1
|
Rodríguez-Molino P, González Martínez S, Bustamante Amador J, Mellado-Sola I, Montes Martín L, Falces-Romero I, García López-Hortelano M, Hurtado-Gallego J, Mellado MJ, Grasa C, Sainz T. Schistosomiasis in migrant children and adolescents in a paediatric tropical referral unit in Spain: diagnosis and long-term management challenges. Eur J Pediatr 2024; 183:4457-4465. [PMID: 39143347 DOI: 10.1007/s00431-024-05623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/28/2024] [Accepted: 05/19/2024] [Indexed: 08/16/2024]
Abstract
Globalisation and population movement have led to an increasing number of migrant children residing in areas non-endemic for schistosomiasis. However, diagnosing and managing schistosomiasis in children remain controversial. This study aims to investigate the prevalence of schistosomiasis in migrant children and to describe the diagnostic approach and management strategies, including long-term follow-up, to explore the potential role of serological tests in evaluating treatment response. We conducted a retrospective descriptive study spanning from January 2014-July 2021 at a referral unit for Paediatric Tropical Diseases in Madrid (Spain). The study included patients under 18 years diagnosed with schistosomiasis. Of 679 children screened for schistosomiasis, 73 (10.8%) tested positive. The median age was 16.3 years [IQR 9-17.6], 74% male. The majority originated from Sub-Saharan Africa (47%) and Asia (47%). Only 40% presented with symptoms, with gastrointestinal (18%) and cutaneous (17%) manifestations being the most common. Eosinophilia was observed in 43% (median [IQR]: 1103/mm3 [671-1536]), and ova were visualised in the urine of 2/50 (4.0%). Praziquantel treatment was administered to 92%, and 5 patients required retreatment. Follow-up data were available for 58 (80%) over a median period of 9 months [IQR 6-19.8], revealing a progressive decline in eosinophil count, IgE titres, and ELISA optical density. Conclusion: In this series, the prevalence of schistosomiasis among migrant children was significant (10%), highlighting the importance of including serological tests in migrant health screening. The disease is largely asymptomatic, eosinophilia is often absent, and visualisation of ova in urine is exceedingly rare. Eosinophil count, IgE titres, and ELISA optical density could prove valuable as an initial approach for monitoring inflammation during follow-up assessments. What is Known: • The burden of disease related to schistosomiasis is significant, particulary in children, and it is advisable to screen this vulnerable population. What is New: • Eosinophilia may not be present in parasitic infections, so serological tests are crucial for screening migrant children. • Serological monitoring facilitates long-term management of migrant children with schistosomiasis.
Collapse
Affiliation(s)
- Paula Rodríguez-Molino
- General Paediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, Madrid, Spain
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Autonomous University of Madrid (UAM), Madrid, Spain
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | | | - Jorge Bustamante Amador
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Hospital Paediatrics Department, Childhood Infections, El Escorial Hospital, Madrid, Spain
- Health Centre Guzmán el Bueno, SERMAS, Madrid, Spain
| | - Isabel Mellado-Sola
- General Paediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, Madrid, Spain
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Autonomous University of Madrid (UAM), Madrid, Spain
| | | | - Iker Falces-Romero
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Microbiology and Parasitology, La Paz University Hospital, Madrid, Spain
| | - Milagros García López-Hortelano
- General Paediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, Madrid, Spain
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
| | - Jara Hurtado-Gallego
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - María José Mellado
- General Paediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, Madrid, Spain
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Autonomous University of Madrid (UAM), Madrid, Spain
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
| | - Carlos Grasa
- General Paediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, Madrid, Spain
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain
- Autonomous University of Madrid (UAM), Madrid, Spain
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Talía Sainz
- General Paediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, Madrid, Spain.
- La Paz Hospital Research Institute (IdiPAZ), Madrid, Spain.
- Autonomous University of Madrid (UAM), Madrid, Spain.
- Centro de Investigacion en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain.
| |
Collapse
|
2
|
Tadele T, Astatkie A, Tadesse BT, Makonnen E, Aklillu E, Abay SM. Efficacy and safety of praziquantel treatment against Schistosoma mansoni infection among pre-school age children in southern Ethiopia. Trop Med Health 2023; 51:72. [PMID: 38124206 PMCID: PMC10731898 DOI: 10.1186/s41182-023-00562-4] [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: 09/24/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Preventive chemotherapy with a single dose of praziquantel given to an all-at-risk population through mass drug administration is the cornerstone intervention to control and eliminate schistosomiasis as a public health problem. This intervention mainly targets school age children, and pre-school age children (pre-SAC) are excluded from receiving preventive chemotherapy, partly due to scarcity of data on praziquantel treatment outcomes. METHODS We conducted active efficacy and safety surveillance of praziquantel treatment among 240 Schistosoma mansoni-infected pre-SAC who received a single dose of praziquantel (40 mg/kg) in southern Ethiopia. The study outcomes were egg reduction rates (ERR) and cure rates (CRs) four weeks after treatment using the Kato-Katz technique and treatment-associated adverse events (AEs) that occurred within 8 days post-treatment. RESULTS The overall ERR was 93.3% (WHO reference threshold ≥ 90%), while the CR was 85.2% (95% CI = 80.0-89.5%). Baseline S. mansoni infection intensity was significantly associated with CRs, 100% among light infected than moderate (83.4%) or heavy (29.4%) infected children. An increase of 100 in baseline S. mansoni egg count per gram of stool resulted in a 26% (95% CI: 17%, 34%) reduction in the odds of cure. The incidence of experiencing at least one type of AE was 23.1% (95% CI: 18.0%, 29.0%). Stomachache, diarrhea, and nausea were the most common AEs. AEs were mild-to-moderate grade and transient. Pre-treatment moderate (ARR = 3.2, 95% CI: 1.69, 6.14) or heavy infection intensity (ARR = 6.5, 95% CI: 3.62, 11.52) was a significant predictor of AEs (p < 0.001). Sex, age, or soil-transmitted helminth coinfections were not significant predictors of CR or AEs. CONCLUSIONS Single-dose praziquantel is tolerable and effective against S. mansoni infection among pre-SAC, and associated AEs are mostly mild-to-moderate and transient. However, the reduced CR in heavily infected and AEs in one-fourth of S. mansoni-infected pre-SAC underscores the need for safety and efficacy monitoring, especially in moderate-to-high infection settings. Integrating pre-SACs in the national deworming programs is recommended to accelerate the elimination of schistosomiasis as a public health problem.
Collapse
Affiliation(s)
- Tafese Tadele
- School of Public Health, College of Medicine and Health Sciences, Hawassa University, P.O. Box 1560, Hawassa, Ethiopia
| | - Ayalew Astatkie
- School of Public Health, College of Medicine and Health Sciences, Hawassa University, P.O. Box 1560, Hawassa, Ethiopia
| | - Birkneh Tilahun Tadesse
- Department of Pediatrics, College of Medicine and Health Sciences, Hawassa University, P.O. Box 1560, Hawassa, Ethiopia
| | - Eyasu Makonnen
- Center for Innovative Drug Development and Therapeutic Trials for Africa, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
| | - Eleni Aklillu
- Department of Global Public Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Solomon Mequanente Abay
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
| |
Collapse
|
3
|
N'Goran EK, Odiere MR, Assandé Aka R, Ouattara M, Aka NAD, Ogutu B, Rawago F, Bagchus WM, Bödding M, Kourany-Lefoll E, Tappert A, Yin X, Bezuidenhout D, Badenhorst H, Huber E, Dälken B, Haj-Ali Saflo O. Efficacy, safety, and palatability of arpraziquantel (L-praziquantel) orodispersible tablets in children aged 3 months to 6 years infected with Schistosoma in Côte d'Ivoire and Kenya: an open-label, partly randomised, phase 3 trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:867-876. [PMID: 36893784 DOI: 10.1016/s1473-3099(23)00048-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/15/2022] [Accepted: 01/17/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND WHO has underlined the need for a child-friendly treatment for schistosomiasis, a prevalent parasitic disease in low-income and middle-income countries. After successful phase 1 and 2 trials, we aimed to evaluate the efficacy, safety, palatability, and pharmacokinetics of arpraziquantel (L-praziquantel) orodispersible tablets for preschool-aged children. METHODS This open-label, partly randomised, phase 3 study was conducted at two hospitals in Côte d'Ivoire and Kenya. Children with a minimum bodyweight of 5 kg in those aged 3 months to 2 years and 8 kg in those aged 2-6 years were eligible. In cohort 1, participants aged 4-6 years infected with Schistosoma mansoni were randomly assigned (2:1) to receive a single dose of oral arpraziquantel 50 mg/kg (cohort 1a) or oral praziquantel 40 mg/kg (cohort 1b) using a computer-generated randomisation list. Cohorts 2 (aged 2-3 years) and 3 (aged 3 months to 2 years) infected with S mansoni, and the first 30 participants in cohort 4a (aged 3 months to 6 years) infected with Schistosoma haematobium, received a single dose of oral arpraziquantel 50 mg/kg. After follow-up assessments, arpraziquantel was increased to 60 mg/kg (cohort 4b). Laboratory personnel were masked to the treatment group, screening, and baseline values. S mansoni was detected using a point-of-care circulating cathodic antigen urine cassette test and confirmed using the Kato-Katz method. The primary efficacy endpoint was clinical cure rate at 17-21 days after treatment in cohorts 1a and 1b, measured in the modified intention-to-treat population and calculated using the Clopper-Pearson method. This study is registered with ClinicalTrials.gov, NCT03845140. FINDINGS Between Sept 2, 2019, and Aug 7, 2021, 2663 participants were prescreened and 326 were diagnosed with S mansoni or S haematobium. 288 were enrolled (n=100 in cohort 1a, n=50 in cohort 1b, n=30 in cohort 2, n=18 in cohort 3, n=30 in cohort 4a, and n=60 in cohort 4b), but eight participants received antimalarial drugs and were excluded from the efficacy analyses. The median age was 5·1 years (IQR 4·1-6·0) and 132 (47%) of 280 participants were female and 148 (53%) were male. Cure rates with arpraziquantel were similar to those with praziquantel (87·8% [95% CI 79·6-93·5] in cohort 1a vs 81·3% [67·4-91·1] in cohort 1b). No safety concerns were identified during the study. The most common drug-related treatment-emergent adverse events were abdominal pain (41 [14%] of 288 participants), diarrhoea (27 [9%]), vomiting (16 [6%]), and somnolence (21 [7%]). INTERPRETATION Arpraziquantel, a first-line orodispersible tablet, showed high efficacy and favourable safety in preschool-aged children with schistosomiasis. FUNDING The Global Health Innovative Technology Fund, the European and Developing Countries Clinical Trials Partnership, and the healthcare business of Merck KGaA, Darmstadt, Germany (CrossRef Funder ID: 10.13039/100009945).
Collapse
Affiliation(s)
- Eliézer K N'Goran
- Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire, Kisumu, Kenya
| | - Maurice R Odiere
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | | | - Mamadou Ouattara
- Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire, Kisumu, Kenya
| | - N A David Aka
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Bernhards Ogutu
- Kenya Medical Research Institute, Centre for Clinical Research, Nairobi, Kenya
| | - Fredrick Rawago
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Wilhelmina M Bagchus
- Merck Institute of Pharmacometrics, Merck Serono SA (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | | - Elly Kourany-Lefoll
- Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Aliona Tappert
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | - Xiaoyan Yin
- EMD Serono Research and Development Institute Inc (an affiliate of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| | - Deon Bezuidenhout
- Merck Pty Ltd (an affiliate of Merck KGaA, Darmstadt, Germany), Modderfontein, South Africa
| | - Henk Badenhorst
- Merck Pty Ltd (an affiliate of Merck KGaA, Darmstadt, Germany), Modderfontein, South Africa
| | - Eric Huber
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | | | - Okba Haj-Ali Saflo
- Merck SLU (an affiliate of Merck KGaA, Darmstadt, Germany), Madrid, Spain
| |
Collapse
|
4
|
Saad El-Din MI, Gad EL-Hak HN, Ghobashy MA, Elrayess RA. Parasitological and histopathological studies to the effect of aqueous extract of Moringa oleifera Lam. leaves combined with praziquantel therapy in modulating the liver and spleen damage induced by Schistosoma mansoni to male mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:15548-15560. [PMID: 36169837 PMCID: PMC9908685 DOI: 10.1007/s11356-022-23098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/14/2022] [Indexed: 06/16/2023]
Abstract
This study assessed the effectiveness of an aqueous extract of Moringa Oleifera Lam. leaves (MOL) alone or in combination with praziquantel (PZQ) drug targeting-infected mice with Schistosoma mansoni-induced liver and spleen damage. Mice were divided into eight groups control mice group treated orally with saline. PZQ group: non-infected mice treated orally with 300 mg/kg bwt PZQ three consecutive days. MOL group: non-infected mice treated orally with 150 mg/kg bwt MOL extract for 15 days. PZQ/ MOL group: non-infected mice treated orally with 300 mg/kg bwt PZQ for three consecutive days and 150 mg/kg bwt MOL extract for 15 days. IF group: infected mice with 100 cercariae/mouse of the Egyptian strain of S. mansoni. IF/PZQ group infected mice with S. mansoni cercariae and treated orally with 300 mg/kg bwt PZQ for three consecutive days. IF/MOL group: infected mice with S. mansoni cercariae treated orally with 150 mg/kg bwt MOL extract for 15 days. IF/PZQ +MOL group: infected mice with S. mansoni cercariae treated orally with 300 mg/kg bwt PZQ for three consecutive days and 150 mg/kg bwt MOL extract for 15 days. Blood, liver, spleen, worm, and eggs were collected at the end of the experimental period. Treatment of infected mice with MOL and PZQ together significantly reduced the number of ova/g tissue and eliminated the parasites. In addition, the liver and spleen of infected mice showed less histopathological alteration and immunohistochemical expression of nuclear factor kappa β (NF-Kβ). We can conclude that MOL extract combined with PZ has a curative effect on S. mansoni infection and helped to lessen its pathological effects.
Collapse
Affiliation(s)
| | - Heba N. Gad EL-Hak
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Mahi A. Ghobashy
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Ranwa A. Elrayess
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
5
|
Ncube MV, Kabuyaya M, Chimbari MJ. Patient journey and resources mapping to implement a praziquantel mass drug administration program for children aged 5 years and below in resource-limited settings. Syst Rev 2022; 11:227. [PMID: 36271455 PMCID: PMC9585827 DOI: 10.1186/s13643-022-02087-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/28/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The early childhood development of millions of children in some low- and medium-income countries may be compromised by schistosomiasis infections contracted at the age of 5 years and below. Currently, there are no standard guidelines for treating schistosomiasis in children that are 5 years and younger using praziquantel (PZQ), the only drug that the World Health Organization (WHO) recommends for treating schistosomiasis. The review is on processes and resources involved in the treatment of schistosomiasis in children aged 5 years and below. METHODS An electronic search for peer-reviewed articles published in the period from January 2011 to August 2021 was done in the Academic Search Complete, CINAHL with Full Text, Health Source: Nursing/Academic Edition, and MEDLINE databases via EBSCOHost and Google Scholar databases. The search targeted journals that described the treatment of schistosomiasis in children 5 years and below using praziquantel. RESULTS Thirteen studies met the inclusion criteria. The patient journey for treating schistosomiasis in children aged 5 years old and below using PZQ included the following activities: enrolment of the children into the treatment program; clinical examination; diagnosis; taking anthropometric measurements; feeding the children, making the PZQ palatable to the children; administration of PZQ; and monitoring of side effects. There was also a variation in the resources used to treat children aged 5 and below for schistosomiasis. CONCLUSIONS A PZQ mass drug administration program for children aged 5 years old and below in endemic areas should exclude the diagnosis of schistosomiasis before treatment. The resources required in the treatment process should be affordable, and should not require skills and maintenance resources that are beyond those that are available at the primary healthcare level.
