Unit, Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria. .,Sickle Cell Branch, National Heart Lung and Blood Institute, US National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Chidimatembue A, Svigel SS, Mayor A, Aíde P, Nhama A, Nhamussua L, Nhacolo A, Bassat Q, Salvador C, Enosse S, Saifodine A, De Carvalho E, Candrinho B, Zulliger R, Goldman I, Udhayakumar V, Lucchi NW, Halsey ES, Macete E. Molecular surveillance for polymorphisms associated with artemisinin-based combination therapy resistance in Plasmodium falciparum isolates collected in Mozambique, 2018. Malar J 2021; 20:398. [PMID: 34641867 PMCID: PMC8507114 DOI: 10.1186/s12936-021-03930-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/26/2021] [Indexed: 11/10/2022] Open
Abstract
Background Due to the threat of emerging anti-malarial resistance, the World Health Organization recommends incorporating surveillance for molecular markers of anti-malarial resistance into routine therapeutic efficacy studies (TESs). In 2018, a TES of artemether-lumefantrine (AL) and artesunate-amodiaquine (ASAQ) was conducted in Mozambique, and the prevalence of polymorphisms in the pfk13, pfcrt, and pfmdr1 genes associated with drug resistance was investigated. Methods Children aged 6–59 months were enrolled in four study sites. Blood was collected and dried on filter paper from participants who developed fever within 28 days of initial malaria treatment. All samples were first screened for Plasmodium falciparum using a multiplex real-time PCR assay, and polymorphisms in the pfk13, pfcrt, and pfmdr1 genes were investigated by Sanger sequencing. Results No pfk13 mutations, associated with artemisinin partial resistance, were observed. The only pfcrt haplotype observed was the wild type CVMNK (codons 72–76), associated with chloroquine sensitivity. Polymorphisms in pfmdr1 were only observed at codon 184, with the mutant 184F in 43/109 (39.4%) of the samples, wild type Y184 in 42/109 (38.5%), and mixed 184F/Y in 24/109 (22.0%). All samples possessed N86 and D1246 at these two codons. Conclusion In 2018, no markers of artemisinin resistance were documented. Molecular surveillance should continue to monitor the prevalence of these markers to inform decisions on malaria treatment in Mozambique.
Collapse
Affiliation(s)
| | - Samaly S Svigel
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Alfredo Mayor
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología Y Salud Pública (CIBERESP), Madrid, Spain
| | - Pedro Aíde
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Abel Nhama
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Lídia Nhamussua
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Arsénio Nhacolo
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.,Pediatric Infectious Diseases Unit, Pediatrics Department, Hospital Sant Joan de Déu (University of Barcelona), Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología Y Salud Pública (CIBERESP), Madrid, Spain
| | - Crizólgo Salvador
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Sónia Enosse
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Abuchahama Saifodine
- United States President's Malaria Initiative, US Agency for International Development, Maputo, Mozambique
| | | | - Baltazar Candrinho
- National Malaria Control Programme, Ministry of Health, Maputo, Mozambique
| | - Rose Zulliger
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA.,United States President's Malaria Initiative, Centers for Disease Control and Prevention, Maputo, Mozambique
| | - Ira Goldman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Venkatachalam Udhayakumar
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Naomi W Lucchi
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Eric S Halsey
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA.,United States President's Malaria Initiative, Atlanta, GA, USA
| | - Eusébio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique. .,National Directorate of Public Health, Ministry of Health, Maputo, Mozambique.
| |
Collapse
|
48
|
Noreen N, Ullah A, Salman SM, Mabkhot Y, Alsayari A, Badshah SL. New insights into the spread of resistance to artemisinin and its analogues. J Glob Antimicrob Resist 2021; 27:142-149. [PMID: 34517141 DOI: 10.1016/j.jgar.2021.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 08/19/2021] [Accepted: 09/02/2021] [Indexed: 12/22/2022] Open
Abstract
Plasmodium falciparum, the causative agent of malaria, has been developing resistance to several drugs worldwide for more than five decades. Initially, resistance was against drugs such as chloroquine, pyrimethamine, sulfadoxine, mefloquine and quinine. Research studies are now reporting parasites with resistance to the most effective and novel drug used against malaria infection worldwide, namely artemisinin. For this reason, the first-line treatment strategy of artemisinin-based combination therapy is becoming unsuccessful in areas where drug resistance is highly prevalent. The increase in artemisinin-resistant P. falciparum strains has threatened international efforts to eliminate malarial infections and to reduce the disease burden. Detection of several phenotypes that display artemisinin resistance, specification of basic genetic factors, the discovery of molecular pathways, and evaluation of its clinical outcome are possible by the current series of research on genomics and transcriptomic levels in Asia and Africa. In artemisinin resistance, slow parasite clearance among malaria-infected patients and enhanced in vitro survival of parasites occurs at the early ring stage. This resistance is due to single nucleotide polymorphisms within the Kelch 13 gene of the parasite and is related to significantly upregulated resistance signalling pathways; thus, the pro-oxidant action of artemisinins can be antagonised. New strategies are required to halt the spread of artemisinin-resistant malarial parasites.
