51
|
Shumbej T, Menu S, Girum T, Bekele F, Gebru T, Worku M, Dendir A, Solomon A, Kahase D, Alemayehu M. Impact of annual preventive mass chemotherapy for soil-transmitted helminths among primary school children in an endemic area of Gurage zone: a prospective cross-sectional study. Res Rep Trop Med 2019; 10:109-118. [PMID: 31308788 PMCID: PMC6616309 DOI: 10.2147/rrtm.s208473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/19/2019] [Indexed: 01/01/2023] Open
Abstract
Background and aim School-based preventive mass chemotherapy has been a key component of Ethiopia's national plan for the control of soil-transmitted helminths. Without an impact evaluation on the impact of a deworming program on infection levels, it is unclear whether the deworming program warrants levels of environmental transmission of infection. This study aimed to determine the impact of annual preventive mass chemotherapy for soil-transmitted helminths among schoolchildren in an endemic area of Gurage zone, south-central Ethiopia. Methods A repeated school-based quantitative prospective cross-sectional method was employed. Data were collected from study participants selected using systematic sampling with probability proportional to size at baseline and after annual treatment. Fresh stool samples were collected and processed using the Kato─Katz technique at the Wolkite University parasitology laboratory. SPSS-21 was used for data management and analysis. Changes in parasitological variables after treatment were estimated. Results Overall, 41.1% prevalence and 22.3% mean geometric infection-intensity reduction were found. Reductions in prevalence of Schistosoma mansoni and hookworms were 13.2% and 15.3%, respectively. Similarly, decreases in prevalence were seen in Ascaris lumbricoides and Trichuris trichiura, representing 94.4% and 80.0% reduction rates, respectively, while 25.9% of the children had heavy S. mansoni (≥400 eggs per gram) infections at baseline, which were reduced to 4.5% after annual treatment. Geometric mean infection intensity–reduction rates for hookworms, A. lumbricoides, and T. trichiura were 80.8%, 20.2%, and 96.7%, respectively. Conclusion Annual mass chemotherapy failed to clear soil-transmitted helminths completely in the present study. However, it resulted in a substantial reduction in overall prevalence and infection intensity. Therefore, other than deworming for school children, interventions such as access to improved personal hygiene and environmental hygiene in school should be emphasized to interrupt transmission.
Collapse
Affiliation(s)
- Teha Shumbej
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Sofia Menu
- Department of Medicine, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Tadele Girum
- Department of Public Health, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Fitsum Bekele
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Teklemichael Gebru
- Department of Public Health, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Meron Worku
- Department of Public Health, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Andamlak Dendir
- Department of Public Health, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Absra Solomon
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Daniel Kahase
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| | - Mihret Alemayehu
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wolkite University, Wolkite, Ethiopia
| |
Collapse
|
52
|
Wepnje GB, Anchang-Kimbi JK, Ndassi VD, Lehman LG, Kimbi HK. Schistosoma haematobium infection status and its associated risk factors among pregnant women in Munyenge, South West Region, Cameroon following scale-up of communal piped water sources from 2014 to 2017: a cross-sectional study. BMC Public Health 2019; 19:392. [PMID: 30971223 PMCID: PMC6458650 DOI: 10.1186/s12889-019-6659-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 03/14/2019] [Indexed: 11/24/2022] Open
Abstract
Background In 2014, a study in Munyenge revealed a high prevalence of urogenital schistosomiasis (UGS) among pregnant women. This study investigated he prevalence and risk factors of UGS in pregnancy following scale-up of piped water sources from 2014 to 2017. Secondly, we compared stream usage, stream contact behaviour, infection rate and intensity with the findings of 2014. Methods Consenting pregnant women reporting for antenatal care (ANC) in the different health facilities were enrolled consecutively between November 2016 and January 2018. Information on age, gravidity status, residence, marital status, educational level, occupation, household water source, frequency of contact with water and stream activities were obtained using a semi-structured questionnaire. Urine samples were examined for the presence of microhaematuria and S. haematobium ova using test strip and filtration/microscopy methods respectively. Data were analysed using univariate and multivariate regression analyses and relative risk reductions calculated. Results Of the 368 women enrolled, 22.3% (82) were diagnosed with UGS. Marital status (single) (aOR = 2.24, 95% CI: 1.04–4.79), primary - level of education (aOR = 2.0; 95% CI: 1.04–3.85) and domestic activity and bathing in the stream (aOR = 3.3; 95% CI: 1.83–6.01) increased risk of S. haematobium infection. Meanwhile, fewer visits (< 3 visits/week) to stream (aOR = 0.35, 95% CI = 0.17–0.74) reduced exposure to infection. Piped water usage was associated with reduced stream usage and eliminated the risk of infection among women who used safe water only. Compared with the findings of 2014, stream usage (RRR = 23 95% CI: 19–28), frequency (≥ 3 visits) (RRR = 69 95% CI: 59–77) and intensity of contact with water (RRR = 37 95% CI = 22–49) has reduced. Similarly, we observed a decrease in infection rate (RRR = 52, 95% CI = 40–62) and prevalence of heavy egg intensity (RRR = 71, 95% CI = 53–81). Conclusion Following increased piped water sources in Munyenge, S. haematobium infection has declined due to reduced stream contact. This has important implication in the control of UGS in pregnancy. Electronic supplementary material The online version of this article (10.1186/s12889-019-6659-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Godlove Bunda Wepnje
- Department of Zoology and Animal Physiology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon
| | - Judith Kuoh Anchang-Kimbi
- Department of Zoology and Animal Physiology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon.
| | - Vicky Daonyle Ndassi
- Department of Zoology and Animal Physiology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon
| | - Leopold Gustave Lehman
- Department of Animal Biology, Faculty of Science, University of Douala, P.O. Box 24157, Douala, Cameroon
| | - Helen Kuokuo Kimbi
- Department of Medical Laboratory Science, Faculty of Health Sciences, University of Bamenda, P.O. Box 39, Bambili, Cameroon
| |
Collapse
|
53
|
Sousa MS, van Dam GJ, Pinheiro MCC, de Dood CJ, Peralta JM, Peralta RHS, Daher EDF, Corstjens PLAM, Bezerra FSM. Performance of an Ultra-Sensitive Assay Targeting the Circulating Anodic Antigen (CAA) for Detection of Schistosoma mansoni Infection in a Low Endemic Area in Brazil. Front Immunol 2019; 10:682. [PMID: 31019510 PMCID: PMC6458306 DOI: 10.3389/fimmu.2019.00682] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/13/2019] [Indexed: 12/03/2022] Open
Abstract
Techniques with high sensitivity and specificity are required for an accurate diagnosis in low-transmission settings, where the conventional parasitological methods are insensitive. We determined the accuracy of an up-converting phosphor-lateral flow circulating anodic antigen (UCP-LF CAA) assay in urine and serum for Schistosoma mansoni diagnosis in low-prevalence settings in Ceará, Brazil, before and after praziquantel treatment. Clinical samples of a total of 258 individuals were investigated by UCP-LF CAA, point-of-care-circulating cathodic antigen (POC-CCA), soluble worm antigen preparation (SWAP)-ELISA and Kato-Katz (KK); a selection of 128 stools by real-time PCR technique. Three and 6-weeks after treatment, samples were collected and evaluated by detection Schistosoma circulating antigens (CAA and CCA). The UCP-LF CAA assays detected 80 positives (31%) with urine and 82 positives (31.8%) with serum. The urine POC-CCA and serum SWAP-ELISA assays detected 30 (11.6%) and 107 (40.7%) positives, respectively. The Kato-Katz technique revealed only 4 positive stool samples (1.6%). Among the 128 individuals with complete data records, 19 cases were identified by PCR (14.8%); Sensitivities and specificities of the UCP-LF CAA assays, determined versus a combined reference standard based on CCA/KK/PCR positivity, ranged from 60-68% to 68-77%, respectively. In addition only for comparative purposes, sensitivities of the different assays were determined vs. a comparative reference based on CAA/KK/PCR positivity, showing the highest sensitivity for the urine CAA assay (80%), followed by the serum CAA (70.9%), SWAP-ELISA (43.6%), PCR (34.5%), POC-CCA (29.1%), whilst triplicate Kato-Katz thick smears had a very low sensitivity (3.6%). CAA concentrations were higher in serum than in urine and were significantly correlated. There was a significant decrease in urine and serum CAA levels 3 and 6-weeks after treatment. The UCP-LF CAA assays revealed 33 and 28 S. mansoni-infected patients at the 3- and 6-week post-treatment follow-up, respectively. The UCP-LF CAA assays show high sensitivity for the diagnosis of S. mansoni in low-endemicity settings. It detects a considerably higher number of infections than microscopy, POC-CCA or PCR. Also it shows to be very useful for evaluating cure rates after treatment. Hence, the UCP-LF CAA assay is a robust and promising diagnostic approach in low-transmission settings.
