search
Back to results

Spatial Repellents for Aedes-borne Virus Control in Sri Lanka (AEGIS ABV)

Primary Purpose

Arbovirus Infections

Status
Not yet recruiting
Phase
Not Applicable
Locations
Sri Lanka
Study Type
Interventional
Intervention
Transfluthrin
Placebo
Sponsored by
University of Notre Dame
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional prevention trial for Arbovirus Infections focused on measuring Dengue, Zika, Chikungunya, Spatial Repellent, Transfluthrin, Vector-borne diseases, Mosquito vectors, Incidence

Eligibility Criteria

6 Months - undefined (Child, Adult, Older Adult)All SexesAccepts Healthy Volunteers

LONGITUDINAL SEROCONVERSION Individual Level

Inclusion Criteria:

  • ≥ 4 - 16 years of age
  • Plans to stay in residence and/or study area for a minimum of 24 months
  • Resident of household or frequent visitor (~20% of day hours in house / month)

Exclusion Criteria:

  • < 4 and > 16 years of age
  • Plans to leave residence and/or study area within next 24 months
  • Temporary visitor to household (<20% of day hours in house/ month)

FEBRILE SURVEILLANCE Household Level

Inclusion Criteria:

  • Adult head of households agrees to census, health visits and logging resident symptoms when febrile (or in the case of suspected Zika in the absence of fever, presenting with rash, arthralgia, arthritis or non-purulent conjunctivitis).
  • Individuals spend a minimum of 4hrs per week during the daytime hours or sleep in the house.

Exclusion Criteria:

  • Adult head of households does not agree to census, health visits or logging symptoms of residents.
  • Households where study personnel identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile).
  • Sites where no residents spend time during the day (i.e. work 7d a week outside the home).

FEBRILE SURVEILLANCE Individual Level

Inclusion Criteria:

  • ≥ 6mo of age.
  • Fever at the time of presentation or report of feverishness within the previous 24 hours or presenting with a rash, arthralgia, arthritis or non-purulent conjunctivitis (suspicion of ZIKA determined by project physician)
  • Individual who spends a minimum of 4 hours per week within the household or sleeps in the house.

Exclusion Criteria:

  • < 6mo of age.
  • No fever at time of presentation or report of feverishness within the previous 24 hours or not reporting with a rash, arthralgia, arthritis or non-purulent conjunctivitis
  • Individuals who have spent less than 4 hours in the household during the week prior to illness.

ENTOMOLOGICAL MONITORING Household Level

Inclusion Criteria:

  • Adult head of household agrees to surveys.
  • Properties where study personnel do not identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile).

Exclusion Criteria:

  • Adult head of household does not agree to surveys.
  • Properties where study personnel identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile).

SPATIAL REPELLENT INTERVENTION Household Level

Inclusion Criteria:

  • Adult head of households agrees to have intervention applied inside the home and to provide access to team member at 4-week intervals to change products.
  • Properties where study personnel do not identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile).

Exclusion Criteria:

  • Adult head of household does not agree to Mosquito ShieldTM deployment or study team access.
  • Properties where study personnel identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile).

Sites / Locations

  • Epidemiology Unit, Ministry of Health
  • Clinical Trials Unit

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Placebo Comparator

Arm Label

Spatial Repellent

Placebo

Arm Description

Transfluthrin

Inert ingredients

Outcomes

Primary Outcome Measures

Incidence of Aedes-borne virus (ABV) infection in the 'longitudinal cohort'.
The primary endpoint is the fraction of monotypic or seronegative individuals in the 'longitudinal cohort' who seroconvert to an arbovirus during the follow-up period post randomization with intervention. Here, the intervention follow-up period is 2 years after initial deployment of SR or placebo. There will be 3 blood samplings from longitudinal cohort participants for measure of seroconversion: one for baseline serostatus characterization (T0), a second at 12 months (T1) and a third at 24 months (T2) from time of initial placement of intervention.