Collapse
Affiliation(s)
- Mhlengi Vella Ncube
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa. .,Medical Affairs Institute, Johannesburg, South Africa.
| | - Muhubiri Kabuyaya
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Moses John Chimbari
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
6
|
Birhanu G. Schistosoma Mansoni Infection and Therapeutic Efficacy of Praziquantel in Preschool-Aged Children. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.103901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Schistosomiasis is one of the extreme scientific results which can be greater usual in sub-Saharan Africa. It impacts growth, bodily fitness, and cognitive function, mainly in children. The maximum not unusual place method to manipulate schistosomiasis is preventive chemotherapy. Though World Health Organization (WHO) recommends praziquantel for the manage and remedy of schistosomiasis, preschool-aged children (PSAC) are excluded from populace remedy applications specifically due to paucity of statistics on reaction rate. The findings of preceding research accomplished to evaluate the impact of chemotherapy on schistosomiasis confirmed a few variation. This indicates the need for in addition research in one of the kind populations.
Collapse
|
7
|
Akrasi W, Brah AS, Essuman MA, Osei V, Boye A. Adverse drug effects among students following mass de-worming exercise involving administration of Praziquantel and Albendazole in KEEA Municipality, Ghana. PLoS Negl Trop Dis 2022; 16:e0010680. [PMID: 36094964 PMCID: PMC9499283 DOI: 10.1371/journal.pntd.0010680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/22/2022] [Accepted: 07/20/2022] [Indexed: 11/18/2022] Open
Abstract
Background
To manage the deleterious effects of parasitic infections such as lymphatic filariasis (LF) and schistosomiasis among school children, most countries including Ghana make use of mass drug administration (MDA). Although MDA has proven effective in reducing worm burden, unfortunately adverse drug effects (ADEs) post-MDA are derailing the gains and also remain poorly monitored. The study assessed incidence and factors associated with ADEs among students following a school-based mass de-worming exercise involving administration of Praziquantel (PZQT) and Albendazole (ADZ) against LF and SCH at Komenda-Edina-Eguafo-Abirem (KEEA) Municipal.
Methodology
After fulfilling all ethical obligations, a total of 598 students aged 5–20 years who received PZQT or ADZ monotherapy or a combination of the two (PZQT + ADZ) as part of the mass de-worming exercise were recruited through quota and random sampling. Bodyweight and height of students were measured and body mass index (BMI) calculated. Students were orally interviewed to obtain information such as age, sex, intake of diet before taking drugs. Subsequently, students were monitored over 24 hours post-MDA for cases of ADEs. Descriptive statistics and logistic regression analysis using SPSS version 26 was used to describe data collected and to determine associations between incidence of ADEs and predictor variables.
Principal findings
Out of the 598 students, 243 (40.64%) represented by 124 males (51.03%) and 119 females (48.97%) with mean (SD) age of 13.43 (2.74) years experienced one or more forms of ADE. In decreasing order, the detected ADEs included headache (64.6%), Abdominal pain (48.6%), fever (30.0%), diarrhea (21.4%) and itching (12.8%). Multivariable statistical analysis showed that age 5–9 years (OR: 2.01, p = 0.041) and underweight (OR: 2.02, p = 0.038) were associated with incidence of ADEs. Compared with students who received combination therapy, students who received ADZ only (OR: 0.05, p < 0.001) and PZQT only (OR: 0.26, p < 0.001) had low cases of ADEs. Gender and diet intake before MDA were not associated with ADE incidence.
Conclusion
ADE incidence was common among students in the KEEA municipality. Age, underweight, and double dosing were associated with increase in ADE incidence, while gender and food intake were not associated with increase in ADE incidence. The Disease Control Unit of the Ghana Health Service should incorporate stringent ADE monitoring in post-MDA surveillance in the National MDA program in order to be able to detect, manage and report ADEs to inform planning for future MDA programs. Such initiatives will help not only in improving effectiveness of MDA programs but also identify high risk groups and exact strategies to reduce negative influence of ADE on MDA coverage and anthelminthic drug compliance.
Collapse
Affiliation(s)
- Wisdom Akrasi
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Augustine Suurinobah Brah
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Mainprice Akuoko Essuman
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Viona Osei
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Alex Boye
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
- * E-mail:
| |
Collapse
|
8
|
Summers S, Bhattacharyya T, Allan F, Stothard JR, Edielu A, Webster BL, Miles MA, Bustinduy AL. A review of the genetic determinants of praziquantel resistance in Schistosoma mansoni: Is praziquantel and intestinal schistosomiasis a perfect match? FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.933097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease (NTD) caused by parasitic trematodes belonging to the Schistosoma genus. The mainstay of schistosomiasis control is the delivery of a single dose of praziquantel (PZQ) through mass drug administration (MDA) programs. These programs have been successful in reducing the prevalence and intensity of infections. Due to the success of MDA programs, the disease has recently been targeted for elimination as a public health problem in some endemic settings. The new World Health Organization (WHO) treatment guidelines aim to provide equitable access to PZQ for individuals above two years old in targeted areas. The scale up of MDA programs may heighten the drug selection pressures on Schistosoma parasites, which could lead to the emergence of PZQ resistant schistosomes. The reliance on a single drug to treat a disease of this magnitude is worrying should drug resistance develop. Therefore, there is a need to detect and track resistant schistosomes to counteract the threat of drug resistance to the WHO 2030 NTD roadmap targets. Until recently, drug resistance studies have been hindered by the lack of molecular markers associated with PZQ resistance. This review discusses recent significant advances in understanding the molecular basis of PZQ action in S. mansoni and proposes additional genetic determinants associated with PZQ resistance. PZQ resistance will also be analyzed in the context of alternative factors that may decrease efficacy within endemic field settings, and the most recent treatment guidelines recommended by the WHO.
Collapse
|
9
|
Kabatende J, Barry A, Mugisha M, Ntirenganya L, Bergman U, Bienvenu E, Aklillu E. Safety of Praziquantel and Albendazole Coadministration for the Control and Elimination of Schistosomiasis and Soil-Transmitted Helminths Among Children in Rwanda: An Active Surveillance Study. Drug Saf 2022; 45:909-922. [PMID: 35819751 PMCID: PMC9360141 DOI: 10.1007/s40264-022-01201-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Introduction School-based preventive chemotherapy (Deworming) with praziquantel and albendazole to control and eliminate schistosomiasis and soil-transmitted helminths as public health problems is recommended by the World Health Organization (WHO). Safety monitoring during mass drug administration (MDA) is imperative but data from sub-Saharan Africa are scarce. Objective The aim of this active safety surveillance study was to identify the incidence, type, severity, and risk factors for adverse events (AEs) following mass administration of praziquantel and albendazole. Methods Overall, 8037 school children aged 5–15 years in Rwanda were enrolled. Baseline sociodemographic, medical history and any pre-existing clinical symptoms were recorded. Participants received a single dose of praziquantel and albendazole during MDA. AEs were actively monitored on days 1, 2, and 7 post MDA. Results Overall, 3196 AEs were reported by 1658 children; 91.3%, 8.4%, and 0.3% of the AEs were mild, moderate, and severe, respectively, and most resolved within 3 days. Headache (21%), dizziness or fainting (15.2 %), nausea (12.8%) and stomach pain (12.2%) were the most common AEs. The overall cumulative incidence of experiencing at least one type of AE was 20.6% (95% confidence interval [CI] 19.7–21.5%), being significantly higher (p < 0.001) in children with pre-MDA clinical events (27.5%, 95% CI 25.4–29.6%) than those without (18.7%, 95% CI 17.7–19.7%). Females, older age, having pre-MDA events, types of food taken before MDA and taking two or more praziquantel tablets were significant predictors of AEs. Conclusions Praziquantel and albendazole MDA is safe and well-tolerated; however, one in five children experience transient mild to moderate, and in few cases severe, AEs. The incidence of AEs varies significantly between sex and age groups. Pharmacovigilance in the MDA program is recommended for timely detection and management of AEs.
Collapse
Affiliation(s)
- Joseph Kabatende
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden.,Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Abbie Barry
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Michael Mugisha
- College of Medicine and Health Sciences, University of Rwanda, KK 737, Kigali, Rwanda
| | - Lazare Ntirenganya
- Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Ulf Bergman
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Emile Bienvenu
- Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda.,College of Medicine and Health Sciences, University of Rwanda, KK 737, Kigali, Rwanda
| | - Eleni Aklillu
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden.
| |
Collapse
|
10
|
Salas-Coronas J, Pérez Pérez A, Roure S, Sánchez Peinador C, Santos Larrégola L, Arranz Izquierdo J, Bocanegra C, García López Hortelano M, García Vázquez E, Moza Moriñigo H, Azkune Galparsoro H. [Consensus document for the management of schistosomiasis in Primary Care]. Aten Primaria 2022; 54:102408. [PMID: 35753207 PMCID: PMC9249679 DOI: 10.1016/j.aprim.2022.102408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/14/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
La esquistosomiasis humana es la enfermedad parasitaria con mayor morbimortalidad a nivel mundial después de la malaria. Es endémica en más de 78 países tropicales y subtropicales, sobre todo de África Subsahariana, estimándose que 236 millones de personas están infectadas. Puede causar graves complicaciones de salud a nivel genitourinario y hepatoesplénico, llegando a ocasionar la muerte de 300.000 personas cada año. El número de casos importados en los países occidentales se ha ido incrementado en los últimos años debido a la llegada de un importante número de migrantes procedentes de regiones endémicas y de un creciente número de viajeros que han visitado las mismas. Por otro lado, recientemente se han comunicado brotes de transmisión autóctona en Córcega (Francia) y Almería (España). Por todos estos aspectos, las autoridades sanitarias europeas han recomendado el cribado serológico de la enfermedad en todas las personas migrantes procedentes de zonas endémicas y que lleven menos de 5 años en Europa. Dado que atención primaria es habitualmente el primer punto de contacto de estas personas con el sistema sanitario, los médicos deben conocer los principales aspectos de la enfermedad, y ser dotados de los medios necesarios para su diagnóstico y tratamiento. Este documento ha sido elaborado por profesionales pertenecientes a 5 sociedades científicas de atención primaria (SEMFyC, SEMG, SEMERGEN), Pediatría (SEIP) y Medicina Tropical y Salud Internacional (SEMTSI), con objeto de establecer unas recomendaciones claras para el diagnóstico y el manejo de la esquistosomiasis en atención primaria.
Collapse
Affiliation(s)
- Joaquín Salas-Coronas
- Unidad de Medicina Tropical, Hospital Universitario Poniente, SEMTSI, El Ejido (Almería), España.
| | - Alejandra Pérez Pérez
- Sección de Vigilancia Epidemiológica, Subdirección de Salud Pública de Zaragoza, SEMFYC, Zaragoza, España
| | - Silvia Roure
- Unidad de Salud Internacional Metropolitana Norte de Barcelona, PROSICS Metropolitana Nord, Servicio de Enfermedades Infecciosas, Hospital Universitari Germans Trias i Pujol, SEMTSI, Badalona, España
| | - Carmen Sánchez Peinador
- Centro de Salud de Cantalejo, Segovia, Grupo de Enfermedades Infecciosas, SEMG, Cantalejo, España
| | | | - Javier Arranz Izquierdo
- CS Escola Graduada, IBSALUT, Institut d'Investigació Sanitària de les Illes Balears. (IDISBA), SEMTSI, Palma de Mallorca, España
| | - Cristina Bocanegra
- Unidad de Medicina Tropical y Salud Internacional Vall d'Hebron-Drassanes. PROSICS Barcelona, Servicio de Enfermedades Infecciosas Hospital Universitario Vall d'Hebron, SEMTSI, Barcelona, España
| | - Milagros García López Hortelano
- Servicio de Pediatría, Enfermedades Infecciosa y Tropicales, Hospital Universitario Infantil La Paz - Hospital Carlos III, CIBERINFEC, ISCIII, SEIP, Madrid, España
| | - Elisa García Vázquez
- Unidad de Enfermedades Infecciosas, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB, Departamento de Medicina. Facultad de Medicina de la Universidad de Murcia, SEMTSI, Murcia, España
| | - Helena Moza Moriñigo
- FEA Medicina Preventiva y Salud Pública, Hospital Universitario Príncipe de Asturias, Madrid, SEMTSI, Alcalá de Henares, España
| | - Harkaitz Azkune Galparsoro
- Servicio Enfermedades Infecciosas, Hospital Universitario de Donostia/Donostia unibertsitate ospitalea, EHU-UPV, Biodonostia, Gipuzkoa, SEMTSI, Donostia-San Sebastian, España
| |
Collapse
|
11
|
|
12
|
Prevalence of Schistosoma mansoni and Associated Risk Factors in Human and Biomphalaria Snails in Ethiopia: A Systematic Review and Meta-analysis. Acta Parasitol 2022; 67:31-48. [PMID: 34259986 DOI: 10.1007/s11686-021-00449-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/28/2021] [Indexed: 01/27/2023]
Abstract
PURPOSE Schistosomiasis is one of the leading chronic parasitic diseases in Ethiopia. We aimed to summarize the prevalence of S. mansoni in human and Biomphalaria snails as well as risk factors in Ethiopia. METHODS Literature search was carried out from Scopus, Google Scholar, Science Direct and PubMed which reported Schistosoma mansoni infection in human and Biomphalaria snails in Ethiopia. The overall prevalence was estimated by a random-effect model and heterogeneities among studies were assessed by I2 test. RESULTS A total of 178,251 participants and 1,097 snails were tested for the presence of S. mansoni in the eligible studies. The pooled prevalence of S. mansoni was 32.5% (95% CI 28.0-37.0) and 15.9% (95% CI - 5.6-37.5) in human and Biomphalaria snails in Ethiopia, respectively. The highest pooled prevalence was 43.2% (95% CI 27.3-59.1) in South Nations and National Peoples region while the lowest pooled prevalence was 25% (95% CI 19.3-30.7) observed in the Afar region. Interestingly, the pooled prevalence of S. mansoni declined from 40.7% (95% CI 33-48.4) to 22.4% (95% CI 18.5-26.3) after the launching of mass drug administration (MDA). The analysis of risk factors showed that swimming habit (OR, 2.78; 95% CI 2.35-3.21) and participation in irrigation (OR, 2.69; 95% CI 1.45-3.73) were independent predictors for S. mansoni infection. CONCLUSION This study revealed that about one-third of human and nearly 16% of Biomphalaria snails were infected with S. mansoni in Ethiopia. This review indicated that the prevalence of S. mansoni declined after the implementation of MDA. This study highlights the importance of further integrated approaches for better control of schistosomiasis in Ethiopia.