Collapse
Affiliation(s)
- Mariana Silva Sousa
- Medical Sciences Post Graduate Program, Department of Internal Medicine, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
- Parasitology and Mollusks Biology Research Laboratory, Department of Clinical Analysis and Toxicology, Federal University of Ceará, Fortaleza, Brazil
| | - Govert J. van Dam
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Marta Cristhiany Cunha Pinheiro
- Parasitology and Mollusks Biology Research Laboratory, Department of Clinical Analysis and Toxicology, Federal University of Ceará, Fortaleza, Brazil
| | - Claudia J. de Dood
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Jose Mauro Peralta
- Department of Immunology, Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Elizabeth de Francesco Daher
- Medical Sciences Post Graduate Program, Department of Internal Medicine, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Paul L. A. M. Corstjens
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Fernando Schemelzer Moraes Bezerra
- Medical Sciences Post Graduate Program, Department of Internal Medicine, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
- Parasitology and Mollusks Biology Research Laboratory, Department of Clinical Analysis and Toxicology, Federal University of Ceará, Fortaleza, Brazil
- Pathology Post Graduate Program, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
54
|
Tweyongyere R, Nassanga BR, Muhwezi A, Odongo M, Lule SA, Nsubuga RN, Webb EL, Cose SC, Elliott AM. Effect of Schistosoma mansoni infection and its treatment on antibody responses to measles catch-up immunisation in pre-school children: A randomised trial. PLoS Negl Trop Dis 2019; 13:e0007157. [PMID: 30763405 PMCID: PMC6392333 DOI: 10.1371/journal.pntd.0007157] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 02/27/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Schistosoma infection is associated with immune modulation that can influence responses to non-schistosome antigens. Vaccine responses may be impaired in S. mansoni-infected individuals. We investigated effects of S. mansoni infection on responses to childhood measles catch-up immunisation and of praziquantel treatment on this outcome in a randomised trial. METHODOLOGY The Immune Modulation and Childhood Immunisation (IMoChI) study was based in Entebbe, Uganda. Children aged 3-5 years (193 S. mansoni-infected and 61 uninfected) were enrolled. Infected children were randomised in a 1:1:1 ratio to receive praziquantel 2 weeks before, at time of, or 1 week after, measles catch-up immunisation. Plasma anti-measles IgG was measured at enrolment, 1 week and 24 weeks after measles immunisation. Primary outcomes were IgG levels and percentage of participants with levels considered protective against measles. RESULTS Anti-measles IgG levels increased following immunisation, but at 1 week post-immunisation S. mansoni-infected, compared to uninfected, children had lower levels of anti-measles IgG (adjusted geometric mean ratio (aGMR) 0.4 [95% CI 0.2-0.7]) and the percentage with protective antibody levels was also lower (adjusted odds ratio 0.1 [0-0.9]). Among S. mansoni-infected children, anti-measles IgG one week post-immunisation was higher among those treated with praziquantel than among those who were not yet treated (treatment before immunisation, aGMR 2.3 [1.5-4.8]; treatment at immunisation aGMR 1.8 [1.1-3.5]). At 24 weeks post-immunisation, IgG levels did not differ between the trial groups, but tended to be lower among previously-infected children who were still S mansoni stool-positive than among those who became stool-negative. CONCLUSIONS AND SIGNIFICANCE Our findings suggest that S. mansoni infection among pre-school children is associated with a reduced antibody response to catch-up measles immunisation, and that praziquantel treatment improves the response. S. mansoni infection may contribute to impaired vaccine responses in endemic populations; effective schistosomiasis control may be beneficial for vaccine efficacy. This should be further explored. TRIAL REGISTRATION ISRCTN87107592.
Collapse
Affiliation(s)
- Robert Tweyongyere
- Department of Veterinary Pharmacy Clinical and Comparative Medicine, Makerere University, Kampala, Uganda
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Beatrice R. Nassanga
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Allan Muhwezi
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Matthew Odongo
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Swaib A. Lule
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Rebecca N. Nsubuga
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Emily L. Webb
- London School of Hygiene & Tropical Medicine, Keppel Street, London United Kingdom
| | - Stephen C. Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, Keppel Street, London United Kingdom
| | - Alison M. Elliott
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, Keppel Street, London United Kingdom
| |
Collapse
|
55
|
Abstract
Parasitic infections of the gut have major implications for child health, but many questions remain unanswered. Protozoal parasites, especially cryptosporidiosis and giardiasis, cause diarrhoea and contribute to impaired growth, neurocognitive development and mortality. Entamoeba histolytica causes dysentery and may have more subtle effects on child growth. Helminth infections are mostly asymptomatic, and untargeted mass deworming has not been shown to be beneficial. However, children with heavy infections certainly benefit from antihelminthic treatment. Hepatosplenic schistosomiasis is a neglected problem on a massive scale, which causes portal hypertension and lifelong morbidity in individuals who get infected in childhood. Neurocysticercosis causes epilepsy and is a significant consequence of taeniasis solium, another neglected disease which is entirely preventable. Parasitic infections of the gut contribute to child health problems on a large scale. Fresh approaches are needed to prevention and treatment.
Collapse
Affiliation(s)
- Kapula Chifunda
- a Tropical Gastroenterology and Nutrition group , University of Zambia School of Medicine , Lusaka , Zambia
| | - Paul Kelly
- a Tropical Gastroenterology and Nutrition group , University of Zambia School of Medicine , Lusaka , Zambia.,b Blizard Institute, Barts and The London School of Medicine , Queen Mary University of London , London , UK
| |
Collapse
|
56
|
Atalabi TE, Adubi TO. The epidemiology and chemotherapeutic approaches to the control of urinary schistosomiasis in school-age children (SAC): a systematic review. BMC Infect Dis 2019; 19:73. [PMID: 30658583 PMCID: PMC6339440 DOI: 10.1186/s12879-018-3647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 12/21/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Human schistosomiases are acute and chronic infectious diseases of poverty. Currently, epidemiological data of urinary schistosomiasis (US) in school-age children (SAC) and adults are often reported together making it difficult to ascertain the true status of the disease. Based on this premise, we set out to carry out this review. METHOD To achieve this aim, we carried out a computer-aided search of PubMed, Web of Science, Science Direct, African Journals OnLine (AJOL) and the database of World Health Organization. However, the information obtained from these sources was supplemented with additional literatures from Mendeley, Research Gate, and Google. RESULTS The search yielded 183 literatures of which 93 full text research, review and online articles were deemed fit for inclusion. Our key findings showed that: (1) of all World Health Organization (WHO) Regions, Africa is the most endemic zone for US, with Kenya and Senegal recording the highest prevalence and mean intensity respectively; (2) SAC within the range of 5-16 years contribute most significantly to the transmission cycle of US globally; (3) gender is a factor to watch out for, with male often recording the highest prevalence and intensity of infection; (4) contact with open, potentially infested water sources contribute significantly to transmission; (5) parental factors (occupation and education status) predispose SAC to US; (6) economic vis a vis ecological factors play a key role in infection transmission; and (7) in the last decade, a treatment coverage of 45% was never achieved globally for SAC or non-SAC treatment category for urinary schistosomiasis. CONCLUSION In view of the WHO strategic plan to eliminate schistosomiasis by 2020 and the findings from this review, it is obvious that this goal, in the face of realities, might not be achieved. It is imperative that annual control programmes be scaled up marginally, particularly in the African region of WHO. While US-based researches should be sponsored at the grass-root level to unveil hidden endemic foci, adequate facilities for Water, Sanitation, and Hygiene (WASH) should be put in place in all schools globally.
Collapse
Affiliation(s)
- Tolulope Ebenezer Atalabi
- Department of Biological Sciences, Federal University, Dutsin-Ma, Km 65, P.M.B. 5001, Dutsin-Ma, Katsina State Nigeria
| | - Taiwo Oluwakemi Adubi
- Department of Biological Sciences and Biotechnology, College of Pure and Applied Sciences, Caleb University, Imota, Lagos State Nigeria
| |
Collapse
|
57
|
Weber CJ, Hargan-Calvopiña J, Graef KM, Manner CK, Dent J. WIPO Re:Search-A Platform for Product-Centered Cross-Sector Partnerships for the Elimination of Schistosomiasis. Trop Med Infect Dis 2019; 4:E11. [PMID: 30634429 PMCID: PMC6473617 DOI: 10.3390/tropicalmed4010011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/26/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Schistosomiasis is an acute and chronic disease that affects over 200 million people worldwide, and with over 700 million people estimated to be at risk of contracting this disease, it is a pressing issue in global health. However, research and development (R&D) to develop new approaches to preventing, diagnosing, and treating schistosomiasis has been relatively limited. Praziquantel, a drug developed in the 1970s, is the only agent used in schistosomiasis mass drug administration (MDA) campaigns, indicating a critical need for a diversified therapeutic pipeline. Further, gaps in the vaccine and diagnostic pipelines demonstrate a need for early-stage innovation in all areas of schistosomiasis product R&D. As a platform for public-private partnerships (PPPs), the WIPO Re:Search consortium engages the private sector in early-stage R&D for neglected diseases by forging mutually beneficial collaborations and facilitating the sharing of intellectual property (IP) assets between the for-profit and academic/non-profit sectors. The Consortium connects people, resources, and ideas to fill gaps in neglected disease product development pipelines by leveraging the strengths of these two sectors. Using WIPO Re:Search as an example, this article highlights the opportunities for the PPP model to play a key role in the elimination of schistosomiasis.
Collapse
Affiliation(s)
- Callie J Weber
- BIO Ventures for Global Health, 2101 Fourth Avenue, Suite 1950, Seattle, WA 98121, USA.
| | | | - Katy M Graef
- BIO Ventures for Global Health, 2101 Fourth Avenue, Suite 1950, Seattle, WA 98121, USA.
| | - Cathyryne K Manner
- BIO Ventures for Global Health, 2101 Fourth Avenue, Suite 1950, Seattle, WA 98121, USA.
| | - Jennifer Dent
- BIO Ventures for Global Health, 2101 Fourth Avenue, Suite 1950, Seattle, WA 98121, USA.
| |
Collapse
|
58
|
Bakuza J. Demographic Factors Driving Schistosomiasis and Soil-Transmitted Helminthiases in Milola Ward, Lindi District, Tanzania: A Useful Guide for Launching Intervention Programmes. East Afr Health Res J 2018; 2:156-167. [PMID: 34308187 PMCID: PMC8279248 DOI: 10.24248/eahrj-d-18-00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/12/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Current information on the distribution of and risk factors for schistosomiasis and soil-transmitted helminthiases is scarce for most areas of southern Tanzania, including Milola Ward in Lindi District. This study was initiated to establish the status of these infections in Milola Ward and to assess how they vary with demographic factors. METHODS From September to October 2014, 2 sets of stool and urine samples were collected from residents of Milola Ward. The Kato-Katz technique was used to examine stool samples for faecal-borne parasites, and the filtration technique was used to examine urine for urinary schistosomes. A total of 195 individuals aged 5 to 90 years were enrolled in the study; 190 (97%) participants submitted adequate urine samples, of whom 107 (56%) were female and 83 (43%) were male. Of the 195 participants who took part in the initial sampling exercise, 158 (81%) provided adequate stool samples; 121 (77%) of these were adults, and the rest (n=37, 23%) were children. Only 53 urine and 26 faecal samples were obtained in the second round of sampling, and due to marked inconsistencies, these have been excluded from the analysis. Mean parasite abundance was analysed for its association with demographic factors, such as age and sex. RESULTS Three varieties of parasite were detected, namely, Schistosoma haematobium in 44 (23%) of 190 urine samples, hookworms in 12 (8%) of 158 stool samples, and Trichuris trichiura in 6 (4%) of 158 stool samples. The difference in S. haematobium prevalence between male and female participants (27 of 107 females, 25% vs 17 of 83 males, 20%) was not statistically significant (Kruskal-Wallis test, P=.47). Linear regression analysis of S. haematobium infection with age showed a significant association, with children having higher infection intensities than adults (P<.001). S. haematobium prevalence and intensity did not vary significantly between villages (intensity [Kruskal-Wallis test], P=.95; prevalence, P=.88). DISCUSSION These data confirm that in this setting, the mean age of peak helminthiasis prevalence decreases as transmission pressure increases, with non-school children below 18 years old being most at risk of acquiring parasitic infections. This was the first baseline survey of parasitic infections in Milola Ward, so the results will be crucial for guiding control efforts against parasitic diseases in the area.
