1
|
Kagaya W. Low-density Plasmodium falciparum infection: "Even a parasite will turn". Parasitol Int 2025; 107:103052. [PMID: 39986449 DOI: 10.1016/j.parint.2025.103052] [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: 10/31/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
As global malaria control and elimination efforts have resulted in the suppression of Plasmodium falciparum infections, low-density Plasmodium falciparum infections present a significant challenge. These infections, frequently characterized as "submicroscopic" or "asymptomatic", contribute to the persistent transmission in endemic regions. Recent advancements in molecular diagnostic methodologies have enhanced the detection of these infections and elucidated the nature of previously unrecognized infections. These infections harbor smaller populations of parasites; however, the risk of disease progression and transmission remains substantial. The observed infections could be an alternative survival mechanism of this parasite. Thus, control measures should be redesigned to address these infections, rather than merely expanding the current tools. This review provides an overview of the issues surrounding the detection and monitoring of these infections and their importance for infected individuals and populations, with further emphasis on control measures for malaria elimination.
Collapse
Affiliation(s)
- Wataru Kagaya
- Department of Eco-Epidemiology, Institute of Tropical Medicine (Nekken), Nagasaki University, 1-12-4, Sakamoto, Nagasaki, Nagasaki 852-8523, Japan.
| |
Collapse
|
2
|
Yukich J, Doum D, McIver DJ, Richardson JH, Sovannanoroth S, Lobo NF, Tatarsky A. Willingness to pay for a mosquito bite prevention 'forest pack' in Cambodia: results of a discrete choice experiment. Malar J 2024; 23:392. [PMID: 39702135 DOI: 10.1186/s12936-024-05224-2] [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: 07/24/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Progress towards malaria elimination in the Greater Mekong Subregion has left much of the residual malaria transmission concentrated among forest-exposed populations for whom traditional domicile focused malaria vector control is unlikely to be effective. New tools to protect these populations from vector biting outdoors are needed. METHODS Alongside implementation research on the deployment of a "forest pack" consisting of a volatile pyrethroid (transfluthrin)-based spatial repellent (VPSR), a picaridin-based topical repellent and etofenprox treatment of clothing, an assessment was made of participant willingness to pay for the forest packs and variants of the packs using a discrete choice experiment. RESULTS Participants showed willingness to pay for forest packs consistent with full-cost recovery for VPSR devices. The inclusion of a full malaria season's worth of VPSR devices increased the willingness to pay for a forest pack by 15% (p = 0.061). At a price of approximately 10 USD, approximately 50% of participants were willing to pay for a forest pack which included a full season's worth of VPSR. CONCLUSION Forest packs which include VPSR are likely to be acceptable to the target forest-exposed populations, and those which include VPSR products may even have potential for commercial sales or some cost-recovery.
Collapse
Affiliation(s)
- Joshua Yukich
- Tulane University School of Public Health and Tropical Medicine, New Orleans, USA
| | - Dyna Doum
- Health Forefront Organization, Phnom Penh, Cambodia
| | - David J McIver
- Malaria Elimination Initiative, Institute for Global Health Sciences, University of California San Francisco, San Francisco, USA.
| | | | - Siv Sovannanoroth
- Cambodia National Centre for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Neil F Lobo
- Malaria Elimination Initiative, Institute for Global Health Sciences, University of California San Francisco, San Francisco, USA
- Eck Institute for Global Health, University of Notre Dame, South Bend, USA
| | - Allison Tatarsky
- Malaria Elimination Initiative, Institute for Global Health Sciences, University of California San Francisco, San Francisco, USA
| |
Collapse
|
3
|
Lek D, Sokomar N, Samphornarann T, Rideout J, Hassan SED, Bunkea T, Ath SS, Seng R, Hustedt J, Peto TJ, Hughes J, Kimmen K, Dy K, Adhikari B. Impact of targeted drug administration and intermittent preventive treatment for forest goers using artesunate-pyronaridine to control malaria outbreaks in Cambodia. Trop Med Health 2024; 52:42. [PMID: 38863067 PMCID: PMC11165738 DOI: 10.1186/s41182-024-00607-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024] Open
Abstract
INTRODUCTION The national malaria programme of Cambodia targets the rapid elimination of all human malaria by 2025. As clinical cases decline to near-elimination levels, a key strategy is the rapid identification of malaria outbreaks triggering effective action to interrupt local transmission. We report a comprehensive, multipronged management approach in response to a 2022 Plasmodium falciparum outbreak in Kravanh district, western Cambodia. METHODS The provincial health department of Pursat in conjunction with the Center for Parasitology, Entomology and Malaria Control (CNM) identified villages where transmission was occurring using clinical records, and initiated various interventions, including the distribution of insecticide-treated bed nets, running awareness campaigns, and implementing fever screening with targeted drug administration. Health stations were set up at forest entry points, and later, targeted drug administrations with artesunate-pyronaridine (Pyramax) and intermittent preventive treatment for forest goers (IPTf) were implemented in specific village foci. Data related to adherence and adverse events from IPTf and TDA were collected. The coverage rates of interventions were calculated, and local malaria infections were monitored. RESULTS A total of 942 individuals were screened through active fever surveillance in villages where IPTf and TDA were conducted. The study demonstrated high coverage and adherence rates in the targeted villages, with 92% (553/600) coverage in round one and 65% (387/600) in round two. Adherence rate was 99% (551/553) in round one and 98% (377/387) in round two. The study found that forest goers preferred taking Pyramax over repeated testing consistent with the coverage rates: 92% in round one compared to 65% in round two. All individuals reachable through health stations or mobile teams reported complete IPTf uptake. No severe adverse events were reported. Only six individuals reported mild adverse events, such as loss of energy, fever, abdominal pain, diarrhoea, and muscle aches. Two individuals attributed their symptoms to heavy alcohol intake following prophylaxis. CONCLUSIONS The targeted malaria outbreak response demonstrated high acceptability, safety, and feasibility of the selected interventions. Malaria transmission was rapidly controlled using the available community resources. This experience suggests the effectiveness of the programmatic response for future outbreaks.
Collapse
Affiliation(s)
- Dysoley Lek
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia.
- School of Public Health, National Institute of Public Health, Phnom Penh, Cambodia.
| | - Nguon Sokomar
- Cambodia Malaria Elimination Project2, University Research Company Ltd., Phnom Penh, Cambodia
| | - Top Samphornarann
- Cambodia Malaria Elimination Project2, University Research Company Ltd., Phnom Penh, Cambodia
| | - Jeanne Rideout
- Cambodia Malaria Elimination Project2, University Research Company Ltd., Phnom Penh, Cambodia
| | - Saad El-Din Hassan
- Cambodia Malaria Elimination Project2, University Research Company Ltd., Phnom Penh, Cambodia
| | - Tol Bunkea
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Saing Sam Ath
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Rothpisey Seng
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - John Hustedt
- Cambodia Malaria Elimination Project2, University Research Company Ltd., Phnom Penh, Cambodia
- FHI 360, Phnom Penh, Cambodia
| | - Thomas J Peto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jayme Hughes
- Clinton Health Access Initiative, Phnom Penh, Cambodia
| | - Ke Kimmen
- Provincial Health Department, Pursat, Cambodia
| | - Khoy Dy
- Clinton Health Access Initiative, Phnom Penh, Cambodia
| | - Bipin Adhikari
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Conradis-Jansen F, Tripura R, Peto TJ, Callery JJ, Adhikari B, Ean M, Jongdeepaisal M, Pell C, Khonputsa P, Murgia R, Sovannaroth S, Müller O, Cheah PY, Dondorp AM, von Seidlein L, Maude RJ. Community engagement among forest goers in a malaria prophylaxis trial: implementation challenges and implications. Malar J 2023; 22:178. [PMID: 37291578 PMCID: PMC10248338 DOI: 10.1186/s12936-023-04610-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Malaria transmission in Southeast Asia is increasingly confined to forests, where marginalized groups are exposed primarily through their work. Anti-malarial chemoprophylaxis may help to protect these people. This article examines the effectiveness and practical challenges of engaging forest-goers to participate in a randomized controlled clinical trial of anti-malarial chemoprophylaxis with artemether-lumefantrine (AL) versus a control (multivitamin, MV) for malaria in northeast Cambodia. METHODS The impact of engagement in terms of uptake was assessed as the proportion of people who participated during each stage of the trial: enrolment, compliance with trial procedures, and drug intake. During the trial, staff recorded the details of engagement meetings, including the views and opinions of participants and community representatives, the decision-making processes, and the challenges addressed during implementation. RESULTS In total, 1613 participants were assessed for eligibility and 1480 (92%) joined the trial, 1242 (84%) completed the trial and received prophylaxis (AL: 82% vs MV: 86%, p = 0.08); 157 (11%) were lost to follow-up (AL: 11% vs MV: 11%, p = 0.79); and 73 (5%) discontinued the drug (AL-7% vs MV-3%, p = 0.005). The AL arm was associated with discontinuation of the study drug (AL: 48/738, 7% vs 25/742, 3%; p = 0.01). Females (31/345, 9%) were more likely (42/1135, 4%) to discontinue taking drugs at some point in the trial (p = 0.005). Those (45/644, 7%) who had no previous history of malaria infection were more likely to discontinue the study drug than those (28/836, 3%) who had a history of malaria (p = 0.02). Engagement with the trial population was demanding because many types of forest work are illegal; and the involvement of an engagement team consisting of representatives from the local administration, health authorities, community leaders and community health workers played a significant role in building trust. Responsiveness to the needs and concerns of the community promoted acceptability and increased confidence in taking prophylaxis among participants. Recruitment of forest-goer volunteers to peer-supervise drug administration resulted in high compliance with drug intake. The development of locally-appropriate tools and messaging for the different linguistic and low-literacy groups was useful to ensure participants understood and adhered to the trial procedures. It was important to consider forest-goers` habits and social characteristics when planning the various trial activities. CONCLUSIONS The comprehensive, participatory engagement strategy mobilized a wide range of stakeholders including study participants, helped build trust, and overcame potential ethical and practical challenges. This locally-adapted approach was highly effective as evidenced by high levels of trial enrolment, compliance with trial procedures and drug intake.
Collapse
Affiliation(s)
- Franca Conradis-Jansen
- Heidelberg Institute of Global Health, Medical School, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - James J Callery
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Mom Ean
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Monnaphat Jongdeepaisal
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Christopher Pell
- Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Global Health, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Global Health Program, Amsterdam, The Netherlands
| | - Panarasri Khonputsa
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Riccardo Murgia
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Siv Sovannaroth
- Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Olaf Müller
- Heidelberg Institute of Global Health, Medical School, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Richard J Maude
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
- The Open University, Milton Keynes, UK.
| |
Collapse
|
5
|
Chang MA, Impoinvil D, Hamre KES, Dalexis PE, Mérilien JB, Dismer AM, Fouché B, Desir L, Holmes K, Lafortune W, Herman C, Rogier E, Noland GS, Young AJ, Druetz T, Ashton R, Eisele TP, Cohen J, van den Hoogen L, Stresman G, Drakeley C, Pothin E, Cameron E, Battle KE, Williamson J, Telfort MA, Lemoine JF. Acceptability, Feasibility, Drug Safety, and Effectiveness of a Pilot Mass Drug Administration with a Single Round of Sulfadoxine-Pyrimethamine Plus Primaquine and Indoor Residual Spraying in Communities with Malaria Transmission in Haiti, 2018. Am J Trop Med Hyg 2023; 108:1127-1139. [PMID: 37160282 PMCID: PMC10540127 DOI: 10.4269/ajtmh.22-0623] [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: 09/28/2022] [Accepted: 02/24/2023] [Indexed: 05/11/2023] Open
Abstract
For a malaria elimination strategy, Haiti's National Malaria Control Program piloted a mass drug administration (MDA) with indoor residual spraying (IRS) in 12 high-transmission areas across five communes after implementing community case management and strengthened surveillance. The MDA distributed sulfadoxine-pyrimethamine and single low-dose primaquine to eligible residents during house visits. The IRS campaign applied pirimiphos-methyl insecticide on walls of eligible houses. Pre- and post-campaign cross-sectional surveys were conducted to assess acceptability, feasibility, drug safety, and effectiveness of the combined interventions. Stated acceptability for MDA before the campaign was 99.2%; MDA coverage estimated at 10 weeks post-campaign was 89.6%. Similarly, stated acceptability of IRS at baseline was 99.9%; however, household IRS coverage was 48.9% because of the high number of ineligible houses. Effectiveness measured by Plasmodium falciparum prevalence at baseline and 10 weeks post-campaign were similar: 1.31% versus 1.43%, respectively. Prevalence of serological markers were similar at 10 weeks post-campaign compared with baseline, and increased at 6 months. No severe adverse events associated with the MDA were identified in the pilot; there were severe adverse events in a separate, subsequent campaign. Both MDA and IRS are acceptable and feasible interventions in Haiti. Although a significant impact of a single round of MDA/IRS on malaria transmission was not found using a standard pre- and post-intervention comparison, it is possible there was blunting of the peak transmission. Seasonal malaria transmission patterns, suboptimal IRS coverage, and low baseline parasitemia may have limited the effectiveness or the ability to measure effectiveness.
Collapse
Affiliation(s)
- Michelle A. Chang
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Daniel Impoinvil
- Entomology Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Karen E. S. Hamre
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
- CDC Foundation, Atlanta, Georgia
| | | | - Jean-Baptiste Mérilien
- Programme National de Contrôle de la Malaria, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - Amber M. Dismer
- Emergency Response and Recovery Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | - Kathleen Holmes
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Willy Lafortune
- Programme National de Contrôle de la Malaria, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - Camelia Herman
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Eric Rogier
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Alyssa J. Young
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Thomas Druetz
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Ruth Ashton
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Thomas P. Eisele
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Justin Cohen
- Clinton Health Access Initiative, Washington, District of Columbia
| | | | - Gillian Stresman
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Chris Drakeley
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Emilie Pothin
- Clinton Health Access Initiative, Washington, District of Columbia
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ewan Cameron
- School of Public Health, Curtin University, Bentley, Australia
| | - Katherine E. Battle
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, Washington
| | - John Williamson
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Marc-Aurèle Telfort
- Programme National de Contrôle de la Malaria, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - Jean Frantz Lemoine
- Programme National de Contrôle de la Malaria, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| |
Collapse
|
6
|
Moss S, Mańko E, Krishna S, Campino S, Clark TG, Last A. How has mass drug administration with dihydroartemisinin-piperaquine impacted molecular markers of drug resistance? A systematic review. Malar J 2022; 21:186. [PMID: 35690758 PMCID: PMC9188255 DOI: 10.1186/s12936-022-04181-y] [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: 01/17/2022] [Accepted: 05/10/2022] [Indexed: 11/10/2022] Open
Abstract
The World Health Organization (WHO) recommends surveillance of molecular markers of resistance to anti-malarial drugs. This is particularly important in the case of mass drug administration (MDA), which is endorsed by the WHO in some settings to combat malaria. Dihydroartemisinin-piperaquine (DHA-PPQ) is an artemisinin-based combination therapy which has been used in MDA. This review analyses the impact of MDA with DHA-PPQ on the evolution of molecular markers of drug resistance. The review is split into two parts. Section I reviews the current evidence for different molecular markers of resistance to DHA-PPQ. This includes an overview of the prevalence of these molecular markers in Plasmodium falciparum Whole Genome Sequence data from the MalariaGEN Pf3k project. Section II is a systematic literature review of the impact that MDA with DHA-PPQ has had on the evolution of molecular markers of resistance. This systematic review followed PRISMA guidelines. This review found that despite being a recognised surveillance tool by the WHO, the surveillance of molecular markers of resistance following MDA with DHA-PPQ was not commonly performed. Of the total 96 papers screened for eligibility in this review, only 20 analysed molecular markers of drug resistance. The molecular markers published were also not standardized. Overall, this warrants greater reporting of molecular marker prevalence following MDA implementation. This should include putative pfcrt mutations which have been found to convey resistance to DHA-PPQ in vitro.
Collapse
Affiliation(s)
- Sophie Moss
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | - Emilia Mańko
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Sanjeev Krishna
- Institute of Infection and Immunity, St George's University of London, London, UK
| | - Susana Campino
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Taane G Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Anna Last
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
7
|
Bassat Q, Maïga-Ascofaré O, May J, Clain J, Mombo-Ngoma G, Groger M, Adegnika AA, Agobé JCD, Djimde A, Mischlinger J, Ramharter M. Challenges in the clinical development pathway for triple and multiple drug combinations in the treatment of uncomplicated falciparum malaria. Malar J 2022; 21:61. [PMID: 35193586 PMCID: PMC8864855 DOI: 10.1186/s12936-022-04079-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/06/2022] [Indexed: 11/23/2022] Open
Abstract
The addition of a third anti-malarial drug matching the pharmacokinetic characteristics of the slowly eliminated partner drug in artemisinin-based combination therapy (ACT) has been proposed as new therapeutic paradigm for the treatment of uncomplicated falciparum malaria. These triple artemisinin-based combination therapy (TACT) should in theory more effectively prevent the development and spread of multidrug resistance than current ACT. Several clinical trials evaluating TACT—or other multidrug anti-malarial combination therapy (MDACT)—have been reported and more are underway. From a regulatory perspective, these clinical development programmes face a strategic dilemma: pivotal clinical trials evaluating TACT are designed to test for non-inferiority of efficacy compared to standard ACT as primary endpoint. While meeting the endpoint of non-inferior efficacy, TACT are consistently associated with a slightly higher frequency of adverse drug reactions than currently used ACT. Moreover, the prevention of the selection of specific drug resistance—one of the main reasons for TACT development—is beyond the scope of even large-scale clinical trials. This raises important questions: if equal efficacy is combined with poorer tolerability, how can then the actual benefit of these drug combinations be demonstrated? How should clinical development plans be conceived to provide objective evidence for or against an improved management of patients and effective prevention of anti-malarial drug resistance by TACT? What are the objective criteria to ultimately convince regulators to approve these new products? In this Opinion paper, the authors discuss the challenges for the clinical development of triple and multidrug anti-malarial combination therapies and the hard choices that need to be taken in the further clinical evaluation and future implementation of this new treatment paradigm.
