1
|
Bohissou FET, Sondo P, Inoue J, Rouamba T, Kaboré B, Nassa GJW, Kambou AES, Traoré TE, Asua V, Borrmann S, Tinto H, Held J. Evolution of Pfdhps and Pfdhfr mutations before and after adopting seasonal malaria chemoprevention in Nanoro, Burkina Faso. Sci Rep 2024; 14:24224. [PMID: 39414909 PMCID: PMC11484836 DOI: 10.1038/s41598-024-75369-2] [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: 06/04/2024] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
Seasonal Malaria Chemoprevention consisting of monthly administration of amodiaquine/sulfadoxine-pyrimethamine to children aged 3-59 months during the transmission season could promote SP-resistance. Mutations in dihydrofolate reductase (Pfdhfr) and dihydropteroate synthase (Pfdhps) genes were assessed before and after SMC adoption in Burkina Faso. A total of 769 dried blood spots were selected from studies conducted in Nanoro, Burkina Faso, between 2010 and 2020. Of those, 299 were pre-SMC (2010-2012) and 470 were post-SMC-samples. Pfdhps and Pfdhfr genes were PCR-amplified and sequenced. A systematic review/meta-analysis of published studies conducted in Burkina Faso (2009-2023) was additionally performed. In Nanoro, the prevalence of Pfdhfr triple mutations (CIRNI) rose from 43.6% pre-SMC to 89.4% post-SMC (p < 0.0001). There was no mutation in Pfdhfr 164 and Pfdhps 540; Pfdhps A437G mutation increased from 63.9% (2010-2012) to 84.7% (2020) (p < 0.0001). The VAGKGS haplotype was 2.8% (2020). Pfdhfr/Pfdhps quintuple mutant IRN-436A437G rose from 18.6% (2010-2012) to 58.3% (2020) (p < 0.0001). Meta-analysis results from Burkina Faso showed an increase in mutations at Pfdhfr N51I, C59R, S108N, and Pfdhps A437G after SMC adoption. Post-SMC, the pyrimethamine-resistance marker prevalence increased, while the sulfadoxine-resistance marker prevalence remained stable. Detection of emerging PfdhpsVAGKGS haplotypes in 2020 underscores the importance of continuous SP-resistance monitoring.
Collapse
Affiliation(s)
- Francis Emmanuel Towanou Bohissou
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
- Centre de Recherche Entomologique de Cotonou (CREC), Cotonou, Benin
| | - Paul Sondo
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Juliana Inoue
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Berenger Kaboré
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | | | - A Elisée Sié Kambou
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Tiampan Edwig Traoré
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Victor Asua
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Steffen Borrmann
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, (CERMEL), Lambaréné, Gabon
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso.
| | - Jana Held
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.
- Centre de Recherches Médicales de Lambaréné, (CERMEL), Lambaréné, Gabon.
| |
Collapse
|
2
|
Huang S, Baker K, Ibinaiye T, Oresanya O, Nnaji C, Richardson S. Impact of seasonal malaria chemoprevention based on the number of medicines doses received on malaria burden among children aged 3-59 months in Nigeria: A propensity score-matched analysis. Trop Med Int Health 2024; 29:668-679. [PMID: 38842452 DOI: 10.1111/tmi.14019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
BACKGROUND Seasonal malaria chemoprevention using sulfadoxine-pyrimethamine plus amodiaquine (sulfadoxine-pyrimethamine plus amodiaquine on Day 1 and amodiaquine on both Day 2 and Day 3) is delivered to children aged 3-59 months in areas of highly season malaria transmission. While the overall population-level impact of seasonal malaria chemoprevention on malaria control has been documented in various countries and time periods, there is no clear evidence regarding seasonal malaria chemoprevention impact based on the number of medicine doses children receive in one cycle in routine programmatic conditions. METHODS Data were extracted from Nigeria's routinely collected seasonal malaria chemoprevention end-of-round coverage surveys (2021, 2022). We matched seasonal malaria chemoprevention-targeted children who received specific numbers of seasonal malaria chemoprevention medicines with those who did not receive any doses of seasonal malaria chemoprevention medicines (non-sulfadoxine-pyrimethamine plus amodiaquine) using multiple sets of propensity score matches. We performed multilevel logistic regression for each matched group to evaluate the association between the number of doses of seasonal malaria chemoprevention medicines and monthly confirmed malaria cases (caregiver-reported malaria infection diagnosed by rapid diagnostic test at a health facility following the penultimate cycle of seasonal malaria chemoprevention). RESULTS Among 21,621 SMC-targeted children, 9.7% received non-sulfadoxine-pyrimethamine plus amodiaquine, 0.5% received only Day 1 sulfadoxine-pyrimethamine plus amodiaquine, 1.0% received Day 1 sulfadoxine-pyrimethamine plus amodiaquine and either Day 2 amodiaquine or Day 3 amodiaquine (sulfadoxine-pyrimethamine plus amodiaquine + amodiaquine), and 88.8% received Day 1 sulfadoxine-pyrimethamine plus amodiaquine and both Day 2 and Day 3 amodiaquine (sulfadoxine-pyrimethamine plus amodiaquine + amodiaquine + amodiaquine). Children receiving only Day 1 sulfadoxine-pyrimethamine plus amodiaquine did not have significant lower odds of rapid diagnostic tests-confirmed malaria than those receiving non-sulfadoxine-pyrimethamine plus amodiaquine (OR 0.77, 0.42-1.42). However, children receiving sulfadoxine-pyrimethamine plus amodiaquine + amodiaquine had significantly lower odds of rapid diagnostic tests-confirmed malaria than those receiving non-sulfadoxine-pyrimethamine plus amodiaquine (OR 0.42, 95% CI 0.28-0.63). Similarly, children receiving sulfadoxine-pyrimethamine plus amodiaquine + amodiaquine + amodiaquine also had significantly lower odds of rapid diagnostic test-confirmed malaria than those receiving non-sulfadoxine-pyrimethamine plus amodiaquine (OR 0.54, 95% CI 0.47-0.62). CONCLUSION Adherence to at least one daily dose of amodiaquine administration following receipt of Day 1 sulfadoxine-pyrimethamine plus amodiaquine by eligible children is crucial to ensure the effectiveness of seasonal malaria chemoprevention. This demonstrates the importance of enhancing caregiver awareness regarding the importance of amodiaquine and identifying barriers toward amodiaquine administration at the community level.
Collapse
Affiliation(s)
- Sikai Huang
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Kevin Baker
- Malaria Consortium UK, The Green House, London, UK
- Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | | | | | - Chuks Nnaji
- Malaria Consortium UK, The Green House, London, UK
| | - Sol Richardson
- Vanke School of Public Health, Tsinghua University, Beijing, China
| |
Collapse
|
3
|
Faye SLB, Lugand MM, Offianan AT, Dossou-Yovo A, Kouadio DKM, Pinto F. Field testing of user-friendly perennial malaria chemoprevention packaging in Benin, Côte d'Ivoire and Mozambique. Malar J 2024; 23:157. [PMID: 38773567 PMCID: PMC11106929 DOI: 10.1186/s12936-024-04977-0] [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: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Perennial malaria chemoprevention (PMC) aims to protect children at risk from severe malaria by the administration of anti-malarial drugs to children of defined ages throughout the year. Sulfadoxine-pyrimethamine (SP) has been widely used for chemoprevention in Africa and a child-friendly dispersible tablet formulation has recently become available. METHODS This qualitative non-interventional observational study was conducted in Benin, Côte d'Ivoire, and Mozambique between February and June 2022. Prototype blister packs, dispensing boxes and job aids designed to support dispersible SP deployment for PMC were evaluated using focus group discussions (FGD) and semi-structured in-depth individual interviews (IDI) with health authorities, health personnel, community health workers (CHWs) and caregivers. The aim was to evaluate knowledge and perceptions of malaria and chemoprevention, test understanding of the tools and identify gaps in understanding, satisfaction, user-friendliness and acceptability, and assess the potential role of CHWs in PMC implementation. Interviews were transcribed and imported to ATLAS.ti for encoding and categorization. Thematic content analysis used deductive and inductive coding with cross-referencing of findings between countries and participants to enrich data interpretation. Continuous comparison across the IDI and FGD permitted iterative, collaborative development of materials. RESULTS Overall, 106 participants completed IDIs and 70 contributed to FGDs. Malaria was widely recognised as the most common disease affecting children, and PMC was viewed as a positive intervention to support child health. The role of CHWs was perceived differently by the target groups, with caregivers appreciating their trusted status in the community, whereas health authorities preferred clinic-based deployment of PMC by health professionals. Empirical testing of the prototype blister packs, dispensing boxes and job aids highlighted the context-specific expectations of respondents, such as familiar situations and equipment, and identified areas of confusion or low acceptance. A key finding was the need for a clear product identity reflecting malaria. CONCLUSION Simple modifications profoundly affected the perception of PMC and influenced acceptability. Iterative quantitative investigation resulted in PMC-specific materials suited to the local context and socio-cultural norms of the target population with the aim of increasing access to chemoprevention in children most at risk of severe malaria.
Collapse
Affiliation(s)
- Sylvain Landry Birane Faye
- Laboratoire de Sociologie, Anthropologie, Psychologie (LASAP), Department of Sociology, Cheikh Anta DIOP University (UCAD), Dakar, Senegal
| | - Maud Majeres Lugand
- Medicines for Malaria Venture, 20 Route de Pré-Bois, PO Box 1826, 1215, Geneva 15, Switzerland.
| | - André Touré Offianan
- Department of Parasitology & Mycology, Institut Pasteur of Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Aurélie Dossou-Yovo
- Directorate of Health Training and Research, Ministry of Health, Cotonou, Benin
| | - Dieudonné Kouakou M'Bra Kouadio
- Département d'Anthropologie et de Sociologie/Centre de Recherche Pour le Développement, Université Alassane Ouattara, Bouaké, Côte d'Ivoire
| | | |
Collapse
|
4
|
Bisanzio D, Keita MS, Camara A, Guilavogui T, Diallo T, Barry H, Preston A, Bangoura L, Mbounga E, Florey LS, Taton JL, Fofana A, Reithinger R. Malaria trends in districts that were targeted and not-targeted for seasonal malaria chemoprevention in children under 5 years of age in Guinea, 2014-2021. BMJ Glob Health 2024; 9:e013898. [PMID: 38413098 PMCID: PMC10900330 DOI: 10.1136/bmjgh-2023-013898] [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: 09/07/2023] [Accepted: 01/26/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Seasonal malaria chemoprevention (SMC) is a main intervention to prevent and reduce childhood malaria. Since 2015, Guinea has implemented SMC targeting children aged 3-59 months (CU5) in districts with high and seasonal malaria transmission. OBJECTIVE We assessed the programmatic impact of SMC in Guinea's context of scaled up malaria intervention programming by comparing malaria-related outcomes in 14 districts that had or had not been targeted for SMC. METHODS Using routine health management information system data, we compared the district-level monthly test positivity rate (TPR) and monthly uncomplicated and severe malaria incidence for the whole population and disaggregated age groups (<5 years and ≥5 years of age). Changes in malaria indicators through time were analysed by calculating the district-level compound annual growth rate (CAGR) from 2014 to 2021; we used statistical analyses to describe trends in tested clinical cases, TPR, uncomplicated malaria incidence and severe malaria incidence. RESULTS The CAGR of TPR of all age groups was statistically lower in SMC (median=-7.8%) compared with non-SMC (median=-3.0%) districts. Similarly, the CAGR in uncomplicated malaria incidence was significantly lower in SMC (median=1.8%) compared with non-SMC (median=11.5%) districts. For both TPR and uncomplicated malaria incidence, the observed difference was also significant when age disaggregated. The CAGR of severe malaria incidence showed that all age groups experienced a decline in severe malaria in both SMC and non-SMC districts. However, this decline was significantly higher in SMC (median=-22.3%) than in non-SMC (median=-5.1%) districts for the entire population, as well as both CU5 and people over 5 years of age. CONCLUSION Even in an operational programming context, adding SMC to the malaria intervention package yields a positive epidemiological impact and results in a greater reduction in TPR, as well as the incidence of uncomplicated and severe malaria in CU5.
Collapse
Affiliation(s)
- Donal Bisanzio
- RTI International, Washington, District of Columbia, USA
| | | | - Alioune Camara
- Programme National de la Lutte contre le Paludisme, Ministère de la Santé et de l'Hygiène Publique, Conakry, Guinea
| | | | | | | | | | - Lamine Bangoura
- President's Malaria Initiative, US Agency for International Development, Conakry, Guinea
| | - Eliane Mbounga
- President's Malaria Initiative, US Agency for International Development, Conakry, Guinea
| | - Lia S Florey
- US Agency for International Development, Washington, District of Columbia, USA
| | | | | | | |
Collapse
|
5
|
Mwaiswelo R, Ngasala B, Chaky F, Molteni F, Mohamed A, Lazaro S, Samwel B, Mmbando BP. Dihydroartemisinin-piperaquine effectiveness for seasonal malaria chemoprevention in settings with extended seasonal malaria transmission in Tanzania. Sci Rep 2024; 14:2143. [PMID: 38273019 PMCID: PMC10810795 DOI: 10.1038/s41598-024-52706-z] [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: 08/29/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Abstract
Effectiveness of dihydroartemisinin-piperaquine (DP) as seasonal malaria chemoprevention (SMC) was assessed in Nanyumbu and Masasi Districts. Between March and June 2021, children aged 3-59 months were enrolled in a cluster randomized study. Children in the intervention clusters received a monthly, 3-days course of DP for three consecutive months regardless of malaria infection status, and those in the control clusters received no intervention. Malaria infection was assessed at before the first-round and at 7 weeks after the third-round of DP in both arms. Malaria prevalence after the third-round of DP administration was the primary outcome. Chi-square tests and logistic regression model were used to compare proportions and adjust for explanatory variables. Before the intervention, malaria prevalence was 13.7% (161/1171) and 18.2% (212/1169) in the intervention and control clusters, respectively, p < 004. Malaria prevalence declined to 5.8% (60/1036) in the intervention clusters after three rounds of DP, and in the control clusters it declined to 9.3% (97/1048), p = 0.003. Unadjusted and adjusted prevalence ratios between the intervention and control arms were 0.42 (95%CI 0.32-0.55, p < 0.001) and 0.77 (95%CI 0.53-1.13, p = 0.189), respectively. SMC using DP was effective for control of malaria in the two Districts.Trial registration: NCT05874869, https://clinicaltrials.gov/ 25/05/2023.