Secondary Outcome Measures

Clinically apparent cases of Aedes-borne virus (ABV) disease.
Clinically apparent is defined as an acute infection that causes overt symptoms (fever, rash, etc.) indicating virus circulation in the blood. For the longitudinal cohort participants, acute and convalescent blood sampling based on time of health facility visit when febrile throughout the intervention period. For other household members participating in febrile surveillance, case definition measured and reported whenever they visit designated health facilities throughout the intervention period.
Adult female Aedes aegypti indoor abundance.
Measured by comparing adult female Aedes aegypti indoor abundance in households using Procopak mosquito aspiration with active and placebo product receiving standard entomological surveillance and control procedures by the local Ministry of Health, as an indicator for reduced mosquito house entry due to effect of product. Indoor mosquito collections in enrolled households once every 28 days during intervention.
Adult female Aedes aegypti blood fed rate.
Measured by comparing adult female Aedes aegypti blood fed rate in households with active and placebo product receiving standard entomological surveillance and control procedures by the local Ministry of Health, as an indicator for reduced mosquito human contact due to effect of product. Direct mosquito abdominal observation by microscopy from samples taken by Procopak aspiration during indoor mosquito collections in enrolled households once every 28 days during intervention.
Diversion of Aedes aegypti mosquitoes into untreated houses.
Measured by comparing adult female Aedes aegypti abundance using Procopak mosquito aspiration in untreated households adjacent to treatment clusters (with active product) to untreated households adjacent to placebo clusters as an indicator for mosquito diversion due to effect of product. Indoor mosquito collections in enrolled households once every 28 days during intervention.
Overall incidence of Aedes-borne virus (ABV) infection.
Measured by the seroconversion rates of all children enrolled in the trial, independent of order of infection (i.e., including tertiary and quaternary infections). Based on blood samples taken for longitudinal seroconversion and febrile surveillance from time of initial placement of intervention.

Full Information

First Posted
July 6, 2022
Last Updated
March 21, 2023
Sponsor
University of Notre Dame
Collaborators
SC Johnson, A Family Company, fhiClinical, Ministry of Health, Sri Lanka, University of Sri Jayewardenepura, Sri Lanka, University of Washington, RemediumOne
search

1. Study Identification

Unique Protocol Identification Number
NCT05452447
Brief Title
Spatial Repellents for Aedes-borne Virus Control in Sri Lanka
Acronym
AEGIS ABV
Official Title
A Cluster Randomized, Placebo Control Trial to Evaluate the Efficacy of a Spatial Repellent (Mosquito ShieldTM) Against Aedes-borne Virus Infection Among Children ≥ 4-16 Years of Age in the Gampaha District, Sri Lanka
Study Type
Interventional

2. Study Status

Record Verification Date
July 2022
Overall Recruitment Status
Not yet recruiting
Study Start Date
March 2023 (Anticipated)
Primary Completion Date
February 2025 (Anticipated)
Study Completion Date
February 2025 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
University of Notre Dame
Collaborators
SC Johnson, A Family Company, fhiClinical, Ministry of Health, Sri Lanka, University of Sri Jayewardenepura, Sri Lanka, University of Washington, RemediumOne

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
The primary objective of the study is to demonstrate and quantify the protective efficacy (PE) of a single SR product, in reducing DENV infection and active Aedes-borne virus (ABV) disease in human cohorts. The study design will be a prospective, cluster randomized controlled trial (cRCT). Although not a specific objective of this project, an overall goal is to allow for official recommendations (or not) from the World Health Organization (WHO) for the use of SRs in public health. A WHO global policy recommendation will establish evaluation systems of SR products to regulate efficacy evaluations, thereby increasing quality, overall use and a consequent reduction in disease.
Detailed Description
The study will be a prospective, cRCT, participant and observer-blinded, placebo-controlled trial in a site endemic for ABV to measure the impact of a SR product on new ABV virus infections. Clusters of households, each cluster containing 110-120 residents testing negative for antibodies against DENV (seronegative) or positive to a single DENV infection (monotypic), will be selected from three MOH areas in the district of Gampaha: Negambo, Wattala, Kelaniya. All participating houses in each cluster will be monitored entomologically for adult Aedes aegypti surveys for 3 months before deployment of the SR intervention and monthly after the intervention is in place. Entomological surveys will include monitoring of indoor Ae. aegypti adult population densities and blood-fed status. DENV infection in study participants will be assessed by serologic testing of scheduled longitudinal blood samples (primary outcome) and passively by monitoring febrile persons for acute Dengue illness (secondary outcome). Seroconversion to DENV from baseline (pre-intervention) and follow-up (post-intervention) samples as well as ABV active disease rates will be compared between active intervention and placebo (control) clusters. Testing and confirmation of Zika virus (ZIKV) and Chikungunya virus (CHIKV) infection at baseline and during the intervention phase of the trial will be dependent on circulation history/detection in study area during study period. The spatial repellent (SR) will be a new formulation of transfluthrin. This active ingredient (AI) is widely used in mosquito coils and other household pest control products worldwide. The new formulation is a passive emanator that will release the AI over a period of up to four weeks, Mosquito ShieldTM. The emanator will consist of a pre-treated piece of cellulose acetate or other medium, which will be positioned within consenting households according to manufacturer specifications of 2 units/9m2. A placebo product of matched design with inert ingredients will be applied similarly. The Mosquito ShieldTM and placebo products for this study will be designed and provided by S.C. Johnson, INC. A Family Company.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Arbovirus Infections
Keywords
Dengue, Zika, Chikungunya, Spatial Repellent, Transfluthrin, Vector-borne diseases, Mosquito vectors, Incidence