Collapse
Affiliation(s)
- Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ICREA, Passeig de Lluís Companys 23, 08010, Barcelona, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Oumou Maïga-Ascofaré
- Kumasi Center for Collaborative Research, Kumasi, Ghana.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Partner Site Hamburg- Lübeck-Borstel-Riems, Hamburg- Lübeck-Borstel-Riems, Germany
| | - Jürgen May
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Partner Site Hamburg- Lübeck-Borstel-Riems, Hamburg- Lübeck-Borstel-Riems, Germany
| | - Jerôme Clain
- Université de Paris, Centre National de Référence du Paludisme, Hôpital Bichat- Claude Bernard, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Ghyslain Mombo-Ngoma
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Partner Site Hamburg- Lübeck-Borstel-Riems, Hamburg- Lübeck-Borstel-Riems, Germany.,Centre de Recherches de Lambaréné, Lambaréné, Gabon
| | - Mirjam Groger
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Partner Site Hamburg- Lübeck-Borstel-Riems, Hamburg- Lübeck-Borstel-Riems, Germany
| | - Ayôla A Adegnika
- Centre de Recherches de Lambaréné, Lambaréné, Gabon.,Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | | | - Abdoulaye Djimde
- Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Johannes Mischlinger
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Partner Site Hamburg- Lübeck-Borstel-Riems, Hamburg- Lübeck-Borstel-Riems, Germany
| | - Michael Ramharter
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany. .,German Center for Infection Research, Partner Site Hamburg- Lübeck-Borstel-Riems, Hamburg- Lübeck-Borstel-Riems, Germany. .,Centre de Recherches de Lambaréné, Lambaréné, Gabon.
| | | |
Collapse
|
8
|
Abstract
BACKGROUND Studies evaluating mass drug administration (MDA) in malarious areas have shown reductions in malaria immediately following the intervention. However, these effects vary by endemicity and are not sustained. Since the 2013 version of this Cochrane Review on this topic, additional studies have been published. OBJECTIVES Primary objectives To assess the sustained effect of MDA with antimalarial drugs on: - the reduction in malaria transmission in moderate- to high-transmission settings; - the interruption of transmission in very low- to low-transmission settings. Secondary objective To summarize the risk of drug-associated adverse effects following MDA. SEARCH METHODS We searched several trial registries, citation databases, conference proceedings, and reference lists for relevant articles up to 11 February 2021. We also communicated with researchers to identify additional published and unpublished studies. SELECTION CRITERIA Randomized controlled trials (RCTs) and non-randomized studies comparing MDA to no MDA with balanced co-interventions across study arms and at least two geographically distinct sites per study arm. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trials for eligibility and extracted data. We calculated relative risk (RR) and rate ratios with corresponding 95% confidence intervals (CIs) to compare prevalence and incidence, respectively, in MDA compared to no-MDA groups. We stratified analyses by malaria transmission and by malaria species. For cluster-randomized controlled trials (cRCTs), we adjusted standard errors using the intracluster correlation coefficient. We assessed the certainty of the evidence using the GRADE approach. For non-randomized controlled before-and-after (CBA) studies, we summarized the data using difference-in-differences (DiD) analyses. MAIN RESULTS Thirteen studies met our criteria for inclusion. Ten were cRCTs and three were CBAs. Cluster-randomized controlled trials Moderate- to high-endemicity areas (prevalence ≥ 10%) We included data from two studies conducted in The Gambia and Zambia. At one to three months after MDA, the Plasmodium falciparum (hereafter, P falciparum) parasitaemia prevalence estimates may be higher compared to control but the CIs included no effect (RR 1.76, 95% CI 0.58 to 5.36; Zambia study; low-certainty evidence); parasitaemia incidence was probably lower (RR 0.61, 95% CI 0.40 to 0.92; The Gambia study; moderate-certainty evidence); and confirmed malaria illness incidence may be substantially lower, but the CIs included no effect (rate ratio 0.41, 95% CI 0.04 to 4.42; Zambia study; low-certainty evidence). At four to six months after MDA, MDA showed little or no effect on P falciparum parasitaemia prevalence (RR 1.18, 95% CI 0.89 to 1.56; The Gambia study; moderate-certainty evidence) and, no persisting effect was demonstrated with parasitaemia incidence (rate ratio 0.91, 95% CI 0.55 to 1.50; The Gambia study). Very low- to low-endemicity areas (prevalence < 10%) Seven studies from Cambodia, Laos, Myanmar (two studies), Vietnam, Zambia, and Zanzibar evaluated the effects of multiple rounds of MDA on P falciparum. Immediately following MDA (less than one month after MDA), parasitaemia prevalence was reduced (RR 0.12, 95% CI 0.03 to 0.52; one study; low-certainty evidence). At one to three months after MDA, there was a reduction in both parasitaemia incidence (rate ratio 0.37, 95% CI 0.21 to 0.55; 1 study; moderate-certainty evidence) and prevalence (RR 0.25, 95% CI 0.15 to 0.41; 7 studies; low-certainty evidence). For confirmed malaria incidence, absolute rates were low, and it is uncertain whether MDA had an effect on this outcome (rate ratio 0.58, 95% CI 0.12 to 2.73; 2 studies; very low-certainty evidence). For P falciparum prevalence, the relative differences declined over time, from RR 0.63 (95% CI 0.36 to 1.12; 4 studies) at four to six months after MDA, to RR 0.86 (95% CI 0.55 to 1.36; 5 studies) at 7 to 12 months after MDA. Longer-term prevalence estimates showed overall low absolute risks, and relative effect estimates of the effect of MDA on prevalence varied from RR 0.82 (95% CI 0.20 to 3.34) at 13 to 18 months after MDA, to RR 1.25 (95% CI 0.25 to 6.31) at 31 to 36 months after MDA in one study. Five studies from Cambodia, Laos, Myanmar (2 studies), and Vietnam evaluated the effect of MDA on Plasmodium vivax (hereafter, P vivax). One month following MDA, P vivax prevalence was lower (RR 0.18, 95% CI 0.08 to 0.40; 1 study; low-certainty evidence). At one to three months after MDA, there was a reduction in P vivax prevalence (RR 0.15, 95% CI 0.10 to 0.24; 5 studies; low-certainty evidence). The immediate reduction on P vivax prevalence was not sustained over time, from RR 0.78 (95% CI 0.63 to 0.95; 4 studies) at four to six months after MDA, to RR 1.12 (95% CI 0.94 to 1.32; 5 studies) at 7 to 12 months after MDA. One of the studies in Myanmar provided estimates of longer-term effects, where overall absolute risks were low, ranging from RR 0.81 (95% CI 0.44 to 1.48) at 13 to 18 months after MDA, to RR 1.20 (95% CI 0.44 to 3.29) at 31 to 36 months after MDA. Non-randomized studies Three CBA studies were conducted in moderate- to high-transmission areas in Burkina Faso, Kenya, and Nigeria. There was a reduction in P falciparum parasitaemia prevalence in MDA groups compared to control groups during MDA (DiD range: -15.8 to -61.4 percentage points), but the effect varied at one to three months after MDA (DiD range: 14.9 to -41.1 percentage points). AUTHORS' CONCLUSIONS: In moderate- to high-transmission settings, no studies reported important effects on P falciparum parasitaemia prevalence within six months after MDA. In very low- to low-transmission settings, parasitaemia prevalence and incidence were reduced initially for up to three months for both P falciparum and P vivax; longer-term data did not demonstrate an effect after four months, but absolute risks in both intervention and control groups were low. No studies provided evidence of interruption of malaria transmission.
Collapse
Affiliation(s)
- Monica P Shah
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jimee Hwang
- U.S. President's Malaria Initiative, Malaria Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
- Global Health Group, University of California San Francisco, San Francisco, USA
| | - Leslie Choi
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Kim A Lindblade
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - S Patrick Kachur
- Department of Population and Family Health, Columbia University Medical Center, New York, NY, USA
| | - Meghna Desai
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
9
|
Efforts Made to Eliminate Drug-Resistant Malaria and Its Challenges. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5539544. [PMID: 34497848 PMCID: PMC8421183 DOI: 10.1155/2021/5539544] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023]
Abstract
Since 2000, a good deal of progress has been made in malaria control. However, there is still an unacceptably high burden of the disease and numerous challenges limiting advancement towards its elimination and ultimate eradication. Among the challenges is the antimalarial drug resistance, which has been documented for almost all antimalarial drugs in current use. As a result, the malaria research community is working on the modification of existing treatments as well as the discovery and development of new drugs to counter the resistance challenges. To this effect, many products are in the pipeline and expected to be marketed soon. In addition to drug and vaccine development, mass drug administration (MDA) is under scientific scrutiny as an important strategy for effective utilization of the developed products. This review discusses the challenges related to malaria elimination, ongoing approaches to tackle the impact of drug-resistant malaria, and upcoming antimalarial drugs.
Collapse
|
10
|
Rasmussen C, Alonso P, Ringwald P. Current and emerging strategies to combat antimalarial resistance. Expert Rev Anti Infect Ther 2021; 20:353-372. [PMID: 34348573 DOI: 10.1080/14787210.2021.1962291] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Since the spread of chloroquine resistance in Plasmodium falciparum in the 1960s, recommendations have been made on how to respond to antimalarial resistance. Only with the advent of artemisinin partial resistance were large scale efforts made in the Greater Mekong Subregion to carry out recommendations in a coordinated and well-funded manner. Independent emergence of parasites partially resistant to artemisinins has now been reported in Rwanda. AREAS COVERED We reviewed past recommendations and activities to respond to resistance as well as the research ongoing into new ways to stop or delay the spread of resistant parasites. EXPERT OPINION Inadequate information limits the options and support for a strong, coordinated response to artemisinin partial resistance in Africa, making better phenotypic and genotypic surveillance a priority. A response to resistance needs to address factors that may have hastened the emergence and could speed the spread, including overuse of drugs and lack of access to quality treatment. New ways to use the existing treatments in the response to resistance such as multiple first-lines are currently impeded by the limited number of drugs available.
Collapse
Affiliation(s)
| | - Pedro Alonso
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | - Pascal Ringwald
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| |
Collapse
|
11
|
Maude RJ, Tripura R, Ean M, Sokha M, Peto TJ, Callery JJ, Imwong M, Vongpromek R, Tarning J, Mukaka M, Waithira N, Soviet O, von Seidlein L, Sovannaroth S. Study protocol: an open-label individually randomised controlled trial to assess the efficacy of artemether-lumefantrine prophylaxis for malaria among forest goers in Cambodia. BMJ Open 2021; 11:e045900. [PMID: 34233975 PMCID: PMC8264911 DOI: 10.1136/bmjopen-2020-045900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/15/2021] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION In the Greater Mekong Subregion, adults are at highest risk for malaria. The most relevant disease vectors bite during daytime and outdoors which makes forest work a high-risk activity for malaria. The absence of effective vector control strategies and limited periods of exposure during forest visits suggest that chemoprophylaxis could be an appropriate strategy to protect forest goers against malaria. METHODS AND ANALYSIS The protocol describes an open-label randomised controlled trial of artemether-lumefantrine (AL) versus multivitamin as prophylaxis against malaria among forest goers aged 16-65 years in rural northeast Cambodia. The primary objective is to compare the efficacy of the artemisinin combination therapy AL versus a multivitamin preparation as defined by the 28-day PCR parasite positivity rate and incidence of confirmed clinical malaria of any species. The sample size is 2200 patient-episodes of duration 1 month in each arm. The duration of follow-up and prophylaxis for each participant is 1, 2 or 3 consecutive 28-day periods, followed by a further 28 days of post-exposure prophylaxis, depending on whether they continue to visit the forest. Analysis will be done both by intention to treat and per protocol. ETHICS AND DISSEMINATION All participants will provide written, informed consent. Ethical approval was obtained from the Oxford Tropical Research Ethics Committee and the Cambodia National Ethics Committee for Health Research. Results will be disseminated by peer-reviewed open access publication together with open data. TRIAL REGISTRATION NUMBER NCT04041973; Pre-result.
Collapse
Affiliation(s)
- Richard James Maude
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
- The Open University, Milton Keynes, UK
| | - Rupam Tripura
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mom Ean
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Meas Sokha
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thomas Julian Peto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James John Callery
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mallika Imwong
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ranitha Vongpromek
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Asia-Pacific Regional Centre, WorldWide Antimalarial Resistance Network, Bangkok, Thailand
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mavuto Mukaka
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Naomi Waithira
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Oung Soviet
- Provincial Health Department, Stung Treng, Cambodia
| | - Lorenz von Seidlein
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Siv Sovannaroth
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| |
Collapse
|
12
|
Vilakati S, Mngadi N, Benjamin-Chung J, Dlamini N, Dufour MSK, Whittemore B, Bhangu K, Prach LM, Baltzell K, Nhlabathi N, Malambe C, Dlamini B, Helb D, Greenhouse B, Maphalala G, Pindolia D, Kalungero M, Tesfa G, Gosling R, Ntshalintshali N, Kunene S, Hsiang MS. Effectiveness and safety of reactive focal mass drug administration (rfMDA) using dihydroartemisinin-piperaquine to reduce malaria transmission in the very low-endemic setting of Eswatini: a pragmatic cluster randomised controlled trial. BMJ Glob Health 2021; 6:e005021. [PMID: 34193475 PMCID: PMC8246301 DOI: 10.1136/bmjgh-2021-005021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/30/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION To reduce malaria transmission in very low-endemic settings, screening and treatment near index cases (reactive case detection (RACD)), is widely practised, but the rapid diagnostic tests (RDTs) used miss low-density infections. Reactive focal mass drug administration (rfMDA) may be safe and more effective. METHODS We conducted a pragmatic cluster randomised controlled trial in Eswatini, a very low-endemic setting. 77 clusters were randomised to rfMDA using dihydroartemisin-piperaquine (DP) or RACD involving RDTs and artemether-lumefantrine. Interventions were delivered by the local programme. An intention-to-treat analysis was used to compare cluster-level cumulative confirmed malaria incidence among clusters with cases. Secondary outcomes included safety and adherence. RESULTS From September 2015 to August 2017, 222 index cases from 47 clusters triggered 46 RACD events and 64 rfMDA events. RACD and rfMDA were delivered to 1455 and 1776 individuals, respectively. Index case coverage was 69.5% and 62.4% for RACD and rfMDA, respectively. Adherence to DP was 98.7%. No serious adverse events occurred. For rfMDA versus RACD, cumulative incidences (per 1000 person-years) of all malaria were 2.11 (95% CI 1.73 to 2.59) and 1.97 (95% CI 1.57 to 2.47), respectively; and of locally acquired malaria, they were 1.29 (95% CI 1.00 to 1.67) and 0.97 (95% CI 0.71 to 1.34), respectively. Adjusting for imbalance in baseline incidence, incidence rate ratio for rfMDA versus RACD was 0.95 (95% CI 0.55 to 1.65) for all malaria and 0.82 (95% CI 0.40 to 1.71) for locally acquired malaria. Similar results were obtained in a per-protocol analysis that excluded clusters with <80% index case coverage. CONCLUSION In a very low-endemic, real-world setting, rfMDA using DP was safe, but did not lower incidence compared with RACD, potentially due to insufficient coverage and/or power. To assess impact of interventions in very low-endemic settings, improved coverage, complementary interventions and adaptive ring trial designs may be needed. TRIAL REGISTRATION NUMBER NCT02315690.
Collapse
Affiliation(s)
| | | | - Jade Benjamin-Chung
- Epidemiology & Biostatistics, University of California, Berkeley, California, USA
- Malaria Elimination Initiative, University of California, San Francisco, California, USA
| | - Nomcebo Dlamini
- National Malaria Program, Ministry of Health, Manzini, Eswatini
| | - Mi-Suk Kang Dufour
- Epidemiology & Biostatistics, University of California, Berkeley, California, USA
- Medicine, University of California, San Francisco, California, USA
| | - Brooke Whittemore
- Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Lisa M Prach
- Malaria Elimination Initiative, University of California, San Francisco, California, USA
| | - Kimberly Baltzell
- Family Health Care Nursing, University of California, San Francisco, California, USA
| | | | | | | | - Danica Helb
- Medicine, University of California, San Francisco, California, USA
| | - Bryan Greenhouse
- Medicine, University of California, San Francisco, California, USA
| | - Gugu Maphalala
- National Clinical Laboratory Services, Mbabane, Swaziland
| | | | | | - Getahun Tesfa
- Paediatrics, Raleigh Fitkin Memorial Hospital, Manzini, Swaziland
| | - Roly Gosling
- Malaria Elimination Initiative, University of California, San Francisco, California, USA
| | | | - Simon Kunene
- National Malaria Program, Ministry of Health, Manzini, Eswatini
| | - Michelle S Hsiang
- Malaria Elimination Initiative, University of California, San Francisco, California, USA
- Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Pediatrics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
13
|
Ean M, Sanann N, Callery JJ, Pell C, Peto TJ, Tripura R, Cheah PY. Theory of change: Drama and arts-based community engagement for malaria research and elimination in Cambodia. Wellcome Open Res 2021; 6:46. [PMID: 34041367 PMCID: PMC8127021 DOI: 10.12688/wellcomeopenres.16574.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Across the Greater Mekong Sub-region, malaria persists in isolated communities along international borders. Arts and drama have been used to reach to communities in Cambodia to engage them in malaria research, prevention and control. The “Village Drama Against Malaria” (VDAM) project was conducted in north eastern and western Cambodia: Stung Treng; Battambang and Pailin provinces during 2016 to 2019. In total, VDAM reached 55 rural villages, 2,378 student participants and 43,502 audience members. Methods: This article presents the results of two stakeholder-led evaluation workshops in which participants collaboratively developed theories of change to better understand the potential and actual impact of arts and drama-based activities on malaria in these communities. The workshops had a particular focus on identifying areas for monitoring and evaluation so that impact can be measured. Workshop participants included village malaria workers, community leaders, professional and student drama performers, and representatives from the local health authorities and the national malaria control programme. Results: Five broad areas were identified as relevant for monitoring and evaluation: logistical and practical challenges; embeddedness and reach of engagement; health knowledge and confidence of young people; effectiveness of communications; impact on malaria. These areas align well with the monitoring and evaluation conducted to date and point to additional opportunities for data collection. Conclusions: The findings from these workshops will inform future engagement strategies, for example, we may engage a smaller number of young people but over a longer period and more in-depth.
