1
|
Maluleke T, Benecke R, Oladejo S, Feulner G, Kern S, Lister J, McClelland G, Njoki M, Noack P, Petruccione F, Rajaratnam K, Waitt C, Rosenkranz B, Pillai G. Cross-disciplinary mathematical modelling to benefit healthcare - could clinical pharmacology play an enabling role? Br J Clin Pharmacol 2024; 90:2509-2516. [PMID: 39082394 DOI: 10.1111/bcp.16192] [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: 02/07/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 09/28/2024] Open
Abstract
Clinical pharmacology is often the nexus in any cross-disciplinary team that is seeking solutions for human healthcare issues. The use and application of real-world data and artificial intelligence to better understand our ecosystem has influenced our view at the world and how we do things. This has resulted in remarkable advancements in the healthcare space and development of personalized medicines with great attributes from the application of models and simulations, contributing to a more efficient healthcare development process. A cross-disciplinary symposium was held in December 2023, whereby experts from different scientific disciplines engaged in a high-level discussion on the opportunities and challenges of mathematical models in different fields, possible future developments and decision making. A strong interlink amongst the disciplines represented was apparent, with clinical pharmacology identified as the one which integrates various scientific disciplines. Deliberate and strategic cross-disciplinary dialogues are required to break out of the silos and implement integration for efficiency and cost-effectiveness of novel interventions.
Collapse
Affiliation(s)
- Tirhani Maluleke
- F. Hoffmann La-Roche, Johannesburg, South Africa
- Fundisa African Academy of Medicines Development, Cape Town, South Africa
| | - Rohan Benecke
- Division of Clinical Pharmacology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sunday Oladejo
- School for Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Georg Feulner
- Potsdam Institute for Climate Impact Research, Member of the Leibniz Association, Potsdam, Germany
| | - Steve Kern
- Global Health Labs, Bellevue, Bellevue, Washington, USA
| | | | | | - Miriam Njoki
- Science For Africa (SFA) Foundation, Nairobi, Kenya
| | - Patrick Noack
- University of Applied Sciences, Weihenstephan-Triesdorf, Germany
| | - Francesco Petruccione
- School for Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
- National Institute for Theoretical and Computational Sciences (NITheCS), Stellenbosch, South Africa
| | - Kanshukan Rajaratnam
- School for Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Catriona Waitt
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Bernd Rosenkranz
- Fundisa African Academy of Medicines Development, Cape Town, South Africa
- Division of Clinical Pharmacology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Institute for Clinical Pharmacology and Toxicology, Charité Universitätsmedizin, Berlin, Germany
| | - Goonaseela Pillai
- CP+ Associates GmbH, Basel, Switzerland
- Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
- Pharmacometrics Africa NPC, Cape Town, South Africa
| |
Collapse
|
2
|
Pooda SH, Hien DFDS, Pagabeleguem S, Heinrich AP, Porciani A, Sagna AB, Zela L, Percoma L, Lefèvre T, Dabiré RK, Koffi AA, Düring RA, Pennetier C, Moiroux N, Mouline K. Impact of blood meals taken on ivermectin-treated livestock on survival and egg production of the malaria vector Anopheles coluzzii under laboratory conditions. PLoS One 2024; 19:e0308293. [PMID: 39146278 PMCID: PMC11326554 DOI: 10.1371/journal.pone.0308293] [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: 11/24/2023] [Accepted: 07/20/2024] [Indexed: 08/17/2024] Open
Abstract
Treatment of livestock with endectocides such as ivermectin is viewed as a complementary vector control approach to address residual transmission of malaria. However, efficacy of this treatment may vary between animal species. Hence, our purpose was to investigate the effects of ivermectin treatments of common livestock species on life history traits of the opportunistic malaria vector Anopheles coluzzii. Sheep, goats and pigs were treated using injectable veterinary ivermectin formulation at the species-specific doses (recommended dose for all species and high dose in pig). Mosquito batches were exposed to treated and control (not injected) animals at different days after treatment. Daily mosquito mortality was recorded and fecundity assessed through the count of gravid females and the number of eggs they developed. The recommended dose of ivermectin induced a significant decrease in mosquito survival for up to 7 days after injection (DAI), with a decrease of 89.7%, 66.7%, and 48.4% in treated pigs, goats and sheep, respectively, compared to control animals. In treated pigs, the triple therapeutic dose decreased mosquito survival of 68.97% relatively to controls up to 14 DAI. The average number in gravid females Anopheles that survived after feeding on treated animals were reduced when blood-meals were taken on sheep (2.57% and 42.03% at 2 and 7 DAI), or on goats (decrease of the 28.28% and 73.64% respectively at 2 and 7 DAI). This study shows that ivermectin treatments to animals negatively impacts An. coluzzii life history traits and could reduce vector densities in areas where livestock live near humans. However, due to short-term efficacy of single dose treatments, repeated treatments and potentially increased dosages would be required to span the transmission season. The use of long-acting ivermectin formulations is discussed as a mean for extending efficacy while remaining cost effective.
Collapse
Affiliation(s)
- Sié Hermann Pooda
- Université de Dédougou, Dédougou (UDDG), Dédougou, Burkina Faso
- Insectarium de Bobo-Dioulasso-Campagne d'Eradication de la Mouche Tsé-tsé et de la Tryapnosomose (IBD-CETT), Bobo-Dioulasso, Burkina Faso
| | | | - Soumaïla Pagabeleguem
- Université de Dédougou, Dédougou (UDDG), Dédougou, Burkina Faso
- Insectarium de Bobo-Dioulasso-Campagne d'Eradication de la Mouche Tsé-tsé et de la Tryapnosomose (IBD-CETT), Bobo-Dioulasso, Burkina Faso
| | - Andre Patrick Heinrich
- Institute of Soil Science and Soil Conservation, Research Center for Biosystems, Land Use and Nutrition (iFZ), Justus Liebig University Giessen, Giessen, Germany
| | - Angélique Porciani
- Unité Mixte sur les Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), Université de Montpellier, IRD, CNRS, Montpellier, France
| | - André Barembaye Sagna
- Unité Mixte sur les Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Lamidi Zela
- Centre International de Recherche-Développement sur l'Elevage en zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
| | - Lassane Percoma
- Insectarium de Bobo-Dioulasso-Campagne d'Eradication de la Mouche Tsé-tsé et de la Tryapnosomose (IBD-CETT), Bobo-Dioulasso, Burkina Faso
| | - Thierry Lefèvre
- Unité Mixte sur les Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Roch Kounbobr Dabiré
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso, Burkina Faso
| | | | - Rolf-Alexander Düring
- Institute of Soil Science and Soil Conservation, Research Center for Biosystems, Land Use and Nutrition (iFZ), Justus Liebig University Giessen, Giessen, Germany
| | - Cédric Pennetier
- Unité Mixte sur les Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Nicolas Moiroux
- Unité Mixte sur les Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Karine Mouline
- Unité Mixte sur les Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), Université de Montpellier, IRD, CNRS, Montpellier, France
| |
Collapse
|
3
|
Cramer EY, Nguyen XQ, Hertz JC, Nguyen DV, Quang HH, Mendenhall IH, Lover AA. Measuring effects of ivermectin-treated cattle on potential malaria vectors in Vietnam: A cluster-randomized trial. PLoS Negl Trop Dis 2024; 18:e0012014. [PMID: 38683855 PMCID: PMC11098492 DOI: 10.1371/journal.pntd.0012014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/16/2024] [Accepted: 02/19/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Malaria elimination using current tools has stalled in many areas. Ivermectin (IVM) is a broad-antiparasitic drug and mosquitocide and has been proposed as a tool for accelerating progress towards malaria elimination. Under laboratory conditions, IVM has been shown to reduce the survival of adult Anopheles populations that have fed on IVM-treated mammals. Treating cattle with IVM has been proposed as an important contribution to malaria vector management, however, the impacts of IVM in this One Health use case have been untested in field trials in Southeast Asia. METHODS Through a randomized village-based trial, this study quantified the effect of IVM-treated cattle on anopheline populations in treated vs. untreated villages in Central Vietnam. Local zebu cattle in six rural villages were included in this study. In three villages, cattle were treated with IVM at established veterinary dosages, and in three additional villages cattle were left as untreated controls. For the main study outcome, the mosquito populations in all villages were sampled using cattle-baited traps for six nights before, and six nights after a 2-day IVM-administration (intervention) period. Anopheline species were characterized using taxonomic keys. The impact of the intervention was analyzed using a difference-in-differences (DID) approach with generalized estimating equations (with negative binomial distribution and robust errors). This intervention was powered to detect a 50% reduction in total nightly Anopheles spp. vector catches from cattle-baited traps. Given the unusual diversity in anopheline populations, exploratory analyses examined taxon-level differences in the ecological population diversity. RESULTS Across the treated villages, 1,112 of 1,523 censused cows (73% overall; range 67% to 83%) were treated with IVM. In both control and treated villages, there was a 30% to 40% decrease in total anophelines captured in the post-intervention period as compared to the pre-intervention period. In the control villages, there were 1,873 captured pre-intervention and 1,079 captured during the post-intervention period. In the treated villages, there were 1,594 captured pre-intervention, and 1,101 captured during the post-intervention period. The difference in differences model analysis comparing total captures between arms was not statistically significant (p = 0.61). Secondary outcomes of vector population diversity found that in three villages (one control and two treatment) Brillouin's index increased, and in three villages (two control and one treatment) Brillouin's index decreased. When examining biodiversity by trapping-night, there were no clear trends in treated or untreated vector populations. Additionally, there were no clear trends when examining the components of biodiversity: richness and evenness. CONCLUSIONS The ability of this study to quantify the impacts of IVM treatment was limited due to unexpectedly large spatiotemporal variability in trapping rates; an area-wide decrease in trapping counts across all six villages post-intervention; and potential spillover effects. However, this study provides important data to directly inform future studies in the GMS and beyond for IVM-based vector control.
Collapse
Affiliation(s)
- Estee Y. Cramer
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts-Amherst, Massachusetts, United States of America
| | - Xuan Quang Nguyen
- Institute of Malariology-Parasitology & Entomology Quy Nhon, Ministry of Health, Vietnam
| | | | - Do Van Nguyen
- Institute of Malariology-Parasitology & Entomology Quy Nhon, Ministry of Health, Vietnam
| | - Huynh Hong Quang
- Institute of Malariology-Parasitology & Entomology Quy Nhon, Ministry of Health, Vietnam
| | - Ian H. Mendenhall
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Andrew A. Lover
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts-Amherst, Massachusetts, United States of America
| |
Collapse
|
4
|
Kositz C, Vasileva H, Mohammed N, Achan J, Dabira ED, D'Alessandro U, Bradley J, Marks M. Risk factors for non-participation in ivermectin and dihydroartemisinin-piperaquine mass drug administration for malaria control in the MASSIV trial. Malar J 2024; 23:54. [PMID: 38383367 PMCID: PMC10882911 DOI: 10.1186/s12936-024-04878-2] [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: 11/12/2023] [Accepted: 02/10/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Mass Drug Administration (MDA) has become a mainstay for the control of several diseases over the last two decades. Successful implementation of MDA programmes requires community participation and can be threatened by systematic non-participation. Such concerns are particularly pertinent for MDA programmes against malaria, as they require multi-day treatment over several consecutive months. Factors associated with non-participation to the MDA campaign with ivermectin (IVM) and dihydroartemisinin-piperaquine (DHP) implemented within the MASSIV cluster randomized trial were determined. METHODS Coverage data was extracted from the MASSIV trial study database, with every datapoint being a directly observed therapy (DOT). A complete month of MDA was classified as receiving all three daily doses of treatment. For both ivermectin and DHP, ordinal logistic regression was used to identify individual and household level variables associated with non-participation. RESULTS For ivermectin, 51.5% of eligible participants received all 3 months of treatment while 30.7% received either one or two complete months. For DHP, 56.7% of eligible participants received all 3 months of treatment and 30.5% received either one or two complete months. Children aged 5-15 years and adults aged more than 50 years were more likely to receive at least one complete month of MDA than working age adults, both for ivermectin (aOR 4.3, 95% CI 3.51-5.28 and aOR of 2.26, 95% CI 1.75-2.95) and DHP (aOR 2.47, 95%CI 2.02-3.02 and aOR 1.33, 95%CI 1.01-1.35), respectively. Members of households where the head received a complete month of MDA were more likely to themselves have received a complete month of MDA, both for ivermectin (aOR 1.71, 95%CI 1.35-2.14) and for DHP (aOR 1.64, 95%CI 1.33-2.04). CONCLUSION Personal and household-level variables were associated with participation in the MDA programme for malaria control. Specific strategies to (increase participation amongst some groups may be important to ensure maximum impact of MDA strategies in achieving malaria elimination. TRIAL REGISTRATION The MASSIV trial is registered under NCT03576313.