Collapse
|
13
|
Hailegebriel T, Nibret E, Munshea A. Efficacy of Praziquantel for the Treatment of Human Schistosomiasis in Ethiopia: A Systematic Review and Meta-Analysis. J Trop Med 2021; 2021:2625255. [PMID: 34966433 PMCID: PMC8712180 DOI: 10.1155/2021/2625255] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/04/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Schistosomiasis is one of the neglected tropical diseases causing a serious human health problem in Ethiopia. Praziquantel is the only drug that has been used for the treatment of human schistosomiasis in the country. In line with this, the efficacy of praziquantel has been evaluated in a few interventional studies in the country, but there is a lack in systematically gathered and analyzed information for policymakers. The aim of this systematic review and meta-analysis was to provide a summary of the efficacy of praziquantel for the treatment of human schistosomiasis in Ethiopia. METHODS We conducted a literature search from ScienceDirect, PubMed/Medlin, and Google Scholar databases. A total of 140 articles published in English from 1980 to June 2021 were accessed and 15 of them were eligible for this meta-analysis. The meta-analysis was conducted using Stata 14 software, "metan command." The heterogeneities among studies were evaluated using I 2 test. RESULTS A total of 140 articles were reviewed, but only 15 of them fulfilled the inclusion criteria. The polled cure rate of 40 mg/kg praziquantel was 89.2% (95% CI: 85.4-93.1) and 93.6% (95% CI: 80.6-106) among Schistosoma mansoni and S. haematobium, respectively. Similarly, the mean egg reduction rates of 40 mg/kg praziquantel were 90.2% and 85% among S. mansoni and S. haematobium infected subjects, respectively. The common adverse events observed after receiving praziquantel include abdominal pain, vomiting, headache, diarrhea, and bloody stool. CONCLUSION This systematic review and meta-analysis has indicated that praziquantel is still an appropriate drug for the treatment of human schistosomiasis in Ethiopia.
Collapse
Affiliation(s)
- Tamirat Hailegebriel
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
- Biotechnology Research Institute (BRI), Bahir Dar University, Bahir Dar, Ethiopia
| | - Endalkachew Nibret
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
- Biotechnology Research Institute (BRI), Bahir Dar University, Bahir Dar, Ethiopia
| | - Abaineh Munshea
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
- Biotechnology Research Institute (BRI), Bahir Dar University, Bahir Dar, Ethiopia
| |
Collapse
|
14
|
Nalugwa A, Tukahebwa EM, Olsen A, Nuwaha F. Regression of Schistosoma mansoni associated morbidity among Ugandan preschool children following praziquantel treatment: A randomised trial. PLoS One 2021; 16:e0259338. [PMID: 34780499 PMCID: PMC8592404 DOI: 10.1371/journal.pone.0259338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/06/2021] [Indexed: 11/18/2022] Open
Abstract
Preschool children suffer from morbidity attributable to Schistosoma mansoni. We compared a single and double dose of praziquantel treatment on the regression of S. mansoni associated morbidity in children less than six years in Uganda. We measured the sizes of spleen and liver as well as liver fibrosis before treatment and 8 months after treatment among children who either received one dose (n = 201) or two doses (n = 184) of praziquantel (standard oral dose of 40 mg/kg body weight). Heamoglobin measurements were also taken. Overall, liver enlargement reduced from 52.2% (95% CI (Confidence interval) 45.1, 59.3) to 17.9% (95% CI 12.9, 23.9) with a single dose and from 48.4 (95% CI 40.9, 55.8) to 17.9% (95% CI 12.7, 24.3) with a double dose and there was no significant difference between the changes in proportion of children with enlarged liver between the two treatment groups. The proportion of children with enlarged spleen was not significantly reduced in the group treated with either one or two doses, 47.8% (95% CI 41.7, 54.9) to 45.3% (95% CI 38.3, 52.4) and 48.4% (95% CI 40.9,55.8) to 40.8% 95% CI 33.6, 48.2), respectively. Liver fibrosis detected among children getting single dose (n = 9) or double doses (n = 13) resolved after treatment with praziquantel. The number of children with low heamoglobin significantly reduced from 51.2% (95% CI 44.1, 58.3) to 0.5% (0.2, 0.8) and 61.4% (95% CI 53.9,68.5) to 1.1% (95% CI 0.1, 3.9) after single and double dose treatment, respectively. These results suggest that there is no evidence of a difference in effect between one dose of praziquantel and two doses in reversing morbidity attributable to S. mansoni among children less than six years of age.
Collapse
Affiliation(s)
- Allen Nalugwa
- Child Health and Development Centre, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Annette Olsen
- Parasitology and Aquatic Diseases, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fred Nuwaha
- Disease Control and Prevention, Makerere University, Kampala, Uganda
| |
Collapse
|
15
|
Mesenchymal Stromal Cells: an Antimicrobial and Host-Directed Therapy for Complex Infectious Diseases. Clin Microbiol Rev 2021; 34:e0006421. [PMID: 34612662 DOI: 10.1128/cmr.00064-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is an urgent need for new antimicrobial strategies for treating complex infections and emerging pathogens. Human mesenchymal stromal cells (MSCs) are adult multipotent cells with antimicrobial properties, mediated through direct bactericidal activity and modulation of host innate and adaptive immune cells. More than 30 in vivo studies have reported on the use of human MSCs for the treatment of infectious diseases, with many more studies of animal MSCs in same-species models of infection. MSCs demonstrate potent antimicrobial effects against the major classes of human pathogens (bacteria, viruses, fungi, and parasites) across a wide range of infection models. Mechanistic studies have yielded important insight into their immunomodulatory and bactericidal activity, which can be enhanced through various forms of preconditioning. MSCs are being investigated in over 80 clinical trials for difficult-to-treat infectious diseases, including sepsis and pulmonary, intra-abdominal, cutaneous, and viral infections. Completed trials consistently report MSCs to be safe and well tolerated, with signals of efficacy against some infectious diseases. Although significant obstacles must be overcome to produce a standardized, affordable, clinical-grade cell therapy, these studies suggest that MSCs may have particular potential as an adjunct therapy in complex or resistant infections.
Collapse
|
16
|
N'Diaye M, Keita BF, Danfakha F, Keita F, Keita G, Senghor CS, Diop B, Diawara L, Bessin F, Vernet C, Barbier D, Dewavrin P, Klotz F. A 12-year follow-up of intestinal schistosomiasis in pre-school-aged children in Assoni Village, Eastern Senegal. Infect Dis Poverty 2021; 10:89. [PMID: 34176498 PMCID: PMC8237420 DOI: 10.1186/s40249-021-00867-8] [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] [Received: 02/24/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022] Open
Abstract
Background To monitor the prevalence of schistosomiasis in school-aged children (SAC), the National Bilharzia Control Program (PNLB) was set up by the Senegalese authorities; however, geographically isolated Bedik ethnic groups that did not benefit from this program were found to be heavily infected with Schistosoma mansoni. This observation led us to implement a new schistosomiasis control program in 2008 under the aegis of the non-governmental organization “Le Kaïcedrat” and in partnership with the PNLB/WHO to monitor the prevalence of schistosomiasis in this area. In the village of Assoni, where 100% of SAC were infected, analysis of the stools of pre-school-aged children (PSAC) showed that they were massively infected, so we decided to focus our program on them. Methods From 2008 to 2020, we (i) monitored the prevalence of S. mansoni in PSAC in Assoni using double-stool smear preparation, (ii) treated the infected PSAC with a standard dose of praziquantel 40 mg/kg, (iii) ran educational campaigns each year in the village, and (iv) built latrines to improve sanitation and reduce schistosomiasis transmission. Linear regression was used to examine the trend in the annual schistosomiasis prevalence and a two-sided of Chi-squared test was used to compare prevalence between the different age groups of PSAC. Results We observed an extremely high prevalence of schistosomiasis (78%) in PSAC before implementation of the program in 2008. Contamination occurred in very young children, as 64.3% of children under 2 years old were infected. Moreover, prevalence increased with age and reached 96.8% in children 4 to < 6 years old. Our annual interventions in Assoni Village raised awareness among villagers that water bodies were areas of significant infestation, allowed the building of 88 latrines and led to a decrease in prevalence in PSAC as only 11% of these children were infected in 2020. Conclusion Our study allowed Assoni to be the first village in Senegal to treat PSAC since 2014, but only on an individual basis. It also shows that schistosomiasis is difficult to eradicate and that multi-sectorial actions are required to keep its prevalence at a low level. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40249-021-00867-8.
Collapse
Affiliation(s)
- Monique N'Diaye
- Normandy University, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France. .,Faculty of Pharmaceutical Sciences, Department of Biodiversity Health, Microbiology, Biotechnology, UNICAEN, Caen, France. .,NGO Le Kaïcedrat, Paris, France.
| | | | | | - Fili Keita
- Health Post, Bandafassi, Kedougou District, Dakar, Senegal
| | - Gérald Keita
- Health Post, Bandafassi, Kedougou District, Dakar, Senegal
| | | | - Bocar Diop
- Programme National de Lutte Contre Les Bilharzioses (PNLB), Ministry of Health, Dakar, Senegal
| | - Lamine Diawara
- Neglected Tropical Diseases, Inter-Country Support Team for West Africa, World Health Organization, Ouagadougou, Burkina Faso
| | - François Bessin
- Faculty of Pharmaceutical Sciences, Department of Biodiversity Health, Microbiology, Biotechnology, UNICAEN, Caen, France.,NGO Le Kaïcedrat, Paris, France
| | - Charlotte Vernet
- Faculty of Pharmaceutical Sciences, Department of Biodiversity Health, Microbiology, Biotechnology, UNICAEN, Caen, France.,NGO Le Kaïcedrat, Paris, France
| | - Dominique Barbier
- Faculty of Pharmaceutical Sciences, Department of Biodiversity Health, Microbiology, Biotechnology, UNICAEN, Caen, France.,NGO Le Kaïcedrat, Paris, France
| | | | - Francis Klotz
- NGO Le Kaïcedrat, Paris, France.,Military Hospital Val-de-Grâce, Paris, France
| |
Collapse
|
17
|
Ghazy RM, Tahoun MM, Abdo SM, El-Badry AA, Hamdy NA. Evaluation of Praziquantel Effectivenss After Decades of Prolonged Use in an Endemic Area in Egypt. Acta Parasitol 2021; 66:81-90. [PMID: 32729003 DOI: 10.1007/s11686-020-00242-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 06/16/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Praziquantel (PZQ) is the preferred drug for schistomiasis treatment because of its safety. As PZQ is used for mass drug administration (MDA) in schistosomiasis endemic areas, the effectiveness of the drug, used solely for decades, should be continuously monitored to detect drug resistance. We aimed to assess the effectiveness of PZQ to cure Schistosoma mansoni infection and to reduce the intensity of infection in an endemic area by estimating the cure rate (CR), egg reduction rate (ERR), and comparing these estimates to the levels recommended by the World Health Organization (WHO). METHODS A total of 342 children aged 5-15 years living in Kafr-El-Sheikh were screened for S. mansoni infection. Stool samples were examined microscopically using Kato-Katz (KK) technique. Among the screened children, 106 children had S. mansoni ova in stool, 100 of them received the first dose of PZQ (40 mg/kg). Four weeks later, 96 of 100 children received the second dose of PZQ. Stool samples, collected 4 weeks after each dose of PZQ, were examined using KK. The effectiveness of PZQ was assessed based on ERR and CR. RESULTS CR after the first dose of PZQ was 66.7%, increased to 79.12% after second dose (X2 = 3.05, P = 0.08). Median egg count before treatment was 30.00 (6.00-744), that significantly decreased after two doses of PZQ to 0.00 (0.00-221.33) (Z = 8.29, P = 0.001). Children aged 10-15 years showed higher CR (91.3%) than those aged 5-9 years (OR = 5.25, CI 1.58-17.40). CONCLUSIONS PZQ is still an effective agent against S. mansoni in endemic areas, achieving a high CR and ERR with predominantly low intensity of infection. Age is a main predictor of response to PZQ.
Collapse
Affiliation(s)
- Ramy Mohamed Ghazy
- Department of Tropical Health, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Mohamed Mostafa Tahoun
- Department of Epidemiology, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Sarah Mohamed Abdo
- Department of Medical Parasitology, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Ayman A El-Badry
- Department of Medical Parasitology, Kasr Al-Ainy Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Noha Alaa Hamdy
- Department of Pharmacy Practice, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| |
Collapse
|
18
|
Fukushige M, Chase-Topping M, Woolhouse MEJ, Mutapi F. Efficacy of praziquantel has been maintained over four decades (from 1977 to 2018): A systematic review and meta-analysis of factors influence its efficacy. PLoS Negl Trop Dis 2021; 15:e0009189. [PMID: 33730095 PMCID: PMC7968639 DOI: 10.1371/journal.pntd.0009189] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/01/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The antihelminthic drug praziquantel has been used as the drug of choice for treating schistosome infection for more than 40 years. Although some epidemiological studies have reported low praziquantel efficacy in cure rate (CR) and/or egg reduction rate (ERR), there is no consistent robust evidence of the development of schistosome resistance to praziquantel (PZQ). There is need to determine factors that lead to variable treatment CR and/or ERR. Therefore, we conducted a systematic review and meta-analysis to review CR and ERR as well as identify their predictors. METHODOLOGY/PRINCIPAL FINDINGS In this systematic review and meta-analysis, a literature review was conducted using Biosis Citation Index, Data Citation Index, MEDLINE, and Web of Science Core Collection all of which were provided through Web of Science. Alongside these, EMBASE, and CAB abstracts were searched to identify relevant articles. Random effect meta-regression models were used to identify the factors that influence CR and/or ERR by considering differences in host characteristics and drug dose. In total, 12,127 potential articles were screened and 146 eligible articles (published from 1979 to 2020) were identified and included for the meta-analysis. We found that there has been no significant reduction in CR or ERR over the study period. The results showed more variability in CR, compared with ERR which was more consistent and remained high. The results showed a positive effect of "PZQ treatment dose" with the current recommended dose of 40 mg/kg body weight achieving 57% to 88% CR depending on schistosome species, age of participants, and number of parasitological samples used for diagnosis, and ERR of 95%. CONCLUSIONS/SIGNIFICANCE Based on a review of over 40 years of research there is no evidence to support concerns about schistosomes developing resistance to PZQ. These results indicate that PZQ remains effective in treating schistosomiasis.
Collapse
Affiliation(s)
- Mizuho Fukushige
- Institute of Immunology and Infection Research, Centre for Immunity, Infection & Evolution, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Margo Chase-Topping
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Francisca Mutapi
- Institute of Immunology and Infection Research, Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
19
|
Ramzy RMR, Rabiee A, Abd Elaziz KM, Campbell CH, Kittur N, Colley DG, Haggag AA. Test, Treat, Track, Test, and Treat Active Surveillance toward Elimination of Schistosomiasis: A Feasibility Study. Am J Trop Med Hyg 2020; 103:1572-1577. [PMID: 32662392 PMCID: PMC7543836 DOI: 10.4269/ajtmh.20-0156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We assessed the feasibility of using a test, treat, track, test, and treat (5T) active surveillance strategy to identify and treat individuals with schistosomiasis in three very low-prevalence villages in Kafr El Sheikh Governorate, Egypt. Primary index cases (PICs) were identified using the point-of-care circulating cathodic antigen (POC-CCA) assay in schools, in rural health units (retesting individuals with positive Kato–Katz examinations over the previous 6 months), and at potential water transmission sites identified by PICs and field observations. Primary cases identified potential second-generation cases—people with whom they shared water activities—who were then tracked, tested, and treated if infected. Those sharing water activities with second-generation cases were also tested. The yield of PICs from the three venues were 128 of 3,576 schoolchildren (3.6%), 42 of 696 in rural health units (6.0%), and 83 of 1,156 at water contact sites (7.2%). There were 118 second- and 19 third-generation cases identified. Persons testing positive were treated with praziquantel. Of 388 persons treated, 368 (94.8%) had posttreatment POC-CCA tests 3–4 weeks after treatment, and 81.8% (301) became negative. The 67 persons remaining positive had negative results after a second treatment. Therefore, all those found positive, treated, and followed up were negative following one or two treatments. Analysis of efforts as expressed in person-hours indicates that 4,459 person-hours were required for these 5T activities, with nearly 65% of that time spent carrying out interviews, treatments, and evaluations following treatment. The 5T strategy appears feasible and acceptable as programs move toward elimination.