Collapse
Affiliation(s)
- Mom Ean
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
| | - Nou Sanann
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand.,University Research Company, Phnom Penh, Cambodia
| | - James J Callery
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
| | - Christopher Pell
- 3Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,The Ethox Centre, University of Oxford, Oxford, UK
| |
Collapse
|
14
|
Ean M, Sanann N, Callery JJ, Pell C, Peto TJ, Tripura R, Cheah PY. Theory of change: Drama and arts-based community engagement for malaria research and elimination in Cambodia. Wellcome Open Res 2021; 6:46. [PMID: 34041367 PMCID: PMC8127021 DOI: 10.12688/wellcomeopenres.16574.1] [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] [Accepted: 05/06/2021] [Indexed: 01/24/2024] Open
Abstract
Background: Across the Greater Mekong Sub-region, malaria persists in isolated communities along international borders. Arts and drama have been used to reach to communities in Cambodia to engage them in malaria research, prevention and control. The "Village Drama Against Malaria" (VDAM) project was conducted in north eastern and western Cambodia: Stung Treng; Battambang and Pailin provinces during 2016 to 2019. In total, VDAM reached 55 rural villages, 2,378 student participants and 43,502 audience members. Methods: This article presents the results of two stakeholder-led evaluation workshops in which participants collaboratively developed theories of change to better understand the potential and actual impact of arts and drama-based activities on malaria in these communities. The workshops had a particular focus on identifying areas for monitoring and evaluation so that impact can be measured. Workshop participants included village malaria workers, community leaders, professional and student drama performers, and representatives from the local health authorities and the national malaria control programme. Results: Five broad areas were identified as relevant for monitoring and evaluation: logistical and practical challenges; embeddedness and reach of engagement; health knowledge and confidence of young people; effectiveness of communications; impact on malaria. These areas align well with the monitoring and evaluation conducted to date and point to additional opportunities for data collection. Conclusions: The findings from these workshops will inform future engagement strategies, for example, we may engage a smaller number of young people but over a longer period and more in-depth.
Collapse
Affiliation(s)
- Mom Ean
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
| | - Nou Sanann
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
- University Research Company, Phnom Penh, Cambodia
| | - James J. Callery
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
| | - Christopher Pell
- 3Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Thomas J. Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- The Ethox Centre, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Kyaw SS, Delmas G, Drake TL, Celhay O, Pan-Ngum W, Pukrittayakamee S, Lubell Y, Aguas RJ, Maude RJ, White LJ, Nosten F. Estimating the programmatic cost of targeted mass drug administration for malaria in Myanmar. BMC Public Health 2021; 21:826. [PMID: 33926399 PMCID: PMC8082869 DOI: 10.1186/s12889-021-10842-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 04/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Mass drug administration (MDA) has received growing interest to accelerate the elimination of multi-drug resistant malaria in the Greater Mekong Subregion. Targeted MDA, sometimes referred to as focal MDA, is the practice of delivering MDA to high incidence subpopulations only, rather than the entire population. The potential effectiveness of delivering targeted MDA was demonstrated in a recent intervention in Kayin State, Myanmar. Policymakers and funders need to know what resources are required if MDA, targeted or otherwise, is to be included in elimination packages beyond existing malaria interventions. This study aims to estimate the programmatic cost and the unit cost of targeted MDA in Kayin State, Myanmar. Methods We used financial data from a malaria elimination initiative, conducted in Kayin State, to estimate the programmatic costs of the targeted MDA component using a micro-costing approach. Three activities (community engagement, identification of villages for targeted MDA, and conducting mass treatment in target villages) were evaluated. We then estimated the programmatic costs of implementing targeted MDA to support P. falciparum malaria elimination in Kayin State. A costing tool was developed to aid future analyses. Results The cost of delivering targeted MDA within an integrated malaria elimination initiative in eastern Kayin State was approximately US$ 910,000. The cost per person reached, distributed among those in targeted and non-targeted villages, for the MDA component was US$ 2.5. Conclusion This cost analysis can assist policymakers in determining the resources required to clear malaria parasite reservoirs. The analysis demonstrated the value of using financial data from research activities to predict programmatic implementation costs of targeting MDA to different numbers of target villages. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-021-10842-5.
Collapse
Affiliation(s)
- Shwe Sin Kyaw
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Gilles Delmas
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Mae Sot, Thailand
| | - Tom L Drake
- Department for International Development, London, UK
| | - Olivier Celhay
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wirichada Pan-Ngum
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sasithon Pukrittayakamee
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yoel Lubell
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ricardo J Aguas
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Richard James Maude
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,Harvard TH Chan School of Public Health, Harvard University, Boston, USA
| | - Lisa J White
- Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, Oxford, UK
| | - Francois Nosten
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Mae Sot, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Choosing interventions to eliminate forest malaria: preliminary results of two operational research studies inside Cambodian forests. Malar J 2021; 20:51. [PMID: 33472630 PMCID: PMC7818569 DOI: 10.1186/s12936-020-03572-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/24/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Rapid elimination of Plasmodium falciparum malaria in Cambodia is a goal with both national and international significance. Transmission of malaria in Cambodia is limited to forest environments, and the main population at risk consists of forest-goers who rely on forest products for income or sustenance. The ideal interventions to eliminate malaria from this population are unknown. METHODS In two forested regions of Cambodia, forest-goers were trained to become forest malaria workers (FMWs). In one region, FMWs performed mass screening and treatment, focal screening and treatment, and passive case detection inside the forest. In the other region, FMWs played an observational role for the first year, to inform the choice of intervention for the second year. In both forests, FMWs collected blood samples and questionnaire data from all forest-goers they encountered. Mosquito collections were performed in each forest. RESULTS Malaria prevalence by PCR was high in the forest, with 2.3-5.0% positive for P. falciparum and 14.6-25.0% positive for Plasmodium vivax among forest-goers in each study site. In vectors, malaria prevalence ranged from 2.1% to 9.6%, but no P. falciparum was observed. Results showed poor performance of mass screening and treatment, with sensitivity of rapid diagnostic tests equal to 9.1% (95% CI 1.1%, 29.2%) for P. falciparum and 4.4% (95% CI 1.6%, 9.2%) for P. vivax. Malaria infections were observed in all demographics and throughout the studied forests, with no clear risk factors emerging. CONCLUSIONS Malaria prevalence remains high among Cambodian forest-goers, but performance of rapid diagnostic tests is poor. More adapted strategies to this population, such as intermittent preventive treatment of forest goers, should be considered.
Collapse
|
17
|
Nice J, Nahusenay H, Eckert E, Eisele TP, Ashton RA. Estimating malaria chemoprevention and vector control coverage using program and campaign data: A scoping review of current practices and opportunities. J Glob Health 2020; 10:020413. [PMID: 33110575 PMCID: PMC7568932 DOI: 10.7189/jogh.10.020413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Accurate estimation of intervention coverage is a vital component of malaria program monitoring and evaluation, both for process evaluation (how well program targets are achieved), and impact evaluation (whether intervention coverage had an impact on malaria burden). There is growing interest in maximizing the utility of program data to generate interim estimates of intervention coverage in the periods between large-scale cross-sectional surveys (the gold standard). As such, this study aimed to identify relevant concepts and themes that may guide future optimization of intervention coverage estimation using routinely collected data, or data collected during and following intervention campaigns, with a particular focus on strategies to define the denominator. METHODS We conducted a scoping review of current practices to estimate malaria intervention coverage for insecticide-treated nets (ITNs); indoor residual spray (IRS); intermittent preventive treatment in pregnancy (IPTp); mass drug administration (MDA); and seasonal malaria chemoprevention (SMC) interventions; case management was excluded. Multiple databases were searched for relevant articles published from January 1, 2015 to June 1, 2018. Additionally, we identified and included other guidance relevant to estimating population denominators, with a focus on innovative techniques. RESULTS While program data have the potential to provide intervention coverage data, there are still substantial challenges in selecting appropriate denominators. The review identified a lack of consistency in how coverage was defined and reported for each intervention type, with denominator estimation methods not clearly or consistently reported, and denominator estimates rarely triangulated with other data sources to present the feasible range of denominator values and consequently the range of likely coverage estimates. CONCLUSIONS Though household survey-based estimates of intervention coverage remain the gold standard, efforts should be made to further standardize practices for generating interim measurements of intervention coverage from program data, and for estimating and reporting population denominators. This includes fully describing any projections or adjustments made to existing census or population data, exploring opportunities to validate available data by comparing with other sources, and explaining how the denominator has been restricted (or not) to reflect exclusion criteria.
Collapse
Affiliation(s)
- Johanna Nice
- MEASURE Evaluation, Centre for Applied Malaria Research and Evaluation, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Honelgn Nahusenay
- MEASURE Evaluation, Centre for Applied Malaria Research and Evaluation, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Erin Eckert
- U.S. President's Malaria Initiative, United States Agency for International Development, Washington, D.C., USA
- RTI International, Washington, D.C., USA
| | - Thomas P Eisele
- Centre for Applied Malaria Research and Evaluation, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Ruth A Ashton
- MEASURE Evaluation, Centre for Applied Malaria Research and Evaluation, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
18
|
Imwong M, Dhorda M, Myo Tun K, Thu AM, Phyo AP, Proux S, Suwannasin K, Kunasol C, Srisutham S, Duanguppama J, Vongpromek R, Promnarate C, Saejeng A, Khantikul N, Sugaram R, Thanapongpichat S, Sawangjaroen N, Sutawong K, Han KT, Htut Y, Linn K, Win AA, Hlaing TM, van der Pluijm RW, Mayxay M, Pongvongsa T, Phommasone K, Tripura R, Peto TJ, von Seidlein L, Nguon C, Lek D, Chan XHS, Rekol H, Leang R, Huch C, Kwiatkowski DP, Miotto O, Ashley EA, Kyaw MP, Pukrittayakamee S, Day NPJ, Dondorp AM, Smithuis FM, Nosten FH, White NJ. Molecular epidemiology of resistance to antimalarial drugs in the Greater Mekong subregion: an observational study. THE LANCET. INFECTIOUS DISEASES 2020; 20:1470-1480. [PMID: 32679084 PMCID: PMC7689289 DOI: 10.1016/s1473-3099(20)30228-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/19/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The Greater Mekong subregion is a recurrent source of antimalarial drug resistance in Plasmodium falciparum malaria. This study aimed to characterise the extent and spread of resistance across this entire region between 2007 and 2018. METHODS P falciparum isolates from Myanmar, Thailand, Laos, and Cambodia were obtained from clinical trials and epidemiological studies done between Jan 1, 2007, and Dec 31, 2018, and were genotyped for molecular markers (pfkelch, pfcrt, pfplasmepsin2, and pfmdr1) of antimalarial drug resistance. Genetic relatedness was assessed using microsatellite and single nucleotide polymorphism typing of flanking sequences around target genes. FINDINGS 10 632 isolates were genotyped. A single long pfkelch Cys580Tyr haplotype (from -50 kb to +31·5 kb) conferring artemisinin resistance (PfPailin) now dominates across the eastern Greater Mekong subregion. Piperaquine resistance associated with pfplasmepsin2 gene amplification and mutations in pfcrt downstream of the Lys76Thr chloroquine resistance locus has also developed. On the Thailand-Myanmar border a different pfkelch Cys580Tyr lineage rose to high frequencies before it was eliminated. Elsewhere in Myanmar the Cys580Tyr allele remains widespread at low allele frequencies. Meanwhile a single artemisinin-resistant pfkelch Phe446Ile haplotype has spread across Myanmar. Despite intense use of dihydroartemisinin-piperaquine in Kayin state, eastern Myanmar, both in treatment and mass drug administrations, no selection of piperaquine resistance markers was observed. pfmdr1 amplification, a marker of resistance to mefloquine, remains at low prevalence across the entire region. INTERPRETATION Artemisinin resistance in P falciparum is now prevalent across the Greater Mekong subregion. In the eastern Greater Mekong subregion a multidrug resistant P falciparum lineage (PfPailin) dominates. In Myanmar a long pfkelch Phe446Ile haplotype has spread widely but, by contrast with the eastern Greater Mekong subregion, there is no indication of artemisinin combination therapy (ACT) partner drug resistance from genotyping known markers, and no evidence of spread of ACT resistant P falciparum from the east to the west. There is still a window of opportunity to prevent global spread of ACT resistance. FUNDING Thailand Science Research and Innovation, Initiative 5%, Expertise France, Wellcome Trust.
Collapse
Affiliation(s)
- Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Mehul Dhorda
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Worldwide Antimalarial Resistance Network, Bangkok, Thailand
| | - Kyaw Myo Tun
- Department of Preventive and Social Medicine, Defence Services Medical Academy, Yangon, Myanmar
| | - Aung Myint Thu
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Aung Pyae Phyo
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Myanmar Oxford Clinical Research Unit, Yangon, Myanmar
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Kanokon Suwannasin
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Chanon Kunasol
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Suttipat Srisutham
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jureeporn Duanguppama
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | | | - Aungkana Saejeng
- Bureau of Vector-borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Rungniran Sugaram
- Bureau of Vector-borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Nongyao Sawangjaroen
- Department of Microbiology, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Kreepol Sutawong
- Buntharik Hospital, Amphoe Buntharik, Ubon Ratchathani, Thailand
| | - Kay Thwe Han
- Department of Medical Research, Ministry of Health and Sports, Yangon, Myanmar
| | - Ye Htut
- Department of Medical Research, Ministry of Health and Sports, Yangon, Myanmar
| | - Khin Linn
- Department of Medical Research, Ministry of Health and Sports, Yangon, Myanmar
| | - Aye Aye Win
- Department of Tropical and Infectious Diseases, University of Medicine 1, Yangon, Myanmar
| | - Tin M Hlaing
- Defence Services Medical Research Centre, Naypyitaw, Myanmar
| | - Rob W van der Pluijm
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mayfong Mayxay
- Institute of Research and Education Development, University of Health Sciences, Ministry of Health, Vientiane, Laos; Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Vientiane, Laos
| | - Tiengkham Pongvongsa
- Savannakhet Provincial Health Department, Phonsavangnuea village, Kaysone-Phomvihan district, Savannakhet, Laos
| | - Koukeo Phommasone
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Vientiane, Laos
| | - Rupam Tripura
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thomas J Peto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lorenz von Seidlein
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chea Nguon
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Dysoley Lek
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Xin Hui S Chan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Huy Rekol
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Cheah Huch
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Dominic P Kwiatkowski
- Wellcome Sanger Institute, Hinxton, UK; Medical Research Council Centre for Genomics and Global Health, Big Data Institute, University of Oxford, Oxford, UK
| | - Olivo Miotto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Wellcome Sanger Institute, Hinxton, UK; Medical Research Council Centre for Genomics and Global Health, Big Data Institute, University of Oxford, Oxford, UK
| | - Elizabeth A Ashley
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Vientiane, Laos
| | - Myat Phone Kyaw
- Department of Medical Research, Myanmar Health Network Organization, Yangon, Myanmar
| | - Sasithon Pukrittayakamee
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; The Royal Society of Thailand, Dusit, Bangkok, Thailand
| | - Nicholas P J Day
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Frank M Smithuis
- Myanmar Oxford Clinical Research Unit, Yangon, Myanmar; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Medical Action Myanmar, Yangon, Myanmar
| | - Francois H Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Gupta H, Galatas B, Chidimatembue A, Huijben S, Cisteró P, Matambisso G, Nhamussua L, Simone W, Bassat Q, Ménard D, Ringwald P, Rabinovich NR, Alonso PL, Saúte F, Aide P, Mayor A. Effect of mass dihydroartemisinin-piperaquine administration in southern Mozambique on the carriage of molecular markers of antimalarial resistance. PLoS One 2020; 15:e0240174. [PMID: 33075062 PMCID: PMC7571678 DOI: 10.1371/journal.pone.0240174] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mass drug administration (MDA) can rapidly reduce the burden of Plasmodium falciparum (Pf). However, concerns remain about its contribution to select for antimalarial drug resistance. METHODS We used Sanger sequencing and real-time PCR to determine the proportion of molecular markers associated with antimalarial resistance (k13, pfpm2, pfmdr1 and pfcrt) in Pf isolates collected before (n = 99) and after (n = 112) the implementation of two monthly MDA rounds with dihydroartemisinin-piperaquine (DHAp) for two consecutive years in Magude district of Southern Mozambique. RESULTS None of the k13 polymorphisms associated with artemisinin resistance were observed in the Pf isolates analyzed. The proportion of Pf isolates with multiple copies of pfpm2, an amplification associated with piperaquine resistance, was similar in pre- (4.9%) and post-MDA groups (3.4%; p = 1.000). No statistically significant differences were observed between pre- and post-MDA groups in the proportion of Pf isolates neither with mutations in pfcrt and pfmdr1 genes, nor with the carriage of pfmdr1 multiple copies (p>0.05). CONCLUSIONS This study does not show any evidence of increased frequency of molecular makers of antimalarial resistance after MDA with DHAp in southern Mozambique where markers of antimalarial resistance were absent or low at the beginning of the intervention.
