1
|
Risch F, Kazakov A, Specht S, Pfarr K, Fischer PU, Hoerauf A, Hübner MP. The long and winding road towards new treatments against lymphatic filariasis and onchocerciasis. Trends Parasitol 2024; 40:829-845. [PMID: 39122645 DOI: 10.1016/j.pt.2024.07.005] [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: 05/30/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024]
Abstract
Although lymphatic filariasis and onchocerciasis have been targeted for global elimination, these helminth infections are still a major public health problem across the tropics and subtropics. Despite decades of research, treatment options remain limited and drugs that completely clear the infections, and can be used on a large scale, are still unavailable. In the present review we discuss the strengths and weaknesses of currently available treatments and new ones in development. Novel candidates (corallopyronin A, DNDi-6166, emodepside, and oxfendazole) are currently moving through (pre)clinical development, while the development of two candidates (AWZ1066S and ABBV-4083/flubentylosin) was recently halted. The preclinical R&D pipeline for filarial infections continues to be limited, and recent setbacks highlight the importance of continuous drug discovery and testing.
Collapse
Affiliation(s)
- Frederic Risch
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Alexander Kazakov
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Sabine Specht
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Kenneth Pfarr
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Peter U Fischer
- Division of Infectious Diseases, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany.
| |
Collapse
|
2
|
Njenga SM, Kanyi H, Okoyo C, Githinji E, Mwatele C, Matendechero SH, Omondi WP, Gitahi PN, Owaga C, Onsongo JK, Gass K. Triple-drug therapy with ivermectin, diethylcarbamazine and albendazole for the acceleration of lymphatic filariasis elimination in Kenya: Programmatic implementation and results of the first impact assessment. PLoS Negl Trop Dis 2024; 18:e0011942. [PMID: 38976718 PMCID: PMC11257386 DOI: 10.1371/journal.pntd.0011942] [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: 01/25/2024] [Revised: 07/18/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
The World Health Organization (WHO) endorsed the use of triple-drug mass drug administration (MDA) regimen with ivermectin, diethylcarbamazine (DEC) and albendazole (commonly abbreviated as IDA) to accelerate the elimination of lymphatic filariasis (LF) as a public health problem in settings where onchocerciasis is not co-endemic. The National Programme for Elimination of LF (NPELF) in Kenya was among the first adopters of the IDA-MDA and two annual rounds were provided in 2018 and 2019 to the residents of Lamu County and Jomvu sub-County in the coast region. This study documented the feasibility of successfully delivering the two rounds of IDA-MDA. An operational research study was undertaken to determine efficient sampling strategies, indicators, and the appropriate population groups that could be used for the monitoring and evaluation of LF programs using IDA-MDA for the elimination of the disease as a public health problem. Two cross-sectional surveys were conducted at baseline in 2018 before IDA-MDA and an impact assessment 17 months after the second round of IDA-MDA. The reported epidemiological treatment coverage was at least 80% in all implementation units during each round of IDA-MDA. Blood samples were tested for filarial antigenemia using commercial Filariasis Test Strips (FTS) and any individual found to be positive was tested again at night for the presence of microfilariae in finger prick blood smears using microscopy. The overall prevalence of circulating filarial antigen (CFA) was relatively low at the baseline survey with Jomvu having 1.39% (95% CI: 0.91, 2.11) and Lamu having 0.48% (95% CI: 0.21, 1.13). Significant reduction in CFA prevalence was observed during the impact assessment after the two annual rounds of mass treatment. The overall relative reduction (%) in CFA prevalence following the two rounds of MDA with IDA was significant in both Jomvu (52.45%, Z = -2.46, P < 0.02) and Lamu (52.71%, Z = -1.97, P < 0.05). Heterogeneity, however, was observed in the CFA prevalence reduction between random and purposive clusters, as well as between adult and child populations. The results of the impact assessment survey offered strong evidence that it was safe to stop the IDA-MDA in the two EUs because transmission appears to have been interrupted. It is also important to implement a post-treatment surveillance system which would enable efficient detection of any recrudescence of LF transmission at a sub-evaluation unit level. Our findings show that IDA-MDA may be considered for acceleration of LF elimination in other settings where onchocerciasis is not co-endemic.
Collapse
Affiliation(s)
- Sammy M. Njenga
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | - Henry Kanyi
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | - Collins Okoyo
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
- Department of Epidemiology, Statistics and Informatics, Kenya Medical Research Institute, Nairobi, Kenya
| | - Edward Githinji
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | - Cassian Mwatele
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | | | - Wyckliff P. Omondi
- Vector-Borne and Neglected Tropical Diseases Unit, Ministry of Health, Nairobi, Kenya
| | - Patrick N. Gitahi
- Vector-Borne and Neglected Tropical Diseases Unit, Ministry of Health, Nairobi, Kenya
| | | | | | - Katherine Gass
- NTD Support Center, Task Force for Global Health, Atlanta, Georgia, United States of America
| |
Collapse
|
3
|
Mayfield HJ, Sartorius B, Sheridan S, Howlett M, Martin BM, Thomsen R, Tofaeono-Pifeleti R, Viali S, Graves PM, Lau CL. Ongoing transmission of lymphatic filariasis in Samoa 4.5 years after one round of triple-drug mass drug administration. PLoS Negl Trop Dis 2024; 18:e0012236. [PMID: 38935622 PMCID: PMC11210818 DOI: 10.1371/journal.pntd.0012236] [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: 01/13/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Lymphatic filariasis (LF) remains a significant global issue. To eliminate LF as a public health problem, the World Health Organization (WHO) recommends multiple rounds of mass drug administration (MDA). In certain scenarios, including when elimination targets have not been met with two-drug MDA, triple-drug MDA (using ivermectin, diethylcarbamazine and albendazole) is recommended. In this study, we report on antigen (Ag) and microfilaria (Mf) prevalence in eight primary sampling units (PSUs) in Samoa 4.5 years after one round of triple-drug MDA. METHODOLOGY In 2023, community surveys were conducted in eight PSUs that had been surveyed previously in 2018 (between 1.5 and 3.5 months post triple-drug MDA) and 2019 (six to eight-months post triple-drug MDA). Fifteen houses were randomly selected in each PSU with household members aged ≥ 5 years invited to participate. Blood samples were tested for Ag and Mf. PRINCIPAL FINDINGS Ag-positive participants were observed in six of the eight PSUs, and Ag prevalence was significantly above the 1% threshold in four PSUs. The presence of Mf-positive participants in five PSUs confirms the presence of residual active infections. CONCLUSIONS/SIGNIFICANCE This study provides evidence of persistent LF transmission in Samoa 4.5 years after one round of triple-drug MDA, confirming that one round was insufficient for interruption of transmission in this setting. Our findings highlight the negative impact of delaying MDA rounds, for example, due to public health emergencies.
Collapse
Affiliation(s)
- Helen J. Mayfield
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Benn Sartorius
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Sarah Sheridan
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Maddison Howlett
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Beatris Mario Martin
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | - Satupaitea Viali
- School of Medicine, National University of Samoa, Apia, Samoa
- Oceania University of Medicine Samoa, Apia, Samoa
| | - Patricia M. Graves
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Queensland, Australia
| | - Colleen L. Lau
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
4
|
Vasconcelos A, King JD, Nunes-Alves C, Anderson R, Argaw D, Basáñez MG, Bilal S, Blok DJ, Blumberg S, Borlase A, Brady OJ, Browning R, Chitnis N, Coffeng LE, Crowley EH, Cucunubá ZM, Cummings DAT, Davis CN, Davis EL, Dixon M, Dobson A, Dyson L, French M, Fronterre C, Giorgi E, Huang CI, Jain S, James A, Kim SH, Kura K, Lucianez A, Marks M, Mbabazi PS, Medley GF, Michael E, Montresor A, Mutono N, Mwangi TS, Rock KS, Saboyá-Díaz MI, Sasanami M, Schwehm M, Spencer SEF, Srivathsan A, Stawski RS, Stolk WA, Sutherland SA, Tchuenté LAT, de Vlas SJ, Walker M, Brooker SJ, Hollingsworth TD, Solomon AW, Fall IS. Accelerating Progress Towards the 2030 Neglected Tropical Diseases Targets: How Can Quantitative Modeling Support Programmatic Decisions? Clin Infect Dis 2024; 78:S83-S92. [PMID: 38662692 PMCID: PMC11045030 DOI: 10.1093/cid/ciae082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
Over the past decade, considerable progress has been made in the control, elimination, and eradication of neglected tropical diseases (NTDs). Despite these advances, most NTD programs have recently experienced important setbacks; for example, NTD interventions were some of the most frequently and severely impacted by service disruptions due to the coronavirus disease 2019 (COVID-19) pandemic. Mathematical modeling can help inform selection of interventions to meet the targets set out in the NTD road map 2021-2030, and such studies should prioritize questions that are relevant for decision-makers, especially those designing, implementing, and evaluating national and subnational programs. In September 2022, the World Health Organization hosted a stakeholder meeting to identify such priority modeling questions across a range of NTDs and to consider how modeling could inform local decision making. Here, we summarize the outputs of the meeting, highlight common themes in the questions being asked, and discuss how quantitative modeling can support programmatic decisions that may accelerate progress towards the 2030 targets.
Collapse
Affiliation(s)
- Andreia Vasconcelos
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, United Kingdom
- Centre for Global Health Research, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, United Kingdom
| | - Jonathan D King
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Cláudio Nunes-Alves
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, United Kingdom
| | - Roy Anderson
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Daniel Argaw
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Maria-Gloria Basáñez
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Shakir Bilal
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - David J Blok
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Seth Blumberg
- Francis I. Proctor Foundation, University of California, San Francisco, California, USA
| | - Anna Borlase
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Oliver J Brady
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Centre for the Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Raiha Browning
- The Department of Statistics, The University of Warwick, Coventry, United Kingdom
| | - Nakul Chitnis
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Luc E Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emily H Crowley
- Zeeman Institute for System Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Zulma M Cucunubá
- Departamento de Epidemiología Clínica y Bioestadística, Facultad de Medicina, Universidad Pontificia Javeriana, Bogotá, Colombia
| | - Derek A T Cummings
- Department of Biology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Christopher Neil Davis
- Zeeman Institute for System Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Emma Louise Davis
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Matthew Dixon
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Andrew Dobson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Louise Dyson
- Zeeman Institute for System Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Michael French
- Schistosomiasis Control Initiative, Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, United Kingdom
- RTI International, Washington, D.C., USA
| | - Claudio Fronterre
- CHICAS, Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
| | - Emanuele Giorgi
- CHICAS, Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
| | - Ching-I Huang
- Zeeman Institute for System Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Saurabh Jain
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Ananthu James
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sung Hye Kim
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Klodeta Kura
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Ana Lucianez
- Communicable Diseases Prevention, Control, and Elimination, Pan American Health Organization, Washington D.C., USA
| | - Michael Marks
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Pamela Sabina Mbabazi
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Graham F Medley
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Edwin Michael
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Antonio Montresor
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Nyamai Mutono
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Thumbi S Mwangi
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Kat S Rock
- Zeeman Institute for System Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Martha-Idalí Saboyá-Díaz
- Communicable Diseases Prevention, Control, and Elimination, Pan American Health Organization, Washington D.C., USA
| | - Misaki Sasanami
- Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
| | - Markus Schwehm
- ExploSYS GmbH, Interdisciplinary Institute for Exploratory Systems, Leinfelden-Echterdingen, Germany
| | - Simon E F Spencer
- Centre for the Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ariktha Srivathsan
- Francis I. Proctor Foundation, University of California, San Francisco, California, USA
| | - Robert S Stawski
- Institute of Public Health and Wellbeing, School of Health and Social Care, University of Essex, Essex, United Kingdom
| | - Wilma A Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Samuel A Sutherland
- Zeeman Institute for System Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Warwick Medical School, The University of Warwick, Coventry, United Kingdom
| | | | - Sake J de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Martin Walker
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | | | - T Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, United Kingdom
| | - Anthony W Solomon
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| | - Ibrahima Socé Fall
- Global Neglected Tropical Diseases Programme, World Health Organization, Geneva, Switzerland
| |
Collapse
|
5
|
Touloupou P, Fronterre C, Cano J, Prada JM, Smith M, Kontoroupis P, Brown P, Rivera RC, de Vlas SJ, Gunawardena S, Irvine MA, Njenga SM, Reimer L, Seife F, Sharma S, Michael E, Stolk WA, Pulan R, Spencer SEF, Hollingsworth TD. An Ensemble Framework for Projecting the Impact of Lymphatic Filariasis Interventions Across Sub-Saharan Africa at a Fine Spatial Scale. Clin Infect Dis 2024; 78:S108-S116. [PMID: 38662704 PMCID: PMC11045016 DOI: 10.1093/cid/ciae071] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Lymphatic filariasis (LF) is a neglected tropical disease targeted for elimination as a public health problem by 2030. Although mass treatments have led to huge reductions in LF prevalence, some countries or regions may find it difficult to achieve elimination by 2030 owing to various factors, including local differences in transmission. Subnational projections of intervention impact are a useful tool in understanding these dynamics, but correctly characterizing their uncertainty is challenging. METHODS We developed a computationally feasible framework for providing subnational projections for LF across 44 sub-Saharan African countries using ensemble models, guided by historical control data, to allow assessment of the role of subnational heterogeneities in global goal achievement. Projected scenarios include ongoing annual treatment from 2018 to 2030, enhanced coverage, and biannual treatment. RESULTS Our projections suggest that progress is likely to continue well. However, highly endemic locations currently deploying strategies with the lower World Health Organization recommended coverage (65%) and frequency (annual) are expected to have slow decreases in prevalence. Increasing intervention frequency or coverage can accelerate progress by up to 5 or 6 years, respectively. CONCLUSIONS While projections based on baseline data have limitations, our methodological advancements provide assessments of potential bottlenecks for the global goals for LF arising from subnational heterogeneities. In particular, areas with high baseline prevalence may face challenges in achieving the 2030 goals, extending the "tail" of interventions. Enhancing intervention frequency and/or coverage will accelerate progress. Our approach facilitates preimplementation assessments of the impact of local interventions and is applicable to other regions and neglected tropical diseases.
Collapse
Affiliation(s)
| | | | - Jorge Cano
- Expanded Special Project for Elimination of Neglected Tropical Diseases (ESPEN), WHO Regional Office for Africa, Brazzaville, Democratic Republic of the Congo
| | - Joaquin M Prada
- School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Morgan Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | | - Paul Brown
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Rocio Caja Rivera
- Center for Global Health Infectious Disease Research, University of South Florida, Tampa, USA
| | - Sake J de Vlas
- Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Michael A Irvine
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, Canada
| | - Sammy M Njenga
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Lisa Reimer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Fikre Seife
- Disease Prevention and Control Directorate, Federal Ministry of Health, Addis Ababa, Ethiopia
| | - Swarnali Sharma
- Department of Mathematics, Vijaygarh Jyotish Ray College, Kolkata, India
| | - Edwin Michael
- Center for Global Health Infectious Disease Research, University of South Florida, Tampa, USA
| | - Wilma A Stolk
- Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Pulan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Simon E F Spencer
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - T Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
James A, Coffeng LE, Blok DJ, King JD, de Vlas SJ, Stolk WA. Predictive Value of Microfilariae-Based Stop-MDA Thresholds After Triple Drug Therapy With IDA Against Lymphatic Filariasis in Treatment-Naive Indian Settings. Clin Infect Dis 2024; 78:S131-S137. [PMID: 38662696 PMCID: PMC11045019 DOI: 10.1093/cid/ciae019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
Mass drug administration (MDA) of antifilarial drugs is the main strategy for the elimination of lymphatic filariasis (LF). Recent clinical trials indicated that the triple-drug therapy with ivermectin, diethylcarbamazine, and albendazole (IDA) is much more effective against LF than the widely used two-drug combinations (albendazole plus either ivermectin or diethylcarbamazine). For IDA-based MDA, the stop-MDA decision is made based on microfilariae (mf) prevalence in adults. In this study, we assess how the probability of eventually reaching elimination of transmission depends on the critical threshold used in transmission assessment surveys (TAS-es) to define whether transmission was successfully suppressed and triple-drug MDA can be stopped. This analysis focuses on treatment-naive Indian settings. We do this for a range of epidemiological and programmatic contexts, using the established LYMFASIM model for transmission and control of LF. Based on our simulations, a single TAS, one year after the last MDA round, provides limited predictive value of having achieved suppressed transmission, while a higher MDA coverage increases elimination probability, thus leading to a higher predictive value. Every additional TAS, conditional on previous TAS-es being passed with the same threshold, further improves the predictive value for low values of stop-MDA thresholds. An mf prevalence threshold of 0.5% corresponding to TAS-3 results in ≥95% predictive value even when the MDA coverage is relatively low.