7. Study Design

Primary Purpose
Prevention
Study Phase
Not Applicable
Interventional Study Model
Parallel Assignment
Masking
ParticipantInvestigator
Allocation
Randomized
Enrollment
14430 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
Spatial Repellent
Arm Type
Experimental
Arm Description
Transfluthrin
Arm Title
Placebo
Arm Type
Placebo Comparator
Arm Description
Inert ingredients
Intervention Type
Device
Intervention Name(s)
Transfluthrin
Intervention Description
Passive emanator with formulated transfluthrin
Intervention Type
Device
Intervention Name(s)
Placebo
Intervention Description
Passive emanator with formulated inert ingredients
Primary Outcome Measure Information:
Title
Incidence of Aedes-borne virus (ABV) infection in the 'longitudinal cohort'.
Description
The primary endpoint is the fraction of monotypic or seronegative individuals in the 'longitudinal cohort' who seroconvert to an arbovirus during the follow-up period post randomization with intervention. Here, the intervention follow-up period is 2 years after initial deployment of SR or placebo. There will be 3 blood samplings from longitudinal cohort participants for measure of seroconversion: one for baseline serostatus characterization (T0), a second at 12 months (T1) and a third at 24 months (T2) from time of initial placement of intervention.
Time Frame
24 months
Secondary Outcome Measure Information:
Title
Clinically apparent cases of Aedes-borne virus (ABV) disease.
Description
Clinically apparent is defined as an acute infection that causes overt symptoms (fever, rash, etc.) indicating virus circulation in the blood. For the longitudinal cohort participants, acute and convalescent blood sampling based on time of health facility visit when febrile throughout the intervention period. For other household members participating in febrile surveillance, case definition measured and reported whenever they visit designated health facilities throughout the intervention period.
Time Frame
24 months
Title
Adult female Aedes aegypti indoor abundance.
Description
Measured by comparing adult female Aedes aegypti indoor abundance in households using Procopak mosquito aspiration with active and placebo product receiving standard entomological surveillance and control procedures by the local Ministry of Health, as an indicator for reduced mosquito house entry due to effect of product. Indoor mosquito collections in enrolled households once every 28 days during intervention.
Time Frame
24 months
Title
Adult female Aedes aegypti blood fed rate.
Description
Measured by comparing adult female Aedes aegypti blood fed rate in households with active and placebo product receiving standard entomological surveillance and control procedures by the local Ministry of Health, as an indicator for reduced mosquito human contact due to effect of product. Direct mosquito abdominal observation by microscopy from samples taken by Procopak aspiration during indoor mosquito collections in enrolled households once every 28 days during intervention.
Time Frame
24 months
Title
Diversion of Aedes aegypti mosquitoes into untreated houses.
Description
Measured by comparing adult female Aedes aegypti abundance using Procopak mosquito aspiration in untreated households adjacent to treatment clusters (with active product) to untreated households adjacent to placebo clusters as an indicator for mosquito diversion due to effect of product. Indoor mosquito collections in enrolled households once every 28 days during intervention.
Time Frame
24 months
Title
Overall incidence of Aedes-borne virus (ABV) infection.
Description
Measured by the seroconversion rates of all children enrolled in the trial, independent of order of infection (i.e., including tertiary and quaternary infections). Based on blood samples taken for longitudinal seroconversion and febrile surveillance from time of initial placement of intervention.
Time Frame
24 months
Other Pre-specified Outcome Measures:
Title
Adverse Events (AEs) and Serious Adverse Events (SAEs).
Description
Measured by solicited and unsolicited reports from both the longitudinal cohort and febrile surveillance cohort during the trial period. Mean, minimum and maximum frequency and percentage of AEs and SAEs across clusters among enrolled subjects will be summarized by treatment arm.
Time Frame
24 months
Title
Incidence of Aedes-borne virus (ABV) infection in subjects residing in households within treatment clusters but without SR product.
Description
Measured by comparing Aedes-borne virus infection rates between subjects residing in households with SR product in treatment clusters and subjects from the same clusters who did not agree to the SR application in their households but are receiving standard entomological surveillance and control procedures by the local ministry of health, as an indicator of community effect due to effect of product.
Time Frame
24 months
Title
Clinically apparent cases of Aedes-borne virus (ABV) disease in subjects residing in households within treatment clusters but without SR product.
Description
Measured by comparing Aedes-borne virus disease case rates between subjects residing in households with SR product in households in treatment clusters and individuals from the same clusters who did not agree to the SR application in their households but are receiving standard entomological surveillance and control procedures by the local ministry of health, as an indicator of community effect due to effect of product.
Time Frame
24 months
Title
Adult female Aedes aegypti indoor abundance using Procopak mosquito aspiration in households within treatment clusters but without SR product.
Description
Measured by comparing adult female Aedes aegypti indoor abundance in households with SR product in treatment clusters and households from the same clusters who did not agree to the SR application but are receiving standard entomological surveillance and control procedures by the local ministry of health, as an indicator of community effect to effect of product. Indoor mosquito collections in enrolled households once every 28 days during intervention
Time Frame
24 months
Title
Adult female Aedes aegypti blood fed rate using Procopak mosquito aspiration in households within treatment clusters but without SR product.
Description
Measured by comparing adult female Aedes aegypti blood fed rate in households with SR product in treatment clusters and households from the same clusters who did not agree to the SR application but are receiving standard entomological surveillance and control procedures by the local ministry of health, as an indicator of community effect to effect of product. Samples from indoor mosquito collections in enrolled households once every 28 days during intervention.
Time Frame
24 months