Collapse
Affiliation(s)
- Himanshu Gupta
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
| | - Beatriz Galatas
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | | | - Silvie Huijben
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
| | - Pau Cisteró
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
| | | | - Lidia Nhamussua
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | - Wilson Simone
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | - Quique Bassat
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
- ICREA, Pg. Lluís Companys, Barcelona, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Pediatric Infectious Diseases Unit, Pediatrics Department, Hospital Sant Joan de Déu (University of Barcelona), Barcelona, Spain
| | - Didier Ménard
- Institut Pasteur, Paris, France
- INSERM U1201, Paris, France
- CNRS ERL9195, Paris, France
| | - Pascal Ringwald
- World Health Organization (WHO), Global Malaria Programme, Geneva, Switzerland
| | - N. Regina Rabinovich
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Pedro L. Alonso
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | - Francisco Saúte
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
- National Institute of Health, Ministry of Health, Manhica, Mozambique
| | - Alfredo Mayor
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
- ICREA, Pg. Lluís Companys, Barcelona, Spain
| |
Collapse
|
20
|
Fraser M, Miller JM, Silumbe K, Hainsworth M, Mudenda M, Hamainza B, Moonga H, Chizema Kawesha E, Mercer LD, Bennett A, Schneider K, Slater HC, Eisele TP, Guinovart C. Evaluating the Impact of Programmatic Mass Drug Administration for Malaria in Zambia Using Routine Incidence Data. J Infect Dis 2020; 225:1415-1423. [PMID: 32691047 PMCID: PMC9016426 DOI: 10.1093/infdis/jiaa434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/17/2020] [Indexed: 01/24/2023] Open
Abstract
Background In 2016, the Zambian National Malaria Elimination Centre started programmatic mass drug administration (pMDA) campaigns with dihydroartemisinin-piperaquine as a malaria elimination tool in Southern Province. Two rounds were administered, 2 months apart (coverage 70% and 57%, respectively). We evaluated the impact of 1 year of pMDA on malaria incidence using routine data. Methods We conducted an interrupted time series with comparison group analysis on monthly incidence data collected at the health facility catchment area (HFCA) level, with a negative binomial model using generalized estimating equations. Programmatic mass drug administration was conducted in HFCAs with greater than 50 cases/1000 people per year. Ten HFCAs with incidence rates marginally above this threshold (pMDA group) were compared with 20 HFCAs marginally below (comparison group). Results The pMDA HFCAs saw a 46% greater decrease in incidence at the time of intervention than the comparison areas (incidence rate ratio = 0.536; confidence interval = 0.337–0.852); however, incidence increased toward the end of the season. No HFCAs saw a transmission interruption. Conclusions Programmatic mass drug administration, implemented during 1 year with imperfect coverage in low transmission areas with suboptimal vector control coverage, significantly reduced incidence. However, elimination will require additional tools. Routine data are important resources for programmatic impact evaluations and should be considered for future analyses.
Collapse
Affiliation(s)
- Maya Fraser
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
| | | | | | - Michael Hainsworth
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
| | - Mutinta Mudenda
- National Malaria Elimination Centre, Zambia Ministry of Health, Lusaka, Zambia
| | - Busiku Hamainza
- National Malaria Elimination Centre, Zambia Ministry of Health, Lusaka, Zambia
| | - Hawela Moonga
- National Malaria Elimination Centre, Zambia Ministry of Health, Lusaka, Zambia
| | | | - Laina D Mercer
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
| | - Adam Bennett
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
- University of California San Francisco, San Francisco, California, USA
| | - Kammerle Schneider
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
| | - Hannah C Slater
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
| | - Thomas P Eisele
- Center for Applied Malaria Research and Evaluation, Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Caterina Guinovart
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Seattle, Washington, USA
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Wellems TE, Sá JM, Su XZ, Connelly SV, Ellis AC. 'Artemisinin Resistance': Something New or Old? Something of a Misnomer? Trends Parasitol 2020; 36:735-744. [PMID: 32586776 DOI: 10.1016/j.pt.2020.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 01/02/2023]
Abstract
Artemisinin and its derivatives (ART) are crucial first-line antimalarial drugs that rapidly clear parasitemia, but recrudescences of the infection frequently follow ART monotherapy. For this reason, ART must be used in combination with one or more partner drugs that ensure complete cure. The ability of malaria parasites to survive ART monotherapy may relate to an innate growth bistability phenomenon whereby a fraction of the drug-exposed population enters into metabolic quiescence (dormancy) as persister forms. Characterization of the events that underlie entry and waking from persistence may lead to lasting breakthroughs in malaria chemotherapy that can prevent recrudescences and protect the future of ART-based combination therapies.
Collapse
Affiliation(s)
- Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sean V Connelly
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela C Ellis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Millar J, Toh KB, Valle D. To screen or not to screen: an interactive framework for comparing costs of mass malaria treatment interventions. BMC Med 2020; 18:149. [PMID: 32552743 PMCID: PMC7304173 DOI: 10.1186/s12916-020-01609-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 04/28/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Mass drug administration and mass-screen-and-treat interventions have been used to interrupt malaria transmission and reduce burden in sub-Saharan Africa. Determining which strategy will reduce costs is an important challenge for implementers; however, model-based simulations and field studies have yet to develop consensus guidelines. Moreover, there is often no way for decision-makers to directly interact with these data and/or models, incorporate local knowledge and expertise, and re-fit parameters to guide their specific goals. METHODS We propose a general framework for comparing costs associated with mass drug administrations and mass screen and treat based on the possible outcomes of each intervention and the costs associated with each outcome. We then used publicly available data from six countries in western Africa to develop spatial-explicit probabilistic models to estimate intervention costs based on baseline malaria prevalence, diagnostic performance, and sociodemographic factors (age and urbanicity). In addition to comparing specific scenarios, we also develop interactive web applications which allow managers to select data sources and model parameters, and directly input their own cost values. RESULTS The regional-level models revealed substantial spatial heterogeneity in malaria prevalence and diagnostic test sensitivity and specificity, indicating that a "one-size-fits-all" approach is unlikely to maximize resource allocation. For instance, urban communities in Burkina Faso typically had lower prevalence rates compared to rural communities (0.151 versus 0.383, respectively) as well as lower diagnostic sensitivity (0.699 versus 0.862, respectively); however, there was still substantial regional variation. Adjusting the cost associated with false negative diagnostic results to included additional costs, such as delayed treated and potential lost wages, undermined the overall costs associated with MSAT. CONCLUSIONS The observed spatial variability and dependence on specified cost values support not only the need for location-specific intervention approaches but also the need to move beyond standard modeling approaches and towards interactive tools which allow implementers to engage directly with data and models. We believe that the framework demonstrated in this article will help connect modeling efforts and stakeholders in order to promote data-driven decision-making for the effective management of malaria, as well as other diseases.
Collapse
Affiliation(s)
- Justin Millar
- School of Forest Resources and Conservation, University of Florida, Gainesville, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, USA.
| | - Kok Ben Toh
- Emerging Pathogens Institute, University of Florida, Gainesville, USA
- School of Natural Resources and Environment, University of Florida, Gainesville, USA
| | - Denis Valle
- School of Forest Resources and Conservation, University of Florida, Gainesville, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, USA
| |
Collapse
|
23
|
Bannister-Tyrrell M, Krit M, Sluydts V, Tho S, Sokny M, Mean V, Kim S, Menard D, Grietens KP, Abrams S, Hens N, Coosemans M, Bassat Q, van Hensbroek MB, Durnez L, Van Bortel W. Households or Hotspots? Defining Intervention Targets for Malaria Elimination in Ratanakiri Province, Eastern Cambodia. J Infect Dis 2020; 220:1034-1043. [PMID: 31028393 PMCID: PMC6688056 DOI: 10.1093/infdis/jiz211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/25/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Malaria "hotspots" have been proposed as potential intervention units for targeted malaria elimination. Little is known about hotspot formation and stability in settings outside sub-Saharan Africa. METHODS Clustering of Plasmodium infections at the household and hotspot level was assessed over 2 years in 3 villages in eastern Cambodia. Social and spatial autocorrelation statistics were calculated to assess clustering of malaria risk, and logistic regression was used to assess the effect of living in a malaria hotspot compared to living in a malaria-positive household in the first year of the study on risk of malaria infection in the second year. RESULTS The crude prevalence of Plasmodium infection was 8.4% in 2016 and 3.6% in 2017. Living in a hotspot in 2016 did not predict Plasmodium risk at the individual or household level in 2017 overall, but living in a Plasmodium-positive household in 2016 strongly predicted living in a Plasmodium-positive household in 2017 (Risk Ratio, 5.00 [95% confidence interval, 2.09-11.96], P < .0001). There was no consistent evidence that malaria risk clustered in groups of socially connected individuals from different households. CONCLUSIONS Malaria risk clustered more clearly in households than in hotspots over 2 years. Household-based strategies should be prioritized in malaria elimination programs in this region.
Collapse
Affiliation(s)
| | | | - Vincent Sluydts
- Institute of Tropical Medicine, Antwerp.,University of Antwerp, Belgium
| | - Sochantha Tho
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh
| | - Mao Sokny
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh
| | - Vanna Mean
- Ratanakiri Provincial Health Department, Banlung
| | | | | | | | - Steven Abrams
- University of Antwerp, Belgium.,University of Hasselt, Belgium
| | - Niel Hens
- University of Antwerp, Belgium.,University of Hasselt, Belgium
| | | | - Quique Bassat
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique.,Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | | | - Lies Durnez
- Institute of Tropical Medicine, Antwerp.,University of Antwerp, Belgium
| | | |
Collapse
|
24
|
Mathema VB, Nakeesathit S, White NJ, Dondorp AM, Imwong M. Genome-wide microsatellite characteristics of five human Plasmodium species, focusing on Plasmodium malariae and P. ovale curtisi. Parasite 2020; 27:34. [PMID: 32410726 PMCID: PMC7227371 DOI: 10.1051/parasite/2020034] [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] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
Microsatellites can be utilized to explore genotypes, population structure, and other genomic features of eukaryotes. Systematic characterization of microsatellites has not been a focus for several species of Plasmodium, including P. malariae and P. ovale, as the majority of malaria elimination programs are focused on P. falciparum and to a lesser extent P. vivax. Here, five human malaria species (P. falciparum, P. vivax, P. malariae, P. ovale curtisi, and P. knowlesi) were investigated with the aim of conducting in-depth categorization of microsatellites for P. malariae and P. ovale curtisi. Investigation of reference genomes for microsatellites with unit motifs of 1-10 base pairs indicates high diversity among the five Plasmodium species. Plasmodium malariae, with the largest genome size, displays the second highest microsatellite density (1421 No./Mbp; 5% coverage) next to P. falciparum (3634 No./Mbp; 12% coverage). The lowest microsatellite density was observed in P. vivax (773 No./Mbp; 2% coverage). A, AT, and AAT are the most commonly repeated motifs in the Plasmodium species. For P. malariae and P. ovale curtisi, microsatellite-related sequences are observed in approximately 18-29% of coding sequences (CDS). Lysine, asparagine, and glutamic acids are most frequently coded by microsatellite-related CDS. The majority of these CDS could be related to the gene ontology terms "cell parts," "binding," "developmental processes," and "metabolic processes." The present study provides a comprehensive overview of microsatellite distribution and can assist in the planning and development of potentially useful genetic tools for further investigation of P. malariae and P. ovale curtisi epidemiology.
Collapse
Affiliation(s)
- Vivek Bhakta Mathema
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University 10400 Bangkok Thailand
| | - Supatchara Nakeesathit
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University 10400 Bangkok Thailand
| | - Nicholas J. White
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University 10400 Bangkok Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford OX1 2JD Oxford United Kingdom
| | - Arjen M. Dondorp
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University 10400 Bangkok Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford OX1 2JD Oxford United Kingdom
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University 10400 Bangkok Thailand
| |
Collapse
|
25
|
Peto TJ, Tripura R, Maude RJ. Strengthen Village Malaria Reporting to Better Target Reservoirs of Persistent Infections in Southeast Asia. Clin Infect Dis 2020; 68:1066-1067. [PMID: 30219841 PMCID: PMC6399432 DOI: 10.1093/cid/ciy793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Richard J Maude
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| |
Collapse
|
26
|
Hsiang MS, Ntuku H, Roberts KW, Dufour MSK, Whittemore B, Tambo M, McCreesh P, Medzihradsky OF, Prach LM, Siloka G, Siame N, Gueye CS, Schrubbe L, Wu L, Scott V, Tessema S, Greenhouse B, Erlank E, Koekemoer LL, Sturrock HJW, Mwilima A, Katokele S, Uusiku P, Bennett A, Smith JL, Kleinschmidt I, Mumbengegwi D, Gosling R. Effectiveness of reactive focal mass drug administration and reactive focal vector control to reduce malaria transmission in the low malaria-endemic setting of Namibia: a cluster-randomised controlled, open-label, two-by-two factorial design trial. Lancet 2020; 395:1361-1373. [PMID: 32334702 PMCID: PMC7184675 DOI: 10.1016/s0140-6736(20)30470-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/23/2020] [Accepted: 02/25/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND In low malaria-endemic settings, screening and treatment of individuals in close proximity to index cases, also known as reactive case detection (RACD), is practised for surveillance and response. However, other approaches could be more effective for reducing transmission. We aimed to evaluate the effectiveness of reactive focal mass drug administration (rfMDA) and reactive focal vector control (RAVC) in the low malaria-endemic setting of Zambezi (Namibia). METHODS We did a cluster-randomised controlled, open-label trial using a two-by-two factorial design of 56 enumeration area clusters in the low malaria-endemic setting of Zambezi (Namibia). We randomly assigned these clusters using restricted randomisation to four groups: RACD only, rfMDA only, RAVC plus RACD, or rfMDA plus RAVC. RACD involved rapid diagnostic testing and treatment with artemether-lumefantrine and single-dose primaquine, rfMDA involved presumptive treatment with artemether-lumefantrine, and RAVC involved indoor residual spraying with pirimiphos-methyl. Interventions were administered within 500 m of index cases. To evaluate the effectiveness of interventions targeting the parasite reservoir in humans (rfMDA vs RACD), in mosquitoes (RAVC vs no RAVC), and in both humans and mosquitoes (rfMDA plus RAVC vs RACD only), an intention-to-treat analysis was done. For each of the three comparisons, the primary outcome was the cumulative incidence of locally acquired malaria cases. This trial is registered with ClinicalTrials.gov, number NCT02610400. FINDINGS Between Jan 1, 2017, and Dec 31, 2017, 55 enumeration area clusters had 1118 eligible index cases that led to 342 interventions covering 8948 individuals. The cumulative incidence of locally acquired malaria was 30·8 per 1000 person-years (95% CI 12·8-48·7) in the clusters that received rfMDA versus 38·3 per 1000 person-years (23·0-53·6) in the clusters that received RACD; 30·2 per 1000 person-years (15·0-45·5) in the clusters that received RAVC versus 38·9 per 1000 person-years (20·7-57·1) in the clusters that did not receive RAVC; and 25·0 per 1000 person-years (5·2-44·7) in the clusters that received rfMDA plus RAVC versus 41·4 per 1000 person-years (21·5-61·2) in the clusters that received RACD only. After adjusting for imbalances in baseline and implementation factors, the incidence of malaria was lower in clusters receiving rfMDA than in those receiving RACD (adjusted incidence rate ratio 0·52 [95% CI 0·16-0·88], p=0·009), lower in clusters receiving RAVC than in those that did not (0·48 [0·16-0·80], p=0·002), and lower in clusters that received rfMDA plus RAVC than in those receiving RACD only (0·26 [0·10-0·68], p=0·006). No serious adverse events were reported. INTERPRETATION In a low malaria-endemic setting, rfMDA and RAVC, implemented alone and in combination, reduced malaria transmission and should be considered as alternatives to RACD for elimination of malaria. FUNDING Novartis Foundation, Bill & Melinda Gates Foundation, and Horchow Family Fund.
Collapse
Affiliation(s)
- Michelle S Hsiang
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| | - Henry Ntuku
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Kathryn W Roberts
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Mi-Suk Kang Dufour
- Division of Prevention Science, University of California San Francisco, San Francisco, CA, USA
| | - Brooke Whittemore
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Munyaradzi Tambo
- Multidisciplinary Research Centre, University of Namibia, Windhoek, Namibia
| | - Patrick McCreesh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Oliver F Medzihradsky
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Lisa M Prach
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Griffith Siloka
- Zambezi Ministry of Health and Social Services, Katima, Namibia
| | - Noel Siame
- Zambezi Ministry of Health and Social Services, Katima, Namibia
| | - Cara Smith Gueye
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Leah Schrubbe
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Lindsey Wu
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Valerie Scott
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Sofonias Tessema
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Bryan Greenhouse
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Erica Erlank
- Wits Research Institute for Malaria, South African Medical Research Council Collaborating Centre for Multi-Disciplinary Research on Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lizette L Koekemoer
- Wits Research Institute for Malaria, South African Medical Research Council Collaborating Centre for Multi-Disciplinary Research on Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Hugh J W Sturrock
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Agnes Mwilima
- Zambezi Ministry of Health and Social Services, Katima, Namibia
| | - Stark Katokele
- National Vector-Borne Diseases Control Programme, Namibia Ministry of Health and Social Services, Windhoek, Namibia
| | - Petrina Uusiku
- National Vector-Borne Diseases Control Programme, Namibia Ministry of Health and Social Services, Windhoek, Namibia
| | - Adam Bennett
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer L Smith
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA
| | - Immo Kleinschmidt
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Wits Research Institute for Malaria, South African Medical Research Council Collaborating Centre for Multi-Disciplinary Research on Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Southern African Development Community, Malaria Elimination Eight Secretariat, Windhoek, Namibia
| | - Davis Mumbengegwi
- Multidisciplinary Research Centre, University of Namibia, Windhoek, Namibia
| | - Roly Gosling
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, CA, USA; Multidisciplinary Research Centre, University of Namibia, Windhoek, Namibia
| |
Collapse
|
27
|
Lek D, Callery JJ, Nguon C, Debackere M, Sovannaroth S, Tripura R, Wojnarski M, Piola P, Khean ST, Manion K, Nguon S, Kunkel A, Vernaeve L, Peto TJ, Dantzer E, Davoeung C, Etienne W, Dondorp AM, Tuseo L, von Seidlein L, Guintran JO. Tools to accelerate falciparum malaria elimination in Cambodia: a meeting report. Malar J 2020; 19:151. [PMID: 32293452 PMCID: PMC7161105 DOI: 10.1186/s12936-020-03197-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
Cambodia targets malaria elimination by 2025. Rapid elimination will depend on successfully identifying and clearing malaria foci linked to forests. Expanding and maintaining universal access to early diagnosis and effective treatment remains the key to malaria control and ultimately malaria elimination in the Greater Mekong Subregion (GMS) in the foreseeable future. Mass Drug Administration (MDA) holds some promise in the rapid reduction of Plasmodium falciparum infections, but requires considerable investment of resources and time to mobilize the target communities. Furthermore, the most practical drug regimen for MDA in the GMS—three rounds of DHA/piperaquine—has lost some of its efficacy. Mass screening and treatment benefits asymptomatic P. falciparum carriers by clearing chronic infections, but in its current form holds little promise for malaria elimination. Hopes that “highly sensitive” diagnostic tests would provide substantial advances in screen and treat programmes have been shown to be misplaced. To reduce the burden on P. falciparum and Plasmodium vivax infections in people working in forested areas novel approaches to the use of malaria prophylaxis in forest workers should be explored. During an October 2019 workshop in Phnom Penh researchers and policymakers reviewed evidence of acceptability, feasibility and effectiveness of interventions to target malaria foci and interrupt P. falciparum transmission and discussed operational requirements and conditions for programmatic implementation.