Collapse
Affiliation(s)
- Ananthu James
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Luc E Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - David J Blok
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jonathan D King
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Sake J de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Wilma A Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Prada JM, Touloupou P, Kebede B, Giorgi E, Sime H, Smith M, Kontoroupis P, Brown P, Cano J, Farkas H, Irvine M, Reimer L, Caja Rivera R, de Vlas SJ, Michael E, Stolk WA, Pulan R, Spencer SEF, Hollingsworth TD, Seife F. Subnational Projections of Lymphatic Filariasis Elimination Targets in Ethiopia to Support National Level Policy. Clin Infect Dis 2024; 78:S117-S125. [PMID: 38662702 PMCID: PMC11045027 DOI: 10.1093/cid/ciae072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Lymphatic filariasis (LF) is a debilitating, poverty-promoting, neglected tropical disease (NTD) targeted for worldwide elimination as a public health problem (EPHP) by 2030. Evaluating progress towards this target for national programmes is challenging, due to differences in disease transmission and interventions at the subnational level. Mathematical models can help address these challenges by capturing spatial heterogeneities and evaluating progress towards LF elimination and how different interventions could be leveraged to achieve elimination by 2030. METHODS Here we used a novel approach to combine historical geo-spatial disease prevalence maps of LF in Ethiopia with 3 contemporary disease transmission models to project trends in infection under different intervention scenarios at subnational level. RESULTS Our findings show that local context, particularly the coverage of interventions, is an important determinant for the success of control and elimination programmes. Furthermore, although current strategies seem sufficient to achieve LF elimination by 2030, some areas may benefit from the implementation of alternative strategies, such as using enhanced coverage or increased frequency, to accelerate progress towards the 2030 targets. CONCLUSIONS The combination of geospatial disease prevalence maps of LF with transmission models and intervention histories enables the projection of trends in infection at the subnational level under different control scenarios in Ethiopia. This approach, which adapts transmission models to local settings, may be useful to inform the design of optimal interventions at the subnational level in other LF endemic regions.
Collapse
Affiliation(s)
- Joaquin M Prada
- Department of Comparative Biomedical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | - Biruck Kebede
- RTI International, 3040 E Cornwallis Rd, Research Triangle Park, North Carolina 27709, USA
| | | | - Heven Sime
- Malaria and Neglected Tropical Diseases Research Team, Bacterial, Parasitic and Zoonotic Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Morgan Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | | - Paul Brown
- Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Jorge Cano
- Expanded Special Project for Elimination of Neglected Tropical Diseases (ESPEN), WHO Regional Office for Africa, Brazzaville, Democratic Republic of the Congo
| | - Hajnal Farkas
- Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Mike Irvine
- Faculty of Science, BC Centre for Disease Control, Vancouver, Canada
| | - Lisa Reimer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Rocio Caja Rivera
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Sake J de Vlas
- Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Edwin Michael
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Wilma A Stolk
- Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Pulan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Simon E F Spencer
- Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - T Déirdre Hollingsworth
- Nuffield Department of Medicine, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Fikre Seife
- Disease Prevention and Control Directorate, Federal Ministry of Health, Addis Ababa, Ethiopia
| |
Collapse
|
8
|
Kura K, Stolk WA, Basáñez MG, Collyer BS, de Vlas SJ, Diggle PJ, Gass K, Graham M, Hollingsworth TD, King JD, Krentel A, Anderson RM, Coffeng LE. How Does the Proportion of Never Treatment Influence the Success of Mass Drug Administration Programs for the Elimination of Lymphatic Filariasis? Clin Infect Dis 2024; 78:S93-S100. [PMID: 38662701 PMCID: PMC11045024 DOI: 10.1093/cid/ciae021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Mass drug administration (MDA) is the cornerstone for the elimination of lymphatic filariasis (LF). The proportion of the population that is never treated (NT) is a crucial determinant of whether this goal is achieved within reasonable time frames. METHODS Using 2 individual-based stochastic LF transmission models, we assess the maximum permissible level of NT for which the 1% microfilaremia (mf) prevalence threshold can be achieved (with 90% probability) within 10 years under different scenarios of annual MDA coverage, drug combination and transmission setting. RESULTS For Anopheles-transmission settings, we find that treating 80% of the eligible population annually with ivermectin + albendazole (IA) can achieve the 1% mf prevalence threshold within 10 years of annual treatment when baseline mf prevalence is 10%, as long as NT <10%. Higher proportions of NT are acceptable when more efficacious treatment regimens are used. For Culex-transmission settings with a low (5%) baseline mf prevalence and diethylcarbamazine + albendazole (DA) or ivermectin + diethylcarbamazine + albendazole (IDA) treatment, elimination can be reached if treatment coverage among eligibles is 80% or higher. For 10% baseline mf prevalence, the target can be achieved when the annual coverage is 80% and NT ≤15%. Higher infection prevalence or levels of NT would make achieving the target more difficult. CONCLUSIONS The proportion of people never treated in MDA programmes for LF can strongly influence the achievement of elimination and the impact of NT is greater in high transmission areas. This study provides a starting point for further development of criteria for the evaluation of NT.
Collapse
Affiliation(s)
- Klodeta Kura
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, United Kingdom
| | - Wilma A Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maria-Gloria Basáñez
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, United Kingdom
| | - Benjamin S Collyer
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, United Kingdom
| | - Sake J de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter J Diggle
- Centre for Health Informatics, Computing and Statistics, Lancaster University, Lancaster, United Kingdom
| | - Katherine Gass
- Neglected Tropical Diseases Support Center, Task Force for Global Health, Decatur, Georgia, USA
| | - Matthew Graham
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Centre for Mathematical Modelling of Infectious Disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - T Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Jonathan D King
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Alison Krentel
- Bruyère Research Institute, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Canada
| | - Roy M Anderson
- London Centre for Neglected Tropical Disease Research, London, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, United Kingdom
| | - Luc E Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Antony Oliver MC, Graham M, Gass KM, Medley GF, Clark J, Davis EL, Reimer LJ, King JD, Pouwels KB, Hollingsworth TD. Reducing the Antigen Prevalence Target Threshold for Stopping and Restarting Mass Drug Administration for Lymphatic Filariasis Elimination: A Model-Based Cost-effectiveness Simulation in Tanzania, India and Haiti. Clin Infect Dis 2024; 78:S160-S168. [PMID: 38662697 PMCID: PMC11045020 DOI: 10.1093/cid/ciae108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The Global Programme to Eliminate Lymphatic Filariasis (GPELF) aims to reduce and maintain infection levels through mass drug administration (MDA), but there is evidence of ongoing transmission after MDA in areas where Culex mosquitoes are the main transmission vector, suggesting that a more stringent criterion is required for MDA decision making in these settings. METHODS We use a transmission model to investigate how a lower prevalence threshold (<1% antigenemia [Ag] prevalence compared with <2% Ag prevalence) for MDA decision making would affect the probability of local elimination, health outcomes, the number of MDA rounds, including restarts, and program costs associated with MDA and surveys across different scenarios. To determine the cost-effectiveness of switching to a lower threshold, we simulated 65% and 80% MDA coverage of the total population for different willingness to pay per disability-adjusted life-year averted for India ($446.07), Tanzania ($389.83), and Haiti ($219.84). RESULTS Our results suggest that with a lower Ag threshold, there is a small proportion of simulations where extra rounds are required to reach the target, but this also reduces the need to restart MDA later in the program. For 80% coverage, the lower threshold is cost-effective across all baseline prevalences for India, Tanzania, and Haiti. For 65% MDA coverage, the lower threshold is not cost-effective due to additional MDA rounds, although it increases the probability of local elimination. Valuing the benefits of elimination to align with the GPELF goals, we find that a willingness to pay per capita government expenditure of approximately $1000-$4000 for 1% increase in the probability of local elimination would be required to make a lower threshold cost-effective. CONCLUSIONS Lower Ag thresholds for stopping MDAs generally mean a higher probability of local elimination, reducing long-term costs and health impacts. However, they may also lead to an increased number of MDA rounds required to reach the lower threshold and, therefore, increased short-term costs. Collectively, our analyses highlight that lower target Ag thresholds have the potential to assist programs in achieving lymphatic filariasis goals.
Collapse
Affiliation(s)
- Mary Chriselda Antony Oliver
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Matthew Graham
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Katherine M Gass
- Neglected Tropical Diseases Support Centre, The Task Force for Global Health, Decatur, Georgia, USA
| | - Graham F Medley
- Centre for Mathematical Modelling of Infectious Disease and Department of Global Health and Development, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jessica Clark
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Emma L Davis
- Mathematics Institute and the Zeeman Institute for Systems Biology and Infectious Disease Epidemiological Research, University of Warwick, Coventry, United Kingdom
| | - Lisa J Reimer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jonathan D King
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Koen B Pouwels
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - T Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Voronin D, Tricoche N, Peguero R, Kaminska AM, Ghedin E, Sakanari JA, Lustigman S. Repurposed Drugs That Activate Autophagy in Filarial Worms Act as Effective Macrofilaricides. Pharmaceutics 2024; 16:256. [PMID: 38399310 PMCID: PMC10891619 DOI: 10.3390/pharmaceutics16020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Onchocerciasis and lymphatic filariasis are two neglected tropical diseases caused by filarial nematodes that utilize insect vectors for transmission to their human hosts. Current control strategies are based on annual or biannual mass drug administration (MDA) of the drugs Ivermectin or Ivermectin plus Albendazole, respectively. These drug regimens kill the first-stage larvae of filarial worms (i.e., microfilariae) and interrupt the transmission of infections. MDA programs for these microfilaricidal drugs must be given over the lifetime of the filarial adult worms, which can reach 15 years in the case of Onchocerca volvulus. This is problematic because of suboptimal responses to ivermectin in various endemic regions and inefficient reduction of transmission even after decades of MDA. There is an urgent need for the development of novel alternative treatments to support the 2030 elimination goals of onchocerciasis and lymphatic filariasis. One successful approach has been to target Wolbachia, obligatory endosymbiotic bacteria on which filarial worms are dependent for their survival and reproduction within the human host. A 4-6-week antibiotic therapy with doxycycline, for example, resulted in the loss of Wolbachia that subsequently led to extensive apoptosis of somatic cells, germline, embryos, and microfilariae, as well as inhibition of fourth-stage larval development. However, this long-course regimen has limited use in MDA programs. As an alternative approach to the use of bacteriostatic antibiotics, in this study, we focused on autophagy-inducing compounds, which we hypothesized could disturb various pathways involved in the interdependency between Wolbachia and filarial worms. We demonstrated that several such compounds, including Niclosamide, an FDA-approved drug, Niclosamide ethanolamine (NEN), and Rottlerin, a natural product derived from Kamala trees, significantly reduced the levels of Wolbachia in vitro. Moreover, when these compounds were used in vivo to treat Brugia pahangi-infected gerbils, Niclosamide and NEN significantly decreased adult worm survival, reduced the release of microfilariae, and decreased embryonic development depending on the regimen and dose used. All three drugs given orally significantly reduced Wolbachia loads and induced an increase in levels of lysosome-associated membrane protein in worms from treated animals, suggesting that Niclosamide, NEN, and Rottlerin were effective in causing drug-induced autophagy in these filarial worms. These repurposed drugs provide a new avenue for the clearance of adult worms in filarial infections.
Collapse
Affiliation(s)
- Denis Voronin
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20892, USA;
| | - Nancy Tricoche
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Ricardo Peguero
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Anna Maria Kaminska
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20892, USA;
| | - Judy A. Sakanari
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA;
| | - Sara Lustigman
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| |
Collapse
|
11
|
Freitas LT, Khan MA, Uddin A, Halder JB, Singh-Phulgenda S, Raja JD, Balakrishnan V, Harriss E, Rahi M, Brack M, Guérin PJ, Basáñez MG, Kumar A, Walker M, Srividya A. The lymphatic filariasis treatment study landscape: A systematic review of study characteristics and the case for an individual participant data platform. PLoS Negl Trop Dis 2024; 18:e0011882. [PMID: 38227595 PMCID: PMC10817204 DOI: 10.1371/journal.pntd.0011882] [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: 09/05/2023] [Revised: 01/26/2024] [Accepted: 12/22/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Lymphatic filariasis (LF) is a neglected tropical disease (NTD) targeted by the World Health Organization for elimination as a public health problem (EPHP). Since 2000, more than 9 billion treatments of antifilarial medicines have been distributed through mass drug administration (MDA) programmes in 72 endemic countries and 17 countries have reached EPHP. Yet in 2021, nearly 900 million people still required MDA with combinations of albendazole, diethylcarbamazine and/or ivermectin. Despite the reliance on these drugs, there remain gaps in understanding of variation in responses to treatment. As demonstrated for other infectious diseases, some urgent questions could be addressed by conducting individual participant data (IPD) meta-analyses. Here, we present the results of a systematic literature review to estimate the abundance of IPD on pre- and post-intervention indicators of infection and/or morbidity and assess the feasibility of building a global data repository. METHODOLOGY We searched literature published between 1st January 2000 and 5th May 2023 in 15 databases to identify prospective studies assessing LF treatment and/or morbidity management and disease prevention (MMDP) approaches. We considered only studies where individual participants were diagnosed with LF infection or disease and were followed up on at least one occasion after receiving an intervention/treatment. PRINCIPAL FINDINGS We identified 138 eligible studies from 23 countries, having followed up an estimated 29,842 participants after intervention. We estimate 14,800 (49.6%) IPD on pre- and post-intervention infection indicators including microfilaraemia, circulating filarial antigen and/or ultrasound indicators measured before and after intervention using 8 drugs administered in various combinations. We identified 33 studies on MMDP, estimating 6,102 (20.4%) IPD on pre- and post-intervention clinical morbidity indicators only. A further 8,940 IPD cover a mixture of infection and morbidity outcomes measured with other diagnostics, from participants followed for adverse event outcomes only or recruited after initial intervention. CONCLUSIONS The LF treatment study landscape is heterogeneous, but the abundance of studies and related IPD suggest that establishing a global data repository to facilitate IPD meta-analyses would be feasible and useful to address unresolved questions on variation in treatment outcomes across geographies, demographics and in underrepresented groups. New studies using more standardized approaches should be initiated to address the scarcity and inconsistency of data on morbidity management.
