35
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The epidemiology of subclinical malaria infections in South-East Asia: findings from cross-sectional surveys in Thailand–Myanmar border areas, Cambodia, and Vietnam

      research-article
      , , , , , , , , , , , , , , , , , , , , , , , , , , , , ,
      Malaria Journal
      BioMed Central
      Malaria, P. falciparum, P. vivax, Sub-microscopic, Epidemiology, South-East Asia, Myanmar, Thailand, Cambodia, Vietnam, Greater Mekong Sub-region

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          The importance of the submicroscopic reservoir of Plasmodium infections for malaria elimination depends on its size, which is generally considered small in low transmission settings. The precise estimation of this reservoir requires more sensitive parasite detection methods. The prevalence of asymptomatic, sub-microscopic malaria was assessed by a sensitive, high blood volume quantitative real-time polymerase chain reaction method in three countries of the Greater Mekong Sub-region.

          Methods

          Cross-sectional surveys were conducted in three villages in western Cambodia, four villages along the Thailand–Myanmar border and four villages in southwest Vietnam. Malaria parasitaemia was assessed by Plasmodium falciparum/pan malaria rapid diagnostic tests (RDTs), microscopy and a high volume ultra-sensitive real-time polymerase chain reaction (HVUSqPCR: limit of detection 22 parasites/mL). All villagers older than 6 months were invited to participate.

          Results

          A census before the surveys identified 7355 residents in the study villages. Parasite prevalence was 224/5008 (4 %) by RDT, 229/5111 (5 %) by microscopy, and 988/4975 (20 %) when assessed by HVUSqPCR. Of these 164 (3 %) were infected with P. falciparum, 357 (7 %) with Plasmodium vivax, 56 (1 %) with a mixed infection, and 411 (8 %) had parasite densities that were too low for species identification. A history of fever, male sex, and age of 15 years or older were independently associated with parasitaemia in a multivariate regression model stratified by site.

          Conclusion

          Light microscopy and RDTs identified only a quarter of all parasitaemic participants. The asymptomatic Plasmodium reservoir is considerable, even in low transmission settings. Novel strategies are needed to eliminate this previously under recognized reservoir of malaria transmission.

          Electronic supplementary material

          The online version of this article (doi:10.1186/s12936-015-0906-x) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references18

          • Record: found
          • Abstract: found
          • Article: not found

          Epidemiology and infectivity of Plasmodium falciparum and Plasmodium vivax gametocytes in relation to malaria control and elimination.

          Malaria remains a major cause of morbidity and mortality in the tropics, with Plasmodium falciparum responsible for the majority of the disease burden and P. vivax being the geographically most widely distributed cause of malaria. Gametocytes are the sexual-stage parasites that infect Anopheles mosquitoes and mediate the onward transmission of the disease. Gametocytes are poorly studied despite this crucial role, but with a recent resurgence of interest in malaria elimination, the study of gametocytes is in vogue. This review highlights the current state of knowledge with regard to the development and longevity of P. falciparum and P. vivax gametocytes in the human host and the factors influencing their distribution within endemic populations. The evidence for immune responses, antimalarial drugs, and drug resistance influencing infectiousness to mosquitoes is reviewed. We discuss how the application of molecular techniques has led to the identification of submicroscopic gametocyte carriage and to a reassessment of the human infectious reservoir. These components are drawn together to show how control measures that aim to reduce malaria transmission, such as mass drug administration and a transmission-blocking vaccine, might better be deployed.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A review of malaria diagnostic tools: microscopy and rapid diagnostic test (RDT).

            The absolute necessity for rational therapy in the face of rampant drug resistance places increasing importance on the accuracy of malaria diagnosis. Giemsa microscopy and rapid diagnostic tests (RDTs) represent the two diagnostics most likely to have the largest impact on malaria control today. These two methods, each with characteristic strengths and limitations, together represent the best hope for accurate diagnosis as a key component of successful malaria control. This review addresses the quality issues with current malaria diagnostics and presents data from recent rapid diagnostic test trials. Reduction of malaria morbidity and drug resistance intensity plus the associated economic loss of these two factors require urgent scaling up of the quality of parasite-based diagnostic methods. An investment in anti-malarial drug development or malaria vaccine development should be accompanied by a parallel commitment to improve diagnostic tools and their availability to people living in malarious areas.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Microsatellite markers reveal a spectrum of population structures in the malaria parasite Plasmodium falciparum.