Collapse
Affiliation(s)
- Dysoley Lek
- Centre for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - James J Callery
- Mahidol-Oxford University Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Chea Nguon
- Centre for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | | | - Siv Sovannaroth
- Centre for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Rupam Tripura
- Mahidol-Oxford University Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Marius Wojnarski
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | | | - Soy Ty Khean
- University Research Company Ltd., Phnom Penh, Cambodia.,London School of Hygiene and Tropical Medicine, London, UK
| | | | - Sokomar Nguon
- University Research Company Ltd., Phnom Penh, Cambodia
| | - Amber Kunkel
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia.,Emerging Diseases Epidemiology Unit, Institut Pasteur, Paris, France
| | | | - Thomas J Peto
- Mahidol-Oxford University Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Emily Dantzer
- University of California at San Francisco, San Francisco, USA
| | | | | | - Arjen M Dondorp
- Mahidol-Oxford University Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | | | - Lorenz von Seidlein
- Mahidol-Oxford University Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand. .,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | - Jean-Olivier Guintran
- Centre for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia. .,Mahidol-Oxford University Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
28
|
Phommasone K, van Leth F, Peto TJ, Landier J, Nguyen TN, Tripura R, Pongvongsa T, Lwin KM, Kajeechiwa L, Thwin MM, Parker DM, Wiladphaingern J, Nosten S, Proux S, Nguon C, Davoeung C, Rekol H, Adhikari B, Promnarate C, Chotivanich K, Hanboonkunupakarn B, Jittmala P, Cheah PY, Dhorda M, Imwong M, Mukaka M, Peerawaranun P, Pukrittayakamee S, Newton PN, Thwaites GE, Day NPJ, Mayxay M, Hien TT, Nosten FH, Cobelens F, Dondorp AM, White NJ, von Seidlein L. Mass drug administrations with dihydroartemisinin-piperaquine and single low dose primaquine to eliminate Plasmodium falciparum have only a transient impact on Plasmodium vivax: Findings from randomised controlled trials. PLoS One 2020; 15:e0228190. [PMID: 32023293 PMCID: PMC7001954 DOI: 10.1371/journal.pone.0228190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/08/2020] [Indexed: 11/30/2022] Open
Abstract
Background Mass administrations of antimalarial drugs (MDA) have reduced the incidence and prevalence of P. falciparum infections in a trial in the Greater Mekong Subregion. Here we assess the impact of the MDA on P. vivax infections. Methods Between May 2013 and July 2017, four villages in each Myanmar, Vietnam, Cambodia and Lao PDR were selected based on high prevalence of P. falciparum infections. Eight of the 16 villages were randomly assigned to receive MDA consisting of three-monthly rounds of three-day courses of dihydroartemisinin-piperaquine and, except in Cambodia, a single low-dose of primaquine. Cross-sectional surveys were conducted at quarterly intervals to detect Plasmodium infections using ultrasensitive qPCR. The difference in the cumulative incidence between the groups was assessed through a discrete time survival approach, the difference in prevalence through a difference-in-difference analysis, and the difference in the number of participants with a recurrence of P. vivax infection through a mixed-effect logistic regression. Results 3,790 (86%) residents in the intervention villages participated in at least one MDA round, of whom 2,520 (57%) participated in three rounds. The prevalence of P. vivax infections fell from 9.31% to 0.89% at month 3 but rebounded by six months to 5.81%. There was no evidence that the intervention reduced the cumulative incidence of P.vivax infections (95% confidence interval [CI] Odds ratio (OR): 0.29 to 1.36). Similarly, there was no evidence of MDA related reduction in the number of participants with at least one recurrent infection (OR: 0.34; 95% CI: 0.08 to 1.42). Conclusion MDA with schizontocidal drugs had a lasting effect on P. falciparum infections but only a transient effect on the prevalence of P. vivax infections. Radical cure with an 8-aminoquinoline will be needed for the rapid elimination of vivax malaria.
Collapse
Affiliation(s)
- Koukeo Phommasone
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Department of Global Health, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
- Amsterdam Institute for Global Health & Development, Amsterdam, Netherlands
| | - Frank van Leth
- Department of Global Health, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
- Amsterdam Institute for Global Health & Development, Amsterdam, Netherlands
| | - Thomas J. Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Jordi Landier
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Institut de Recherche pour le Développement (IRD), Institut national de la santé et de la recherche médical (INSERM), Aix-Marseille Université · SESSTIM, Marseille, France
| | - Thuy-Nhien Nguyen
- Oxford University Clinical Research Unit, Wellcome Trust Major Oversea Programme, Ho Chi Minh City, Vietnam
| | - Rupam Tripura
- Department of Global Health, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Tiengkham Pongvongsa
- Savannakhet Provincial Health Department, Savannakhet Province, Lao People’s Demographic Republic
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Khin Maung Lwin
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ladda Kajeechiwa
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - May Myo Thwin
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Daniel M. Parker
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Department of Population Health and Disease Prevention, University of California, Irvine, California, United States of America
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Suphak Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Chea Nguon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | | | - Huy Rekol
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Cholrawee Promnarate
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Worldwide Antimalarial Resistance Network (WWARN) Asia Regional Centre, Mahidol University, Bangkok, Thailand
| | - Kesinee Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Podjanee Jittmala
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Worldwide Antimalarial Resistance Network (WWARN) Asia Regional Centre, Mahidol University, Bangkok, Thailand
| | - Mallika Imwong
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Pimnara Peerawaranun
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sasithon Pukrittayakamee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- The Royal Society of Thailand, Dusit, Bangkok, Thailand
| | - Paul N. Newton
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Guy E. Thwaites
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
- Oxford University Clinical Research Unit, Wellcome Trust Major Oversea Programme, Ho Chi Minh City, Vietnam
| | - Nicholas P. J. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Mayfong Mayxay
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Institute of Research and Education Development, University of Health Sciences, Vientiane, Lao People’s Demographic Republic
| | - Tran Tinh Hien
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
- Oxford University Clinical Research Unit, Wellcome Trust Major Oversea Programme, Ho Chi Minh City, Vietnam
| | - Francois H. Nosten
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Frank Cobelens
- Department of Global Health, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
- Amsterdam Institute for Global Health & Development, Amsterdam, Netherlands
| | - Arjen M. Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, England, United Kingdom
- * E-mail:
| |
Collapse
|
29
|
Ivermectin as a novel complementary malaria control tool to reduce incidence and prevalence: a modelling study. THE LANCET. INFECTIOUS DISEASES 2020; 20:498-508. [PMID: 31948767 DOI: 10.1016/s1473-3099(19)30633-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/30/2019] [Accepted: 10/21/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ivermectin is a potential new vector control tool to reduce malaria transmission. Mosquitoes feeding on a bloodmeal containing ivermectin have a reduced lifespan, meaning they are less likely to live long enough to complete sporogony and become infectious. We aimed to estimate the effect of ivermectin on malaria transmission in various scenarios of use. METHODS We validated an existing population-level mathematical model of the effect of ivermectin mass drug administration (MDA) on the mosquito population and malaria transmission against two datasets: clinical data from a cluster- randomised trial done in Burkina Faso in 2015 wherein ivermectin was given to individuals taller than 90 cm and entomological data from a study of mosquito outcomes after ivermectin MDA for onchocerciasis or lymphatic filariasis in Burkina Faso, Senegal, and Liberia between 2008 and 2013. We extended the existing model to include a range of complementary malaria interventions (seasonal malaria chemoprevention and MDA with dihydroartemisinin-piperaquine) and to incorporate new data on higher doses of ivermectin with a longer mosquitocidal effect. We consider two ivermectin regimens: a single dose of 400 μg/kg (1 × 400 μg/kg) and three consecutive daily doses of 300 μg/kg per day (3 × 300 μg/kg). We simulated the effect of these two doses in a range of usage scenarios in different transmission settings (highly seasonal, seasonal, and perennial). We report percentage reductions in clinical incidence and slide prevalence. FINDINGS We estimate that MDA with ivermectin will reduce prevalence and incidence and is most effective in areas with highly seasonal transmission. In a highly seasonal moderate transmission setting, three rounds of ivermectin only MDA at 3 × 300 μg/kg (rounds spaced 1 month apart) and 70% coverage is predicted to reduce clinical incidence by 71% and prevalence by 34%. We predict that adding ivermectin MDA to seasonal malaria chemoprevention in this setting would reduce clinical incidence by an additional 77% in children younger than 5 years compared with seasonal malaria chemoprevention alone; adding ivermectin MDA to MDA with dihydroartemisinin-piperaquine in this setting would reduce incidence by an additional 75% and prevalence by an additional 64% (all ages) compared with MDA with dihydroartemisinin-piperaquine alone. INTERPRETATION Our modelling predictions suggest that ivermectin could be a valuable addition to the malaria control toolbox, both in areas with persistently high transmission where existing interventions are insufficient and in areas approaching elimination to prevent resurgence. FUNDING Imperial College Junior Research Fellowship.
Collapse
|
30
|
Ethics and Antimalarial Drug Resistance. ETHICS AND DRUG RESISTANCE: COLLECTIVE RESPONSIBILITY FOR GLOBAL PUBLIC HEALTH 2020. [PMCID: PMC7586435 DOI: 10.1007/978-3-030-27874-8_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
There has been impressive progress in malaria control and treatment over the past two decades. One of the most important factors in the decline of malaria-related mortality has been the development and deployment of highly effective treatment in the form of artemisinin-based combination therapies (ACTs). However, recent reports suggest that these gains stand the risk of being reversed due to the emergence of ACT resistance in the Greater Mekong Subregion and the threat of this resistance spreading to Africa, where the majority of the world’s malaria cases occur, with catastrophic consequences. This chapter provides an overview of strategies proposed by malaria experts to tackle artemisinin-resistant malaria, and some of the most important practical ethical issues presented by each of these interventions. The proposed strategies include mass antimalarial drug administrations in selected populations, and mandatory screening of possibly infected individuals prior to entering an area free of artemisinin-resistant malaria. We discuss ethical issues such as tensions between the wishes of individuals versus the broader goal of malaria elimination, and the risks of harm to interventional populations, and conclude by proposing a set of recommendations.
Collapse
|
31
|
Kobylinski KC, Jittamala P, Hanboonkunupakarn B, Pukrittayakamee S, Pantuwatana K, Phasomkusolsil S, Davidson SA, Winterberg M, Hoglund RM, Mukaka M, van der Pluijm RW, Dondorp A, Day NPJ, White NJ, Tarning J. Safety, Pharmacokinetics, and Mosquito-Lethal Effects of Ivermectin in Combination With Dihydroartemisinin-Piperaquine and Primaquine in Healthy Adult Thai Subjects. Clin Pharmacol Ther 2019; 107:1221-1230. [PMID: 31697848 PMCID: PMC7285759 DOI: 10.1002/cpt.1716] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022]
Abstract
Mass administration of antimalarial drugs and ivermectin are being considered as potential accelerators of malaria elimination. The safety, tolerability, pharmacokinetics, and mosquito‐lethal effects of combinations of ivermectin, dihydroartemisinin‐piperaquine, and primaquine were evaluated. Coadministration of ivermectin and dihydroartemisinin‐piperaquine resulted in increased ivermectin concentrations with corresponding increases in mosquito‐lethal effect across all subjects. Exposure to piperaquine was also increased when coadministered with ivermectin, but electrocardiograph QT‐interval prolongation was not increased. One subject had transiently impaired liver function. Ivermectin mosquito‐lethal effect was greater than predicted previously against the major Southeast Asian malaria vectors. Both Anopheles dirus and Anopheles minimus mosquito mortality was increased substantially (20‐fold and 35‐fold increase, respectively) when feeding on volunteer blood after ivermectin administration compared with in vitro ivermectin‐spiked blood. This suggests the presence of ivermectin metabolites that impart mosquito‐lethal effects. Further studies of this combined approach to accelerate malaria elimination are warranted.
Collapse
Affiliation(s)
- Kevin C Kobylinski
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand.,Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Podjanee Jittamala
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sasithon Pukrittayakamee
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,The Royal Society of Thailand, Dusit, Bangkok, Thailand
| | - Kanchana Pantuwatana
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Siriporn Phasomkusolsil
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Silas A Davidson
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand.,Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Markus Winterberg
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Richard M Hoglund
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Mavuto Mukaka
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Rob W van der Pluijm
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Arjen Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Nicholas P J Day
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Peerawaranun P, Landier J, Nosten FH, Nguyen TN, Hien TT, Tripura R, Peto TJ, Phommasone K, Mayxay M, Day NPJ, Dondorp A, White N, von Seidlein L, Mukaka M. Intracluster correlation coefficients in the Greater Mekong Subregion for sample size calculations of cluster randomized malaria trials. Malar J 2019; 18:428. [PMID: 31852499 PMCID: PMC6921387 DOI: 10.1186/s12936-019-3062-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sample size calculations for cluster randomized trials are a recognized methodological challenge for malaria research in pre-elimination settings. Positively correlated responses from the participants in the same cluster are a key feature in the estimated sample size required for a cluster randomized trial. The degree of correlation is measured by the intracluster correlation coefficient (ICC) where a higher coefficient suggests a closer correlation hence less heterogeneity within clusters but more heterogeneity between clusters. METHODS Data on uPCR-detected Plasmodium falciparum and Plasmodium vivax infections from a recent cluster randomized trial which aimed at interrupting malaria transmission through mass drug administrations were used to calculate the ICCs for prevalence and incidence of Plasmodium infections. The trial was conducted in four countries in the Greater Mekong Subregion, Laos, Myanmar, Vietnam and Cambodia. Exact and simulation approaches were used to estimate ICC values for both the prevalence and the incidence of parasitaemia. In addition, the latent variable approach to estimate ICCs for the prevalence was utilized. RESULTS The ICCs for prevalence ranged between 0.001 and 0.082 for all countries. The ICC from the combined 16 villages in the Greater Mekong Subregion were 0.26 and 0.21 for P. falciparum and P. vivax respectively. The ICCs for incidence of parasitaemia ranged between 0.002 and 0.075 for Myanmar, Cambodia and Vietnam. There were very high ICCs for incidence in the range of 0.701 to 0.806 in Laos during follow-up. CONCLUSION ICC estimates can help researchers when designing malaria cluster randomized trials. A high variability in ICCs and hence sample size requirements between study sites was observed. Realistic sample size estimates for cluster randomized malaria trials in the Greater Mekong Subregion have to assume high between cluster heterogeneity and ICCs. This work focused on uPCR-detected infections; there remains a need to develop more ICC references for trials designed around prevalence and incidence of clinical outcomes. Adequately powered trials are critical to estimate the benefit of interventions to malaria in a reliable and reproducible fashion. TRIAL REGISTRATION ClinicalTrials.govNCT01872702. Registered 7 June 2013. Retrospectively registered. https://clinicaltrials.gov/ct2/show/NCT01872702.
Collapse
Affiliation(s)
- Pimnara Peerawaranun
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand
| | - Jordi Landier
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.,Aix-Marseille University, IRD, INSERM, SESSTIM, Marseille, France
| | - Francois H Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thuy-Nhien Nguyen
- Oxford University Clinical Research Unit, Wellcome Trust Major Oversea Programme, Ho Chi Minh City, Vietnam
| | - Tran Tinh Hien
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Oxford University Clinical Research Unit, Wellcome Trust Major Oversea Programme, Ho Chi Minh City, Vietnam
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Department of Global Health, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Koukeo Phommasone
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR.,Amsterdam Institute for Global Health & Development, Amsterdam, Netherlands
| | - Mayfong Mayxay
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR.,Institute of Research and Education Development, University of Health Sciences, Vientiane, Lao PDR
| | - Nicholas P J Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Arjen Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nick White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 60th Anniversary Chalermprakiat Building, 3rd Floor, 420/6 Ratchawithi Rd, Ratchathewi District, Bangkok, 10400, Thailand. .,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Kagaya W, Gitaka J, Chan CW, Kongere J, Md Idris Z, Deng C, Kaneko A. Malaria resurgence after significant reduction by mass drug administration on Ngodhe Island, Kenya. Sci Rep 2019; 9:19060. [PMID: 31836757 PMCID: PMC6910941 DOI: 10.1038/s41598-019-55437-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
Although WHO recommends mass drug administration (MDA) for malaria elimination, further evidence is required for understanding the obstacles for the optimum implementation of MDA. Just before the long rain in 2016, two rounds of MDA with artemisinin/piperaquine (Artequick) and low-dose primaquine were conducted with a 35-day interval for the entire population of Ngodhe Island (~500 inhabitants) in Lake Victoria, Kenya, which is surrounded by areas with moderate and high transmission. With approximately 90% compliance, Plasmodium prevalence decreased from 3% to 0% by microscopy and from 10% to 2% by PCR. However, prevalence rebounded to 9% by PCR two months after conclusion of MDA. Besides the remained local transmission, parasite importation caused by human movement likely contributed to the resurgence. Analyses of 419 arrivals to Ngodhe between July 2016 and September 2017 revealed Plasmodium prevalence of 4.6% and 16.0% by microscopy and PCR, respectively. Risk factors for infection among arrivals included age (0 to 5 and 11 to 15 years), and travelers from Siaya County, located to the north of Ngodhe Island. Parasite importation caused by human movement is one of major obstacles to sustain malaria elimination, suggesting the importance of cross-regional initiatives together with local vector control.