Collapse
Affiliation(s)
- Christian Kositz
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, UK.
| | - Hristina Vasileva
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, UK
| | - Nuredin Mohammed
- Disease Control and Elimination, Medical Research Council Unit Gambia at the London School of Hygiene and Tropical Medicine (MRCG at LSHTM), Fajara, The Gambia
| | - Jane Achan
- Disease Control and Elimination, Medical Research Council Unit Gambia at the London School of Hygiene and Tropical Medicine (MRCG at LSHTM), Fajara, The Gambia
| | - Edgard Diniba Dabira
- Disease Control and Elimination, Medical Research Council Unit Gambia at the London School of Hygiene and Tropical Medicine (MRCG at LSHTM), Fajara, The Gambia
| | - Umberto D'Alessandro
- Disease Control and Elimination, Medical Research Council Unit Gambia at the London School of Hygiene and Tropical Medicine (MRCG at LSHTM), Fajara, The Gambia
| | - John Bradley
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London, UK
| | - Michael Marks
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, UK
- Hospital for Tropical Diseases, University College London Hospital, London, UK
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
5
|
Zeleke G, Duchateau L, Yewhalaw D, Suleman S, Devreese M. In-vitro susceptibility and ex-vivo evaluation of macrocyclic lactone endectocides sub-lethal concentrations against Plasmodium vivax oocyst development in Anopheles arabiensis. Malar J 2024; 23:26. [PMID: 38238768 PMCID: PMC10797976 DOI: 10.1186/s12936-024-04845-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/05/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Asymptomatic malaria transmission has become a public health concern across malaria-endemic Africa including Ethiopia. Specifically, Plasmodium vivax is more efficient at transmitting earlier in the infection and at lower densities than Plasmodium falciparum. Consequently, a greater proportion of individuals infected with P. vivax can transmit without detectable gametocytaemia. Mass treatment of livestock with macrocyclic lactones (MLs), e.g., ivermectin and doramectin, was suggested as a complementary malaria vector tool because of their insecticidal effects. However, the effects of MLs on P. vivax in Anopheles arabiensis has not yet been fully explored. Hence, comparative in-vitro susceptibility and ex-vivo studies were conducted to evaluate the effects of ivermectin, doramectin and moxidectin sub-lethal concentrations on P. vivax oocyst development in An. arabiensis. METHODS The 7-day sub-lethal concentrations of 25% (LC25) and 5% (LC5) were determined from in-vitro susceptibility tests on female An. arabiensis in Hemotek® membrane feeding assay. Next, an ex-vivo study was conducted using P. vivax gametocytes infected patient's blood spiked with the LC25 and LC5 of the MLs. At 7-days post-feeding, each mosquito was dissected under a dissection stereo microscope, stained with 0.5% (w/v) mercurochrome solution, and examined for the presence of P. vivax oocysts. Statistical analysis was based on a generalized mixed model with binomially distributed error terms. RESULTS A 7-day lethal concentration of 25% (LC25, in ng/mL) of 7.1 (95% CI: [6.3;8.0]), 20.0 (95%CI:[17.8;22.5]) and 794.3 (95%CI:[716.4;1516.3]) were obtained for ivermectin, doramectin and moxidectin, respectively. Similarly, a lethal concentration of 5% (LC5, in ng/mL) of 0.6 (95% CI: [0.5;0.7]), 1.8 (95% CI:[1.6;2.0]) and 53.7 (95% CI:[ 48.4;102.5]) were obtained respectively for ivermectin, doramectin and moxidectin. The oocyst prevalence in treatment and control groups did not differ significantly (p > 0.05) from each other. Therefore, no direct effect of ML endectocides on P. vivax infection in An. arabiensis mosquitoes was observed at the sub-lethal concentration (LC25 and LC5). CONCLUSIONS The effects of ivermectin and doramectin on malaria parasite is more likely via indirect effects, particularly by reducing the vectors lifespan and causing mortality before completing the parasite's sporogony cycle or reducing their vector capacity as it affects the locomotor activity of the mosquito.
Collapse
Affiliation(s)
- Gemechu Zeleke
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke, Belgium
- Jimma University Laboratory of Drug Quality (JuLaDQ), and School of Pharmacy, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Luc Duchateau
- Biometrics Research Center, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke, Belgium
| | - Delenasaw Yewhalaw
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Sultan Suleman
- Jimma University Laboratory of Drug Quality (JuLaDQ), and School of Pharmacy, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Mathias Devreese
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke, Belgium.
| |
Collapse
|
6
|
Sagna AB, Zéla L, Ouedraogo COW, Pooda SH, Porciani A, Furnival-Adams J, Lado P, Somé AF, Pennetier C, Chaccour CJ, Dabiré RK, Mouline K. Ivermectin as a novel malaria control tool: Getting ahead of the resistance curse. Acta Trop 2023; 245:106973. [PMID: 37352998 DOI: 10.1016/j.actatropica.2023.106973] [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: 04/07/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
Reduction in malaria clinical cases is strongly dependent on the ability to prevent Anopheles infectious bites. Vector control strategies using long-lasting insecticidal nets and indoor residual spraying with insecticides have contributed to significantly reduce the incidence of malaria in many endemic countries, especially in the Sub-Saharan region. However, global progress in reducing malaria cases has plateaued since 2015 mostly due to the increased insecticide resistance and behavioral changes in Anopheles vectors. Additional control strategies are thus required to further reduce the burden of malaria and contain the spread of resistant and invasive Anopheles vectors. The use of endectocides such as ivermectin as an additional malaria control tool is now receiving increased attention, driven by its different mode of action compared to insecticides used so far and its excellent safety record for humans. In this opinion article, we discuss the advantages and disadvantages of using ivermectin for malaria control with a focus on the risk of selecting ivermectin resistance in malaria vectors. We also highlight the importance of understanding how ivermectin resistance could develop in mosquitoes and what its underlying mechanisms and associated molecular markers are, and propose a research agenda to manage this phenomenon.
Collapse
Affiliation(s)
- André B Sagna
- MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France.
| | - Lamidi Zéla
- Centre International de Recherche-Développement sur l'Elevage en zone Subhumide, Bobo-Dioulasso, Burkina Faso
| | - Cheick Oumar W Ouedraogo
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l'Ouest, Centre National de Recherche Scientifique et Technologique, Bobo-Dioulasso, Burkina Faso
| | - Sié H Pooda
- Centre International de Recherche-Développement sur l'Elevage en zone Subhumide, Bobo-Dioulasso, Burkina Faso; Université de Dédougou, Dédougou, Burkina Faso
| | | | | | - Paula Lado
- Center for Vector-borne Infectious Diseases, Colorado State University, Fort Collins, CO, USA
| | - Anyirékun F Somé
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l'Ouest, Centre National de Recherche Scientifique et Technologique, Bobo-Dioulasso, Burkina Faso
| | - Cédric Pennetier
- MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France
| | - Carlos J Chaccour
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Madrid, Spain; Universidad de Navarra, Pamplona, Spain
| | - Roch K Dabiré
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l'Ouest, Centre National de Recherche Scientifique et Technologique, Bobo-Dioulasso, Burkina Faso
| | - Karine Mouline
- MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France
| |
Collapse
|
7
|
Hutchins H, Bradley J, Pretorius E, Teixeira da Silva E, Vasileva H, Jones RT, Ndiath MO, Dit Massire Soumare H, Mabey D, Nante EJ, Martins C, Logan JG, Slater H, Drakeley C, D'Alessandro U, Rodrigues A, Last AR. Protocol for a cluster randomised placebo-controlled trial of adjunctive ivermectin mass drug administration for malaria control on the Bijagós Archipelago of Guinea-Bissau: the MATAMAL trial. BMJ Open 2023; 13:e072347. [PMID: 37419638 PMCID: PMC10335573 DOI: 10.1136/bmjopen-2023-072347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
INTRODUCTION As malaria declines, innovative tools are required to further reduce transmission and achieve elimination. Mass drug administration (MDA) of artemisinin-based combination therapy (ACT) is capable of reducing malaria transmission where coverage of control interventions is already high, though the impact is short-lived. Combining ACT with ivermectin, an oral endectocide shown to reduce vector survival, may increase its impact, while also treating ivermectin-sensitive co-endemic diseases and minimising the potential impact of ACT resistance in this context. METHODS AND ANALYSIS MATAMAL is a cluster-randomised placebo-controlled trial. The trial is being conducted in 24 clusters on the Bijagós Archipelago, Guinea-Bissau, where the peak prevalence of Plasmodium falciparum (Pf) parasitaemia is approximately 15%. Clusters have been randomly allocated to receive MDA with dihydroartemisinin-piperaquine and either ivermectin or placebo. The primary objective is to determine whether the addition of ivermectin MDA is more effective than dihydroartemisinin-piperaquine MDA alone in reducing the prevalence of P. falciparum parasitaemia, measured during peak transmission season after 2 years of seasonal MDA. Secondary objectives include assessing prevalence after 1 year of MDA; malaria incidence monitored through active and passive surveillance; age-adjusted prevalence of serological markers indicating exposure to P. falciparum and anopheline mosquitoes; vector parous rates, species composition, population density and sporozoite rates; prevalence of vector pyrethroid resistance; prevalence of artemisinin resistance in P. falciparum using genomic markers; ivermectin's impact on co-endemic diseases; coverage estimates; and the safety of combined MDA. ETHICS AND DISSEMINATION The trial has been approved by the London School of Hygiene and Tropical Medicine's Ethics Committee (UK) (19156) and the Comite Nacional de Eticas de Saude (Guinea-Bissau) (084/CNES/INASA/2020). Results will be disseminated in peer-reviewed publications and in discussion with the Bissau-Guinean Ministry of Public Health and participating communities. TRIAL REGISTRATION NUMBER NCT04844905.
Collapse
Affiliation(s)
- Harry Hutchins
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK
| | - John Bradley
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Elizabeth Pretorius
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
| | - Eunice Teixeira da Silva
- Projecto de Saúde Bandim, Bissau, Guinea-Bissau
- Ministério de Saúde Pública, Bissau, Guinea-Bissau
| | - Hristina Vasileva
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Robert T Jones
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
| | | | | | - David Mabey
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Ernesto Jose Nante
- Programa Nacional de Luta Contra o Paludismo, Ministério de Saúde, Bissau, Guinea-Bissau
| | | | - James G Logan
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
- Arctech Innovation, London, UK
| | | | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Amabelia Rodrigues
- Projecto de Saúde Bandim, Bissau, Guinea-Bissau
- Ministério de Saúde Pública, Bissau, Guinea-Bissau
| | - Anna R Last
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
8
|
Anwar MN, Hickson RI, Mehra S, Price DJ, McCaw JM, Flegg MB, Flegg JA. Optimal Interruption of P. vivax Malaria Transmission Using Mass Drug Administration. Bull Math Biol 2023; 85:43. [PMID: 37076740 PMCID: PMC10115738 DOI: 10.1007/s11538-023-01153-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/30/2023] [Indexed: 04/21/2023]
Abstract
Plasmodium vivax is the most geographically widespread malaria-causing parasite resulting in significant associated global morbidity and mortality. One of the factors driving this widespread phenomenon is the ability of the parasites to remain dormant in the liver. Known as 'hypnozoites', they reside in the liver following an initial exposure, before activating later to cause further infections, referred to as 'relapses'. As around 79-96% of infections are attributed to relapses from activating hypnozoites, we expect it will be highly impactful to apply treatment to target the hypnozoite reservoir (i.e. the collection of dormant parasites) to eliminate P. vivax. Treatment with radical cure, for example tafenoquine or primaquine, to target the hypnozoite reservoir is a potential tool to control and/or eliminate P. vivax. We have developed a deterministic multiscale mathematical model as a system of integro-differential equations that captures the complex dynamics of P. vivax hypnozoites and the effect of hypnozoite relapse on disease transmission. Here, we use our multiscale model to study the anticipated effect of radical cure treatment administered via a mass drug administration (MDA) program. We implement multiple rounds of MDA with a fixed interval between rounds, starting from different steady-state disease prevalences. We then construct an optimisation model with three different objective functions motivated on a public health basis to obtain the optimal MDA interval. We also incorporate mosquito seasonality in our model to study its effect on the optimal treatment regime. We find that the effect of MDA interventions is temporary and depends on the pre-intervention disease prevalence (and choice of model parameters) as well as the number of MDA rounds under consideration. The optimal interval between MDA rounds also depends on the objective (combinations of expected intervention outcomes). We find radical cure alone may not be enough to lead to P. vivax elimination under our mathematical model (and choice of model parameters) since the prevalence of infection eventually returns to pre-MDA levels.
Collapse
Affiliation(s)
- Md Nurul Anwar
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
- Department of Mathematics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Roslyn I Hickson
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
- Australian Institute of Tropical Health and Medicine, and College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia
- CSIRO, Townsville, Australia
| | - Somya Mehra
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
| | - David J Price
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - James M McCaw
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Mark B Flegg
- School of Mathematics, Monash University, Melbourne, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
9
|
Chaccour C, Casellas A, Hammann F, Ruiz-Castillo P, Nicolas P, Montaña J, Mael M, Selvaraj P, Duthaler U, Mrema S, Kakolwa M, Lyimo I, Okumu F, Marathe A, Schürch R, Elobolobo E, Sacoor C, Saute F, Xia K, Jones C, Rist C, Maia M, Rabinovich NR. BOHEMIA: Broad One Health Endectocide-based Malaria Intervention in Africa-a phase III cluster-randomized, open-label, clinical trial to study the safety and efficacy of ivermectin mass drug administration to reduce malaria transmission in two African settings. Trials 2023; 24:128. [PMID: 36810194 PMCID: PMC9942013 DOI: 10.1186/s13063-023-07098-2] [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: 11/19/2022] [Accepted: 01/17/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Residual malaria transmission is the result of adaptive mosquito behavior that allows malaria vectors to thrive and sustain transmission in the presence of good access to bed nets or insecticide residual spraying. These behaviors include crepuscular and outdoor feeding as well as intermittent feeding upon livestock. Ivermectin is a broadly used antiparasitic drug that kills mosquitoes feeding on a treated subject for a dose-dependent period. Mass drug administration with ivermectin has been proposed as a complementary strategy to reduce malaria transmission. METHODS A cluster randomized, parallel arm, superiority trial conducted in two settings with distinct eco-epidemiological conditions in East and Southern Africa. There will be three groups: human intervention, consisting of a dose of ivermectin (400 mcg/kg) administered monthly for 3 months to all the eligible population in the cluster (>15 kg, non-pregnant and no medical contraindication); human and livestock intervention, consisting human treatment as above plus treatment of livestock in the area with a single dose of injectable ivermectin (200 mcg/kg) monthly for 3 months; and controls, consisting of a dose of albendazole (400 mg) monthly for 3 months. The main outcome measure will be malaria incidence in a cohort of children under five living in the core of each cluster followed prospectively with monthly RDTs DISCUSSION: The second site for the implementation of this protocol has changed from Tanzania to Kenya. This summary presents the Mozambique-specific protocol while the updated master protocol and the adapted Kenya-specific protocol undergo national approval in Kenya. BOHEMIA will be the first large-scale trial evaluating the impact of ivermectin-only mass drug administration to humans or humans and cattle on local malaria transmission TRIAL REGISTRATION: ClinicalTrials.gov NCT04966702 . Registered on July 19, 2021. Pan African Clinical Trials Registry PACTR202106695877303.