10. Eligibility

Sex
All
Minimum Age & Unit of Time
6 Months
Accepts Healthy Volunteers
Accepts Healthy Volunteers
Eligibility Criteria
LONGITUDINAL SEROCONVERSION Individual Level Inclusion Criteria: ≥ 4 - 16 years of age Plans to stay in residence and/or study area for a minimum of 24 months Resident of household or frequent visitor (~20% of day hours in house / month) Exclusion Criteria: < 4 and > 16 years of age Plans to leave residence and/or study area within next 24 months Temporary visitor to household (<20% of day hours in house/ month) FEBRILE SURVEILLANCE Household Level Inclusion Criteria: Adult head of households agrees to census, health visits and logging resident symptoms when febrile (or in the case of suspected Zika in the absence of fever, presenting with rash, arthralgia, arthritis or non-purulent conjunctivitis). Individuals spend a minimum of 4hrs per week during the daytime hours or sleep in the house. Exclusion Criteria: Adult head of households does not agree to census, health visits or logging symptoms of residents. Households where study personnel identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile). Sites where no residents spend time during the day (i.e. work 7d a week outside the home). FEBRILE SURVEILLANCE Individual Level Inclusion Criteria: ≥ 6mo of age. Fever at the time of presentation or report of feverishness within the previous 24 hours or presenting with a rash, arthralgia, arthritis or non-purulent conjunctivitis (suspicion of ZIKA determined by project physician) Individual who spends a minimum of 4 hours per week within the household or sleeps in the house. Exclusion Criteria: < 6mo of age. No fever at time of presentation or report of feverishness within the previous 24 hours or not reporting with a rash, arthralgia, arthritis or non-purulent conjunctivitis Individuals who have spent less than 4 hours in the household during the week prior to illness. ENTOMOLOGICAL MONITORING Household Level Inclusion Criteria: Adult head of household agrees to surveys. Properties where study personnel do not identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile). Exclusion Criteria: Adult head of household does not agree to surveys. Properties where study personnel identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile). SPATIAL REPELLENT INTERVENTION Household Level Inclusion Criteria: Adult head of households agrees to have intervention applied inside the home and to provide access to team member at 4-week intervals to change products. Properties where study personnel do not identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile). Exclusion Criteria: Adult head of household does not agree to Mosquito ShieldTM deployment or study team access. Properties where study personnel identify a security risk (i.e., site where drugs are sold, residents are always drunk or hostile).
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
John P Grieco, Ph.D.
Phone
5743617572
Email
jgrieco@nd.edu
First Name & Middle Initial & Last Name or Official Title & Degree
Nicole L Achee, Ph.D.
Phone
5746311561
Email
nachee@nd.edu
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
John P Grieco, Ph.D.
Organizational Affiliation
University of Notre Dame
Official's Role
Study Director
First Name & Middle Initial & Last Name & Degree
Korelege Hasitha Aravinda Tissera, M.D.
Organizational Affiliation
Epidemiology Unit, Ministry of Health, Sri Lanka
Official's Role
Principal Investigator
Facility Information:
Facility Name
Epidemiology Unit, Ministry of Health
City
Colombo
State/Province
West
ZIP/Postal Code
00100
Country
Sri Lanka
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Hasitha Tisseara, Dr.
Email
dr_korelege@yahoo.co.uk
Facility Name
Clinical Trials Unit
City
Ragama
State/Province
West
ZIP/Postal Code
01010
Country
Sri Lanka
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Asita de Silva, Prof.
Email
asita@kln.ac.lk