Collapse
Affiliation(s)
- Noreen Noreen
- Department of Chemistry, Islamia College University, Peshawar 25120, Pakistan
| | - Asad Ullah
- Department of Chemistry, Islamia College University, Peshawar 25120, Pakistan
| | | | - Yahia Mabkhot
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia.
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Syed Lal Badshah
- Department of Chemistry, Islamia College University, Peshawar 25120, Pakistan.
| |
Collapse
|
49
|
de Haan F, Bolarinwa OA, Guissou R, Tou F, Tindana P, Boon WPC, Moors EHM, Cheah PY, Dhorda M, Dondorp AM, Ouedraogo JB, Mokuolu OA, Amaratunga C. To what extent are the antimalarial markets in African countries ready for a transition to triple artemisinin-based combination therapies? PLoS One 2021; 16:e0256567. [PMID: 34464398 PMCID: PMC8407563 DOI: 10.1371/journal.pone.0256567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/09/2021] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Triple artemisinin-based combination therapies (TACTs) are being developed as a response to artemisinin and partner drug resistance in the treatment of falciparum malaria in Southeast Asia. In African countries, where current artemisinin-based combination therapies (ACTs) are still effective, TACTs have the potential to benefit the larger community and future patients by mitigating the risk of drug resistance. This study explores the extent to which the antimalarial drug markets in African countries are ready for a transition to TACTs. METHODS A qualitative study was conducted in Nigeria and Burkina Faso and comprised in-depth interviews (n = 68) and focus group discussions (n = 11) with key actor groups in the innovation system of antimalarial therapies. RESULTS Evidence of ACT failure in African countries and explicit support for TACTs by the World Health Organization (WHO) and international funders were perceived important determinants for the market prospects of TACTs in Nigeria and Burkina Faso. At the country level, slow regulatory and implementation procedures were identified as potential barriers towards rapid TACTs deployment. Integrating TACTs in public sector distribution channels was considered relatively straightforward. More challenges were expected for integrating TACTs in private sector distribution channels, which are characterized by patient demand and profit motives. Finally, several affordability and acceptability issues were raised for which ACTs were suggested as a benchmark. CONCLUSION The market prospects of TACTs in Nigeria and Burkina Faso will depend on the demonstration of the added value of TACTs over ACTs, their advocacy by the WHO, the inclusion of TACTs in financial and regulatory arrangements, and their alignment with current distribution and deployment practices. Further clinical, health-economic and feasibility studies are required to inform decision makers about the broader implications of a transition to TACTs in African counties. The recent reporting of artemisinin resistance and ACT failure in Africa might change important determinants of the market readiness for TACTs.
Collapse
Affiliation(s)
- Freek de Haan
- Copernicus Institute of Sustainable Development, Utrecht University, Utrecht, The Netherlands
| | | | - Rosemonde Guissou
- Institut de Recherche en Sciences de la Sante, Bobo-Dioulasso, Burkina Faso
| | - Fatoumata Tou
- Institut des Sciences et Techniques, Bobo-Dioulasso, Burkina Faso
| | - Paulina Tindana
- School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Wouter P. C. Boon
- Copernicus Institute of Sustainable Development, Utrecht University, Utrecht, The Netherlands
| | - Ellen H. M. Moors
- Copernicus Institute of Sustainable Development, Utrecht University, Utrecht, The Netherlands
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Arjen M. Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jean Bosco Ouedraogo
- Institut de Recherche en Sciences de la Sante, Bobo-Dioulasso, Burkina Faso
- Institut des Sciences et Techniques, Bobo-Dioulasso, Burkina Faso
| | | | - Chanaki Amaratunga
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Discrete Survival Model Analysis of Plasmodium falciparum Response to Artemisinin-Based Combination Therapies among Children in Regions of Varying Malaria Transmission in Cameroon. Pathogens 2021; 10:pathogens10091106. [PMID: 34578139 PMCID: PMC8465257 DOI: 10.3390/pathogens10091106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022] Open
Abstract
The need to monitor changes in parasite clearance following treatment with artemisinin-based combination therapies (ACTs) is important in the containment of drug resistance. This study aimed to model Plasmodium falciparum response to ACTs among children in two different transmission settings (Mutengene and Garoua) in Cameroon. Using the step function, a discrete-time survival model was fitted with all the covariates included that might play a role in parasite clearance. The probability of clearing parasites within 24 h following treatment was 21.6% and 70.3% for younger children aged 6 to 59 months and 29.3% and 59.8% for older children aged 60 to 120 months in Mutengene and Garoua, respectively. After two days of treatment, the conditional probability of clearing parasites given that they were not cleared on day 1 was 76.7% and 96.6% for children aged 6–59 months and 83.1% and 93.5% for children aged 60–120 months in Mutengene and Garoua, respectively. The model demonstrated that the ecological setting, age group and pretreatment serum levels of creatinine and alanine aminotransferase were the main factors that significantly influenced parasite clearance in vivo after administration of ACTs (p < 0.05). The findings highlight the need for further investigations on host differential response to ACTs in current practice.
Collapse
|