Collapse
Affiliation(s)
- Luzia T. Freitas
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
| | | | - Azhar Uddin
- ICMR-Vector Control Research Centre, Puducherry, India
| | - Julia B. Halder
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Sauman Singh-Phulgenda
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | - Eli Harriss
- The Knowledge Centre, Bodleian Health Care Libraries, University of Oxford, Oxford, United Kingdom
| | - Manju Rahi
- ICMR-Vector Control Research Centre, Puducherry, India
| | - Matthew Brack
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Philippe J. Guérin
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Maria-Gloria Basáñez
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
| | - Ashwani Kumar
- Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Martin Walker
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- Infectious Diseases Data Observatory, University of Oxford, Oxford, United Kingdom
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | | |
Collapse
|
12
|
Clark J, Davis EL, Prada JM, Gass K, Krentel A, Hollingsworth TD. How correlations between treatment access and surveillance inclusion impact neglected tropical disease monitoring and evaluation-A simulated study. PLoS Negl Trop Dis 2023; 17:e0011582. [PMID: 37672518 PMCID: PMC10506705 DOI: 10.1371/journal.pntd.0011582] [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: 09/08/2022] [Revised: 09/18/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
Neglected tropical diseases (NTDs) largely impact marginalised communities living in tropical and subtropical regions. Mass drug administration is the leading intervention method for five NTDs; however, it is known that there is lack of access to treatment for some populations and demographic groups. It is also likely that those individuals without access to treatment are excluded from surveillance. It is important to consider the impacts of this on the overall success, and monitoring and evaluation (M&E) of intervention programmes. We use a detailed individual-based model of the infection dynamics of lymphatic filariasis to investigate the impact of excluded, untreated, and therefore unobserved groups on the true versus observed infection dynamics and subsequent intervention success. We simulate surveillance in four groups-the whole population eligible to receive treatment, the whole eligible population with access to treatment, the TAS focus of six- and seven-year-olds, and finally in >20-year-olds. We show that the surveillance group under observation has a significant impact on perceived dynamics. Exclusion to treatment and surveillance negatively impacts the probability of reaching public health goals, though in populations that do reach these goals there are no signals to indicate excluded groups. Increasingly restricted surveillance groups over-estimate the efficacy of MDA. The presence of non-treated groups cannot be inferred when surveillance is only occurring in the group receiving treatment.
Collapse
Affiliation(s)
- Jessica Clark
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, Scotland
- Big Data Institute, Neglected Tropical Disease Modelling Consortium, University of Oxford, Oxford, England
| | - Emma L. Davis
- Big Data Institute, Neglected Tropical Disease Modelling Consortium, University of Oxford, Oxford, England
| | - Joaquin M. Prada
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, England
| | - Katherine Gass
- Neglected Tropical Diseases Support Center, Task Force for Global Health, Decatur, Georgia, United States of America
| | - Alison Krentel
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
- Bruyère Research Institute, Ottawa, Canada
| | - T. Déirdre Hollingsworth
- Big Data Institute, Neglected Tropical Disease Modelling Consortium, University of Oxford, Oxford, England
| |
Collapse
|
13
|
Dinesh RJ, Srividya A, Subramanian S, Krishnamoorthy K, Sabesan S, Raghorte MC, Kumar A, Jambulingam P. Coverage evaluation of mass drug administration with triple drug regimen in an evaluation unit in Nagpur district of Maharashtra, India. PLoS Negl Trop Dis 2023; 17:e0011588. [PMID: 37676897 PMCID: PMC10484419 DOI: 10.1371/journal.pntd.0011588] [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: 08/17/2022] [Accepted: 08/11/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Triple drug regimen (IDA; Ivermectin, Diethylcarbamazine, Albendazole) recommended for accelerating elimination of lymphatic filariasis was launched in India in December 2018. Nagpur district in Maharashtra was one of the first five districts where this strategy was introduced. The National Vector Borne Disease Control Programme (NVBDCP) at the district reported ~85.0% treatment coverage in the first round of mass drug administration (MDA) with IDA implemented in EU-2 in Nagpur district in January 2019. As per the national guideline, a coverage evaluation survey was carried out and both quantitative and qualitative data were collected to assess the treatment coverage, the level of community preparation and identify the gaps, if any, for improvement. METHODOLOGY A Coverage Evaluation Survey (CES) following the WHO recommended protocol was conducted in one of the two evaluation units (EU-2) in Nagpur district in March 2019. Coverage Sample Builder (CSB) V2.9 tool was used to calculate the sample size, select sites and estimate drug coverage. The CSB tool followed a two-stage cluster sampling procedure to select 30 primary sampling units (ward/village as a cluster) and a list of random numbers for selecting households (HHs) in each cluster. The results were analyzed for operational indicators. Stata ver. 14.0 software was used to construct the 95% confidence limits accounting for clustering. RESULTS A total of 1601 individuals aged 5-85 years of both gender from 328 HHs were surveyed from the 30 randomly selected clusters in EU-2. The mean age was 33.8±17.6 years. Among the surveyed population, 78.0% received the drugs (programme reach) and 66.1% consumed the drugs (survey coverage). Survey coverage was significantly higher in rural (82.6%) than in urban (59.4%) and peri-urban (58.6%) areas (P<0.001). Directly observed treatment (DOT) among the surveyed population was 51.6%. Adverse events were reported among 6.9% respondents who reported to have consumed the drugs. CONCLUSION The IDA based MDA strategy could achieve just the required level of treatment coverage (~65%) in EU-2, Nagpur district, which had previously undergone several rounds of DA-MDAs (Diethylcarbamazine, Albendazole). Having achieved an effective treatment coverage of >80% in rural areas, the coverage in urban and peri-urban areas need to be improved in order to attain the impact of IDA-MDA. It is imperative to strengthen drug delivery and community preparation activities along with improved DOT especially in urban and peri-urban areas to achieve the required level of treatment coverage. Addition of ivermectin did not have any additional perceived adverse events.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ashwani Kumar
- ICMR-Vector Control Research Centre, Puducherry, India
| | | |
Collapse
|
14
|
Chhonker YS, Bjerum C, Bala V, Ouattara AF, Koudou BG, Gabo TP, Alshehri A, Meïté A, Fischer PU, Weil GJ, King CL, Budge PJ, Murry DJ. Pharmacokinetics of Moxidectin combined with Albendazole or Albendazole plus Diethylcarbamazine for Bancroftian Filariasis. PLoS Negl Trop Dis 2023; 17:e0011567. [PMID: 37616301 PMCID: PMC10482275 DOI: 10.1371/journal.pntd.0011567] [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: 02/24/2023] [Revised: 09/06/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Moxidectin (MOX) is a milbemycin endectocide recently approved by the U.S. FDA for the treatment of onchocerciasis in persons at least 12 years of age. MOX has been shown to have a good safety profile in recent clinical trials. The efficacy of MOX for the treatment of lymphatic filariasis (LF) and its potential use in mass drug administration protocols for the elimination of LF is currently under evaluation. In the context of a clinical trial, we investigated the pharmacokinetics and drug interactions of a combination of MOX plus albendazole (ALB) with or without diethylcarbamazine (DEC) compared to ivermectin (IVM) plus ALB with or without DEC in the following four different treatment arms: (I) IVM (0.2mg/kg) plus DEC (6 mg/kg) and ALB (400mg); (II) IVM plus ALB; (III) MOX (8 mg) plus DEC and ALB; and (IV) MOX plus ALB. Drug concentrations were determined using validated liquid chromatography-mass spectrometric methods. Pharmacokinetic parameters were determined using standard non-compartmental analysis methods. Statistical analysis was performed using JMP software. Fifty-eight of 164 study participants (53 men and five women) were included with ages ranging from 18 to 63 yrs (mean = 37). MOX apparent oral clearance (Cl/F) ranged from 0.7 to 10.8 L/hr with Cmax values ranging from 20.8 to 314.5 ng/mL. The mean (range) area under the curve (AUC)0-∞ for MOX, 3405 ng*hr/mL (742-11376), and IVM 1906 ng*hr/mL (692-5900), varied over a ~15.3 and ~8.5-fold range, respectively. The geometric mean ratio for Cmax, AUC0-t, and AUC0-∞ were within the no-drug interaction range of 80-125% for all drugs. This indicates that the addition of MOX to ALB alone or ALB plus DEC for LF therapy did not alter the drug exposure of co-administered drugs compared to IVM combinations. Clinical Trial Registration: NCT04410406, https://clinicaltrials.gov/.
Collapse
Affiliation(s)
- Yashpal S. Chhonker
- Clinical Pharmacology Laboratory, Dept of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Catherine Bjerum
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Veenu Bala
- Clinical Pharmacology Laboratory, Dept of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Allassane F. Ouattara
- Centre Suisse de Recherche Scientifique en Côte d’Ivoire (CSRS), Abidjan, Ivory Coast
| | - Benjamin G. Koudou
- Centre Suisse de Recherche Scientifique en Côte d’Ivoire (CSRS), Abidjan, Ivory Coast
| | - Toki P. Gabo
- Centre Hospitalier Regional d’Agboville, Côte d’Ivoire
| | - Abdullah Alshehri
- Clinical Pharmacology Laboratory, Dept of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Abdoulaye Meïté
- Programme National de la Lutte Contre la Schistosomiase, les Geohelminthiases et la Filariose Lymphatique, Ivory Coast
| | - Peter U. Fischer
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gary J. Weil
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Christopher L. King
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Veterans Affairs Research Service, Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Philip J. Budge
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Daryl J. Murry
- Clinical Pharmacology Laboratory, Dept of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
15
|
Risch F, Scheunemann JF, Reichwald JJ, Lenz B, Ehrens A, Gal J, Fercoq F, Koschel M, Fendler M, Hoerauf A, Martin C, Hübner MP. The efficacy of the benzimidazoles oxfendazole and flubendazole against Litomosoides sigmodontis is dependent on the adaptive and innate immune system. Front Microbiol 2023; 14:1213143. [PMID: 37440891 PMCID: PMC10335397 DOI: 10.3389/fmicb.2023.1213143] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/09/2023] [Indexed: 07/15/2023] Open
Abstract
Filarial nematodes can cause debilitating diseases such as lymphatic filariasis and onchocerciasis. Oxfendazole (OXF) is one promising macrofilaricidal candidate with improved oral availability compared to flubendazole (FBZ), and OXF is currently under preparation for phase 2 clinical trials in filariasis patients. This study aimed to investigate the immune system's role during treatment with OXF and FBZ and explore the potential to boost the treatment efficacy via stimulation of the immune system. Wild type (WT) BALB/c, eosinophil-deficient ΔdblGata1, IL-4r/IL-5-/-, antibody-deficient μMT and B-, T-, NK-cell and ILC-deficient Rag2/IL-2rγ-/- mice were infected with the rodent filaria Litomosoides sigmodontis and treated with an optimal and suboptimal regimen of OXF and FBZ for up to 5 days. In the second part, WT mice were treated for 2-3 days with a combination of OXF and IL-4, IL-5, or IL-33. Treatment of WT mice reduced the adult worm burden by up to 94% (OXF) and 100% (FBZ) compared to vehicle controls. In contrast, treatment efficacy was lower in all immunodeficient strains with a reduction of up to 90% (OXF) and 75% (FBZ) for ΔdblGata1, 50 and 92% for IL-4r/IL-5-/-, 64 and 78% for μMT or 0% for Rag2/IL-2rγ-/- mice. The effect of OXF on microfilariae and embryogenesis displayed a similar pattern, while FBZ's ability to prevent microfilaremia was independent of the host's immune status. Furthermore, flow cytometric analysis revealed strain-and treatment-specific immunological changes. The efficacy of a shortened 3-day treatment of OXF (-33% adult worms vs. vehicle) could be boosted to a 91% worm burden reduction via combination with IL-5, but not IL-4 or IL-33. Our results suggest that various components of the immune system support the filaricidal effect of benzimidazoles in vivo and present an opportunity to boost treatment efficacy.
Collapse
Affiliation(s)
- Frederic Risch
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Johanna F. Scheunemann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Julia J. Reichwald
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Benjamin Lenz
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Alexandra Ehrens
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Joséphine Gal
- Unité Molécules de Communication et Adaptation des Microorganismes, Sorbonne Université, Muséum national d’Histoire naturelle, CNRS, Paris, France
| | - Frédéric Fercoq
- Unité Molécules de Communication et Adaptation des Microorganismes, Sorbonne Université, Muséum national d’Histoire naturelle, CNRS, Paris, France
| | - Marianne Koschel
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Martina Fendler
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Coralie Martin
- Unité Molécules de Communication et Adaptation des Microorganismes, Sorbonne Université, Muséum national d’Histoire naturelle, CNRS, Paris, France
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
16
|
Alshehri A, Chhonker YS, Bala V, Edi C, Bjerum CM, Koudou BG, John LN, Mitjà O, Marks M, King CL, Murry DJ. Population pharmacokinetic model of ivermectin in mass drug administration against lymphatic filariasis. PLoS Negl Trop Dis 2023; 17:e0011319. [PMID: 37262040 DOI: 10.1371/journal.pntd.0011319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/19/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Ivermectin (IVM) is a broad-spectrum anthelmintic drug used to treat diseases caused by filarial worms, such as onchocerciasis and lymphatic filariasis (LF). IVM is part of a triple-drug therapy used by the Mass Drug Administration (MDA) as a preventive strategy to eradicate LF in sub-Saharan Africa. The drug shows high variability in drug exposure in previous pharmacokinetic studies. This study aims to build a population pharmacokinetic (PopPK) model to identify and quantify the possible sources of the variability of IVM exposure after a single-oral dose in LF-infected subjects and healthy individuals. METHODOLOGY / PRINCIPAL FINDINGS In this analysis, 724 samples were collected from treatment-naïve Wuchereria bancrofti-infected (n = 32) and uninfected (n = 24) adults living in Côte d'Ivoire who had received one dose of IVM as a part of triple-drug therapy. PopPK analysis was conducted using Phoenix NLME 8.3 software. The Monte Carlo simulation based on the final model was performed to simulate drug exposure among different dosing groups (200 μg/kg, 18 mg, and 36 mg). A two-compartment model with zero-order dose input into the absorption compartment with a lag time function followed by first-order absorption and linear elimination best described the IVM's pharmacokinetic (PK) parameters. The final model identifies that the PK parameters of IVM are not affected by LF infection. Sex was a significant covariate on the peripheral volume of distribution (Vp/F, 53% lower in men than in women). IVM drug exposure shows linear pharmacokinetic behavior among the simulated dosing groups with similar drug exposure based on sex. CONCLUSION/SIGNIFICANCE We have developed a PopPk model to describe and identify possible sources of the variability of IVM exposure. To our knowledge, this is the first PopPK study of IVM in patients with LF. TRIAL REGISTRATION NCT02845713; NCT03664063.
Collapse
Affiliation(s)
- Abdullah Alshehri
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Yashpal S Chhonker
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Veenu Bala
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Constant Edi
- Centre Suisse de Recherche Scientifique en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Catherine M Bjerum
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Benjamin G Koudou
- Centre Suisse de Recherche Scientifique en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- Université Nangui Abrogoua, Abidjan, Côte d'Ivoire
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Lucy N John
- Barcelona Institute for Global Health-University of Barcelona, Barcelona, Spain
- School of Medicine and Health Sciences, University of Papua New Guinea, Port Moresby, Papua New Guinea
- National Department of Health, Port Moresby, Papua New Guinea
| | - Oriol Mitjà
- Barcelona Institute for Global Health-University of Barcelona, Barcelona, Spain
- School of Medicine and Health Sciences, University of Papua New Guinea, Port Moresby, Papua New Guinea
| | - Michael Marks
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Hospital for Tropical Diseases, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Veterans Affairs Research Service, Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Daryl J Murry
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
17
|
Shaw C, McLure A, Graves PM, Lau CL, Glass K. Lymphatic filariasis endgame strategies: Using GEOFIL to model mass drug administration and targeted surveillance and treatment strategies in American Samoa. PLoS Negl Trop Dis 2023; 17:e0011347. [PMID: 37200375 DOI: 10.1371/journal.pntd.0011347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 05/31/2023] [Accepted: 04/29/2023] [Indexed: 05/20/2023] Open
Abstract
American Samoa underwent seven rounds of mass drug administration (MDA) for lymphatic filariasis (LF) from 2000-2006, but subsequent surveys found evidence of ongoing transmission. American Samoa has since undergone further rounds of MDA in 2018, 2019, and 2021; however, recent surveys indicate that transmission is still ongoing. GEOFIL, a spatially-explicit agent-based LF model, was used to compare the effectiveness of territory-wide triple-drug MDA (3D-MDA) with targeted surveillance and treatment strategies. Both approaches relied on treatment with ivermectin, diethylcarbamazine, and albendazole. We simulated three levels of whole population coverage for 3D-MDA: 65%, 73%, and 85%, while the targeted strategies relied on surveillance in schools, workplaces, and households, followed by targeted treatment. In the household-based strategies, we simulated 1-5 teams travelling village-to-village and offering antigen (Ag) testing to randomly selected households in each village. If an Ag-positive person was identified, treatment was offered to members of all households within 100m-1km of the positive case. All simulated interventions were finished by 2027 and their effectiveness was judged by their 'control probability'-the proportion of simulations in which microfilariae prevalence decreased between 2030 and 2035. Without future intervention, we predict Ag prevalence will rebound. With 3D-MDA, a 90% control probability required an estimated ≥ 4 further rounds with 65% coverage, ≥ 3 rounds with 73% coverage, or ≥ 2 rounds with 85% coverage. While household-based strategies were substantially more testing-intensive than 3D-MDA, they could offer comparable control probabilities with substantially fewer treatments; e.g. three teams aiming to test 50% of households and offering treatment to a 500m radius had approximately the same control probability as three rounds of 73% 3D-MDA, but used < 40% the number of treatments. School- and workplace-based interventions proved ineffective. Regardless of strategy, reducing Ag prevalence below the 1% target threshold recommended by the World Health Organization was a poor indicator of the interruption of LF transmission, highlighting the need to review blanket elimination targets.