Collapse
Affiliation(s)
- Wataru Kagaya
- Department of Parasitology & Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka City University, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Jesse Gitaka
- Department of Clinical Medicine, Mount Kenya University, PO Box 342-01000, Thika, Kenya
| | - Chim W Chan
- Department of Parasitology & Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka City University, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.,Island Malaria Group, Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, 171 65, Solna, Stockholm, Sweden.,Department of Anthropology, Binghamton University, Binghamton, NY, 13905, USA
| | - James Kongere
- Nairobi Research Station, Nagasaki University Institute of Tropical Medicine-Kenya Medical Research Institute (NUITM-KEMRI) Project, Institute of Tropical Medicine (NEKKEN), Nagasaki University, PO Box 19993-00202, Nairobi, Kenya
| | - Zulkarnain Md Idris
- Island Malaria Group, Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, 171 65, Solna, Stockholm, Sweden.,Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000, Kuala Lumpur, Malaysia
| | - Changsheng Deng
- Science and Technology Park, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Akira Kaneko
- Department of Parasitology & Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka City University, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan. .,Island Malaria Group, Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, 171 65, Solna, Stockholm, Sweden. .,Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
34
|
Nsanzabana C. Strengthening Surveillance Systems for Malaria Elimination by Integrating Molecular and Genomic Data. Trop Med Infect Dis 2019; 4:E139. [PMID: 31816974 PMCID: PMC6958499 DOI: 10.3390/tropicalmed4040139] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/05/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
Unprecedented efforts in malaria control over the last 15 years have led to a substantial decrease in both morbidity and mortality in most endemic settings. However, these progresses have stalled over recent years, and resurgence may cause dramatic impact on both morbidity and mortality. Nevertheless, elimination efforts are currently going on with the objective of reducing malaria morbidity and mortality by 90% and malaria elimination in at least 35 countries by 2030. Strengthening surveillance systems is of paramount importance to reach those targets, and the integration of molecular and genomic techniques into routine surveillance could substantially improve the quality and robustness of data. Techniques such as polymerase chain reaction (PCR) and quantitative PCR (qPCR) are increasingly available in malaria endemic countries, whereas others such as sequencing are already available in a few laboratories. However, sequencing, especially next-generation sequencing (NGS), requires sophisticated infrastructure with adequate computing power and highly trained personnel for data analysis that require substantial investment. Different techniques will be required for different applications, and cost-effective planning must ensure the appropriate use of available resources. The development of national and sub-regional reference laboratories could help in minimizing the resources required in terms of equipment and trained staff. Concerted efforts from different stakeholders at national, sub-regional, and global level are needed to develop the required framework to establish and maintain these reference laboratories.
Collapse
Affiliation(s)
- Christian Nsanzabana
- Department of Medicine, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; ; Tel.: +41-61-284-82-52
- University of Basel, P.O. Box, CH-4003 Basel, Switzerland
| |
Collapse
|
35
|
Imwong M, Madmanee W, Suwannasin K, Kunasol C, Peto TJ, Tripura R, von Seidlein L, Nguon C, Davoeung C, Day NPJ, Dondorp AM, White NJ. Asymptomatic Natural Human Infections With the Simian Malaria Parasites Plasmodium cynomolgi and Plasmodium knowlesi. J Infect Dis 2019; 219:695-702. [PMID: 30295822 PMCID: PMC6376906 DOI: 10.1093/infdis/jiy519] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/30/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In Southeast Asia, Plasmodium knowlesi, a parasite of long-tailed macaques (Macaca fascicularis), is an important cause of human malaria. Plasmodium cynomolgi also commonly infects these monkeys, but only one naturally acquired symptomatic human case has been reported previously. METHODS Malariometric studies involving 5422 subjects (aged 6 months to 65 years) were conducted in 23 villages in Pailin and Battambang, western Cambodia. Parasite detection and genotyping was conducted on blood samples, using high-volume quantitative PCR (uPCR). RESULTS Asymptomatic malaria parasite infections were detected in 1361 of 14732 samples (9.2%). Asymptomatic infections with nonhuman primate malaria parasites were found in 21 individuals living close to forested areas; P. cynomolgi was found in 11, P. knowlesi was found in 8, and P. vivax and P. cynomolgi were both found in 2. Only 2 subjects were female, and 14 were men aged 20-40 years. Geometric mean parasite densities were 3604 parasites/mL in P. cynomolgi infections and 52488 parasites/mL in P. knowlesi infections. All P. cynomolgi isolates had wild-type dihydrofolate reductase genes, in contrast to the very high prevalence of mutations in the human malaria parasites. Asymptomatic reappearance of P. cynomolgi occurred in 2 subjects 3 months after the first infection. CONCLUSIONS Asymptomatic naturally acquired P. cynomolgi and P. knowlesi infections can both occur in humans. CLINICAL TRIALS REGISTRATION NCT01872702.
Collapse
Affiliation(s)
- Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Mahidol University, Bangkok, Thailand.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanassanan Madmanee
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kanokon Suwannasin
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Chanon Kunasol
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thomas J Peto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Rupam Tripura
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Lorenz von Seidlein
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Chea Nguon
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh
| | | | - Nicholas P J Day
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, United Kingdom
| |
Collapse
|
36
|
Peto TJ, Tripura R, Sanann N, Adhikari B, Callery J, Droogleever M, Heng C, Cheah PY, Davoeung C, Nguon C, von Seidlein L, Dondorp AM, Pell C. The feasibility and acceptability of mass drug administration for malaria in Cambodia: a mixed-methods study. Trans R Soc Trop Med Hyg 2019; 112:264-271. [PMID: 29917147 PMCID: PMC6044409 DOI: 10.1093/trstmh/try053] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/16/2018] [Indexed: 01/27/2023] Open
Abstract
Background Mass drug administrations (MDAs) are part of the World Health Organization’s Plasmodium falciparum elimination strategy for the Greater Mekong Subregion (GMS). In Cambodia, a 2015–2017 clinical trial evaluated the effectiveness of MDA. This article explores factors that influence the feasibility and acceptability of MDA, including seasonal timing, financial incentives and the delivery model. Methods Quantitative data were collected through structured questionnaires from the heads of 163 households. Qualitative data were collected through 25 semi-structured interviews and 5 focus group discussions with villagers and local health staff. Calendars of village activities were created and meteorological and malaria treatment records were collected. Results MDA delivered house-to-house or at a central point, with or without compensation, were equally acceptable and did not affect coverage. People who knew about the rationale for the MDA, asymptomatic infections and transmission were more likely to participate. In western Cambodia, MDA delivered house-to-house by volunteers at the end of the dry season may be most practicable but requires the subsequent treatment of in-migrants to prevent reintroduction of infections. Conclusions For MDA targeted at individual villages or village clusters it is important to understand local preferences for community mobilisation, delivery and timing, as several models of MDA are feasible.
Collapse
Affiliation(s)
- Thomas J Peto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Rupam Tripura
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Nou Sanann
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Bipin Adhikari
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - James Callery
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mark Droogleever
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Chhouen Heng
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Chan Davoeung
- Battambang Provincial Health Department, Mohatep Street, Battambang, Cambodia
| | - Chea Nguon
- National Centre for Parasitology, Entomology and Malaria Control, 477 Betong, Khan Sen Sok, Phnom Penh, Cambodia
| | - Lorenz von Seidlein
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Christopher Pell
- Centre for Social Science and Global Health, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Kaehler N, Adhikari B, Cheah PY, von Seidlein L, Day NPJ, Paris DH, Tanner M, Pell C. Prospects and strategies for malaria elimination in the Greater Mekong Sub-region: a qualitative study. Malar J 2019; 18:203. [PMID: 31221145 PMCID: PMC6585139 DOI: 10.1186/s12936-019-2835-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/09/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND As malaria elimination becomes a goal in malaria-endemic nations, questions of feasibility become critical. This article explores the potential challenges associated with this goal and future strategies for malaria elimination in the Greater Mekong Sub-region. METHODS Thirty-two semi-structured interviews were conducted with policy makers (n = 17) and principal investigators (n = 15) selected based on their involvement in malaria prevention, control and elimination in the GMS. Interviews were audio-recorded and transcribed for qualitative content (thematic) analysis using QSR NVivo. RESULTS All respondents described current malaria control and elimination strategies, such as case detection and management, prevention and strengthening of surveillance systems as critical and of equal priority. Aware of the emergence of multi-drug resistance in the GMS, researchers and policy makers outlined the need for additional elimination tools. As opposed to a centralized strategy, more targeted and tailored approaches to elimination were recommended. These included targeting endemic areas, consideration for local epidemiology and malaria species, and strengthening the peripheral health system. A decline in malaria transmission could lead to complacency amongst funders and policy makers resulting in a reduction or discontinuation of support for malaria elimination. Strong commitment of policymakers combined with strict monitoring and supervision by funders were considered pivotal to successful elimination programmes. CONCLUSION Against a backdrop of increasing anti-malarial resistance and decreasing choices of anti-malarial regimens, policy makers and researchers stressed the urgency of finding new malaria elimination strategies. There was consensus that multi-pronged strategies and approaches are needed, that no single potential tool/strategy can be appropriate to all settings. Hence there is a need to customize malaria control and elimination strategies based on the better surveillance data.
Collapse
Affiliation(s)
- Nils Kaehler
- Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, 4051, Basel, Switzerland.
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Bipin Adhikari
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Phaik Yeong Cheah
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- The Ethox Centre, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Lorenz von Seidlein
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Nicholas P J Day
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Daniel H Paris
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, 4051, Basel, Switzerland
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, 4051, Basel, Switzerland
| | - Christopher Pell
- Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam, The Netherlands
- Centre for Social Science and Global Health, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Ndong IC, Okyere D, Enos JY, Amambua-Ngwa A, Merle CSC, Nyarko A, Koram KA, Ahorlu CS. Challenges and perceptions of implementing mass testing, treatment and tracking in malaria control: a qualitative study in Pakro sub-district of Ghana. BMC Public Health 2019; 19:695. [PMID: 31170964 PMCID: PMC6554894 DOI: 10.1186/s12889-019-7037-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/23/2019] [Indexed: 01/13/2023] Open
Abstract
Background Malaria remains endemic in Ghana despite several interventions. Studies have demonstrated very high levels of asymptomatic malaria parasitaemia in both under-five and school-age children. Mass testing, treatment and tracking (MTTT) of malaria in communities is being proposed for implementation with the argument that it can reduce parasite load, amplify gains from the other control interventions and consequently lead to elimination. However, challenges associated with implementing MTTT such as feasibility, levels of coverage to be achieved for effectiveness, community perceptions and cost implications need to be clearly understood. This qualitative study was therefore conducted in an area with on-going MTTT to assess community and health workers’ perceptions about feasibility of scale-up and effectiveness to guide scale-up decisions. Methods This qualitative study employed purposive sampling to select the study participants. Ten focus group discussions (FGDs) were conducted in seven communities; eight with community members (n = 80) and two with health workers (n = 14). In addition, two in-depth interviews (IDI) were conducted, one with a Physician Assistant and another with a Laboratory Technician at the health facility. All interviews were recorded, transcribed, translated and analyzed using QSR NVivo 12. Results Both health workers and community members expressed positive perceptions about the feasibility of implementation and effectiveness of MTTT as an intervention that could reduce the burden of malaria in the community. MTTT implementation was perceived to have increased sensitisation about malaria, reduced the incidence of malaria, reduced household expenditure on malaria and alleviated the need to travel long distances for healthcare. Key challenges to implementation were doubts about the expertise of trained Community-Based Health Volunteers (CBHVs) to diagnose and treat malaria appropriately, side effects of Artemisinin-based Combination Therapies (ACTs) and misconceptions that CBHVs could infect children with epilepsy. Conclusion The study demonstrated that MTTT was perceived to be effective in reducing malaria incidence and related hospital visits in participating communities. MTTT was deemed useful in breaking financial and geographical barriers to accessing healthcare. The interventions were feasible and acceptable to community members, despite observed challenges to implementation such as concerns about CBHVs’ knowledge and skills and reduced revenue from internally generated funds (IGF) of the health facility.
Collapse
Affiliation(s)
- Ignatius Cheng Ndong
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana. .,Department of Biochemistry, Faculty of Science, Catholic University of Cameroon, Bamenda, Cameroon.
| | - Daniel Okyere
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Juliana Yartey Enos
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Alfred Amambua-Ngwa
- Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Corinne Simone C Merle
- Special Programme for Research & Training in Tropical Diseases (TDR), World Health Organization, Geneva, Switzerland
| | - Alexander Nyarko
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Clinical Pathology, Noguchi Memorial Institute for Medical Research, College of health Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Ansah Koram
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Collins Stephan Ahorlu
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
39
|
Kaehler N, Adhikari B, Cheah PY, Day NPJ, Paris DH, Tanner M, Pell C. The promise, problems and pitfalls of mass drug administration for malaria elimination: a qualitative study with scientists and policymakers. Int Health 2019; 11:166-176. [PMID: 30395228 PMCID: PMC6484636 DOI: 10.1093/inthealth/ihy079] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/01/2018] [Accepted: 09/09/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The emergence of artemisinin resistance in the Greater Mekong Subregion (GMS) has prompted urgent containment measures. One possible approach is mass drug administration (MDA). This article explores attitudes towards and perceptions of MDA for malaria elimination among policymakers and leading malariologists. METHODS Thirty-two semistructured interviews (SSI) were conducted with policymakers (n=17) and principal investigators (n=15) selected based on their involvement in malaria prevention, control and elimination in the GMS. Interviews were audio recorded and transcribed for qualitative content (thematic) analysis using NVivo (QSR International, Doncaster, Victoria, Australia). RESULTS Researchers and policymakers described reluctance and consequently delays to pilot MDA for malaria elimination. Most policymakers and some researchers reported concerns around the evidence base, citing a lack of data on its effectiveness and appropriate target populations. There were also worries about promoting resistance. Other issues included a previous lack of support from the World Health Organization, past MDAs, the remoteness of target populations and challenges explaining the rationale for MDA. CONCLUSIONS The complex rationale for MDA for malaria elimination, mistaking pilot studies for implementation, past experiences with MDA, difficulties in selecting appropriate sites and the WHO's lack of clear backing undermined the support for MDA for malaria elimination.
Collapse
Affiliation(s)
- Nils Kaehler
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Bipin Adhikari
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Phaik Yeong Cheah
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Ethox Centre, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, UK
| | - Nicholas P J Day
- Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel H Paris
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Christopher Pell
- Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam Health Technology Centre, Tower C4, Paasheuvelweg 25, BP Amsterdam, The Netherlands
- Centre for Social Science and Global Health, University of Amsterdam, Nieuwe Achtergracht 166, WV, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Parker DM, Tun STT, White LJ, Kajeechiwa L, Thwin MM, Landier J, Chaumeau V, Corbel V, Dondorp AM, von Seidlein L, White NJ, Maude RJ, Nosten F. Potential herd protection against Plasmodium falciparum infections conferred by mass antimalarial drug administrations. eLife 2019; 8:e41023. [PMID: 30990166 PMCID: PMC6467567 DOI: 10.7554/elife.41023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 03/25/2019] [Indexed: 11/13/2022] Open
Abstract
The global malaria burden has decreased over the last decade and many nations are attempting elimination. Asymptomatic malaria infections are not normally diagnosed or treated, posing a major hurdle for elimination efforts. One solution to this problem is mass drug administration (MDA), with success depending on adequate population participation. Here, we present a detailed spatial and temporal analysis of malaria episodes and asymptomatic infections in four villages undergoing MDA in Myanmar. In this study, individuals from neighborhoods with low MDA adherence had 2.85 times the odds of having a malaria episode post-MDA in comparison to those from high adherence neighborhoods, regardless of individual participation, suggesting a herd effect. High mosquito biting rates, living in a house with someone else with malaria, or having an asymptomatic malaria infection were also predictors of clinical episodes. Spatial clustering of non-adherence to MDA, even in villages with high overall participation, may frustrate elimination efforts.