Collapse
Affiliation(s)
- Jared Bakuza
- Department of Biological Sciences, Dar es Salaam University College of Education, Dar es Salaam, Tanzania
| |
Collapse
|
59
|
Buchter V, Hess J, Gasser G, Keiser J. Assessment of tegumental damage to Schistosoma mansoni and S. haematobium after in vitro exposure to ferrocenyl, ruthenocenyl and benzyl derivatives of oxamniquine using scanning electron microscopy. Parasit Vectors 2018; 11:580. [PMID: 30400935 PMCID: PMC6219169 DOI: 10.1186/s13071-018-3132-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/02/2018] [Indexed: 12/30/2022] Open
Abstract
Background Schistosomiasis is one of the most harmful parasitic diseases worldwide, praziquantel being the only drug in widespread use to treat it. We recently demonstrated that ferrocenyl, ruthenocenyl and benzyl derivatives of oxamniquine (Fc-OXA, Rc-OXA and Bn-OXA) are promising antischistosomal drug candidates. Methods In this study we assessed the tegumental damage of these three derivatives of oxamniquine using scanning electron microscopy. Adult Schistosoma mansoni and S. haematobium were exposed to a concentration of 100 μM of each drug and incubated for 4–120 h, according to their onset of action and activity. Results While on S. mansoni the fastest acting compound was Fc-OXA, which revealed high activity after 4 h of incubation, on S. haematobium, Rc-OXA revealed the quickest onset, being lethal on all males within 24 h. In both species studied, the three derivatives showed the same patterns of tegumental damage consisting of blebs, sloughing and tegument rupturing all over the body. Additionally, on S. mansoni distinct patterns of tegumental damage were observed for each of the compounds: tissue ruptures in the gynaecophoric canal for Fc-OXA, loss of spines for Rc-OXA and oral sucker rupture for Bn-OXA. Conclusions Our study confirmed that Fc-OXA, Rc-OXA and Bn-OXA are promising broad spectrum antischistosomal drug candidates. All derivatives show fast in vitro activity against S. mansoni and S. haematobium while validating the previous finding that the parent drug oxamniquine is less active in vitro under the conditions described. This work sets the base for further studies on the identification of a lead oxamniquine derivative, with the aim of identifying a molecule with the potential to become a new drug for human use.
Collapse
Affiliation(s)
- Valentin Buchter
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health institute, Socinstrasse 57, 4051, Basel, CH, Switzerland.,University of Basel, P.O. Box, 4003, Basel, CH, Switzerland
| | - Jeannine Hess
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, CH, Switzerland
| | - Gilles Gasser
- Chimie ParisTech, PSL University, Laboratory of Inorganic Chemical Biology, F-75005, Paris, France
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health institute, Socinstrasse 57, 4051, Basel, CH, Switzerland. .,University of Basel, P.O. Box, 4003, Basel, CH, Switzerland.
| |
Collapse
|
60
|
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: 1.9] [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
|
61
|
Bonate PL, Wang T, Passier P, Bagchus W, Burt H, Lüpfert C, Abla N, Kovac J, Keiser J. Extrapolation of praziquantel pharmacokinetics to a pediatric population: a cautionary tale. J Pharmacokinet Pharmacodyn 2018; 45:747-762. [PMID: 30218416 PMCID: PMC6182730 DOI: 10.1007/s10928-018-9601-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 07/13/2018] [Indexed: 11/29/2022]
Abstract
L-praziquantel (PZQ) pharmacokinetic data were analyzed from two relative bioavailability Phase 1 studies in adult, healthy subjects with two new oral dispersion tablet (ODT) formulations of L-PZQ administered under various combinations of co-administration with food, water, and/or crushing. Linear mixed effects models adequately characterized the noncompartmental estimates of the pharmacokinetic profiles in both studies. Dose, food, and formulation were found to significantly affect L-PZQ exposure in both studies. The model for AUC was then extrapolated to children 2–5 years old accounting for enzyme maturation and weight. The predicted exposures were compared to an external Phase 1 study conducted by the Swiss Tropical and Public Health Institute using a currently marketed formulation (Cesol 600 mg immediate-release tablets) and found to be substantially lower than observed. A root cause analysis was completed to identify the reason for failure of the models. Various scenarios were proposed and tested. Two possible reasons for the failure were identified. One reason was that the model did not account for the reduced hepatic clearance seen in patients compared to the healthy volunteer population used to build the model. The second possible reason was that PZQ absorption appears sensitive to meal composition and the model did not account for differences in meals between a standardized Phase 1 unit and clinical sites in Africa. Further studies are needed to confirm our hypotheses.
Collapse
Affiliation(s)
| | - Tianli Wang
- Astellas, 1 Astellas Way, Northbrook, IL, 60062, USA.,Alkermes, Waltham, MA, 02451, USA
| | - Paul Passier
- Astellas, 1 Astellas Way, Northbrook, IL, 60062, USA.,Galapagos BV, Zernikedreef 16, Leiden, The Netherlands
| | - Wilhelmina Bagchus
- Merck Serono SA, Merck Institute for Pharmacometrics (A Subsidiary of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | - Howard Burt
- Simcyp (a Certara company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Christian Lüpfert
- Merck KGaA, Translational Quantitative Pharmacology, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Nada Abla
- Merck Global Health Institute, Ares Trading S.A. (A Subsidiary of Merck KGaA, Darmstadt, Germany), 1262, Eysins, Switzerland
| | - Jana Kovac
- Swiss Tropical and Public Health Institute, Socinstr. 57, CH-4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Socinstr. 57, CH-4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
62
|
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
|
63
|
Macklin G, Stanton MC, Tchuem-Tchuenté LA, Stothard JR. A pilot study using wearable global positioning system data loggers to compare water contact levels: Schistosoma haematobium infection in pre-school-age children (PSAC) and their mothers at Barombi Kotto, Cameroon. Trans R Soc Trop Med Hyg 2018; 112:361-365. [PMID: 29992295 DOI: 10.1093/trstmh/try059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/27/2018] [Indexed: 12/22/2022] Open
Abstract
Background Barombi Kotto, Cameroon serves as a reference location for assessing intervention strategies against Schistosoma haematobium. Methods As part of a pilot study, the whole community was treated with praziquantel, inclusive of pre-school-age children (PSAC) and their mothers. One year later, egg-patent infections were reassessed and water contact patterns of 12 pairs of PSAC and their mothers were measured with global positioning system (GPS) data loggers. Results A substantial reduction in general infection prevalence, from 44.8% to 12.2%, was observed but certain PSAC and mothers continued to have egg-patent infections. Analysis of GPS data demonstrated similar water contact levels between the child and mother groups, although certain individuals were numerical outliers. Conclusions This study shows the potential of GPS data loggers to clarify the at-risk status of PSAC and mothers.
Collapse
Affiliation(s)
- Grace Macklin
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | | | - J Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
64
|
Ondigo BN, Muok EMO, Oguso JK, Njenga SM, Kanyi HM, Ndombi EM, Priest JW, Kittur N, Secor WE, Karanja DMS, Colley DG. Impact of Mothers' Schistosomiasis Status During Gestation on Children's IgG Antibody Responses to Routine Vaccines 2 Years Later and Anti-Schistosome and Anti-Malarial Responses by Neonates in Western Kenya. Front Immunol 2018; 9:1402. [PMID: 29967622 PMCID: PMC6015899 DOI: 10.3389/fimmu.2018.01402] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/06/2018] [Indexed: 02/04/2023] Open
Abstract
The potential consequences of parasitic infections on a person’s immune responsiveness to unrelated antigens are often conjectured upon in relationship to allergic responses and autoimmune diseases. These considerations sometimes extend to whether parasitic infection of pregnant women can influence the outcomes of responses by their offspring to the immunizations administered during national Expanded Programs of Immunization. To provide additional data to these discussions, we have enrolled 99 close-to-term pregnant women in western Kenya and determined their Schistosoma mansoni and Plasmodium falciparum infection status. At 2 years of age, when the initial immunization schedule was complete, we determined their children’s IgG antibody levels to tetanus toxoid, diphtheria toxoid, and measles nucleoprotein (N-protein) antigens using a multiplex assay. We also monitored antibody responses during the children’s first 2 years of life to P. falciparum MSP119 (PfMSP119), S. mansoni Soluble Egg Antigen (SEA), Ascaris suum hemoglobin (AsHb), and Strongyloides stercoralis (SsNIE). Mothers’ infections with either P. falciparum or S. mansoni had no impact on the level of antibody responses of their offspring or the proportion of offspring that developed protective levels of antibodies to either tetanus or diphtheria antigens at 2 years of age. However, children born of S. mansoni-positive mothers and immunized for measles at 9 months of age had significantly lower levels of anti-measles N-protein antibodies when they were 2 years old (p = 0.007) and a lower proportion of these children (62.5 vs. 90.2%, OR = 0.18, 95% CI = 0.04–0.68, p = 0.011) were considered positive for measles N-protein antibodies. Decreased levels of measles antibodies may render these children more susceptible to measles infection than children whose mothers did not have schistosomiasis. None of the children demonstrated responses to AsHb or SsNIE during the study period. Anti-SEA and anti-PfMSP119 responses suggested that 6 and 70% of the children acquired schistosomes and falciparum malaria, respectively, during the first 2 years of life.