Collapse
Affiliation(s)
- Callum Shaw
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT, Australia
| | - Angus McLure
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT, Australia
| | - Patricia M Graves
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland, Australia
| | - Colleen L Lau
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Kathryn Glass
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
18
|
Sharma S, Smith ME, Bilal S, Michael E. Evaluating elimination thresholds and stopping criteria for interventions against the vector-borne macroparasitic disease, lymphatic filariasis, using mathematical modelling. Commun Biol 2023; 6:225. [PMID: 36849730 PMCID: PMC9971242 DOI: 10.1038/s42003-022-04391-9] [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: 09/12/2021] [Accepted: 12/20/2022] [Indexed: 03/01/2023] Open
Abstract
We leveraged the ability of EPIFIL transmission models fit to field data to evaluate the use of the WHO Transmission Assessment Survey (TAS) for supporting Lymphatic Filariasis (LF) intervention stopping decisions. Our results indicate that understanding the underlying parasite extinction dynamics, particularly the protracted transient dynamics involved in shifts to the extinct state, is crucial for understanding the impacts of using TAS for determining the achievement of LF elimination. These findings warn that employing stopping criteria set for operational purposes, as employed in the TAS strategy, without a full consideration of the dynamics of extinction could seriously undermine the goal of achieving global LF elimination.
Collapse
Affiliation(s)
- Swarnali Sharma
- Christian Medical College, IDA Scudder Road, Vellore, Tamil Nadu, 632004, India.
| | - Morgan E Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, South Bend, IN, USA
| | - Shakir Bilal
- Center for Global Health Infectious Disease Research, University of South Florida, Tampa, FL, USA
| | - Edwin Michael
- Center for Global Health Infectious Disease Research, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
19
|
Ehrens A, Schiefer A, Krome AK, Becker T, Rox K, Neufeld H, Aden T, Wagner KG, Müller R, Grosse M, Stadler M, König GM, Kehraus S, Alt S, Hesterkamp T, Hübner MP, Pfarr K, Hoerauf A. Pharmacology and early ADMET data of corallopyronin A, a natural product with macrofilaricidal anti-wolbachial activity in filarial nematodes. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.983107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Corallopyronin A (CorA), a natural product antibiotic of Corallococcus coralloides, inhibits the bacterial DNA-dependent RNA polymerase. It is active against the essential Wolbachia endobacteria of filarial nematodes, preventing development, causing sterility and killing adult worms. CorA is being developed to treat the neglected tropical diseases onchocerciasis and lymphatic filariasis caused by Wolbachia-containing filariae. For this, we have completed standard Absorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) studies. In Caco-2 assays, CorA had good adsorption values, predicting good transport from the intestines, but may be subject to active efflux. In fed-state simulated human intestinal fluid (pH 5.0), CorA half-life was >139 minutes, equivalent to the stability in buffer (pH 7.4). CorA plasma-stability was >240 minutes, with plasma protein binding >98% in human, mouse, rat, dog, mini-pig and monkey plasma. Clearance in human and dog liver microsomes was low (35.2 and 42 µl/min/mg, respectively). CorA was mainly metabolized via phase I reactions, i.e., oxidation, and to a minimal extent via phase II reactions. In contrast to rifampicin, CorA does not induce CYP3A4 resulting in a lower drug-drug-interaction potential. Apart from inhibition of CYP2C9, no impact of CorA on enzymes of the CYP450 system was detected. Off-target profiling resulted in three hits (inhibition/activation) for the A3 and PPARγ receptors and COX1 enzyme; thus, potential drug-drug interactions could occur with antidiabetic medications, COX2 inhibitors, angiotensin AT1 receptor antagonists, vitamin K-antagonists, and antidepressants. In vivo pharmacokinetic studies in Mongolian gerbils and rats demonstrated excellent intraperitoneal and oral bioavailability (100%) with fast absorption and high distribution in plasma. No significant hERG inhibition was detected and no phototoxicity was seen. CorA did not induce gene mutations in bacteria (Ames test) nor chromosomal damage in human lymphocytes (micronucleus test). Thus, CorA possesses an acceptable in vitro early ADMET profile; supported by previous in vivo experiments in mice, rats and Mongolian gerbils in which all animals tolerated CorA daily administration for 7-28 days. The non-GLP package will guide selection and planning of regulatory-conform GLP models prior to a first-into-human study.
Collapse
|
20
|
McLure A, Graves PM, Lau C, Shaw C, Glass K. Modelling lymphatic filariasis elimination in American Samoa: GEOFIL predicts need for new targets and six rounds of mass drug administration. Epidemics 2022; 40:100591. [DOI: 10.1016/j.epidem.2022.100591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/03/2022] Open
|
21
|
Ehrens A, Hoerauf A, Hübner MP. Current perspective of new anti-Wolbachial and direct-acting macrofilaricidal drugs as treatment strategies for human filariasis. GMS INFECTIOUS DISEASES 2022; 10:Doc02. [PMID: 35463816 PMCID: PMC9006451 DOI: 10.3205/id000079] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Filarial diseases like lymphatic filariasis and onchocerciasis belong to the Neglected Tropical Diseases and remain a public health problem in endemic countries. Lymphatic filariasis and onchocerciasis can lead to stigmatizing pathologies and present a socio-economic burden for affected people and their endemic countries. Current treatment recommendations by the WHO include mass drug administration with ivermectin for the treatment of onchocerciasis and a combination of ivermectin, albendazole and diethylcarbamazine (DEC) for the treatment of lymphatic filariasis in areas that are not co-endemic for onchocerciasis or loiasis. Limitations of these treatment strategies are due to potential severe adverse events in onchocerciasis and loiasis patients following DEC or ivermectin treatment, respectively, the lack of a macrofilaricidal efficacy of those drugs and the risk of drug resistance development. Thus, to achieve the elimination of transmission of onchocerciasis and the elimination of lymphatic filariasis as a public health problem by 2030, the WHO defined in its roadmap that new alternative treatment strategies with macrofilaricidal compounds are required. Within a collaboration of the non-profit organizations Drugs for Neglected Diseases initiative (DNDi), the Bill & Melinda Gates Foundation, and partners from academia and industry, several new promising macrofilaricidal drug candidates were identified, which will be discussed in this review.
Collapse
Affiliation(s)
- Alexandra Ehrens
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
22
|
Clark J, Stolk WA, Basáñez MG, Coffeng LE, Cucunubá ZM, Dixon MA, Dyson L, Hampson K, Marks M, Medley GF, Pollington TM, Prada JM, Rock KS, Salje H, Toor J, Hollingsworth TD. How modelling can help steer the course set by the World Health Organization 2021-2030 roadmap on neglected tropical diseases. Gates Open Res 2022; 5:112. [PMID: 35169682 PMCID: PMC8816801 DOI: 10.12688/gatesopenres.13327.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 01/12/2023] Open
Abstract
The World Health Organization recently launched its 2021-2030 roadmap, Ending the Neglect to Attain the Sustainable Development Goals , an updated call to arms to end the suffering caused by neglected tropical diseases. Modelling and quantitative analyses played a significant role in forming these latest goals. In this collection, we discuss the insights, the resulting recommendations and identified challenges of public health modelling for 13 of the target diseases: Chagas disease, dengue, gambiense human African trypanosomiasis (gHAT), lymphatic filariasis (LF), onchocerciasis, rabies, scabies, schistosomiasis, soil-transmitted helminthiases (STH), Taenia solium taeniasis/ cysticercosis, trachoma, visceral leishmaniasis (VL) and yaws. This piece reflects the three cross-cutting themes identified across the collection, regarding the contribution that modelling can make to timelines, programme design, drug development and clinical trials.
Collapse
Affiliation(s)
- Jessica Clark
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Institute of Biodiversity, Animal Health & Comparative Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Wilma A. Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - María-Gloria Basáñez
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - Luc E. Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Zulma M. Cucunubá
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - Matthew A. Dixon
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
- Schistosomiasis Control Initiative Foundation, London, SE11 5DP, UK
| | - Louise Dyson
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Katie Hampson
- Institute of Biodiversity, Animal Health & Comparative Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Michael Marks
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Graham F. Medley
- Centre for Mathematical Modelling of Infectious Disease, London School of Hygiene & Tropical Medicine, 15-17 Tavistock Place, London, WC1H 9SH, UK
| | - Timothy M. Pollington
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
| | - Joaquin M. Prada
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, UK
| | - Kat S. Rock
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - T. Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| |
Collapse
|
23
|
Abuelazm MT, Abdelazeem B, Badr H, Gamal M, Ashraf M, Abd‐elsalam S. Efficacy and Safety of Triple Therapy Versus Dual Therapy for Lymphatic Filariasis: A Systematic Review and Meta‐Analysis. Trop Med Int Health 2022; 27:226-235. [PMID: 35080325 DOI: 10.1111/tmi.13727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Basel Abdelazeem
- McLaren Health Care, Flint/ Michigan State University Michigan USA
| | - Helmy Badr
- Faculty of Medicine Tanta University Tanta Egypt
| | | | | | | |
Collapse
|
24
|
Drug associations as alternative and complementary therapy for neglected tropical diseases. Acta Trop 2022; 225:106210. [PMID: 34687644 DOI: 10.1016/j.actatropica.2021.106210] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/02/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022]
Abstract
The present paper aims to establish different treatments for neglected tropical disease by a survey on drug conjugations and possible fixed-dose combinations (FDC) used to obtain alternative, safer and more effective treatments. The source databases used were Science Direct and PubMed/Medline, in the intervals between 2015 and 2021 with the drugs key-words or diseases, like "schistosomiasis", "praziquantel", "malaria", "artesunate", "Chagas' disease", "benznidazole", "filariasis", diethylcarbamazine", "ivermectin", " albendazole". 118 works were the object of intense analysis, other articles and documents were used to increase the quality of the studies, such as consensuses for harmonizing therapeutics and historical articles. As a result, an effective NTD control can be achieved when different public health approaches are combined with interventions guided by the epidemiology of each location and the availability of appropriate measures to detect, prevent and control disease. It was also possible to verify that the FDCs promote a simplification of the therapeutic regimen, which promotes better patient compliance and enables a reduction in the development of parasitic resistance, requiring further studies aimed at resistant strains, since the combined APIs usually act by different mechanisms or at different target sites. In addition to eliminating the process of developing a new drug based on the identification and validation of active compounds, which is a complex, long process and requires a strong long-term investment, other advantages that FDCs have are related to productive gain and gain from the industrial plant, which can favor and encourage the R&D of new FDCs not only for NTDs but also for other diseases that require the use of more than one drug.
Collapse
|
25
|
Clark J, Stolk WA, Basáñez MG, Coffeng LE, Cucunubá ZM, Dixon MA, Dyson L, Hampson K, Marks M, Medley GF, Pollington TM, Prada JM, Rock KS, Salje H, Toor J, Hollingsworth TD. How modelling can help steer the course set by the World Health Organization 2021-2030 roadmap on neglected tropical diseases. Gates Open Res 2021; 5:112. [PMID: 35169682 PMCID: PMC8816801 DOI: 10.12688/gatesopenres.13327.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 01/12/2023] Open
Abstract
The World Health Organization recently launched its 2021-2030 roadmap, Ending the Neglect to Attain the Sustainable Development Goals , an updated call to arms to end the suffering caused by neglected tropical diseases. Modelling and quantitative analyses played a significant role in forming these latest goals. In this collection, we discuss the insights, the resulting recommendations and identified challenges of public health modelling for 13 of the target diseases: Chagas disease, dengue, gambiense human African trypanosomiasis (gHAT), lymphatic filariasis (LF), onchocerciasis, rabies, scabies, schistosomiasis, soil-transmitted helminthiases (STH), Taenia solium taeniasis/ cysticercosis, trachoma, visceral leishmaniasis (VL) and yaws. This piece reflects the three cross-cutting themes identified across the collection, regarding the contribution that modelling can make to timelines, programme design, drug development and clinical trials.
Collapse
Affiliation(s)
- Jessica Clark
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Institute of Biodiversity, Animal Health & Comparative Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Wilma A. Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - María-Gloria Basáñez
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - Luc E. Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Zulma M. Cucunubá
- London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - Matthew A. Dixon
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
- Schistosomiasis Control Initiative Foundation, London, SE11 5DP, UK
| | - Louise Dyson
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Katie Hampson
- Institute of Biodiversity, Animal Health & Comparative Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Michael Marks
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Graham F. Medley
- Centre for Mathematical Modelling of Infectious Disease, London School of Hygiene & Tropical Medicine, 15-17 Tavistock Place, London, WC1H 9SH, UK
| | - Timothy M. Pollington
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
| | - Joaquin M. Prada
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, UK
| | - Kat S. Rock
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - T. Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| |
Collapse
|
26
|
Population pharmacokinetics of diethylcarbamazine in patients with lymphatic filariasis and healthy individuals. Antimicrob Agents Chemother 2021; 65:e0031721. [PMID: 34310218 DOI: 10.1128/aac.00317-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Diethylcarbamazine (DEC) is a drug of choice to treat lymphatic filariasis (LF) either used alone or in combination as mass drug administration (MDA) preventive strategies. The objective of this study was to develop a population pharmacokinetic model for DEC in subjects infected with lymphatic filariasis (LF) compared to healthy individuals, and to evaluate the effect of covariates on the volume of distribution (V/F) and oral clearance (CL/F) of DEC. This was an open-label cohort study of treatment naïve Wuchereria bancrofti-infected (n=32) and uninfected (n=24) adults residing in the Agboville district of Côte d'Ivoire. The population pharmacokinetic model for DEC was built using Phoenix NLME 8.0 software. The covariates included in the model building process were age, gender, bodyweight, infection status, creatinine clearance (CrCl), aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. A total of 56 adults were enrolled in the study and a total of 728 samples were obtained over 168 hours. A one-compartment linear pharmacokinetic model with first-order absorption with an absorption lag-time (Tlag) best described the data. After determining the pharmacokinetics (PK) parameters of DEC, body weight and gender were found to be the significant covariates for DEC V/F. The final population pharmacokinetic model adequately described the pharmacokinetics of DEC in the studied population. Model-based simulation indicated that the body weight significantly impacted the exposure in both the male and female population. This analysis may further support the drug-drug interaction model development of DEC with different co-administered drugs/agents in disease control programs.