Collapse
Affiliation(s)
- Daniel M Parker
- Department of Population Health and Disease PreventionUniversity of CaliforniaIrvineUnited States
| | - Sai Thein Than Tun
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
| | - Lisa J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
| | - Ladda Kajeechiwa
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
| | - May Myo Thwin
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
| | - Jordi Landier
- Institut de Recherche pour le DéveloppementUniversity of MontpellierMontpellierFrance
| | - Victor Chaumeau
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre Hospitalier Universitaire de MontpellierMontpellierFrance
- Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle IRD 224-CNRS 5290UM1-UM2, Institut de Recherche pour le Développement (IRD)University of MontpellierMontpellierFrance
| | - Vincent Corbel
- Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle IRD 224-CNRS 5290UM1-UM2, Institut de Recherche pour le Développement (IRD)University of MontpellierMontpellierFrance
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
| | - Lorenz von Seidlein
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
| | - Richard J Maude
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
- Harvard TH Chan School of Public HealthHarvard UniversityHarvardUnited States
| | - François Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUnited kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityNakhon PathomThailand
| |
Collapse
|
41
|
Rijal KR, Adhikari B, Adhikari N, Dumre SP, Banjara MS, Shrestha UT, Banjara MR, Singh N, Ortegea L, Lal BK, Thakur GD, Ghimire P. Micro-stratification of malaria risk in Nepal: implications for malaria control and elimination. Trop Med Health 2019; 47:21. [PMID: 30976192 PMCID: PMC6437900 DOI: 10.1186/s41182-019-0148-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/14/2019] [Indexed: 11/10/2022] Open
Abstract
Background A significant reduction in malaria cases over the recent years in Nepal has encouraged the government to adopt a goal of "malaria-free nation by 2025." Nevertheless, to achieve this goal, it is critical to identify the epidemiological burden of malaria by specific regions and areas for an effective targeted intervention. The main objective of this study was to estimate the risk of malaria at Village Development Committee (VDC) level in Nepal based on disease, vector, parasite, and geography. Methods In 2012, the micro-stratification of malaria risk was carried out in 75 districts of Nepal. Instruments such as a questionnaire, case record forms, and guidelines for malaria micro-stratification were developed and pre-tested for necessary adaptations. Village Development Committee (VDC)-wise malaria data were analyzed using exploratory statistics and were stratified by geographical variables that contributed to the risk of malaria. To understand the transmission risk at VDC level, overlay analysis was done using ArcGIS 10. To ensure transparent, reproducible, and comprehensible risk assessment, standard scoring method was selected and utilized for data from 2009 to 2011. Thus identified, three major variables (key determinants) were given weights (wt.) accordingly to stratification of the malaria risk (disease burden, "0.3" wt.; ecology/vector transmission, "0.5" wt.; and vulnerability-population movement, "0.2" wt.). Malaria risk in a VDC was determined based on the overall scores and classified into four categories: no risk, low risk, moderate risk, and high risk. Results Analyzing the overall risk based on scoring of the total VDCs (n = 3976), 54 (1.36%), 201 (5.06%), 999 (25.13%), and 2718 (68.36%) were identified as high-, moderate-, low-, and no-risk categories for malaria, respectively. Based on the population statistics, 3.62%, 9.79%, 34.52%, and 52.05% of the country's total population live in high-risk, moderate-risk, low-risk, and no-risk VDCs for malaria, respectively. Our micro-stratification study estimates are 100,000 population at high risk. Regional distribution showed that the majority of the high-risk VDCs were identified in the Far- and Mid-western regions (19 and 18 VDCs) followed by Central and Western regions (10 and 7 VDCs) with no high-risk VDCs in the Eastern region. Similarly, 77, 59, 27, 24, and 14 VDCs of the Central, Mid-western, Western, Eastern, and Far-western regions, respectively, were found under moderate malaria risk. Of the low-risk VDCs, 353, 215, 191, 148, and 92 were respectively from the Central, Eastern, Western, Far-western, and Mid-western regions. Conclusions The current micro-stratification study provides insights on malaria risk up to the VDC level. This will help the malaria elimination program to target interventions at the local level thereby ensuring the best utilization of available resources to substantially narrowed-down target areas. With further updates and refinement, the micro-stratification approach can be employed to identify the risk areas up to smaller units within the VDCs (ward and villages).
Collapse
Affiliation(s)
- Komal Raj Rijal
- 1Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Bipin Adhikari
- 2Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nabaraj Adhikari
- 1Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Shyam Prakash Dumre
- 3Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | | | | | - Megha Raj Banjara
- 1Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Nihal Singh
- World Health Organization (WHO), Country Office, Kathmandu, Nepal
| | - Leonard Ortegea
- 6Global Malaria Program, World Health Organization Headquarters, Geneva, Switzerland
| | - Bibek Kumar Lal
- 7Epidemiology and Disease Control Division (EDCD), Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | - Garib Das Thakur
- 7Epidemiology and Disease Control Division (EDCD), Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | - Prakash Ghimire
- 1Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal.,World Health Organization (WHO), Country Office, Kathmandu, Nepal
| |
Collapse
|
42
|
Pell CL, Adhikari B, Myo Thwin M, Kajeechiwa L, Nosten S, Nosten FH, Sahan KM, Smithuis FM, Nguyen TN, Hien TT, Tripura R, Peto TJ, Sanann N, Nguon C, Pongvongsa T, Phommasone K, Mayxay M, Mukaka M, Peerawaranun P, Kaehler N, Cheah PY, Day NPJ, White NJ, Dondorp AM, von Seidlein L. Community engagement, social context and coverage of mass anti-malarial administration: Comparative findings from multi-site research in the Greater Mekong sub-Region. PLoS One 2019; 14:e0214280. [PMID: 30908523 PMCID: PMC6433231 DOI: 10.1371/journal.pone.0214280] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/11/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Between 2013 and 2017, targeted malaria elimination (TME), a package of interventions that includes mass drug administration (MDA)-was piloted in communities with reservoirs of asymptomatic P. falciparum across the Greater Mekong sub-Region (GMS). Coverage in target communities is a key determinant of the effectiveness of MDA. Drawing on mixed methods research conducted alongside TME pilot studies, this article examines the impact of the community engagement, local social context and study design on MDA coverage. METHODS AND FINDINGS Qualitative and quantitative data were collected using questionnaire-based surveys, semi-structured and in-depth interviews, focus group discussions, informal conversations, and observations of study activities. Over 1500 respondents were interviewed in Myanmar, Vietnam, Cambodia and Laos. Interview topics included attitudes to malaria and experiences of MDA. Overall coverage of mass anti-malarial administration was high, particularly participation in at least a single round (85%). Familiarity with and concern about malaria prompted participation in MDA; as did awareness of MDA and familiarity with the aim of eliminating malaria. Fear of adverse events and blood draws discouraged people. Hence, community engagement activities sought to address these concerns but their impact was mediated by the trust relationships that study staff could engender in communities. In contexts of weak healthcare infrastructure and (cash) poverty, communities valued the study's ancillary care and the financial compensation. However, coverage did not necessarily decrease in the absence of cash compensation. Community dynamics, affected by politics, village conformity, and household decision-making also affected coverage. CONCLUSIONS The experimental nature of TME presented particular challenges to achieving high coverage. Nonetheless, the findings reflect those from studies of MDA under implementation conditions and offer useful guidance for potential regional roll-out of MDA: it is key to understand target communities and provide appropriate information in tailored ways, using community engagement that engenders trust.
Collapse
Affiliation(s)
- Christopher L. Pell
- Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam, The Netherlands
- Centre for Social Science and Global Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - May Myo Thwin
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ladda Kajeechiwa
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Suphak Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Francois H. Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Sorbonne Universités, UPMC Univ Paris 06, UPMC UMRS CR7, Paris, France
| | - Kate M. Sahan
- Ethox Centre and Wellcome Centre for Ethics and Humanities, Nuffield Department of Population Health, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Frank M. Smithuis
- Medical Action Myanmar, Yangon, Myanmar
- Myanmar Oxford Clinical Research Unit, Yangon, Myanmar
| | - Thuy-Nhien Nguyen
- Oxford University Clinical Research Unit, Wellcome Trust Asia Programme, Ho Chi Minh City, Vietnam
| | - Tran Tinh Hien
- Oxford University Clinical Research Unit, Wellcome Trust Asia Programme, Ho Chi Minh City, Vietnam
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas J. Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nou Sanann
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Chea Nguon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | | | - Koukeo Phommasone
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Vientiane, Lao PDR
| | - Mayfong Mayxay
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Vientiane, Lao PDR
- Institute of Research and Educational Development, University of Health Sciences, Vientiane, Lao PDR
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Pimnara Peerawaranun
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nils Kaehler
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Ethox Centre and Wellcome Centre for Ethics and Humanities, Nuffield Department of Population Health, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Nicholas P. J. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Arjen M. Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
The role of monitoring and evaluation to ensure functional access to community-based early diagnosis and treatment in a malaria elimination programme in Eastern Myanmar. Malar J 2019; 18:50. [PMID: 30795764 PMCID: PMC6387481 DOI: 10.1186/s12936-019-2677-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/14/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Improving access to early diagnosis and treatment (EDT) has increasingly proven to be a major contributor to decreasing malaria incidence in low-transmission settings. The Malaria Elimination Task Force (METF) has deployed malaria posts set up in Eastern Myanmar, providing free uninterrupted community-based access to EDT in more than 1200 villages. Ensuring high quality services are provided by these malaria posts is essential to reaching elimination targets. The present study aimed to determine the functionality of the malaria posts in the METF programme. METHODS This report analysed routinely collected data (weekly reports, individual consultation, diagnostic test quality control) and data collected specifically during monitoring and evaluation visits using descriptive statistics and univariate logistic regression. The presence of major dysfunctions (stock-outs and reported closing; likely to impair the ability of the population to access EDT) or minor dysfunctions (no formal METF training, lack of regular salary, forms and manual not on-site, and low frequency of supervisor visits) and the ability to anticipate dysfunctions through analysis of weekly reports were assessed. RESULTS A total of 65% of malaria posts had no major dysfunction identified during monitoring and evaluation visits, while 86% of malaria posts were fully stocked with tests and medicines used for treatment. Diagnosis was correctly conducted with few false positives and rare mis-speciation of results. Malaria post worker knowledge of malaria treatments showed few gaps, mostly in the treatment of more complex presentations. Malaria posts were well utilized in the population, with 94% of consultations occurring within the first 3 days of fever. In the regression analysis, reported stock-outs and delayed weekly reports were associated with observed major and minor dysfunctions in monitoring and evaluation visits, emphasizing the need to reinforce support to malaria post supervisors, who were responsible for the local logistics of supply and data transmission and day-to-day supervision. CONCLUSION The malaria posts operating under the METF programme perform to a high standard, with the majority offering uninterrupted access to diagnosis and treatment, and high service uptake in the villages serviced by the programme. However, programme operations can be strengthened by increasing malaria post supervisor visits and re-training malaria post workers.
Collapse
|
44
|
von Seidlein L, Peto TJ, Landier J, Nguyen TN, Tripura R, Phommasone K, Pongvongsa T, Lwin KM, Keereecharoen L, Kajeechiwa L, Thwin MM, Parker DM, Wiladphaingern J, Nosten S, Proux S, Corbel V, Tuong-Vy N, Phuc-Nhi TL, Son DH, Huong-Thu PN, Tuyen NTK, Tien NT, Dong LT, Hue DV, Quang HH, Nguon C, Davoeung C, Rekol H, Adhikari B, Henriques G, Phongmany P, Suangkanarat P, Jeeyapant A, Vihokhern B, van der Pluijm RW, Lubell Y, White LJ, Aguas R, Promnarate C, Sirithiranont P, Malleret B, Rénia L, Onsjö C, Chan XH, Chalk J, Miotto O, Patumrat K, Chotivanich K, Hanboonkunupakarn B, Jittmala P, Kaehler N, Cheah PY, Pell C, Dhorda M, Imwong M, Snounou G, Mukaka M, Peerawaranun P, Lee SJ, Simpson JA, Pukrittayakamee S, Singhasivanon P, Grobusch MP, Cobelens F, Smithuis F, Newton PN, Thwaites GE, Day NPJ, Mayxay M, Hien TT, Nosten FH, Dondorp AM, White NJ. The impact of targeted malaria elimination with mass drug administrations on falciparum malaria in Southeast Asia: A cluster randomised trial. PLoS Med 2019; 16:e1002745. [PMID: 30768615 PMCID: PMC6377128 DOI: 10.1371/journal.pmed.1002745] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The emergence and spread of multidrug-resistant Plasmodium falciparum in the Greater Mekong Subregion (GMS) threatens global malaria elimination efforts. Mass drug administration (MDA), the presumptive antimalarial treatment of an entire population to clear the subclinical parasite reservoir, is a strategy to accelerate malaria elimination. We report a cluster randomised trial to assess the effectiveness of dihydroartemisinin-piperaquine (DP) MDA in reducing falciparum malaria incidence and prevalence in 16 remote village populations in Myanmar, Vietnam, Cambodia, and the Lao People's Democratic Republic, where artemisinin resistance is prevalent. METHODS AND FINDINGS After establishing vector control and community-based case management and following intensive community engagement, we used restricted randomisation within village pairs to select 8 villages to receive early DP MDA and 8 villages as controls for 12 months, after which the control villages received deferred DP MDA. The MDA comprised 3 monthly rounds of 3 daily doses of DP and, except in Cambodia, a single low dose of primaquine. We conducted exhaustive cross-sectional surveys of the entire population of each village at quarterly intervals using ultrasensitive quantitative PCR to detect Plasmodium infections. The study was conducted between May 2013 and July 2017. The investigators randomised 16 villages that had a total of 8,445 residents at the start of the study. Of these 8,445 residents, 4,135 (49%) residents living in 8 villages, plus an additional 288 newcomers to the villages, were randomised to receive early MDA; 3,790 out of the 4,423 (86%) participated in at least 1 MDA round, and 2,520 out of the 4,423 (57%) participated in all 3 rounds. The primary outcome, P. falciparum prevalence by month 3 (M3), fell by 92% (from 5.1% [171/3,340] to 0.4% [12/2,828]) in early MDA villages and by 29% (from 7.2% [246/3,405] to 5.1% [155/3,057]) in control villages. Over the following 9 months, the P. falciparum prevalence increased to 3.3% (96/2,881) in early MDA villages and to 6.1% (128/2,101) in control villages (adjusted incidence rate ratio 0.41 [95% CI 0.20 to 0.84]; p = 0.015). Individual protection was proportional to the number of completed MDA rounds. Of 221 participants with subclinical P. falciparum infections who participated in MDA and could be followed up, 207 (94%) cleared their infections, including 9 of 10 with artemisinin- and piperaquine-resistant infections. The DP MDAs were well tolerated; 6 severe adverse events were detected during the follow-up period, but none was attributable to the intervention. CONCLUSIONS Added to community-based basic malaria control measures, 3 monthly rounds of DP MDA reduced the incidence and prevalence of falciparum malaria over a 1-year period in areas affected by artemisinin resistance. P. falciparum infections returned during the follow-up period as the remaining infections spread and malaria was reintroduced from surrounding areas. Limitations of this study include a relatively small sample of villages, heterogeneity between villages, and mobility of villagers that may have limited the impact of the intervention. These results suggest that, if used as part of a comprehensive, well-organised, and well-resourced elimination programme, DP MDA can be a useful additional tool to accelerate malaria elimination. TRIAL REGISTRATION ClinicalTrials.gov NCT01872702.
Collapse
Affiliation(s)
- Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas J. Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jordi Landier
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Institut de Recherche pour le Développement, Aix–Marseille University, INSERM, SESSTIM, Marseille, France
| | - Thuy-Nhien Nguyen
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Koukeo Phommasone
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Amsterdam Institute for Global Health & Development, Amsterdam, The Netherlands
| | - Tiengkham Pongvongsa
- Savannakhet Provincial Health Department, Savannakhet Province, Lao People’s Democratic Republic
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Khin Maung Lwin
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Lilly Keereecharoen
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ladda Kajeechiwa
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - May Myo Thwin
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Daniel M. Parker
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Department of Population Health and Disease Prevention, University of California, Irvine, Irvine, California, United States of America
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Suphak Nosten
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Vincent Corbel
- Maladies Infectieuses et Vecteurs: Écologie, Génétique, Evolution et Contrôle, Institut de Recherche pour le Développement, Université Montpellier, Montpellier, France
| | - Nguyen Tuong-Vy
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Truong Le Phuc-Nhi
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Do Hung Son
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Pham Nguyen Huong-Thu
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Kim Tuyen
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Nguyen Thanh Tien
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Le Thanh Dong
- Institute of Malariology, Parasitology, and Entomology, Ho Chi Minh City, Vietnam
| | - Dao Van Hue
- Center for Malariology, Parasitology and Entomology, Ninh Thuan Province, Vietnam
| | - Huynh Hong Quang
- Institute of Malariology, Parasitology, and Entomology, Quy Nhon, Vietnam
| | - Chea Nguon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | | | - Huy Rekol
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gisela Henriques
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Panom Phongmany
- Savannakhet Provincial Health Department, Savannakhet Province, Lao People’s Democratic Republic
| | - Preyanan Suangkanarat
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
| | - Atthanee Jeeyapant
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Benchawan Vihokhern
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rob W. van der Pluijm
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Yoel Lubell
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lisa J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ricardo Aguas
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Cholrawee Promnarate
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- WWARN Asia Regional Centre, Mahidol University, Bangkok, Thailand
| | - Pasathorn Sirithiranont
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Benoit Malleret
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Carl Onsjö
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Faculty of Medicine and Health Sciences, Linköping University, Linköping, Linköping, Sweden
| | - Xin Hui Chan
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jeremy Chalk
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Olivo Miotto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Krittaya Patumrat
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kesinee Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Podjanee Jittmala
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nils Kaehler
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Christopher Pell
- Amsterdam Institute for Global Health & Development, Amsterdam, The Netherlands
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- WWARN Asia Regional Centre, Mahidol University, Bangkok, Thailand
| | - Mallika Imwong
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Georges Snounou
- CEA–Université Paris Sud 11–INSERM U1184, IDMIT, Direction de la Recherche Fondamentale, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Fontenay-aux-Roses, France
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pimnara Peerawaranun
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sue J. Lee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julie A. Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Sasithon Pukrittayakamee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Royal Society of Thailand, Bangkok, Thailand
| | - Pratap Singhasivanon
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Martin P. Grobusch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank Cobelens
- Amsterdam Institute for Global Health & Development, Amsterdam, The Netherlands
| | | | - Paul N. Newton
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
| | - Guy E. Thwaites
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Nicholas P. J. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mayfong Mayxay
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Institute of Research and Education Development, University of Health Sciences, Vientiane, Lao People’s Democratic Republic
| | - Tran Tinh Hien
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Institut de Recherche pour le Développement, Aix–Marseille University, INSERM, SESSTIM, Marseille, France
| | - Francois H. Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Arjen M. Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Morris U, Msellem MI, Mkali H, Islam A, Aydin-Schmidt B, Jovel I, Shija SJ, Khamis M, Ali SM, Hodzic L, Magnusson E, Poirot E, Bennett A, Sachs MC, Tarning J, Mårtensson A, Ali AS, Björkman A. A cluster randomised controlled trial of two rounds of mass drug administration in Zanzibar, a malaria pre-elimination setting-high coverage and safety, but no significant impact on transmission. BMC Med 2018; 16:215. [PMID: 30526588 PMCID: PMC6287359 DOI: 10.1186/s12916-018-1202-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/29/2018] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Mass drug administration (MDA) has the potential to interrupt malaria transmission and has been suggested as a tool for malaria elimination in low-endemic settings. This study aimed to determine the effectiveness and safety of two rounds of MDA in Zanzibar, a pre-elimination setting. METHODS A cluster randomised controlled trial was conducted in 16 areas considered as malaria hotspots, with an annual parasite index of > 0.8%. The areas were randomised to eight intervention and eight control clusters. The intervention included two rounds of MDA with dihydroartemisinin-piperaquine and single low-dose primaquine 4 weeks apart in May-June 2016. Primary and secondary outcomes were cumulative confirmed malaria case incidences 6 months post-MDA and parasite prevalences determined by PCR 3 months post-MDA. Additional outcomes included intervention coverage, treatment adherence, occurrence of adverse events, and cumulative incidences 3, 12, and 16 months post-MDA. RESULTS Intervention coverage was 91.0% (9959/10944) and 87.7% (9355/10666) in the first and second rounds, respectively; self-reported adherence was 82.0% (881/1136) and 93.7% (985/1196). Adverse events were reported in 11.6% (147/1268) and 3.2% (37/1143) of post-MDA survey respondents after both rounds respectively. No serious adverse event was reported. No difference in cumulative malaria case incidence was observed between the control and intervention arms 6 months post-MDA (4.2 and 3.9 per 1000 population; p = 0.94). Neither was there a difference in PCR-determined parasite prevalences 3 months post-MDA (1.4% and 1.7%; OR = 1.0, p = 0.94), although having received at least the first MDA was associated with reduced odds of malaria infection (aOR = 0.35; p = 0.02). Among confirmed malaria cases at health facilities, 26.0% and 26.3% reported recent travel outside Zanzibar in the intervention and control shehias (aOR ≥ 85; p ≤ 0.001). CONCLUSIONS MDA was implemented with high coverage, adherence, and tolerability. Despite this, no significant impact on transmission was observed. The findings suggest that two rounds of MDA in a single year may not be sufficient for a sustained impact on transmission in a pre-elimination setting, especially when the MDA impact is restricted by imported malaria. Importantly, this study adds to the limited evidence for the use of MDA in low transmission settings in sub-Saharan Africa. TRIAL REGISTRATION ClinicalTrials.gov, NCT02721186 (registration date: March 29, 2016).