Collapse
Affiliation(s)
- Richard Mwaiswelo
- Department of Microbiology, Immunology, and Parasitology, Faculty of Medicine, Hubert Kairuki Memorial University, Dar es Salaam, Tanzania.
| | - Billy Ngasala
- Department of Medical Parasitology and Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Frank Chaky
- National Malaria Control Programme, Ministry of Health, Dodoma, Tanzania
| | | | - Ally Mohamed
- National Malaria Control Programme, Ministry of Health, Dodoma, Tanzania
| | - Samwel Lazaro
- National Malaria Control Programme, Ministry of Health, Dodoma, Tanzania
| | - Bushukatale Samwel
- Department of Medical Parasitology and Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Bruno P Mmbando
- National Institute for Medical Research, Tanga Research Centre, Tanga, Tanzania
| |
Collapse
|
6
|
Khan J, Suau Sans M, Okot F, Rom Ayuiel A, Magoola J, Rassi C, Huang S, Mubiru D, Bonnington C, Baker K, Ahmed J, Nnaji C, Richardson S. A quasi-experimental study to estimate effectiveness of seasonal malaria chemoprevention in Aweil South County in Northern Bahr El Ghazal, South Sudan. Malar J 2024; 23:33. [PMID: 38267985 PMCID: PMC10810022 DOI: 10.1186/s12936-024-04853-x] [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: 06/12/2023] [Accepted: 01/16/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Seasonal malaria chemoprevention (SMC) is an effective intervention to prevent malaria in children in locations where the burden of malaria is high and transmission is seasonal. There is growing evidence suggesting that SMC with sulfadoxine-pyrimethamine and amodiaquine can retain its high level of effectiveness in East and Southern Africa despite resistance concerns. This study aims to generate evidence on the effectiveness of SMC when delivered under programmatic conditions in an area with an unknown anti-malarial drug resistance profile in the Northern Bahr el-Ghazal region of South Sudan. METHODS A non-randomized quasi experimental study was conducted to compare an intervention county with a control county. Five monthly SMC cycles were delivered between July and November 2022, targeting about 19,000 children 3-59 months old. Data were obtained from repeated cross-sectional household surveys of caregivers of children aged 3-59 months using cluster sampling. Wave 1 survey took place in both counties before SMC implementation; Waves 2 and 3 took place after the second and fourth monthly SMC cycles. Difference-in-differences analyses were performed by fitting logistic regression models with interactions between county and wave. RESULTS A total of 2760 children were sampled in the study across the three survey waves in both study counties. Children in the intervention arm had 70% lower odds of caregiver-reported fever relative to those in the control arm during the one-month period prior to Wave 2 (OR: 0.30, 95% CI 0.12-0.70, p = 0.003), and 37% lower odds in Wave 3 (OR: 0.63, 95% CI 0.22-1.59, p = 0.306) after controlling for baseline difference between counties in Wave 1. Odds of caregiver-reported RDT-confirmed malaria were 82% lower in the previous 1-month period prior to Wave 2 (OR: 0.18, 95% CI 0.07-0.49, p = 0.001) and Wave 3 (OR: 0.18, 95% CI 0.06-0.54, p = 0.003). CONCLUSION These results show high effectiveness of SMC using SPAQ in terms of reducing malaria disease during the high transmission season in children 3-59 month. Despite the promising results, additional evidence and insights from chemoprevention efficacy cohort studies, and analyses of relevant resistance markers, are required to assess the suitability of SMC for this specific context.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sikai Huang
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | | | | | - Kevin Baker
- Malaria Consortium, London, UK
- Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | | | | | - Sol Richardson
- Malaria Consortium, London, UK
- Vanke School of Public Health, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Thwing J, Williamson J, Cavros I, Gutman JR. Systematic Review and Meta-Analysis of Seasonal Malaria Chemoprevention. Am J Trop Med Hyg 2024; 110:20-31. [PMID: 38081050 PMCID: PMC10793029 DOI: 10.4269/ajtmh.23-0481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 10/23/2023] [Indexed: 01/05/2024] Open
Abstract
Seasonal malaria chemoprevention (SMC) for children under 5 years of age for up to four monthly cycles during malaria transmission season was recommended by the WHO in 2012 and has been implemented in 13 countries in the Sahel, reaching more than 30 million children annually. Malaria control programs implementing SMC have asked the WHO to consider expanding the age range or number of monthly cycles. We conducted a systematic review and meta-analysis of SMC among children up to 15 years of age and up to six monthly cycles. Twelve randomized studies were included, with outcomes stratified by age (< 5/≥ 5 years), by three or four versus five or six cycles, and by drug where possible. Drug regimens included sulfadoxine-pyrimethamine + amodiaquine, amodiaquine-artesunate, and sulfadoxine-pyrimethamine + artesunate. Included studies were all conducted in Sahelian countries in which high-grade resistance to sulfadoxine-pyrimethamine was rare and in zones with parasite prevalence ranging from 1% to 79%. Seasonal malaria chemoprevention resulted in substantial reductions in uncomplicated malaria incidence measured during that transmission season (rate ratio: 0.27, 95% CI: 0.25-0.29 among children < 5 years; rate ratio: 0.27, 95% CI: 0.25-0.30 among children ≥ 5 years) and in the prevalence of malaria parasitemia measured within 4-6 weeks from the final SMC cycle (risk ratio: 0.38, 95% CI: 0.34-0.43 among children < 5 years; risk ratio: 0.23, 95% CI: 0.11-0.48 among children ≥ 5 years). In high-transmission zones, SMC resulted in a moderately reduced risk of any anemia (risk ratio: 0.77, 95% CI: 0.72-0.83 among children < 5 years; risk ratio: 0.70, 95% CI: 0.52-0.95 among children ≥ 5 years [one study]). Children < 10 years of age had a moderate reduction in severe malaria (risk ratio: 0.53, 95% CI: 0.37-0.76) but no evidence of a mortality reduction. The evidence suggests that in areas in which sulfadoxine-pyrimethamine and amodiaquine remained efficacious, SMC effectively reduced malaria disease burden among children both < 5 and ≥ 5 years old and that the number of cycles should be commensurate with the length of the transmission season, up to six cycles.
Collapse
Affiliation(s)
- Julie Thwing
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - John Williamson
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Irene Cavros
- U.S. President’s Malaria Initiative, Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Julie R. Gutman
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
8
|
Phiri KS, Khairallah C, Kwambai TK, Bojang K, Dhabangi A, Opoka R, Idro R, Stepniewska K, van Hensbroek MB, John CC, Robberstad B, Greenwood B, Kuile FOT. Post-discharge malaria chemoprevention in children admitted with severe anaemia in malaria-endemic settings in Africa: a systematic review and individual patient data meta-analysis of randomised controlled trials. Lancet Glob Health 2024; 12:e33-e44. [PMID: 38097295 PMCID: PMC10733130 DOI: 10.1016/s2214-109x(23)00492-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Severe anaemia is associated with high in-hospital mortality among young children. In malaria-endemic areas, surviving children also have an increased risk of mortality or readmission after hospital discharge. We conducted a systematic review and individual patient data meta-analysis to determine the efficacy of monthly post-discharge malaria chemoprevention in children recovering from severe anaemia. METHODS This analysis was conducted according to PRISMA-IPD guidelines. We searched multiple databases on Aug 28, 2023, without date or language restrictions, for randomised controlled trials comparing monthly post-discharge malaria chemoprevention with placebo or standard of care among children (aged <15 years) admitted with severe anaemia in malaria-endemic Africa. Trials using daily or weekly malaria prophylaxis were not eligible. The investigators from all eligible trials shared pseudonymised datasets, which were standardised and merged for analysis. The primary outcome was all-cause mortality during the intervention period. Analyses were performed in the modified intention-to-treat population, including all randomly assigned participants who contributed to the endpoint. Fixed-effects two-stage meta-analysis of risk ratios (RRs) was used to generate pooled effect estimates for mortality. Recurrent time-to-event data (readmissions or clinic visits) were analysed using one-stage mixed-effects Prentice-Williams-Peterson total-time models to obtain hazard ratios (HRs). This study is registered with PROSPERO, CRD42022308791. FINDINGS Our search identified 91 articles, of which 78 were excluded by title and abstract, and a further ten did not meet eligibility criteria. Three double-blind, placebo-controlled trials, including 3663 children with severe anaemia, were included in the systematic review and meta-analysis; 3507 (95·7%) contributed to the modified intention-to-treat analysis. Participants received monthly sulfadoxine-pyrimethamine until the end of the malaria transmission season (mean 3·1 courses per child [range 1-6]; n=1085; The Gambia), monthly artemether-lumefantrine given at the end of weeks 4 and 8 post discharge (n=1373; Malawi), or monthly dihydroartemisinin-piperaquine given at the end of weeks 2, 6, and 10 post discharge (n=1049; Uganda and Kenya). During the intervention period, post-discharge malaria chemoprevention was associated with a 77% reduction in mortality (RR 0·23 [95% CI 0·08-0·70], p=0·0094, I2=0%) and a 55% reduction in all-cause readmissions (HR 0·45 [95% CI 0·36-0·56], p<0·0001) compared with placebo. The protective effect was restricted to the intervention period and was not sustained after the direct pharmacodynamic effect of the drugs had waned. The small number of trials limited our ability to assess heterogeneity, its sources, and publication bias. INTERPRETATION In malaria-endemic Africa, post-discharge malaria chemoprevention reduces mortality and readmissions in recently discharged children recovering from severe anaemia. Post-discharge malaria chemoprevention could be a valuable strategy for the management of this group at high risk. Future research should focus on methods of delivery, options to prolong the protection duration, other hospitalised groups at high risk, and interventions targeting non-malarial causes of post-discharge morbidity. FUNDING The Research-Council of Norway and the Bill-&-Melinda-Gates-Foundation through the Worldwide-Antimalarial-Research-Network.
Collapse
Affiliation(s)
- Kamija S Phiri
- School of Global and Public Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi; Training and Research Unit of Excellence, Blantyre, Malawi
| | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Titus K Kwambai
- Division of Parasitic Diseases and Malaria, Global Health Center, Centers for Disease Control and Prevention, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Kalifa Bojang
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Aggrey Dhabangi
- Makerere University College of Health Sciences, Kampala, Uganda
| | - Robert Opoka
- Makerere University College of Health Sciences, Kampala, Uganda; Aga Khan University, Medical College, Nairobi, Kenya
| | - Richard Idro
- Makerere University College of Health Sciences, Kampala, Uganda
| | - Kasia Stepniewska
- Worldwide Antimalarial Resistance Network (WWARN), Oxford, UK; Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Infectious Diseases Data Observatory (IDDO), Oxford, UK
| | - Michael Boele van Hensbroek
- Amsterdam Centre for Global Child Health, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bjarne Robberstad
- Section for Ethics and Health Economics, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.
| |
Collapse
|
9
|
Obeng-Aboagye E, Frimpong A, Amponsah JA, Danso SE, Owusu EDA, Ofori MF. Inflammatory cytokines as potential biomarkers for early diagnosis of severe malaria in children in Ghana. Malar J 2023; 22:220. [PMID: 37525227 PMCID: PMC10388454 DOI: 10.1186/s12936-023-04652-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Severe malaria (SM) is a fatal multi-system disease which accounted for an estimated 619,000 deaths in 2021. Less than 30% of children presenting with SM are diagnosed and treated promptly, resulting in increased mortality and neurologic impairments in survivors. Studies have identified cytokine profiles that differentiate the various clinical manifestations of malaria (severe and uncomplicated). However, the diagnostic capability of these cytokines in differentiating between the disease states in terms of cut-off values has not yet been determined. METHODS The plasma levels of 22 pro-inflammatory cytokines (Eotaxin/CCL 11, interferon-gamma (IFN-γ), interleukin (IL)- 2, IL-6, IL-1β, IL-12p40/p70, IL-17A, RANTES, MCP-1, IL-15, IL-5, IL-1RA, IL-2R, IFN-α, IP-10, TNF, MIG, MIP-1α, MIP-1β, IL-7, IL-8 and Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF), and 3 anti-inflammatory cytokines-(IL-4, IL-13 and IL-10) in patients with SM, uncomplicated malaria (UM) and other febrile conditions, were measured and compared using the Human Cytokine Magnetic 25-Plex Panel. The receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of these cytokines. RESULTS The level of the pro-inflammatory cytokine, IL-17A, was significantly higher in the SM group as compared to the UM group. Levels of the anti-inflammatory cytokines however did not differ significantly among the SM and UM groups. Only IL-1β and IL-17A showed good diagnostic potential after ROC curve analysis. CONCLUSION The data show that levels of pro-inflammatory cytokines correlate with malaria disease severity. IL-1β and IL-17A showed good diagnostic potentials and can be considered for use in clinical practice to target treatment.
Collapse
Affiliation(s)
- Elizabeth Obeng-Aboagye
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Augustina Frimpong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.
| | - Jones Amo Amponsah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | | | - Ewurama D A Owusu
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
10
|
Beshir KB, Muwanguzi J, Nader J, Mansukhani R, Traore A, Gamougam K, Ceesay S, Bazie T, Kolie F, Lamine MM, Cairns M, Snell P, Scott S, Diallo A, Merle CS, NDiaye JL, Razafindralambo L, Moroso D, Ouedraogo JB, Zongo I, Kessely H, Doumagoum D, Bojang K, Ceesay S, Loua K, Maiga H, Dicko A, Sagara I, Laminou IM, Ogboi SJ, Eloike T, Milligan P, Sutherland CJ. Prevalence of Plasmodium falciparum haplotypes associated with resistance to sulfadoxine-pyrimethamine and amodiaquine before and after upscaling of seasonal malaria chemoprevention in seven African countries: a genomic surveillance study. THE LANCET. INFECTIOUS DISEASES 2023; 23:361-370. [PMID: 36328000 DOI: 10.1016/s1473-3099(22)00593-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/08/2022] [Accepted: 09/02/2022] [Indexed: 12/26/2022]
Abstract
BACKGROUND Seasonal malaria chemoprevention is used in 13 countries in the Sahel region of Africa to prevent malaria in children younger than 5 years. Resistance of Plasmodium falciparum to seasonal malaria chemoprevention drugs across the region is a potential threat to this intervention. METHODS Between December, 2015, and March, 2016, and between December, 2017, and March, 2018, immediately following the 2015 and 2017 malaria transmission seasons, community surveys were done among children younger than 5 years and individuals aged 10-30 years in districts implementing seasonal malaria chemoprevention with sulfadoxine-pyrimethamine and amodiaquine in Burkina Faso, Chad, Guinea, Mali, Nigeria, Niger and The Gambia. Dried blood samples were collected and tested for P falciparum DNA by PCR. Resistance-associated haplotypes of the P falciparum genes crt, mdr1, dhfr, and dhps were identified by quantitative PCR and sequencing of isolates from the collected samples, and survey-weighted prevalence and prevalence ratio between the first and second surveys were estimated for each variant. FINDINGS 5130 (17·5%) of 29 274 samples from 2016 and 2176 (7·6%) of 28 546 samples from 2018 were positive for P falciparum on quantitative PCR. Among children younger than 5 years, parasite carriage decreased from 2844 of 14 345 samples (19·8% [95% CI 19·2-20·5]) in 2016 to 801 of 14 019 samples (5·7% [5·3-6·1]) in 2018 (prevalence ratio 0·27 [95% CI 0·24-0·31], p<0·0001). Genotyping found no consistent evidence of increasing prevalence of amodiaquine resistance-associated variants of crt and mdr1 between 2016 and 2018. The dhfr haplotype IRN (consisting of 51Ile-59Arg-108Asn) was common at both survey timepoints, but the dhps haplotype ISGEAA (431Ile-436Ser-437Gly-540Glu-581Ala-613Ala), crucial for resistance to sulfadoxine-pyrimethamine, was always rare. Parasites carrying amodiaquine resistance-associated variants of both crt and mdr1 together with dhfr IRN and dhps ISGEAA occurred in 0·05% of isolates. The emerging dhps haplotype VAGKGS (431Val-436Ala-437Gly-540Lys-581Gly-613Ser) was present in four countries. INTERPRETATION In seven African countries, evidence of a significant reduction in parasite carriage among children receiving seasonal malaria chemoprevention was found 2 years after intervention scale-up. Combined resistance-associated haplotypes remained rare, and seasonal malaria chemoprevention with sulfadoxine-pyrimethamine and amodiaquine is expected to retain effectiveness. The threat of future erosion of effectiveness due to dhps variant haplotypes requires further monitoring. FUNDING Unitaid.
Collapse
Affiliation(s)
- Khalid B Beshir
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Julian Muwanguzi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Johanna Nader
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK; Department of Genetics and Bioinformatics, Division of Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Raoul Mansukhani
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Aliou Traore
- Malaria Research and Training Centre, University of Bamako, Bamako, Mali
| | | | - Sainey Ceesay
- Medical Research Council Laboratories, London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Thomas Bazie
- Institute of Health Science Research, Bobo-Dioulasso, Burkina Faso
| | - Fassou Kolie
- Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | | | - Matt Cairns
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Paul Snell
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Susana Scott
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Corinne S Merle
- Special Programme for Research & Training in Tropical Diseases, WHO, Geneva, Switzerland
| | | | | | - Diego Moroso
- Malaria Consortium, Kampala, Uganda; UK Foreign, Commonwealth, & Development Office, Lagos, Nigeria
| | | | - Issaka Zongo
- Institute of Health Science Research, Bobo-Dioulasso, Burkina Faso
| | - Hamit Kessely
- Centre de Support en Santé Internationale, N'Djamena, Chad
| | | | - Kalifa Bojang
- Medical Research Council Laboratories, London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Serign Ceesay
- Medical Research Council Laboratories, London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Kovana Loua
- Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Hamma Maiga
- Malaria Research and Training Centre, University of Bamako, Bamako, Mali
| | - Alassane Dicko
- Malaria Research and Training Centre, University of Bamako, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Centre, University of Bamako, Bamako, Mali
| | | | | | - Tony Eloike
- Jedima International Health Consult, Lagos, Nigeria
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Colin J Sutherland
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
11
|
Hospital-based surveillance of severe paediatric malaria in two malaria transmission ecological zones of Burkina Faso. Malar J 2023; 22:6. [PMID: 36609299 PMCID: PMC9817371 DOI: 10.1186/s12936-022-04433-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 12/24/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND In the current context of tailoring interventions to maximize impact, it is important that current data of clinical epidemiology guide public health programmes and health workers in the management of severe disease. This study aimed at describing the burden of severe malaria at hospital level in two areas with distinct malaria transmission intensity. METHODS A hospital-based surveillance was established in two regional hospitals located in two areas exposed to different malaria transmission. Data on paediatric severe malaria admissions were recorded using standardized methods from August 2017 to August 2018 with an interruption during the dry season from April to June 2018. RESULTS In total, 921 children with severe malaria cases were enrolled in the study. The mean age was 33.9 (± 1.3) and 36.8 (± 1.6) months in lower malaria transmission (LMT) and higher malaria transmission (HMT) areas (p = 0.15), respectively. The geometric mean of asexual P. falciparum density was significantly higher in the LMT area compared to the HMT area: 22,861 trophozoites/µL (95% CI 17,009.2-30,726.8) vs 11,291.9 trophozoites/µL (95% CI 8577.9-14,864.5). Among enrolled cases, coma was present in 70 (9.2%) participants. 196 patients (21.8%) presented with two or more convulsions episodes prior to admission. Severe anaemia was present in 448 children (49.2%). Other clinical features recorded included 184 (19.9%) cases of lethargy, 99 (10.7%) children with incoercible vomiting, 80 (8.9%) patients with haemoglobinuria, 43 (4.8%) children with severe hypoglycaemia, 37 (4.0%) cases where child was unable to drink/suck, 11 (1.2%) cases of patients with circulatory collapse/shock, and 8 cases (0.9%) of abnormal bleeding (epistaxis). The adjusted odds of presenting with coma, respiratory distress, haemoglobinuria, circulatory collapse/shock and hypoglycaemia were significantly higher (respectively 6.5 (95%CI 3.4-12.1); 1.8 (95%CI 1.0-3.2); 2.7 (95%CI 1.6-4.3); 5.9 (95%CI 1.3-27.9); 1.9 (95%CI 1.0-3.6)) in children living in the HMT area compared to those residing in the LMT area. Overall, forty-four children died during hospitalization (case fatality rate 5.0%) with the highest fatalities in children admitted with respiratory distress (26.0%) and those with hypoglycaemia (25.0%). CONCLUSION The study showed that children in the HMT area have a higher risk of presenting with coma, shock/dehydration, haemoglobinuria, hypoglycaemia, and respiratory distress. Case-fatality rate is higher among patients with respiratory distress or hypoglycaemia. Hospital surveillance provides a reliable and sustainable means to monitor the clinical presentation of severe malaria and tailor the training needs and resources allocation for case management.