Collapse
Affiliation(s)
- Carlos Chaccour
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- Universidda de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Madrid, Spain
| | - Aina Casellas
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Felix Hammann
- University Hospital of Bern, Inselspital, Bern, Switzerland
| | | | - Patricia Nicolas
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Julia Montaña
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Mary Mael
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | | | | | | | | | - Issa Lyimo
- Ifakara Health Institute, Ifakara, Tanzania
| | | | | | - Roger Schürch
- Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Eldo Elobolobo
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | | | - Francisco Saute
- Centro de Investigação em Saúde de Manhiça, Manhica, Mozambique
| | - Kang Xia
- Virginia Polytechnic Institute and State University, Blacksburg, USA
| | | | - Cassidy Rist
- Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Marta Maia
- KEMRI Wellcome Trust Research Programme, Kilifi, Kenya
| | - N. Regina Rabinovich
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- TH Chan Harvard School of Public Health, Boston, USA
| |
Collapse
|
10
|
Sturm A, Vos MW, Henderson R, Eldering M, Koolen KMJ, Sheshachalam A, Favia G, Samby K, Herreros E, Dechering KJ. Barcoded Asaia bacteria enable mosquito in vivo screens and identify novel systemic insecticides and inhibitors of malaria transmission. PLoS Biol 2021; 19:e3001426. [PMID: 34928952 PMCID: PMC8726507 DOI: 10.1371/journal.pbio.3001426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/04/2022] [Accepted: 12/03/2021] [Indexed: 11/18/2022] Open
Abstract
This work addresses the need for new chemical matter in product development for control of pest insects and vector-borne diseases. We present a barcoding strategy that enables phenotypic screens of blood-feeding insects against small molecules in microtiter plate-based arrays and apply this to discovery of novel systemic insecticides and compounds that block malaria parasite development in the mosquito vector. Encoding of the blood meals was achieved through recombinant DNA-tagged Asaia bacteria that successfully colonised Aedes and Anopheles mosquitoes. An arrayed screen of a collection of pesticides showed that chemical classes of avermectins, phenylpyrazoles, and neonicotinoids were enriched for compounds with systemic adulticide activity against Anopheles. Using a luminescent Plasmodium falciparum reporter strain, barcoded screens identified 48 drug-like transmission-blocking compounds from a 400-compound antimicrobial library. The approach significantly increases the throughput in phenotypic screening campaigns using adult insects and identifies novel candidate small molecules for disease control. This study presents a barcoding strategy that enables high-throughput phenotypic screens of blood-feeding insects against small molecules in microtiter plate-based arrays and applies this to the discovery of novel systemic insecticides and compounds that block malaria parasite development in the mosquito vector.
Collapse
|
11
|
Reiker T, Golumbeanu M, Shattock A, Burgert L, Smith TA, Filippi S, Cameron E, Penny MA. Emulator-based Bayesian optimization for efficient multi-objective calibration of an individual-based model of malaria. Nat Commun 2021; 12:7212. [PMID: 34893600 PMCID: PMC8664949 DOI: 10.1038/s41467-021-27486-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 11/18/2021] [Indexed: 11/21/2022] Open
Abstract
Individual-based models have become important tools in the global battle against infectious diseases, yet model complexity can make calibration to biological and epidemiological data challenging. We propose using a Bayesian optimization framework employing Gaussian process or machine learning emulator functions to calibrate a complex malaria transmission simulator. We demonstrate our approach by optimizing over a high-dimensional parameter space with respect to a portfolio of multiple fitting objectives built from datasets capturing the natural history of malaria transmission and disease progression. Our approach quickly outperforms previous calibrations, yielding an improved final goodness of fit. Per-objective parameter importance and sensitivity diagnostics provided by our approach offer epidemiological insights and enhance trust in predictions through greater interpretability.
Collapse
Affiliation(s)
- Theresa Reiker
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Monica Golumbeanu
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Andrew Shattock
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Lydia Burgert
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Thomas A Smith
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Ewan Cameron
- Malaria Atlas Project, Big Data Institute, University of Oxford, Oxford, UK
- Curtin University, Perth, Australia
- Telethon Kids Institute, Perth Children's Hospital, Perth, Australia
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
12
|
Marathe A, Shi R, Mendez-Lopez A, Hu Z, Lewis B, Rabinovich R, Chaccour CJ, Rist C. Potential impact of 5 years of ivermectin mass drug administration on malaria outcomes in high burden countries. BMJ Glob Health 2021; 6:bmjgh-2021-006424. [PMID: 34764146 PMCID: PMC8587489 DOI: 10.1136/bmjgh-2021-006424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/18/2021] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION The global progress against malaria has slowed significantly since 2017. As the current malaria control tools seem insufficient to get the trend back on track, several clinical trials are investigating ivermectin mass drug administration (iMDA) as a potential additional vector control tool; however, the health impacts and cost-effectiveness of this new strategy remain unclear. METHODS We developed an analytical tool based on a full factorial experimental design to assess the potential impact of iMDA in nine high burden sub-Saharan African countries. The simulated iMDA regimen was assumed to be delivered monthly to the targeted population for 3 months each year from 2023 to 2027. A broad set of parameters of ivermectin efficacy, uptake levels and global intervention scenarios were used to predict averted malaria cases and deaths. We then explored the potential averted treatment costs, expected implementation costs and cost-effectiveness ratios under different scenarios. RESULTS In the scenario where coverage of malaria interventions was maintained at 2018 levels, we found that iMDA in these nine countries has the potential to reverse the predicted growth of malaria burden by averting 20-50 million cases and 36 000-90 000 deaths with an assumed efficacy of 20%. If iMDA has an efficacy of 40%, we predict between 40-99 million cases and 73 000-179 000 deaths will be averted with an estimated net cost per case averted between US$2 and US$7, and net cost per death averted between US$1460 and US$4374. CONCLUSION This study measures the potential of iMDA to reverse the increasing number of malaria cases for several sub-Saharan African countries. With additional efficacy information from ongoing clinical trials and country-level modifications, our analytical tool can help determine the appropriate uptake strategies of iMDA by calculating potential marginal gains and costs under different scenarios.
Collapse
Affiliation(s)
- Achla Marathe
- Network Systems Science and Advanced Computing Division, Biocomplexity Institute, University of Virginia, Charlottesville, Virginia, USA.,Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Ruoding Shi
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Ana Mendez-Lopez
- Department of Preventive Medicine, Public Health and Microbiology, Autonomous University of Madrid, Madrid, Spain.,Barcelona Institute for Global Health, ISGlobal, Barcelona, Spain
| | - Zhihao Hu
- Department of Statistics, Virginia Tech, Blacksburg, Virginia, USA
| | - Bryan Lewis
- Network Systems Science and Advanced Computing Division, Biocomplexity Institute, University of Virginia, Charlottesville, Virginia, USA
| | | | - Carlos J Chaccour
- Barcelona Institute for Global Health, ISGlobal, Barcelona, Spain.,Universitat de Navarra, Pamplona, Spain
| | - Cassidy Rist
- Department of Population Health Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
13
|
Broekhuizen H, Fehr A, Nieto-Sanchez C, Muela J, Peeters-Grietens K, Smekens T, Kalleh M, Rijndertse E, Achan J, D'Alessandro U. Costs and barriers faced by households seeking malaria treatment in the Upper River Region, The Gambia. Malar J 2021; 20:368. [PMID: 34530823 PMCID: PMC8447575 DOI: 10.1186/s12936-021-03898-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 08/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria transmission in The Gambia decreased substantially over the last 20 years thanks to the scale-up of control interventions. However, malaria prevalence is still relatively high in eastern Gambia and represents both a health and a financial burden for households. This study aims to quantify the out-of-pocket costs and productivity losses of seeking malaria treatment at household level. METHODS A household survey was carried out through in-person interviews. Respondents were asked about malaria prevention methods, their treatment-seeking behaviour, and any costs incurred for transport, services, food, and/or overnight stays. A bottom-up costing approach was used to calculate the unit cost of treatment and a tobit regression approach to investigate cost drivers. RESULTS The survey included 864 respondents, mainly subsistence farmers. Most respondents (87%) considered malaria to be a problem affecting their ability to perform their regular duties. Respondents preferred going to a health facility for treatment. The primary reason for not going was related to costs; 70% of respondents incurred costs for seeking health care, with a median of £3.62 (IQR: £1.73 to £6.10). The primary driver of cost was living in one of the villages that are off the main road and/or far from health facilities. 66% reported productivity loss of 5 working days on average during a malaria episode of them or their child. CONCLUSIONS Although malaria prevalence is decreasing and treatment is provided free of charge, households seeking treatment are confronted with out-of-pocket expenditures and lost working days; particularly in remote villages.
Collapse
Affiliation(s)
- Henk Broekhuizen
- Dept. Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands. .,Dept. Health and Society, Wageningen University and Research, Wageningen, The Netherlands.
| | - Alexandra Fehr
- Department of Sociology and Anthropology, Faculty of Social and Behavioural Science, University of Amsterdam, Amsterdam, The Netherlands.,Medical Anthropology Unit, Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Claudia Nieto-Sanchez
- Medical Anthropology Unit, Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Koen Peeters-Grietens
- Medical Anthropology Unit, Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium.,School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Tom Smekens
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Esmé Rijndertse
- Dept. Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jane Achan
- MRC The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia.,Malaria Research Consortium, London, UK
| | - Umberto D'Alessandro
- MRC The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
14
|
Wamaket N, Khamprapa O, Chainarin S, Thamsawet P, Ninsaeng U, Thongsalee S, Suwan V, Sakolvaree J, Takhampunya R, Davidson SA, McCardle PW, Sa-Angchai P, Mukaka M, Kiattibutr K, Khamsiriwatchara A, Nguitragool W, Sattabongkot J, Sirichaisinthop J, Kobylinski KC. Anopheles bionomics in a malaria endemic area of southern Thailand. Parasit Vectors 2021; 14:378. [PMID: 34315509 PMCID: PMC8317318 DOI: 10.1186/s13071-021-04870-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/02/2021] [Indexed: 12/26/2022] Open
Abstract
Background Ivermectin mass drug administration (MDA) could accelerate malaria elimination in the Greater Mekong Subregion. This study was performed to characterize the bionomics of Anopheles in Surat Thani province, Thailand. Methods Mosquitoes were collected via human landing collections between February and October 2019. Anopheles mosquitoes were morphologically identified to species. Primary Anopheles malaria vectors were dissected to assess parity status, and a subset were evaluated for molecular identification and Plasmodium detection. Results A total of 17,348 mosquitoes were collected during the study period; of these, 5777 were Anopheles mosquitoes. Morphological studies identified 15 Anopheles species, of which the most abundant were Anopheles minimus (s.l.) (87.16%, n = 5035), An. dirus s.l. (7.05%, n = 407) and An. barbirostris s.l. (2.86%, n = 165). Molecular identification confirmed that of the An. minimus s.l. mosquitoes collected, 99.80% were An. minimus (s.s.) (n = 484) and 0.2% were An. aconitus (n = 1), of the An. dirus (s.l.) collected, 100% were An. baimaii (n = 348), and of the An. maculatus (s.l.) collected, 93.62% were An. maculatus (s.s.) (n = 44) and 6.38% were An. sawadwongporni (n = 3). No Anopheles mosquito tested was Plasmodium positive (0/879). An average of 11.46 Anopheles were captured per collector per night. There were differences between species in hour of collection (Kruskal–Wallis H-test: χ2 = 80.89, P < 0.0001, n = 5666), with more An. barbirostris (s.l.) and An. maculatus (s.l.) caught earlier compared to An. minimus (s.l.) (P = 0.0001 and P < 0.0001, respectively) and An. dirus (s.l.) (P = 0.0082 and P < 0.001, respectively). The proportion of parous An. minimus (s.l.) captured by hour increased throughout the night (Wald Chi-square: χ2 = 17.31, P = 0.000, odds ratio = 1.0535, 95% confidence interval 1.0279–1.0796, n = 3400). Overall, An. minimus (s.l.) parity was 67.68% (2375/3509) with an intra-cluster correlation of 0.0378. A power calculation determined that an An. minimus (s.l.) parity reduction treatment effect size = 34%, with four clusters per treatment arm and a minimum of 300 mosquitoes dissected per cluster, at an α = 0.05, will provide 82% power to detect a significant difference following ivermectin MDA. Conclusions The study area in Surat Thani province is an ideal location to evaluate the impact of ivermectin MDA on An. minimus parity. Graphical abstract ![]()
Collapse
Affiliation(s)
- Narenrit Wamaket
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand.,Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Oranicha Khamprapa
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand.,Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Sittinont Chainarin
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand.,Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Panisa Thamsawet
- Surat Thani Vector-Borne Diseases Control Center 11.3, Muang, Surat Thani, Thailand
| | - Ubolrat Ninsaeng
- Surat Thani Vector-Borne Diseases Control Center 11.3, Muang, Surat Thani, Thailand
| | - Suttipong Thongsalee
- Surat Thani Vector-Borne Diseases Control Center 11.3, Muang, Surat Thani, Thailand
| | - Veerast Suwan
- Surat Thani Vector-Borne Diseases Control Center 11.3, Muang, Surat Thani, Thailand
| | - Jira Sakolvaree
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand
| | - Ratree Takhampunya
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand
| | - Silas A Davidson
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand
| | - Patrick W McCardle
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand
| | - Patiwat Sa-Angchai
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Mavuto Mukaka
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kirakorn Kiattibutr
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Amnat Khamsiriwatchara
- Center of Excellence for Biomedical and Public Health Informatics, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Wang Nguitragool
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand.,Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | | | - Kevin C Kobylinski
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Ratchathewi, Bangkok, Thailand.
| |
Collapse
|
15
|
A systematic review and an individual patient data meta-analysis of ivermectin use in children weighing less than fifteen kilograms: Is it time to reconsider the current contraindication? PLoS Negl Trop Dis 2021; 15:e0009144. [PMID: 33730099 PMCID: PMC7968658 DOI: 10.1371/journal.pntd.0009144] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/13/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Oral ivermectin is a safe broad spectrum anthelminthic used for treating several neglected tropical diseases (NTDs). Currently, ivermectin use is contraindicated in children weighing less than 15 kg, restricting access to this drug for the treatment of NTDs. Here we provide an updated systematic review of the literature and we conducted an individual-level patient data (IPD) meta-analysis describing the safety of ivermectin in children weighing less than 15 kg. METHODOLOGY/PRINCIPAL FINDINGS A systematic review was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) for IPD guidelines by searching MEDLINE via PubMed, Web of Science, Ovid Embase, LILACS, Cochrane Database of Systematic Reviews, TOXLINE for all clinical trials, case series, case reports, and database entries for reports on the use of ivermectin in children weighing less than 15 kg that were published between 1 January 1980 to 25 October 2019. The protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO): CRD42017056515. A total of 3,730 publications were identified, 97 were selected for potential inclusion, but only 17 sources describing 15 studies met the minimum criteria which consisted of known weights of children less than 15 kg linked to possible adverse events, and provided comprehensive IPD. A total of 1,088 children weighing less than 15 kg were administered oral ivermectin for one of the following indications: scabies, mass drug administration for scabies control, crusted scabies, cutaneous larva migrans, myiasis, pthiriasis, strongyloidiasis, trichuriasis, and parasitic disease of unknown origin. Overall a total of 1.4% (15/1,088) of children experienced 18 adverse events all of which were mild and self-limiting. No serious adverse events were reported. CONCLUSIONS/SIGNIFICANCE Existing limited data suggest that oral ivermectin in children weighing less than 15 kilograms is safe. Data from well-designed clinical trials are needed to provide further assurance.