Collapse
|
27
|
Gangwar M, Jha R, Goyal M, Srivastava M. Biochemical characterization of Recombinase A from Wolbachia endosymbiont of filarial nematode Brugia malayi (wBmRecA). Int J Parasitol 2021; 51:841-853. [PMID: 34273392 DOI: 10.1016/j.ijpara.2021.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 11/17/2022]
Abstract
Lymphatic filariasis is a debilitating disease that affects over 890 million people in 49 countries. A lack of vaccines, non-availability of adulticidal drugs, the threat of emerging drug resistance against available chemotherapeutics and an incomplete understanding of the immunobiology of the disease have sustained the problem. Characterization of Wolbachia proteins, the bacterial endosymbiont which helps in the growth and development of filarial worms, regulates fecundity in female worms and mediates immunopathogenesis of Lymphatic Filariasis, is an important approach to gain insights into the immunopathogenesis of the disease. In this study, we carried out extensive biochemical characterization of Recombinase A from Wolbachia of the filarial nematode Brugia malayi (wBmRecA) using an Electrophoretic Mobility Shift Assay, an ATP binding and hydrolysis assay, DNA strand exchange reactions, DAPI displacement assay and confocal microscopy, and evaluated anti-filarial activity of RecA inhibitors. Confocal studies showed that wBmRecA was expressed and localised within B. malayi microfilariae (Mf) and uteri and lateral chord of adult females. Recombinant wBmRecA was biochemically active and showed intrinsic binding capacity towards both single-stranded DNA and double-stranded DNA that were enhanced by ATP, suggesting ATP-induced cooperativity. wBmRecA promoted ATP hydrolysis and DNA strand exchange reactions in a concentration-dependent manner, and its binding to DNA was sensitive to temperature, pH and salt concentration. Importantly, the anti-parasitic drug Suramin, and Phthalocyanine tetrasulfonate (PcTs)-based inhibitors Fe-PcTs and 3,4-Cu-PcTs, inhibited wBmRecA activity and affected the motility and viability of Mf. The addition of Doxycycline further enhanced microfilaricidal activity of wBmRecA, suggesting potential synergism. Taken together, the omnipresence of wBmRecA in B. malayi life stages and the potent microfilaricidal activity of RecA inhibitors suggest an important role of wBmRecA in filarial pathogenesis.
Collapse
Affiliation(s)
- Mamta Gangwar
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ruchi Jha
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Manish Goyal
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Mrigank Srivastava
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
28
|
Davis EL, Prada J, Reimer LJ, Hollingsworth TD. Modelling the Impact of Vector Control on Lymphatic Filariasis Programs: Current Approaches and Limitations. Clin Infect Dis 2021; 72:S152-S157. [PMID: 33905475 PMCID: PMC8201547 DOI: 10.1093/cid/ciab191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Vector control is widely considered an important tool for lymphatic filariasis (LF) elimination but is not usually included in program budgets and has often been secondary to other policy questions in modelling studies. Evidence from the field demonstrates that vector control can have a large impact on program outcomes and even halt transmission entirely, but implementation is expensive. Models of LF have the potential to inform where and when resources should be focused, but often simplify vector dynamics and focus on capturing human prevalence trends, making them comparatively ill-designed for direct analysis of vector control measures. We review the recent modelling literature and present additional results using a well-established model, highlighting areas of agreement between model predictions and field evidence, and discussing the possible determinants of existing disagreements. We conclude that there are likely to be long-term benefits of vector control, both on accelerating programs and preventing resurgence.
Collapse
Affiliation(s)
- E L Davis
- Big Data Institute, University of Oxford, Oxford, UK
| | - J Prada
- University of Surrey, Guildford,UK
| | - L J Reimer
- Liverpool School of Tropical Medicine, Liverpool,UK
| | | |
Collapse
|
29
|
Salonga PKN, Mendoza VMP, Mendoza RG, Belizario VY. A mathematical model of the dynamics of lymphatic filariasis in Caraga Region, the Philippines. ROYAL SOCIETY OPEN SCIENCE 2021; 8:201965. [PMID: 34234950 PMCID: PMC8242838 DOI: 10.1098/rsos.201965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 06/07/2021] [Indexed: 06/13/2023]
Abstract
Despite being one of the first countries to implement mass drug administration (MDA) for elimination of lymphatic filariasis (LF) in 2001 after a pilot study in 2000, the Philippines is yet to eliminate the disease as a public health problem with 6 out of the 46 endemic provinces still implementing MDA for LF as of 2018. In this work, we propose a mathematical model of the transmission dynamics of LF in the Philippines and a control strategy for its elimination using MDA. Sensitivity analysis using the Latin hypercube sampling and partial rank correlation coefficient methods suggests that the infected human population is most sensitive to the treatment parameters. Using the available LF data in Caraga Region from the Philippine Department of Health, we estimate the treatment rates r 1 and r 2 using the least-squares parameter estimation technique. Parameter bootstrapping showed small variability in the parameter estimates. Finally, we apply optimal control theory with the objective of minimizing the infected human population and the corresponding implementation cost of MDA, using the treatment coverage γ as the control parameter. Simulation results highlight the importance of maintaining a high MDA coverage per year to effectively minimize the infected population by the year 2030.
Collapse
Affiliation(s)
- Pamela Kim N. Salonga
- Institute of Mathematics, University of the Philippines Diliman, Quezon City, Philippines
- Natural Sciences Research Institute, University of the Philippines Diliman, Quezon City, Philippines
| | - Victoria May P. Mendoza
- Institute of Mathematics, University of the Philippines Diliman, Quezon City, Philippines
- Natural Sciences Research Institute, University of the Philippines Diliman, Quezon City, Philippines
| | - Renier G. Mendoza
- Institute of Mathematics, University of the Philippines Diliman, Quezon City, Philippines
- Natural Sciences Research Institute, University of the Philippines Diliman, Quezon City, Philippines
| | - Vicente Y. Belizario
- College of Public Health and Neglected Tropical Diseases Study Group, National Institutes of Health, University of the Philippines Manila, Philippines
| |
Collapse
|
30
|
Lammie PJ, Gass KM, King J, Deming MS, Addiss DG, Biswas G, Ottesen EA, Henderson R. Evolution of the monitoring and evaluation strategies to support the World Health Organization's Global Programme to Eliminate Lymphatic Filariasis. Int Health 2021; 13:S65-S70. [PMID: 33349883 PMCID: PMC7753166 DOI: 10.1093/inthealth/ihaa084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/05/2020] [Accepted: 10/16/2020] [Indexed: 12/02/2022] Open
Abstract
The Global Programme to Eliminate Lymphatic Filariasis (GPELF) was established with the ambitious goal of eliminating LF as a public health problem. The remarkable success of the GPELF over the past 2 decades in carrying out its principal strategy of scaling up and scaling down mass drug administration has relied first on the development of a rigorous monitoring and evaluation (M&E) framework and then the willingness of the World Health Organization and its community of partners to modify this framework in response to the practical experiences of national programmes. This flexibility was facilitated by the strong partnership that developed among researchers, LF programme managers and donors willing to support the necessary research agenda. This brief review summarizes the historical evolution of the GPELF M&E strategies and highlights current research needed to achieve the elimination goal.
Collapse
Affiliation(s)
- Patrick J Lammie
- Division of Parasitic Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA 30333, USA
| | - Katherine M Gass
- NTD Support Center, Task Force for Global Health, 325 Swanton Way, Decatur, GA 30030, USA
| | - Jonathan King
- NTD Department, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland
| | - Michael S Deming
- Division of Parasitic Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA 30333, USA
| | - David G Addiss
- Division of Parasitic Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA 30333, USA
| | - Gautam Biswas
- NTD Department, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland
| | - Eric A Ottesen
- NTD Support Center, Task Force for Global Health, 325 Swanton Way, Decatur, GA 30030, USA
| | - Ralph Henderson
- NTD Support Center, Task Force for Global Health, 325 Swanton Way, Decatur, GA 30030, USA
| |
Collapse
|
31
|
Kapa DR, Mohamed AJ. Progress and impact of 20 years of a lymphatic filariasis elimination programme in South-East Asia. Int Health 2021; 13:S17-S21. [PMID: 33349881 PMCID: PMC7753165 DOI: 10.1093/inthealth/ihaa056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/21/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023] Open
Abstract
Background The South-East Asia regional programme to eliminate lymphatic filariasis (LF) was launched in 2000. This study presents the progress and impact of the programme in the region. Methods Mass drug administration (MDA) and morbidity management data were accessed from the WHO preventive chemotherapy databank. The status of the programme in the nine South-East Asia countries was reviewed and progress was assessed. The impact of the programme on LF disease burden was estimated on the basis of the effectiveness of the MDA drugs against microfilaraemia and chronic disease. Results Under the MDA programme, 8.1 billion treatments were delivered in nine countries and 5.7 billion treatments were consumed by the target population during 2001–2018. Three of nine countries eliminated LF. Bangladesh is poised to reach its elimination goal by 2021. In the other five countries, 38–76% of intervention units completed intervention and surveillance is in progress. The MDA programme prevented or cured 74.9 million infections, equivalent to an 84.2% reduction. Close to 1 million lymphoedema patients and 0.5 million hydrocele patients were reported and are being provided with the minimum package of care. Conclusions The South-East Asia region's LF elimination programme reduced the burden of LF appreciably and is moving towards achieving the elimination goal in the next 8–10 y.
Collapse
Affiliation(s)
| | - Ahmed Jamsheed Mohamed
- Depa rtment of Communicable Diseases, WHO Regional Office for South-East Asia, New Delhi 110002, India
| |
Collapse
|
32
|
Bjerum CM, Ouattara AF, Aboulaye M, Kouadio O, Marius VK, Andersen BJ, Weil GJ, Koudou BG, King CL. Efficacy and Safety of a Single Dose of Ivermectin, Diethylcarbamazine, and Albendazole for Treatment of Lymphatic Filariasis in Côte d'Ivoire: An Open-label Randomized Controlled Trial. Clin Infect Dis 2021; 71:e68-e75. [PMID: 31641754 PMCID: PMC7583415 DOI: 10.1093/cid/ciz1050] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/21/2019] [Indexed: 12/30/2022] Open
Abstract
Background Improved drug regimens are needed to accelerate elimination of lymphatic filariasis in Africa. This study determined whether a single co-administered dose of ivermectin plus diethylcarbamazine plus albendazole [IDA] is noninferior to standard 3 annual doses of ivermectin plus albendazole (IA) used in many LF-endemic areas of Africa. Methods Treatment-naive adults with Wuchereria bancrofti microfilaremia in Côte d’Ivoire were randomized to receive a single dose of IDA (n = 43) or 3 annual doses of IA (n = 52) in an open-label, single-blinded trial. The primary endpoint was the proportion of participants who were microfilaria (Mf) negative at 36 months. Secondary endpoints were Mf clearance at 6, 12, and 24 months; inactivation of adult worm nests; and safety. Results At 36 months posttreatment with IDA, 18/33 (55%; 95% CI, 38–72%) cleared Mf versus 33/42 (79%; 67–91%) with IA (P = .045). At 6 and 12 months IDA was superior to IA in clearing Mf (89% [77–99%] and 71% [56–85%]), respectively, versus 34% (20–48%) and 26% (14–42%) (P < .001). IDA was equivalent to IA at 24 months (61% [45–77%] vs 54% [38–72%]; P = .53). IDA was superior to IA for inactivating adult worms at all time points. Both treatments were well tolerated, and there were no serious adverse events. Conclusions A single dose of IDA was superior to 2 doses of IA in reducing the overall Mf burden by 24 months. Reinfection may have contributed to the lack of sustained clearance of Mf with IDA. Clinical Trials Registration NCT02974049.
Collapse
Affiliation(s)
- Catherine M Bjerum
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Allassane F Ouattara
- Centre Suisse de Recherche Scientifique en Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Nangui Abrogoua, Abidjan, Côte d'Ivoire
| | - Méité Aboulaye
- Programme National de la Lutte Contre la Schistosomiase, Les Geohelminthiases et la Filariose Lymphatique, Abidjan, Côte d'Ivoire
| | - Olivier Kouadio
- Centre Suisse de Recherche Scientifique en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Vanga K Marius
- Universite Alassane Ouattara Centre Hospitalier Universitaire de Bouake, Bouaké, Côte d'Ivoire
| | - Britt J Andersen
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Gary J Weil
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Benjamin G Koudou
- Centre Suisse de Recherche Scientifique en Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Nangui Abrogoua, Abidjan, Côte d'Ivoire
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Veterans Affairs Research Service, Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
33
|
Walker M, Hamley JID, Milton P, Monnot F, Kinrade S, Specht S, Pedrique B, Basáñez MG. Supporting drug development for neglected tropical diseases using mathematical modelling. Clin Infect Dis 2021; 73:e1391-e1396. [PMID: 33893482 PMCID: PMC8442785 DOI: 10.1093/cid/ciab350] [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: 12/18/2020] [Indexed: 11/14/2022] Open
Abstract
Drug-based interventions are at the heart of global efforts to reach elimination as a public health problem (trachoma, soil-transmitted helminthiases, schistosomiasis, lymphatic filariasis) or elimination of transmission (onchocerciasis) for 5 of the most prevalent neglected tropical diseases tackled via the World Health Organization preventive chemotherapy strategy. While for some of these diseases there is optimism that currently available drugs will be sufficient to achieve the proposed elimination goals, for others—particularly onchocerciasis—there is a growing consensus that novel therapeutic options will be needed. Since in this area no high return of investment is possible, minimizing wasted money and resources is essential. Here, we use illustrative results to show how mathematical modeling can guide the drug development pathway, yielding resource-saving and efficiency payoffs, from the refinement of target product profiles and intended context of use to the design of clinical trials.
Collapse
Affiliation(s)
- Martin Walker
- Department of Pathobiology and Population Sciences and London Centre for Neglected Tropical Disease Research, Royal Veterinary College, UK.,MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology and London Centre for Neglected Tropical Disease Research, Imperial College London, UK
| | - Jonathan I D Hamley
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology and London Centre for Neglected Tropical Disease Research, Imperial College London, UK
| | - Philip Milton
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology and London Centre for Neglected Tropical Disease Research, Imperial College London, UK
| | - Frédéric Monnot
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Sally Kinrade
- Medicines Development for Global Health, Southbank VIC, Australia
| | - Sabine Specht
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Bélen Pedrique
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Maria-Gloria Basáñez
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology and London Centre for Neglected Tropical Disease Research, Imperial College London, UK
| |
Collapse
|
34
|
Prada JM, Stolk WA, Davis EL, Touloupou P, Sharma S, Muñoz J, Caja Rivera RM, Reimer LJ, Michael E, de Vlas SJ, Hollingsworth TD. Delays in lymphatic filariasis elimination programmes due to COVID-19, and possible mitigation strategies. Trans R Soc Trop Med Hyg 2021; 115:261-268. [PMID: 33515454 PMCID: PMC7928650 DOI: 10.1093/trstmh/trab004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022] Open
Abstract
Background In view of the current global coronavirus disease 2019 pandemic, mass drug administration interventions for neglected tropical diseases, including lymphatic filariasis (LF), have been halted. We used mathematical modelling to estimate the impact of delaying or cancelling treatment rounds and explore possible mitigation strategies. Methods We used three established LF transmission models to simulate infection trends in settings with annual treatment rounds and programme delays in 2020 of 6, 12, 18 or 24 months. We then evaluated the impact of various mitigation strategies upon resuming activities. Results The delay in achieving the elimination goals is on average similar to the number of years the treatment rounds are missed. Enhanced interventions implemented for as little as 1 y can allow catch-up on the progress lost and, if maintained throughout the programme, can lead to acceleration of up to 3 y. Conclusions In general, a short delay in the programme does not cause a major delay in achieving the goals. Impact is strongest in high-endemicity areas. Mitigation strategies such as biannual treatment or increased coverage are key to minimizing the impact of the disruption once the programme resumes and lead to potential acceleration should these enhanced strategies be maintained.