Collapse
|
12
|
Bakai TA, Thomas A, Iwaz J, Atcha-Oubou T, Tchadjobo T, Khanafer N, Rabilloud M, Voirin N. Effectiveness of seasonal malaria chemoprevention in three regions of Togo: a population-based longitudinal study from 2013 to 2020. Malar J 2022; 21:400. [PMID: 36587191 PMCID: PMC9804945 DOI: 10.1186/s12936-022-04434-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In 2012, the World Health Organization (WHO) recommended seasonal malaria chemoprevention (SMC) in areas of high seasonal transmission. Though implemented since 2013, the effectiveness of SMC in Togo was never evaluated. METHODS This study concerned routine data from 2013 to 2020 mass SMC campaigns for children under five in all health facilities of three Regions of Togo. Treatment coverage, reasons for non-treatment, and SMC-attributable adverse reactions were analysed per year and treatment round. Random effect logistic models estimated SMC effectiveness per health district, year, and treatment round. RESULTS The overall coverage was 98% (7,971,877 doses for 8,129,668 children). Contraindication was the main reason for non-administration. Over the study period, confirmed malaria cases decreased from 11,269 (1st round of 2016) to 1395 (4th round of 2020). Only 2,398 adverse reactions were reported (prevalence: 3/10,000), but no severe Lyell syndrome or Stevens-Johnson-type skin reaction. Compared to 2016, malaria prevalence decrease was estimated at 22.6% in 2017 (p < 0.001) and 75% in 2020 (p < 0.001). SMC effectiveness ranged from 76.6% (2nd round) to 96.2% (4th round) comparison with the 1st round. CONCLUSIONS SMC reduced significantly malaria cases among children under five. The results reassure all actors and call for effort intensification to reach the WHO goals for 2030.
Collapse
Affiliation(s)
- Tchaa A. Bakai
- grid.25697.3f0000 0001 2172 4233Université de Lyon, Lyon, France ,grid.7849.20000 0001 2150 7757Université Lyon 1, 69100 Villeurbanne, France ,grid.413852.90000 0001 2163 3825Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, 69003 Lyon, France ,grid.462854.90000 0004 0386 3493Équipe Biostatistique-Santé, Laboratoire de Biométrie et Biologie Évolutive, CNRS UMR 5558, 69100 Villeurbanne, France ,EPIMOD (Epidemiology and Modelling in Infectious Diseases), 01240 Lent, France ,Programme National de Lutte contre le Paludisme (PNLP), 01 BP 518, Lomé, Togo ,grid.412180.e0000 0001 2198 4166Service d’Hygiène, Épidémiologie et Prévention, Hôpital Édouard Herriot, Hospices Civils de Lyon, 69003 Lyon, France
| | - Anne Thomas
- grid.25697.3f0000 0001 2172 4233Université de Lyon, Lyon, France ,grid.7849.20000 0001 2150 7757Université Lyon 1, 69100 Villeurbanne, France ,grid.413852.90000 0001 2163 3825Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, 69003 Lyon, France ,grid.462854.90000 0004 0386 3493Équipe Biostatistique-Santé, Laboratoire de Biométrie et Biologie Évolutive, CNRS UMR 5558, 69100 Villeurbanne, France ,EPIMOD (Epidemiology and Modelling in Infectious Diseases), 01240 Lent, France
| | - Jean Iwaz
- grid.25697.3f0000 0001 2172 4233Université de Lyon, Lyon, France ,grid.7849.20000 0001 2150 7757Université Lyon 1, 69100 Villeurbanne, France ,grid.413852.90000 0001 2163 3825Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, 69003 Lyon, France ,grid.462854.90000 0004 0386 3493Équipe Biostatistique-Santé, Laboratoire de Biométrie et Biologie Évolutive, CNRS UMR 5558, 69100 Villeurbanne, France
| | - Tinah Atcha-Oubou
- Programme National de Lutte contre le Paludisme (PNLP), 01 BP 518, Lomé, Togo
| | - Tchassama Tchadjobo
- Programme National de Lutte contre le Paludisme (PNLP), 01 BP 518, Lomé, Togo
| | - Nagham Khanafer
- grid.412180.e0000 0001 2198 4166Service d’Hygiène, Épidémiologie et Prévention, Hôpital Édouard Herriot, Hospices Civils de Lyon, 69003 Lyon, France
| | - Muriel Rabilloud
- grid.25697.3f0000 0001 2172 4233Université de Lyon, Lyon, France ,grid.7849.20000 0001 2150 7757Université Lyon 1, 69100 Villeurbanne, France ,grid.413852.90000 0001 2163 3825Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, 69003 Lyon, France ,grid.462854.90000 0004 0386 3493Équipe Biostatistique-Santé, Laboratoire de Biométrie et Biologie Évolutive, CNRS UMR 5558, 69100 Villeurbanne, France
| | - Nicolas Voirin
- EPIMOD (Epidemiology and Modelling in Infectious Diseases), 01240 Lent, France
| |
Collapse
|
13
|
Ahorlu CS, Ndong IC, Okyere D, Mensah BA, Chu CE, Enos JY, Abuaku B. The Effect of Mass Testing, Treatment and Tracking on the Prevalence of Febrile Illness in Children under 15 in Ghana. Pathogens 2022; 11:pathogens11101118. [PMID: 36297175 PMCID: PMC9609179 DOI: 10.3390/pathogens11101118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/16/2022] [Accepted: 09/24/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Malaria remains a serious threat to children under 15 years of age in sub-Sahara Africa. Mass testing, treatment and tracking (MTTT) of malaria has been reported to reduce parasite load significantly. However, the impact of MTTT on the prevalence of febrile illnesses in children under 15 is not yet clear. This study explores the impact of MTTT complemented by prompt home-based management of malaria on febrile illnesses and their treatment in children under 15 years old. Methods: A cohort of 460 children under 15 years were recruited from the Pakro subdistrict in Ghana during a community-wide implementation of a quarterly MTTT intervention. The MTTT implementation involved testing all household members for malaria using RDTs, and positive cases were treated with Artemisinin-based combination therapy (ACT). Febrile illnesses among this cohort in the two weeks prior to the prevalence survey at baseline and endline were recorded to constitute date for analysis. Results: The prevalence of febrile illnesses, such chills, convulsion, fever, diarrhoea, headache, vomit, cough/rashes or stomachache, etc., were recorded). Asymptomatic parasitaemia prevalence at baseline was 53.3%, which dropped to 44.1% at evaluation. An overall decrease in the parasitaemia prevalence of 33.0% (OR = 0.67, CI = 0.50, 0.89) was observed at evaluation compared to baseline after adjusting for age, ITN use and temperature. A 67% decrease in severe anaemia cases (Hb < 7) was observed at evaluation. Conclusion: Our findings suggest that implementing MTTT complemented by home-based timely management of malaria does not only reduce febrile illnesses and for that matter malaria prevalence, but could also reduce severe anaemia in children under 15 years old.
Collapse
Affiliation(s)
- Collins Stephen Ahorlu
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
- Correspondence:
| | - Ignatius Cheng Ndong
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
- Department of Biochemistry, Faculty of Science, Catholic University of Cameroon, Bamenda P.O. Box 572, Cameroon
| | - Daniel Okyere
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
| | - Benedicta A. Mensah
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
| | - Chuo Ennestine Chu
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
- Department of Biochemistry, Faculty of Science, Catholic University of Cameroon, Bamenda P.O. Box 572, Cameroon
| | - Juliana Y. Enos
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
| | - Benjamin Abuaku
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 581, Ghana
| |
Collapse
|
14
|
Thiam LG, Nyarko PB, Ansah F, Niang M, Awandare GA, Aniweh Y. Phenotypic characterization of Ghanaian P. falciparum clinical isolates reveals a homogenous parasite population. Front Immunol 2022; 13:1009252. [PMID: 36211335 PMCID: PMC9537689 DOI: 10.3389/fimmu.2022.1009252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/06/2022] [Indexed: 01/26/2023] Open
Abstract
Background Erythrocyte invasion by P. falciparum involves functionally overlapping interactions between the parasite's ligands and the erythrocyte surface receptors. While some P. falciparum isolates necessarily engage the sialic acid (SA) moieties of the erythrocytes during the invasion, others use ligands whose binding is independent of SA for successful invasion. Deciphering the major pathway used by P. falciparum clinical isolates represent a key step toward developing an efficient blood stage malaria vaccine. Methods We collected a total of 156 malaria-infected samples from Ghanaian children aged 2 to 14 years and used a two-color flow cytometry-based invasion assay to assess the invasion phenotype diversity of Ghanaian P. falciparum clinical isolates. Anti-human CR1 antibodies were used to determine the relative contribution of the PfRh4-CR1 interaction in the parasites invasion phenotype and RT-qPCR was used to assess the expression levels of key invasion-related ligands. Results Our findings show no clear association between demographic or clinical data and existing reports on the malaria transmission intensity. The complete invasion data obtained for 156 isolates, showed the predominance of SA-independent pathways in Ghanaian clinical isolates. Isolates from Hohoe and Navrongo had the highest diversity in invasion profile. Our data also confirmed that the PfRh4-CR1 mediated alternative pathway is important in Ghanaian clinical isolates. Furthermore, the transcript levels of ten invasion-related genes obtained in the study showed little variations in gene expression profiles within and between parasite populations across sites. Conclusion Our data suggest a low level of phenotypic diversity in Ghanaian clinical isolates across areas of varying endemicity and further highlight its importance in the quest for new intervention strategies, such as the investigation of blood-stage vaccine targets, particularly those targeting specific pathways and able to trigger the stimulation of broadly neutralizing invasion antibodies.
Collapse
Affiliation(s)
- Laty G. Thiam
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Prince B. Nyarko
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Felix Ansah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Makhtar Niang
- Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
15
|
Factors Influencing Second and Third Dose Observance during Seasonal Malaria Chemoprevention (SMC): A Quantitative Study in Burkina Faso, Mali and Niger. Trop Med Infect Dis 2022; 7:tropicalmed7090214. [PMID: 36136625 PMCID: PMC9503675 DOI: 10.3390/tropicalmed7090214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
This study aims to evaluate the factors influencing the adherence to the 2nd and 3rd doses of Amodiaquine (AQ) during seasonal malaria chemoprevention (SMC) in Burkina Faso, Mali, and Niger. Overall, 3132 people were interviewed during surveys between 2019 and 2020 in 15 health districts. In Burkina Faso, Mali, and Niger, the proportions of non-adherence were 4.15%, 5.60%, and 13.30%, respectively, for the 2nd dose and 3.98%, 5.60% and 14.39% for the 3rd dose. The main cause of non-adherence to the 2nd and 3rd doses was other illnesses in 28.5% and 29.78%, respectively, in Burkina Faso, 5.35% and 5.35% in Mali and 1.6% and 0.75% in Niger. It was followed by vomiting in 12.24% and 10.63% for Burkina and 2.45% and 3.78% in Niger. The last cause was refusal in 6.12% and 4.25% in Burkina, 33.9% and 15.25% in Mali and 0.8% and 1.51% in Niger. Non-adherence of doses related to parents was primarily due to their absence in 28.5% and 27.65% in Burkina, 16.07% and 16.07% in Mali and 7.37% and 6.06% in Niger. Traveling was the second cause related to parents in 12.24% and 12.76% in Burkina, 19.64% and 19.64% in Mali and 0.81% and 0.75% in Niger. Non-adherence related to community distributors was mainly due to missing the doses in 4.08% and 4.25% in Burkina, 23.21% and 23.21% in Mali, 77.04% and 76.51% in Niger. Our study reported very small proportions of non-adherence to 2nd and 3rd doses of SMC and identified the main causes of non-adherence. These findings will provide helpful information for policymakers and public health authorities to improve adherence to SMC
Collapse
|
16
|
Nikiema S, Soulama I, Sombié S, Tchouatieu AM, Sermé SS, Henry NB, Ouedraogo N, Ouaré N, Ily R, Ouédraogo O, Zongo D, Djigma FW, Tiono AB, Sirima SB, Simporé J. Seasonal Malaria Chemoprevention Implementation: Effect on Malaria Incidence and Immunity in a Context of Expansion of P. falciparum Resistant Genotypes with Potential Reduction of the Effectiveness in Sub-Saharan Africa. Infect Drug Resist 2022; 15:4517-4527. [PMID: 35992756 PMCID: PMC9386169 DOI: 10.2147/idr.s375197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Seasonal Malaria Chemoprevention (SMC), which combines amodiaquine (AQ) with sulfadoxine-pyrimethamine (SP), is an effective and promising strategy, recommended by WHO, for controlling malaria morbidity and mortality in areas of intense seasonal transmission. Despite the effectiveness of this strategy, a number of controversies regarding the impact of the development of malaria-specific immunity and challenges of the strategy in the context of increasing and expanding antimalarial drugs resistance but also the limited coverage of the SMC in children make the relevance of the SMC questionable, especially in view of the financial and logistical investments. Indeed, the number of malaria cases in the target group, children under 5 years old, has increased while the implementation of SMC is been extended in several African countries. This ambivalence of the SMC strategy, the increase in the prevalence of malaria cases suggests the need to evaluate the SMC and understand some of the factors that may hinder the success of this strategy in the implementation areas. The present review discusses the impact of the SMC on malaria morbidity, parasite resistance to antimalarial drugs, molecular and the immunity affecting the incidence of malaria in children. This approach will contribute to improving the malaria control strategy in highly seasonal transmission areas where the SMC is implemented.
Collapse
Affiliation(s)
- Séni Nikiema
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
- Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
| | - Issiaka Soulama
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
- Biomedical and Public Health Department, Institut de Recherche en Sciences de la Santé (IRSS)/Centre National de Recherche Scientifiques et Technologiques (CNRST), Ouagadougou, Burkina Faso
| | - Salif Sombié
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
| | - André-Marie Tchouatieu
- Access and Product Management – Chemoprevention Department, Medicines for Malaria Venture (MMV), Geneva, Switzerland
| | - Samuel Sindie Sermé
- Direction Scientifique, Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
| | - Noëlie Béré Henry
- Direction Scientifique, Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
| | - Nicolas Ouedraogo
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
| | - Nathalie Ouaré
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
- Institut Supérieur des Sciences de la santé (IN.S.SA), Université Nazi Boni, Bobo Dioulasso, Burkina Faso
| | - Raissa Ily
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
- Institut Supérieur des Sciences de la santé (IN.S.SA), Université Nazi Boni, Bobo Dioulasso, Burkina Faso
| | - Oumarou Ouédraogo
- Biomedical and Public Health Department, Institut de Recherche en Sciences de la Santé (IRSS)/Centre National de Recherche Scientifiques et Technologiques (CNRST), Ouagadougou, Burkina Faso
| | - Dramane Zongo
- Biomedical and Public Health Department, Institut de Recherche en Sciences de la Santé (IRSS)/Centre National de Recherche Scientifiques et Technologiques (CNRST), Ouagadougou, Burkina Faso
| | - Florencia Wendkuuni Djigma
- Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
| | - Alfred B Tiono
- Research Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP)/Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
| | - Sodiomon B Sirima
- Direction Scientifique, Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
| | - Jacques Simporé
- Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
- Centre de recherche biomoléculaire Pietro Annigoni (CERBA), Ouagadougou, Burkina Faso
| |
Collapse
|
17
|
Effect of seasonal malaria chemoprevention in children between 5 and 9 years old in Kita and Bafoulabe districts, Mali. Parasite Epidemiol Control 2022; 18:e00258. [PMID: 35789762 PMCID: PMC9249800 DOI: 10.1016/j.parepi.2022.e00258] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/22/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background Seasonal malaria chemoprevention (SMC) has been widely expanded in Mali since its recommendation by the the World Health Organization in 2012. SMC guidelines currently target children between three months and five years of age. The SMC initiative has been largely successful. Children at least five years of age are not currently covered by current SMC guidelines but bear a considerable portion of the malaria burden. For this reason, this study sought to determine the feasibility and effectiveness for extending SMC to children aged 5–9 years. Methods A non-randomized, pre-post study was performed with an intervention district (Kita) and a comparison district (Bafoulabe). Children aged 3–59 months received SMC in both comparison districts, and children aged 60–120 months received SMC in the intervention district. SMC was delivered as sulfadoxine-pyriméthamine plus amodiaquine (SP-AQ) at monthly intervals from July to October in 2017 and 2018 during the historical transmission seasons. Baseline and endline cross-sectional surveys were conducted in both comparison districts. A total of 200 household surveys were conducted at each of the four monthly SMC cycles to determine adherence and tolerance to SMC in the intervention district. Results In July 2017, 633 children aged 60–120 months old were enrolled at the Kita and Bafoulabe study sites (n = 310 and n = 323, respectively). Parasitemia prevalence was similar in the intervention and comparison districts prior the SMC campaign (27.7% versus 21.7%, p = 0.07). Mild anemia was observed in 14.2% children in Kita and in 10.5% of children in Bafoulabé. At the Kita site, household surveys showed an SMC coverage rate of 89.1% with a response rate of 93.3% among child caregivers. The most common adverse event reported by parents was drowsiness (11.8%). One year following SMC implementation in the older age group in Kita, the coverage of three doses per round was 81.2%. Between the baseline and endline surveys, there was a reduction in parasitemia prevalence of 40% (OR = 0.60, CI: 0.41–0.89). Malaria molecular resistance was low in the intervention district following the intervention. A significant reduction in the prevalence of parasitemia in children 60 to 120 months was observed in the intervention district, but the prevalance of clinical malaria remained relatively constant. Conclusion This study shows that the prospect of extending SMC coverage to children between five and nine years old is encouraging. The reduction in the parasitemia could also warrant consideration for adapting SMC policy to account for extended malaria transmission seasons.