Collapse
|
16
|
Duthaler U, Weber M, Hofer L, Chaccour C, Maia M, Müller P, Krähenbühl S, Hammann F. The pharmacokinetics and drug-drug interactions of ivermectin in Aedes aegypti mosquitoes. PLoS Pathog 2021; 17:e1009382. [PMID: 33730100 PMCID: PMC7968666 DOI: 10.1371/journal.ppat.1009382] [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] [Received: 09/18/2020] [Accepted: 02/15/2021] [Indexed: 11/29/2022] Open
Abstract
Mosquitoes are vectors of major diseases such as dengue fever and malaria. Mass drug administration of endectocides to humans and livestock is a promising complementary approach to current insecticide-based vector control measures. The aim of this study was to establish an insect model for pharmacokinetic and drug-drug interaction studies to develop sustainable endectocides for vector control. Female Aedes aegypti mosquitoes were fed with human blood containing either ivermectin alone or ivermectin in combination with ketoconazole, rifampicin, ritonavir, or piperonyl butoxide. Drug concentrations were quantified by LC-MS/MS at selected time points post-feeding. Primary pharmacokinetic parameters and extent of drug-drug interactions were calculated by pharmacometric modelling. Lastly, the drug effect of the treatments was examined. The mosquitoes could be dosed with a high precision (%CV: ≤13.4%) over a range of 0.01–1 μg/ml ivermectin without showing saturation (R2: 0.99). The kinetics of ivermectin were characterised by an initial lag phase of 18.5 h (CI90%: 17.0–19.8 h) followed by a slow zero-order elimination rate of 5.5 pg/h (CI90%: 5.1–5.9 pg/h). By contrast, ketoconazole, ritonavir, and piperonyl butoxide were immediately excreted following first order elimination, whereas rifampicin accumulated over days in the mosquitoes. Ritonavir increased the lag phase of ivermectin by 11.4 h (CI90%: 8.7–14.2 h) resulting in an increased exposure (+29%) and an enhanced mosquitocidal effect. In summary, this study shows that the pharmacokinetics of drugs can be investigated and modulated in an Ae. aegypti animal model. This may help in the development of novel vector-control interventions and further our understanding of toxicology in arthropods. Mosquitoes are responsible for the transmission of pathogens, which cause diseases that are of major health significance such as dengue fever and malaria. Preventive strategies involving the use of insecticides, however, have led to the emergence of resistant mosquitoes. Consequently, development of complementary approaches is urgently needed to stop the spread of these pathogens. Our study reports on a pioneering approach to investigate how well drugs are taken up by the mosquitoes and how long they reside in their body. We focused on ivermectin, which is toxic for mosquitoes, and several drugs that interfere with drug metabolising enzymes. We demonstrated that the exposure of drugs can be precisely determined in individual mosquitoes and that drugs interact with each other in the same way as observed in vertebrates. In this regard, we were able to increase the exposure and mosquito toxicity of ivermectin by co-administering ritonavir, a broad-spectrum inhibitor of drug metabolising enzymes. This study establishes Aedes mosquitoes as a new model organism for pharmacokinetic studies. It opens the door for the investigation of novel insecticide strategies and optimisation of lead compounds against mosquitoes.
Collapse
Affiliation(s)
- Urs Duthaler
- Division of Clinical Pharmacology & Toxicology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Division of Clinical Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- * E-mail:
| | - Michael Weber
- Division of Clinical Pharmacology & Toxicology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Division of Clinical Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Lorenz Hofer
- Swiss Tropical and Public Health institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Carlos Chaccour
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Facultad de Medicina, Universidad de Navarra, Pamplona, Spain
- Ifakara Health Institute, Ifakara, United Republic of Tanzania
| | - Marta Maia
- Kenyan Medical Research Institute, Wellcome Trust Research Programme, Department of Biosciences, Kilifi, Kenya
- University of Oxford, Nuffield Department of Medicine, Centre for Global Health and Tropical Medicine, Oxford, United Kingdom
| | - Pie Müller
- Swiss Tropical and Public Health institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Division of Clinical Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Felix Hammann
- Division of Clinical Pharmacology & Toxicology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Division of Clinical Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Clinical Pharmacology and Toxicology, Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| |
Collapse
|
17
|
Spatial and genetic clustering of Plasmodium falciparum and Plasmodium vivax infections in a low-transmission area of Ethiopia. Sci Rep 2020; 10:19975. [PMID: 33203956 PMCID: PMC7672087 DOI: 10.1038/s41598-020-77031-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/02/2020] [Indexed: 11/23/2022] Open
Abstract
The distribution of malaria infections is heterogeneous in space and time, especially in low transmission settings. Understanding this clustering may allow identification and targeting of pockets of transmission. In Adama district, Ethiopia, Plasmodium falciparum and P. vivax malaria patients and controls were examined, together with household members and immediate neighbors. Rapid diagnostic test and quantitative PCR (qPCR) were used for the detection of infections that were genetically characterized by a panel of microsatellite loci for P. falciparum (26) and P. vivax (11), respectively. Individuals living in households of clinical P. falciparum patients were more likely to have qPCR detected P. falciparum infections (22.0%, 9/41) compared to individuals in control households (8.7%, 37/426; odds ratio, 2.9; 95% confidence interval, 1.3–6.4; P = .007). Genetically related P. falciparum, but not P. vivax infections showed strong clustering within households. Genotyping revealed a marked temporal cluster of P. falciparum infections, almost exclusively comprised of clinical cases. These findings uncover previously unappreciated transmission dynamics and support a rational approach to reactive case detection strategies for P. falciparum in Ethiopia.
Collapse
|
18
|
Harrington LC, Foy BD, Bangs MJ. Considerations for Human Blood-Feeding and Arthropod Exposure in Vector Biology Research: An Essential Tool for Investigations and Disease Control. Vector Borne Zoonotic Dis 2020; 20:807-816. [PMID: 32905735 DOI: 10.1089/vbz.2020.2620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Eventually there may be a broadly acceptable, even perfected, substitute for the human host requirement for direct feeding experiments by arthropods, most notably mosquitoes. However, for now, direct and indirect feeding on human volunteers is an important, if not essential, tool in vector biology research (VBR). This article builds on the foundational publication by Achee et al. (2015) covering considerations for the use of human participants in VBR pursuits. The authors introduced methods involving human participation in VBR, while detailing human-landing collections (catches) as a prime example. Benedict et al. (2018) continued this theme with an overview of human participation and considerations for research that involves release of mosquito vectors into the environment. In this study, we discuss another important aspect of human use in VBR activities: considerations addressing studies that require an arthropod to feed on a live human host. Using mosquito studies as our principal example, in this study, we discuss the tremendous importance and value of this approach to support and allow study of a wide variety of factors and interactions related to our understanding of vector-borne diseases and their control. This includes establishment of laboratory colonies for test populations, characterization of essential nutrients that contribute to mosquito fitness, characterization of blood-feeding (biting) behavior and pathogen transmission, parameterization for modeling transmission dynamics, evaluation of human host attraction and/or agents that repel, and the effectiveness of antivector or parasite therapeutic drug studies.
Collapse
Affiliation(s)
| | - Brian D Foy
- Department of Microbiology, Immunology & Pathology, Arthropod-Borne and Infectious Diseases Laboratory Fort Collins, Colorado State University, Fort Collins, Colorado, USA
| | - Michael J Bangs
- Public Health & Malaria Control, PT Freeport Indonesia/International SOS, Kuala Kencana, Indonesia.,Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
19
|
Suh E, Grossman MK, Waite JL, Dennington NL, Sherrard-Smith E, Churcher TS, Thomas MB. The influence of feeding behaviour and temperature on the capacity of mosquitoes to transmit malaria. Nat Ecol Evol 2020; 4:940-951. [PMID: 32367033 PMCID: PMC7334094 DOI: 10.1038/s41559-020-1182-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022]
Abstract
Insecticide-treated bed nets reduce malaria transmission by limiting contact between mosquito vectors and human hosts when mosquitoes feed during the night. However, malaria vectors can also feed in the early evening and in the morning when people are not protected. Here, we explored how the timing of blood feeding interacts with environmental temperature to influence the capacity of Anopheles mosquitoes to transmit the human malaria parasite Plasmodium falciparum. In laboratory experiments, we found no effect of biting time itself on the proportion of mosquitoes that became infectious (vector competence) at constant temperature. However, when mosquitoes were maintained under more realistic fluctuating temperatures, there was a significant increase in competence for mosquitoes feeding in the evening (18:00), and a significant reduction in competence for those feeding in the morning (06:00), relative to those feeding at midnight (00:00). These effects appear to be due to thermal sensitivity of malaria parasites during the initial stages of parasite development within the mosquito, and the fact that mosquitoes feeding in the evening experience cooling temperatures during the night, whereas mosquitoes feeding in the morning quickly experience warming temperatures that are inhibitory to parasite establishment. A transmission dynamics model illustrates that such differences in competence could have important implications for malaria prevalence, the extent of transmission that persists in the presence of bed nets, and the epidemiological impact of behavioural resistance. These results indicate that the interaction of temperature and feeding behaviour could be a major ecological determinant of the vectorial capacity of malaria mosquitoes.
Collapse
Affiliation(s)
- Eunho Suh
- Center for Infectious Disease Dynamics, Department of Entomology, Penn State University, University Park, PA, USA.
| | - Marissa K Grossman
- Center for Infectious Disease Dynamics, Department of Entomology, Penn State University, University Park, PA, USA
| | - Jessica L Waite
- Center for Infectious Disease Dynamics, Department of Entomology, Penn State University, University Park, PA, USA.,Green Mountain Antibodies, Burlington, VT, USA
| | - Nina L Dennington
- Center for Infectious Disease Dynamics, Department of Entomology, Penn State University, University Park, PA, USA
| | - Ellie Sherrard-Smith
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Thomas S Churcher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Matthew B Thomas
- Center for Infectious Disease Dynamics, Department of Entomology, Penn State University, University Park, PA, USA
| |
Collapse
|
20
|
Karbwang J, Na‐Bangchang K. The Role of Clinical Pharmacology in Chemotherapy of Multidrug‐Resistant
Plasmodium falciparum. J Clin Pharmacol 2020; 60:830-847. [DOI: 10.1002/jcph.1589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/21/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Juntra Karbwang
- Graduate Program in Bioclinical SciencesChulabhorn International College of MedicineThammasat University (Rangsit Campus) Pathumthani Thailand
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and CholangiocarcinomaThammasat University (Rangsit Campus) Pathumthani Thailand
- Drug Discovery and Development Center, Office of Advanced Science and TechnologyThammasat University (Rangsit Campus) Pathumthani Thailand
- Department of Clinical Product developmentNagasaki Institute of Tropical MedicineNagasaki University Nagasaki Japan
| | - Kesara Na‐Bangchang
- Graduate Program in Bioclinical SciencesChulabhorn International College of MedicineThammasat University (Rangsit Campus) Pathumthani Thailand
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and CholangiocarcinomaThammasat University (Rangsit Campus) Pathumthani Thailand
- Drug Discovery and Development Center, Office of Advanced Science and TechnologyThammasat University (Rangsit Campus) Pathumthani Thailand
| |
Collapse
|
21
|
Singh L, Fontinha D, Francisco D, Mendes AM, Prudêncio M, Singh K. Molecular Design and Synthesis of Ivermectin Hybrids Targeting Hepatic and Erythrocytic Stages of Plasmodium Parasites. J Med Chem 2020; 63:1750-1762. [PMID: 32011136 DOI: 10.1021/acs.jmedchem.0c00033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ivermectin is a powerful endectocide, which reduces the incidence of vector-borne diseases. Besides its strong insecticidal effect on mosquito vectors of the disease, ivermectin inhibits Plasmodium falciparum sporogonic and blood stage development and impairs Plasmodium berghei development inside hepatocytes, both in vitro and in vivo. Herein, we present the first report on structural modification of ivermectin to produce dual-action molecular hybrids with good structure-dependent in vitro activity against both the hepatic and erythrocytic stages of P. berghei and P. falciparum infection, suggesting inclusion of ivermectin antimalarial hybrids in malaria control strategies. The most active hybrid displayed over threefold and 10-fold higher in vitro activity than ivermectin against hepatic and blood stage infections, respectively. Although an overwhelming insecticidal effect against Anopheles stephensi mosquitoes in laboratory conditions was not noticed, in silico docking analysis supports allosteric binding to glutamate-gated chloride channels similar to ivermectin.
Collapse
Affiliation(s)
- Lovepreet Singh
- Department of Chemistry , Guru Nanak Dev University , Amritsar 143 005 , India
| | - Diana Fontinha
- Instituto de Medicina Molecular , Faculdade de Medicina da Universidade de Lisboa , Av. Prof. Egas Moniz , Lisboa 1649-028 , Portugal
| | - Denise Francisco
- Instituto de Medicina Molecular , Faculdade de Medicina da Universidade de Lisboa , Av. Prof. Egas Moniz , Lisboa 1649-028 , Portugal
| | - Antonio M Mendes
- Instituto de Medicina Molecular , Faculdade de Medicina da Universidade de Lisboa , Av. Prof. Egas Moniz , Lisboa 1649-028 , Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular , Faculdade de Medicina da Universidade de Lisboa , Av. Prof. Egas Moniz , Lisboa 1649-028 , Portugal
| | - Kamaljit Singh
- Department of Chemistry , Guru Nanak Dev University , Amritsar 143 005 , India
| |
Collapse
|
22
|
Billingsley P, Binka F, Chaccour C, Foy B, Gold S, Gonzalez-Silva M, Jacobson J, Jagoe G, Jones C, Kachur P, Kobylinski K, Last A, Lavery JV, Mabey D, Mboera D, Mbogo C, Mendez-Lopez A, Rabinovich NR, Rees S, Richards F, Rist C, Rockwood J, Ruiz-Castillo P, Sattabongkot J, Saute F, Slater H, Steer A, Xia K, Zullinger R. A Roadmap for the Development of Ivermectin as a Complementary Malaria Vector Control Tool. Am J Trop Med Hyg 2020; 102:3-24. [PMID: 31971144 PMCID: PMC7008306 DOI: 10.4269/ajtmh.19-0620] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
In the context of stalling progress against malaria, resistance of mosquitoes to insecticides, and residual transmission, mass drug administration (MDA) of ivermectin, an endectocide used for neglected tropical diseases (NTDs), has emerged as a promising complementary vector control method. Ivermectin reduces the life span of Anopheles mosquitoes that feed on treated humans and/or livestock, potentially decreasing malaria parasite transmission when administered at the community level. Following the publication by WHO of the preferred product characteristics for endectocides as vector control tools, this roadmap provides a comprehensive view of processes needed to make ivermectin available as a vector control tool by 2024 with a completely novel mechanism of action. The roadmap covers various aspects, which include 1) the definition of optimal dosage/regimens for ivermectin MDA in both humans and livestock, 2) the risk of resistance to the drug and environmental impact, 3) ethical issues, 4) political and community engagement, 5) translation of evidence into policy, and 6) operational aspects of large-scale deployment of the drug, all in the context of a drug given as a prevention tool acting at the community level. The roadmap reflects the insights of a multidisciplinary group of global health experts who worked together to elucidate the path to inclusion of ivermectin in the toolbox against malaria, to address residual transmission, counteract insecticide resistance, and contribute to the end of this deadly disease.