Collapse
Affiliation(s)
- Reda M R Ramzy
- National Nutrition Institute, General Organization for Teaching Hospitals and Institutes, Cairo, Egypt
| | - Amal Rabiee
- Ministry of Health and Population, Cairo, Egypt
| | - Khaled M Abd Elaziz
- Department of Community, Environmental and Occupational Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Carl H Campbell
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Nupur Kittur
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Daniel G Colley
- Department of Microbiology, University of Georgia, Athens, Georgia.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | | |
Collapse
|
20
|
Acute pancytopenia due to praziquantel treatment for cerebral cysticercosis: A rare case report. J Infect Chemother 2020; 26:1082-1085. [PMID: 32600852 DOI: 10.1016/j.jiac.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 11/21/2022]
Abstract
Praziquantel (PZQ) has been commonly used to treat diverse parasitic infections for over thirty years. Many studies have confirmed its efficacy for the treatment of cysticercosis and the side effects. We reported a rare case of a 56-year-old Chinese man with cerebral cysticercosis. He had experienced acute pancytopenia two times following PZQ treatment (40 mg/kg per day for five days) and gradually recovered after PZQ withdrawal, which was an adverse effect of PZQ that was not previously reported in the literatures. It is suggested that medical observation and dynamic monitoring of PBC should be maintained throughout the entire PZQ therapy course until two weeks after the drug withdrawal, especially in elderly people and those receiving increasing dosages.
Collapse
|
21
|
Olliaro PL, Coulibaly JT, Garba A, Halleux C, Keiser J, King CH, Mutapi F, N’Goran EK, Raso G, Scherrer AU, Sousa-Figueiredo JC, Stete K, Utzinger J, Vaillant MT. Efficacy and safety of single-dose 40 mg/kg oral praziquantel in the treatment of schistosomiasis in preschool-age versus school-age children: An individual participant data meta-analysis. PLoS Negl Trop Dis 2020; 14:e0008277. [PMID: 32569275 PMCID: PMC7360067 DOI: 10.1371/journal.pntd.0008277] [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: 12/27/2019] [Revised: 07/14/2020] [Accepted: 04/08/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Better knowledge of the efficacy and safety of single-dose 40 mg/kg oral praziquantel in preschool-age children is required, should preventive chemotherapy programs for schistosomiasis be expanded to include this age group. METHODOLOGY We analyzed individual participant-level data from 16 studies (13 single-arm or cohort studies and three randomized trials), amounting to 683 preschool-age children (aged <6 years) and 2,010 school-age children (aged 6-14 years). Children had a documented Schistosoma mansoni or S. haematobium infection, were treated with single 40 mg/kg oral praziquantel, and assessed between 21 and 60 days post-treatment. Efficacy was expressed as arithmetic mean and individual egg reduction rate (ERR) and meta-analyzed using general linear models and mixed models. Safety was summarized using reported adverse events (AEs). PRINCIPAL FINDINGS Preschool-age children had significantly lower baseline Schistosoma egg counts and more losses to follow-up compared to school-age children. No difference in efficacy was found between preschool- and school-age children using a general linear model of individual-participant ERR with baseline log-transformed egg count as covariate and study, age, and sex as fixed variables, and a mixed model with a random effect on the study. Safety was reported in only four studies (n = 1,128 individuals); few AEs were reported in preschool-age children 4 and 24 hours post-treatment as well as at follow-up. Three severe but not serious AEs were recorded in school-age children during follow-up. CONCLUSIONS/SIGNIFICANCE There is no indication that single-dose 40 mg/kg oral praziquantel would be less efficacious and less safe in preschool-age children compared to school-age children, with the caveat that only few randomized comparisons exist between the two age groups. Preventive chemotherapy might therefore be extended to preschool-age children, with proper monitoring of its efficacy and safety.
Collapse
Affiliation(s)
- Piero L. Olliaro
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jean T. Coulibaly
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Amadou Garba
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Christine Halleux
- UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases (TDR), World Health Organization, Geneva, Switzerland
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Charles H. King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
- Schistosomiasis Consortium for Operational Research and Evaluation, University of Georgia, Athens, Georgia, United States of America
| | - Francisca Mutapi
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - Eliézer K. N’Goran
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Giovanna Raso
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Alexandra U. Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - José Carlos Sousa-Figueiredo
- Department of Life Sciences, Natural History Museum, Wolfson Wellcome Biomedical Laboratories, London, United Kingdom
- Centro de Investigação em Saúde de Angola, Hospital Provincial, Bengo, Angola
| | - Katarina Stete
- Division of Infectious Diseases, Department of Medicine II, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Michel T. Vaillant
- Centre of Competences for Methodology and Statistics, Luxembourg Institute of Health, Strassen, Luxembourg
- * E-mail:
| |
Collapse
|
22
|
Faust CL, Osakunor DNM, Downs JA, Kayuni S, Stothard JR, Lamberton PHL, Reinhard-Rupp J, Rollinson D. Schistosomiasis Control: Leave No Age Group Behind. Trends Parasitol 2020; 36:582-591. [PMID: 32430274 DOI: 10.1016/j.pt.2020.04.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 01/11/2023]
Abstract
Despite accelerating progress towards schistosomiasis control in sub-Saharan Africa, several age groups have been eclipsed by current treatment and monitoring strategies that mainly focus on school-aged children. As schistosomiasis poses a threat to people of all ages, unfortunate gaps exist in current treatment coverage and associated monitoring efforts, preventing subsequent health benefits to preschool-aged children as well as certain adolescents and adults. Expanding access to younger ages through the forthcoming pediatric praziquantel formulation and improving treatment coverage in older ages is essential. This should occur alongside formal inclusion of these groups in large-scale monitoring and evaluation activities. Current omission of these age groups from treatment and monitoring exacerbates health inequities and has long-term consequences for sustainable schistosomiasis control.
Collapse
Affiliation(s)
- Christina L Faust
- Wellcome Centre for Integrative Parasitology, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Derick N M Osakunor
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK
| | - Jennifer A Downs
- Center for Global Health, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sekeleghe Kayuni
- Department of Tropical Disease Biology, Centre for Neglected Tropical Diseases, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; MASM Medi Clinics Limited, Medical Aid Society of Malawi (MASM), Blantyre, Malawi
| | - J Russell Stothard
- Department of Tropical Disease Biology, Centre for Neglected Tropical Diseases, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Poppy H L Lamberton
- Wellcome Centre for Integrative Parasitology, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | | | - David Rollinson
- Global Schistosomiasis Alliance, Natural History Museum, London SW7 5BD, UK
| |
Collapse
|
23
|
Lemos M, Pedro JM, Fançony C, Moura S, Brito M, Nery SV, Sousa CP, Barros H. Schistosomiasis and soil-transmitted helminthiasis preventive chemotherapy: Adverse events in children from 2 to 15 years in Bengo province, Angola. PLoS One 2020; 15:e0229247. [PMID: 32160223 PMCID: PMC7065752 DOI: 10.1371/journal.pone.0229247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/29/2020] [Indexed: 01/29/2023] Open
Abstract
Preventive chemotherapy campaigns with praziquantel and albendazole are being implemented in Angola, as a high priority public health intervention. However, there are no published data regarding adverse events associated with these medications. In this context, we analysed adverse events due to co-administration of praziquantel and albendazole in endemic areas of schistosomiasis and soil-transmitted helminths in Bengo, Angola. In the context of a targeted drug administration, between December 2012 and September 2013, we conducted two surveys after co-administrating single oral doses of praziquantel and albendazole tablets to children 2 to 15 years of age. About 24 hours after each treatment, participants answered a questionnaire about adverse events. At baseline, 605 children (55.0% male; mean age: 9.7 years) were treated; 460 were interviewed and 257 (55.9%) reported at least one adverse event, 62.3% (160/257) of children being infected with schistosoma haematobium. After six months of treatment, among 339 children surveyed, 184 (54.3%) reported adverse events, with 49.5% (91/184) of infected children. Adverse events were most common in preschool-aged children, with no significant difference between genders. The most frequent adverse events in the two surveys were abdominal pain (18.5%, 25.7%), headache (20.9%, 23.0%) and dizziness (15.7%, 19.8%). Children aged 12 to 15 years (adjusted OR = 0.40, p = 0.040) and those with mixed infection (adjusted OR = 0.04, p = 0.011) had lower odds of adverse events. After the second treatment, those with heavy infection (adjusted OR = 2.72, p = 0.018) and aged 9-11 years (adjusted OR = 2.01, p = 0.049) had significantly fewer adverse events. About 2.0% of children experienced severe adverse events. This study adds evidence that preventive chemotherapy for schistosomiasis and soil-transmitted helminths control is safe, but cases of adverse events are expected. Standardized methodologies to discriminate drug-related adverse events from the clinical manifestations of the infections are needed.
Collapse
Affiliation(s)
- Manuel Lemos
- CISA—Centro de Investigação em Saúde de Angola, Caxito, Angola
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Departamento de Saúde Pública, Faculdade de Medicina, Universidade Agostinho Neto, Luanda, Angola
| | - Joao M. Pedro
- CISA—Centro de Investigação em Saúde de Angola, Caxito, Angola
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - Cláudia Fançony
- CISA—Centro de Investigação em Saúde de Angola, Caxito, Angola
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - Sofia Moura
- CISA—Centro de Investigação em Saúde de Angola, Caxito, Angola
| | - Miguel Brito
- CISA—Centro de Investigação em Saúde de Angola, Caxito, Angola
- H&TRC—Health and Technology Research Center, Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisboa, Portugal
| | - Susana Vaz Nery
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Carlos Pinto Sousa
- Departamento de Saúde Pública, Faculdade de Medicina, Universidade Agostinho Neto, Luanda, Angola
| | - Henrique Barros
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
24
|
Dejon-Agobé JC, Edoa JR, Honkpehedji YJ, Zinsou JF, Adégbitè BR, Ngwese MM, Mangaboula A, Lell B, Grobusch MP, Mordmüller B, Adegnika AA. Schistosoma haematobium infection morbidity, praziquantel effectiveness and reinfection rate among children and young adults in Gabon. Parasit Vectors 2019; 12:577. [PMID: 31823806 PMCID: PMC6905022 DOI: 10.1186/s13071-019-3836-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 12/03/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Sub-Saharan Africa carries most of the global burden of schistosomiasis. To optimize disease control and reduce morbidity, precise data are needed for control measures adapted to the local epidemiological situation. The objective of this study is to provide baseline information on schistosomiasis dynamics, including praziquantel (PZQ) treatment outcome in children and young adults living in the vicinity of Lambaréné, Gabon. METHODS Eligible volunteers were included into a prospective longitudinal study. Urine filtration technique was used to detect eggs in urine for schistosomiasis diagnosis. Subjects were treated with 60 mg of PZQ once per month for three consecutive months, and the outcome was assessed by cure rate (CR) and egg reduction rate (ERR). RESULTS A total of 328 volunteers were enrolled in the study with a mean (± SD) age of 12.2 ± 4.7 years-old. The female-to-male ratio was 0.99. Out of 258 participants in total, 45% had schistosomiasis during the survey and 43% presented with heavy infections. The incidences of haematuria and schistosomiasis were 0.11 and 0.17 person-years, respectively. After the first and third dose of PZQ, overall ERR of 93% and 95% were found, respectively; while the CR were 78% and 88%, respectively. Both ERR (100 vs 88%) and CR (90 vs 68%) were higher among females than males after the first dose. The CR increased for both groups after the third dose to 95% and 80%, respectively. After the first PZQ dose, ERR was higher for heavy compared to light infections (94 vs 89%), while the CR was higher for light than for heavy infections (87 vs 59%). After the third PZQ dose, ERR increased only for light infections to 99%, while CR increased to 98% and 75% for light and for heavy infections, respectively. The reinfection rate assessed at a mean of 44.6 weeks post-treatment was 25%. CONCLUSIONS The prevalence of schistosomiasis is moderate in communities living in the vicinity of Lambaréné, where a subpopulation with a high risk of reinfection bears most of the burden of the disease. To improve schistosomiasis control in this scenario, we suggest education of these high-risk groups to seek themselves a one-year PZQ treatment. Trial registration clinicaltrials.gov Identifier NCT02769103. Registered 11 May 2016, retrospectively registered. https://clinicaltrials.gov/ct2/show/NCT02769013.
Collapse
Affiliation(s)
- Jean Claude Dejon-Agobé
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Yabo Josiane Honkpehedji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeannot Fréjus Zinsou
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Ance Mangaboula
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine 1, Medical University of Vienna, Vienna, Austria
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Partner Site, Tübingen, Germany
| | - Martin Peter Grobusch
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Partner Site, Tübingen, Germany
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Partner Site, Tübingen, Germany
| | - Ayôla Akim Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Partner Site, Tübingen, Germany
- German Center for Infection Research, Tübingen, Germany
| |
Collapse
|
25
|
Ncube MV, Chimbari MJ. A prospective risk assessment of the implementation of a schistosomiasis preventive mass drug administration for children aged five years and below in the uMkhanyakude district of KwaZulu-Natal. BMC Health Serv Res 2019; 19:685. [PMID: 31590663 PMCID: PMC6781343 DOI: 10.1186/s12913-019-4507-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/03/2019] [Indexed: 11/17/2022] Open
Abstract
Background Schistosomiasis is endemic in the uMkhanyakude district of KwaZulu-Natal, South Africa. The South Africa Department of Health (DoH) has decided to implement a schistosomiasis preventive mass drug administration program in all affected parts of the country. Quality management is part of the strategic objectives of the treatment program. We conducted a risk assessment and developed guidelines for the quality management of a schistosomiasis preventive treatment program for children aged 5 years and below in the uMkhanyakude District of KwaZulu-Natal. Methods We conducted a scenario planning exercise by interviewing 10 child health experts from the uMkhanyakude Health District to establish potential risks associated with a planned schistosomiasis preventive control treatment program for children aged 5 years old and below. The risks were analyzed using a modified Failure Mode and Effect Analysis (FMEA). An FMEA table was produced to guide the quality management of the planned schistosomiasis preventive control treatment program for children aged 5 years and below in the uMkhanyakude Health District. Results We identified potential risks, failure modes and possible failure corrective/preventive measures in the following activities that would be part of the mass treatment of children aged 5 years and below infected with schistosomiasis in the uMkhanyakude District. These included enrolment of children into the treatment program; general health checks; weight and height measurements; administration of drugs; reporting of side effects and monitoring and evaluation. Conclusion We were able to use FMEA guide quality management and identify potential risks associated with the planned schistosomiasis preventive treatment program for children aged 5 years old and below in the uMkhanyakude District of KwaZulu-Natal. The FMEA for this program will be useful to the quality management of schistosomiasis preventive treatment programs for this age group in other similar settings.