Collapse
|
18
|
Frimpong A, Owusu EDA, Amponsah JA, Obeng-Aboagye E, van der Puije W, Frempong AF, Kusi KA, Ofori MF. Cytokines as Potential Biomarkers for Differential Diagnosis of Sepsis and Other Non-Septic Disease Conditions. Front Cell Infect Microbiol 2022; 12:901433. [PMID: 35811678 PMCID: PMC9260692 DOI: 10.3389/fcimb.2022.901433] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 01/08/2023] Open
Abstract
Sepsis defined as a dysregulated immune response is a major cause of morbidity in children. In sub-Saharan Africa, the clinical features of sepsis overlap with other frequent infections such as malaria, thus sepsis is usually misdiagnosed in the absence of confirmatory tests. Therefore, it becomes necessary to identify biomarkers that can be used to distinguish sepsis from other infectious diseases. We measured and compared the plasma levels of 18 cytokines (Th1 [GM-CSF, IFN-γ, TNF-α, IL-1β, 1L-2, IL-6, IL-8, IL-12/IL-23p40, IL-15], Th2[IL-4, IL-5, IL-13), Th17 [IL17A], Regulatory cytokine (IL-10) and 7 chemokines (MCP-1/CCL2, MIP-1α/CCL3, MIP-1β/CCL4, RANTES/CCL5, Eotaxin/CCL11, MIG/CXCL9 and IP-10/CXCL10 using the Human Cytokine Magnetic 25-Plex Panel in plasma samples obtained from children with sepsis, clinical malaria and other febrile conditions. Children with sepsis had significantly higher levels of IL-1β, IL-12 and IL-17A compared to febrile controls but lower levels of MIP1-β/CCL4, RANTES/CCL5 and IP10/CXCL10 when compared to children with malaria and febrile controls. Even though levels of most inflammatory responses were higher in malaria compared to sepsis, children with sepsis had a higher pro-inflammatory to anti-inflammatory ratio which seemed to be mediated by mostly monocytes. A principal component analysis and a receiver operator characteristic curve analysis, identified seven potential biomarkers; IL-1β, IL-7, IL-12, IL-1RA, RANTES/CCL5, MIP1β/CCL4 and IP10/CXCL10 that could discriminate children with sepsis from clinical malaria and other febrile conditions. The data suggests that sepsis is associated with a higher pro-inflammatory environment. These pro-inflammatory cytokines/chemokines could further be evaluated for their diagnostic potential to differentiate sepsis from malaria and other febrile conditions in areas burdened with infectious diseases.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- *Correspondence: Augustina Frimpong, ; Michael Fokuo Ofori,
| | - Ewurama D. A. Owusu
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Jones Amo Amponsah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Elizabeth Obeng-Aboagye
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - William van der Puije
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Abena Fremaah Frempong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- *Correspondence: Augustina Frimpong, ; Michael Fokuo Ofori,
| |
Collapse
|
19
|
Kirakoya-Samadoulougou F, De Brouwere V, Fokam AF, Ouédraogo M, Yé Y. Assessing the effect of seasonal malaria chemoprevention on malaria burden among children under 5 years in Burkina Faso. Malar J 2022; 21:143. [PMID: 35524310 PMCID: PMC9074217 DOI: 10.1186/s12936-022-04172-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background In 2014, the Burkina Faso government launched the Seasonal Malaria Chemoprevention (SMC) programme. Expected benefit was a 75% reduction of all malaria episodes and a 75% drop of severe malaria episodes. This study assessed SMC efficiency on malaria morbidity in the country after 2 years of implementation. Methods Quasi-experimental design comparing changes in outcomes during the high transmission period (August–November) between SMC and non-SMC health districts before (2013–2014) and after intervention (two rounds in 2015 and 2016). Health indicators (number of uncomplicated malaria cases (UM) and severe malaria cases (SM)) from 19 health districts (8 in intervention and 11 in comparison group) were extracted from the District Health Information System (DHIS2)-based platform including health facilities data. Effect on incidence was assessed by fitting difference-in difference mixed-effects negative binomial regression model at a log scale. Results The two rounds of SMC were associated with a reduction of UM incidence (ratio of incidence rate ratio (IRR) 69% (95% CI 55–86%); p = 0.001) and SM incidence (ratio of IRR = 73% (55–95%), p = 0.018) among under five children. Conclusion The two rounds of SMC had a significant effect on the reduction of malaria cases in under five children. This additional evidence on the effectiveness of SMC, using routine data, support the need to sustain its implementation and consider expansion to eligible areas not yet covered. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04172-z.
Collapse
Affiliation(s)
- Fati Kirakoya-Samadoulougou
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium.
| | - Vincent De Brouwere
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Mady Ouédraogo
- Institut national de la statistique et de la démographie (INSD), Ouagadougou, Burkina Faso
| | - Yazoumé Yé
- ICF, 530 Gaither Road, Rockville, MD, 20850, USA
| |
Collapse
|
20
|
Chu X, Yan P, Zhang N, Chen N, Liu Y, Feng L, Li M, Zhang Z, Wang Q, Wang S, Yang K. The efficacy and safety of intermittent preventive treatment with sulphadoxine-pyrimethamine vs artemisinin-based drugs for malaria: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg 2021; 116:298-309. [PMID: 34651193 DOI: 10.1093/trstmh/trab158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/27/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Malaria is one of the most serious global problems. The objective of this study is to assess whether intermittent preventive treatment (IPT) using artemisinin-based combination therapies (ACTs) was a promising alternative to IPT with sulphadoxine-pyrimethamine (IPT-SP). METHODS We searched the following sources up to 12 August 2020: PubMed, The Cochrane Library, Embase, Web of Science, CNKI, CBM, VIP and WanFang Database from inception. The randomized controlled trials comparing SP with ACTs for malaria were included. Data were pooled using Stata.14 software. We performed subgroup analysis based on the different types of ACTs groups and participants. RESULTS A total of 13 studies comprising 5180 people were included. The meta-analysis showed that ACTs had the lower risk of number of any parasitemia (RR=0.46; 95% CI 0.22 to 0.96, p=0.039; I2=90.50%, p<0.001), early treatment failure (RR=0.17; 95% CI 0.06 to 0.48, p<0.001; I2=66.60%, p=0.011) and late treatment failure (RR=0.34; 95% CI 0.13 to 0.92, p<0.001; I2=87.80%, p<0.001) compared with SP. There was no significant difference in adequate clinical response, average hemoglobin and adverse neonatal outcomes. CONCLUSION Combinations with ACTs appear promising as suitable alternatives for IPT-SP.
Collapse
Affiliation(s)
- Xiajing Chu
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Peijing Yan
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610044, China
| | - Na Zhang
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Nan Chen
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Yang Liu
- Institute for Health Toxicology, School of Public Health, Lanzhou University, Lanzhou,730000, China
| | - Lufang Feng
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Meixuan Li
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Ziyao Zhang
- School of Foreign Language, Lanzhou University of Arts and Science, Lanzhou, 730000, China
| | - Qi Wang
- Health Policy PhD Program, McMaster University, Hamilton, Ontario, L8S 4K1, Canada.,McMaster Health Forum, McMaster University, Hamilton, Ontario, L8S 4L6, Canada.,Department of Health Research Methods, Evidence and Impact, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | | | - Kehu Yang
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| |
Collapse
|
21
|
Abstract
BACKGROUND Intermittent preventive treatment could help prevent malaria in infants (IPTi) living in areas of moderate to high malaria transmission in sub-Saharan Africa. The World Health Organization (WHO) policy recommended IPTi in 2010, but its adoption in countries has been limited. OBJECTIVES To evaluate the effects of intermittent preventive treatment (IPT) with antimalarial drugs to prevent malaria in infants living in malaria-endemic areas. SEARCH METHODS We searched the following sources up to 3 December 2018: the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (the Cochrane Library), MEDLINE (PubMed), Embase (OVID), LILACS (Bireme), and reference lists of articles. We also searched the metaRegister of Controlled Trials (mRCT) and the WHO International Clinical Trials Registry Platform (ICTRP) portal for ongoing trials up to 3 December 2018. SELECTION CRITERIA We included randomized controlled trials (RCTs) that compared IPT to placebo or no intervention in infants (defined as young children aged between 1 to 12 months) in malaria-endemic areas. DATA COLLECTION AND ANALYSIS The primary outcome was clinical malaria (fever plus asexual parasitaemia). Two review authors independently assessed trials for inclusion, evaluated the risk of bias, and extracted data. We summarized dichotomous outcomes and count data using risk ratios (RR) and rate ratios respectively, and presented all measures with 95% confidence intervals (CIs). We extracted protective efficacy values and their 95% CIs; when an included trial did not report this data, we calculated these values from the RR or rate ratio with its 95% CI. Where appropriate, we combined data in meta-analyses and assessed the certainty of the evidence using the GRADE approach. MAIN RESULTS We included 12 trials that enrolled 19,098 infants; all were conducted in sub-Saharan Africa. Three trials were cluster-RCTs. IPTi with sulfadoxine-pyrimethamine (SP) was evaluated in 10 trials from 1999 to 2013 (n = 15,256). Trials evaluating ACTs included dihydroartemisinin-piperaquine (1 trial, 147 participants; year 2013), amodiaquine-artesunate (1 study, 684 participants; year 2008), and SP-artesunate (1 trial, 676 participants; year 2008). The earlier studies evaluated IPTi with SP, and were conducted in Tanzania (in 1999 and 2006), Mozambique (2004), Ghana (2004 to 2005), Gabon (2005), Kenya (2008), and Mali (2009). One trial evaluated IPTi with amodiaquine in Tanzania (2000). Later studies included three conducted in Kenya (2008), Tanzania (2008), and Uganda (2013), evaluating IPTi in multiple trial arms that included artemisinin-based combination therapy (ACT). Although the effect size varied over time and between drugs, overall IPTi impacts on the incidence of clinical malaria overall, with a 30% reduction (rate ratio 0.70, 0.62 to 0.80; 10 studies, 10,602 participants). The effect of SP appeared to attenuate over time, with trials conducted after 2009 showing little or no effect of the intervention. IPTi with SP probably resulted in fewer episodes of clinical malaria (rate ratio 0.78, 0.69 to 0.88; 8 trials, 8774 participants, moderate-certainty evidence), anaemia (rate ratio 0.82, 0.68 to 0.98; 6 trials, 7438 participants, moderate-certainty evidence), parasitaemia (rate ratio 0.66, 0.56 to 0.79; 1 trial, 1200 participants, moderate-certainty evidence), and fewer hospital admissions (rate ratio 0.85, 0.78 to 0.93; 7 trials, 7486 participants, moderate-certainty evidence). IPTi with SP probably made little or no difference to all-cause mortality (risk ratio 0.93, 0.74 to 1.15; 9 trials, 14,588 participants, moderate-certainty evidence). Since 2009, IPTi trials have evaluated ACTs and indicate impact on clinical malaria and parasitaemia. A small trial of DHAP in 2013 shows substantive effects on clinical malaria (RR 0.42, 0.33 to 0.54; 1 trial, 147 participants, moderate-certainty evidence) and parasitaemia (moderate-certainty evidence). AUTHORS' CONCLUSIONS In areas of sub-Saharan Africa, giving antimalarial drugs known to be effective against the malaria parasite at the time to infants as IPT probably reduces the risk of clinical malaria, anaemia, and hospital admission. Evidence from SP studies over a 19-year period shows declining efficacy, which may be due to increasing drug resistance. Combinations with ACTs appear promising as suitable alternatives for IPTi.
Collapse
Affiliation(s)
- Ekpereonne B Esu
- Department of Public Health, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Chioma Oringanje
- GIDP Entomology and Insect Science, University of Tucson, Tucson, Arizona, USA
| | - Martin M Meremikwu
- Department of Paediatrics, University of Calabar Teaching Hospital, Calabar, Nigeria
| |
Collapse
|
22
|
Ofori MF, Kploanyi EE, Mensah BA, Dickson EK, Kyei-Baafour E, Gyabaa S, Tetteh M, Koram KA, Abuaku BK, Ghansah A. Ex vivo Sensitivity Profile of Plasmodium falciparum Clinical Isolates to a Panel of Antimalarial Drugs in Ghana 13 Years After National Policy Change. Infect Drug Resist 2021; 14:267-276. [PMID: 33536768 PMCID: PMC7850388 DOI: 10.2147/idr.s295277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/09/2021] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Malaria continues to be a major health issue globally with almost 85% of the global burden and deaths borne by sub-Saharan Africa and India. Although the current artemisinin derived combination therapies in Ghana are still efficacious against the Plasmodium falciparum (Pf) parasite, compounding evidence of artemisinin and amodiaquine resistance establish the need for a full, up-to-date understanding and monitoring of antimalarial resistance to provide evidence for planning control strategies. MATERIALS AND METHODS The study was cross-sectional and was conducted during the peak malaria transmission seasons of 2015, 2016, and 2017 in two ecological zones of Ghana. Study participants included children aged 6 months to 14 years. Using ex vivo 4,6-diamidino-2-phenylindole (DAPI) drug sensitivity assay, 330 Pf isolates were used to investigate susceptibility to five antimalarial drugs: chloroquine (CQ), amodiaquine (AMD) dihydroartemisinin (DHA), artesunate (ART) and mefloquine (MFQ). RESULTS The pooled geometric mean IC50S (GMIC50) of the five drugs against the parasites from Cape Coast and Begoro were 15.5, 42.4, 18.9, 4.6 and 27.3nM for CQ, AMD, DHA, ART, and MFQ, respectively. The GMIC50 values for CQ (p<0.001), ART (p<0.011) and DHA (p<0.018) were significantly higher for Cape Coast isolates as compared to Begoro isolates. However, GMIC50 estimates for MFQ (p<0.022) were significantly higher for Begoro isolates. Positive correlations were found between each pair of drugs with the weakest found between MFQ and DHA (r = 0.34;p<0.001), and the strongest between ART and DHA (r =0.66; p<0.001). CONCLUSION The parasites showed reduced sensitivities to three (AMD, DHA and MFQ) out of the five drugs assessed. The study also demonstrated the continual return of chloroquine-sensitive parasites after 13 years of its withdrawal as the first-line drug for the treatment of uncomplicated malaria in Ghana. The ex vivo DAPI assay is a reliable method for assessing antimalarial drug sensitivities of Pf field isolates under field settings.