12. IPD Sharing Statement

Plan to Share IPD
Yes
IPD Sharing Plan Description
Analytical datasets will be anonymized and GPS tag-blurred to remove sensitive information prior to sharing.
IPD Sharing Time Frame
The data and supporting information will be made available 12 months following completion of data analysis and will remain open access in the public domain.
IPD Sharing Access Criteria
Open-access repository distributed under the terms of the Creative Commons Attribution (CC-BY) License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Citations:
PubMed Identifier
22583679
Citation
Achee NL, Bangs MJ, Farlow R, Killeen GF, Lindsay S, Logan JG, Moore SJ, Rowland M, Sweeney K, Torr SJ, Zwiebel LJ, Grieco JP. Spatial repellents: from discovery and development to evidence-based validation. Malar J. 2012 May 14;11:164. doi: 10.1186/1475-2875-11-164.
Results Reference
background
PubMed Identifier
19653497
Citation
Achee NL, Sardelis MR, Dusfour I, Chauhan KR, Grieco JP. Characterization of spatial repellent, contact irritant, and toxicant chemical actions of standard vector control compounds. J Am Mosq Control Assoc. 2009 Jun;25(2):156-67. doi: 10.2987/08-5831.1.
Results Reference
background
PubMed Identifier
8395596
Citation
Ansari MZ, Shope RE, Malik S. Evaluation of vero cell lysate antigen for the ELISA of flaviviruses. J Clin Lab Anal. 1993;7(4):230-7. doi: 10.1002/jcla.1860070408.
Results Reference
background
PubMed Identifier
18788903
Citation
Comach G, Blair PJ, Sierra G, Guzman D, Soler M, de Quintana MC, Bracho-Labadie M, Camacho D, Russell KL, Olson JG, Kochel TJ. Dengue virus infections in a cohort of schoolchildren from Maracay, Venezuela: a 2-year prospective study. Vector Borne Zoonotic Dis. 2009 Feb;9(1):87-92. doi: 10.1089/vbz.2007.0213. Epub 2008 Sep 12.
Results Reference
background
PubMed Identifier
19891873
Citation
Forshey BM, Morrison AC, Cruz C, Rocha C, Vilcarromero S, Guevara C, Camacho DE, Alava A, Madrid C, Beingolea L, Suarez V, Comach G, Kochel TJ. Dengue virus serotype 4, northeastern Peru, 2008. Emerg Infect Dis. 2009 Nov;15(11):1815-8. doi: 10.3201/eid1511.090663. Erratum In: Emerg Infect Dis. 2010 Jan;16(1):177.
Results Reference
background
PubMed Identifier
18816269
Citation
Erlanger TE, Keiser J, Utzinger J. Effect of dengue vector control interventions on entomological parameters in developing countries: a systematic review and meta-analysis. Med Vet Entomol. 2008 Sep;22(3):203-21. doi: 10.1111/j.1365-2915.2008.00740.x.
Results Reference
background
PubMed Identifier
14695086
Citation
Getis A, Morrison AC, Gray K, Scott TW. Characteristics of the spatial pattern of the dengue vector, Aedes aegypti, in Iquitos, Peru. Am J Trop Med Hyg. 2003 Nov;69(5):494-505.
Results Reference
background
PubMed Identifier
17684562
Citation
Grieco JP, Achee NL, Chareonviriyaphap T, Suwonkerd W, Chauhan K, Sardelis MR, Roberts DR. A new classification system for the actions of IRS chemicals traditionally used for malaria control. PLoS One. 2007 Aug 8;2(8):e716. doi: 10.1371/journal.pone.0000716.