Collapse
Affiliation(s)
- Bartholomew N Ondigo
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya.,Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya
| | - Erick M O Muok
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - John K Oguso
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Sammy M Njenga
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Henry M Kanyi
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Eric M Ndombi
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya.,Department of Pathology, Kenyatta University, Nairobi, Kenya
| | - Jeffrey W Priest
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Nupur Kittur
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - William Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Diana M S Karanja
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Daniel G Colley
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States.,Department of Microbiology, University of Georgia, Athens, GA, United States
| |
Collapse
|
65
|
A single dose of the antineoplastics hydroxyurea or cisplatin has praziquantel-like effects on Schistosoma mansoni worms and host mouse liver. Biomed Pharmacother 2018; 99:570-575. [PMID: 29902867 DOI: 10.1016/j.biopha.2018.01.098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 11/21/2022] Open
Abstract
Increasing resistance to praziquantel, the only available antischistosomal drug, is always developed by schistosomes. The recent description of stem cell-like neoblasts in schistosomes led to the idea of applying antineoplastic drugs as antischistosomal agents that may inhibit stem cell divisions and retard worm regeneration. Here, we explored the in vitro and in vivo effect of some antineoplastic drugs on S. mansoni worm and the host mouse liver. S. mansoni worms' viability was tested after exposure to either praziquantel or one of the antitumor drugs (hydroxyurea, cisplatin, methotrexate, and colchicine) in vitro for 24 and 48 h. The effect of two of them (hydroxyurea and cisplatin) on worm burden, tegument ultrastructure, and host liver structure and function was tested in vivo in S. mansoni-infected mouse model. All drugs affected variably the worm burden in vitro. Hydroxyurea and cisplatin, like praziquantel, damaged the worm tegument, reduced worm burden, and viable schistosome eggs, decreased anti-schistosome IgG, reduced egg-induced hepatic granuloma size and cellularity, restored liver organization and improved liver function as represented by serum alanine aminotransferase and albumin. In conclusions, a single dose of hydroxyurea and cisplatin had anti-schistosome effects and may offer a safe promising alternative to control of schistosomiasis. A direct link between antitumor drugs and inhibition of schistosome neoblasts remains to be proven.
Collapse
|
66
|
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: 4.3] [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
|
67
|
Coulibaly JT, Panic G, Yapi RB, Kovač J, Barda B, N'Gbesso YK, Hattendorf J, Keiser J. Efficacy and safety of ascending doses of praziquantel against Schistosoma haematobium infection in preschool-aged and school-aged children: a single-blind randomised controlled trial. BMC Med 2018; 16:81. [PMID: 29855373 PMCID: PMC5984412 DOI: 10.1186/s12916-018-1066-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/01/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Despite decades of experience with praziquantel treatment in school-aged children (SAC) and adults, we still face considerable knowledge gaps relevant to the successful treatment of preschool-aged children (PSAC). This study aimed to assess the efficacy and safety of escalating praziquantel dosages in PSAC infected with Schistosoma haematobium. METHODS We conducted a randomised, dose-finding trial in PSAC (2-5 years) and as comparator a cohort of SAC (6-15 years) infected with S. haematobium in Côte d'Ivoire. A total of 186 PSAC and 195 SAC were randomly assigned to 20, 40 or 60 mg/kg praziquantel or placebo. The nature of the dose-response relationship in terms of cure rate (CR) was the primary objective. Egg reduction rate (ERR) and tolerability were secondary outcomes. CRs and ERRs were assessed using triplicate urine filtration over 3 consecutive days. Available-case analysis was performed including all participants with primary endpoint data. RESULTS A total of 170 PSAC and 174 SAC received treatment. Almost 90% of PSAC and three quarters of SAC were lightly infected with S. haematobium. Follow-up data were available for 157 PSAC and 166 SAC. In PSAC, CRs of praziquantel were 85.7% (30/35), 78.0% (32/41) and 68.3% (28/41) at 20, 40 and 60 mg/kg and 47.5% (19/40) for placebo. In SAC, CRs were 10.8% for placebo (4/37), 55.6% for 20 mg/kg (25/45), 68.3% for 40 mg/kg (28/41) and 60.5% for 60 mg/kg (26/43). ERRs based on geometric means ranged between 96.5% (60 mg/kg) and 98.3% (20 mg/kg) in PSAC and between 97.6% (20 mg/kg and 60 mg/kg) and 98.6% (40 mg/kg) in SAC. Adverse events were mild and transient. CONCLUSIONS Praziquantel revealed dose-independent efficacy against light infections of S. haematobium. Over the dose range tested, praziquantel displayed a ceiling effect with the highest response for 20 mg/kg in PSAC. In SAC maximum efficacy was obtained with 40 mg/kg praziquantel. Further investigations are required in children with moderate to heavy infections. TRIAL REGISTRATION This trial is registered with International Standard Randomised Controlled Trial Number ISRCTN15280205 .
Collapse
Affiliation(s)
- Jean T Coulibaly
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland.,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, P.O. Box, CH-4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Richard B Yapi
- Centre Suisse de Recherches Scientifiques, Abidjan, Côte d'Ivoire
| | - Jana Kovač
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Beatrice Barda
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Yves K N'Gbesso
- Departement d'Agboville, Centre de Santé Urbain d'Azaguié, Azaguié, Côte d'Ivoire
| | - Jan Hattendorf
- University of Basel, Basel, Switzerland.,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, P.O. Box, CH-4002, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
68
|
Pasche V, Laleu B, Keiser J. Screening a repurposing library, the Medicines for Malaria Venture Stasis Box, against Schistosoma mansoni. Parasit Vectors 2018; 11:298. [PMID: 29764454 PMCID: PMC5952519 DOI: 10.1186/s13071-018-2855-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022] Open
Abstract
Background The development of new treatments against schistosomiasis is imperative but lacks commercial interest. Drug repurposing represents a suitable strategy to identify potential treatments, which have already unblocked several essential steps along the drug development path, hence reducing costs and timelines. Promoting this approach, the Medicines for Malaria Venture (MMV) recently distributed a drug repurposing library of 400 advanced lead candidates (Stasis Box). Methods All 400 compounds were initially tested in vitro against the larval stage of Schistosoma mansoni at 10 μM. Hits progressed to screening on adult worms and were further characterised for IC50, cytotoxicity and selectivity. Ten lead compounds were tested in mice harbouring a chronic S. mansoni infection. Results Eleven of the 37 compounds active on the larval stage were also highly active on adult worms in vitro (IC50 = 2.0–7.5 μM). IC50 values on adult S. mansoni decreased substantially in the presence of albumin (7.5–123.5 μM). Toxicity to L6 and MRC cells was moderate. A moderate worm burden reduction of 51.6% was observed for MMV690534, while the other 9 compounds showed low activity. None of the in vivo results were statistically significant (P > 0.05). Conclusions Phenotypic screening of advanced lead compounds is a simple and resource-low method to identify novel anthelminthics. None of the promising hits of the Stasis Box identified in vitro against S. mansoni yielded acceptable worm burden reductions in vivo, which might be due to the high plasma protein binding. Since the in vitro hits interfere with different drug targets, they might provide a starting point for target based screening and structure-activity relationship studies.
Collapse
Affiliation(s)
- Valérian Pasche
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland.,University of Basel, P.O. Box, CH-4003, Basel, Switzerland
| | - Benoît Laleu
- Medicines for Malaria Venture (MMV), PO Box 1826, 20, Route de Pré-Bois, 1215, Geneva 15, Switzerland
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, CH-4002, Basel, Switzerland. .,University of Basel, P.O. Box, CH-4003, Basel, Switzerland.
| |
Collapse
|
69
|
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: 7.1] [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
|
70
|
The COUNTDOWN Study Protocol for Expansion of Mass Drug Administration Strategies against Schistosomiasis and Soil-Transmitted Helminthiasis in Ghana. Trop Med Infect Dis 2018; 3:tropicalmed3010010. [PMID: 30720777 PMCID: PMC6136637 DOI: 10.3390/tropicalmed3010010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Current international policy for schistosomiasis and soil-transmitted helminthiasis (STH) control emphasises mass administration of deworming drugs in school-based programmes. However, this approach is insufficient to control the transmission of these diseases, and their burden in non-school cohorts is recognised, albeit under-researched. This research will investigate the feasibility and acceptability of expanding access to praziquantel (PZQ) against schistosomiasis, and albendazole (ALB) against STH, to communities in selected transmission settings in Ghana. (2) Methods: A three-site longitudinal study will be implemented to investigate the effectiveness of expanding treatment strategies for PZQ and ALB to community members. In the context of community mass drug administration (to preschool children, school non-attending children, and adults, including pregnant women), the intervention will be assessed in a random sample of community members, at baseline with follow-up at 6, 12, and 18 months. In each community, 658 participants will be enrolled, and 314 followed up at each time point. The primary outcome measure is the prevalence of infection of Schistosoma haematobium and/or S. mansoni at study endpoint, as assessed by longitudinal surveys. Secondary outcomes are to quantify the infection of schistosomiasis and STH infections in non-treated cohorts, reductions in prevalence of STH, and intensity of schistosomiasis and STH, and treatment coverage. Nested within this study will be qualitative, cost-benefit, and cost-effectiveness evaluations that will explore accessibility, feasibility, and economic impact of expanded treatment from different complementary perspectives. (3) Discussion: Using a multidisciplinary approach, this study will generate evidence for improved availability, acceptability, affordability, and accessibility to deworming drugs against schistosomiasis and STH to individuals and communities in Ghana. This is likely to have considerable research, programmatic, and political value to contribute evidence for national programme policy development within Ghana, and, more broadly, World Health Organization policy development.