Collapse
Affiliation(s)
- Michael Fokuo Ofori
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Emma E Kploanyi
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Benedicta A Mensah
- Epidemiology Department, Noguchi Memorial Institute for Medical Research,University of Ghana, Legon, Accra, Ghana
| | - Emmanuel K Dickson
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Eric Kyei-Baafour
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Sampson Gyabaa
- Ewim Polyclinic, Ghana Health Service, Cape Coast, Ghana
| | - Mary Tetteh
- Begoro District Hospital, Ghana Health Service, Begoro, Ghana
| | - Kwadwo A Koram
- Epidemiology Department, Noguchi Memorial Institute for Medical Research,University of Ghana, Legon, Accra, Ghana
| | - Benjamin K Abuaku
- Epidemiology Department, Noguchi Memorial Institute for Medical Research,University of Ghana, Legon, Accra, Ghana
| | - Anita Ghansah
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
- Parasitology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
23
|
Kwambai TK, Dhabangi A, Idro R, Opoka R, Watson V, Kariuki S, Kuya NA, Onyango ED, Otieno K, Samuels AM, Desai MR, Boele van Hensbroek M, Wang D, John CC, Robberstad B, Phiri KS, Ter Kuile FO. Malaria Chemoprevention in the Postdischarge Management of Severe Anemia. N Engl J Med 2020; 383:2242-2254. [PMID: 33264546 PMCID: PMC9115866 DOI: 10.1056/nejmoa2002820] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Children who have been hospitalized with severe anemia in areas of Africa in which malaria is endemic have a high risk of readmission and death within 6 months after discharge. No prevention strategy specifically addresses this period. METHODS We conducted a multicenter, two-group, randomized, placebo-controlled trial in nine hospitals in Kenya and Uganda to determine whether 3 months of malaria chemoprevention could reduce morbidity and mortality after hospital discharge in children younger than 5 years of age who had been admitted with severe anemia. All children received standard in-hospital care for severe anemia and a 3-day course of artemether-lumefantrine at discharge. Two weeks after discharge, children were randomly assigned to receive dihydroartemisinin-piperaquine (chemoprevention group) or placebo, administered as 3-day courses at 2, 6, and 10 weeks after discharge. Children were followed for 26 weeks after discharge. The primary outcome was one or more hospital readmissions for any reason or death from the time of randomization to 6 months after discharge. Conditional risk-set modeling for recurrent events was used to calculate hazard ratios with the use of the Prentice-Williams-Peterson total-time approach. RESULTS From May 2016 through May 2018, a total of 1049 children underwent randomization; 524 were assigned to the chemoprevention group and 525 to the placebo group. From week 3 through week 26, a total of 184 events of readmission or death occurred in the chemoprevention group and 316 occurred in the placebo group (hazard ratio, 0.65; 95% confidence interval [CI], 0.54 to 0.78; P<0.001). The lower incidence of readmission or death in the chemoprevention group than in the placebo group was restricted to the intervention period (week 3 through week 14) (hazard ratio, 0.30; 95% CI, 0.22 to 0.42) and was not sustained after that time (week 15 through week 26) (hazard ratio, 1.13; 95% CI, 0.87 to 1.47). No serious adverse events were attributed to dihydroartemisinin-piperaquine. CONCLUSIONS In areas with intense malaria transmission, 3 months of postdischarge malaria chemoprevention with monthly dihydroartemisinin-piperaquine in children who had recently received treatment for severe anemia prevented more deaths or readmissions for any reason after discharge than placebo. (Funded by the Research Council of Norway and the Centers for Disease Control and Prevention; ClinicalTrials.gov number, NCT02671175.).
Collapse
Affiliation(s)
- Titus K Kwambai
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Aggrey Dhabangi
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Richard Idro
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Robert Opoka
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Victoria Watson
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Simon Kariuki
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Nickline A Kuya
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Eric D Onyango
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Kephas Otieno
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Aaron M Samuels
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Meghna R Desai
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Michael Boele van Hensbroek
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Duolao Wang
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Chandy C John
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Bjarne Robberstad
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Kamija S Phiri
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Feiko O Ter Kuile
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| |
Collapse
|
24
|
Oppong M, Lamptey H, Kyei-Baafour E, Aculley B, Ofori EA, Tornyigah B, Kweku M, Ofori MF. Prevalence of sickle cell disorders and malaria infection in children aged 1-12 years in the Volta Region, Ghana: a community-based study. Malar J 2020; 19:426. [PMID: 33228681 PMCID: PMC7684914 DOI: 10.1186/s12936-020-03500-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/15/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Alterations in the structure of haemoglobin (Hb) are usually brought about by point mutations affecting one or, in some cases, two codons encoding amino acids of the globin chains. One in three Ghanaians are said to have sickle cell disorders, whereas malaria continues to be one of the leading causes of mortality among children. This study determined the prevalence of sickle cell disorders and malaria infection among children aged 1-12 years in the Volta Region. METHODS This was a community-based cross-sectional survey that involved 938 children aged 1-12 years selected from three districts, one each from the 3 geographical zones of the Volta Region using a multistage sampling method. Demographic information was collected using a standard questionnaire and anthropometric indices were measured. Isoelectric focusing (IEF) electrophoresis was used to determine the Hb genotypes and sub-microscopic parasites were determined by PCR. RESULTS The prevalence of sickling screening positive was 16.0% with an overall prevalence of sickle cell disorders being 2.0%. Among the individual genotypes making up the sickle cell disorders, genotype HbSF was the highest (0.9% as compared to 0.2%; HbSS, 0.6%; HbSC and 0.3%; HbSCF). Microscopic Plasmodium falciparum parasitaemia was detected among 5.5% of the children and 14.2% sub-microscopic prevalence by PCR. Children with sickle cell disorders were more likely to have sub-microscopic parasitaemia (AOR = 5.51 95%CI (2.15, 14.10), p < 0.001) as well as anaemia (AOR = 3.03 95% CI (1.04, 8.82), p = 0.042), compared to those with normal genotypes. There was no significant difference observed between sickle cell disorders and growth and development of the children screened. CONCLUSIONS Sickle cell disorders were significantly associated with sub-microscopic parasitaemia as well as anaemia in this study. Establishment of sickle cell clinics in the district and regional hospitals will help in the management of children with the disorder and also generate a national database on sickle cell disorders. National neonatal screening policies must also be put in place to help in early detection and management of these disorders.
Collapse
Affiliation(s)
- Mavis Oppong
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Hohoe, Ghana
| | - Helena Lamptey
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Eric Kyei-Baafour
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Belinda Aculley
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ebenezer Addo Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Bernard Tornyigah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Margaret Kweku
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Hohoe, Ghana
| | - Michael F Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
25
|
Lopez-Perez M, van der Puije W, Castberg FC, Ofori MF, Hviid L. Binding of human serum proteins to Plasmodium falciparum-infected erythrocytes and its association with malaria clinical presentation. Malar J 2020; 19:362. [PMID: 33032607 PMCID: PMC7545873 DOI: 10.1186/s12936-020-03438-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/01/2020] [Indexed: 11/10/2022] Open
Abstract
Background The pathogenesis of Plasmodium falciparum malaria is related to the ability of parasite‑infected erythrocytes (IEs) to adhere to the vascular endothelium (cytoadhesion/sequestration) or to surrounding uninfected erythrocytes (rosetting). Both processes are mediated by the expression of members of the clonally variant PfEMP1 parasite protein family on the surface of the IEs. Recent evidence obtained with laboratory-adapted clones indicates that P. falciparum can exploit human serum factors, such as IgM and α2-macroglobulin (α2M), to increase the avidity of PfEMP1-mediated binding to erythrocyte receptors, as well as to evade host PfEMP1-specific immune responses. It has remained unclear whether PfEMP1 variants present in field isolates share these characteristics, and whether they are associated with clinical malaria severity. These issues were investigated here. Methods Children 1–12 years reporting with P. falciparum malaria to Hohoe Municipal Hospital, Ghana were enrolled in the study. Parasites from children with uncomplicated (UM) and severe malaria (SM) were collected. Binding of α2M and IgM from non-immune individuals to erythrocytes infected by P. falciparum isolates from 34 children (UM and SM) were analysed by flow cytometry. Rosetting in the presence of IgM or α2M was also evaluated. Experimental results were analysed according to the clinical presentation of the patients. Results Clinical data from 108 children classified as UM (n = 54) and SM cases (n = 54) were analysed. Prostration, severe malaria anaemia, and hyperparasitaemia were the most frequent complications. Three children were diagnosed with cerebral malaria, and one child died. Parasite isolates from UM (n = 14) and SM (n = 20) children were analysed. Most of the field isolates bound non-immune IgM (33/34), whereas the α2M-binding was less common (23/34). Binding of both non-immune IgM and α2M was higher but not significant in IEs from children with SM than from children with UM. In combination, IgM and α2M supported rosette formation at levels similar to that observed in the presence of 10% human serum. Conclusions The results support the hypothesis that binding of non-immune IgM and/or α2M to IEs facilitates rosette formation and perhaps contributes to P. falciparum malaria severity.
Collapse
Affiliation(s)
- Mary Lopez-Perez
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - William van der Puije
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana.,West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Legon, Ghana.,Centre for Medical Parasitology, Department of Infectious Diseases and Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Filip C Castberg
- Centre for Medical Parasitology, Department of Infectious Diseases and Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Michael F Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Lars Hviid
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Centre for Medical Parasitology, Department of Infectious Diseases and Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
26
|
Maiga H, Gaudart J, Sagara I, Diarra M, Bamadio A, Djimde M, Coumare S, Sangare B, Dicko Y, Tembely A, Traore D, Dicko A, Lasry E, Doumbo O, Djimde AA. Two-Year Scale-Up of Seasonal Malaria Chemoprevention Reduced Malaria Morbidity among Children in the Health District of Koutiala, Mali. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E6639. [PMID: 32932990 PMCID: PMC7558455 DOI: 10.3390/ijerph17186639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/03/2020] [Accepted: 09/05/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Previous controlled studies demonstrated seasonal malaria chemoprevention (SMC) reduces malaria morbidity by >80% in children aged 3-59 months. Here, we assessed malaria morbidity after large-scale SMC implementation during a pilot campaign in the health district of Koutiala, Mali. METHODS Starting in August 2012, children received three rounds of SMC with sulfadoxine-pyrimethamine (SP) and amodiaquine (AQ). From July 2013 onward, children received four rounds of SMC. Prevalence of malaria infection, clinical malaria and anemia were assessed during two cross-sectional surveys conducted in August 2012 and June 2014. Investigations involved 20 randomly selected clusters in 2012 against 10 clusters in 2014. RESULTS Overall, 662 children were included in 2012, and 670 in 2014. Children in 2014 versus those surveyed in 2012 showed reduced proportions of malaria infection (12.4% in 2014 versus 28.7% in 2012 (p = 0.001)), clinical malaria (0.3% versus 4.2%, respectively (p < 0.001)), and anemia (50.1% versus 67.4%, respectively (p = 0.001)). A propensity score approach that accounts for environmental differences showed that SMC conveyed a significant protective effect against malaria infection (IR = 0.01, 95% CI (0.0001; 0.09), clinical malaria (OR = 0.25, 95% CI (0.06; 0.85)), and hemoglobin concentration (β = 1.3, 95% CI (0.69; 1.96)) in 2012 and 2014, respectively. CONCLUSION SMC significantly reduced frequency of malaria infection, clinical malaria and anemia two years after SMC scale-up in Koutiala.
Collapse
Affiliation(s)
- Hamma Maiga
- Institut National de Santé Publique, Bamako BP: 1771, Mali;
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Jean Gaudart
- Aix Marseille Univ, APHM, IRD, INSERM, UMR1252 SESSTIM Sciences Economiques & Sociales de la Santé & Traitement de l’Information Médicale, Hop Timone, BioSTIC, Biostatistic & ICT, 13385 Marseille, France;
| | - Issaka Sagara
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
- Aix Marseille Univ, APHM, IRD, INSERM, UMR1252 SESSTIM Sciences Economiques & Sociales de la Santé & Traitement de l’Information Médicale, Hop Timone, BioSTIC, Biostatistic & ICT, 13385 Marseille, France;
| | - Modibo Diarra
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Amadou Bamadio
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Moussa Djimde
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Samba Coumare
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Boubou Sangare
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Yeyia Dicko
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Aly Tembely
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Djibril Traore
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Alassane Dicko
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| | - Estrella Lasry
- Médecins Sans Frontières (MSF), New York, NY 10006, USA;
| | - Ogobara Doumbo
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
- Aix Marseille Univ, APHM, IRD, INSERM, UMR1252 SESSTIM Sciences Economiques & Sociales de la Santé & Traitement de l’Information Médicale, Hop Timone, BioSTIC, Biostatistic & ICT, 13385 Marseille, France;
| | - Abdoulaye A. Djimde
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako BP: 1805, Mali; (I.S.); (M.D.); (A.B.); (M.D.); (S.C.); (B.S.); (Y.D.); (A.T.); (D.T.); (A.D.); (O.D.)
| |
Collapse
|
27
|
Abstract
Vaccine trials for infectious diseases take place in a milieu of trust in which scientists, regulatory institutions, and volunteers trust each other to play traditional roles. This milieu of trust emerges from a combination of preexisting linkages embedded in the local and national political context. Using the case of failed vaccine trials in Hohoe, Ghana, we explore this milieu of trust by employing the concept of tandems of trust and control, with a particular focus on the perceived characteristics of the disease and the linkages formed. An analysis of qualitative interviews collected in Hohoe following the West Africa Ebola outbreak of 2014-2016 shows that the trust/control nexus in vaccine trials precedes the implementation of those trials, while both the characteristics of Ebola and the political context shaped the formation and breakdown of relationships in the trial network.
Collapse
|
28
|
Esu EB, Oringanje C, Meremikwu MM, Cochrane Infectious Diseases Group. Intermittent preventive treatment for malaria in infants. Cochrane Database Syst Rev 2019; 12:CD011525. [PMID: 31792925 PMCID: PMC6887842 DOI: 10.1002/14651858.cd011525.pub2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Intermittent preventive treatment could help prevent malaria in infants (IPTi) living in areas of moderate to high malaria transmission in sub-Saharan Africa. The World Health Organization (WHO) policy recommended IPTi in 2010, but its adoption in countries has been limited. OBJECTIVES To evaluate the effects of intermittent preventive treatment (IPT) with antimalarial drugs to prevent malaria in infants living in malaria-endemic areas. SEARCH METHODS We searched the following sources up to 3 December 2018: the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (the Cochrane Library), MEDLINE (PubMed), Embase (OVID), LILACS (Bireme), and reference lists of articles. We also searched the metaRegister of Controlled Trials (mRCT) and the WHO International Clinical Trials Registry Platform (ICTRP) portal for ongoing trials up to 3 December 2018. SELECTION CRITERIA We included randomized controlled trials (RCTs) that compared IPT to placebo or no intervention in infants (defined as young children aged between 1 to 12 months) in malaria-endemic areas. DATA COLLECTION AND ANALYSIS The primary outcome was clinical malaria (fever plus asexual parasitaemia). Two review authors independently assessed trials for inclusion, evaluated the risk of bias, and extracted data. We summarized dichotomous outcomes and count data using risk ratios (RR) and rate ratios respectively, and presented all measures with 95% confidence intervals (CIs). We extracted protective efficacy values and their 95% CIs; when an included trial did not report this data, we calculated these values from the RR or rate ratio with its 95% CI. Where appropriate, we combined data in meta-analyses and assessed the certainty of the evidence using the GRADE approach. MAIN RESULTS We included 12 trials that enrolled 19,098 infants; all were conducted in sub-Saharan Africa. Three trials were cluster-RCTs. IPTi with sulfadoxine-pyrimethamine (SP) was evaluated in 10 trials from 1999 to 2013 (n = 15,256). Trials evaluating ACTs included dihydroartemisinin-piperaquine (1 trial, 147 participants; year 2013), amodiaquine-artesunate (1 study, 684 participants; year 2008), and SP-artesunate (1 trial, 676 participants; year 2008). The earlier studies evaluated IPTi with SP, and were conducted in Tanzania (in 1999 and 2006), Mozambique (2004), Ghana (2004 to 2005), Gabon (2005), Kenya (2008), and Mali (2009). One trial evaluated IPTi with amodiaquine in Tanzania (2000). Later studies included three conducted in Kenya (2008), Tanzania (2008), and Uganda (2013), evaluating IPTi in multiple trial arms that included artemisinin-based combination therapy (ACT). Although the effect size varied over time and between drugs, overall IPTi impacts on the incidence of clinical malaria overall, with a 27% reduction (rate ratio 0.73, 0.65 to 0.82; 10 studies, 10,602 participants). The effect of SP appeared to attenuate over time, with trials conducted after 2009 showing little or no effect of the intervention. IPTi with SP probably resulted in fewer episodes of clinical malaria (rate ratio 0.79, 0.74 to 0.85; 8 trials, 8774 participants, moderate-certainty evidence), anaemia (rate ratio 0.82, 0.68 to 0.98; 6 trials, 7438 participants, moderate-certainty evidence), parasitaemia (rate ratio 0.66, 0.56 to 0.79; 1 trial, 1200 participants, moderate-certainty evidence), and fewer hospital admissions (rate ratio 0.85, 0.78 to 0.93; 7 trials, 7486 participants, moderate-certainty evidence). IPTi with SP probably made little or no difference to all-cause mortality (risk ratio 0.93, 0.74 to 1.15; 9 trials, 14,588 participants, moderate-certainty evidence). Since 2009, IPTi trials have evaluated ACTs and indicate impact on clinical malaria and parasitaemia. A small trial of DHAP in 2013 shows substantive effects on clinical malaria (RR 0.42, 0.33 to 0.54; 1 trial, 147 participants, moderate-certainty evidence) and parasitaemia (moderate-certainty evidence). AUTHORS' CONCLUSIONS In areas of sub-Saharan Africa, giving antimalarial drugs known to be effective against the malaria parasite at the time to infants as IPT probably reduces the risk of clinical malaria, anaemia, and hospital admission. Evidence from SP studies over a 19-year period shows declining efficacy, which may be due to increasing drug resistance. Combinations with ACTs appear promising as suitable alternatives for IPTi. 2 December 2019 Up to date All studies incorporated from most recent search All eligible published studies found in the last search (3 Dec, 2018) were included.