Results Reference
background
PubMed Identifier
16506566
Citation
Grieco JP, Achee NL, Sardelis MR, Chauhan KR, Roberts DR. A novel high-throughput screening system to evaluate the behavioral response of adult mosquitoes to chemicals. J Am Mosq Control Assoc. 2005 Dec;21(4):404-11. doi: 10.2987/8756-971X(2006)21[404:ANHSST]2.0.CO;2.
Results Reference
background
PubMed Identifier
10925798
Citation
Grieco JP, Achee NL, Andre RG, Roberts DR. A comparison study of house entering and exiting behavior of Anopheles vestitipennis (Diptera: Culicidae) using experimental huts sprayed with DDT or deltamethrin in the southern district of Toledo, Belize, C.A. J Vector Ecol. 2000 Jun;25(1):62-73.
Results Reference
background
PubMed Identifier
2472746
Citation
Gubler DJ. Aedes aegypti and Aedes aegypti-borne disease control in the 1990s: top down or bottom up. Charles Franklin Craig Lecture. Am J Trop Med Hyg. 1989 Jun;40(6):571-8. doi: 10.4269/ajtmh.1989.40.571. No abstract available.
Results Reference
background
PubMed Identifier
9665979
Citation
Gubler DJ. Dengue and dengue hemorrhagic fever. Clin Microbiol Rev. 1998 Jul;11(3):480-96. doi: 10.1128/CMR.11.3.480.
Results Reference
background
PubMed Identifier
11827812
Citation
Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century. Trends Microbiol. 2002 Feb;10(2):100-3. doi: 10.1016/s0966-842x(01)02288-0.
Results Reference
background
PubMed Identifier
18982804
Citation
Hapuarachchi HA, Bandara KB, Hapugoda MD, Williams S, Abeyewickreme W. Laboratory confirmation of dengue and chikungunya co-infection. Ceylon Med J. 2008 Sep;53(3):104-5. doi: 10.4038/cmj.v53i3.252. No abstract available.
Results Reference
background
PubMed Identifier
19955565
Citation
Hapuarachchi HC, Bandara KB, Sumanadasa SD, Hapugoda MD, Lai YL, Lee KS, Tan LK, Lin RT, Ng LF, Bucht G, Abeyewickreme W, Ng LC. Re-emergence of Chikungunya virus in South-east Asia: virological evidence from Sri Lanka and Singapore. J Gen Virol. 2010 Apr;91(Pt 4):1067-76. doi: 10.1099/vir.0.015743-0. Epub 2009 Dec 2.
Results Reference
background
PubMed Identifier
8916809
Citation
Hayes CG, Phillips IA, Callahan JD, Griebenow WF, Hyams KC, Wu SJ, Watts DM. The epidemiology of dengue virus infection among urban, jungle, and rural populations in the Amazon region of Peru. Am J Trop Med Hyg. 1996 Oct;55(4):459-63. doi: 10.4269/ajtmh.1996.55.459.
Results Reference
background
PubMed Identifier
2540664
Citation
Innis BL, Nisalak A, Nimmannitya S, Kusalerdchariya S, Chongswasdi V, Suntayakorn S, Puttisri P, Hoke CH. An enzyme-linked immunosorbent assay to characterize dengue infections where dengue and Japanese encephalitis co-circulate. Am J Trop Med Hyg. 1989 Apr;40(4):418-27. doi: 10.4269/ajtmh.1989.40.418.
Results Reference
background
PubMed Identifier
19193262
Citation
Kanakaratne N, Wahala WM, Messer WB, Tissera HA, Shahani A, Abeysinghe N, de-Silva AM, Gunasekera M. Severe dengue epidemics in Sri Lanka, 2003-2006. Emerg Infect Dis. 2009 Feb;15(2):192-9. doi: 10.3201/eid1502.080926.