Collapse
Affiliation(s)
- Joaquín M Prada
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Wilma A Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emma L Davis
- Big Data Institute, Li Ka Shing Center for Health Information and Discovery, Headington, Oxford, UK
| | - Panayiota Touloupou
- Department of Statistics, University of Warwick, Coventry, UK.,School of Mathematics, University of Birmingham, Birmingham, UK
| | - Swarnali Sharma
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Johanna Muñoz
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rocio M Caja Rivera
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Center for Global Health Infectious Disease Research, University of South Florida, Tampa, FL, USA
| | - Lisa J Reimer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Edwin Michael
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Center for Global Health Infectious Disease Research, University of South Florida, Tampa, FL, USA
| | - Sake J de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - T Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Center for Health Information and Discovery, Headington, Oxford, UK
| |
Collapse
|
35
|
Jambulingam P, Kuttiatt VS, Krishnamoorthy K, Subramanian S, Srividya A, Raju HKK, Rahi M, Somani RK, Suryaprakash MK, Dwivedi GP, Weil GJ. An open label, block randomized, community study of the safety and efficacy of co-administered ivermectin, diethylcarbamazine plus albendazole vs. diethylcarbamazine plus albendazole for lymphatic filariasis in India. PLoS Negl Trop Dis 2021; 15:e0009069. [PMID: 33591979 PMCID: PMC7909694 DOI: 10.1371/journal.pntd.0009069] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 02/26/2021] [Accepted: 12/12/2020] [Indexed: 12/16/2022] Open
Abstract
Background Better drug regimens for mass drug administration (MDA) could accelerate the Global Programme to Eliminate Lymphatic Filariasis (LF). This community study was designed to compare the safety and efficacy of MDA with IDA (ivermectin, diethylcarbamazine and albendazole) or DA (diethylcarbamazine and albendazole) in India. Methodology/Principal findings This two-armed, open-labelled, block randomised, community study was conducted in LF endemic villages in Yadgir district, Karnataka, India. Consenting participants ≥5 years of age were tested for circulating filarial antigenemia (CFA) and microfilaremia (Mf) before treatment with a single oral dose of IDA or DA. Adverse events (AEs) were monitored actively for two days and passively for five more days. Persons with positive CFA or Mf tests at baseline were retested 12-months post-treatment to assess treatment efficacy. Baseline CFA and Mf-rates were 26.4% and 6.9% in IDA and 24.5% and 6.4% in DA villages respectively. 4758 and 4160 participants received IDA and DA. Most AEs were mild after both treatments; fewer than 0.1% of participants experienced AEs with severity > grade 1. No serious AEs were observed. Fever, headache and dizziness were the most common AEs. AE rates were slightly higher after IDA than DA (8.3% vs. 6.4%, P<0.01). AEs were more frequent in females and Mf-positives after either treatment, but significantly more frequent after IDA (40.5% vs 20.2%, P < 0.001). IDA was more effective for clearing Mf than DA (84% vs. 61.8%, P < 0.001). Geometric mean Mf counts per 60μl in retested Mf-positives decreased by 96.4% from 11.8 after IDA and by 90.0% from 9.5 after DA. Neither treatment was effective for clearing CFA. Conclusions/Significance IDA had an acceptable safety profile and was more effective for clearing Mf than DA. With adequate compliance and medical support to manage AEs, IDA has the potential to accelerate LF elimination in India. Trial registration Clinical Trial Registry of India (CTRI No/2016/10/007399) Lymphatic filariasis (LF) is a major neglected tropical disease that is caused by filarial nematode worms. The strategies of the Global Programme to Eliminate Lymphatic Filariasis, launched in 2000, are mass drug administration (MDA) of antifilarial medications to kill the parasites and reduce transmission and morbidity management and disability prevention for those who are already affected by the disease. Recent clinical trials have shown that a single co-administered dose of ivermectin, diethylcarbamazine and albendazole (IDA) is more effective for clearing microfilariae (Mf) from the blood than the traditional two-drug regimen (DA). That is important, because blood Mf are essential for mosquitoes to transmit the parasite. As part of a large multicenter study, we assessed the safety of IDA and compared the efficacy of IDA and DA for clearing parasites from the blood. We treated almost 9,000 people in Wuchereria bancrofti endemic villages with either IDA or DA. Adverse events (AE) were monitored actively for two days and passively for another five days. AE rates were slightly higher after IDA than DA, but AEs were mild and self-limited. Infected persons, adults and females had higher AE rates in both treatment areas. We retested infected persons one year after treatment. IDA was significantly more effective for clearing Mf and reducing blood Mf counts than DA. Neither treatment was effective for clearing circulating filarial antigenemia. Our large study showed that IDA was well tolerated and more effective than DA. This new treatment has the potential to hasten LF elimination in India and many other countries.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Manju Rahi
- Indian Council of Medical Research, New Delhi, India
| | - Roopali K. Somani
- Department of Clinical Pharmacology & Therapeutics, Nizams Institute of Medical Sciences, Hyderabad, India
| | | | | | - Gary J. Weil
- Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
36
|
Dickson BFR, Graves PM, Aye NN, Nwe TW, Wai T, Win SS, Shwe M, Douglass J, Wood P, Wangdi K, McBride WJ. Risk factors for lymphatic filariasis and mass drug administration non-participation in Mandalay Region, Myanmar. Parasit Vectors 2021; 14:72. [PMID: 33482891 PMCID: PMC7821648 DOI: 10.1186/s13071-021-04583-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myanmar commenced a lymphatic filariasis (LF) elimination programme in 2000. Whilst the country has made considerable progress since then, a number of districts have demonstrated persistent transmission after many rounds of mass drug administration (MDA). The causes of unsuccessful MDA have been examined elsewhere; however, there remains little information on the factors that contribute in Myanmar. METHODS We conducted an analysis of factors associated with persistent infection, LF-related hydrocoele and MDA participation in an area with ongoing transmission in 2015. A cross-sectional household survey was undertaken in 24 villages across four townships of Mandalay Region. Participants were screened for circulating filarial antigen (CFA) using immunochromatographic tests and, if positive, for microfilaria by night-time thick blood slide. Individuals 15 year and older were assessed for filariasis morbidity (lymphoedema and, if male, hydrocoele) by ultrasound-assisted clinical examination. A pre-coded questionnaire was used to assess risk factors for LF and for non-participation (never taking MDA). Significant variables identified in univariate analyses were included in separate step-wise multivariate logistic regressions for each outcome. RESULTS After adjustment for covariates and survey design, being CFA positive was significantly associated with age [odds ratio (OR) 1.03, 95% CI 1.01-1.06), per year], male gender (OR 3.14, 1.27-7.76), elevation (OR 0.96, 0.94-0.99, per metre) and the density of people per household room (OR 1.59, 1.31-1.92). LF-related hydrocoele was associated with age (OR 1.06, 1.03-1.09, per year) and residing in Amarapura Township (OR 8.93, 1.37-58.32). Never taking MDA was associated with male gender [OR 6.89 (2.13-22.28)] and age, particularly in females, with a significant interaction term. Overall, compared to those aged 30-44 years, the proportion never taking MDA was higher in all age groups (OR highest in those < 5 years and > 60 years, ranging from 3.37 to 12.82). Never taking MDA was also associated with residing in Amarapura township (OR 2.48, 1.15-5.31), moving to one's current village from another (OR 2.62, 1.12-6.11) and ever having declined medication (OR 11.82, 4.25-32.91). Decreased likelihood of never taking MDA was associated with a higher proportion of household members being present during the last MDA round (OR 0.16, 0.03-0.74) and the number visits by the MDA programme (OR 0.69, 0.48-1.00). CONCLUSIONS These results contribute to the understanding of LF and MDA participation-related risk factors and will assist Myanmar to improve its elimination and morbidity management programmes.
Collapse
Affiliation(s)
- Benjamin F R Dickson
- College of Public Health, Medical and Veterinary Sciences, Division of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.
| | - Patricia M Graves
- College of Medicine & Dentistry, Division of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,James Cook University and World Health Organization Collaborating Centre for Vector-Borne and Neglected Tropical Diseases, Townsville, QLD, Australia
| | - Ni Ni Aye
- Vector Borne Disease Control Unit, Ministry of Health and Sport, Naypyitaw, Myanmar
| | - Thet Wai Nwe
- Vector Borne Disease Control Unit, Ministry of Health and Sport, Naypyitaw, Myanmar
| | - Tint Wai
- Regional Vector Borne Disease Control Unit, Ministry of Health and Sport, Mandalay, Myanmar
| | | | | | - Janet Douglass
- College of Medicine & Dentistry, Division of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,James Cook University and World Health Organization Collaborating Centre for Vector-Borne and Neglected Tropical Diseases, Townsville, QLD, Australia
| | - Peter Wood
- College of Medicine & Dentistry, Division of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Kinley Wangdi
- Department of Global Health, Research School of Population Health, ANU College of Health & Medicine, The Australian National University, Canberra, ACT, Australia
| | - William J McBride
- College of Public Health, Medical and Veterinary Sciences, Division of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
37
|
Hardwick RJ, Werkman M, Truscott JE, Anderson RM. Stochastic challenges to interrupting helminth transmission. Epidemics 2021; 34:100435. [PMID: 33571786 DOI: 10.1016/j.epidem.2021.100435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/27/2020] [Accepted: 01/10/2021] [Indexed: 01/29/2023] Open
Abstract
Predicting the effect of different programmes designed to control both the morbidity induced by helminth infections and parasite transmission is greatly facilitated by the use of mathematical models of transmission and control impact. In such models, it is essential to account for the many sources of uncertainty - natural, or otherwise - to ensure robustness in prediction and to accurately depict variation around an expected outcome. In this paper, we investigate how well the standard deterministic models match the predictions made using individual-based stochastic simulations. We also explore how well concepts which derive from deterministic models, such as 'breakpoints' in transmission, apply in the stochastic world. Employing an individual-based stochastic model framework we also investigate how transmission and control are affected by the migration of infected people into a defined community. To give our study focus we consider the control of soil-transmitted helminths (STH) by mass drug administration (MDA), though our methodology is readily applicable to the other helminth species such as the schistosome parasites and the filarial worms. We show it is possible to theoretically define a 'stochastic breakpoint' where much noise surrounds the expected deterministic breakpoint. We also discuss the concept of the 'interruption of transmission' independent of the 'breakpoint' concept where analyses of model behaviour illustrate the current limitations of deterministic models to account for the 'fade-out' or transmission extinction behaviour in simulations. Our analysis of migration confirms a relationship between the critical infected human migration rate scale (i.e., order of magnitude) per unit of time and the death rate of infective stages that are released into the free-living environment. This relationship is shown to determine the likelihood that control activities aim at chemotherapeutic treatment of the human host will eliminate transmission. The development of a new stochastic simulation code for STH in the form of a publicly-available open-source python package which includes features to incorporate many population stratifications, different control interventions including mass drug administration (with defined frequency, coverage levels and compliance patterns) and inter-village human migration is also described.
Collapse
Affiliation(s)
- Robert J Hardwick
- London Centre for Neglected Tropical Disease Research (LCNTDR), Department of Infectious Disease Epidemiology, St. Mary's Campus, Imperial College London, London WC2 1PG, UK; The DeWorm3 Project, the Natural History Museum of London, London SW7 5BD, UK; MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, UK.
| | - Marleen Werkman
- London Centre for Neglected Tropical Disease Research (LCNTDR), Department of Infectious Disease Epidemiology, St. Mary's Campus, Imperial College London, London WC2 1PG, UK; The DeWorm3 Project, the Natural History Museum of London, London SW7 5BD, UK; MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, UK
| | - James E Truscott
- London Centre for Neglected Tropical Disease Research (LCNTDR), Department of Infectious Disease Epidemiology, St. Mary's Campus, Imperial College London, London WC2 1PG, UK; The DeWorm3 Project, the Natural History Museum of London, London SW7 5BD, UK; MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, UK
| | - Roy M Anderson
- London Centre for Neglected Tropical Disease Research (LCNTDR), Department of Infectious Disease Epidemiology, St. Mary's Campus, Imperial College London, London WC2 1PG, UK; The DeWorm3 Project, the Natural History Museum of London, London SW7 5BD, UK; MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, UK
| |
Collapse
|
38
|
Tyagi R, Bulman CA, Cho-Ngwa F, Fischer C, Marcellino C, Arkin MR, McKerrow JH, McNamara CW, Mahoney M, Tricoche N, Jawahar S, Janetka JW, Lustigman S, Sakanari J, Mitreva M. An Integrated Approach to Identify New Anti-Filarial Leads to Treat River Blindness, a Neglected Tropical Disease. Pathogens 2021; 10:71. [PMID: 33466870 PMCID: PMC7830784 DOI: 10.3390/pathogens10010071] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 11/17/2022] Open
Abstract
Filarial worms cause multiple debilitating diseases in millions of people worldwide, including river blindness. Currently available drugs reduce transmission by killing larvae (microfilariae), but there are no effective cures targeting the adult parasites (macrofilaricides) which survive and reproduce in the host for very long periods. To identify effective macrofilaricides, we carried out phenotypic screening of a library of 2121 approved drugs for clinical use against adult Brugia pahangi and prioritized the hits for further studies by integrating those results with a computational prioritization of drugs and associated targets. This resulted in the identification of 18 hits with anti-macrofilaricidal activity, of which two classes, azoles and aspartic protease inhibitors, were further expanded upon. Follow up screening against Onchocerca spp. (adult Onchocerca ochengi and pre-adult O. volvulus) confirmed activity for 13 drugs (the majority having IC50 < 10 μM), and a counter screen of a subset against L. loa microfilariae showed the potential to identify selective drugs that prevent adverse events when co-infected individuals are treated. Stage specific activity was also observed. Many of these drugs are amenable to structural optimization, and also have known canonical targets, making them promising candidates for further optimization that can lead to identifying and characterizing novel anti-macrofilarial drugs.
Collapse
Affiliation(s)
- Rahul Tyagi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, 4523 Clayton Ave., St. Louis, MO 63110, USA;
| | - Christina A. Bulman
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th Street, San Francisco, CA 94158, USA; (C.A.B.); (C.F.); (M.R.A.)
| | - Fidelis Cho-Ngwa
- ANDI Centre of Excellence for Onchocerciasis Drug Research, Biotechnology Unit, Faculty of Science, University of Buea, Buea CM-00237, Cameroon;
| | - Chelsea Fischer
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th Street, San Francisco, CA 94158, USA; (C.A.B.); (C.F.); (M.R.A.)
| | - Chris Marcellino
- Division of Neurocritical Care and Hospital Neurology, Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th Street, San Francisco, CA 94158, USA; (C.A.B.); (C.F.); (M.R.A.)
| | - James H. McKerrow
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA 92093, USA;
| | - Case W. McNamara
- Calibr, a Division of The Scripps Research Institute, 11119 Torrey Pines Road, La Jolla, CA 92037, USA;
| | - Matthew Mahoney
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA; (M.M.); (J.W.J.)
| | - Nancy Tricoche
- Lindsley F. Kimball Research Institute, New York City, NY 10065, USA; (N.T.); (S.J.); (S.L.)
| | - Shabnam Jawahar
- Lindsley F. Kimball Research Institute, New York City, NY 10065, USA; (N.T.); (S.J.); (S.L.)
| | - James W. Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA; (M.M.); (J.W.J.)
| | - Sara Lustigman
- Lindsley F. Kimball Research Institute, New York City, NY 10065, USA; (N.T.); (S.J.); (S.L.)
| | - Judy Sakanari
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th Street, San Francisco, CA 94158, USA; (C.A.B.); (C.F.); (M.R.A.)
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, 4523 Clayton Ave., St. Louis, MO 63110, USA;
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave., St. Louis, MO 63108, USA
| |
Collapse
|
39
|
Das L, Mathiarasan L, Krishnakumari A. Assessment of the impact of morbidity management and disability prevention for lymphatic filariasis on the disease burden in villupuram district of Tamil Nadu, India. Indian J Community Med 2021; 46:657-661. [PMID: 35068729 PMCID: PMC8729271 DOI: 10.4103/ijcm.ijcm_12_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022] Open
Abstract
Background: The global program to eliminate lymphatic filariasis (GPELF) was started in 2003 with two strategies: the mass drug administration (MDA) to interrupt disease transmission and the morbidity management and disability prevention (MMDP) to provide the basic hygienic care to filariasis lymphedema patients. Among the two strategies, the MDA is well advanced and got the desired results, but the MMDP is lagging due to poor execution. Objectives: To assess the awareness of MMDP and ongoing morbidity management practices by lymphedema patients and to estimate the impacts of the MMDP on the prevalence and severity of lymphedema. Materials and Methods: This study was conducted among 100 lymphedema patients in 7 filariasis endemic villages of Villupuram district, Tamil Nadu, India through interviews using a structured, pretested questionnaire. The grading and adenolymphangitis (ADL) attack determination were done by a clinician. The impact was assessed in terms of changes in the lymphedema grades, frequency of ADL attacks, and changes in the burden. Results: Of the 100 patients, 70% were aware of the program and among them, only 48% were practising MMDP regularly (i.e. two times per day). The majority of them (80%) were taking treatment during ADL attacks. The overall lymphedema grades reversal and progression were observed in 13% and 52% of cases, respectively. Conclusion: This study has revealed that the second arm of the GPELF, “MMDP” has not yielded the desired results as evidenced by the incidence of frequent ADL attacks and advancement of lymphedema grades.