Collapse
Affiliation(s)
- Ekpereonne B Esu
- College of Medical Sciences, University of CalabarDepartment of Public HealthCalabarCross River StateNigeria
| | - Chioma Oringanje
- University of TucsonGIDP Entomology and Insect ScienceTucsonArizonaUSA85721
| | - Martin M Meremikwu
- University of Calabar Teaching HospitalDepartment of PaediatricsPMB 1115CalabarCross River StateNigeria
| | | |
Collapse
|
29
|
Abuaku B, Duah-Quashie NO, Quaye L, Matrevi SA, Quashie N, Gyasi A, Owusu-Antwi F, Malm K, Koram K. Therapeutic efficacy of artesunate-amodiaquine and artemether-lumefantrine combinations for uncomplicated malaria in 10 sentinel sites across Ghana: 2015-2017. Malar J 2019; 18:206. [PMID: 31234874 PMCID: PMC6591907 DOI: 10.1186/s12936-019-2848-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/19/2019] [Indexed: 11/10/2022] Open
Abstract
Background Routine surveillance on the therapeutic efficacy of artemisinin-based combination therapy (ACT) has been ongoing in Ghana since 2005. The sixth round of surveillance was conducted between 2015 and 2017 to determine the therapeutic efficacy of artesunate–amodiaquine (AS–AQ) and artemether–lumefantrine (AL) in 10 sentinel sites across the country. Methods The study was a one-arm, prospective, evaluation of the clinical, parasitological, and haematological responses to directly observed treatment with AS–AQ and AL among children 6 months to 9 years old with uncomplicated falciparum malaria. The WHO 2009 protocol on surveillance of anti-malaria drug efficacy was used for the study with primary outcomes as prevalence of day 3 parasitaemia and clinical and parasitological cure rates on day 28. Secondary outcomes assessed included patterns of fever and parasite clearance as well as changes in haemoglobin concentration. Results Day 3 parasitaemia was absent in all sites following treatment with AS–AQ whilst only one person (0.2%) was parasitaemic on day 3 following treatment with AL. Day 28 PCR-corrected cure rates following treatment with AS–AQ ranged between 96.7% (95% CI 88.5–99.6) and 100%, yielding a national rate of 99.2% (95% CI 97.7–99.7). Day 28 PCR-corrected cure rates following treatment with AL ranged between 91.3% (95% CI 79.2–97.6) and 100%, yielding a national rate of 96% (95% CI 93.5–97.6). Prevalence of fever declined by 88.4 and 80.4% after first day of treatment with AS–AQ and AL, respectively, whilst prevalence of parasitaemia on day 2 was 2.1% for AS–AQ and 1.5% for AL. Gametocytaemia was maintained at low levels (< 5%) during the 3 days of treatment. Post-treatment mean haemoglobin concentration was significantly higher than pre-treatment concentration following treatment with either AS–AQ or AL. Conclusions The therapeutic efficacy of AS–AQ and AL is over 90% in sentinel sites across Ghana. The two anti-malarial drugs therefore remain efficacious in the treatment of uncomplicated malaria in the country and continue to achieve rapid fever and parasite clearance as well as low gametocyte carriage rates and improved post-treatment mean haemoglobin concentration. Electronic supplementary material The online version of this article (10.1186/s12936-019-2848-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benjamin Abuaku
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana.
| | - Nancy O Duah-Quashie
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana
| | - Lydia Quaye
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana
| | - Sena A Matrevi
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana
| | - Neils Quashie
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana.,Centre for Tropical Clinical Pharmacology and Therapeutics, University of Ghana Medical School, P. O. Box GP4236, Accra, Ghana
| | - Akosua Gyasi
- National Malaria Control Programme, Public Health Division, Ghana Health Service, Accra, Ghana
| | | | - Keziah Malm
- National Malaria Control Programme, Public Health Division, Ghana Health Service, Accra, Ghana
| | - Kwadwo Koram
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana
| |
Collapse
|
30
|
Castberg FC, Sarbah EW, Koram KA, Opoku N, Ofori MF, Styrishave B, Hviid L, Kurtzhals JAL. Malaria causes long-term effects on markers of iron status in children: a critical assessment of existing clinical and epidemiological tools. Malar J 2018; 17:464. [PMID: 30537973 PMCID: PMC6290551 DOI: 10.1186/s12936-018-2609-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/01/2018] [Indexed: 01/13/2023] Open
Abstract
Background Most epidemiological studies on the interplay between iron deficiency and malaria risk classify individuals as iron-deficient or iron-replete based on inflammation-dependent iron markers and adjustment for inflammation by using C-reactive protein (CRP) or α-1-acid glycoprotein (AGP). The validity of this approach and the usefulness of fibroblast growth factor 23 (FGF23) as a proposed inflammation-independent iron marker were tested. Methods Conventional iron markers and FGF23 were measured in children with acute falciparum malaria and after 1, 2, 4, and 6 weeks. Children, who were transfused or received iron supplementation in the follow-up period, were excluded, and iron stores were considered to be stable throughout. Ferritin levels 6 weeks after admission were used as a reference for admission iron status and compared with iron markers at different time points. Results There were long-term perturbations in iron markers during convalescence from acute malaria. None of the tested iron parameters, including FGF23, were independent of inflammation. CRP and AGP normalized faster than ferritin after malaria episodes. Conclusion Malaria may bias epidemiological studies based on inflammation-dependent iron markers. Better markers of iron status during and after inflammation are needed in order to test strategies for iron supplementation in populations at risk of malaria.
Collapse
Affiliation(s)
- Filip C Castberg
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Edem W Sarbah
- Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - Kwadwo A Koram
- Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - Nicholas Opoku
- Hohoe Municipality Hospital, Hohoe, Ghana.,School of Public Health, University of Health and Allied Sciences, Ho, Ghana
| | - Michael F Ofori
- Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - Bjarne Styrishave
- Toxicology and Drug Metabolism Group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Hviid
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Centre for Medical Parasitology, Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Jørgen A L Kurtzhals
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark.
| |
Collapse
|
31
|
Dinko B, Amakpa E, Kweku M, Amoah P, Tampuori J, Adjuik M, Awandare GA, Deitsch KW. Plasmodium falciparum malaria cases detected for prompt treatment by rapid diagnostic tests in the Ho Teaching Hospital of the Volta Region of Ghana. Parasite Epidemiol Control 2018; 3:e00072. [PMID: 29988323 PMCID: PMC6020104 DOI: 10.1016/j.parepi.2018.e00072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Prompt diagnosis and effective treatment of malaria cases with efficacious drugs is an important strategy in the management and control of malaria in endemic populations. As part of a study investigating the factors modulating the development of Plasmodium falciparum gametocytes in the human host, we assessed the rate of RDT positivity of patients in different departments of the Ho Teaching Hospital and the relation with age and anaemia. MATERIALS AND METHODS Eight-hundred and ten individuals attending clinic at various departments within the Ho Teaching Hospital were screened for malaria antigenaemia using RDT as a point-of-entry investigation. RDT positive individuals were immediately treated for malaria whereas RDT negative individuals were treated for other ailments. Haematological analyses were performed for 69 of these patients and the relationship between RDT results and haemoglobin levels were investigated. RESULTS The overall RDT positivity rate was 19.8% (160/810) of all individuals screened. There was no significant difference in the haemoglobin levels of RDT-positive and RDT-negative individuals (p value = 0.272). The highest number of attendees screened was children in the paediatric outpatient department and paediatric ward, 62% (507/810), with RDT positivity rate of 17% (91/507). We found the highest RDT positivity rate of 51% (19/37) in the male medical ward. CONCLUSIONS This study shows that RDT is a useful tool in promoting prompt diagnosis and management of malaria and though children form a majority of hospital attendees and malaria infections, the frequency of malaria detection may be higher in adults as compared to children.
Collapse
Affiliation(s)
- Bismarck Dinko
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Volta Region, Ghana
| | - Eric Amakpa
- Department of Obstetrics and Gyaenocology, Ho Teaching Hospital, Ho, Volta Regiona, Ghana
| | - Margaret Kweku
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Volta Region, Ghana
| | - Paul Amoah
- Diagnostic Laboratory, Ho Teaching Hospital, Ho, Volta Regiona, Ghana
| | - John Tampuori
- Department of Urology, Ho Teaching Hospital, Ho, Volta Regiona, Ghana
| | - Martin Adjuik
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Volta Region, Ghana
| | - Gordon A. Awandare
- Department of Biochemistry, Cell and Molecular Biology and West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon-Accra, Ghana
| | - Kirk W. Deitsch
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
32
|
Dinko B, Ansah F, Agyare-Kwabi C, Tagboto S, Amoah LE, Urban BC, Sutherland CJ, Awandare GA, Williamson KC, Binka FN, Deitsch KW. Gametocyte Development and Carriage in Ghanaian Individuals with Uncomplicated Plasmodium falciparum Malaria. Am J Trop Med Hyg 2018; 99:57-64. [PMID: 29692310 PMCID: PMC6085798 DOI: 10.4269/ajtmh.18-0077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/23/2018] [Indexed: 01/29/2023] Open
Abstract
Plasmodium falciparum gametocytes develop over 9-12 days while sequestered in deep tissues. On emergence into the bloodstream, they circulate for varied amounts of time during which certain host factors might influence their further development. We aimed to evaluate the potential association of patient clinical parameters with gametocyte development and carriage via in vivo methods. Seventy-two patients were enrolled from three hospitals in the Volta region of Ghana in 2016. Clinical parameters were documented for all patients, and gametocyte prevalence by microscopy was estimated at 12.5%. By measuring RNA transcripts representing two distinct gametocyte developmental stages using reverse transcriptase quantitative polymerase chain reaction (RT-qPCR), we obtained a more precise estimate of gametocyte carriage while also inferring gametocyte maturation. Fifty-three percent of the study participants harbored parasites expressing transcripts of the immature gametocyte-specific gene (PF3D7_1477700), whereas 36% harbored PF3D7_1438800 RNA-positive parasites, which is enriched in mid and mature gametocytes, suggesting the presence of more immature stages. Linear logistic regression showed that patients older than 5 years but less than 16 years were more likely to carry gametocytes expressing both PF3D7_1477700 and PF3D7_1438800 compared with younger participants, and gametocytemia was more likely in mildly anemic individuals compared with those with severe/moderate anemia. These data provide further evidence that a greater number of malaria patients harbor gametocytes than typically estimated by microscopy and suggest a possible association between age, fever, anemia, and gametocytemia.
Collapse
Affiliation(s)
- Bismarck Dinko
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Felix Ansah
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Comfort Agyare-Kwabi
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Senyo Tagboto
- Department of Internal Medicine, School of Medicine, University of Health and Allied Sciences, Ho, Volta Region, Ghana
| | - Linda Eva Amoah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Britta C. Urban
- Faculty of Biological Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Colin J. Sutherland
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Kim C. Williamson
- Microbiology and Immunology Department, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Fred N. Binka
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Volta Region, Ghana
| | - Kirk W. Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York
| |
Collapse
|
33
|
Kinetics of antibody responses to PfRH5-complex antigens in Ghanaian children with Plasmodium falciparum malaria. PLoS One 2018; 13:e0198371. [PMID: 29883485 PMCID: PMC5993283 DOI: 10.1371/journal.pone.0198371] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/17/2018] [Indexed: 11/21/2022] Open
Abstract
Plasmodium falciparum PfRH5 protein binds Ripr, CyRPA and Pf113 to form a complex that is essential for merozoite invasion of erythrocytes. The inter-genomic conservation of the PfRH5 complex proteins makes them attractive blood stage vaccine candidates. However, little is known about how antibodies to PfRH5, CyRPA and Pf113 are acquired and maintained in naturally exposed populations, and the role of PfRH5 complex proteins in naturally acquired immunity. To provide such data, we studied 206 Ghanaian children between the ages of 1–12 years, who were symptomatic, asymptomatic or aparasitemic and healthy. Plasma levels of antigen-specific IgG and IgG subclasses were measured by ELISA at several time points during acute disease and convalescence. On the day of admission with acute P. falciparum malaria, the prevalence of antibodies to PfRH5-complex proteins was low compared to other merozoite antigens (EBA175, GLURP-R0 and GLURP-R2). At convalescence, the levels of RH5-complex-specific IgG were reduced, with the decay of PfRH5-specific IgG being slower than the decay of IgG specific for CyRPA and Pf113. No correlation between IgG levels and protection against P. falciparum malaria was observed for any of the PfRH5 complex proteins. From this we conclude that specific IgG was induced against proteins from the PfRH5-complex during acute P. falciparum malaria, but the prevalence was low and the IgG levels decayed rapidly after treatment. These data indicate that the levels of IgG specific for PfRH5-complex proteins in natural infections in Ghanaian children were markers of recent exposure only.
Collapse
|
34
|
Druetz T. Evaluation of direct and indirect effects of seasonal malaria chemoprevention in Mali. Sci Rep 2018; 8:8104. [PMID: 29802375 PMCID: PMC5970148 DOI: 10.1038/s41598-018-26474-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/10/2018] [Indexed: 01/24/2023] Open
Abstract
Randomized controlled trials have established that seasonal malaria chemoprevention (SMC) in children is a promising strategy to reduce malaria transmission in Sahelian West Africa. This strategy was recently introduced in a dozen countries, and about 12 million children received SMC in 2016. However, evidence on SMC effectiveness under routine programme conditions is sparse. We aim to measure the effects of the nationwide SMC programme in Mali on the prevalence of malaria and anemia in children 6-59 months. We used data from the 2015 nationally representative malaria indicator survey. A post-test only with non-randomized control group study was designed. We fitted a generalized structural equation model that controlled for potential bias on observed and non-observed variables (endogenous treatment effect model). Having received SMC reduced by 44% (95% CI [0.39-0.49]) the risk of having a positive rapid diagnostic test for malaria. In addition, the programme indirectly reduced by 18% the risk of moderate-to-severe anemia (95% CI [0.15-0.21]). SMC in Mali has substantial protective effects under routine nationwide programme conditions. Endogenous treatment effects analyses can contribute to rigorously measuring the effectiveness of health programmes and to bridging a widening gap in evaluation methods to measure progress towards achieving malaria elimination.
Collapse
Affiliation(s)
- Thomas Druetz
- Center for Applied Malaria Research and Evaluation, Department of Tropical Medicine, Tulane University, New Orleans, USA.
- Department of Social and Preventive Medicine, School of Public Health, University of Montreal, Montreal, Canada.
| |
Collapse
|
35
|
Thera MA, Kone AK, Tangara B, Diarra E, Niare S, Dembele A, Sissoko MS, Doumbo OK. School-aged children based seasonal malaria chemoprevention using artesunate-amodiaquine in Mali. Parasite Epidemiol Control 2018; 3:96-105. [PMID: 29988270 PMCID: PMC6011810 DOI: 10.1016/j.parepi.2018.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 11/01/2022] Open
Abstract
INTRODUCTION Malaria is still a public health problem in Africa. Seasonal Malaria Chemoprevention (SMC) is an efficient control strategy recommended by WHO that targets children under five year old living in areas of seasonal malaria transmission. SMC uses the combination amodiaquine (AQ) - sulfadoxine-pyrimethamine (SP). However SP selects rapidly drug resistant parasites. And malaria burden may increase in older children where SMC is implemented. We initiated a pilot study to assess an alternative approach to SMC in older children in Mali. METHODS A randomized open-label clinical trial was conducted to test the efficacy and safety of SMC using artesunate - amodiaquine in school aged children in Mali. Two hundred pupils aged 6-15 years old were enrolled and randomized into two arms of 100 each, to receive either artesunate-amodiaquine (ASAQ) monthly or no intervention. Both arms were followed and clinical malaria were diagnosed and treated with arthemeter-lumefanthrine as recommended by Mali National Malaria Control Program. ASAQ was administered 3 days under study team direct observation and during 4 consecutive months starting in October 2013. Follow up was continued until April 2014. RESULTS Overall, 20 cases of uncomplicated clinical malaria were encountered in the Control arm and three cases in the ASAQ arm, showing a protective efficacy of 85% 95% CI [80.1-89.9] against clinical malaria. Protective efficacy against malaria infection was 69.6% 95% CI [58.6-21.4]. No effect on anemia was observed. ASAQ was well tolerated. Most common solicited adverse events were abdominal pain and headaches of mild intensity in respectively 64% and 44% of children that swallowed ASAQ. CONCLUSION ASAQ is effective and well tolerated as SMC targeting older children in a peri urban setting in Mali. Its administration at schools is a feasible and accepted strategy to deliver the intervention.