Collapse
Affiliation(s)
| | - Fred Binka
- University of Health and Allied Sciences
| | | | | | | | | | | | | | | | | | | | - Anna Last
- London School of Hygiene and Tropical Medicine
| | | | - David Mabey
- London School of Hygiene and Tropical Medicine
| | | | | | | | | | | | | | - Cassidy Rist
- Virginia-Maryland College of Veterinary Medicine at Virginia Tech
| | | | | | | | | | | | | | - Kang Xia
- School of Plant and Environmental Sciences, Virginia Tech
| | - Rose Zullinger
- US President’s Malaria Initiative/US Centers for Disease Control and Prevention
| |
Collapse
|
23
|
Teboh-Ewungkem MI, Ngwa GA. Fighting malaria with ivermectin: a novel malaria control tool. THE LANCET. INFECTIOUS DISEASES 2020; 20:394-395. [PMID: 31948766 DOI: 10.1016/s1473-3099(19)30691-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 10/25/2022]
Affiliation(s)
| | - Gideon A Ngwa
- Department of Mathematics, University of Buea, Buea, PO Box 63, Cameroon
| |
Collapse
|
24
|
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: 41] [Impact Index Per Article: 10.3] [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
|
25
|
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: 2.2] [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
|
26
|
Smit MR, Ochomo EO, Aljayyoussi G, Kwambai TK, Abong’o BO, Bousema T, Waterhouse D, Bayoh NM, Gimnig JE, Samuels AM, Desai MR, Phillips-Howard PA, Kariuki SK, Wang D, Ward SA, ter Kuile FO. Human Direct Skin Feeding Versus Membrane Feeding to Assess the Mosquitocidal Efficacy of High-Dose Ivermectin (IVERMAL Trial). Clin Infect Dis 2019; 69:1112-1119. [PMID: 30590537 PMCID: PMC6743833 DOI: 10.1093/cid/ciy1063] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Ivermectin is being considered for mass drug administration for malaria, due to its ability to kill mosquitoes feeding on recently treated individuals. In a recent trial, 3-day courses of 300 and 600 mcg/kg/day were shown to kill Anopheles mosquitoes for at least 28 days post-treatment when fed patients' venous blood using membrane feeding assays. Direct skin feeding on humans may lead to higher mosquito mortality, as ivermectin capillary concentrations are higher. We compared mosquito mortality following direct skin and membrane feeding. METHODS We conducted a mosquito feeding study, nested within a randomized, double-blind, placebo-controlled trial of 141 adults with uncomplicated malaria in Kenya, comparing 3 days of ivermectin 300 mcg/kg/day, ivermectin 600 mcg/kg/day, or placebo, all co-administered with 3 days of dihydroartemisinin-piperaquine. On post-treatment day 7, direct skin and membrane feeding assays were conducted using laboratory-reared Anopheles gambiae sensu stricto. Mosquito survival was assessed daily for 28 days post-feeding. RESULTS Between July 20, 2015, and May 7, 2016, 69 of 141 patients participated in both direct skin and membrane feeding (placebo, n = 23; 300 mcg/kg/day, n = 24; 600 mcg/kg/day, n = 22). The 14-day post-feeding mortality for mosquitoes fed 7 days post-treatment on blood from pooled patients in both ivermectin arms was similar with direct skin feeding (mosquitoes observed, n = 2941) versus membrane feeding (mosquitoes observed, n = 7380): cumulative mortality (risk ratio 0.99, 95% confidence interval [CI] 0.95-1.03, P = .69) and survival time (hazard ratio 0.96, 95% CI 0.91-1.02, P = .19). Results were consistent by sex, by body mass index, and across the range of ivermectin capillary concentrations studied (0.72-73.9 ng/mL). CONCLUSIONS Direct skin feeding and membrane feeding on day 7 resulted in similar mosquitocidal effects of ivermectin across a wide range of drug concentrations, suggesting that the mosquitocidal effects seen with membrane feeding accurately reflect those of natural biting. Membrane feeding, which is more patient friendly and ethically acceptable, can likely reliably be used to assess ivermectin's mosquitocidal efficacy. CLINICAL TRIALS REGISTRATION NCT02511353.
Collapse
Affiliation(s)
- Menno R Smit
- Liverpool School of Tropical Medicine, United Kingdom
| | - Eric O Ochomo
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu
| | | | - Titus K Kwambai
- Liverpool School of Tropical Medicine, United Kingdom
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu
- Kenya Ministry of Health, Kisumu County, Kisumu
| | - Bernard O Abong’o
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu
| | - Teun Bousema
- Radboud University Medical Center, Nijmegen, The Netherlands
- London School of Hygiene and Tropical Medicine, United Kingdom
| | | | - Nabie M Bayoh
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, Georgia
| | - John E Gimnig
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, Georgia
| | - Aaron M Samuels
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, Georgia
| | - Meghna R Desai
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, Georgia
| | | | - Simon K Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu
| | - Duolao Wang
- Liverpool School of Tropical Medicine, United Kingdom
| | | | | |
Collapse
|
27
|
Selvaraj P, Suresh J, Wenger EA, Bever CA, Gerardin J. Reducing malaria burden and accelerating elimination with long-lasting systemic insecticides: a modelling study of three potential use cases. Malar J 2019; 18:307. [PMID: 31488139 PMCID: PMC6727392 DOI: 10.1186/s12936-019-2942-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/28/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND While bed nets and insecticide spraying have had significant impact on malaria burden in many endemic regions, outdoor vector feeding and insecticide resistance may ultimately limit their contribution to elimination and control campaigns. Complementary vector control methods such as endectocides or systemic insecticides, where humans or animals are treated with drugs that kill mosquitoes upon ingestion via blood meal, are therefore generating much interest. This work explores the conditions under which long-lasting systemic insecticides would have a substantial impact on transmission and burden. METHODS Hypothetical long-lasting systemic insecticides with effective durations ranging from 14 to 90 days are simulated using an individual-based mathematical model of malaria transmission. The impact of systemic insecticides when used to complement existing vector control and drug campaigns is evaluated in three settings-a highly seasonal high-transmission setting, a near-elimination setting with seasonal travel to a high-risk area, and a near-elimination setting in southern Africa. RESULTS At 60% coverage, a single round of long-lasting systemic insecticide with effective duration of at least 60 days, distributed at the start of the season alongside a seasonal malaria chemoprevention campaign in a high-transmission setting, results in further burden reduction of 30-90% depending on the sub-populations targeted. In a near-elimination setting where transmission is sustained by seasonal travel to a high-risk area, targeting high-risk travellers with systemic insecticide with effective duration of at least 30 days can result in likely elimination even if intervention coverage is as low as 50%. In near-elimination settings with robust vector control, the addition of a 14-day systemic insecticide alongside an anti-malarial in mass drug administration (MDA) campaigns can decrease the necessary MDA coverage from about 85% to the more easily achievable 65%. CONCLUSIONS While further research into the safety profile of systemic insecticides is necessary before deployment, models predict that long-lasting systemic insecticides can play a critical role in reducing burden or eliminating malaria in a range of contexts with different target populations, existing malaria control methods, and transmission intensities. Continued investment in lengthening the duration of systemic insecticides and improving their safety profile is needed for this intervention to achieve its fullest potential.
Collapse
Affiliation(s)
| | | | | | | | - Jaline Gerardin
- Institute for Disease Modeling, Bellevue, WA, USA. .,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
28
|
Siewe Fodjo JN, Kugler M, Hotterbeekx A, Hendy A, Van Geertruyden JP, Colebunders R. Would ivermectin for malaria control be beneficial in onchocerciasis-endemic regions? Infect Dis Poverty 2019; 8:77. [PMID: 31439040 PMCID: PMC6706915 DOI: 10.1186/s40249-019-0588-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/09/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There is accumulating evidence supporting the use of ivermectin as a malaria control tool. Recent findings from the repeat ivermectin mass drug administrations for control of malaria trial demonstrated a reduced incidence of malaria in villages which received repeated ivermectin mass drug administration (MDA; six doses) compared to those who had only one round of ivermectin. Several other studies investigating the benefits of ivermectin for malaria purposes are ongoing/planned. MAIN TEXT While ivermectin MDA offers promising perspectives in the fight against malaria, we highlight the added benefits and anticipated challenges of conducting future studies in onchocerciasis-endemic regions, which are confronted with a substantial disease burden including onchocerciasis-associated epilepsy. Increasing the frequency of ivermectin MDA in such places may reduce the burden of both malaria and onchocerciasis, and allow for more entomological investigations on both the Anopheles mosquitoes and the blackflies. Upfront, acceptability and feasibility studies are needed to assess the endorsement by the local populations, as well as the programmatic feasibility of implementing ivermectin MDA several times a year. CONCLUSIONS Onchocerciasis-endemic sites would doubly benefit from ivermectin MDA interventions, as these will alleviate onchocerciasis-associated morbidity and mortality, while potentially curbing malaria transmission. Involving onchocerciasis programs and other relevant stakeholders in the malaria/ivermectin research agenda would foster the implementation of pluri-annual MDA in target communities.
Collapse
Affiliation(s)
- Joseph Nelson Siewe Fodjo
- Global Health Institute, University of Antwerp, Kinsbergen centrum, Doornstraat 331, 2610, Antwerp, Belgium
| | - Marina Kugler
- Global Health Institute, University of Antwerp, Kinsbergen centrum, Doornstraat 331, 2610, Antwerp, Belgium
| | - An Hotterbeekx
- Global Health Institute, University of Antwerp, Kinsbergen centrum, Doornstraat 331, 2610, Antwerp, Belgium
| | - Adam Hendy
- Department of Pathology, University of Texas Medical Branch, Galveston, USA
| | | | - Robert Colebunders
- Global Health Institute, University of Antwerp, Kinsbergen centrum, Doornstraat 331, 2610, Antwerp, Belgium.
| |
Collapse
|
29
|
Duthaler U, Suenderhauf C, Gaugler S, Vetter B, Krähenbühl S, Hammann F. Development and validation of an LC-MS/MS method for the analysis of ivermectin in plasma, whole blood, and dried blood spots using a fully automatic extraction system. J Pharm Biomed Anal 2019; 172:18-25. [DOI: 10.1016/j.jpba.2019.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/05/2019] [Indexed: 10/27/2022]
|
30
|
Ivermectin Impairs the Development of Sexual and Asexual Stages of Plasmodium falciparum In Vitro. Antimicrob Agents Chemother 2019; 63:AAC.00085-19. [PMID: 31109978 DOI: 10.1128/aac.00085-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022] Open
Abstract
Ivermectin is the drug of choice for many parasitic infections, with more than one billion doses being distributed in onchocerciasis programs. The drug has been put into focus recently by the malaria community because of its potential to kill blood-sucking mosquitoes, thereby reducing malaria transmission. However, the activity of ivermectin against the malaria parasite itself has been only partly investigated. This study aimed to investigate the in vitro activity of ivermectin against asexual and sexual stages of Plasmodium falciparum Both asexual and late-stage gametocytes were incubated with ivermectin and control drugs in vitro The growth-inhibiting effects were assessed for asexual stages of different Plasmodium falciparum laboratory strains and culture-adapted clinical isolates using the histidine-rich protein 2 enzyme-linked immunosorbent assay technique. The effect against stage IV/V gametocytes was evaluated based on ATP quantification. Ivermectin showed activities at nanomolar concentrations against asexual stages (50% inhibitory concentration of ∼100 nM) and stage IV/V gametocytes (500 nM) of P. falciparum Stage-specific assays suggested that ivermectin arrests the parasite cycle at the trophozoite stage. Ivermectin might add a feature to its "wonder drug" properties with activity against asexual stages of the malaria parasite Plasmodium falciparum The observed activities might be difficult to reach with current regimens but will be more relevant with future high-dose regimens under investigation. Further studies should be performed to confirm these results in vitro and in vivo.