Collapse
|
40
|
Schiefer A, Hübner MP, Krome A, Lämmer C, Ehrens A, Aden T, Koschel M, Neufeld H, Chaverra-Muñoz L, Jansen R, Kehraus S, König GM, Pogorevc D, Müller R, Stadler M, Hüttel S, Hesterkamp T, Wagner K, Pfarr K, Hoerauf A. Corallopyronin A for short-course anti-wolbachial, macrofilaricidal treatment of filarial infections. PLoS Negl Trop Dis 2020; 14:e0008930. [PMID: 33284808 PMCID: PMC7746275 DOI: 10.1371/journal.pntd.0008930] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/17/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Current efforts to eliminate the neglected tropical diseases onchocerciasis and lymphatic filariasis, caused by the filarial nematodes Onchocerca volvulus and Wuchereria bancrofti or Brugia spp., respectively, are hampered by lack of a short-course macrofilaricidal–adult-worm killing–treatment. Anti-wolbachial antibiotics, e.g. doxycycline, target the essential Wolbachia endosymbionts of filariae and are a safe prototype adult-worm-sterilizing and macrofilaricidal regimen, in contrast to standard treatments with ivermectin or diethylcarbamazine, which mainly target the microfilariae. However, treatment regimens of 4–5 weeks necessary for doxycycline and contraindications limit its use. Therefore, we tested the preclinical anti-Wolbachia drug candidate Corallopyronin A (CorA) for in vivo efficacy during initial and chronic filarial infections in the Litomosoides sigmodontis rodent model. CorA treatment for 14 days beginning immediately after infection cleared >90% of Wolbachia endosymbionts from filariae and prevented development into adult worms. CorA treatment of patently infected microfilaremic gerbils for 14 days with 30 mg/kg twice a day (BID) achieved a sustained reduction of >99% of Wolbachia endosymbionts from adult filariae and microfilariae, followed by complete inhibition of filarial embryogenesis resulting in clearance of microfilariae. Combined treatment of CorA and albendazole, a drug currently co-administered during mass drug administrations and previously shown to enhance efficacy of anti-Wolbachia drugs, achieved microfilarial clearance after 7 days of treatment at a lower BID dose of 10 mg/kg CorA, a Human Equivalent Dose of 1.4 mg/kg. Importantly, this combination led to a significant reduction in the adult worm burden, which has not yet been published with other anti-Wolbachia candidates tested in this model. In summary, CorA is a preclinical candidate for filariasis, which significantly reduces treatment times required to achieve sustained Wolbachia depletion, clearance of microfilariae, and inhibition of embryogenesis. In combination with albendazole, CorA is robustly macrofilaricidal after 7 days of treatment and fulfills the Target Product Profile for a macrofilaricidal drug. Infections with filarial roundworms can cause the disfiguring human neglected tropical diseases onchocerciasis and lymphatic filariasis. Treatment of these diseases is limited, as there is no well-tolerated treatment available that kills the adult worms after a short-term regimen. Thus, mass drug administrations (MDA) are performed with drugs that temporarily clear the microfilariae, the filarial offspring, to inhibit the transmission of the disease. As these MDA treatments have to be given 1–2 times per year for many years, the goal to eliminate onchocerciasis and lymphatic filariasis is hampered. In the present study we investigated a novel preclinical candidate for the treatment of filariasis. Corallopyronin A (CorA) is a natural compound that clears the essential Wolbachia endobacteria of filariae. Using the Litomosoides sigmodontis rodent model of filariasis we demonstrated that 2 weeks of CorA treatment clears Wolbachia endosymbionts in vivo, leading to a maintained clearance of microfilariae by inhibition of filarial embryogenesis. Combination therapy of CorA with the MDA drug albendazole allowed lower CorA doses and shortened treatment to 7 days. More importantly, it also led to the death of the adult filariae. Portfolios (Target Product Profiles) of new drugs against filariae should show adult killing efficacy like CorA.
Collapse
Affiliation(s)
- Andrea Schiefer
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Anna Krome
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Bonn, Germany
| | - Christine Lämmer
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Alexandra Ehrens
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Tilman Aden
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Marianne Koschel
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Helene Neufeld
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | | | - Rolf Jansen
- Department Microbial Drugs, Helmholtz Center for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Gabriele M. König
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Domen Pogorevc
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
| | - Rolf Müller
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
| | - Marc Stadler
- Department Microbial Drugs, Helmholtz Center for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Stephan Hüttel
- Department Microbial Drugs, Helmholtz Center for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Thomas Hesterkamp
- Translational Project Management Office (TPMO), German Center for Infection Research, Braunschweig, Germany
| | - Karl Wagner
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Bonn, Germany
| | - Kenneth Pfarr
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
- * E-mail:
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
41
|
Nepal G, Kharel G, Shing YK, Ojha R, Jamarkattel S, Yadav JK, Vyas H, Poudyal D, Sah R. The dangers of mass drug administration of albendazole in Nepal, a Neurocysticercosis-endemic region. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2020; 6:22. [PMID: 33110612 PMCID: PMC7585209 DOI: 10.1186/s40794-020-00122-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/14/2020] [Indexed: 11/21/2022]
Abstract
The majority of cases of Neurocysticercosis (NCC) are asymptomatic. Injudicious use of antihelmintics like albendazole (ALB) can cause cyst degeneration and perilesional inflammation, thus rendering asymptomatic individuals symptomatic with seizures, headache, vascular events, or cerebral edema. Mass drug administration (MDA) using ALB is a very common practice in developing countries like Nepal to contain transmission of soil-transmitted helminths (STH) and lymphatic filariasis (LF). Although the benefits of ALB-based MDA in the general population cannot be undermined, there can be severe consequences in certain groups, especially those with latent NCC. In this commentary, we discuss the effect it may have on such patients, and suggest potential solutions.
Collapse
Affiliation(s)
- Gaurav Nepal
- Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj Rd, Kathmandu, 44600 Nepal
| | - Ghanshyam Kharel
- Department of Neurology, National Academy of Medical Sciences, Mahaboudha, Kathmandu, 44600 Nepal
| | - Yow Ka Shing
- Department of Internal Medicine, National University Hospital, 5 Lower Kent Ridge Rd, Singapore, 119074 Singapore
| | - Rajeev Ojha
- Department of Neurology, Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj Rd, Kathmandu, 44600 Nepal
| | - Sujan Jamarkattel
- Department of Infectious Diseases, Tufts Medical Centre, 800 Washington St, Boston, MA 02111 USA
| | - Jayant Kumar Yadav
- Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj Rd, Kathmandu, 44600 Nepal
| | - Himani Vyas
- All India Institute of Medical Sciences, Sri Aurobindo Marg, Ansari Nagar, New Delhi, Delhi, 110029 India
| | - Dhiraj Poudyal
- Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj Rd, Kathmandu, 44600 Nepal
| | - Ranjit Sah
- Department of Microbiology, Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj Rd, Kathmandu, 44600 Nepal
| |
Collapse
|
42
|
Oxfendazole mediates macrofilaricidal efficacy against the filarial nematode Litomosoides sigmodontis in vivo and inhibits Onchocerca spec. motility in vitro. PLoS Negl Trop Dis 2020; 14:e0008427. [PMID: 32628671 PMCID: PMC7365463 DOI: 10.1371/journal.pntd.0008427] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 07/16/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
A major impediment to eliminate lymphatic filariasis and onchocerciasis is the lack of effective short-course macrofilaricidal drugs or regimens that are proven to be safe for both infections. In this study we tested oxfendazole, an anthelmintic shown to be well tolerated in phase 1 clinical trials. In vitro, oxfendazole exhibited modest to marginal motility inhibition of adult worms of Onchocerca gutturosa, pre-adult worms of Onchocerca volvulus and Onchocerca lienalis microfilariae. In vivo, five days of oral treatments provided sterile cure with up to 100% macrofilaricidal efficacy in the murine Litomosoides sigmodontis model of filariasis. In addition, 10 days of oral treatments with oxfendazole inhibited filarial embryogenesis in patent L. sigmodontis-infected jirds and subsequently led to a protracted but complete clearance of microfilaremia. The macrofilaricidal effect observed in vivo was selective, as treatment with oxfendazole of microfilariae-injected naïve mice was ineffective. Based on pharmacokinetic analysis, the driver of efficacy is the maintenance of a minimal efficacious concentration of approximately 100 ng/ml (based on subcutaneous treatment at 25 mg/kg in mice). From animal models, the human efficacious dose is predicted to range from 1.5 to 4.1 mg/kg. Such a dose has already been proven to be safe in phase 1 clinical trials. Oxfendazole therefore has potential to be efficacious for treatment of human filariasis without causing adverse reactions due to drug-induced microfilariae killing. Onchocerciasis and lymphatic filariasis represent two debilitating filarial diseases that belong to the neglected tropical diseases. The current efforts to eliminate those diseases is hampered by the lack of short-course macrofilaricidal drugs, i.e. drugs that kill the adult worms, or regimens that are proven to be safe for both diseases. In the present study we demonstrate that the anthelmintic drug oxfendazole, currently used in veterinary medicine against intestinal helminths, has excellent efficacy in the Litomosoides sigmodontis rodent model of filariasis. Oxfendazole caused complete clearance of adult filariae after a short oral regimen in vivo. Oxfendazole was not directly active against the circulating filarial progeny, the microfilariae, suggesting that drug-induced serious adverse events due to the clearance of microfilariae are unlikely. Human dose was predicted based on the efficacy in the rodent model, the calculation estimated a low efficacious dose, which has already been shown to be safe in phase 1 clinical trials. Thus, oxfendazole represents a promising drug candidate for the treatment of human filarial diseases such as onchocerciasis and lymphatic filariasis.
Collapse
|
43
|
Prada JM, Davis EL, Touloupou P, Stolk WA, Kontoroupis P, Smith ME, Sharma S, Michael E, de Vlas SJ, Hollingsworth TD. Elimination or Resurgence: Modelling Lymphatic Filariasis After Reaching the 1% Microfilaremia Prevalence Threshold. J Infect Dis 2020; 221:S503-S509. [PMID: 31853554 PMCID: PMC7289550 DOI: 10.1093/infdis/jiz647] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The low prevalence levels associated with lymphatic filariasis elimination pose a challenge for effective disease surveillance. As more countries achieve the World Health Organization criteria for halting mass treatment and move on to surveillance, there is increasing reliance on the utility of transmission assessment surveys (TAS) to measure success. However, the long-term disease outcomes after passing TAS are largely untested. Using 3 well-established mathematical models, we show that low-level prevalence can be maintained for a long period after halting mass treatment and that true elimination (0% prevalence) is usually slow to achieve. The risk of resurgence after achieving current targets is low and is hard to predict using just current prevalence. Although resurgence is often quick (<5 years), it can still occur outside of the currently recommended postintervention surveillance period of 4-6 years. Our results highlight the need for ongoing and enhanced postintervention monitoring, beyond the scope of TAS, to ensure sustained success.
Collapse
Affiliation(s)
- Joaquin M Prada
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Emma L Davis
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, Mathematics Institute and School of Life Sciences, University of Warwick, Coventry, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Headington, Oxford, UK
| | | | - Wilma A Stolk
- Department of Public Health, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Periklis Kontoroupis
- Department of Public Health, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Morgan E Smith
- Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Swarnali Sharma
- Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Edwin Michael
- Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Sake J de Vlas
- Department of Public Health, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - T Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Headington, Oxford, UK
| |
Collapse
|
44
|
Safety and efficacy of co-administered diethylcarbamazine, albendazole and ivermectin during mass drug administration for lymphatic filariasis in Haiti: Results from a two-armed, open-label, cluster-randomized, community study. PLoS Negl Trop Dis 2020; 14:e0008298. [PMID: 32511226 PMCID: PMC7302858 DOI: 10.1371/journal.pntd.0008298] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/18/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
In Haiti, 22 communes still require mass drug administration (MDA) to eliminate lymphatic filariasis (LF) as a public health problem. Several clinical trials have shown that a single oral dose of ivermectin (IVM), diethylcarbamazine (DEC) and albendazole (ALB) (IDA) is more effective than DEC plus ALB (DA) for clearing Wuchereria bancrofti microfilariae (Mf). We performed a cluster-randomized community study to compare the safety and efficacy of IDA and DA in an LF-endemic area in northern Haiti. Ten localities were randomized to receive either DA or IDA. Participants were monitored for adverse events (AE), parasite antigenemia, and microfilaremia. Antigen-positive participants were retested one year after MDA to assess treatment efficacy. Fewer participants (11.0%, 321/2917) experienced at least one AE after IDA compared to DA (17.3%, 491/2844, P<0.001). Most AEs were mild, and the three most common AEs reported were headaches, dizziness and abdominal pain. Serious AEs developed in three participants who received DA. Baseline prevalence for filarial antigenemia was 8.0% (239/3004) in IDA localities and 11.5% (344/2994) in DA localities (<0.001). Of those with positive antigenemia, 17.6% (42/239) in IDA localities and 20.9% (72/344, P = 0.25) in DA localities were microfilaremic. One year after treatment, 84% percent of persons with positive filarial antigen tests at baseline could be retested. Clearance rates for filarial antigenemia were 20.5% (41/200) after IDA versus 25.4% (74/289) after DA (P = 0.3). However, 94.4% (34/36) of IDA recipients and 75.9% (44/58) of DA recipients with baseline microfilaremia were Mf negative at the time of retest (P = 0.02). Thus, MDA with IDA was at least as well tolerated and significantly more effective for clearing Mf compared to the standard DA regimen in this study. Effective MDA coverage with IDA could accelerate the elimination of LF as a public health problem in the 22 communes that still require MDA in Haiti.
Collapse
|
45
|
Burgert-Brucker CR, Zoerhoff KL, Headland M, Shoemaker EA, Stelmach R, Karim MJ, Batcho W, Bougouma C, Bougma R, Benjamin Didier B, Georges N, Marfo B, Lemoine JF, Pangaribuan HU, Wijayanti E, Coulibaly YI, Doumbia SS, Rimal P, Salissou AB, Bah Y, Mwingira U, Nshala A, Muheki E, Shott J, Yevstigneyeva V, Ndayishimye E, Baker M, Kraemer J, Brady M. Risk factors associated with failing pre-transmission assessment surveys (pre-TAS) in lymphatic filariasis elimination programs: Results of a multi-country analysis. PLoS Negl Trop Dis 2020; 14:e0008301. [PMID: 32479495 PMCID: PMC7289444 DOI: 10.1371/journal.pntd.0008301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 06/11/2020] [Accepted: 04/16/2020] [Indexed: 11/20/2022] Open
Abstract
Achieving elimination of lymphatic filariasis (LF) as a public health problem requires a minimum of five effective rounds of mass drug administration (MDA) and demonstrating low prevalence in subsequent assessments. The first assessments recommended by the World Health Organization (WHO) are sentinel and spot-check sites-referred to as pre-transmission assessment surveys (pre-TAS)-in each implementation unit after MDA. If pre-TAS shows that prevalence in each site has been lowered to less than 1% microfilaremia or less than 2% antigenemia, the implementation unit conducts a TAS to determine whether MDA can be stopped. Failure to pass pre-TAS means that further rounds of MDA are required. This study aims to understand factors influencing pre-TAS results using existing programmatic data from 554 implementation units, of which 74 (13%) failed, in 13 countries. Secondary data analysis was completed using existing data from Bangladesh, Benin, Burkina Faso, Cameroon, Ghana, Haiti, Indonesia, Mali, Nepal, Niger, Sierra Leone, Tanzania, and Uganda. Additional covariate data were obtained from spatial raster data sets. Bivariate analysis and multilinear regression were performed to establish potential relationships between variables and the pre-TAS result. Higher baseline prevalence and lower elevation were significant in the regression model. Variables statistically significantly associated with failure (p-value ≤0.05) in the bivariate analyses included baseline prevalence at or above 5% or 10%, use of Filariasis Test Strips (FTS), primary vector of Culex, treatment with diethylcarbamazine-albendazole, higher elevation, higher population density, higher enhanced vegetation index (EVI), higher annual rainfall, and 6 or more rounds of MDA. This paper reports for the first time factors associated with pre-TAS results from a multi-country analysis. This information can help countries more effectively forecast program activities, such as the potential need for more rounds of MDA, and prioritize resources to ensure adequate coverage of all persons in areas at highest risk of failing pre-TAS.