Collapse
Affiliation(s)
- Mahamadou A. Thera
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| | - Abdoulaye K. Kone
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| | - Bourama Tangara
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| | - Elizabeth Diarra
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| | - Sirama Niare
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| | - Abdramane Dembele
- Service of Psychiatry, University and Hospital Center of Point G, Bamako, Mali
| | - Mahamadou S. Sissoko
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| | - Ogobara K. Doumbo
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, USTTB, Point G, BP 1805 Bamako, Mali
| |
Collapse
|
36
|
Wangdi K, Furuya-Kanamori L, Clark J, Barendregt JJ, Gatton ML, Banwell C, Kelly GC, Doi SAR, Clements ACA. Comparative effectiveness of malaria prevention measures: a systematic review and network meta-analysis. Parasit Vectors 2018; 11:210. [PMID: 29587882 PMCID: PMC5869791 DOI: 10.1186/s13071-018-2783-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/06/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Malaria causes significant morbidity and mortality worldwide. There are several preventive measures that are currently employed, including insecticide-treated nets (ITNs, including long-lasting insecticidal nets and insecticidal-treated bed nets), indoor residual spraying (IRS), prophylactic drugs (PD), and untreated nets (UN). However, it is unclear which measure is the most effective for malaria prevention. We therefore undertook a network meta-analysis to compare the efficacy of different preventive measures on incidence of malaria infection. METHODS A systematic literature review was undertaken across four medical and life sciences databases (PubMed, Cochrane Central, Embase, and Web of Science) from their inception to July 2016 to compare the effectiveness of different preventive measures on malaria incidence. Data from the included studies were analysed for the effectiveness of several measures against no intervention (NI). This was carried out using an automated generalized pairwise modeling (GPM) framework for network meta-analysis to generate mixed treatment effects against a common comparator of no intervention (NI). RESULTS There were 30 studies that met the inclusion criteria from 1998-2016. The GPM framework led to a final ranking of effectiveness of measures in the following order from best to worst: PD, ITN, IRS and UN, in comparison with NI. However, only ITN (RR: 0.49, 95% CI: 0.32-0.74) showed precision while other methods [PD (RR: 0.24, 95% CI: 0.004-15.43), IRS (RR: 0.55, 95% CI: 0.20-1.56) and UN (RR: 0.73, 95% CI: 0.28-1.90)] demonstrating considerable uncertainty associated with their point estimates. CONCLUSION Current evidence is strong for the protective effect of ITN interventions in malaria prevention. Even though ITNs were found to be the only preventive measure with statistical support for their effectiveness, the role of other malaria control measures may be important adjuncts in the global drive to eliminate malaria.
Collapse
Affiliation(s)
- Kinley Wangdi
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Luis Furuya-Kanamori
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Justin Clark
- Centre for Research in Evidence-Based Practice (CREBP), Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland Australia
| | - Jan J. Barendregt
- School of Public Health, The University of Queensland, Brisbane, Queensland Australia
- Epigear International Pty Ltd, Sunrise Beach, Queensland Australia
| | - Michelle L. Gatton
- School of Public Health & Social Work, Queensland University of Technology, Brisbane, Queensland Australia
| | - Cathy Banwell
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Gerard C. Kelly
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Suhail A. R. Doi
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Archie C. A. Clements
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| |
Collapse
|
37
|
Druetz T, Corneau-Tremblay N, Millogo T, Kouanda S, Ly A, Bicaba A, Haddad S. Impact Evaluation of Seasonal Malaria Chemoprevention under Routine Program Implementation: A Quasi-Experimental Study in Burkina Faso. Am J Trop Med Hyg 2017; 98:524-533. [PMID: 29260654 PMCID: PMC5929206 DOI: 10.4269/ajtmh.17-0599] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Seasonal malaria chemoprevention (SMC) for children < 5 is a strategy that is gaining popularity in West African countries. Although its efficacy to reduce malaria incidence has been demonstrated in trials, the effects of SMC implemented in routine program conditions, outside of experimental contexts, are unknown. In 2014 and 2015, a survey was conducted in 1,311 households located in Kaya District (Burkina Faso) where SMC had been recently introduced. All children < 72 months were tested for malaria and anemia. A pre–post study with control group was designed to measure SMC impact during high transmission season. A difference-in-differences approach was coupled in the analysis with propensity score weighting to control for observable and time-invariant nonobservable confounding factors. SMC reduced the parasitemia point and period prevalence by 3.3 and 24% points, respectively; this translated into protective effects of 51% and 62%. SMC also reduced the likelihood of having moderate to severe anemia by 32%, and history of recent fever by 46%. Self-reported coverage for children at the first cycle was 83%. The SMC program was successfully added to a package of interventions already in place. To our knowledge, with prevalence < 10% during the peak of the transmission season, this is the first time that malaria can be reported as hypo-endemic in a sub-Sahelian setting in Burkina Faso. SMC has great potential, and along with other interventions, it could contribute to approaching the threshold where elimination strategies will be envisioned in Burkina Faso.
Collapse
Affiliation(s)
- Thomas Druetz
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | | | - Tieba Millogo
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
| | - Seni Kouanda
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
| | - Antarou Ly
- Department of Preventive and Social Medicine, Faculty of Medicine, Laval University, Quebec City, Canada
| | - Abel Bicaba
- Société d'Études et de Recherches en Santé Publique, Ouagadougou, Burkina Faso
| | - Slim Haddad
- Department of Preventive and Social Medicine, Faculty of Medicine, Laval University, Quebec City, Canada
| |
Collapse
|
38
|
Coldiron ME, Von Seidlein L, Grais RF. Seasonal malaria chemoprevention: successes and missed opportunities. Malar J 2017; 16:481. [PMID: 29183327 PMCID: PMC5704360 DOI: 10.1186/s12936-017-2132-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/23/2017] [Indexed: 11/10/2022] Open
Abstract
Seasonal malaria chemoprevention (SMC) was recommended in 2012 for young children in the Sahel during the peak malaria transmission season. Children are given a single dose of sulfadoxine/pyrimethamine combined with a 3-day course of amodiaquine, once a month for up to 4 months. Roll-out and scale-up of SMC has been impressive, with 12 million children receiving the intervention in 2016. There is evidence of its overall benefit in routine implementation settings, and a meta-analysis of clinical trial data showed a 75% decrease in clinical malaria compared to placebo. SMC is not free of shortcomings. Its target zone includes many hard-to-reach areas, both because of poor infrastructure and because of political instability. Treatment adherence to a 3-day course of preventive treatment has not been fully documented, and could prove challenging. As SMC is scaled up, integration into a broader, community-based paradigm which includes other preventive and curative activities may prove beneficial, both for health systems and for recipients.
Collapse
|
39
|
Maiga H, Lasry E, Diarra M, Sagara I, Bamadio A, Traore A, Coumare S, Bahonan S, Sangare B, Dicko Y, Diallo N, Tembely A, Traore D, Niangaly H, Dao F, Haidara A, Dicko A, Doumbo OK, Djimde AA. Seasonal Malaria Chemoprevention with Sulphadoxine-Pyrimethamine and Amodiaquine Selects Pfdhfr-dhps Quintuple Mutant Genotype in Mali. PLoS One 2016; 11:e0162718. [PMID: 27662368 PMCID: PMC5035027 DOI: 10.1371/journal.pone.0162718] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/26/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Seasonal malaria chemoprevention (SMC) with sulphadoxine-pyrimethamine (SP) plus amodiaquine (AQ) is being scaled up in Sahelian countries of West Africa. However, the potential development of Plasmodium falciparum resistance to the respective component drugs is a major concern. METHODS Two cross-sectional surveys were conducted before (August 2012) and after (June 2014) a pilot implementation of SMC in Koutiala, Mali. Children aged 3-59 months received 7 rounds of curative doses of SP plus AQ over two malaria seasons. Genotypes of P. falciparum Pfdhfr codons 51, 59 and 108; Pfdhps codons 437 and 540, Pfcrt codon 76 and Pfmdr1codon 86 were analyzed by PCR on DNA from samples collected before and after SMC, and in non-SMC patient population as controls (November 2014). RESULTS In the SMC population 191/662 (28.9%) and 85/670 (12.7%) of children were P. falciparum positive by microscopy and were included in the molecular analysis before (2012) and after SMC implementation (2014), respectively. In the non-SMC patient population 220/310 (71%) were successfully PCR analyzed. In the SMC children, the prevalence of all molecular markers of SP resistance increased significantly after SMC including the Pfdhfr-dhps quintuple mutant genotype, which was 1.6% before but 7.1% after SMC (p = 0.02). The prevalence of Pfmdr1-86Y significantly decreased from 26.7% to 15.3% (p = 0.04) while no significant change was seen for Pfcrt 76T. In 2014, prevalence of all molecular markers of SP resistance were significantly higher among SMC children compared to the non-SMC population patient (p < 0.01). No Pfdhfr-164 mutation was found neither at baseline nor post SMC. CONCLUSION SMC increased the prevalence of molecular markers of P. falciparum resistance to SP in the treated children. However, there was no significant increase of these markers of resistance in the general parasite population after 2 years and 7 rounds of SMC.
Collapse
Affiliation(s)
- Hamma Maiga
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Estrella Lasry
- Médecins Sans Frontières (MSF), New York, New York, United States of America
| | - Modibo Diarra
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Amadou Bamadio
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Aliou Traore
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Samba Coumare
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | | | - Boubou Sangare
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Yeyia Dicko
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Nouhoum Diallo
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Aly Tembely
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Djibril Traore
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Hamidou Niangaly
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - François Dao
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Aboubecrine Haidara
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Alassane Dicko
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
| | - Abdoulaye A. Djimde
- Malaria Research and Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Odontostomatology and Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Mali
- * E-mail:
| |
Collapse
|
40
|
Opoku EC, Olsen A, Browne E, Hodgson A, Awoonor-Williams JK, Yelifari L, Williams J, Magnussen P. Impact of combined intermittent preventive treatment of malaria and helminths on anaemia, sustained attention, and recall in Northern Ghanaian schoolchildren. Glob Health Action 2016; 9:32197. [PMID: 27633035 PMCID: PMC5025525 DOI: 10.3402/gha.v9.32197] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/03/2016] [Accepted: 08/19/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The benefits of integrated control of malaria, schistosomiasis, and soil-transmitted helminth infections have not been fully explored in Ghanaian schoolchildren. OBJECTIVE To assess the impact of co-administered artemether-lumefantrine plus albendazole, and artemether-lumefantrine plus albendazole plus praziquantel compared to albendazole plus praziquantel on anaemia, sustained attention, and recall in schoolchildren. DESIGN This three-arm, open-label intervention study was carried out in Ghana among class three schoolchildren. Artemether-lumefantrine and albendazole were co-administered to 131 schoolchildren in Study Arm 1; artemether-lumefantrine, albendazole, and praziquantel to 90 children in Study Arm 2 versus albendazole and praziquantel to 127 children in Control Arm 3. Medicines were administered to all children at least 30 min after a meal. A HemoCue(®) photometer was used to measure haemoglobin (Hb), while the code transmission test (CTT), adapted from the Test of Everyday Attention for Children (TEA-Ch), was used to measure sustained attention and recall before-and-after interventions in June 2011 and June 2012. RESULTS We observed significant malaria parasite prevalence reductions of 62.8 and 59.2% in Study Arm 1 from 24.2 to 9.0%, p<0.01, and 59.2% in Study Arm 2 from 26.7 to 10.9%, p<0.01), respectively, compared to 8.93% in Control Arm 3 (from 34.7 to 31.6%, p>0.05). Meanwhile, anaemia prevalence reduced significantly (p<0.01) in all three study arms after interventions by 38.4% (from 19.8 to 12.2%), 20.7% (from 26.6 to 21.1%), and 36.0% (from 28.3 to 18.1%) in Study Arms 1, 2, and 3, respectively. Although the interventions had no significant effects on Hb levels, anaemia prevalence reduced insignificantly by 38.4 and 20.7% in Study Arms 1 and 2, respectively, compared to 36.0% in Control Arm 3. Among schoolchildren in Study Arms 1 and 2, mean CTT score improved significantly after interventions by 10.4% (from 3.18 to 3.55, p=0.01) and 20.5% (from 2.83 to 3.56, p=0.01) respectively, compared to 5.75% in Control Arm 3 (from 2.95 to 3.13, p=0.09). Likewise, mean recall test score improvements after interventions were 16.9% (from 2.07 to 2.49, p=0.01) and 27.9% (from 1.91 to 2.65, p=0.01) in Study Arms 1 and 2, respectively, compared to 18.3% (from 1.92 to 2.35, p=0.01) in Control Arm 3. CONCLUSION Combined intermittent preventive treatment of malaria and deworming reduced prevalence of anaemia and improved sustained attention and recall in schoolchildren. Best results for sustained attention and recall were seen in Study Arm 2.
Collapse
Affiliation(s)
- Ernest Cudjoe Opoku
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana.,Copenhagen School of Global Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Annette Olsen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Edmund Browne
- Department of Community Health, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Abraham Hodgson
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana
| | | | - Lawrence Yelifari
- Regional Health Directorate, Ghana Health Service, Bolgatanga, Ghana
| | - John Williams
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana
| | - Pascal Magnussen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Onyeneho NG, Igweonu OU. Anaemia is typical of pregnancies: capturing community perception and management of anaemia in pregnancy in Anambra State, Nigeria. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2016; 35:29. [PMID: 27581730 PMCID: PMC5026019 DOI: 10.1186/s41043-016-0066-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/20/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Anaemia during pregnancy continues to constitute significant challenge to maternal health in Nigeria and contributes substantially to the worsening maternal mortality ratio (MMR) in Nigeria despite a global reduction in MMR in response to effort to improve safe motherhood. The incidence of anaemia during pregnancy is still high (>40 %) in Nigeria, and attitudes and management practices are yet unclear as the peoples' understanding of the phenomenon remains unclear. This study explored the perceptions/attitudes on anaemia during pregnancy and practices to prevent and/or manage it in Anambra State. METHODS In-depth interview and focus group discussion data were collected from health workers and mothers who delivered within 6 months preceding the study and from mothers and husbands of women who delivered within 6 months preceding the study, respectively. RESULTS The people expressed some knowledge of anaemia, being common in pregnancies. However, some expressed the view that anaemia being a typical sign of pregnancy cannot be prevented. Some mothers expressed desire for focused antenatal care services to control anaemia but lamented the attitude of the health workers, who make access to these interventions difficult. CONCLUSIONS Control of anaemia in pregnancy should start with providing health education to pregnant women and their partners, who reinforce what the women are told during antenatal care, and with training health workers for friendlier attitudes to clients.
Collapse
Affiliation(s)
- Nkechi G. Onyeneho
- Department of Sociology/Anthropology, University of Nigeria, Nsukka, Enugu State Nigeria
- Takemi Program in International Health, Department of Global Health and Population, Harvard TH Chan School of Public Health, 677 Huntington Avenue, Boston, MA USA
| | - Obianuju U. Igweonu
- Social Science Unit, School of General Studies, University of Nigeria, Nsukka, Enugu State Nigeria
| |
Collapse
|
42
|
Dinko B, Ayivor-Djanie R, Abugri J, Agboli E, Kye-Duodu G, Tagboto S, Tampuori J, Adzaku F, Binka FN, Awandare GA. Comparison of malaria diagnostic methods in four hospitals in the Volta region of Ghana. MALARIAWORLD JOURNAL 2016; 7:5. [PMID: 38601360 PMCID: PMC11003212 DOI: 10.5281/zenodo.10797112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Background Rapid diagnostic tests (RDTs) and microscopy are routinely used for the diagnosis of malaria in Ghana. DNA-based polymerase chain reaction (PCR) is not yet used routinely. We compared diagnostic methods and tested the sensitivities of different malaria diagnostic methods against PCR. Materials and methods Study participants from four hospitals with a suspicion of malaria donated finger -prick blood for RDT and blood film examination. In addition, a blood spot was collected for PCR analysis, prior to treatment. Retrospective species-specific PCR was performed on all samples collected. Results Using PCR we found an overall malaria prevalence of 39% among the 211 evaluable blood spots (83/211) and this ranged between 6-61% across the four hospitals. Of the 164 participants with RDT data, malaria prevalence was 57% (94/164), ranging from 3-100% from the four hospitals. Microscopy was the least sensitive with a parasite prevalence of 21% (25/119) of the evaluable 119 participants, varying from 9 to 35% across three health facilities. By comparison, we found the sensitivities and specificities of RDT results when compared to PCR to be slightly higher than microscopy compared to PCR. These were 56.4% versus 41.7% and 90% versus 81.9%, respectively, but generally lower than expected. Ninety-five percent of the PCR-detected infections were P. falciparum, while 4% were mixed species infections of P. falciparum and P. malariae, with the remaining being a mono-infection of P. malariae. Conclusions While using PCR as a gold standard, we found RDT to be more reliable in diagnosing malaria than microscopy. In addition, a majority of malaria-treated cases were not supported by PCR diagnosis, leading to possible overtreatment. Pragmatic strategies are needed to ensure suspected malaria cases are accurately diagnosed before treatment.