Collapse
Affiliation(s)
- Mhlengi Vella Ncube
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu- Natal, Durban, South Africa.
| | - Moses John Chimbari
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu- Natal, Durban, South Africa
| |
Collapse
|
26
|
Schistosoma mansoni infection among preschool age children attending Erer Health Center, Ethiopia and the response rate to praziquantel. BMC Res Notes 2019; 12:211. [PMID: 30953565 PMCID: PMC6451229 DOI: 10.1186/s13104-019-4246-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/01/2019] [Indexed: 11/19/2022] Open
Abstract
Objective Preschool age children (PSAC) are excluded from community based praziquantel treatment programs mainly due to paucity of evidence on the magnitude of schistosomiasis, efficacy and safety of this treatment in PSAC. The aim of this study is to assess Schistosoma mansoni infection rate and evaluate response to praziquantel in PSAC. A facility based longitudinal study was employed from April to June 2016 at Erer Health Center, Eastern Ethiopia. Stool sample was examined for schistosomiasis in 236 PSAC and repeated after 4 weeks post-treatment in positive individuals. Treatment outcomes were recorded and interpreted. Results Out of the 236 study participants, 59 (25%) were infected with S. mansoni. Praziquantel treatment (40 mg/kg) resulted in 96.4% cure rate and 99.4% egg reduction rate. Children of 3–5 year old were significantly affected with S. mansoni infection. Nausea and fatigue were common mild adverse events within 4 h of treatment however moderate and severe adverse events and allergic reactions were not observed. In conclusion, praziquantel at 40 mg/kg, the dose utilized in standard care for school age children, is tolerable and efficacious in the treatment of S. mansoni infection in PSAC, which calls for the healthcare system to provide appropriate service for this population. Electronic supplementary material The online version of this article (10.1186/s13104-019-4246-8) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Kimani BW, Mbugua AK, Kihara JH, Ng’ang’a M, Njomo DW. Safety, efficacy and acceptability of praziquantel in the treatment of Schistosoma haematobium in pre-school children of Kwale County, Kenya. PLoS Negl Trop Dis 2018; 12:e0006852. [PMID: 30332403 PMCID: PMC6205662 DOI: 10.1371/journal.pntd.0006852] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 10/29/2018] [Accepted: 09/17/2018] [Indexed: 11/18/2022] Open
Abstract
Background The recommended strategy for control of schistosomiasis is preventive chemotherapy with praziquantel (PZQ). Pre-school children (PSC) are excluded from population treatment programs. In high endemic areas, these children are also at risk, and require treatment with PZQ. The Government of Kenya initiated the National School-Based Deworming Programme (NSBDP) where PSC in Early Childhood Development Education (ECDE) Centers are only eligible for treatment with albendazole (ABZ) but not with PZQ. Methodology/Principal findings 400 PSC were enrolled, from 10 randomly selected ECDE Centers in Kwale County, Kenya where children were treated with crushed PZQ tablets mixed with orange juice, at a single dose of 40 mg/kg. Adverse events were assessed 24 hours post-treatment through questionnaires administered to the parents or guardians. Acceptability was determined by observing if the child spat and/ or vomited all or part of the PZQ dose immediately after treatment. Efficacy was assessed by examining urine samples for Schistosoma haematobium eggs in the 5 weeks post-treatment follow-up. Children testing negative for S. haematobium during the follow-up were considered cured. Egg reduction rate (ERR) was calculated as the decrement in the infection intensity (group’s geometric mean egg counts per 10 ml of urine) following treatment expressed as a proportion of the pre-treatment infection intensity. Before treatment, 80 out of the 400 children enrolled in the study tested positive for S. haematobium (20.0% (95% confidence interval (CI) 16.4–24.2%). Of these, 41 had infections of heavy intensity (51.3%) while the rest (48.7%) were of light intensity. Five weeks post-treatment, 10 children who had heavy intensity infection were diagnosed with S. haematobium (prevalence: 2.5% (95% CI 1.5–4.9%). Infection intensities decreased significantly from 45.9 (95% CI: 31.0–68.0) eggs/ 10 ml urine to1.4 (95% CI: 1.1–1.7) eggs/ 10 ml urine during pre-and post-treatment respectively. The ERR was 96.9%. There were no severe adverse events during follow up 24 hours post treatment. Treatment tolerability among the 400 children was high as none of the children spat and/ or vomited as observed in this study. Conclusion/Significance The study revealed that crushed PZQ is safe and effective in the treatment of urogenital schistosomiasis in this age group. It is therefore recommended that PZQ should be administered to the PSC in Kwale County. Control of schistosome infections is through treatment of infected people with a single dose of the anti-helminth drug praziquantel (PZQ) which is safe, highly efficacious, and can reverse schistosome-related morbidity particularly in the early stages of disease progression. However pre-school children are normally excluded due to the belief that these children are not sufficiently exposed to infective water to experience high infection rates. This could lead to clinical manifestation of the disease and the lack of safety data on praziquantel in this age group. Due to this we investigated the safety, efficacy and acceptability of praziquantel in Kwale County, Kenya. We examined urine samples from 400 preschool children. They were treated with crushed praziquantel (40mg/kg) mixed with orange juice and the efficacy of the treatment was determined 5 weeks after treatment. Acceptability was determined by whether the child spat and/ or vomited the treatment through the direct observed treatment (DOT).No child spat or vomited during treatment. Safety of the treatment was assessed by interviewing the parents of the treated children for adverse events (e.g., abdominal pain, dizziness, and headache). The treatment was well tolerated and most of the parasites were cleared by praziquantel.
Collapse
Affiliation(s)
- Bridget W. Kimani
- Center for Microbiology Research (CMR), Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
- Institute of Tropical Medicine and Infectious Diseases, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
- * E-mail:
| | - Amos K. Mbugua
- Institute of Tropical Medicine and Infectious Diseases, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
| | - Jimmy H. Kihara
- Institute of Tropical Medicine and Infectious Diseases, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
- Division of Vector Borne Diseases & Neglected Tropical Diseases (DVBD-NTD), Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Murima Ng’ang’a
- Division of Vector Borne Diseases & Neglected Tropical Diseases (DVBD-NTD), Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Doris W. Njomo
- Eastern and Southern Africa Center of International Parasite Control (ESACIPAC), Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| |
Collapse
|
28
|
Mutsaka-Makuvaza MJ, Matsena-Zingoni Z, Tshuma C, Ray S, Zhou XN, Webster B, Midzi N. Reinfection of urogenital schistosomiasis in pre-school children in a highly endemic district in Northern Zimbabwe: a 12 months compliance study. Infect Dis Poverty 2018; 7:102. [PMID: 30268157 PMCID: PMC6162945 DOI: 10.1186/s40249-018-0483-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
Background In light of the shift to aiming for schistosomiasis elimination, the following are needed: data on reinfection patterns, participation, and sample submission adherence of all high-risk age groups to intervention strategies. This study was conducted to assess prevalence, reinfections along with consecutive participation, sample submission adherence, and effect of treatment on schistosomiasis prevalence in children aged five years and below in an endemic district in Zimbabwe, over one year. Methods The study was conducted from February 2016–February 2017 in Madziwa area, Shamva district. Following community mobilisation, mothers brought their children aged 5 years and below for recruitment at baseline and also urine sample collection at baseline, 3, 6, 9 and 12 months follow up surveys. At each time point, urine was tested for urogenital schistosomiasis by urine filtration and children found positive received treatment. Schistosoma haematobium prevalence, reinfections as well as children participation, and urine sample submission at each visit were assessed at each time point for one year. Results Of the 535 children recruited from the five communities, 169 (31.6%) participated consecutively at all survey points. The highest mean number of samples submitted was 2.9 among communities and survey points. S. haematobium prevalence significantly reduced from 13.3% at baseline to 2.8% at 12 months for all participants and from 24.9% at baseline to 1.8% at 12 months (P < 0.001) for participants coming at all- time points. Among the communities, the highest baseline prevalence was found in Chihuri for both the participants coming consecutively (38.5%, 10/26) and all participants (20.4%, 21/103). Reinfections were significantly high at 9 months follow up survey (P = 0.021) and in Mupfure (P = 0.003). New infections significantly decreased over time (P < 0.001). Logistic regression analysis showed that the risk of acquiring schistosomiasis was high in some communities (P < 0.05). Conclusions S. haematobium infections and reinfections are seasonal and depend on micro-geographical settings. The risk of being infected with schistosomes in pre-school aged children increases with increasing age. Sustained treatment of infected individuals in a community reduces prevalence overtime. Participation compliance at consecutive visits and sample submission adherence are important for effective operational control interventions. Electronic supplementary material The online version of this article (10.1186/s40249-018-0483-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masceline Jenipher Mutsaka-Makuvaza
- Department of Medical Microbiology, College of Health Sciences, University of Zimbabwe, P. O. Box A178, Avondale, Harare, Zimbabwe.,National Institute of Health Research, Ministry of Health and Child Care, P.O. Box CY573, Causeway, Harare, Zimbabwe
| | - Zvifadzo Matsena-Zingoni
- National Institute of Health Research, Ministry of Health and Child Care, P.O. Box CY573, Causeway, Harare, Zimbabwe.,Division of Epidemiology and Biostatistics, School of Public Health, Faculty of Health Sciences, University of Witwatersrand, 27 St Andrews' Road, Parktown, Johannesburg, 2193, South Africa
| | - Cremance Tshuma
- Mashonaland Central Provincial Health Office, Ministry of health and Child Care, Bindura, Zimbabwe
| | - Sunanda Ray
- Department of Community Medicine, College of Health Sciences, University of Zimbabwe, P. O. Box A178, Avondale, Harare, Zimbabwe
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, Shanghai, People's Republic of China
| | - Bonnie Webster
- Parasites and Vectors Division, National History Museum, London, UK
| | - Nicholas Midzi
- Department of Medical Microbiology, College of Health Sciences, University of Zimbabwe, P. O. Box A178, Avondale, Harare, Zimbabwe.
| |
Collapse
|
29
|
Abstract
Schistosomiasis (bilharzia) is a neglected tropical disease caused by parasitic flatworms (blood flukes) of the genus Schistosoma, with considerable morbidity in parts of the Middle East, South America, Southeast Asia and, particularly, in sub-Saharan Africa. Infective larvae grow in an intermediate host (fresh-water snails) before penetrating the skin of the definitive human host. Mature adult worms reside in the mesenteric (Schistosoma mansoni and Schistosoma japonicum) or pelvic (Schistosoma haematobium) veins, where female worms lay eggs, which are secreted in stool or urine. Eggs trapped in the surrounding tissues and organs, such as the liver and bladder, cause inflammatory immune responses (including granulomas) that result in intestinal, hepato-splenic or urogenital disease. Diagnosis requires the detection of eggs in excreta or worm antigens in the serum, and sensitive, rapid, point-of-care tests for populations living in endemic areas are needed. The anti-schistosomal drug praziquantel is safe and efficacious against adult worms of all the six Schistosoma spp. infecting humans; however, it does not prevent reinfection and the emergence of drug resistance is a concern. Schistosomiasis elimination will require a multifaceted approach, including: treatment; snail control; information, education and communication; improved water, sanitation and hygiene; accurate diagnostics; and surveillance-response systems that are readily tailored to social-ecological settings.
Collapse
Affiliation(s)
- Donald P McManus
- Immunology Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia.
| | - David W Dunne
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Moussa Sacko
- Department of Diagnostic and Biomedical Research, Institut National de Recherche en Santé Publique, Bamako, Mali
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Birgitte J Vennervald
- Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Shanghai, People's Republic of China
| |
Collapse
|
30
|
Kabuyaya M, Chimbari MJ, Mukaratirwa S. Efficacy of praziquantel treatment regimens in pre-school and school aged children infected with schistosomiasis in sub-Saharan Africa: a systematic review. Infect Dis Poverty 2018; 7:73. [PMID: 29986763 PMCID: PMC6036702 DOI: 10.1186/s40249-018-0448-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Schistosomiasis is a serious public health burden in sub-Saharan Africa. Praziquantel is the only drug recommended by the World Health Organization to treat both urogenital and intestinal schistosomiasis. The reliance on a single drug to treat a disease with such a huge burden has raised concerns of possible drug resistance mainly in endemic areas. This systematic review was conducted to identify gaps and recent progress on the efficacy of different regimens of praziquantel in treating schistosomiasis among children in sub-Saharan Africa where Schistosoma mansoni and S. haematobium are endemic. MAIN TEXT A literature search of peer-reviewed journals was done on Google Scholar, MEDLINE (under EBSCOhost) and PubMed databases using pre-defined search terms and Boolean operators. The search included studies published from 2008 to 2017 (August) with emphasis on the efficacy of praziquantel on S. haematobium and S. mansoni infections among preschool and school children. Nineteen publications satisfied the inclusion criteria for the review. The studies reviewed were from 10 sub-Saharan African countries and 7/19 of the studies (37%) were conducted in Uganda. Seven studies (37%) focused on Schistosoma mansoni, 6/19 (31.5%) on S. haematobium and another 6 on mixed infection. A single standard dose of 40 mg/kg body weight was the most used regimen (9) followed by the repeated single standard dose assessed for efficacy at 3-4 weeks post-treatment. CONCLUSIONS A repeated standard dose of 40 mg/kg achieved satisfactory efficacy compared to a single dose against both parasite species. However, findings on efficacy of repeated doses in co-infection of S. mansoni and S. haematobium were not conclusive. Praziquantel administrated at 60 mg/kg was slightly more efficacious than the 40 mg/kg standard dose. Minor and transitory side-effects were reported for both regimens. The review indicates that further investigations are necessary to conclusively determine efficacy of praziquantel on coinfection of S. haematobium and S. mansoni to formulate concrete guidelines on the use of repeated doses at 40 or 60 mg/kg for treating schistosomiasis. We recommend the use of the egg reduction rate (ERR) formula recommended by the WHO for assessing praziquantel efficacy in order for the results to be comparable for different regions.
Collapse
Affiliation(s)
- Muhubiri Kabuyaya
- Discipline of Public Health Medicine, Howard College, University of KwaZulu-Natal, P.O Box 4041, Durban, South Africa
| | - Moses John Chimbari
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Samson Mukaratirwa
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
31
|
Freer JB, Bourke CD, Durhuus GH, Kjetland EF, Prendergast AJ. Schistosomiasis in the first 1000 days. THE LANCET. INFECTIOUS DISEASES 2018; 18:e193-e203. [PMID: 29170089 DOI: 10.1016/s1473-3099(17)30490-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 07/02/2017] [Accepted: 07/13/2017] [Indexed: 12/29/2022]
Abstract
Infections during the first 1000 days-the period from conception to a child's second birthday-can have lifelong effects on health, because this is a crucial phase of growth and development. There is increasing recognition of the burden and potential effects of schistosomiasis in women of reproductive age and young children. Exposure to schistosomes during pregnancy can modulate infant immune development and schistosomiasis can occur from early infancy, such that the high disease burden found in adolescents is often due to accumulation of infections with long-lived schistosomes from early life. Women of reproductive age and young children are largely neglected in mass drug administration programmes, but early treatment could avert subsequent disease. We evaluate the evidence that early schistosomiasis has adverse effects on birth, growth, and development. We also discuss the case for expanding public health interventions for schistosomiasis in women of reproductive age and preschool-age children, and the need for further research to evaluate the potential of treating women pre-conception to maximise health across the life course.