Collapse
Affiliation(s)
| | - Kathryn L. Zoerhoff
- Global Health Division, RTI International, Washington, DC, United States of America
| | - Maureen Headland
- Global Health Division, RTI International, Washington, DC, United States of America
- Global Health, Population, and Nutrition, FHI 360, Washington, DC, United States of America
| | - Erica A. Shoemaker
- Global Health Division, RTI International, Washington, DC, United States of America
| | - Rachel Stelmach
- Global Health Division, RTI International, Washington, DC, United States of America
| | | | - Wilfrid Batcho
- National Control Program of Communicable Diseases, Ministry of Health, Cotonou, Benin
| | - Clarisse Bougouma
- Lymphatic Filariasis Elimination Program, Ministère de la Santé, Ouagadougou, Burkina Faso
| | - Roland Bougma
- Lymphatic Filariasis Elimination Program, Ministère de la Santé, Ouagadougou, Burkina Faso
| | - Biholong Benjamin Didier
- National Onchocerciasis and Lymphatic Filariasis Control Program, Ministry of Health, Yaounde, Cameroon
| | - Nko'Ayissi Georges
- National Onchocerciasis and Lymphatic Filariasis Control Program, Ministry of Health, Yaounde, Cameroon
| | - Benjamin Marfo
- Neglected Tropical Diseases Programme, Ghana Health Service, Accra, Ghana
| | | | | | - Eksi Wijayanti
- National Institute Health Research & Development, Ministry of Health, Jakarta, Indonesia
| | - Yaya Ibrahim Coulibaly
- Filariasis Unit, International Center of Excellence in Research, Faculty of Medicine and Odontostomatology, Bamako, Mali
| | - Salif Seriba Doumbia
- Filariasis Unit, International Center of Excellence in Research, Faculty of Medicine and Odontostomatology, Bamako, Mali
| | - Pradip Rimal
- Epidemiology and Disease Control Division, Department of Health Service, Kathmandu, Nepal
| | | | - Yukaba Bah
- National Neglected Tropical Disease Program, Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Upendo Mwingira
- Neglected Tropical Disease Control Programme, National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Andreas Nshala
- IMA World Health/Tanzania NTD Control Programme, Uppsala University, & TIBA Fellow, Dar es Salaam, Tanzania
| | - Edridah Muheki
- Programme to Eliminate Lymphatic Filariasis, Ministry of Health, Kampala, Uganda
| | - Joseph Shott
- Division of Neglected Tropical Diseases, Office of Infectious Diseases, Bureau for Global Health, USAID, Washington, DC, United States of America
| | - Violetta Yevstigneyeva
- Division of Neglected Tropical Diseases, Office of Infectious Diseases, Bureau for Global Health, USAID, Washington, DC, United States of America
| | - Egide Ndayishimye
- Global Health, Population, and Nutrition, FHI 360, Washington, DC, United States of America
| | - Margaret Baker
- Global Health Division, RTI International, Washington, DC, United States of America
| | - John Kraemer
- Global Health Division, RTI International, Washington, DC, United States of America
- Georgetown University, Washington, DC, United States of America
| | - Molly Brady
- Global Health Division, RTI International, Washington, DC, United States of America
| |
Collapse
|
46
|
Yokoly FN, Zahouli JBZ, Méite A, Opoku M, Kouassi BL, de Souza DK, Bockarie M, Koudou BG. Low transmission of Wuchereria bancrofti in cross-border districts of Côte d'Ivoire: A great step towards lymphatic filariasis elimination in West Africa. PLoS One 2020; 15:e0231541. [PMID: 32282840 PMCID: PMC7153895 DOI: 10.1371/journal.pone.0231541] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/25/2020] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Lymphatic filariasis (LF) is widely endemic in Côte d'Ivoire, and elimination as public health problem (EPHP) is based on annual mass drug administration (MDA) using ivermectin and albendazole. To guide EPHP efforts, we evaluated Wuchereria bancrofti infection indices among humans, and mosquito vectors after four rounds of MDA in four cross-border health districts of Côte d'Ivoire. METHODOLOGY We monitored people and mosquitoes for W. bancrofti infections in the cross-border health districts of Aboisso, Bloléquin, Odienné and Ouangolodougou, Côte d'Ivoire. W. bancrofti circulating filarial antigen (CFA) was identified using filariasis test strips, and antigen-positive individuals were screened for microfilaremia. Moreover, filarial mosquito vectors were sampled using window exit traps and pyrethrum sprays, and identified morphologically at species level. Anopheles gambiae s.l. and Culex quinquefasciatus females were analyzed for W. bancrofti infection using polymerase chain reaction (PCR) technique. PRINCIPAL FINDINGS Overall, we found a substantial decline in W. bancrofti infection indices after four rounds of MDA compared to pre-MDA baseline data. CFA prevalence fell from 3.38-5.50% during pre-MDA to 0.00-1.53% after MDA interventions. No subjects had detectable levels of CFA in Ouangolodougou. Moreover, post-MDA CFA prevalence was very low, and below the 1% elimination threshold in Aboisso (0.19%) and Odienné (0.49%). Conversely, CFA prevalence remained above 1% in Bloléquin (1.53%). W. bancrofti microfilariae (Mf) were not found in Aboisso, Bloléquin, and Ouangolodougou, except for Odienné with low prevalence (0.16%; n = 613) and microfilaremia of 32.0 Mf/mL. No An. gambiae s.l. and Cx. quinquefasciatus pools were infected with W. bancrofti in Bloléquin and Ouangolodougou, while they exhibited low infection rates in Aboisso (1% and 0.07%), and Odienné (0.08% and 0.08%), respectively. CONCLUSIONS In cross-border areas of Côte d'Ivoire, LF infection indices in humans and mosquito vectors substantially declined after four rounds of MDA. CFA prevalence fell under the World Health Organization (WHO)-established threshold (1%) in Aboisso, Ouangolodougou and Odienné. Moreover, W. bancrofti prevalence in mosquitoes was lower than WHO-established threshold (2%) in all areas. This might suggest the interruption of W. bancrofti transmission, and possible MDA cessation. However, a formal transmission assessment survey (TAS) and molecular xenomonitoring in mosquito vectors should be implemented before eventual MDA cessation. However, MDA should pursue in Bloléquin where W. bancrofti infection prevalence remained above 1%. Our results provided important ramifications for LF control efforts towards EPHP in Côte d'Ivoire.
Collapse
Affiliation(s)
- Firmain N. Yokoly
- Unité de Formation et de Recherche Sciences de la Nature, Université Nangui Abrogoua, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Julien B. Z. Zahouli
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
- Centre d’Entomologie Médicale et Vétérinaire, Université Alassane Ouattara, Bouaké, Côte d’Ivoire
| | - Aboulaye Méite
- Programme National de Lutte contre les Maladies Tropicales Négligées à Chimiothérapie Préventive, Ministère de la Santé, Abidjan, Côte d’Ivoire
| | - Millicent Opoku
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
- European & Developing Countries Clinical Trials Partnership, Cape Town, South Africa
| | - Bernard L. Kouassi
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Dziedzom K. de Souza
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Moses Bockarie
- European & Developing Countries Clinical Trials Partnership, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Benjamin G. Koudou
- Unité de Formation et de Recherche Sciences de la Nature, Université Nangui Abrogoua, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| |
Collapse
|
47
|
Abstract
Neglected parasitic helminth diseases such as onchocerciasis and lymphatic filariasis affect an estimated 145 million people worldwide, creating a serious health burden in endemic areas such as sub-Saharan Africa and India. Although these diseases are not usually lethal, these filarial nematodes, transmitted by blood-feeding insect vectors, cause severe debilitation and cause chronic disability to infected individuals. The adult worms can reproduce from 5 to up to 14 years, releasing millions of microfilariae, juvenile worms, over an infected individual's lifetime. The current treatments for controlling human filarial infections is focused on killing microfilariae, the earliest larval stage. Currently, there is an unmet medical need for treatments consisting of a macrofilaricidal regimen, one that targets the adult stage of the parasite, to increase the rate of elimination, allow for safe use in coendemic regions of Onchocerca volvulus and Loa loa, and to provide a rapid method to resolve reinfections. Herein, recent approaches for targeting human filarial diseases are discussed, including direct acting agents to target parasitic nematodes and antibacterial approaches to target the endosymbiotic bacteria, Wolbachia.
Collapse
Affiliation(s)
- Natalie A. Hawryluk
- Bristol-Myers Squibb, Global Health, 10300 Campus Point Drive, San Diego, California 92121, United States
| |
Collapse
|
48
|
Behrend MR, Basáñez MG, Hamley JID, Porco TC, Stolk WA, Walker M, de Vlas SJ. Modelling for policy: The five principles of the Neglected Tropical Diseases Modelling Consortium. PLoS Negl Trop Dis 2020; 14:e0008033. [PMID: 32271755 PMCID: PMC7144973 DOI: 10.1371/journal.pntd.0008033] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Matthew R. Behrend
- Neglected Tropical Diseases, Bill & Melinda Gates Foundation, Seattle, Washington, United States of America
- Blue Well 8, Seattle, Washington, United States of America
- * E-mail:
| | - María-Gloria Basáñez
- MRC Centre for Global Infectious Disease Analysis and London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Jonathan I. D. Hamley
- MRC Centre for Global Infectious Disease Analysis and London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Travis C. Porco
- Francis I. Proctor Foundation for Research in Ophthalmology, Department of Epidemiology and Biostatistics, and Department of Ophthalmology, University of California, San Francisco, United States of America
| | - Wilma A. Stolk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Martin Walker
- London Centre for Neglected Tropical Disease Research, Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, Hertfordshire, United Kingdom
- London Centre for Neglected Tropical Disease Research and Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Sake J. de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | |
Collapse
|
49
|
Hardy M, Samuela J, Kama M, Tuicakau M, Romani L, Whitfeld MJ, King CL, Weil GJ, Grobler AC, Robinson LJ, Kaldor JM, Steer AC. The safety of combined triple drug therapy with ivermectin, diethylcarbamazine and albendazole in the neglected tropical diseases co-endemic setting of Fiji: A cluster randomised trial. PLoS Negl Trop Dis 2020; 14:e0008106. [PMID: 32176703 PMCID: PMC7098623 DOI: 10.1371/journal.pntd.0008106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/26/2020] [Accepted: 01/31/2020] [Indexed: 11/02/2022] Open
Abstract
Lymphatic filariasis has remained endemic in Fiji despite repeated mass drug administration using the well-established and safe combination of diethylcarbamazine and albendazole (DA) since 2002. In certain settings the addition of ivermectin to this combination (IDA) remains a safe strategy and is more efficacious. However, the safety has yet to be described in scabies and soil-transmitted helminth endemic settings like Fiji. Villages of Rotuma and Gau islands were randomised to either DA or IDA. Residents received weight-based treatment unblinded with standard exclusions. Participants were actively found and asked by a nurse about their health daily for the first two days and then asked to seek review for the next five days if unwell. Anyone with severe symptoms were reviewed by a doctor and any serious adverse event was reported to the Medical Monitor and Data Safety Monitoring Board. Of 3612 enrolled and eligible participants, 1216 were randomised to DA and 2396 to IDA. Age and sex in both groups were representative of the population. Over 99% (3598) of participants completed 7 days follow-up. Adverse events were reported by 600 participants (16.7%), distributed equally between treatment groups, with most graded as mild (93.2%). There were three serious adverse events, all judged not attributable to treatment by an independent medical monitor. Fatigue was the most common symptom reported by 8.5%, with headache, dizziness, nausea and arthralgia being the next four most common symptoms. Adverse events were more likely in participants with microfilaremia (43.2% versus 15.7%), but adverse event frequency was not related to the presence of scabies or soil-transmitted helminth infection. IDA has comparable safety to DA with the same frequency of adverse events experienced following community mass drug administration. The presence of co-endemic infections did not increase adverse events. IDA can be used in community programs where preventative chemotherapy is needed for control of lymphatic filariasis and other neglected tropical diseases.
Collapse
Affiliation(s)
- Myra Hardy
- Tropical Diseases Research Group, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Josaia Samuela
- Fiji Ministry of Health and Medical Services, Suva, Fiji
| | - Mike Kama
- Fiji Ministry of Health and Medical Services, Suva, Fiji
| | | | - Lucia Romani
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Margot J. Whitfeld
- St Vincent’s Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | - Christopher L. King
- Centre for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Gary J. Weil
- Washington University, St. Louis, Missouri, United States of America
| | - Anneke C. Grobler
- Tropical Diseases Research Group, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Leanne J. Robinson
- Vector-borne Diseases and Tropical Public Health, Burnet Institute, Melbourne, Victoria, Australia
| | - John M. Kaldor
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew C. Steer
- Tropical Diseases Research Group, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
50
|
Hedtke SM, Kuesel AC, Crawford KE, Graves PM, Boussinesq M, Lau CL, Boakye DA, Grant WN. Genomic Epidemiology in Filarial Nematodes: Transforming the Basis for Elimination Program Decisions. Front Genet 2020; 10:1282. [PMID: 31998356 PMCID: PMC6964045 DOI: 10.3389/fgene.2019.01282] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/21/2019] [Indexed: 11/25/2022] Open
Abstract
Onchocerciasis and lymphatic filariasis are targeted for elimination, primarily using mass drug administration at the country and community levels. Elimination of transmission is the onchocerciasis target and global elimination as a public health problem is the end point for lymphatic filariasis. Where program duration, treatment coverage, and compliance are sufficiently high, elimination is achievable for both parasites within defined geographic areas. However, transmission has re-emerged after apparent elimination in some areas, and in others has continued despite years of mass drug treatment. A critical question is whether this re-emergence and/or persistence of transmission is due to persistence of local parasites-i.e., the result of insufficient duration or drug coverage, poor parasite response to the drugs, or inadequate methods of assessment and/or criteria for determining when to stop treatment-or due to re-introduction of parasites via human or vector movement from another endemic area. We review recent genetics-based research exploring these questions in Onchocerca volvulus, the filarial nematode that causes onchocerciasis, and Wuchereria bancrofti, the major pathogen for lymphatic filariasis. We focus in particular on the combination of genomic epidemiology and genome-wide associations to delineate transmission zones and distinguish between local and introduced parasites as the source of resurgence or continuing transmission, and to identify genetic markers associated with parasite response to chemotherapy. Our ultimate goal is to assist elimination efforts by developing easy-to-use tools that incorporate genetic information about transmission and drug response for more effective mass drug distribution, surveillance strategies, and decisions on when to stop interventions to improve sustainability of elimination.
Collapse
Affiliation(s)
- Shannon M. Hedtke
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Annette C. Kuesel
- Unicef/UNDP/World Bank/World Health Organization Special Programme for Research and Training in Tropical Diseases (TDR), World Health Organization, Geneva, Switzerland
| | - Katie E. Crawford
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Patricia M. Graves
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, QLD, Australia
| | - Michel Boussinesq
- Unité Mixte Internationale 233 "TransVIHMI", Institut de Recherche pour le Développement (IRD), INSERM U1175, University of Montpellier, Montpellier, France
| | - Colleen L. Lau
- Department of Global Health, Research School of Population Health, Australian National University, Acton, ACT, Australia
| | - Daniel A. Boakye
- Parasitology Department, Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - Warwick N. Grant
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|