Collapse
Affiliation(s)
- Bismarck Dinko
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Reuben Ayivor-Djanie
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - James Abugri
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Eric Agboli
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Ghana
| | - Gideon Kye-Duodu
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Ghana
| | - Senyo Tagboto
- Department of Internal Medicine, School of Medicine, University of Health and Allied Sciences, Ho, Ghana
| | - John Tampuori
- Department of Urology, Volta Regional Hospital, Ho, Ghana
| | - Festus Adzaku
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Fred N Binka
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Ghana
| | - Gordon A Awandare
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
43
|
Hamma M. Impact of seasonal malaria chemoprevention of sulphadoxinepyrimethamine plus amodiaquine on molecular markers resistance of Plasmodium falciparum malaria: A review in West Africa. ACTA ACUST UNITED AC 2016. [DOI: 10.5897/cro15.0098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
44
|
Abuaku B, Duah N, Quaye L, Quashie N, Malm K, Bart-Plange C, Koram K. Therapeutic efficacy of artesunate-amodiaquine and artemether-lumefantrine combinations in the treatment of uncomplicated malaria in two ecological zones in Ghana. Malar J 2016; 15:6. [PMID: 26728096 PMCID: PMC4700572 DOI: 10.1186/s12936-015-1080-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/29/2015] [Indexed: 11/25/2022] Open
Abstract
Background Case management based on prompt diagnosis and adequate treatment using artemisinin-based combination therapy (ACT) remains the main focus of malaria control in Ghana. As part of routine surveillance on the therapeutic efficacy of ACT in Ghana, the efficacy of amodiaquine-artesunate (AS-AQ) and artemether-lumefantrine (AL) were studied in six sentinel sites representing the forest and savannah zones of the country. Methods Three sites representing the two ecological zones studied AS-AQ whilst the other three sites studied AL. In each site, the study was a one-arm prospective evaluation of the clinical, parasitological, and haematological responses to directly observed therapy for uncomplicated malaria with either AS-AQ or AL among children aged 6 months and 9 years. The WHO 2009 protocol for monitoring anti-malarial drug efficacy was used for the study between July 2013 and March 2014. Results Per-protocol analyses on day 28 showed an overall PCR-corrected cure rate of 100 % for AS-AQ and 97.6 % (95 % CI 93.1, 99.5) for AL: 97.2 % (95 % CI 92.0, 99.4) in the forest zone and 100 % in the savannah zone. Kaplan–Meier survival analysis showed similar outcomes. Prevalence of fever decreased by about 75 % after the first day of treatment with each ACT in the two ecological zones. No child studied was parasitaemic on day 3, and gametocytaemia was generally maintained at low levels (<5 %). Post-treatment mean haemoglobin concentrations significantly increased in the two ecological zones. Conclusions Therapeutic efficacy of AS-AQ and AL remains over 90 % in the forest and savannah zones of Ghana. Additionally, post-treatment parasitaemia on day 3 is rare suggesting that artemisinin is still efficacious in Ghana.
Collapse
Affiliation(s)
- Benjamin Abuaku
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Ghana.
| | - Nancy Duah
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Ghana.
| | - Lydia Quaye
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Ghana.
| | - Neils Quashie
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Ghana. .,Centre for Tropical Clinical Pharmacology and Therapeutics, University of Ghana Medical School, P. O. Box GP4236, Accra, Ghana.
| | - Keziah Malm
- National Malaria Control Programme, Public Health Division, Ghana Health Service, Accra, Ghana.
| | - Constance Bart-Plange
- National Malaria Control Programme, Public Health Division, Ghana Health Service, Accra, Ghana.
| | - Kwadwo Koram
- Epidemiology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Ghana.
| |
Collapse
|
45
|
Tagbor H, Antwi GD, Acheampong PR, Bart Plange C, Chandramohan D, Cairns M. Seasonal malaria chemoprevention in an area of extended seasonal transmission in Ashanti, Ghana: an individually randomised clinical trial. Trop Med Int Health 2015; 21:224-35. [PMID: 26578353 PMCID: PMC4982104 DOI: 10.1111/tmi.12642] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Objective To investigate the effectiveness of seasonal malaria chemoprevention (SMC) and community case management with long‐acting artemisinin‐based combination therapies (ACTs) for the control of malaria in areas of extended seasonal malaria transmission. Method Individually randomised, placebo‐controlled trial in the Ashanti Region of Ghana. A total of 2400 children aged 3–59 months received either: (i) a short‐acting ACT for case management of malaria (artemether‐lumefantrine, AL) plus placebo SMC, or (ii) a long‐acting ACT (dihydroartemisinin‐piperaquine, DP) for case management plus placebo SMC or (iii) AL for case management plus active SMC with sulphadoxine‐pyrimethamine and amodiaquine. SMC or placebo was delivered on five occasions during the rainy season. Malaria cases were managed by community health workers, who used rapid diagnostic tests to confirm infection prior to treatment. Results The incidence of malaria was lower in children given SMC during the rainy season. Compared to those given placebo SMC and AL for case management, the adjusted hazard ratio (aHR) was 0.62 (95% CI: 0.41, 0.93), P = 0.020 by intention to treat and 0.53 (95% CI: 0.29, 0.95), P = 0.033 among children given five SMC courses. There were no major differences between groups given different ACTs for case management (aHR DP vs. AL 1.18 (95% CI 0.83, 1.67), P = 0.356). Conclusion SMC may have an important public health impact in areas with a longer transmission season, but further optimisation of SMC schedules is needed to maximise its impact in such settings.
Collapse
Affiliation(s)
- Harry Tagbor
- School of Public Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Centre for Global Health Research, Juaben, Ghana
| | | | | | | | - Daniel Chandramohan
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
| | - Matthew Cairns
- MRC Tropical Epidemiology Group, London School of hygiene and tropical medicine, London, UK
| |
Collapse
|
46
|
Matangila JR, Mitashi P, Inocêncio da Luz RA, Lutumba PT, Van Geertruyden JP. Efficacy and safety of intermittent preventive treatment for malaria in schoolchildren: a systematic review. Malar J 2015; 14:450. [PMID: 26574017 PMCID: PMC4647321 DOI: 10.1186/s12936-015-0988-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/04/2015] [Indexed: 11/10/2022] Open
Abstract
Background Intermittent preventive treatment (IPT) is a proven malaria control strategy in infants and pregnancy. School-aged children represent 26 % of the African population, and an increasing percentage of them are scholarized. Malaria is causing 50 % of deaths in this age group and malaria control efforts may shift the malaria burden to older age groups. Schools have been suggested as a platform for health interventions delivery (deworming, iron-folic acid, nutrients supplementation, (boost-)immunization) and as a possible delivery system for IPT in schoolchildren (IPTsc). However, the current evidence on the efficacy and safety of IPTsc is limited and the optimal therapeutic regimen remains controversial. Methods A systematic search for studies reporting efficacy and safety of IPT in schoolchildren was conducted using PubMed, Web of Science, Clinicaltrials and WHO/ICTRP database, and abstracts from congresses with the following key words: intermittent, preventive treatment AND malaria OR Plasmodium falciparum AND schoolchildren NOT infant NOT pregnancy. Results Five studies were identified. Most IPTsc regimes demonstrated substantial protection against malaria parasitaemia, with dihydroartemisinin-piperaquine (DP) given monthly having the highest protective effect (PE) (94 %; 95 % CI 93–96). Contrarily, SP did not provide any PE against parasitaemia. However, no IPT regimen provided a PE above 50 % in regard to anaemia, and highest protection was provided by SP+ amodiaquine (AQ) given four-monthly (50 %; 95 % CI 41–53). The best protection against clinical malaria was observed in children monthly treated with DP (97 %; 95 % CI 87–98). However, there was no protection when the drug was given three-monthly. No severe adverse events were associated with the drugs used for IPTsc. Conclusion IPTsc may reduce the malaria-related burden in schoolchildren. However, more studies assessing efficacy of IPT in particular against malaria-related anaemia and clinical malaria in schoolchildren must be conducted.
Collapse
Affiliation(s)
- Junior R Matangila
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, BP 747, Kinshasa, XI, Democratic Republic of the Congo. .,Epidemiology for Global Health Institute, University of Antwerp, Campus DrieEiken, Universiteitsplein 1, Wilrijk, 2610, Belgium.
| | - Patrick Mitashi
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, BP 747, Kinshasa, XI, Democratic Republic of the Congo.
| | - Raquel A Inocêncio da Luz
- Epidemiology for Global Health Institute, University of Antwerp, Campus DrieEiken, Universiteitsplein 1, Wilrijk, 2610, Belgium.
| | - Pascal T Lutumba
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, BP 747, Kinshasa, XI, Democratic Republic of the Congo.
| | - Jean-Pierre Van Geertruyden
- Epidemiology for Global Health Institute, University of Antwerp, Campus DrieEiken, Universiteitsplein 1, Wilrijk, 2610, Belgium.
| |
Collapse
|
47
|
Athuman M, Kabanywanyi AM, Rohwer AC, Cochrane Infectious Diseases Group. Intermittent preventive antimalarial treatment for children with anaemia. Cochrane Database Syst Rev 2015; 1:CD010767. [PMID: 25582096 PMCID: PMC4447115 DOI: 10.1002/14651858.cd010767.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Anaemia is a global public health problem. Children under five years of age living in developing countries (mostly Africa and South-East Asia) are highly affected. Although the causes for anaemia are multifactorial, malaria has been linked to anaemia in children living in malaria-endemic areas. Administering intermittent preventive antimalarial treatment (IPT) to children might reduce anaemia, since it could protect children from new Plasmodium parasite infection (the parasites that cause malaria) and allow their haemoglobin levels to recover. OBJECTIVES To assess the effect of IPT for children with anaemia living in malaria-endemic areas. SEARCH METHODS We searched the Cochrane Infectious Diseases Group Specialized Register, Cochrane Central of Controlled Trials (CENTRAL), published in The Cochrane Library; MEDLINE; EMBASE; and LILACS. We also searched the World Health Organization (WHO) International Clinical Trial Registry Platform and metaRegister of Controlled Trials (mRCT) for ongoing trials up to 4 December 2014. SELECTION CRITERIA Randomized controlled trials (RCTs) evaluating the effect of IPT on children with anaemia. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed risk of bias. We analysed data by conducting meta-analyses, stratifying data according to whether participants received iron supplements or not. We used GRADE to assess the quality of evidence. MAIN RESULTS Six trials with 3847 participants met our inclusion criteria. Trials were conducted in areas of low malaria endemicity (three trials), and moderate to high endemicity (three trials). Four trials were in areas of seasonal malaria transmission. Iron was given to all children in two trials, and evaluated in a factorial design in a further two trials.IPT for children with anaemia probably has little or no effect on the proportion anaemic at 12 weeks follow-up (four trials, 2237 participants, (moderate quality evidence).IPT in anaemic children probably increases the mean change in haemoglobin levels from baseline to follow-up at 12 weeks on average by 0.32 g/dL (MD 0.32, 95% CI 0.19 to 0.45; four trials, 1672 participants, moderate quality evidence); and may improve haemoglobin levels at 12 weeks (MD 0.35, 95% CI 0.06 to 0.64; four trials, 1672 participants, low quality evidence). For both of these outcomes, subgroup analysis did not demonstrate a difference between children receiving iron and those that did not.IPT for children with anaemia probably has little or no effect on mortality or hospital admissions at six months (three trials, 3160 participants moderate quality evidence). Subgroup analysis did not show a difference between those children receiving iron supplements and those that did not. AUTHORS' CONCLUSIONS Trials did show a small effect on average haemoglobin levels but this did not appear to translate into an effect on mortality and hospital admissions. Three of the six trials were conducted in low endemicity areas where transmission is low and thus any protective effect is likely to be modest.
Collapse
Affiliation(s)
| | - Abdunoor M Kabanywanyi
- Ifakara Health InstituteP O Box 78373Kiko Avenue, Old Bagamoyo RoadDar‐es‐salaamTanzania
| | - Anke C Rohwer
- Stellenbosch UniversityCentre for Evidence‐based Health Care, Faculty of Medicine and Health SciencesFrancie van Zijl DriveCape TownSouth Africa7505
| | | |
Collapse
|
48
|
Hamdan M, Brabin B, Bates I. Implications of inconsistent anaemia policies for children and adolescents in Africa. Public Health Nutr 2014; 17:2587-94. [PMID: 24477312 PMCID: PMC10282439 DOI: 10.1017/s1368980013003121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 10/15/2013] [Accepted: 10/20/2013] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To evaluate the quality of policies concerning the diagnosis, treatment and prevention of anaemia in children and adolescents; to determine to what extent these are evidence-based; and to use this analysis to inform the policy-making process. SUBJECTS Children and adolescents in sub-Saharan Africa. SETTING Almost 50 % of children and adolescents in sub-Saharan Africa are anaemic, which has profound effects on their intellectual and physical development and their chance of survival. Evidence-based policies are essential to reduce anaemia but because it is caused by an array of interdependent factors, developing policies is challenging. DESIGN Forty-six policy documents concerning the diagnosis, treatment and prevention of anaemia in children and adolescents were identified and analysed. RESULTS There was policy consensus on the usefulness of Fe supplements, the need to treat co-morbidities and the use of blood transfusions for severe anaemia. Information about diagnosis was scarce, and messages regarding the control of anaemia were mixed. Few of the policies were tailored for the African context and they were located on several websites hosted by different health programmes. CONCLUSIONS The weakest aspects of the policies and consequently the priorities for better policy making were: lack of adherence to WHO recommendations for guideline development; little involvement of African practitioners/policy makers in the guideline group and as peer reviewers; and lack of harmonisation, demonstrating the need to establish a single body responsible for developing/revising anaemia policies.
Collapse
Affiliation(s)
- Musa Hamdan
- Medicins Sans Frontières, Amsterdam, The Netherlands
| | - Bernard Brabin
- Tropical Paediatrics, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Imelda Bates
- Tropical Clinical Haematology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| |
Collapse
|
49
|
Nankabirwa JI, Wandera B, Amuge P, Kiwanuka N, Dorsey G, Rosenthal PJ, Brooker SJ, Staedke SG, Kamya MR. Impact of intermittent preventive treatment with dihydroartemisinin-piperaquine on malaria in Ugandan schoolchildren: a randomized, placebo-controlled trial. Clin Infect Dis 2014; 58:1404-12. [PMID: 24621953 PMCID: PMC4001293 DOI: 10.1093/cid/ciu150] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Dihydroartemisinin-piperaquine administered at monthly intervals, but not that dosed once a school term, is a remarkably effective measure for the prevention of incidence of malaria, prevalence of parasitemia, and prevalence of anemia in schoolchildren living in a high-transmission setting. Background. Intermittent preventive treatment (IPT) in schoolchildren offers a promising option for malaria control. However, the optimal drug and dosing regimens for IPT remain to be determined. Methods. We conducted a randomized, double-blind, placebo-controlled trial in 740 schoolchildren aged 6–14 years living in a setting of high malaria transmission in Uganda. Enrolled children were randomized to dihydroartemisinin-piperaquine (DP) given once a month (IPTm), DP given once a school term (4 treatments over 12 months, IPTst), or placebo and followed for 12 months. The primary outcome was the incidence of malaria over 12 months. Secondary outcomes included parasite prevalence and anemia over 12 months. Analyses were conducted on an intention-to-treat basis. Results. In the placebo arm, the incidence of malaria was 0.34 episodes per person-year and the prevalence of parasitemia and anemia was 38% and 20%, respectively. IPTm reduced the incidence of malaria by 96% (95% confidence interval [CI], 88%–99%, P < .0001), the prevalence of asymptomatic parasitemia by 94% (95% CI, 92%–96%, P < .0001), and the prevalence of anemia by 40% (95% CI, 19%–56%, P < .0001). IPTst had no significant effect on the incidence of symptomatic malaria or the prevalence of anemia, but reduced the prevalence of asymptomatic parasitemia by 54% (95% CI, 47%–60%, P < .0001). Conclusions. Monthly IPT with DP offered remarkable protection against clinical malaria, parasitemia, and anemia in schoolchildren living in a high-malaria-transmission setting. Clinical Trials Registration. NCT01231880.
Collapse
|
50
|
Gosling RD, Cairns ME, Chico RM, Chandramohan D. Intermittent preventive treatment against malaria: an update. Expert Rev Anti Infect Ther 2014; 8:589-606. [DOI: 10.1586/eri.10.36] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|