Collapse
|
71
|
Sacolo H, Chimbari M, Kalinda C. Knowledge, attitudes and practices on Schistosomiasis in sub-Saharan Africa: a systematic review. BMC Infect Dis 2018; 18:46. [PMID: 29347919 PMCID: PMC5773048 DOI: 10.1186/s12879-017-2923-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/14/2017] [Indexed: 12/22/2022] Open
Abstract
Background Schistosomiasis remains a global health problem with an estimated 250 million people in 78 countries infected, of whom 85% live in Sub-Saharan Africa. Preventive chemotherapy remains the key public health strategy to combat schistosomiasis worldwide. Recently the WHO emphasized on the use of integrative approaches in the control and elimination of schistosomiasis. However, a detailed understanding of sociocultural factors that may influence the uptake of the intended health activities and services is vital. Thus, our study sought to understand the knowledge, attitudes, perceptions, beliefs and practices about schistosomiasis in various communities in Sub-Saharan Africa. Methods A systematic search of literature for the period 2006–2016 was done on Medline, PubMed, CINAHL, Psych info and Google Scholar using the following key words “Schistosomiasis, S. mansoni, S. haematobium, knowledge, attitudes, perceptions, beliefs and practices in Sub-Saharan Africa” in combination with Bolean operators (OR, AND). In this context, we reviewed studies conducted among school children, community members and caregivers of preschool children. Thematic analysis was utilised for the overall synthesis of the selected studies. This was done after reading the articles in depth. Themes were identified and examined for similarities, differences and contradictions. Results Gaps in schistosomiasis related knowledge and sociocultural barriers towards the uptake of preventive and treatment services among communities in Sub-Saharan Africa were identified. In addition to limited knowledge and negative attitudes, risky water related practices among community members, school children and caregivers of preschool children were identified as key factors promoting transmission of the disease. Conclusion The study concluded that a comprehensive health education programme using contextual and standardised training tools may improve peoples’ knowledge, attitudes and practices in relation to schistosomiasis prevention and control. Findings also highlight the significance of including caregivers in the planning and implementation schistosomiasis control programs targeting pre-school children. Electronic supplementary material The online version of this article (10.1186/s12879-017-2923-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hlengiwe Sacolo
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Howard Campus, Durban, South Africa.
| | - Moses Chimbari
- College of Health Sciences, University of KwaZulu-Natal, Howard Campus, Durban, South Africa
| | - Chester Kalinda
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Howard Campus, Durban, South Africa
| |
Collapse
|
72
|
Cognitive deficits and educational loss in children with schistosome infection-A systematic review and meta-analysis. PLoS Negl Trop Dis 2018; 12:e0005524. [PMID: 29329293 PMCID: PMC5766129 DOI: 10.1371/journal.pntd.0005524] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/23/2017] [Indexed: 12/26/2022] Open
Abstract
Background By means of meta-analysis of information from all relevant epidemiologic studies, we examined the hypothesis that Schistosoma infection in school-aged children (SAC) is associated with educational loss and cognitive deficits. Methodology/Principal findings This review was prospectively registered in the PROSPERO database (CRD42016040052). Medline, Biosis, and Web of Science were searched for studies published before August 2016 that evaluated associations between Schistosoma infection and cognitive or educational outcomes. Cognitive function was defined in four domains—learning, memory, reaction time, and innate intelligence. Educational outcome measures were defined as attendance and scholastic achievement. Risk of bias (ROB) was evaluated using the Newcastle-Ottawa quality assessment scale. Standardized mean differences (SMD) and 95% confidence intervals (CI) were calculated to compare cognitive and educational measures for Schistosoma infected /not dewormed vs. uninfected/dewormed children. Sensitivity analyses by study design, ROB, and sequential exclusion of individual studies were implemented. Thirty studies from 14 countries, including 38,992 SAC between 5–19 years old, were identified. Compared to uninfected children and children dewormed with praziquantel, the presence of Schistosoma infection and/or non-dewormed status was associated with deficits in school attendance (SMD = -0.36, 95%CI: -0.60, -0.12), scholastic achievement (SMD = -0.58, 95%CI: -0.96, -0.20), learning (SMD = -0.39, 95%CI: -0.70, -0.09) and memory (SMD = -0.28, 95%CI: -0.52, -0.04) tests. By contrast, Schistosoma-infected/non-dewormed and uninfected/dewormed children were similar with respect to performance in tests of reaction time (SMD = -0.06, 95%CI: -0.42, 0.30) and intelligence (SMD = -0.25, 95%CI: -0.57, 0.06). Schistosoma infection-associated deficits in educational measures were robust among observational studies, but not among interventional studies. The significance of infection-associated deficits in scholastic achievement was sensitive to ROB. Schistosoma infection-related deficits in learning and memory tests were invariant by ROB and study design. Conclusion/Significance Schistosoma infection/non-treatment was significantly associated with educational, learning, and memory deficits in SAC. Early treatment of children in Schistosoma-endemic regions could potentially mitigate these deficits. Trial registration ClinicalTrials.gov CRD42016040052 Empirical evidence for cognitive or educational benefits of anti-Schistosoma treatment is currently uncertain, despite the recommended practice of wide-scale deworming with praziquantel. We addressed this knowledge gap by synthesizing information from 30 relevant epidemiologic studies reporting on 38,992 children between 5–19 years old from 14 countries. In those studies, Schistosoma infection or non-dewormed status was associated with educational loss and cognitive deficits. Specifically, there were small to moderate deficits in both school attendance and scholastic achievement. Similarly, Schistosoma infection or non-dewormed status was associated with deficits in learning and memory domains of psychometrically tested cognitive function. However, there was no evidence of Schistosoma infection- or non-deworming-associated deficits on tests of innate intelligence or reaction-time. Overall, compared to Schistosoma-uninfected or to dewormed children, the presence of Schistosoma infection or non-dewormed status was associated with educational, learning, and memory deficits in school-aged children. The combined evidence suggests that early treatment of children in Schistosoma-endemic regions could mitigate these deficits.
Collapse
|
73
|
Mass drug administration and the global control of schistosomiasis: successes, limitations and clinical outcomes. Curr Opin Infect Dis 2018; 29:595-608. [PMID: 27584590 DOI: 10.1097/qco.0000000000000312] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Preventive chemotherapy is advocated for the global control and elimination of schistosomiasis. Despite the well known short-term benefits of treating patients for schistosomiasis, the impact of mass drug administration (MDA) campaigns to control the disease in the long term remains unresolved. RECENT FINDINGS Many studies have advocated the success of MDA programs in order to attract donor funds for elimination efforts but such successes are often short-lived given the drug does not alter the life cycle of the organism or prevent reinfection. Within a matter of months to years after halting treatment, the prevalence, intensity of infection and morbidity of disease return to baseline levels. Other mitigating factors contribute to the failings of MDA campaigns namely: poverty, poor drug coverage, poor drug compliance, and, in the case of Asiatic schistosomiasis, zoonotic transmission. Genetic and innate and acquired immunologic mechanisms complicate the epidemiologic picture of schistosomiasis globally, and may contribute indirectly to MDA shortcomings. The possibility of drug resistance is an ever present concern because of the sole reliance on one drug, praziquantel. SUMMARY Preventive chemotherapy is advocated for the global control and elimination of schistosomiasis. The short-term benefits of MDA campaigns are well documented but the long-term benefits are questionable.
Collapse
|
74
|
Nalugwa A, Nuwaha F, Tukahebwa EM, Olsen A. Schistosoma mansoni-Associated Morbidity among Preschool-Aged Children along the Shores of Lake Victoria in Uganda. Trop Med Infect Dis 2017; 2:E58. [PMID: 30270915 PMCID: PMC6082064 DOI: 10.3390/tropicalmed2040058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/02/2022] Open
Abstract
Schistosoma mansoni causes morbidity in human beings, with the highest prevalence in rural sub-Saharan Africa. Prolonged S. mansoni infection with egg deposition in intestinal blood vessels leads to liver and spleen enlargement, and thus chronic morbidity. The objective of this study was to assess whether preschool-aged children develop severe S. mansoni-related morbidity. Parasitological, clinical, and ultrasonographic examinations were carried out in 916 preschool-aged children in five schistosomiasis-endemic districts (Bugiri, Buikwe, Jinja, Mayuge, and Namayingo) along the Lake Victoria shoreline in east-central Uganda. Anaemia and anthropometry measurements were also taken. Using the Kato-Katz technique on one stool sample collected on three consecutive days, 74.9% (686/916) were found infected with S. mansoni; the majority were lightly infected (57.9%), while 22.7% and 19.4% were moderately and heavily infected, respectively. The overall geometric mean intensity (GMI) of infected children was 294.2 eggs per gram faeces. Mayuge and Jinja districts had the highest (51.2%) and lowest (2.2%) number of infected children, respectively. Hookworm infection was found in 7.8% (71/916) of the children. Both liver and spleen were significantly more enlarged in the infected children than in the uninfected children (p < 0.0005), as measured by ultrasonography. Physical palpation of the spleen was more often detected in the uninfected children. A significantly (p < 0.0005) higher proportion of S. mansoni-positive children were anaemic (359/686; 52.3%) compared to the children who had no eggs in their stool samples (81/230; 35.2%). Schistosoma mansoni infection did not have any severe effect on the nutrition status of preschool-aged children. Neither infected nor uninfected children were found to be underweight or stunted. Liver fibrosis with distinct Symmer's 'pipe stems' was found in a few heavily-infected children (0.3%). In a linear multivariable regression analysis, age of the child, anaemia, liver fibrosis, and size of the left liver lobe were associated with S. mansoni intensity of infection (adjusted R² = 0.11; p < 0.0005). Our results demonstrate that S. mansoni-related morbidity does develop in children less than six years of age, and that older children (37⁻60 months) are at higher risk (regression coefficient 0.33; p <0.0005) compared to younger ones (12⁻36 months). We recommend that preschool-aged children be included in the target population for schistosomiasis mass treatment so as to prevent the childhood chronic form of schistosomiasis.
Collapse
Affiliation(s)
- Allen Nalugwa
- Child Health and Development Centre, College of Health Sciences, Makerere University, P.O. Box 6717 Kampala, Uganda.
| | - Fred Nuwaha
- Disease Control and Prevention, School of Public Health, Makerere University, P.O. Box 7062 Kampala, Uganda.
| | - Edridah Muheki Tukahebwa
- Neglected Tropical Diseases, Vector Control Division, Ministry of Health, P.O. Box 1661 Kampala, Uganda.
| | - Annette Olsen
- Parasitology and Aquatic Pathobiology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg C, Denmark.
| |
Collapse
|
75
|
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: 1.9] [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
|
76
|
Won KY, Kanyi HM, Mwende FM, Wiegand RE, Goodhew EB, Priest JW, Lee YM, Njenga SM, Secor WE, Lammie PJ, Odiere MR. Multiplex Serologic Assessment of Schistosomiasis in Western Kenya: Antibody Responses in Preschool Aged Children as a Measure of Reduced Transmission. Am J Trop Med Hyg 2017; 96:1460-1467. [PMID: 28719280 PMCID: PMC5462587 DOI: 10.4269/ajtmh.16-0665] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Currently, impact of schistosomiasis control programs in Schistosoma mansoni–endemic areas is monitored primarily by assessment of parasitologic indicators only. Our study was conducted to evaluate the use of antibody responses as a way to measure the impact of schistosomiasis control programs. A total of 3,612 serum samples collected at three time points from children 1–5 years of age were tested for antibody responses to two schistosome antigens (soluble egg antigen [SEA] and Sm25) by multiplex bead assay. The overall prevalence of antibody responses to SEA was high at baseline (50.0%). After one round of mass drug administration (MDA), there was minimal change in odds of SEA positivity (odds ratio [OR] = 1.02, confidence interval [CI] = 0.79–1.32, P = 0.89). However, after two rounds of treatment, there was a slight decrease in odds of SEA positivity (OR = 0.80, CI = 0.63–1.02, P = 0.08). In contrast to the SEA results, prevalence of antibody responses to Sm25 was lowest at baseline (14.1%) and higher in years 2 (19.8%) and 3 (18.4%). After one round of MDA, odds of Sm25 positivity increased significantly (OR = 1.51, CI = 1.14–2.02, P = 0.005) and remained significantly higher than baseline after two rounds of MDA (OR = 1.37, CI = 1.07–1.76, P = 0.01). There was a significant decrease in the proportion of 1-year-olds with positive SEA responses from 33.1% in year 1 to 13.2% in year 3 and a corresponding decrease in the odds (OR = 3.25, CI = 1.75–6.08, P < 0.001). These results provide preliminary evidence that schistosomiasis program impact can be monitored using serologic responses.