Collapse
Affiliation(s)
- Joseph B Freer
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK.
| | - Claire D Bourke
- Blizard Institute, Queen Mary University of London, London, UK
| | - Gunn H Durhuus
- Department of Internal Medicine, Sorlandet Hospital, Kristiansand, Norway
| | - Eyrun F Kjetland
- Norwegian Centre for Imported and Tropical Diseases, Department of Infectious Diseases Ullevaal, Oslo University Hospital, Oslo, Norway; Discipline of Public Health Medicine, Nelson R Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
32
|
Coulibaly JT, Panic G, Silué KD, Kovač J, Hattendorf J, Keiser J. Efficacy and safety of praziquantel in preschool-aged and school-aged children infected with Schistosoma mansoni: a randomised controlled, parallel-group, dose-ranging, phase 2 trial. LANCET GLOBAL HEALTH 2018; 5:e688-e698. [PMID: 28619227 PMCID: PMC5471607 DOI: 10.1016/s2214-109x(17)30187-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/12/2017] [Accepted: 04/20/2017] [Indexed: 11/25/2022]
Abstract
Background Praziquantel has been the drug of choice for schistosomiasis control for more than 40 years, yet surprisingly, the optimal dose for children younger than 4 years is not known. We aimed to assess the efficacy and safety of escalating praziquantel dosages in preschool-aged children (PSAC). Methods We did a randomised controlled, parallel-group, single-blind, dose-ranging, phase 2 trial in PSAC (2–5 years) and school-aged children (SAC; aged 6–15 years) as a comparator group in southern Côte d'Ivoire. Children were randomly assigned (1:1:1:1) to 20 mg/kg, 40 mg/kg, or 60 mg/kg praziquantel or placebo. Participants, investigators, and laboratory technicians were masked to group assignment, while the investigator providing treatment was aware of the treatment group. The primary objective was to estimate the nature of the dose–response relation in terms of cure rate using the Kato Katz technique. Dose–response curves were estimated using Emax models. Available case analysis was done including all participants with primary endpoint data. This trial is registered with International Standard Randomised Controlled Trial, number ISRCTN15280205. Findings Between Nov 11, 2014, and Feb 18, 2015, 660 PSAC and 225 SAC were assessed for eligibility; of whom 161 (24%) PSAC and 180 (80%) SAC had a detectable Schistosoma mansoni infection. 161 PSAC were randomly allocated of whom 154 received treatment: 42 were assigned to 20 mg/kg praziquantel, of whom 40 received treatment; 38 were assigned to 40 mg/kg praziquantel, of whom 38 received treatment; 41 were assigned to 60 mg/kg praziquantel, of whom 39 received treatment; and 40 were assigned to placebo, of whom 37 received placebo. 180 SAC were randomly allocated of whom 177 received treatment: 49 were assigned to 20 mg/kg praziquantel, of whom 47 received treatment; 46 were assigned to 40 mg/kg praziquantel, of whom 46 received treatment; 42 were assigned to 60 mg/kg praziquantel, of whom 42 received treatment; and 43 were assigned to placebo, of whom 43 received treatment. Follow-up (available-case) data were available for 143 PSAC and 174 SAC. In PSAC, the 20 mg/kg dose resulted in cure in 23 children (62%; 95% CI 44·8–77·5), 40 mg/kg in 26 children (72%; 54·8–85·8), 60 mg/kg in 25 children (71%; 53·7–85·4), and placebo in 13 children (37%; 21·5–55·1). In SAC, the 20 mg/kg dose resulted in cure in 14 children (30%; 95% CI 17·7–45·8), 40 mg/kg in 31 children (69%; 53·4–81·8), 60 mg/kg in 34 children (83%; 67·9–92·8), and placebo in five children (12%; 4·0–25·6). For both age groups, the number of adverse events was similar among the three praziquantel treatment groups, with fewer adverse events observed in the placebo groups. The most common adverse events in PSAC were diarrhoea (11 [9%] of 124) and stomach ache (ten [8%]) and in SAC were diarrhoea (50 [28%] of 177), stomach ache (66 [37%]), and vomiting (26 [15%]) 3 h post treatment. No serious adverse events were reported. Interpretation Praziquantel shows a flat dose-response and overall lower efficacy in PSAC compared with in SAC. In the absence of treatment alternatives, a single dose of praziquantel of 40 mg/kg, recommended by the WHO for S mansoni infections in SAC can be endorsed for PSAC in preventive chemotherapy programmes. Funding European Research Council.
Collapse
Affiliation(s)
- Jean T Coulibaly
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland; Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire
| | - Gordana Panic
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Kigbafori D Silué
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire
| | - Jana Kovač
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Jan Hattendorf
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland.
| |
Collapse
|
33
|
Betson M, Clifford S, Stanton M, Kabatereine NB, Stothard JR. Emergence of Nonfalciparum Plasmodium Infection Despite Regular Artemisinin Combination Therapy in an 18-Month Longitudinal Study of Ugandan Children and Their Mothers. J Infect Dis 2018; 217:1099-1109. [PMID: 29325068 PMCID: PMC5939692 DOI: 10.1093/infdis/jix686] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/05/2018] [Indexed: 12/04/2022] Open
Abstract
As part of a longitudinal cohort investigation of intestinal schistosomiasis and malaria in Ugandan children and their mothers on the shorelines of Lakes Victoria and Albert, we documented risk factors and morbidity associated with nonfalciparum Plasmodium infections and the longitudinal dynamics of Plasmodium species in children. Host age, household location, and Plasmodium falciparum infection were strongly associated with nonfalciparum Plasmodium infections, and Plasmodium malariae infection was associated with splenomegaly. Despite regular artemisinin combination therapy treatment, there was a 3-fold rise in P. malariae prevalence, which was not accountable for by increasing age of the child. Worryingly, our findings reveal the consistent emergence of nonfalciparum infections in children, highlighting the complex dynamics underlying multispecies infections here. Given the growing body of evidence that nonfalciparum malaria infections cause significant morbidity, we encourage better surveillance for nonfalciparum Plasmodium infections, particularly in children, with more sensitive DNA detection methods and improved field-based diagnostics.
Collapse
Affiliation(s)
- Martha Betson
- School of Veterinary Medicine, University of Surrey, Guildford
| | - Sarah Clifford
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Michelle Stanton
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - J Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
34
|
Abla N, Keiser J, Vargas M, Reimers N, Haas H, Spangenberg T. Evaluation of the pharmacokinetic-pharmacodynamic relationship of praziquantel in the Schistosoma mansoni mouse model. PLoS Negl Trop Dis 2017; 11:e0005942. [PMID: 28934207 PMCID: PMC5626502 DOI: 10.1371/journal.pntd.0005942] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 10/03/2017] [Accepted: 09/07/2017] [Indexed: 12/27/2022] Open
Abstract
After more than 40 years of use, Praziquantel (PZQ) still remains the drug of choice for the treatment of intestinal and urogenital schistosomiasis. Its anti-parasitic activity resides primarily in the (R)-enantiomer. Hitherto neither the molecular target nor the pharmacokinetic-pharmacodynamic relationship have been fully elucidated. Here we investigated the efficacy and pharmacokinetics of PZQ in the Schistosoma mansoni mouse model to determine the key factors that drive its efficacy. Dose-response studies with racemic PZQ with or without addition of an irreversible pan-cytochrome P450 (CYP) inhibitor, 1-aminobenzotriazole (ABT), were performed. In addition, efficacy of PZQ in the presence of the CYP inducer, dexamethasone (DEX), was determined. Plasma samples were obtained by tail vein bleeding at 4 time points. The (R)-PZQ levels were determined using a LC-MS/MS method. Non-compartmental pharmacokinetic analysis was performed using PKsolver. In addition, experiments using an enhanced in vitro assay were conducted. We found that the use of ABT increased (R)-PZQ plasma exposures in the systemic circulation by ~10 to 20 fold but the latter were not predictive of efficacy. The use of DEX decreased plasma exposures of (R)-PZQ in the systemic circulation by ~10 fold without reducing efficacy. We extrapolated the (R)-PZQ concentrations in mouse portal vein / mesenteric veins from the systemic exposures and found that a free exposure of (R)-PZQ of ~ 20 μM*h in the portal vein was needed to obtain a worm burden reduction >60%. It is suggested that the high (R)-PZQ concentrations available before the hepatic first pass metabolism drive the efficacy against S. mansoni adult worms residing in the mesenteric veins. It is then possible that the current dosing regimen of 40 mg/kg in preventive chemotherapy programs may provide suboptimal concentrations in low-weight patients such as children, due to smaller total amounts of drug administered, and may consequently result in lower cure rates.
Collapse
Affiliation(s)
- Nada Abla
- Merck Global Health Institute, Ares Trading S.A., a subsidiary of Merck KGaA (Darmstadt, Germany), Coinsins, Switzerland
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Mireille Vargas
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Helmut Haas
- helminGuard, Research Center Borstel, Borstel, Germany
| | - Thomas Spangenberg
- Merck Global Health Institute, Ares Trading S.A., a subsidiary of Merck KGaA (Darmstadt, Germany), Coinsins, Switzerland
- * E-mail:
| |
Collapse
|
35
|
Bustinduy AL, Stothard JR, Friedman JF. Paediatric and maternal schistosomiasis: shifting the paradigms. Br Med Bull 2017; 123:115-125. [PMID: 28910994 PMCID: PMC6279106 DOI: 10.1093/bmb/ldx028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/16/2017] [Indexed: 11/13/2022]
Abstract
BACKGROUND In endemic areas, schistosomiasis causes both overt and subclinical disease in young children and their mothers, as well as in returned travellers. SOURCES OF DATA Key recently published literature. AREAS OF AGREEMENT An action plan for paediatric schistosomiasis and female genital schistosomiasis (FGS) is needed with expanded access to praziquantel (PZQ) treatment required. AREAS OF CONTROVERSY Schistosomiasis-related morbidity is underappreciated. Present and future demand for PZQ treatment is bottlenecked, imbalanced and inequitable. Current dosing, treatment algorithms and access plans are suboptimal with treatment stalled during pregnancy. GROWING POINTS Raised dosing of PZQ (>40 mg/kg) is being explored in young children. Surveillance of female genital schistosomiasis FGS is increasing. Use of PZQ in pregnancy is safe and preventive chemotherapy guidelines are being revised in morbidity- and transmission-control settings. AREAS TIMELY FOR DEVELOPING RESEARCH Shifting focus of population-level control to individual-case management. Detection and prevention of FGS within general health services and integration of PZQ treatment for women and children in antenatal clinics. Feasibility studies assessing alternative and expanded access to PZQ treatment to at-risk children and mothers and pregnant women.
Collapse
Affiliation(s)
- Amaya L Bustinduy
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - J Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Jennifer F Friedman
- Center for International Health Research, Rhode Island Hospital, 55 Claverick Street, Suite 101, Providence, RI 02903, USA
| |
Collapse
|
36
|
Mduluza T, Mutapi F. Putting the treatment of paediatric schistosomiasis into context. Infect Dis Poverty 2017; 6:85. [PMID: 28388940 PMCID: PMC5384153 DOI: 10.1186/s40249-017-0300-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/04/2017] [Indexed: 12/19/2022] Open
Abstract
Despite increased international efforts to control schistosomiasis using preventive chemotherapy, several challenges still exist in reaching the target populations. Until recently, preschool-aged children had been excluded from the recommended target population for mass drug administration, i.e. primary school children aged 6-15 years. Our studies and those of others provided the evidence base for the need to treat preschool-aged children that led to recommendations by the World Health Organization to include preschool-aged children in treatment programmes in 2010. The major challenge now lies in the unavailability of a child-size formulation of the appropriate anthelmintic drug, praziquantel.The currently available formulation of praziquantel presents several problems. First, it is a large tablet, making it difficult for young children and infants to swallow it and thus requires its breaking/crushing to allow for safe uptake. Second, it is bitter so it is often mixed with a sweetener to make it palatable for young children. Third, the current formulation of 600 mg does not allow for flexible dose adjustments for this age group. Thus, there is a need to formulate a child-appropriate praziquantel tablet.This paper discusses the target product profile for paediatric praziquantel, as well as knowledge gaps pertinent to the successful control of schistosome infection and disease in preschool-aged children.
Collapse
Affiliation(s)
- Takafira Mduluza
- Biochemistry Department, University of Zimbabwe, P.O. Box MP167, Mount Pleasant, Harare, Zimbabwe
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Francisca Mutapi
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL UK
| |
Collapse
|
37
|
Zwang J, Olliaro P. Efficacy and safety of praziquantel 40 mg/kg in preschool-aged and school-aged children: a meta-analysis. Parasit Vectors 2017; 10:47. [PMID: 28126024 PMCID: PMC5270314 DOI: 10.1186/s13071-016-1958-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 12/27/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Children carry most of the schistosomiasis burden. While school-aged children are the principal target group of preventive chemotherapy with praziquantel, limited information on efficacy and safety exists for preschool-aged children. METHODS Here, we conducted a meta-analysis of clinical trials of praziquantel for treating children with any form of schistosomiasis. Efficacy was reported as cure rate (CR) and egg reduction rates (ERR); statistical corrections were applied based on methodological disparities across trials to derive the predicted geometrical mean ERR (pERRgm). Safety was reported as frequencies of adverse events. RESULTS Forty-seven comparative and non-comparative studies were identified, enrolling 15,549 children of whom 14,340 (92%) were assessed between 3 and 8 weeks post-treatment with praziquantel 40 mg/kg (the WHO-recommended treatment, n = 8,380, 56%) or comparators (n = 5,960, 44%). The median age was 10 years (range 1-19), 11% (n = 1,694) were preschool-aged. The CR and pERRgm with praziquantel 40 mg/kg were respectively: S. haematobium, 73.6% (95% CI: 63.5-81.40, 25 study arms) and 94.7% (95% CI: 92.7-96.4); S. mansoni, 76.4% (95% CI: 71.5-81.0, 34 arms) and 95.3% (95% CI: 94.2-96.2); S. mansoni/S. haematobium, 67.6% (95% CI: 54.1-80.7, 5 arms) and 93.4% (95% CI: 89.9-96.2); S. japonicum, 94.7% (95% CI: 92.2-98.0) and 98.7% (95% CI: 98.3-99.2). Mixed-effect multivariate analysis found no significant difference between preschool- and school-aged children for CR or pERRgm in S. haematobium (P = 0.309 and P = 0.490, respectively) or S. mansoni (P = 0.982 and P = 0.895) after controlling for time of assessment, formulation, intensity of infection and detection method. Praziquantel was reportedly safe at all ages, with only mild reported adverse events which cleared rapidly after treatment. CONCLUSIONS Praziquantel 40 mg/kg was effective at reducing infection intensity in all Schistosoma species without differences between preschool- and school-aged children. However, conclusions should be tempered because of the limited number of preschool-aged children enrolled, disparities in study procedures and limited information made available in publications, as well as the current imperfect test-of-cure. Also, although reportedly well-tolerated, safety was inconsistently assessed. Studies in target groups, individual-data meta-analysis and standardised methodologies are needed for more robust evidence-base.
Collapse
Affiliation(s)
| | - Piero Olliaro
- UNICEF/UNDP/World Bank/WHO Special Programme for Research & Training in Tropical Diseases (TDR), Geneva, Switzerland
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Pharmacokinetics of the Antischistosomal Lead Ozonide OZ418 in Uninfected Mice Determined by Liquid Chromatography-Tandem Mass Spectrometry. Antimicrob Agents Chemother 2016; 60:7364-7371. [PMID: 27697760 DOI: 10.1128/aac.02394-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 09/27/2016] [Indexed: 11/20/2022] Open
Abstract
One of the major neglected tropical diseases, schistosomiasis, is currently treated and controlled with a single drug, praziquantel. The quest for an alternative drug is fueled by the lack of activity of praziquantel against juvenile Schistosoma worms and the fear of emerging resistance. The synthetic ozonide OZ418 has shown high activity against Schistosoma mansoni, S. haematobium, and S. japonicum in vivo, but its drug disposition remains unknown. To bridge this gap, our study determined the basic pharmacokinetic (PK) parameters of a single oral dose (400 mg/kg of body weight) of OZ418 in uninfected mice. First, a simple liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to quantify OZ418 concentrations in mouse plasma was successfully developed and validated according to U.S. FDA guidelines. This method proved to be selective, accurate (93 to 103%), precise (5 to 16%), and devoid of significant matrix effects (90 to 102%) and provided excellent recovery (101 to 102%). A median peak concentration of 190 (range, 185 to 231) μg/ml was reached at 2 h (2 to 3 h) posttreatment. A naive pooled noncompartmental PK analysis estimated a mean area under the plasma concentration-versus-time curve (AUC) of 9,303 μg h/ml (7,039.2 to 11,908.5 μg h/ml) and a half-life of 38.7 h (20 to 64.6 h). Thus, the OZ418 level in plasma remained well above its in vitro 50% inhibitory concentrations (IC50s) of 27.4 μg/ml (adult S. mansoni worms at 72 h) for at least 75 h. Consistently, OZ418 degraded little in plasma at 37°C (<20% in 121 h) and weakly inhibited cytochrome P450 (CYP450) metabolism (IC50 of 37 to 144 μM). Our results provide a first insight into the disposition of OZ418, paving the way for further studies of its biological fate and effect.