Collapse
|
31
|
Luo J, Zhang P, Liu R, Li X, Hua P, Li S, Zhang T, Zhang T, Fu Y, Song X, Gong T, Zhang Z. Efficient weapon for protracted warfare to malaria: A chondroitin sulfate derivates-containing injectable, ultra-long-lasting meshy-gel system. Carbohydr Polym 2019; 214:131-141. [DOI: 10.1016/j.carbpol.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 11/25/2022]
|
32
|
Foy BD, Alout H, Seaman JA, Rao S, Magalhaes T, Wade M, Parikh S, Soma DD, Sagna AB, Fournet F, Slater HC, Bougma R, Drabo F, Diabaté A, Coulidiaty AGV, Rouamba N, Dabiré RK. Efficacy and risk of harms of repeat ivermectin mass drug administrations for control of malaria (RIMDAMAL): a cluster-randomised trial. Lancet 2019; 393:1517-1526. [PMID: 30878222 PMCID: PMC6459982 DOI: 10.1016/s0140-6736(18)32321-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Ivermectin is widely used in mass drug administrations for controlling neglected parasitic diseases, and can be lethal to malaria vectors that bite treated humans. Therefore, it could be a new tool to reduce plasmodium transmission. We tested the hypothesis that frequently repeated mass administrations of ivermectin to village residents would reduce clinical malaria episodes in children and would be well tolerated with minimal harms. METHODS We invited villages (clusters) in Burkina Faso to participate in a single-blind (outcomes assessor), parallel-assignment, two-arm, cluster-randomised trial over the 2015 rainy season. Villages were assigned (1:1) by random draw to either the intervention group or the control group. In both groups, all eligible participants who consented to the treatment and were at least 90 cm in height received single oral doses of ivermectin (150-200 μg/kg) and albendazole (400 mg), and those in the intervention group received five further doses of ivermectin alone at 3-week intervals thereafter over the 18-week treatment phase. The primary outcome was cumulative incidence of uncomplicated malaria episodes over 18 weeks (analysed on a cluster intention-to-treat basis) in an active case detection cohort of children aged 5 years or younger living in the study villages. This trial is registered with ClinicalTrials.gov, number NCT02509481. FINDINGS Eight villages agreed to participate, and four were randomly assigned to each group. 2712 participants (1333 [49%] males and 1379 [51%] females; median age 15 years [IQR 6-34]), including 590 children aged 5 years or younger, provided consent and were enrolled between May 22 and July 20, 2015 (except for 77 participants enrolled after these dates because of unavailability before the first mass drug administration, travel into the village during the trial, or birth), with 1447 enrolled into the intervention group and 1265 into the control group. 330 (23%) participants in the intervention group and 233 (18%) in the control group met the exclusion criteria for mass drug administration. Most children in the active case detection cohort were not treated because of height restrictions. 14 (4%) children in the intervention group and 10 (4%) in the control group were lost to follow-up. Cumulative malaria incidence was reduced in the intervention group (648 episodes among 327 children; estimated mean 2·00 episodes per child) compared with the control group (647 episodes among 263 children; 2·49 episodes per child; risk difference -0·49 [95% CI -0·79 to -0·21], p=0·0009, adjusted for sex and clustering). The risk of adverse events among all participants did not differ between groups (45 events [3%] among 1447 participants in the intervention group vs 24 events [2%] among 1265 in the control group; risk ratio 1·63 [1·01 to 2·67]; risk difference 1·21 [0·04 to 2·38], p=0·060), and no adverse reactions were reported. INTERPRETATION Frequently repeated mass administrations of ivermectin during the malaria transmission season can reduce malaria episodes among children without significantly increasing harms in the populace. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Brian D Foy
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| | - Haoues Alout
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Jonathan A Seaman
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Sangeeta Rao
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Tereza Magalhaes
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Martina Wade
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Sunil Parikh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Dieudonné D Soma
- Institute of Research in Health Sciences, Western Regional Direction, National Center for Scientific and Technological Research, Bobo-Dioulasso, Burkina Faso; International Mixed Laboratory on Vector Diseases, Bobo-Dioulasso, Burkina Faso
| | - André B Sagna
- International Mixed Laboratory on Vector Diseases, Bobo-Dioulasso, Burkina Faso; Research Institute for Development, Infectious Diseases, and Vectors: Ecology, Genetics, Evolution and Control, National Centre for Scientific Research, University of Montpellier, Montpellier, France
| | - Florence Fournet
- International Mixed Laboratory on Vector Diseases, Bobo-Dioulasso, Burkina Faso; Research Institute for Development, Infectious Diseases, and Vectors: Ecology, Genetics, Evolution and Control, National Centre for Scientific Research, University of Montpellier, Montpellier, France
| | - Hannah C Slater
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Roland Bougma
- National Program for the Fight against Neglected Tropical Diseases, Department of Disease Control, Ministry of Health, Ouagadougou, Burkina Faso
| | - François Drabo
- National Program for the Fight against Neglected Tropical Diseases, Department of Disease Control, Ministry of Health, Ouagadougou, Burkina Faso
| | - Abdoulaye Diabaté
- Institute of Research in Health Sciences, Western Regional Direction, National Center for Scientific and Technological Research, Bobo-Dioulasso, Burkina Faso; International Mixed Laboratory on Vector Diseases, Bobo-Dioulasso, Burkina Faso
| | | | - Nöel Rouamba
- Institute of Research in Health Sciences, Western Regional Direction, National Center for Scientific and Technological Research, Bobo-Dioulasso, Burkina Faso
| | - Roch K Dabiré
- Institute of Research in Health Sciences, Western Regional Direction, National Center for Scientific and Technological Research, Bobo-Dioulasso, Burkina Faso; International Mixed Laboratory on Vector Diseases, Bobo-Dioulasso, Burkina Faso
| |
Collapse
|
33
|
Burrows J, Slater H, Macintyre F, Rees S, Thomas A, Okumu F, Hooft van Huijsduijnen R, Duparc S, Wells TNC. A discovery and development roadmap for new endectocidal transmission-blocking agents in malaria. Malar J 2018; 17:462. [PMID: 30526594 PMCID: PMC6287360 DOI: 10.1186/s12936-018-2598-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/24/2018] [Indexed: 11/10/2022] Open
Abstract
Reaching the overall goal of eliminating malaria requires halting disease transmission. One approach to blocking transmission is to prevent passage of the parasite to a mosquito, by preventing formation or transmission of gametocytes. An alternative approach, pioneered in the veterinary field, is to use endectocides, which are molecules that render vertebrate blood meals toxic for the mosquito vector, also killing the parasite. Field studies and modelling suggest that reducing the lifespan of the mosquito may significantly reduce transmission, given the lengthy maturation process of the parasite. To guide the development of new endectocides, or the reformulation of existing molecules, it is important to construct a framework of the required attributes, commonly called the target candidate profile. Here, using a combination of insights from current endectocides, mathematical models of the malaria transmission dynamics, and known impacts of vector control, a target candidate profile (TCP-6) and a regulatory strategy are proposed for a transmission reducing agent. The parameters chosen can be used to assess the potential of a new medicine, independent of whether it has classical endectocide activity, reduces the insect and parasite lifespan or any combination of all three, thereby constituting an ‘endectocidal transmission blocking’ paradigm.
Collapse
Affiliation(s)
- Jeremy Burrows
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland.
| | - Hannah Slater
- PATH, 2201 Westlake Avenue, Seattle, WA, 98121, USA.,Department of Infectious Disease Epidemiology, MRC Centre for Global Disease Analysis, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - Fiona Macintyre
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Sarah Rees
- Innovative Vector Control Consortium, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Anna Thomas
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Fredros Okumu
- Environmental Health and Ecological Sciences Department, Ifakara Health Institute, Off Mlabani Passage, Ifakara, Morogoro, United Republic of Tanzania.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Republic of South Africa.,Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | | | - Stephan Duparc
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Timothy N C Wells
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| |
Collapse
|
34
|
Gomez SA, Chapman LAC, Dilger E, Courtenay O, Picado A. Estimating the efficacy of community-wide use of systemic insecticides in dogs to control zoonotic visceral leishmaniasis: A modelling study in a Brazilian scenario. PLoS Negl Trop Dis 2018; 12:e0006797. [PMID: 30222756 PMCID: PMC6160230 DOI: 10.1371/journal.pntd.0006797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/27/2018] [Accepted: 08/28/2018] [Indexed: 12/31/2022] Open
Abstract
Systemic insecticides in dogs have been suggested as a public health intervention to prevent human cases of Zoonotic Visceral Leishmaniasis (ZVL). But, currently there are no systemic insecticides for dogs registered against zoo-anthropophilic pool blood feeding phlebotomine flies. We predict the impact of community-wide use of systemic insecticide in dog populations as a public health measure to control transmission of Leishmania infantum to humans using a mathematical model. We developed a Susceptible-Exposed-Infected (SEI) compartmental model to describe L. infantum transmission dynamics in dogs, with a vectorial capacity term to represent transmission between L. infantum-hosting dogs via phlebotomine flies. For Infected (I) dogs two levels of infectiousness were modelled, high infectiousness and low infectiousness. Human incidence was estimated through its relationship to infection in the dog population. We evaluated outcomes from a wide range of scenarios comprising different combinations of initial insecticide efficacy, duration of insecticide efficacy over time, and proportion of the dog population treated (60%, 70% & 80%). The same reduction in human infection incidence can be achieved via different combinations of insecticide efficacy, duration and dog coverage. For example, a systemic insecticide with an initial efficacy of 80% and 6 months above 65% efficacy would require treating at least 70% of the dogs to reduce the human infection incidence by 50%. Sensitivity analysis showed that the model outcome was most sensitive to baseline values of phlebotomine fly daily survival rate and insecticide coverage. Community-wide use of systemic insecticides applied to the “L. infantum canine reservoir” can significantly reduce human incidence of L. infantum infection. The results of this mathematical model can help defining the insecticide target product profile and how the insecticide should be applied to maximise effectiveness. Zoonotic visceral leishmaniasis (ZVL) is a potentially deadly disease in humans caused by Leishmania infantum. This leishmania species can be delivered by pool blood feeding zoo-anthropophilic phlebotomine flies to several mammals, the dog population being recognized as the main reservoir. Transmission from infected dogs to humans is through the bite of female phlebotomine sand flies. The disease is endemic in several countries and Brazil has a high prevalence of cases with over 3000 ZVL cases reported per year. The main, inefficient and highly controversial, control measure in Brazil has been culling sero-positive dogs. The community-wide use of systemic insecticides in dogs could be an alternative to control L. infantum transmission from phlebotomine flies to humans. The rationale is that phlebotomine flies which sampled their blood meals from dogs treated with systemic insecticides would die reducing the risk of L. infantum transmission. To reduce the number of ZVL cases, a large proportion of dogs in the community should be treated and the systemic insecticide used should be effective in killing phlebotomine flies acting as vectors of L. infantum parasites for a significant amount of time. We used a mathematical model mimicking L. infantum transmission to show that this novel vector control strategy could be effective. We identified the combination of different key parameters (e.g. insecticide efficacy, duration and proportion of dogs treated) that could lead to a significant reduction of the risk of L. infantum infection in humans.
Collapse
Affiliation(s)
- Sonia A. Gomez
- ISGlobal-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- * E-mail: (AP); (SAG)
| | - Lloyd A. C. Chapman
- Department of Global Health and Development, London School of Hygiene and Tropical Medicine, London, United Kingdom
- School of Life Sciences, University of Warwick, Gibbet Hill Campus, Coventry, United Kingdom
| | - Erin Dilger
- School of Life Sciences, University of Warwick, Gibbet Hill Campus, Coventry, United Kingdom
| | - Orin Courtenay
- School of Life Sciences, University of Warwick, Gibbet Hill Campus, Coventry, United Kingdom
| | - Albert Picado
- ISGlobal-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- * E-mail: (AP); (SAG)
| |
Collapse
|
35
|
Ng'habi K, Viana M, Matthiopoulos J, Lyimo I, Killeen G, Ferguson HM. Mesocosm experiments reveal the impact of mosquito control measures on malaria vector life history and population dynamics. Sci Rep 2018; 8:13949. [PMID: 30224714 PMCID: PMC6141522 DOI: 10.1038/s41598-018-31805-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/24/2018] [Indexed: 11/29/2022] Open
Abstract
The impact of control measures on mosquito vector fitness and demography is usually estimated from bioassays or indirect variables in the field. Whilst indicative, neither approach is sufficient to quantify the potentially complex response of mosquito populations to combined interventions. Here, large replicated mesocosms were used to measure the population-level response of the malaria vector Anopheles arabiensis to long-lasting insecticidal nets (LLINs) when used in isolation, or combined with insecticidal eave louvers (EL), or treatment of cattle with the endectocide Ivermectin (IM). State-space models (SSM) were fit to these experimental data, revealing that LLIN introduction reduced adult mosquito survival by 91% but allowed population persistence. ELs provided no additional benefit, but IM reduced mosquito fecundity by 59% and nearly eliminated all populations when combined with LLINs. This highlights the value of IM for integrated vector control, and mesocosm population experiments combined with SSM for identifying optimal combinations for vector population elimination.
Collapse
Affiliation(s)
- Kija Ng'habi
- Ifakara Health Institute, Environmental Health and Ecological Sciences, Ifakara, United Republic of Tanzania
- School of Health Sciences, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Mafalda Viana
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Jason Matthiopoulos
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Issa Lyimo
- Ifakara Health Institute, Environmental Health and Ecological Sciences, Ifakara, United Republic of Tanzania
| | - Gerry Killeen
- Ifakara Health Institute, Environmental Health and Ecological Sciences, Ifakara, United Republic of Tanzania
- Liverpool School of Tropical Medicine, Department of Vector Biology, Liverpool, United Kingdom
| | - Heather M Ferguson
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
36
|
Smith NR, Trauer JM, Gambhir M, Richards JS, Maude RJ, Keith JM, Flegg JA. Agent-based models of malaria transmission: a systematic review. Malar J 2018; 17:299. [PMID: 30119664 PMCID: PMC6098619 DOI: 10.1186/s12936-018-2442-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/04/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Much of the extensive research regarding transmission of malaria is underpinned by mathematical modelling. Compartmental models, which focus on interactions and transitions between population strata, have been a mainstay of such modelling for more than a century. However, modellers are increasingly adopting agent-based approaches, which model hosts, vectors and/or their interactions on an individual level. One reason for the increasing popularity of such models is their potential to provide enhanced realism by allowing system-level behaviours to emerge as a consequence of accumulated individual-level interactions, as occurs in real populations. METHODS A systematic review of 90 articles published between 1998 and May 2018 was performed, characterizing agent-based models (ABMs) relevant to malaria transmission. The review provides an overview of approaches used to date, determines the advantages of these approaches, and proposes ideas for progressing the field. RESULTS The rationale for ABM use over other modelling approaches centres around three points: the need to accurately represent increased stochasticity in low-transmission settings; the benefits of high-resolution spatial simulations; and heterogeneities in drug and vaccine efficacies due to individual patient characteristics. The success of these approaches provides avenues for further exploration of agent-based techniques for modelling malaria transmission. Potential extensions include varying elimination strategies across spatial landscapes, extending the size of spatial models, incorporating human movement dynamics, and developing increasingly comprehensive parameter estimation and optimization techniques. CONCLUSION Collectively, the literature covers an extensive array of topics, including the full spectrum of transmission and intervention regimes. Bringing these elements together under a common framework may enhance knowledge of, and guide policies towards, malaria elimination. However, because of the diversity of available models, endorsing a standardized approach to ABM implementation may not be possible. Instead it is recommended that model frameworks be contextually appropriate and sufficiently described. One key recommendation is to develop enhanced parameter estimation and optimization techniques. Extensions of current techniques will provide the robust results required to enhance current elimination efforts.