Collapse
Affiliation(s)
- Kimberly Y Won
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Henry M Kanyi
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | - Faith M Mwende
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | - Ryan E Wiegand
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - E Brook Goodhew
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jeffrey W Priest
- Division of Foodborne, Waterborne and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yeuk-Mui Lee
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sammy M Njenga
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research Institute, Nairobi, Kenya
| | - W Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Patrick J Lammie
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Maurice R Odiere
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| |
Collapse
|
77
|
Reinhard-Rupp J, Klohe K. Developing a comprehensive response for treatment of children under 6 years of age with schistosomiasis: research and development of a pediatric formulation of praziquantel. Infect Dis Poverty 2017; 6:122. [PMID: 28768535 PMCID: PMC5541653 DOI: 10.1186/s40249-017-0336-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/14/2017] [Indexed: 11/10/2022] Open
Abstract
Schistosomiasis is a parasitic disease caused by blood flukes. The disease is caused by an inflammatory reaction to parasite eggs retained in the liver, bladder and reproductive organs. According to 2017 World Health Organization (WHO) estimates 220 million people are potentially infected, from which probably 10% are children under 6 years of age. The regular treatment approach of a single, oral dose of 40 mg/kg body weight with praziquantel however, is difficult for children under the age of 6, leaving them without a treatment option. In order to address this important gap in treatment target populations, an international public-private partnership that works on a not-for-profit basis in the field of drug research and development for schistosomiasis was established in 2012. This is called the Pediatric Praziquantel Consortium. Its mission was and continues to be to develop, register and provide access to a suitable pediatric praziquantel formulation for treating schistosomiasis in preschool-age children (3-6 months up to 6 years). The Target Product Profile for the pediatric formulation of praziquantel that would be suitable to treat children as young as 3-6 months was then defined by a group of experts, including members from the Pediatric Praziquantel Consortium partner organizations as well as experts from WHO (as observer) and schistosomiasis endemic countries. The development of the drug is ongoing and the Pediatric Praziquantel Consortium aims to submit the regulatory dossier for marketing approval in endemic countries and WHO prequalification in 2018/19 with approval and product launch for schistosomiasis pediatric case management in key endemic countries in 2019. Ultimately, the goal is for the product to be considered for a large-scale mass distribution program by 2022.
Collapse
Affiliation(s)
| | - Katharina Klohe
- Global Schistosomiasis Alliance, Westenriederstrasse 10, 80331, Munich, Germany.
| |
Collapse
|
78
|
Kovač J, Vargas M, Keiser J. In vitro and in vivo activity of R- and S- praziquantel enantiomers and the main human metabolite trans-4-hydroxy-praziquantel against Schistosoma haematobium. Parasit Vectors 2017; 10:365. [PMID: 28764732 PMCID: PMC5540299 DOI: 10.1186/s13071-017-2293-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/13/2017] [Indexed: 11/17/2022] Open
Abstract
Background Praziquantel (PZQ) is the mainstay of schistosomiasis control and has been successfully used for decades. However, its mechanism of action is not fully understood. While the majority of studies have been conducted on Schistosoma mansoni, it is not known which enantiomer, R- or S-praziquantel (R-/S-PZQ), is responsible for the activity on Schistosoma haematobium. Methods In vitro and in vivo studies were conducted to evaluate the activity of R- and S-PZQ, racemic PZQ and the main human metabolite, namely trans-4-OH-PZQ, on S. haematobium. IC50 values on adult S. haematobium were determined in vitro. Dose-response relationship studies were performed in golden Syrian hamsters, harbouring a chronic S. haematobium infection. Results R-PZQ displayed the highest activity against adult worms in vitro, revealing an IC50 of 0.007 μg/ml at 4 h and 0.01 μg/ml at 72 h. In contrast, S-PZQ was 501× less active (eudysmic ratio at 4 h), with an IC50 of 3.51 and 3.40 μg/ml (4 and 72 h, respectively). Racemic PZQ and trans-4-OH-PZQ resulted in an IC50 of 0.03 μg/ml and 1.47 μg/ml both at 4 and 72 h, respectively. In vivo, R-PZQ was the most potent drug with worm burden reductions (WBRs) of 98.5, 75.6 and 73.3% at 125.0, 62.5 and 31.0 mg/kg, respectively. A single oral dose of 250.0 mg/kg PZQ resulted in a WBR of 99.3%. S-PZQ was highly active in vivo at 250.0 and 500.0 mg/kg with WBRs of 83.0 and 94.1%, respectively. The lowest tested dose of S-PZQ, 125.0 mg/kg, showed moderate activity (WBR of 46.7%). The calculated ED50 for R- and S-PZQ were 24.7 and 127.6 mg/kg, respectively, with a corresponding eudysmic ratio of 5.17. Conclusion Our data support the theory of R-PZQ driving the antischistosomal activity. Interestingly, also S-PZQ proved to possess a significant activity towards S. haematobium, particularly in vivo.
Collapse
Affiliation(s)
- Jana Kovač
- 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
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
79
|
Spencer SA, Penney JMSJ, Russell HJ, Howe AP, Linder C, Rakotomampianina ALD, Nandimbiniaina AM, Squire SB, Stothard JR, Bustinduy AL, Rahetilahy AM. High burden of Schistosoma mansoni infection in school-aged children in Marolambo District, Madagascar. Parasit Vectors 2017. [PMID: 28646926 PMCID: PMC5483300 DOI: 10.1186/s13071-017-2249-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background A school-based survey was undertaken to assess prevalence and infection intensity of schistosomiasis in school-aged children in the Marolambo District of Madagascar. Methods School-aged children from six purposively selected schools were tested for Schistosoma haematobium by urine filtration and Schistosoma mansoni using circulating cathodic antigen (CCA) and Kato-Katz stool analysis. The investigators did not address soil-transmitted helminths (STH) in this study. Results Of 399 school-aged children screened, 93.7% were infected with S. mansoni based on CCA analysis. Kato-Katz analysis of stool revealed S. mansoni infection in 73.6% (215/ 292). Heavy infections (> 400 eggs per gram) were common (32.1%; 69/ 215), with a mean of 482 eggs per gram of stool. Moderate infection intensities were detected in 31.2% (67/ 215) and light infection intensities in 36.7% (79/ 215) of infected participants. No infection with S. haematobium was detected by urine filtration. Conclusions Intestinal schistosomiasis appears a considerable public health issue in this remote area of Madagascar where there is a pressing need for mass drug administration.
Collapse
Affiliation(s)
- Stephen A Spencer
- Royal United Hospitals Bath NHS Foundation Trust, Bath, UK. .,The University of Manchester Medical School, Manchester, UK.
| | | | | | - Anthony P Howe
- The University of Manchester Medical School, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Moyo VB, Changadeya W, Chiotha S, Sikawa D. Urinary schistosomiasis among preschool children in Malengachanzi, Nkhotakota District, Malawi: Prevalence and risk factors. Malawi Med J 2017; 28:10-4. [PMID: 27217911 DOI: 10.4314/mmj.v28i1.3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM This study was designed to determine the prevalence of and risk factors for schistosomiasis among a group of preschool children in Malawi. Schistosomiasis burden among preschoolers in Malawi is not well documented in the literature. METHODS This study used field research (in the form of a snail survey), laboratory work (urinalysis and microscopy for parasite identification), and questionnaire-guided interviews to determine the prevalence of and risk factors for urinary schistosomiasis among children, aged between 6 and 60 months, in Malengachanzi, Nkhotakota District, Malawi. RESULTS Urinary schistosomiasis prevalence among preschool children was 13%. Of the factors evaluated, only age (P = 0.027) was statistically significantly associated with urinary schistosomiasis risk. Four-year-old preschool children were five times more likely to contract urinary schistosomiasis than twoyear-old children (odds ratio [OR] = 5.255; 95% confidence interval [CI] = 1.014-27.237; P = 0.048). Increased contact with infested water among older children likely explains much of their increased risk. Infestation was evidenced by the presence of infected Bulinus globosus snails in the water contact points surveyed. Multiple regression analysis showed that visiting water contact sites daily (OR = 0.898, 95% CI = 0.185-4.350, P = 0.894), bathing in these sites (OR = 9.462, 95% CI = 0.036-0.00, P = 0.430) and lack of knowledge, among caregivers, regarding the causes of urinary schistosomiasis (OR = 0.235, 95% CI = 0.005-1.102, P = 0.066) posed statistically insignificant risk increases for preschoolers contracting urinary schistosomiasis. CONCLUSIONS Urinary schistosomiasis was prevalent among preschool children in Malengachanzi, Nkhotakota District. Contact with infested water puts these children and the general population at risk of infection and reinfection. Inclusion of preschool children in treatment programmes should be considered imperative, along with safe treatment guidelines. To prevent infection, the population in the area should be provided with health education and safe alternative water sources.