Results Reference
background
PubMed Identifier
15532930
Citation
Kawada H, Maekawa Y, Tsuda Y, Takagi M. Laboratory and field evaluation of spatial repellency with metofluthrin-impregnated paper strip against mosquitoes in Lombok Island, Indonesia. J Am Mosq Control Assoc. 2004 Sep;20(3):292-8.
Results Reference
background
PubMed Identifier
15669387
Citation
Kawada H, Maekawa Y, Tsuda Y, Takagi M. Trial of spatial repellency of metofluthrin-impregnated paper strip against Anopheles and Culex in shelters without walls in Lombok, Indonesia. J Am Mosq Control Assoc. 2004 Dec;20(4):434-7. Erratum In: J Am Mosq Control Assoc. 2005 Mar;21(1):105.
Results Reference
background
PubMed Identifier
16599150
Citation
Kawada H, Maekawa Y, Takagi M. Field trial on the spatial repellency of metofluthrin-impregnated plastic strips for mosquitoes in shelters without walls (beruga) in Lombok, Indonesia. J Vector Ecol. 2005 Dec;30(2):181-5.
Results Reference
background
PubMed Identifier
12147378
Citation
Kochel TJ, Watts DM, Halstead SB, Hayes CG, Espinoza A, Felices V, Caceda R, Bautista CT, Montoya Y, Douglas S, Russell KL. Effect of dengue-1 antibodies on American dengue-2 viral infection and dengue haemorrhagic fever. Lancet. 2002 Jul 27;360(9329):310-2. doi: 10.1016/S0140-6736(02)09522-3.
Results Reference
background
PubMed Identifier
19581242
Citation
Kularatne SA, Gihan MC, Weerasinghe SC, Gunasena S. Concurrent outbreaks of Chikungunya and Dengue fever in Kandy, Sri Lanka, 2006-07: a comparative analysis of clinical and laboratory features. Postgrad Med J. 2009 Jul;85(1005):342-6. doi: 10.1136/pgmj.2007.066746.
Results Reference
background
PubMed Identifier
8654514
Citation
Kuno G. Review of the factors modulating dengue transmission. Epidemiol Rev. 1995;17(2):321-35. doi: 10.1093/oxfordjournals.epirev.a036196. No abstract available.
Results Reference
background
PubMed Identifier
1372617
Citation
Lanciotti RS, Calisher CH, Gubler DJ, Chang GJ, Vorndam AV. Rapid detection and typing of dengue viruses from clinical samples by using reverse transcriptase-polymerase chain reaction. J Clin Microbiol. 1992 Mar;30(3):545-51. doi: 10.1128/jcm.30.3.545-551.1992.
Results Reference
background
PubMed Identifier
17536367
Citation
Lucas JR, Shono Y, Iwasaki T, Ishiwatari T, Spero N, Benzon G. U.S. laboratory and field trials of metofluthrin (SumiOne) emanators for reducing mosquito biting outdoors. J Am Mosq Control Assoc. 2007 Mar;23(1):47-54. doi: 10.2987/8756-971X(2007)23[47:ULAFTO]2.0.CO;2.
Results Reference
background
PubMed Identifier
12224589
Citation
Messer WB, Vitarana UT, Sivananthan K, Elvtigala J, Preethimala LD, Ramesh R, Withana N, Gubler DJ, De Silva AM. Epidemiology of dengue in Sri Lanka before and after the emergence of epidemic dengue hemorrhagic fever. Am J Trop Med Hyg. 2002 Jun;66(6):765-73. doi: 10.4269/ajtmh.2002.66.765.
Results Reference
background
PubMed Identifier
8146129
Citation