Collapse
|
39
|
Bustinduy AL, Friedman JF, Kjetland EF, Ezeamama AE, Kabatereine NB, Stothard JR, King CH. Expanding Praziquantel (PZQ) Access beyond Mass Drug Administration Programs: Paving a Way Forward for a Pediatric PZQ Formulation for Schistosomiasis. PLoS Negl Trop Dis 2016; 10:e0004946. [PMID: 27658198 PMCID: PMC5033572 DOI: 10.1371/journal.pntd.0004946] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Amaya L. Bustinduy
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| | - Jennifer F. Friedman
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Eyrun Floerecke Kjetland
- Norwegian Centre for Imported and Tropical Diseases, Department of Infectious Diseases Ullevaal, Oslo University Hospital, Oslo, Norway
- Discipline of Public Health Medicine, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Amara E. Ezeamama
- Department of Epidemiology and Biostatistics University of Georgia, Athens, Georgia, United States of America
| | | | - J. Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Charles H. King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
40
|
Comparative Assessment of Health Benefits of Praziquantel Treatment of Urogenital Schistosomiasis in Preschool and Primary School-Aged Children. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9162631. [PMID: 27631011 PMCID: PMC5007301 DOI: 10.1155/2016/9162631] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/21/2016] [Accepted: 07/05/2016] [Indexed: 12/15/2022]
Abstract
Schistosomiasis is a major public health problem in Africa. However, it is only recently that its burden has become recognised as a significant component impacting on the health and development of preschool-aged children. A longitudinal study was conducted in Zimbabwean children to determine the effect of single praziquantel treatment on Schistosoma haematobium-related morbidity markers: microhaematuria, proteinuria, and albuminuria. Changes in these indicators were compared in 1-5 years versus 6-10 years age groups to determine if treatment outcomes differed by age. Praziquantel was efficacious at reducing infection 12 weeks after treatment: cure rate = 94.6% (95% CI: 87.9-97.7%). Infection rates remained lower at 12 months after treatment compared to baseline in both age groups. Among treated children, the odds of morbidity at 12 weeks were significantly lower compared to baseline for proteinuria: odds ratio (OR) = 0.54 (95% CI: 0.31-0.95) and albuminuria: OR = 0.05 (95% CI: 0.02-0.14). Microhaematuria significantly reduced 12 months after treatment, and the effect of treatment did not differ by age group: OR = 0.97 (95% CI: 0.50-1.87). In conclusion, praziquantel treatment has health benefits in preschool-aged children exposed to S. haematobium and its efficacy on infection and morbidity is not age-dependent.
Collapse
|
41
|
Population Pharmacokinetics and Pharmacodynamics of Praziquantel in Ugandan Children with Intestinal Schistosomiasis: Higher Dosages Are Required for Maximal Efficacy. mBio 2016; 7:mBio.00227-16. [PMID: 27507822 PMCID: PMC4992966 DOI: 10.1128/mbio.00227-16] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Each year, millions of African children receive praziquantel (PZQ) by mass drug administration (MDA) to treat schistosomiasis at a standard single dose of 40 mg/kg of body weight, a direct extrapolation from studies of adults. A higher dose of 60 mg/kg is also acceptable for refractory cases. We conducted the first PZQ pharmacokinetic (PK) and pharmacodynamic (PD) study in young children comparing dosing. Sixty Ugandan children aged 3 to 8 years old with egg patent Schistosoma mansoni received PZQ at either 40 mg/kg or 60 mg/kg. PK parameters of PZQ racemate and enantiomers (R and S) were quantified. PD outcomes were assessed by standard fecal egg counts and novel schistosome-specific serum (circulating anodic antigen [CAA]) and urine (circulating cathodic antigen [CCA]) antigen assays. Population PK and PD analyses were performed to estimate drug exposure in individual children, and the relationship between drug exposure and parasitological cure was estimated using logistic regression. Monte Carlo simulations were performed to identify better, future dosing regimens. There was marked PK variability between children, but the area under the concentration-time curve (AUC) of PZQ was strongly predictive of the parasitological cure rate (CR). Although no child achieved antigenic cure, which is suggestive of an important residual adult worm burden, higher AUC was associated with greater CAA antigenic decline at 24 days. To optimize the performance of PZQ, analysis of our simulations suggest that higher doses (>60 mg/kg) are needed, particularly in smaller children. Schistosomiasis is a neglected tropical disease, typically associated with chronic morbidity, and its control is a global health priority. Praziquantel (PZQ) is the only available antiparasitic drug and is often given out, as a single oral dose (40 mg/kg), to school-aged children by mass drug administration (MDA) schemes operating within preventive chemotherapy campaigns as endorsed by the World Health Organization (WHO). This current strategy has several limitations. (i) It excludes preschool children who can be patently infected. (ii) It delivers PZQ at a dose directly extrapolated from adult pharmacological studies. To address these problems, we conducted the first pharmacokinetic and pharmacodynamic study of young children within an area of Uganda where Schistosoma mansoni is hyperendemic. Our results demonstrate that a higher dose (>60 mg/kg) is required, especially in smaller children, and draw attention to the need for further optimization of PZQ treatment based on schistosome antigenic assays, which are more sensitive to pharmacodynamic markers.
Collapse
|
42
|
Lo NC, Bogoch II, Blackburn BG, Raso G, N'Goran EK, Coulibaly JT, Becker SL, Abrams HB, Utzinger J, Andrews JR. Comparison of community-wide, integrated mass drug administration strategies for schistosomiasis and soil-transmitted helminthiasis: a cost-effectiveness modelling study. LANCET GLOBAL HEALTH 2016; 3:e629-38. [PMID: 26385302 DOI: 10.1016/s2214-109x(15)00047-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 04/19/2015] [Accepted: 05/14/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND More than 1·5 billion people are affected by schistosomiasis or soil-transmitted helminthiasis. WHO's recommendations for mass drug administration (MDA) against these parasitic infections emphasise treatment of school-aged children, using separate treatment guidelines for these two helminthiases groups. We aimed to evaluate the cost-effectiveness of expanding integrated MDA to the entire community in four settings in Côte d'Ivoire. METHODS We extended previously published, dynamic, age-structured models of helminthiases transmission to simulate costs and disability averted with integrated MDA (of praziquantel and albendazole) for schistosomiasis and soil-transmitted helminthiasis. We calibrated the model to data for prevalence and intensity of species-specific helminth infection from surveys undertaken in four communities in Côte d'Ivoire between March, 1997, and September, 2010. We simulated a 15-year treatment programme with 75% coverage in only school-aged children; school-aged children and preschool-aged children; adults; and the entire community. Treatment costs were estimated at US$0·74 for school-aged children and $1·74 for preschool-aged children and adults. The incremental cost-effectiveness ratio (ICER) was calculated in 2014 US dollars per disability-adjusted life-year (DALY) averted. FINDINGS Expanded community-wide treatment was highly cost effective compared with treatment of only school-aged children (ICER $167 per DALY averted) and WHO guidelines (ICER $127 per DALY averted), and remained highly cost effective even if treatment costs for preschool-aged children and adults were ten times greater than those for school-aged children. Community-wide treatment remained highly cost effective even when elimination of helminth infections was not achieved. These findings were robust across the four diverse communities in Côte d'Ivoire, only one of which would have received annual MDA for both schistosomiasis and soil-transmitted helminthiasis under the latest WHO guidelines. Treatment every 6 months was also highly cost effective in three out of four communities. INTERPRETATION Integrated, community-wide MDA programmes for schistosomiasis and soil-transmitted helminthiasis can be highly cost effective, even in communities with low disease burden in any helminth group. These results support an urgent need to re-evaluate current global guidelines for helminthiases control programmes to include community-wide treatment, increased treatment frequency, and consideration for lowered prevalence thresholds for integrated treatment. FUNDING Stanford University Medical Scholars Programme, Mount Sinai Hospital-University Health Network AMO Innovation Fund.
Collapse
Affiliation(s)
- Nathan C Lo
- Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Isaac I Bogoch
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Division of General Internal Medicine, Toronto General Hospital, University Health Network, Toronto, ON, Canada; Division of Infectious Diseases, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Brian G Blackburn
- Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanna Raso
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Eliézer K N'Goran
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire; Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Jean T Coulibaly
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire; Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Sören L Becker
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Institute of Medical Microbiology and Hygiene, Saarland University, Homburg/Saar, Germany
| | - Howard B Abrams
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Division of General Internal Medicine, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Jürg Utzinger
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
43
|
Walker M, Mabud TS, Olliaro PL, Coulibaly JT, King CH, Raso G, Scherrer AU, Stothard JR, Sousa-Figueiredo JC, Stete K, Utzinger J, Basáñez MG. New approaches to measuring anthelminthic drug efficacy: parasitological responses of childhood schistosome infections to treatment with praziquantel. Parasit Vectors 2016; 9:41. [PMID: 26813154 PMCID: PMC4728951 DOI: 10.1186/s13071-016-1312-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 11/30/2022] Open
Abstract
Background By 2020, the global health community aims to control and eliminate human helminthiases, including schistosomiasis in selected African countries, principally by preventive chemotherapy (PCT) through mass drug administration (MDA) of anthelminthics. Quantitative monitoring of anthelminthic responses is crucial for promptly detecting changes in efficacy, potentially indicative of emerging drug resistance. Statistical models offer a powerful means to delineate and compare efficacy among individuals, among groups of individuals and among populations. Methods We illustrate a variety of statistical frameworks that offer different levels of inference by analysing data from nine previous studies on egg counts collected from African children before and after administration of praziquantel. Results We quantify responses to praziquantel as egg reduction rates (ERRs), using different frameworks to estimate ERRs among population strata, as average responses, and within strata, as individual responses. We compare our model-based average ERRs to corresponding model-free estimates, using as reference the World Health Organization (WHO) 90 % threshold of optimal efficacy. We estimate distributions of individual responses and summarize the variation among these responses as the fraction of ERRs falling below the WHO threshold. Conclusions Generic models for evaluating responses to anthelminthics deepen our understanding of variation among populations, sub-populations and individuals. We discuss the future application of statistical modelling approaches for monitoring and evaluation of PCT programmes targeting human helminthiases in the context of the WHO 2020 control and elimination goals. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1312-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin Walker
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine (St Mary's Campus), Imperial College London, London, W2 1PG, UK. .,London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, Imperial College London, London, W2 1PG, UK.
| | - Tarub S Mabud
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine (St Mary's Campus), Imperial College London, London, W2 1PG, UK. .,Present address: Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| | - Piero L Olliaro
- UNICEF/UNDP/World Bank/WHO Special Programme on Research and Training in Tropical Diseases (TDR), World Health Organization, Av. Appia 20, CH-1211, Geneva 27, Switzerland. .,Centre for Tropical Medicine and Global Health Nuffield Department of Medicine Research Building, University of Oxford Old Road Campus, Roosevelt Drive, Oxford, OX3 7FZ, UK.
| | - Jean T Coulibaly
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland. .,University of Basel, P.O. Box, CH-4003, Basel, Switzerland. .,Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, 22 BP 770, Abidjan 22, Côte d'Ivoire. .,Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303, Abidjan 01, Côte d'Ivoire.
| | - Charles H King
- Center for Global Health and Diseases, Case Western Reserve University, 2109 Adelbert Road, Cleveland, OH, 44106, USA. .,Schistosomiasis Consortium for Operational Research and Evaluation, University of Georgia, 500 D W Brooks Drive, Athens, GA, 30602, USA.
| | - Giovanna Raso
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland. .,University of Basel, P.O. Box, CH-4003, Basel, Switzerland.
| | - Alexandra U Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091, Zurich, Switzerland.
| | - J Russell Stothard
- Department of Parasitology, London School of Tropical Medicine, Liverpool, L3 5QA, UK.
| | - José Carlos Sousa-Figueiredo
- Centro de Investigação em Saúde de Angola (Health Research Center in Angola), Rua direita do Caxito, Hospital Provincial, Bengo, Angola. .,Department of Life Sciences, Natural History Museum, Wolfson Wellcome Biomedical Laboratories, Cromwell Road, London, SW7 5BD, UK.
| | - Katarina Stete
- Center for Infectious Diseases and Travel Medicine, Department of Medicine, University Hospital Freiburg, Hugstetter Strasse 55, D-79106, Freiburg, Germany.
| | - Jürg Utzinger
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland. .,University of Basel, P.O. Box, CH-4003, Basel, Switzerland.
| | - Maria-Gloria Basáñez
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine (St Mary's Campus), Imperial College London, London, W2 1PG, UK. .,London Centre for Neglected Tropical Disease Research, Department of Infectious Disease Epidemiology, Imperial College London, London, W2 1PG, UK.
| |
Collapse
|
44
|
Trastullo R, Dolci LS, Passerini N, Albertini B. Development of flexible and dispersible oral formulations containing praziquantel for potential schistosomiasis treatment of pre-school age children. Int J Pharm 2015; 495:536-550. [PMID: 26386139 DOI: 10.1016/j.ijpharm.2015.09.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/08/2015] [Accepted: 09/11/2015] [Indexed: 01/07/2023]
Abstract
Praziquantel (PZQ), an anthelmintic drug used in developing countries for the treatment of schistosome infections, was processed using the fluid bed wet granulation technology to prepare fast dispersible granules, as an appropriate and flexible dosage form for pre-school-aged children. Granulation experiments were performed incorporating PZQ either in the powder mixture, according to the traditional way, or in the liquid phase containing wetting agents. In the powder mixture several excipients were tested: Flowlac 100 as filler, Galeniq 721 (isomalt) and Neosorb P 100 T (D-sorbitol) as sweeteners and PVP K30 as binder; while in the liquid phase Lutrol F68, Cremophor RH 40 or Tween 80 as surfactants were investigated. Different formulations loaded with 10% w/w (batches 1-8) and 20% w/w of PZQ (batches 9-13) were produced The majority of granules displayed good flow properties and uniform drug content. X-ray powder diffraction showed that PZQ remained in its original crystalline state, while differential scanning calorimetry and Fourier transform-infrared analysis evidenced the formation of chemical interactions among the ingredients. The solubilisation test performed in non-sink condition to reproduce the actual condition in which a child of 4 years takes the medicine revealed that granules quickly formed a very fine suspension in water (dV90=39.9 μm). Although after the granulation process the solubility of raw PZQ was not increased, adding the aqueous suspension to 500 ml of buffer solution of pH 1.5, simulating the fasted state of a child, 50% of the drug was dissolved after 30 min. After granule manipulation with milk and fruit juices, no PZQ degradation was observed during time. Finally, the selected granule formulation provided evidence to be stable even at hot and very humid climate (30°C/75% RH), at least for the examined time.
