16
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The TIRS trial: protocol for a cluster randomized controlled trial assessing the efficacy of preventive targeted indoor residual spraying to reduce Aedes-borne viral illnesses in Merida, Mexico

      research-article

      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

          Current urban vector control strategies have failed to contain dengue epidemics and to prevent the global expansion of Aedes-borne viruses (ABVs: dengue, chikungunya, Zika). Part of the challenge in sustaining effective ABV control emerges from the paucity of evidence regarding the epidemiological impact of any Aedes control method. A strategy for which there is limited epidemiological evidence is targeted indoor residual spraying (TIRS). TIRS is a modification of classic malaria indoor residual spraying that accounts for Aedes aegypti resting behavior by applying residual insecticides on exposed lower sections of walls (< 1.5 m), under furniture, and on dark surfaces.

          Methods/design

          We are pursuing a two-arm, parallel, unblinded, cluster randomized controlled trial to quantify the overall efficacy of TIRS in reducing the burden of laboratory-confirmed ABV clinical disease (primary endpoint). The trial will be conducted in the city of Merida, Yucatan State, Mexico (population ~ 1million), where we will prospectively follow 4600 children aged 2–15 years at enrollment, distributed in 50 clusters of 5 × 5 city blocks each. Clusters will be randomly allocated ( n = 25 per arm) using covariate-constrained randomization. A “fried egg” design will be followed, in which all blocks of the 5 × 5 cluster receive the intervention, but all sampling to evaluate the epidemiological and entomological endpoints will occur in the “yolk,” the center 3 × 3 city blocks of each cluster. TIRS will be implemented as a preventive application (~ 1–2 months prior to the beginning of the ABV season). Active monitoring for symptomatic ABV illness will occur through weekly household visits and enhanced surveillance. Annual sero-surveys will be performed after each transmission season and entomological evaluations of Ae. aegypti indoor abundance and ABV infection rates monthly during the period of active surveillance. Epidemiological and entomological evaluation will continue for up to three transmission seasons.

          Discussion

          The findings from this study will provide robust epidemiological evidence of the efficacy of TIRS in reducing ABV illness and infection. If efficacious, TIRS could drive a paradigm shift in Aedes control by considering Ae. aegypti behavior to guide residual insecticide applications and changing deployment to preemptive control (rather than in response to symptomatic cases), two major enhancements to existing practice.

          Trial registration

          ClinicalTrials.gov NCT04343521. Registered on 13 April 2020. The protocol also complies with the WHO International Clinical Trials Registry Platform (ICTRP) (Additional file 1).

          Primary sponsor

          National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIH/NIAID).

          Related collections

          Most cited references50

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

          Dengue, Urbanization and Globalization: The Unholy Trinity of the 21st Century

          Dengue is the most important arboviral disease of humans with over half of the world’s population living in areas of risk. The frequency and magnitude of epidemic dengue have increased dramatically in the past 40 years as the viruses and the mosquito vectors have both expanded geographically in the tropical regions of the world. There are many factors that have contributed to this emergence of epidemic dengue, but only three have been the principal drivers: 1) urbanization, 2) globalization and 3) lack of effective mosquito control. The dengue viruses have fully adapted to a human-Aedes aegypti-human transmission cycle, in the large urban centers of the tropics, where crowded human populations live in intimate association with equally large mosquito populations. This setting provides the ideal home for maintenance of the viruses and the periodic generation of epidemic strains. These cities all have modern airports through which 10s of millions of passengers pass each year, providing the ideal mechanism for transportation of viruses to new cities, regions and continents where there is little or no effective mosquito control. The result is epidemic dengue. This paper discusses this unholy trinity of drivers, along with disease burden, prevention and control and prospects for the future.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Contemporary status of insecticide resistance in the major Aedes vectors of arboviruses infecting humans

            Both Aedes aegytpi and Ae. albopictus are major vectors of 5 important arboviruses (namely chikungunya virus, dengue virus, Rift Valley fever virus, yellow fever virus, and Zika virus), making these mosquitoes an important factor in the worldwide burden of infectious disease. Vector control using insecticides coupled with larval source reduction is critical to control the transmission of these viruses to humans but is threatened by the emergence of insecticide resistance. Here, we review the available evidence for the geographical distribution of insecticide resistance in these 2 major vectors worldwide and map the data collated for the 4 main classes of neurotoxic insecticide (carbamates, organochlorines, organophosphates, and pyrethroids). Emerging resistance to all 4 of these insecticide classes has been detected in the Americas, Africa, and Asia. Target-site mutations and increased insecticide detoxification have both been linked to resistance in Ae. aegypti and Ae. albopictus but more work is required to further elucidate metabolic mechanisms and develop robust diagnostic assays. Geographical distributions are provided for the mechanisms that have been shown to be important to date. Estimating insecticide resistance in unsampled locations is hampered by a lack of standardisation in the diagnostic tools used and by a lack of data in a number of regions for both resistance phenotypes and genotypes. The need for increased sampling using standard methods is critical to tackle the issue of emerging insecticide resistance threatening human health. Specifically, diagnostic doses and well-characterised susceptible strains are needed for the full range of insecticides used to control Ae. aegypti and Ae. albopictus to standardise measurement of the resistant phenotype, and calibrated diagnostic assays are needed for the major mechanisms of resistance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              House-to-house human movement drives dengue virus transmission.