Collapse
Affiliation(s)
- Ulrika Morris
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mwinyi I. Msellem
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, Tanzania
| | - Humphrey Mkali
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, Tanzania
| | - Atiqul Islam
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Berit Aydin-Schmidt
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Irina Jovel
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shija Joseph Shija
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, Tanzania
| | - Mwinyi Khamis
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, Tanzania
| | - Safia Mohammed Ali
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, Tanzania
| | - Lamija Hodzic
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Magnusson
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eugenie Poirot
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, USA
| | - Adam Bennett
- Malaria Elimination Initiative, Global Health Group, University of California San Francisco, San Francisco, USA
| | - Michael C. Sachs
- Biostatistics Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Bangkok, Thailand
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine, University of Oxford, Oxford, UK
| | - Andreas Mårtensson
- Department of Women’s and Children’s Health, International Maternal and Child Health, Uppsala University, Uppsala, Sweden
| | - Abdullah S. Ali
- Zanzibar Malaria Elimination Programme, Ministry of Health, Zanzibar, Tanzania
| | - Anders Björkman
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Pongvongsa T, Phommasone K, Adhikari B, Henriques G, Chotivanich K, Hanboonkunupakarn B, Mukaka M, Peerawaranun P, von Seidlein L, Day NPJ, White NJ, Dondorp AM, Imwong M, Newton PN, Singhasivanon P, Mayxay M, Pukrittayakamee S. The dynamic of asymptomatic Plasmodium falciparum infections following mass drug administrations with dihydroarteminisin-piperaquine plus a single low dose of primaquine in Savannakhet Province, Laos. Malar J 2018; 17:405. [PMID: 30390647 PMCID: PMC6215638 DOI: 10.1186/s12936-018-2541-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/20/2018] [Indexed: 11/21/2022] Open
Abstract
Background The increase in multidrug resistant Plasmodium falciparum infections threatens the malaria elimination goals in countries within the Greater Mekong Sub-region. A multi-pronged approach assuring access to basic malaria control measures, including insecticide-treated bed nets and early diagnosis and treatment was followed by mass drug administrations (MDA) in southern Savannakhet Province, Laos. The main objective of this study was to evaluate the effectiveness and safety of mass drug administrations as well as their effects on the dynamic of asymptomatic P. falciparum infections in 4 malaria endemic villages. Methods Two villages were randomized to early MDA consisting of 3 rounds of a 3-day course of dihydroartemisinin–piperaquine with a single low dose of primaquine. In the other 2 villages MDA was deferred by 1 year. A total of 1036 residents were enrolled in early MDA villages and 883 in control villages (deferred-MDA). Tri-monthly parasitaemia surveys using uPCR were conducted for a year in the 4 villages. Results Eighty-four percent (872/1036) of the residents participated in the MDAs, of whom 90% (781/872) completed 3 rounds of MDA (9 doses). In intervention villages, the prevalence of asymptomatic P. falciparum infections decreased by 85% after MDA from 4.8% (95% CI 3.4–6.4) at baseline (month 0 or M0) to 0.7% (95% CI 0.3–1.6) at month 12. In control villages there was a decrease of 33% in P. falciparum prevalence between M0: 17.5% (95% CI 15.9–20.3) and M12: 11.6% (95% CI 9.3–14.2). In bivariate and multivariate analyses P. falciparum infections were significantly reduced with early MDA (adjusted incidence rate ratios (AIRR): 0.08, CI 0.01–0.091) and completion of 3 MDA rounds (AIRR: 0.06; CI 0.01–0.66). A quarter of participants (226/872) reported adverse events of which 99% were mild. Conclusion The study found a significant reduction in P. falciparum prevalence and incidence following MDA. MDA was safe, well tolerated, feasible, and achieved high population coverage and adherence. MDAs must be integrated in multi-pronged approaches such as vector control and preventive measures with a focus on specific risk groups such as mobile, migrant population and forest goers for a sustained period to eliminate the remaining parasite reservoirs. Trial registration ClinicalTrials.gov Identifier: NCT01872702
Collapse
Affiliation(s)
- Tiengkham Pongvongsa
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Savannakhet Provincial Health Department, Savannakhet, Savannakhet Province, Laos
| | - Koukeo Phommasone
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Vientiane, Laos
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Gisela Henriques
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kesinee Chotivanich
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Pimnara Peerawaranun
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
| | - Nicholas P J Day
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Mallika Imwong
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Paul N Newton
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Vientiane, Laos.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | | | - Mayfong Mayxay
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Vientiane, Laos.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,Institute of Research and Education Development, University of Health Sciences, Vientiane, Laos
| | - Sasithon Pukrittayakamee
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,The Royal Society, Dusit Palace, Bangkok, Thailand
| |
Collapse
|
47
|
Ghimire P, Rijal KR, Kafle C, Karki BS, Singh N, Ortega L, Thakur GD, Adhikari B. Efficacy of artemether-lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria in Nepal. Trop Dis Travel Med Vaccines 2018; 4:9. [PMID: 30123520 PMCID: PMC6092778 DOI: 10.1186/s40794-018-0068-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/05/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The national treatment guidelines of Nepal have adopted Artemisinin Combination Therapies (ACTs) for the treatment of uncomplicated falciparum malaria since 2004. Emergence of Artemisinin resistance in the Greater Mekong Sub-region (GMS) and beyond may become a threat for Nepal as well. The main objective of this study was to assess the therapeutic efficacy of antimalarial drug artemether-lumefantrine in uncomplicated P. falciparum infected patients at health centers/hospitals treated over the period of 2 years (2013-2014). METHODS Giemsa stained thick and thin smears, prepared from uncomplicated falciparum malaria patients who visited the selected sentinel sites in Nepal during 2013 to 2014 and met the inclusion criteria that included parasitemia (1000-10,000 /μL of blood), were evaluated until 28 days after ACTs treatment, following a World Health Organization (WHO) therapeutic efficacy protocol. Based on the re-occurrence of fever and resurge in parasitemia, the study patients were classified as resistant or susceptible. Blood specimens on filter papers were further analyzed by Polymerase Chain Reaction (PCR), specifically for the K13 propeller gene mutation (a recently identified molecular marker for ACT resistance). RESULTS A total of 56,013 suspected malaria cases were screened for this study. Of which, 120 (0.21%) were infected with falciparum malaria. Out of 120, 28 cases of P. falciparum (28/120; 23.33%) were enrolled in the study, of which 24 cases completed the post-treatment follow up for 28 days. Only one case out of 24 (4%) was identified as a late treatment failure (LTF). K13 mutation, a proxy indicator for ACT resistance in parasites, was not detected on the day 1, which indicates resistance had not yet reached the molecular level. CONCLUSION Only one case of late treatment failure was identified in this study. ACT combination using artemether-lumefantrine was still effective for the treatment of uncomplicated falciparum malaria in Nepal. A close monitoring and supervision for ACT resistance is essential for future malaria treatment in Nepal.
Collapse
Affiliation(s)
- Prakash Ghimire
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Komal Raj Rijal
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Chandramani Kafle
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | | | - Nihal Singh
- World Health Organization, Country office Nepal, UN House, Pulchowk, Lalitpur, Nepal
| | - Leonard Ortega
- Global Malaria Program, World Health Organization headquarters, Geneva, Switzerland
| | | | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
48
|
Chan XHS, Win YN, Mawer LJ, Tan JY, Brugada J, White NJ. Risk of sudden unexplained death after use of dihydroartemisinin-piperaquine for malaria: a systematic review and Bayesian meta-analysis. THE LANCET. INFECTIOUS DISEASES 2018; 18:913-923. [PMID: 29887371 PMCID: PMC6060085 DOI: 10.1016/s1473-3099(18)30297-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dihydroartemisinin-piperaquine is an effective and well tolerated artemisinin-based combination therapy that has been assessed extensively for the prevention and treatment of malaria. Piperaquine, similar to several structurally related antimalarials currently used, can prolong cardiac ventricular repolarisation duration and the electrocardiographic QT interval, leading to concerns about its proarrhythmic potential. We aimed to assess the risk of potentially lethal iatrogenic ventricular arrhythmias in individuals receiving dihydroartemisinin-piperaquine. METHODS We did a systematic review and Bayesian meta-analysis. We searched clinical bibliographic databases (last on May 24, 2017) for studies of dihydroartemisinin-piperaquine in human beings. Further unpublished studies were identified with the WHO Evidence Review Group on the Cardiotoxicity of Antimalarials. We searched for articles containing "dihydroartemisinin-piperaquine" as title, abstract, or subject heading keywords, with synonyms and variant spellings as additional search terms. We excluded animal studies, but did not apply limits on language or publication date. Eligible studies were prospective, randomised, controlled trials or cohort studies in which individuals received at least one 3-day treatment course of dihydroartemisinin-piperaquine for mass drug administration, preventive therapy, or case management of uncomplicated malaria, with follow-up over at least 3 days. At least two independent reviewers screened titles, abstracts, and full texts, agreed study eligibility, and extracted information about study and participant characteristics, adverse event surveillance methodology, dihydroartemisinin-piperaquine exposures, loss-to-follow up, and any deaths after dihydroartemisinin-piperaquine treatment into a standardised database. The risk of sudden unexplained death after dihydroartemisinin-piperaquine with 95% credible intervals (CI) generated by Bayesian meta-analysis was compared with the baseline rate of sudden cardiac death. FINDINGS Our search identified 94 eligible primary studies including data for 197 867 individuals who had received dihydroartemisinin-piperaquine: 154 505 in mass drug administration programmes; 15 188 in 14 studies of repeated courses in preventive therapies and case management of uncomplicated malaria; and 28 174 as single-course treatments of uncomplicated malaria in 76 case-management studies. There was one potentially drug-related sudden unexplained death: a healthy woman aged 16 in Mozambique who developed heart palpitations several hours after the second dose of dihydroartemisinin-piperaquine and collapsed and died on the way to hospital (no autopsy or ECG was done). The median pooled risk estimate of sudden unexplained death after dihydroartemisinin-piperaquine was 1 in 757 950 (95% CI 1 in 2 854 490 to 1 in 209 114). This risk estimate was not higher than the baseline rate of sudden cardiac death (0·7-11·9 per 100 000 person-years or 1 in 1 714 280 to 1 in 100 835 over a 30-day risk period). The risk of bias was low in most studies and unclear in a few. INTERPRETATION Dihydroartemisinin-piperaquine was associated with a low risk of sudden unexplained death that was not higher than the baseline rate of sudden cardiac death. Concerns about repolarisation-related cardiotoxicity need not limit its current use for the prevention and treatment of malaria. FUNDING Wellcome Trust, UK Medical Research Council, WHO, Bill & Melinda Gates Foundation, and University of Oxford.
Collapse
Affiliation(s)
- Xin Hui S Chan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Yan Naung Win
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Defence Services Medical Research Centre & Health and Disease Control Unit, Naypyidaw, Myanmar
| | - Laura J Mawer
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Royal Free London NHS Foundation Trust, London, UK
| | - Jireh Y Tan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Josep Brugada
- Arrhythmia Section, Cardiology Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Spain
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
49
|
Lim R, Tripura R, J Peto T, Sareth M, Sanann N, Davoeung C, Nguon C, Cheah PY. Drama as a community engagement strategy for malaria in rural Cambodia. Wellcome Open Res 2017; 2:95. [PMID: 29062919 PMCID: PMC5645711 DOI: 10.12688/wellcomeopenres.12594.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2017] [Indexed: 06/04/2024] Open
Abstract
Background: Countries in Southeast Asia are working to eliminate multidrug-resistant falciparum malaria, a major cause of mortality in tropical regions. Malaria is declining but transmission persists in many rural areas and among forest workers and isolated populations. In these remote communities, conventional health services and education are limited. Mobilising and educating these populations require new approaches as many people are illiterate and do not attend village meetings. This article describes a qualitative study to assess the feasibility of a drama project as a community engagement strategy. Methods: A drama project was conducted in twenty villages in Cambodia with three key messages: to use insecticide-treated bednets and repellents, to get early diagnosis and treatment, and to learn about risks of forest-acquired malaria. Qualitative interviews were conducted with the drama team members, village malaria workers, local health staffs and villagers, to explore the feasibility of using drama to engage the community and the associated challenges. Results: 29 people were interviewed, which included 18 semi-structured interviews and one focus group discussion. Analysis of the interviews resulted in development of the following seven themes: i) exposure to malaria and engagement activities, ii) readiness and barriers to participation, iii) understanding and learning about malaria using drama, iv) entertainment value and engagement method preferences, v) challenges to community engagement, vi) future participation and vii) sustainability. The event saw a very positive response, with an encouraging average participation rate of 66%. The project faced several challenges including logistic problems, rescheduling due to raining season, and time- and budget-constraints. Conclusions: Our evaluation demonstrated that the drama project was feasible in promoting awareness and understanding of malaria prevention and control. Audience members perceived drama as entertaining and as the preferred choice of engagement activity. Participatory drama could be considered as part of the community engagement for malaria elimination.
Collapse
Affiliation(s)
- Renly Lim
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Quality Use of Medicines and Pharmacy Research Centre, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Faculty of Medicine, University of Amsterdam, Amsterdam, Netherlands
| | - Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ma Sareth
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nou Sanann
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Chea Nguon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- The Ethox Centre, University of Oxford, Oxford, UK
| |
Collapse
|
50
|
Lim R, Tripura R, J Peto T, Sareth M, Sanann N, Davoeung C, Nguon C, Cheah PY. Drama as a community engagement strategy for malaria in rural Cambodia. Wellcome Open Res 2017; 2:95. [PMID: 29062919 PMCID: PMC5645711 DOI: 10.12688/wellcomeopenres.12594.2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2017] [Indexed: 11/23/2022] Open
Abstract
Background: Countries in Southeast Asia are working to eliminate multidrug-resistant falciparum malaria, a major cause of mortality in tropical regions. Malaria is declining but transmission persists in many rural areas and among forest workers and isolated populations. In these remote communities, conventional health services and education are limited. Mobilising and educating these populations require new approaches as many people are illiterate and do not attend village meetings. This article describes a qualitative study to assess the feasibility of a drama project as a community engagement strategy. Methods: A drama project was conducted in twenty villages in Cambodia with three key messages: to use insecticide-treated bednets and repellents, to get early diagnosis and treatment, and to learn about risks of forest-acquired malaria. Qualitative interviews were conducted with the drama team members, village malaria workers, local health staffs and villagers, to explore the feasibility of using drama to engage the community and the associated challenges. Results: 29 people were interviewed, which included 18 semi-structured interviews and one focus group discussion. Analysis of the interviews resulted in development of the following seven themes: i) exposure to malaria and engagement activities, ii) readiness and barriers to participation, iii) understanding and learning about malaria using drama, iv) entertainment value and engagement method preferences, v) challenges to community engagement, vi) future participation and vii) sustainability. The event saw a very positive response, with an encouraging average participation rate of 66%. The project faced several challenges including logistic problems, rescheduling due to raining season, and time- and budget-constraints. Conclusions: Our evaluation demonstrated that the drama project was feasible in promoting awareness and understanding of malaria prevention and control. Audience members perceived drama as entertaining and as the preferred choice of engagement activity. Participatory drama could be considered as part of the community engagement for malaria elimination.
Collapse
Affiliation(s)
- Renly Lim
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Quality Use of Medicines and Pharmacy Research Centre, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Faculty of Medicine, University of Amsterdam, Amsterdam, Netherlands
| | - Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ma Sareth
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nou Sanann
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Chea Nguon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,The Ethox Centre, University of Oxford, Oxford, UK
| |
Collapse
|