Collapse
Affiliation(s)
- Neal R Smith
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| | - James M Trauer
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Manoj Gambhir
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- IBM Research Australia, Melbourne, Australia
| | - Jack S Richards
- Life Sciences, Burnet Institute, Melbourne, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
- Department of Infectious Diseases, Monash University, Melbourne, Australia
| | - 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 Medicine, University of Oxford, Oxford, UK
- Harvard TH Chan School of Public Health, Harvard University, Boston, USA
| | - Jonathan M Keith
- School of Mathematical Sciences, Monash University, Clayton, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, University of Melbourne, Parkville, Australia
| |
Collapse
|
37
|
Challenges and opportunities in controlling mosquito-borne infections. Nature 2018; 559:490-497. [PMID: 30046071 DOI: 10.1038/s41586-018-0318-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/24/2018] [Indexed: 11/08/2022]
Abstract
Mosquito-borne diseases remain a major cause of morbidity and mortality across the tropical regions. Despite much progress in the control of malaria, malaria-associated morbidity remains high, whereas arboviruses-most notably dengue-are responsible for a rising burden of disease, even in middle-income countries that have almost completely eliminated malaria. Here I discuss how new interventions offer the promise of considerable future reductions in disease burden. However, I emphasize that intervention programmes need to be underpinned by rigorous trials and quantitative epidemiological analyses. Such analyses suggest that the long-term goal of elimination is more feasible for dengue than for malaria, even if malaria elimination would offer greater overall health benefit to the public.
Collapse
|
38
|
Miglianico M, Eldering M, Slater H, Ferguson N, Ambrose P, Lees RS, Koolen KMJ, Pruzinova K, Jancarova M, Volf P, Koenraadt CJM, Duerr HP, Trevitt G, Yang B, Chatterjee AK, Wisler J, Sturm A, Bousema T, Sauerwein RW, Schultz PG, Tremblay MS, Dechering KJ. Repurposing isoxazoline veterinary drugs for control of vector-borne human diseases. Proc Natl Acad Sci U S A 2018; 115:E6920-E6926. [PMID: 29967151 PMCID: PMC6055183 DOI: 10.1073/pnas.1801338115] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Isoxazolines are oral insecticidal drugs currently licensed for ectoparasite control in companion animals. Here we propose their use in humans for the reduction of vector-borne disease incidence. Fluralaner and afoxolaner rapidly killed Anopheles, Aedes, and Culex mosquitoes and Phlebotomus sand flies after feeding on a drug-supplemented blood meal, with IC50 values ranging from 33 to 575 nM, and were fully active against strains with preexisting resistance to common insecticides. Based on allometric scaling of preclinical pharmacokinetics data, we predict that a single human median dose of 260 mg (IQR, 177-407 mg) for afoxolaner, or 410 mg (IQR, 278-648 mg) for fluralaner, could provide an insecticidal effect lasting 50-90 days against mosquitoes and Phlebotomus sand flies. Computational modeling showed that seasonal mass drug administration of such a single dose to a fraction of a regional population would dramatically reduce clinical cases of Zika and malaria in endemic settings. Isoxazolines therefore represent a promising new component of drug-based vector control.
Collapse
Affiliation(s)
| | | | - Hannah Slater
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Neil Ferguson
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Pauline Ambrose
- The Liverpool Insect Testing Establishment, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Rosemary S Lees
- The Liverpool Insect Testing Establishment, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | | | - Katerina Pruzinova
- Department of Parasitology, Faculty of Science, Charles University, 116 36 Prague, Czech Republic
| | - Magdalena Jancarova
- Department of Parasitology, Faculty of Science, Charles University, 116 36 Prague, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, 116 36 Prague, Czech Republic
| | | | | | | | - Baiyuan Yang
- California Institute for Biomedical Research, La Jolla, CA 92037
| | | | - John Wisler
- California Institute for Biomedical Research, La Jolla, CA 92037
| | | | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, 6525 Nijmegen, The Netherlands
| | - Robert W Sauerwein
- TropIQ Health Sciences, 6534 Nijmegen, The Netherlands
- Department of Medical Microbiology, Radboud University Medical Center, 6525 Nijmegen, The Netherlands
| | - Peter G Schultz
- California Institute for Biomedical Research, La Jolla, CA 92037;
| | | | | |
Collapse
|
39
|
Cortopassi WA, Celmar Costa Franca T, Krettli AU. A systems biology approach to antimalarial drug discovery. Expert Opin Drug Discov 2018; 13:617-626. [DOI: 10.1080/17460441.2018.1471056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Wilian Augusto Cortopassi
- Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | | | | |
Collapse
|
40
|
Targeting cattle for malaria elimination: marked reduction of Anopheles arabiensis survival for over six months using a slow-release ivermectin implant formulation. Parasit Vectors 2018; 11:287. [PMID: 29728135 PMCID: PMC5935946 DOI: 10.1186/s13071-018-2872-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022] Open
Abstract
Background Mosquitoes that feed on animals can survive and mediate residual transmission of malaria even after most humans have been protected with insecticidal bednets or indoor residual sprays. Ivermectin is a widely-used drug for treating parasites of humans and animals that is also insecticidal, killing mosquitoes that feed on treated subjects. Mass administration of ivermectin to livestock could be particularly useful for tackling residual malaria transmission by zoophagic vectors that evade human-centred approaches. Ivermectin comes from a different chemical class to active ingredients currently used to treat bednets or spray houses, so it also has potential for mitigating against emergence of insecticide resistance. However, the duration of insecticidal activity obtained with ivermectin is critical to its effectiveness and affordability. Results A slow-release formulation for ivermectin was implanted into cattle, causing 40 weeks of increased mortality among Anopheles arabiensis that fed on them. For this zoophagic vector of residual malaria transmission across much of Africa, the proportion surviving three days after feeding (typical mean duration of a gonotrophic cycle in field populations) was approximately halved for 25 weeks. Conclusions This implantable ivermectin formulation delivers stable and sustained insecticidal activity for approximately 6 months. Residual malaria transmission by zoophagic vectors could be suppressed by targeting livestock with this long-lasting formulation, which would be impractical or unacceptable for mass treatment of human populations. Electronic supplementary material The online version of this article (10.1186/s13071-018-2872-y) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Traverso G, Kirtane AR, Schoellhammer CM, Langer R. Translation durch Konvergenz: Drug-Delivery-Forschung in multidisziplinären Teams. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201712512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Ameya R. Kirtane
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | | | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| |
Collapse
|
42
|
Smit MR, Ochomo EO, Aljayyoussi G, Kwambai TK, Abong'o BO, Chen T, Bousema T, Slater HC, Waterhouse D, Bayoh NM, Gimnig JE, Samuels AM, Desai MR, Phillips-Howard PA, Kariuki SK, Wang D, Ward SA, Ter Kuile FO. Safety and mosquitocidal efficacy of high-dose ivermectin when co-administered with dihydroartemisinin-piperaquine in Kenyan adults with uncomplicated malaria (IVERMAL): a randomised, double-blind, placebo-controlled trial. THE LANCET. INFECTIOUS DISEASES 2018; 18:615-626. [PMID: 29602751 DOI: 10.1016/s1473-3099(18)30163-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/09/2018] [Accepted: 02/14/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Ivermectin is being considered for mass drug administration for malaria due to its ability to kill mosquitoes feeding on recently treated individuals. However, standard, single doses of 150-200 μg/kg used for onchocerciasis and lymphatic filariasis have a short-lived mosquitocidal effect (<7 days). Because ivermectin is well tolerated up to 2000 μg/kg, we aimed to establish the safety, tolerability, and mosquitocidal efficacy of 3 day courses of high-dose ivermectin, co-administered with a standard malaria treatment. METHODS We did a randomised, double-blind, placebo-controlled, superiority trial at the Jaramogi Oginga Odinga Teaching and Referral Hospital (Kisumu, Kenya). Adults (aged 18-50 years) were eligible if they had confirmed symptomatic uncomplicated Plasmodium falciparum malaria and agreed to the follow-up schedule. Participants were randomly assigned (1:1:1) using sealed envelopes, stratified by sex and body-mass index (men: <21 vs ≥21 kg/m2; women: <23 vs ≥23 kg/m2), with permuted blocks of three, to receive 3 days of ivermectin 300 μg/kg per day, ivermectin 600 μg/kg per day, or placebo, all co-administered with 3 days of dihydroartemisinin-piperaquine. Blood of patients taken on post-treatment days 0, 2 + 4 h, 7, 10, 14, 21, and 28 was fed to laboratory-reared Anopheles gambiae sensu stricto mosquitoes, and mosquito survival was assessed daily for 28 days after feeding. The primary outcome was 14-day cumulative mortality of mosquitoes fed 7 days after ivermectin treatment (from participants who received at least one dose of study medication). The study is registered with ClinicalTrials.gov, number NCT02511353. FINDINGS Between July 20, 2015, and May 7, 2016, 741 adults with malaria were assessed for eligibility, of whom 141 were randomly assigned to receive ivermectin 600 μg/kg per day (n=47), ivermectin 300 μg/kg per day (n=48), or placebo (n=46). 128 patients (91%) attended the primary outcome visit 7 days post treatment. Compared with placebo, ivermectin was associated with higher 14 day post-feeding mosquito mortality when fed on blood taken 7 days post treatment (ivermectin 600 μg/kg per day risk ratio [RR] 2·26, 95% CI 1·93-2·65, p<0·0001; hazard ratio [HR] 6·32, 4·61-8·67, p<0·0001; ivermectin 300 μg/kg per day RR 2·18, 1·86-2·57, p<0·0001; HR 4·21, 3·06-5·79, p<0·0001). Mosquito mortality remained significantly increased 28 days post treatment (ivermectin 600 μg/kg per day RR 1·23, 1·01-1·50, p=0·0374; and ivermectin 300 μg/kg per day 1·21, 1·01-1·44, p=0·0337). Five (11%) of 45 patients receiving ivermectin 600 μg/kg per day, two (4%) of 48 patients receiving ivermectin 300 μg/kg per day, and none of 46 patients receiving placebo had one or more treatment-related adverse events. INTERPRETATION Ivermectin at both doses assessed was well tolerated and reduced mosquito survival for at least 28 days after treatment. Ivermectin 300 μg/kg per day for 3 days provided a good balance between efficacy and tolerability, and this drug shows promise as a potential new tool for malaria elimination. FUNDING Malaria Eradication Scientific Alliance (MESA) and US Centers for Disease Control and Prevention (CDC).
Collapse
Affiliation(s)
- Menno R Smit
- Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Eric O Ochomo
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | | | - Titus K Kwambai
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Kenya Ministry of Health, Kisumu County, Kisumu, Kenya
| | - Bernard O Abong'o
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Tao Chen
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Teun Bousema
- Radboud University Medical Center, Nijmegen, Netherlands; London School of Hygiene & Tropical Medicine, London, UK
| | - Hannah C Slater
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | | | - Nabie M Bayoh
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, GA, USA
| | - John E Gimnig
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, GA, USA
| | - Aaron M Samuels
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, GA, USA
| | - Meghna R Desai
- US Centers for Disease Control and Prevention, Center for Global Health, Division of Parasitic Diseases and Malaria, Atlanta, GA, USA
| | | | - Simon K Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Duolao Wang
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Steve A Ward
- Liverpool School of Tropical Medicine, Liverpool, UK
| | | |
Collapse
|
43
|
Traverso G, Kirtane AR, Schoellhammer CM, Langer R. Convergence for Translation: Drug-Delivery Research in Multidisciplinary Teams. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/anie.201712512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Ameya R. Kirtane
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | | | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| |
Collapse
|
44
|
Bellinger AM, Jafari M, Grant TM, Zhang S, Slater HC, Wenger EA, Mo S, Lee YAL, Mazdiyasni H, Kogan L, Barman R, Cleveland C, Booth L, Bensel T, Minahan D, Hurowitz HM, Tai T, Daily J, Nikolic B, Wood L, Eckhoff PA, Langer R, Traverso G. Oral, ultra-long-lasting drug delivery: Application toward malaria elimination goals. Sci Transl Med 2017; 8:365ra157. [PMID: 27856796 DOI: 10.1126/scitranslmed.aag2374] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/25/2016] [Indexed: 12/29/2022]
Abstract
Efforts at elimination of scourges, such as malaria, are limited by the logistic challenges of reaching large rural populations and ensuring patient adherence to adequate pharmacologic treatment. We have developed an oral, ultra-long-acting capsule that dissolves in the stomach and deploys a star-shaped dosage form that releases drug while assuming a geometry that prevents passage through the pylorus yet allows passage of food, enabling prolonged gastric residence. This gastric-resident, drug delivery dosage form releases small-molecule drugs for days to weeks and potentially longer. Upon dissolution of the macrostructure, the components can safely pass through the gastrointestinal tract. Clinical, radiographic, and endoscopic evaluation of a swine large-animal model that received these dosage forms showed no evidence of gastrointestinal obstruction or mucosal injury. We generated long-acting formulations for controlled release of ivermectin, a drug that targets malaria-transmitting mosquitoes, in the gastric environment and incorporated these into our dosage form, which then delivered a sustained therapeutic dose of ivermectin for up to 14 days in our swine model. Further, by using mathematical models of malaria transmission that incorporate the lethal effect of ivermectin against malaria-transmitting mosquitoes, we demonstrated that this system will boost the efficacy of mass drug administration toward malaria elimination goals. Encapsulated, gastric-resident dosage forms for ultra-long-acting drug delivery have the potential to revolutionize treatment options for malaria and other diseases that affect large populations around the globe for which treatment adherence is essential for efficacy.