Collapse
Affiliation(s)
- Ramona Trastullo
- Department of Pharmacy and BioTechnology, University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Luisa Stella Dolci
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Nadia Passerini
- Department of Pharmacy and BioTechnology, University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Beatrice Albertini
- Department of Pharmacy and BioTechnology, University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy.
| |
Collapse
|
45
|
Olliaro PL, Vaillant M, Diawara A, Coulibaly JT, Garba A, Keiser J, King CH, Knopp S, Landouré A, N’Goran EK, Raso G, Scherrer AU, Sousa-Figueiredo JC, Stete K, Zhou XN, Utzinger J. Toward Measuring Schistosoma Response to Praziquantel Treatment with Appropriate Descriptors of Egg Excretion. PLoS Negl Trop Dis 2015; 9:e0003821. [PMID: 26086551 PMCID: PMC4473103 DOI: 10.1371/journal.pntd.0003821] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/10/2015] [Indexed: 01/05/2023] Open
Abstract
Background The control of schistosomiasis emphasizes preventive chemotherapy with praziquantel, which aims at decreasing infection intensity and thus morbidity in individuals, as well as transmission in communities. Standardizing methods to assess treatment efficacy is important to compare trial outcomes across settings, and to monitor program effectiveness consistently. We compared customary methods and looked at possible complementary approaches in order to derive suggestions for standardizing outcome measures. Methodology/Principal Findings We analyzed data from 24 studies conducted at African, Asian, and Latin American sites, enrolling overall 4,740 individuals infected with Schistosoma mansoni, S. haematobium, or S. japonicum, and treated with praziquantel at doses of 40–80 mg/kg. We found that group-based arithmetic and geometric means can be used interchangeably to express egg reduction rates (ERR) only if treatment efficacy is high (>95%). For lower levels of efficacy, ERR estimates are higher with geometric than arithmetic means. Using the distribution of individual responses in egg excretion, 6.3%, 1.7% and 4.3% of the subjects treated for S. haematobium, S. japonicum and S. mansoni infection, respectively, had no reduction in their egg counts (ERR = 0). The 5th, 10th, and 25th centiles of the subjects treated for S. haematobium had individual ERRs of 0%, 49.3%, and 96.5%; the corresponding values for S. japonicum were 75%, 99%, and 99%; and for S. mansoni 18.2%, 65.3%, and 99.8%. Using a single rather than quadruplicate Kato-Katz thick smear excluded 19% of S. mansoni-infected individuals. Whilst the effect on estimating ERR was negligible by individual studies, ERR estimates by arithmetic means were 8% lower with a single measurement. Conclusions/Significance Arithmetic mean calculations of Schistosoma ERR are more sensitive and therefore more appropriate to monitor drug performance than geometric means. However, neither are satisfactory to identify poor responders. Group-based response estimated by arithmetic mean and the distribution of individual ERRs are correlated, but the latter appears to be more apt to detect the presence and to quantitate the magnitude of suboptimal responses to praziquantel. To identify whether a person is infected with parasitic worms, stool or urine samples are examined for worm eggs. The drug praziquantel is used against the parasitic disease schistosomiasis. However, there is no definitive agreement as to how the efficacy of praziquantel is best expressed. We put together a database from various studies of the efficacy of praziquantel against schistosomiasis. Efficacy was measured using customary methods: cure rate (CR: percentage of people with eggs in their stool/urine before treatment who became egg-negative after treatment); and egg reduction rate (ERR; percentage reduction in the number of eggs in the stool/urine after treatment, where the mean number of eggs from all people treated is calculated using either geometric or arithmetic means). We found that arithmetic and geometric means can be used interchangeably only if treatment efficacy is very high; arithmetic means are more sensitive to capture drops in efficacy expressed by ERR. A valid complement for drug efficacy monitoring is to study the distribution of individual responses in egg excretion that allows identifying in a single measure both those who had an adequate response to treatment and those who respond less well; e.g., the 5% of the patients with the lowest ERRs.
Collapse
Affiliation(s)
- Piero L. Olliaro
- UNICEF/UNDP/World Bank/WHO Special Programme on Research and Training in Tropical Diseases (TDR), World Health Organization, Geneva, Switzerland
- Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Michel Vaillant
- Centre of Competence for Methodology and Statistics (CCMS), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Aïssatou Diawara
- Department of Biology, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Jean T. Coulibaly
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Amadou Garba
- University of Basel, Basel, Switzerland
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- Réseau International Schistosomoses, Environnement, Aménagement et Lutte (RISEAL-Niger), Niamey, Niger
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Charles H. King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
- Schistosomiasis Consortium for Operational Research and Evaluation, University of Georgia, Athens, Georgia, United States of America
| | - Stefanie Knopp
- University of Basel, Basel, Switzerland
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- Wolfson Wellcome Biomedical Laboratories, Department of Life Sciences, Natural History Museum, London, United Kingdom
| | - Aly Landouré
- Institut National de Recherche en Santé Publique, Bamako, Mali
| | - Eliézer K. N’Goran
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Giovanna Raso
- University of Basel, Basel, Switzerland
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Alexandra U. Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - José Carlos Sousa-Figueiredo
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Katarina Stete
- University of Basel, Basel, Switzerland
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- Center for Infectious Diseases and Travel Medicine, Department of Medicine, University Hospital Freiburg, Freiburg im Breisgau, Germany
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, People’s Republic of China
- Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, WHO Collaborating Center for Malaria, Schistosomiasis and Filariasis, Shanghai, People’s Republic of China
| | - Jürg Utzinger
- University of Basel, Basel, Switzerland
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| |
Collapse
|
46
|
Davis SM, Wiegand RE, Mulama F, Kareko EI, Harris R, Ochola E, Samuels AM, Rawago F, Mwinzi PM, Fox LM, Odiere MR, Won KY. Morbidity associated with schistosomiasis before and after treatment in young children in Rusinga Island, western Kenya. Am J Trop Med Hyg 2015; 92:952-8. [PMID: 25758651 DOI: 10.4269/ajtmh.14-0346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 01/19/2015] [Indexed: 11/07/2022] Open
Abstract
Schistosoma mansoni infection is a major cause of organomegaly and ultimately liver fibrosis in adults. Morbidity in pre-school-aged children is less defined, and they are currently not included in mass drug administration (MDA) programs for schistosomiasis control. We report results of a study of the association of schistosomiasis with organomegaly in a convenience sample of 201 children under 7 years old in Rusinga, Kenya on two cross-sectional visits, before and after praziquantel treatment. Data included stool examination and serology for schistosomiasis, the Niamey ultrasound protocol to stage hepatosplenic morbidity including organomegaly, and potential confounders including malaria. Unadjusted and adjusted Poisson regressions were performed. The baseline prevalence of schistosomiasis by antibody and/or stool was 80.3%. Schistomiasis was associated with hepatomegaly (adjusted prevalence ratio [aPR] = 1.4; 95% confidence interval [CI]: 1.0-2.1) and splenomegaly (aPR = 2.1; 95% CI: 1.2-3.7). The association with hepatomegaly persisted posttreatment (aPR = 1.4; 95% CI: 1.1-1.6). Schistosomiasis was associated with morbidity in this cohort. Efforts to include young children in mass treatment campaigns should intensify.
Collapse
Affiliation(s)
- Stephanie M Davis
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Ryan E Wiegand
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Fridah Mulama
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Edmund Ireri Kareko
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Robert Harris
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Elizabeth Ochola
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Aaron M Samuels
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Fredrick Rawago
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Pauline M Mwinzi
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - LeAnne M Fox
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Maurice R Odiere
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Kimberly Y Won
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Data Management Activity, Division of Parasitic Diseases and Malaria, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia; Kenya Medical Research Institute, Neglected Tropical Diseases Unit, Kisumu, Kenya; Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Radiology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| |
Collapse
|
47
|
Berkowitz AL, Raibagkar P, Pritt BS, Mateen FJ. Neurologic manifestations of the neglected tropical diseases. J Neurol Sci 2015; 349:20-32. [PMID: 25623803 DOI: 10.1016/j.jns.2015.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/24/2014] [Accepted: 01/02/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND The World Health Organization has identified 17 neglected tropical diseases (NTDs) that disproportionately affect the world's poorest populations. The neurologic aspects of many of these NTDs have received relatively little attention. METHODS A review was performed in PubMed (MedLine) for each NTD by disease name, name of its causative organism, and neurology, neurosurgery, neurologist, brain, spinal cord, peripheral nerve, muscle, nervous system, encephalitis, meningitis, encephalopathy, stroke, neuropathy, and myopathy (1968-Sept. 2013). The Oxford Center for Evidence-based Medicine guidelines were used to determine the level of evidence of neurological involvement and treatment based on the reports identified. RESULTS Neurologic manifestations were reported for all NTDs except yaws. Neurologic involvement was described in systematic reviews for four NTDs (Chagas disease, echinococcosis, rabies, cysticercosis) (levels 2a-3a), retrospective cohort studies for six (dengue, human African trypanosomiasis, leishmaniasis, leprosy, onchocerciasis, schistosomiasis) (levels 2b-3b), case series for one (foodborne trematodiasis) (level 4), and case reports for five (Buruli ulcer, dracunculiasis, filariasis, soil-transmitted helminthes, and trachoma). Level 1 evidence for treatment of neurologic manifestations of NTDs was found for human African trypanosomiasis, leprosy, and cysticercosis and level 2 evidence exists for treatment of neurologic involvement in Chagas disease. For the remaining NTDs, treatment of neurologic complications is described in case series and case reports only. CONCLUSIONS Neurologic manifestations of NTDs cause significant morbidity and mortality, although limited evidence exists on how best to treat these neurologic complications. Increased awareness of neurologic manifestations of the NTDs can increase their early identification and treatment, contributing to ongoing elimination and eradication campaigns.
Collapse
Affiliation(s)
- Aaron L Berkowitz
- Brigham and Women's Hospital, Department of Neurology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Pooja Raibagkar
- Brigham and Women's Hospital, Department of Neurology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Massachusetts General Hospital, Department of Neurology, Boston, MA, United States
| | - Bobbi S Pritt
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, United States
| | - Farrah J Mateen
- Harvard Medical School, Boston, MA, United States; Massachusetts General Hospital, Department of Neurology, Boston, MA, United States.
| |
Collapse
|
48
|
Clinical efficacy and tolerability of praziquantel for intestinal and urinary schistosomiasis-a meta-analysis of comparative and non-comparative clinical trials. PLoS Negl Trop Dis 2014; 8:e3286. [PMID: 25412105 PMCID: PMC4238982 DOI: 10.1371/journal.pntd.0003286] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 09/22/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Extensive use of praziquantel for treatment and control of schistosomiasis requires a comprehensive understanding of efficacy and safety of various doses for different Schistosoma species. METHODOLOGY/PRINCIPAL FINDINGS A systematic review and meta-analysis of comparative and non-comparative trials of praziquantel at any dose for any Schistosoma species assessed within two months post-treatment. Of 273 studies identified, 55 were eligible (19,499 subjects treated with praziquantel, control treatment or placebo). Most studied were in school-aged children (64%), S. mansoni (58%), and the 40 mg/kg dose (56%); 68% of subjects were in Africa. Efficacy was assessed as cure rate (CR, n=17,017) and egg reduction rate (ERR, n=13,007); safety as adverse events (AE) incidence. The WHO-recommended dose of praziquantel 40 mg/kg achieved CRs of 94.7% (95%CI 92.2-98.0) for S. japonicum, 77.1% (68.4-85.1) for S. haematobium, 76.7% (95%CI 71.9-81.2) for S. mansoni, and 63.5% (95%CI 48.2-77.0) for mixed S. haematobium/S. mansoni infections. Using a random-effect meta-analysis regression model, a dose-effect for CR was found up to 40 mg/kg for S. mansoni and 30 mg/kg for S. haematobium. The mean ERR was 95% for S. japonicum, 94.1% for S. haematobium, and 86.3% for S. mansoni. No significant relationship between dose and ERR was detected. Tolerability was assessed in 40 studies (12,435 subjects). On average, 56.9% (95%CI 47.4-67.9) of the subjects receiving praziquantel 40 mg/kg experienced an AE. The incidence of AEs ranged from 2.3% for urticaria to 31.1% for abdominal pain. CONCLUSIONS/SIGNIFICANCE The large number of subjects allows generalizable conclusions despite the inherent limitations of aggregated-data meta-analyses. The choice of praziquantel dose of 40 mg/kg is justified as a reasonable compromise for all species and ages, although in a proportion of sites efficacy may be lower than expected and age effects could not be fully explored.
Collapse
|
49
|
Inobaya MT, Olveda RM, Chau TNP, Olveda DU, Ross AGP. Prevention and control of schistosomiasis: a current perspective. Res Rep Trop Med 2014; 2014:65-75. [PMID: 25400499 PMCID: PMC4231879 DOI: 10.2147/rrtm.s44274] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/23/2014] [Indexed: 11/23/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease that ranks second only to malaria in terms of human suffering in the tropics and subtropics. Five species are known to infect man and there are currently over 240 million people infected worldwide. The cornerstone of control to date has been mass drug administration with 40 mg/kg of praziquantel but there are problems with this approach. Human and bovine vaccines are in various stages of development. Integrated control, targeting the life cycle, is the only approach that will lead to sustainability and future elimination.
Collapse
Affiliation(s)
- Marianette T Inobaya
- Department of Health, Research Institute for Tropical Medicine, Muntinlupa City, Philippines
| | - Remigio M Olveda
- Department of Health, Research Institute for Tropical Medicine, Muntinlupa City, Philippines
| | - Thao NP Chau
- Discipline of Public Health, Flinders University, Adelaide, Australia
| | - David U Olveda
- Griffith Health Institute, School of Medical Sciences, Gold Coast Campus, Griffith University, Southport, Australia
| | - Allen GP Ross
- Griffith Health Institute, School of Medical Sciences, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
50
|
Greenberg RM. Schistosome ABC multidrug transporters: From pharmacology to physiology. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2014; 4:301-9. [PMID: 25516841 PMCID: PMC4266782 DOI: 10.1016/j.ijpddr.2014.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The genuine and hypothesized roles of schistosome ABC transporters are reviewed. Evidence suggesting a role for transporters in schistosome drug susceptibility is discussed. Potential roles of ABC transporters in normal schistosome biology are outlined.
Praziquantel (PZQ) is essentially the only drug currently available for treatment and control of schistosomiasis, a disease affecting hundreds of millions worldwide. Though highly effective overall, PZQ has limitations, most notably its significant lack of activity against immature schistosomes. Furthermore, the availability of only a single drug for a disease of this magnitude makes reports of PZQ-resistant isolates particularly troubling. ATP-binding cassette (ABC) multidrug transporters such as P-glycoprotein (Pgp; ABCB1) are efflux transporters that underlie multidrug resistance (MDR); changes in their expression or structure are also associated with drug resistance in parasites, including helminths. This review will discuss the role these transporters might play in modulating schistosome susceptibility to PZQ, and the implications for developing new or repurposed treatments that enhance the efficacy of PZQ. However, in addition to influencing drug susceptibility, ABC transporters play important roles in several critical physiological functions such as excretion and maintenance of permeability barriers. They also transport signaling molecules with high affinity, and several lines of evidence implicate mammalian transporters in a diverse array of physiological functions, including regulation of immune responses. Like their mammalian counterparts, schistosome ABC transporters appear to be involved in functions critical to the parasite, including excretory activity and reproduction, and we hypothesize that they underlie at least some aspects of parasite–host interactions. Thus, in addition to their potential as targets for enhancers of PZQ susceptibility, these transporters might also serve as candidate targets for agents that disrupt the parasite life cycle and act as antischistosomals on their own.
Collapse
Affiliation(s)
- Robert M Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, PA 19104, USA
| |
Collapse
|