Collapse
Affiliation(s)
- V B Moyo
- Master of Environmental Sciences Programme, Faculty of Sciences, Chancellor College, University of Malawi, Zomba, Malawi
| | - W Changadeya
- Master of Environmental Sciences Programme, Faculty of Sciences, Chancellor College, University of Malawi, Zomba, Malawi
| | - S Chiotha
- Master of Environmental Sciences Programme, Faculty of Sciences, Chancellor College, University of Malawi, Zomba, Malawi
| | - D Sikawa
- Department of Aquaculture and Fisheries Science, Lilongwe University of Agriculture and Natural Resources, Lilongwe, Malawi
| |
Collapse
|
81
|
Palasio RGS, Guimarães MCDA, Ohlweiler FP, Tuan R. Molecular and morphological identification of Biomphalaria species from the state of São Paulo, Brazil. Zookeys 2017; 668:11-32. [PMID: 28769640 PMCID: PMC5534528 DOI: 10.3897/zookeys.668.10562] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/21/2017] [Indexed: 11/12/2022] Open
Abstract
DNA barcoding and morphological characters were used to identify adult snails belonging to the genus Biomphalaria from 17 municipalities in the state of São Paulo, Brazil. The DNA barcode analysis also included twenty-nine sequences retrieved from GenBank. The final data set of 104 sequences of the mitochondrial cytochrome oxidase I (COI) gene was analyzed for K2P intraspecific and interspecific divergences, through tree-reconstruction methods (Neighbor-Joining, Maximum Likelihood and Bayesian inference), and by applying different models (ABGD, bPTP, GMYC) to partition the sequences according to the pattern of genetic variation. Twenty-seven morphological parameters of internal organs were used to identify specimens. The molecular taxonomy of Biomphalaria agreed with the morphological identification of specimens from the same collection locality. DNA barcoding may therefore be a useful supporting tool for identifying Biomphalaria snails in areas at risk for schistosomiasis.
Collapse
Affiliation(s)
- Raquel Gardini Sanches Palasio
- Department of Epidemiology, Faculty of Public Health, University of São Paulo, Pinheiros, SP, Brazil
- Biochemistry and Molecular Biology Laboratory, Superintendency for the Control of Endemic Diseases, Rua Paula Sousa, 166, Luz, São Paulo, SP, Brazil
| | - Marisa Cristina de Almeida Guimarães
- Malacology Laboratory, Regional Service SR-02, Superintendency for the Control of Endemic Diseases, São Vicente, Rua João Ramalho, 587, Centro, SP, Brazil
| | - Fernanda Pires Ohlweiler
- Malacology Laboratory, Special Programs Division SR-01, Superintendency for the Control of Endemic Diseases, Rua Cardeal Arcoverde, 2878 - Pinheiros, SP, Brazil
| | - Roseli Tuan
- Biochemistry and Molecular Biology Laboratory, Superintendency for the Control of Endemic Diseases, Rua Paula Sousa, 166, Luz, São Paulo, SP, Brazil
| |
Collapse
|
82
|
Atalabi TE, Adubi TO, Lawal U. Rapid mapping of urinary schistosomiasis: An appraisal of the diagnostic efficacy of some questionnaire-based indices among high school students in Katsina State, northwestern Nigeria. PLoS Negl Trop Dis 2017; 11:e0005518. [PMID: 28369090 PMCID: PMC5391124 DOI: 10.1371/journal.pntd.0005518] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 04/13/2017] [Accepted: 03/21/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In sub-Saharan Africa, over 200 million individuals are estimated to be infected with urinary and intestinal schistosomiasis. In a bid to lay a foundation for effective future control programme, this study was carried out with the aim of assessing the diagnostic efficacy of some questionnaire-based rapid assessment indices of urinary schistosomiasis. METHODOLOGY A total number of 1,363 subjects were enrolled for the study. Questionnaires were administered basically in English and Hausa languages by trained personnel. Following informed consent, terminal urine samples were collected between 09:40 AM and 2:00 PM using clean 20 ml capacity universal bottles. 10μl of each urine residue was examined for the eggs of S. haematobium using x10 objective nose of Motic Binocular Light Microscope (China). PRINCIPAL FINDINGS The average age ± Standard Deviation (SD) of school children examined was 15.30 ± 2.30 years and 40.87% were females. The overall prevalence and geometric mean intensity of S. haematobium infection were 26.41% (24.10─28.85) and 6.59 (5.59─7.75) eggs / 10 ml of urine respectively. Interestingly, a questionnaire equivalence of the prevalence obtained in this survey was 26.41% (24.10─28.85) for Rapid Assessment Procedure based on self-reported blood in urine. The results of correlation analyses demonstrated significant associations between the prevalence of S. haematobium infection and contact with potentially infested open water sources (r = 0.741; P = 0.006). By regression model, cases of respondents with self-reported blood in urine are expected to rise to 24.75% if prevalence of the infection shoots up to 26.5%. CONCLUSIONS/SIGNIFICANCE The best RAP performance was obtained with self-reported blood in urine. Based on the overall prevalence value, the study area was at a "moderate-risk" of endemicity for urinary schistosomiasis. Chemotherapeutic intervention with Praziquantel, the rationale behind rapid assessment procedure for schistosomiasis, has been recommended to be carried out once in every 2 years for such communities.
Collapse
Affiliation(s)
- Tolulope Ebenezer Atalabi
- Department of Biological Sciences, Faculty of Science, Federal University Dutsinma, Dutsin-Ma, Katsina State, Nigeria
| | - Taiwo Oluwakemi Adubi
- Department of Biological Sciences and Biotechnology, College of Pure and Applied Sciences, Caleb University, Imota, Lagos State, Nigeria
| | - Umar Lawal
- Department of Biological Sciences, Faculty of Science, Federal University Dutsinma, Dutsin-Ma, Katsina State, Nigeria
| |
Collapse
|
83
|
Abstract
Early in the history of schistosomiasis research, children under 5 years of age were known to be infected. Although this problem was recognized over 100 years ago, insufficient action has been taken to address this issue. Under current policy, such infected children only receive their first antiparasitic treatment (praziquantel - PZQ) upon entry into primary school as current mass drug administration programmes typically target school-aged children. For many infected children, they will wait up to 6 years before receiving their first medication and significant schistosomiasis-related morbidity may have already established. This inequity would not be accepted for other diseases. To unveil some of the reasons behind this neglect, it is paramount to understand the intricate historical relationship between schistosomiasis and British Imperial medicine, to underline its lasting influence on today's public health priorities. This review presents a perspective on the historical neglect of paediatric schistosomiasis, focusing on important gaps that persist from the early days after discovery of this parasite. Looking to end this inequity, we address several issues that need to be overcome to move forward towards the lasting success of schistosomiasis control and elimination efforts.
Collapse
|
84
|
Campbell SJ, Stothard JR, O'Halloran F, Sankey D, Durant T, Ombede DE, Chuinteu GD, Webster BL, Cunningham L, LaCourse EJ, Tchuem-Tchuenté LA. Urogenital schistosomiasis and soil-transmitted helminthiasis (STH) in Cameroon: An epidemiological update at Barombi Mbo and Barombi Kotto crater lakes assessing prospects for intensified control interventions. Infect Dis Poverty 2017; 6:49. [PMID: 28238285 PMCID: PMC5327519 DOI: 10.1186/s40249-017-0264-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/17/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The crater lakes of Barombi Mbo and Barombi Kotto are well-known transmission foci of schistosomiasis and soil-transmitted helminthiasis having had several important control initiatives previously. To collect contemporary epidemiological information, a cross-sectional survey was undertaken inclusive of: signs and symptoms of disease, individual treatment histories, local water, sanitation and hygiene (WASH)-related factors and malacological surveillance, with molecular characterisation of specimens. METHODS At each lake, a community cross-sectional survey was undertaken using a combination of stool and urine parasitological sampling, and interview with pro-forma questionnaires. A total of 338 children and adults participated. Material from snail and parasite species were characterised by DNA methods. RESULTS Egg-patent prevalence of urogenital schistosomiasis was 8.7% at Barombi Mbo (all light-intensity infections) and 40.1% at Barombi Kotto (21.2% heavy-intensity infections). Intestinal schistosomiasis was absent. At Barombi Kotto, significantly more women reported signs and symptoms associated with female genital schistosomiasis. While there had been extensive recent improvement in WASH-related infrastructure at Barombi Mbo, water contact risk scores were higher among schistosomiasis-infected participants (P < 0.001) and at Barombi Kotto in general (P < 0.001). Across both lakes, mean prevalence of STH was very low (6.3%) evidencing an impressive decrease of 79.0% over the last decade; neither Strongyloides stercoralis nor Ascaris lumbricoides were found. A total of 29 freshwater sampling sites were inspected for snails, 13 in Barombi Mbo and 16 in Barombi Kotto; water chemistry differed significantly (P < 0.0001) between lakes for both mean pH (7.9 v. 9.6) and mean conductivity (64.3 μS v. 202.1 μS) respectively. Only two Bulinus camerunensis found on the central island of Barombi Kotto were observed to shed schistosome cercariae, but schistosome DNA was later detected in Bulinus sampled from both lakes as well as in Indoplanorbis exustus, an invasive species from Asia. CONCLUSIONS STH is currently at very low levels while urogenital schistosomiasis is of greatest concern at Barombi Kotto. This assessment highlights a unique opportunity for further study of the epidemiological dynamics at these crater lakes, to evaluate future intensified interventions both in terms of gaining and sustaining control at Barombi Kotto or in moving towards local interruption of transmission of both diseases at Barombi Mbo.
Collapse
Affiliation(s)
- Suzy J Campbell
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - J Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| | - Faye O'Halloran
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Deborah Sankey
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Timothy Durant
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | | | | | - Bonnie L Webster
- Department of Life Sciences, Parasites and Vectors Division, Natural History Museum, London, SW7 5BD, UK
- London Centre for Neglected Tropical Disease Research, London, UK
| | - Lucas Cunningham
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - E James LaCourse
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Louis-Albert Tchuem-Tchuenté
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
- Laboratory of Parasitology and Ecology, Faculty of Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National Programme for the Control of Schistosomiasis and Intestinal Helminthiasis, Ministry of Public Health, Yaoundé, Cameroon
| |
Collapse
|
85
|
Tchuem Tchuenté LA, Rollinson D, Stothard JR, Molyneux D. Moving from control to elimination of schistosomiasis in sub-Saharan Africa: time to change and adapt strategies. Infect Dis Poverty 2017; 6:42. [PMID: 28219412 PMCID: PMC5319063 DOI: 10.1186/s40249-017-0256-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 02/08/2017] [Indexed: 11/17/2022] Open
Abstract
Schistosomiasis is a water borne parasitic disease of global importance and with ongoing control the disease endemic landscape is changing. In sub-Saharan Africa, for example, the landscape is becoming ever more heterogeneous as there are several species of Schistosoma that respond in different ways to ongoing preventive chemotherapy and the inter-sectoral interventions currently applied. The major focus of preventive chemotherapy is delivery of praziquantel by mass drug administration to those shown to be, or presumed to be, at-risk of infection and disease. In some countries, regional progress may be uneven but in certain locations there are very real prospects to transition from control into interruption of transmission, and ultimately elimination. To manage this transition requires reconsideration of some of the currently deployed diagnostic tools used in surveillance and downward realignment of existing prevalence thresholds to trigger mass treatment. A key challenge will be maintaining and if possible, expanding the current donation of praziquantel to currently overlooked groups, then judging when appropriate to move from mass drug administration to selective treatment. In so doing, this will ensure the health system is adapted, primed and shown to be cost-effective to respond to these changing disease dynamics as we move forward to 2020 targets and beyond.