              Dengue is a mosquito-borne disease of growing global health importance. Prevention efforts focus on mosquito control, with limited success. New insights into the spatiotemporal drivers of dengue dynamics are needed to design improved disease-prevention strategies. Given the restricted range of movement of the primary mosquito vector, Aedes aegypti, local human movements may be an important driver of dengue virus (DENV) amplification and spread. Using contact-site cluster investigations in a case-control design, we demonstrate that, at an individual level, risk for human infection is defined by visits to places where contact with infected mosquitoes is likely, independent of distance from the home. Our data indicate that house-to-house human movements underlie spatial patterns of DENV incidence, causing marked heterogeneity in transmission rates. At a collective level, transmission appears to be shaped by social connections because routine movements among the same places, such as the homes of family and friends, are often similar for the infected individual and their contacts. Thus, routine, house-to-house human movements do play a key role in spread of this vector-borne pathogen at fine spatial scales. This finding has important implications for dengue prevention, challenging the appropriateness of current approaches to vector control. We argue that reexamination of existing paradigms regarding the spatiotemporal dynamics of DENV and other vector-borne pathogens, especially the importance of human movement, will lead to improvements in disease prevention.
                Bookmark

                Author and article information

                Contributors
                gmvazqu@emory.edu
                Journal
                Trials
                Trials
                Trials
                BioMed Central (London )
                1745-6215
                8 October 2020
                8 October 2020
                2020
                : 21
                : 839
                Affiliations
                [1 ]GRID grid.412864.d, ISNI 0000 0001 2188 7788, Unidad Colaborativa de Bioensayos Entomológicos, Campus de Ciencias Biológicas y Agropecuarias, , Universidad Autónoma de Yucatán, ; Merida, Mexico
                [2 ]GRID grid.15276.37, ISNI 0000 0004 1936 8091, Department of Biostatistics, , University of Florida, ; Gainesville, FL 32611 USA
                [3 ]Center for Inference and Dynamics of Infectious Diseases, Seattle, WA 98109 USA
                [4 ]GRID grid.270240.3, ISNI 0000 0001 2180 1622, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, ; Seattle, WA 98109 USA
                [5 ]GRID grid.34477.33, ISNI 0000000122986657, Department of Biostatistics, , University of Washington, ; Seattle, WA 98109 USA
                [6 ]GRID grid.15276.37, ISNI 0000 0004 1936 8091, Emerging Pathogens Institute, University of Florida, ; Gainesville, FL 32611 USA
                [7 ]GRID grid.189967.8, ISNI 0000 0001 0941 6502, Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, , School of Medicine, Emory University, ; Decatur, GA 30030 USA
                [8 ]GRID grid.189967.8, ISNI 0000 0001 0941 6502, Department of Biostatistics and Bioinformatics, , Rollins School of Public Health, Emory University, ; Atlanta, GA 30322 USA
                [9 ]GRID grid.415771.1, ISNI 0000 0004 1773 4764, Health Systems Research Center, , National Institute of Public Health, ; Cuernavaca, Mexico
                [10 ]GRID grid.416738.f, ISNI 0000 0001 2163 0069, Centers for Disease Control and Prevention, ; Atlanta, GA USA
                [11 ]GRID grid.15276.37, ISNI 0000 0004 1936 8091, Department of Biology, , University of Florida, ; Gainesville, FL 32611 USA
                [12 ]GRID grid.189967.8, ISNI 0000 0001 0941 6502, Department of Environmental Sciences, Math and Science Center, , Emory University, ; 400 Dowman Drive, 5th floor, Suite E530, Atlanta, GA 30322 USA
                [13 ]Centro Nacional de Programas Preventivos y Control de Enfermedades (CENAPRECE) Secretaría de Salud Mexico, Mexico City, Mexico
                [14 ]Secretaria de Salud de Yucatan, Merida, Yucatan Mexico
                [15 ]GRID grid.412864.d, ISNI 0000 0001 2188 7788, Centro de Investigaciones Regionales Hideyo Noguchi, , Universidad Autonoma de Yucatan, ; Merida, Mexico
                Author information
                http://orcid.org/0000-0002-0710-270X
                Article
                4780
                10.1186/s13063-020-04780-7
                7542575
                33032661
                0d0f989c-7c55-4fb6-95be-9196f49171fe
                © The Author(s) 2020

                Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

                History
                : 23 July 2020
                : 29 September 2020
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000060, National Institute of Allergy and Infectious Diseases;
                Award ID: U01AI148069
                Award ID: R01 AI139761
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: U54 GM111274
                Award ID: R37 AI0032042
                Funded by: Innovative Vector Control Consortium
                Award ID: DFID:30041-105
                Categories
                Study Protocol
                Custom metadata
                © The Author(s) 2020

                Medicine
                cluster randomized,insecticide,aedes aegypti,dengue,chikungunya,zika,indoor,urban
                Medicine
                cluster randomized, insecticide, aedes aegypti, dengue, chikungunya, zika, indoor, urban

                Comments

                Comment on this article