              Multilocus genotyping of microbial pathogens has revealed a range of population structures, with some bacteria showing extensive recombination and others showing almost complete clonality. The population structure of the protozoan parasite Plasmodium falciparum has been harder to evaluate, since most studies have used a limited number of antigen-encoding loci that are known to be under strong selection. We describe length variation at 12 microsatellite loci in 465 infections collected from 9 locations worldwide. These data reveal dramatic differences in parasite population structure in different locations. Strong linkage disequilibrium (LD) was observed in six of nine populations. Significant LD occurred in all locations with prevalence <1% and in only two of five of the populations from regions with higher transmission intensities. Where present, LD results largely from the presence of identical multilocus genotypes within populations, suggesting high levels of self-fertilization in populations with low levels of transmission. We also observed dramatic variation in diversity and geographical differentiation in different regions. Mean heterozygosities in South American countries (0.3-0.4) were less than half those observed in African locations (0. 76-0.8), with intermediate heterozygosities in the Southeast Asia/Pacific samples (0.51-0.65). Furthermore, variation was distributed among locations in South America (F:(ST) = 0.364) and within locations in Africa (F:(ST) = 0.007). The intraspecific patterns of diversity and genetic differentiation observed in P. falciparum are strikingly similar to those seen in interspecific comparisons of plants and animals with differing levels of outcrossing, suggesting that similar processes may be involved. The differences observed may also reflect the recent colonization of non-African populations from an African source, and the relative influences of epidemiology and population history are difficult to disentangle. These data reveal a range of population structures within a single pathogen species and suggest intimate links between patterns of epidemiology and genetic structure in this organism.
                Bookmark

                Author and article information

                Contributors
                noi@tropmedres.ac
                nhienntt@oucru.org
                Rupam@tropmedres.ac
                Tom@tropmedres.ac
                Sue@tropmedres.ac
                drkhin_mg_lwin@shoklo-unit.com
                Preyanan@tropmedres.ac
                Atthanee@tropmedres.ac
                Benchawan@tropmedres.ac
                klanarongw@afrims.org
                huedvdr@gmail.com
                lethanhdong@gmail.com
                uyennt@oucru.org
                yoel@tropmedres.ac
                lorenz@tropmedres.ac
                mehul.dhorda@wwarn.org
                cholrawee.promnarate@wwarn.org
                georges.snounou@upmc.fr
                Benoit_Malleret@immunol.a-star.edu.sg
                laurentrenia@gmail.com
                lily@shoklo-unit.com
                pratap.sin@mahidol.ac.th
                Pasathorn@tropmedres.ac
                Jem@tropmedres.ac
                cheanguoncnm@gmail.com
                hientt@oucru.org
                Nickd@tropmedres.ac
                nickwdt@tropmedres.ac
                arjen@tropmedres.ac
                francois@tropmedres.ac
                Journal
                Malar J
                Malar. J
                Malaria Journal
                BioMed Central (London )
                1475-2875
                30 September 2015
                30 September 2015
                2015
                : 14
                : 381
                Affiliations
                [ ]Mahidol Oxford 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
                [ ]Oxford University Clinical Research Unit, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam
                [ ]Shoklo Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Tak, Thailand
                [ ]Center for Malariology, Parasitology and Entomology Control, Phan Rang-Thap Cham, Ninh Thuan Province Vietnam
                [ ]Institute of Malariology-Parasitology, Entomology (IMPE) of Ho Chi Minh City, 699 Tran Hung Dao Q5, Ho Chi Minh City, Vietnam
                [ ]Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Churchill Hospital, Oxford, UK
                [ ]WWARN Asia Regional Centre, Mahidol University, Bangkok, Thailand
                [ ]Sorbonne Universités, UPMC Univ Paris 06, UPMC UMRS CR7, 75005 Paris, France
                [ ]Centre d’Immunologie et de Maladies Infectieuses (CIMI), Paris, Institut National de la Santé et de la Recherche Médicale (Inserm) U1135, Centre National de la Recherche Scientifique (CNRS) ERL 8255, 75013 Paris, France
                [ ]Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, 278177 Singapore
                [ ]Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 5 Science Drive 2, Blk MD4, Level 3, Singapore, 117597 Singapore
                [ ]Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
                [ ]National Center for Parasitology, Entomology and Malaria Control, No. 372, Preah Monivong, Phnom Penh, 12302 Cambodia
                [ ]Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
                Article
                906
                10.1186/s12936-015-0906-x
                4590703
                26424000
                7a37539d-ac76-4b47-97a6-5a8501b867fb
                © Imwong et al. 2015

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 10 July 2015
                : 5 September 2015
                Categories
                Research
                Custom metadata
                © The Author(s) 2015

                Infectious disease & Microbiology
                malaria,p. falciparum,p. vivax,sub-microscopic,epidemiology,south-east asia,myanmar,thailand,cambodia,vietnam,greater mekong sub-region

                Comments

                Comment on this article