Collapse
Affiliation(s)
- Andrew M Bellinger
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Lyndra Inc., Watertown, MA 02472, USA
| | - Mousa Jafari
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tyler M Grant
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Lyndra Inc., Watertown, MA 02472, USA
| | - Shiyi Zhang
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hannah C Slater
- Department of Infectious Disease Epidemiology, MRC (Medical Research Council) Centre for Outbreak Analysis and Modelling, Imperial College London, London, U.K
| | | | - Stacy Mo
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Young-Ah Lucy Lee
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hormoz Mazdiyasni
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lawrence Kogan
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ross Barman
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cody Cleveland
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lucas Booth
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Taylor Bensel
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Minahan
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Haley M Hurowitz
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tammy Tai
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Johanna Daily
- Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Boris Nikolic
- Biomatics Capital, 1107 1st Avenue, Apartment 1305, Seattle, WA 98101, USA
| | - Lowell Wood
- Institute for Disease Modeling, Bellevue, WA 98005, USA
| | | | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Media Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Traverso
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
45
|
Sampaio VDS, Rivas GBDS, Kobylinski K, Pinilla YT, Pimenta PFP, Lima JBP, Bruno RV, Lacerda MVG, Monteiro WM. What does not kill it makes it weaker: effects of sub-lethal concentrations of ivermectin on the locomotor activity of Anopheles aquasalis. Parasit Vectors 2017; 10:623. [PMID: 29282130 PMCID: PMC5745606 DOI: 10.1186/s13071-017-2563-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/30/2017] [Indexed: 01/09/2023] Open
Abstract
Background Malaria remains a major public health concern. Vector control measures based solely on insecticide treated nets (ITNs) and indoor residual spraying (IRS) have demonstrated not to be feasible for malaria elimination. It has been shown that ivermectin affects several aspects of Anopheles species biology. Along the Latin American seacoast, Anopheles aquasalis Curry plays an important role in malaria transmission. The observation of mosquitoes locomotor activity under laboratory conditions can reveal details of their daily activity rhythms, which is controlled by an endogenous circadian clock that seems to be influenced by external signals, such as light and temperature. In this study, we assessed basal locomotor activity and the effects of ivermectin on locomotor activity of the American malaria vector, An. aquasalis. Methods Adult females of Anopheles aquasalis used in experiments were three to five days post-emergence. Blood from one single subject was used to provide mosquito meals by membrane feeding assays. Powdered ivermectin compound was used to achieve different concentrations of drug as previously described. Fully engorged mosquitoes were individually placed into glass tubes and provided with 10% sucrose. Each tube was placed into a Locomotor Activity Monitor (LAM). The LAMs were kept inside an incubator under a constant temperature and a 12:12 h light:dark cycle. The average locomotor activity was calculated as the mean number of movements performed per mosquito in the period considered. Intervals of time assessed were adapted from a previous study. One-way ANOVA tests were performed in order to compare means between groups. Additionally, Dunnett’s method was used for post-hoc pairwise means comparisons between each group and control. Stata software version 13 was used for the analysis. Results Anopheles aquasalis showed a nocturnal and bimodal pattern for mosquitoes fed both control blood meals and sub-lethal concentrations of ivermectin. In this species, activity peaks occurred at the beginning of the photophase and scotophase in the control group. The nocturnal activity is evident and higher just after the evening peak and maintains basal levels of locomotion throughout the scotophase. In the entire group analysis, locomotor activity means of experimental sets were significantly lower than control for each period of time evaluated. In the survival group, the locomotor activity means of all treatment sets were lower than control mosquitoes for all intervals of time when both the whole period and scotophase were assessed. When the middle of scotophase was evaluated, means were significantly lower for LC15 and LC25, but not LC5. For the beginning of photophase period, significant differences were detected only between control and LC5. When both the photophase and scotophase were assessed alone, no significant differences were found. Mean locomotor activity was significantly lower for dead group when compared to survival group for all experimental sets when whole period, photophase, and scotophase were assessed. Conclusions Ivermectin seems to decrease locomotor activity of An. aquasalis at sub-lethal concentrations. The effects on locomotor activity increase according at higher ivermectin concentrations and are most evident during the whole scotophase as well as in the beginning and in the end of this phase, and sub-lethal effects may still be observed in the photophase. Findings presented in this study demonstrate that sub-lethal ivermectin effects reduce mosquito locomotor activity, which could diminish vectorial capacity and therefore the malaria transmission. Electronic supplementary material The online version of this article (10.1186/s13071-017-2563-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vanderson de Souza Sampaio
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil. .,Escola de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil. .,Sala de Análise de Situação em Saúde, Fundação de Vigilância em Saúde do Amazonas, Manaus, Brazil.
| | - Gustavo Bueno da Silva Rivas
- Department of Entomology and Nematology, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Kevin Kobylinski
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Yudi Tatiana Pinilla
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Escola de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil
| | | | | | | | - Marcus Vinícius Guimarães Lacerda
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Escola de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil.,Instituto de Pesquisa Leônidas & Maria Deane, Fundação Oswaldo Cruz (Fiocruz), Manaus, Brazil
| | - Wuelton Marcelo Monteiro
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Escola de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil
| |
Collapse
|
46
|
Kobylinski KC, Escobedo-Vargas KS, López-Sifuentes VM, Durand S, Smith ES, Baldeviano GC, Gerbasi RV, Ballard SB, Stoops CA, Vásquez GM. Ivermectin susceptibility, sporontocidal effect, and inhibition of time to re-feed in the Amazonian malaria vector Anopheles darlingi. Malar J 2017; 16:474. [PMID: 29162101 PMCID: PMC5696779 DOI: 10.1186/s12936-017-2125-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Outdoor malaria transmission hinders malaria elimination efforts in the Amazon region and novel vector control tools are needed. Ivermectin mass drug administration (MDA) to humans kills wild Anopheles, targets outdoor-feeding vectors, and can suppress malaria parasite transmission. Laboratory investigations were performed to determine ivermectin susceptibility, sporontocidal effect and inhibition of time to re-feed for the primary Amazonian malaria vector, Anopheles darlingi. METHODS To assess ivermectin susceptibility, various concentrations of ivermectin were mixed in human blood and fed to An. darlingi. Mosquito survival was monitored daily for 7 days and a non-linear mixed effects model with Probit analysis was used to calculate lethal concentrations of ivermectin that killed 50% (LC50), 25% (LC25) and 5% (LC5) of mosquitoes. To examine ivermectin sporonticidal effect, Plasmodium vivax blood samples were collected from malaria patients and offered to mosquitoes without or with ivermectin at the LC50, LC25 or LC5. To assess ivermectin inhibition of mosquito time to re-feed, concentrations of ivermectin predicted to occur after a single oral dose of 200 μg/kg ivermectin were fed to An. darlingi. Every day for 12 days thereafter, individual mosquitoes were given the opportunity to re-feed on a volunteer. Any mosquitoes that re-blood fed or died were removed from the study. RESULTS Ivermectin significantly reduced An. darlingi survivorship: 7-day-LC50 = 43.2 ng/ml [37.5, 48.6], -LC25 = 27.8 ng/ml [20.4, 32.9] and -LC5 = 14.8 ng/ml [7.9, 20.2]. Ivermectin compound was sporontocidal to P. vivax in An. darlingi at the LC50 and LC25 concentrations reducing prevalence by 22.6 and 17.1%, respectively, but not at the LC5. Oocyst intensity was not altered at any concentration. Ivermectin significantly delayed time to re-feed at the 4-h (48.7 ng/ml) and 12-h (26.9 ng/ml) concentrations but not 36-h (10.6 ng/ml) or 60-h (6.3 ng/ml). CONCLUSIONS Ivermectin is lethal to An. darlingi, modestly inhibits sporogony of P. vivax, and delays time to re-feed at concentrations found in humans up to 12 h post drug ingestion. The LC50 value suggests that a higher than standard dose (400-μg/kg) is necessary to target An. darlingi. These results suggest that ivermectin MDA has potential in the Amazon region to aid malaria elimination efforts.
Collapse
Affiliation(s)
- Kevin C Kobylinski
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, 315/6 Rajvithi Road, Bangkok, 10400, Thailand. .,Entomology Branch, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD, 20910, USA.
| | - Karín S Escobedo-Vargas
- Department of Entomology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| | - Victor M López-Sifuentes
- Department of Entomology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| | - Salomón Durand
- Department of Parasitology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| | - Edward S Smith
- Department of Parasitology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| | - G Christian Baldeviano
- Department of Parasitology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| | - Robert V Gerbasi
- Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - Sara-Blythe Ballard
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Rm. W5515, Baltimore, MD, 21205, USA
| | - Craig A Stoops
- Department of Entomology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| | - Gissella M Vásquez
- Department of Entomology, U.S. Naval Medical Research Unit No. 6, Av. Venezuela block 36 s/n, Callao 2, Peru
| |
Collapse
|
47
|
Abstract
This paper summarises key advances and priorities since the 2011 presentation of the Malaria Eradication Research Agenda (malERA), with a focus on the combinations of intervention tools and strategies for elimination and their evaluation using modelling approaches. With an increasing number of countries embarking on malaria elimination programmes, national and local decisions to select combinations of tools and deployment strategies directed at malaria elimination must address rapidly changing transmission patterns across diverse geographic areas. However, not all of these approaches can be systematically evaluated in the field. Thus, there is potential for modelling to investigate appropriate 'packages' of combined interventions that include various forms of vector control, case management, surveillance, and population-based approaches for different settings, particularly at lower transmission levels. Modelling can help prioritise which intervention packages should be tested in field studies, suggest which intervention package should be used at a particular level or stratum of transmission intensity, estimate the risk of resurgence when scaling down specific interventions after local transmission is interrupted, and evaluate the risk and impact of parasite drug resistance and vector insecticide resistance. However, modelling intervention package deployment against a heterogeneous transmission background is a challenge. Further validation of malaria models should be pursued through an iterative process, whereby field data collected with the deployment of intervention packages is used to refine models and make them progressively more relevant for assessing and predicting elimination outcomes.
Collapse
|
48
|
Abstract
Since the turn of the century, a remarkable expansion has been achieved in the range and effectiveness of products and strategies available to prevent, treat, and control malaria, including advances in diagnostics, drugs, vaccines, and vector control. These advances have once again put malaria elimination on the agenda. However, it is clear that even with the means available today, malaria control and elimination pose a formidable challenge in many settings. Thus, currently available resources must be used more effectively, and new products and approaches likely to achieve these goals must be developed. This paper considers tools (both those available and others that may be required) to achieve and maintain malaria elimination. New diagnostics are needed to direct treatment and detect transmission potential; new drugs and vaccines to overcome existing resistance and protect against clinical and severe disease, as well as block transmission and prevent relapses; and new vector control measures to overcome insecticide resistance and more powerfully interrupt transmission. It is also essential that strategies for combining new and existing approaches are developed for different settings to maximise their longevity and effectiveness in areas with continuing transmission and receptivity. For areas where local elimination has been recently achieved, understanding which measures are needed to maintain elimination is necessary to prevent rebound and the reestablishment of transmission. This becomes increasingly important as more countries move towards elimination.
Collapse
|
49
|
Cytochrome P450/ABC transporter inhibition simultaneously enhances ivermectin pharmacokinetics in the mammal host and pharmacodynamics in Anopheles gambiae. Sci Rep 2017; 7:8535. [PMID: 28819225 PMCID: PMC5561046 DOI: 10.1038/s41598-017-08906-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/14/2017] [Indexed: 01/06/2023] Open
Abstract
Mass administration of endectocides, drugs that kill blood-feeding arthropods, has been proposed as a complementary strategy to reduce malaria transmission. Ivermectin is one of the leading candidates given its excellent safety profile. Here we provide proof that the effect of ivermectin can be boosted at two different levels by drugs inhibiting the cytochrome or ABC transporter in the mammal host and the target mosquitoes. Using a mini-pig model, we show that drug-mediated cytochrome P450/ABC transporter inhibition results in a 3-fold increase in the time ivermectin remains above mosquito-killing concentrations. In contrast, P450/ABC transporter induction with rifampicin markedly impaired ivermectin absorption. The same ketoconazole-mediated cytochrome/ABC transporter inhibition also occurs outside the mammal host and enhances the mortality of Anopheles gambiae. This was proven by using the samples from the mini-pig experiments to conduct an ex-vivo synergistic bioassay by membrane-feeding Anopheles mosquitoes. Inhibiting the same cytochrome/xenobiotic pump complex in two different organisms to simultaneously boost the pharmacokinetic and pharmacodynamic activity of a drug is a novel concept that could be applied to other systems. Although the lack of a dose-response effect in the synergistic bioassay warrants further exploration, our study may have broad implications for the control of parasitic and vector-borne diseases.
Collapse
|
50
|
Ivermectin susceptibility and sporontocidal effect in Greater Mekong Subregion Anopheles. Malar J 2017; 16:280. [PMID: 28687086 PMCID: PMC5501099 DOI: 10.1186/s12936-017-1923-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/28/2017] [Indexed: 12/26/2022] Open
Abstract
Background Novel vector control methods that can directly target outdoor malaria transmission are urgently needed in the Greater Mekong Subregion (GMS) to accelerate malaria elimination and artemisinin resistance containment efforts. Ivermectin mass drug administration (MDA) to humans has been shown to effectively kill wild Anopheles and suppress malaria transmission in West Africa. Preliminary laboratory investigations were performed to determine ivermectin susceptibility and sporontocidal effect in GMS Anopheles malaria vectors coupled with pharmacokinetic models of ivermectin at escalating doses. Methods A population-based pharmacokinetic model of ivermectin was developed using pre-existing data from a clinical trial conducted in Thai volunteers at the 200 µg/kg dose. To assess ivermectin susceptibility, various concentrations of ivermectin compound were mixed in human blood meals and blood-fed to Anopheles dirus, Anopheles minimus, Anopheles sawadwongporni, and Anopheles campestris. Mosquito survival was monitored daily for 7 days and a non-linear mixed effects model with probit analyses was used to calculate concentrations of ivermectin that killed 50% (LC50) of mosquitoes for each species. Blood samples were collected from Plasmodium vivax positive patients and offered to mosquitoes with or without ivermectin at the ivermectin LC25 or LC5 for An. dirus and An. minimus. Results The GMS Anopheles displayed a range of susceptibility to ivermectin with species listed from most to least susceptible being An. minimus (LC50 = 16.3 ng/ml) > An. campestris (LC50 = 26.4 ng/ml) = An. sawadwongporni (LC50 = 26.9 ng/ml) > An. dirus (LC50 = 55.6 ng/ml). Mosquito survivorship results, the pharmacokinetic model, and extensive safety data indicated that ivermectin 400 µg/kg is the ideal minimal dose for MDA in the GMS for malaria parasite transmission control. Ivermectin compound was sporontocidal to P. vivax in both An. dirus and An. minimus at the LC25 and LC5 concentrations. Conclusions Ivermectin is lethal to dominant GMS Anopheles malaria vectors and inhibits sporogony of P. vivax at safe human relevant concentrations. The data suggest that ivermectin MDA has potential in the GMS as a vector and transmission blocking control tool to aid malaria elimination efforts. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1923-8) contains supplementary material, which is available to authorized users.
Collapse
|