Monath TP. Dengue: the risk to developed and developing countries. Proc Natl Acad Sci U S A. 1994 Mar 29;91(7):2395-400. doi: 10.1073/pnas.91.7.2395.
Results Reference
background
PubMed Identifier
4031038
Citation
Morens DM, Halstead SB, Repik PM, Putvatana R, Raybourne N. Simplified plaque reduction neutralization assay for dengue viruses by semimicro methods in BHK-21 cells: comparison of the BHK suspension test with standard plaque reduction neutralization. J Clin Microbiol. 1985 Aug;22(2):250-4. doi: 10.1128/jcm.22.2.250-254.1985.
Results Reference
background
PubMed Identifier
20454609
Citation
Morrison AC, Minnick SL, Rocha C, Forshey BM, Stoddard ST, Getis A, Focks DA, Russell KL, Olson JG, Blair PJ, Watts DM, Sihuincha M, Scott TW, Kochel TJ. Epidemiology of dengue virus in Iquitos, Peru 1999 to 2005: interepidemic and epidemic patterns of transmission. PLoS Negl Trop Dis. 2010 May 4;4(5):e670. doi: 10.1371/journal.pntd.0000670.
Results Reference
background
PubMed Identifier
18351798
Citation
Morrison AC, Zielinski-Gutierrez E, Scott TW, Rosenberg R. Defining challenges and proposing solutions for control of the virus vector Aedes aegypti. PLoS Med. 2008 Mar 18;5(3):e68. doi: 10.1371/journal.pmed.0050068.
Results Reference
background
PubMed Identifier
15185957
Citation
Morrison AC, Astete H, Chapilliquen F, Ramirez-Prada C, Diaz G, Getis A, Gray K, Scott TW. Evaluation of a sampling methodology for rapid assessment of Aedes aegypti infestation levels in Iquitos, Peru. J Med Entomol. 2004 May;41(3):502-10. doi: 10.1603/0022-2585-41.3.502.
Results Reference
background
PubMed Identifier
15605653
Citation
Morrison AC, Gray K, Getis A, Astete H, Sihuincha M, Focks D, Watts D, Stancil JD, Olson JG, Blair P, Scott TW. Temporal and geographic patterns of Aedes aegypti (Diptera: Culicidae) production in Iquitos, Peru. J Med Entomol. 2004 Nov;41(6):1123-42. doi: 10.1603/0022-2585-41.6.1123.
Results Reference
background
PubMed Identifier
5984948
Citation
Munasinghe DR, Amarasekera PJ, Fernando CF. An epidemic of dengue-like fever in Ceylon (chikungunya--a clinical and haematological study. Ceylon Med J. 1966 Dec;11(4):129-42. No abstract available.
Results Reference
background
PubMed Identifier
23216844
Citation
Ogoma SB, Moore SJ, Maia MF. A systematic review of mosquito coils and passive emanators: defining recommendations for spatial repellency testing methodologies. Parasit Vectors. 2012 Dec 7;5:287. doi: 10.1186/1756-3305-5-287.
Results Reference
background
PubMed Identifier
25311699
Citation
Syafruddin D, Bangs MJ, Sidik D, Elyazar I, Asih PB, Chan K, Nurleila S, Nixon C, Hendarto J, Wahid I, Ishak H, Bogh C, Grieco JP, Achee NL, Baird JK. Impact of a spatial repellent on malaria incidence in two villages in Sumba, Indonesia. Am J Trop Med Hyg. 2014 Dec;91(6):1079-87. doi: 10.4269/ajtmh.13-0735. Epub 2014 Oct 13.
Results Reference
background

Learn more about this trial

Spatial Repellents for Aedes-borne Virus Control in Sri Lanka

We'll reach out to this number within 24 hrs