Collapse
Affiliation(s)
- Louis-Albert Tchuem Tchuenté
- National Programme for the Control of Schistosomiasis and STH, Ministry of Public Health, Yaoundé, Cameroon. .,Centre for Schistosomiasis and Parasitology, University of Yaoundé I, Yaoundé, Cameroon.
| | - David Rollinson
- Department of Life Sciences, The Natural History Museum, London, SW7 5BD, UK
| | - J Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - David Molyneux
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| |
Collapse
|
86
|
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: 67] [Impact Index Per Article: 8.4] [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
|
87
|
Stothard JR, Campbell SJ, Osei-Atweneboana MY, Durant T, Stanton MC, Biritwum NK, Rollinson D, Ombede DRE, Tchuem-Tchuenté LA. Towards interruption of schistosomiasis transmission in sub-Saharan Africa: developing an appropriate environmental surveillance framework to guide and to support 'end game' interventions. Infect Dis Poverty 2017; 6:10. [PMID: 28088239 PMCID: PMC5237522 DOI: 10.1186/s40249-016-0215-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/12/2016] [Indexed: 01/03/2023] Open
Abstract
Schistosomiasis is a waterborne parasitic disease in sub-Saharan Africa, particularly common in rural populations living in impoverished conditions. With the scale-up of preventive chemotherapy, national campaigns will transition from morbidity- to transmission-focused interventions thus formal investigation of actual or expected declines in environmental transmission is needed as 'end game' scenarios arise. Surprisingly, there are no international or national guidelines to do so in sub-Saharan Africa. Our article therefore provides an introduction to key practicalities and pitfalls in the development of an appropriate environmental surveillance framework. In this context, we discuss how strategies need to be adapted and tailored to the local level to better guide and support future interventions through this transition. As detection of egg-patent infection in people becomes rare, careful sampling of schistosome larvae in freshwater and in aquatic snails with robust species-specific DNA assays will be required. Appropriate metrics, derived from observed prevalence(s) as compared with predetermined thresholds, could each provide a clearer insight into contamination- and exposure-related dynamics. Application could be twofold, first to certify areas currently free from schistosomiasis transmission or second to red-flag recalcitrant locations where extra effort or alternative interventions are needed.
Collapse
Affiliation(s)
- J. Russell Stothard
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA UK
| | - Suzy J. Campbell
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA UK
| | - Mike Y. Osei-Atweneboana
- Department of Environmental Biology and Health, Council for Scientific and Industrial Research-Water Research Insitute, P.O. Box M 32, Accra, Ghana
| | - Timothy Durant
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA UK
| | - Michelle C. Stanton
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA UK
| | | | - David Rollinson
- Department of Life Sciences; Natural History Museum, Cromwell Road, London, SW7 5BD UK
| | - Dieudonné R. Eloundou Ombede
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
- Laboratory of Parasitology and Ecology, Faculty of Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Louis-Albert Tchuem-Tchuenté
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
- Laboratory of Parasitology and Ecology, Faculty of Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National Programme for the Control of Schistosomiasis and Intestinal Helminthiasis, Ministry of Public Health, Yaoundé, Cameroon
| |
Collapse
|
88
|
Al-Shehri H, Stanton MC, LaCourse JE, Atuhaire A, Arinaitwe M, Wamboko A, Adriko M, Kabatereine NB, Stothard JR. An extensive burden of giardiasis associated with intestinal schistosomiasis and anaemia in school children on the shoreline of Lake Albert, Uganda. Trans R Soc Trop Med Hyg 2016; 110:597-603. [PMID: 27864517 DOI: 10.1093/trstmh/trw072] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Water-borne parasitic diseases associated with poverty still blight the lives of African school children. In Uganda, intestinal schistosomiasis is still common along the shoreline of Lake Albert, despite ongoing control, and co-infection with giardiasis and malaria is poorly described. To shed light on putative interactions between diseases, a prospective cross-sectional parasitological survey was undertaken in five primary schools. METHODS Stool samples from 254 school children, aged 5-10 years, were examined by microscopy and rapid diagnostic tests (RDTs), with additional real-time PCR assays for detection of Giardia DNA. A finger-prick blood sample was also taken from each child and tested for malaria, and haemoblobin levels measured. Assocations between diseases and anaemia were assessed. RESULTS Intestinal schistosomiasis (46.5%), giardiasis (41.6%) and malaria (56.2%) were common, and a quarter of children were anaemic (<115 g/L). Up to 87.0% of children were excreting Giardia DNA and the prevalence of heavy infection by real-time PCR (Ct≤19) was 19.5%, being positively associated with light, moderate and heavy egg-patent schistosomiasis, as well as with anaemia. CONCLUSIONS In this setting, an extensive burden of giardiasis was revealed with heavy intensity infections associated with egg-patent intestinal schistosomiasis and anaemia. To improve child health, greater attention on giardiasis is needed along with exploring joined-up actions across diseases that promote better water hygiene and sanitation measures.
Collapse
Affiliation(s)
- Hajri Al-Shehri
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK.,Ministry of Health, Asir District, Kingdom of Saudi Arabia
| | | | | | - Aaron Atuhaire
- Vector Control Division, Ministry of Health, Kampala, Uganda
| | - Moses Arinaitwe
- Vector Control Division, Ministry of Health, Kampala, Uganda
| | - Aida Wamboko
- Vector Control Division, Ministry of Health, Kampala, Uganda
| | - Moses Adriko
- Vector Control Division, Ministry of Health, Kampala, Uganda
| | - Narcis B Kabatereine
- Vector Control Division, Ministry of Health, Kampala, Uganda.,Schistosomiasis Control Initiative, School of Public Health, Imperial College London, London W2 1PG, UK
| | | |
Collapse
|
89
|
Intestinal Schistosomiasis among Primary Schoolchildren in Two On-Shore Communities in Rorya District, Northwestern Tanzania: Prevalence, Intensity of Infection and Associated Risk Factors. J Parasitol Res 2016; 2016:1859737. [PMID: 27822385 PMCID: PMC5086394 DOI: 10.1155/2016/1859737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/15/2016] [Indexed: 12/15/2022] Open
Abstract
In Tanzania, Schistosoma mansoni is of great public health importance. Understanding the prevalence and infection intensity is important for targeted, evidence-based control strategies. This study aimed at studying the prevalence, intensity, and risk factors of S. mansoni among schoolchildren in the study area. A cross-sectional study was conducted in Busanga and Kibuyi villages. Sampled 513 schoolchildren provided stool specimens which were examined using kato-katz method. Pretested questionnaire was used to collect sociodemographic data and associated risk factors. The prevalence of S. mansoni infection was 84.01%, with geometric mean egg intensity of 167.13 (95% CI: 147.19–189.79) eggs per gram of stool (epg). Other parasites detected were Ascaris lumbricoides (1.4%) and hookworms (1.4%). The geometric mean infection intensity in Busanga and Kibuyi were 203.70 (95% CI: 169.67–244.56) and 135.98 (95% CI: 114.33–161.73) epg, respectively. Light, moderate, and heavy infection intensities were 34.11%, 39.91%, and 25.99%, respectively. Village of residence, parent's level of education, toilet use, and treatment history were predictors of infection. The high prevalence and infection intensity in this study were associated with village, parent's level of education, inconsistent toilet use, and treatment history. To control the disease among at-risk groups, these factors need to be considered in designing integrated schistosomiasis control interventions.
Collapse
|
90
|
Abstract
SUMMARYSchistosomiasis, commonly known as bilharzia, is a parasitic disease prevalent in Africa, Asia and South America. The majority of the cases occur in Sub-Saharan Africa where schistosomiasis is a major public health problem impacting on child health and development as well as adult health when infections become chronic. Control of schistosomiasis is by treatment of infected people with the antihelminthic drug praziquantel. Current schistosome control programmes advocated by the World Health Assembly in 2001 are aimed at regular school-based integrated deworming strategies in order to reduce development of severe morbidity, promote school health and to improve cognitive potential of children. Several countries in Africa have now embarked on national scale deworming programmes treating millions of children exposed to schistosomiasis in endemic areas without prior diagnosis of infection through mass drug administration programmes. Implementing such control programmes requires a concerted effort between scientists, policy makers, health practitioners and several other stake holders and of course a receptive community. This paper considers the contributions to global schistosome control efforts made by research conducted in Zimbabwe and the historical context and developments leading to the national schistosomiasis control programme in Zimbabwe giving an example of Getting Research into Policy and Practice.
Collapse
|
91
|
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: 3.8] [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
|
92
|
Wangchuk P, Pearson MS, Giacomin PR, Becker L, Sotillo J, Pickering D, Smout MJ, Loukas A. Compounds Derived from the Bhutanese Daisy, Ajania nubigena, Demonstrate Dual Anthelmintic Activity against Schistosoma mansoni and Trichuris muris. PLoS Negl Trop Dis 2016; 10:e0004908. [PMID: 27490394 PMCID: PMC4973903 DOI: 10.1371/journal.pntd.0004908] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/15/2016] [Indexed: 02/03/2023] Open
Abstract
Background Whipworms and blood flukes combined infect almost one billion people in developing countries. Only a handful of anthelmintic drugs are currently available to treat these infections effectively; there is therefore an urgent need for new generations of anthelmintic compounds. Medicinal plants have presented as a viable source of new parasiticides. Ajania nubigena, the Bhutanese daisy, has been used in Bhutanese traditional medicine for treating various diseases and our previous studies revealed that small molecules from this plant have antimalarial properties. Encouraged by these findings, we screened four major compounds isolated from A. nubigena for their anthelmintic properties. Methodology/Principal Findings Here we studied four major compounds derived from A. nubigena for their anthelmintic properties against the nematode whipworm Trichuris muris and the platyhelminth blood fluke Schistosoma mansoni using the xWORM assay technique. Of four compounds tested, two compounds—luteolin (3) and (3R,6R)-linalool oxide acetate (1)—showed dual anthelmintic activity against S. mansoni (IC50 range = 5.8–36.9 μg/mL) and T. muris (IC50 range = 9.7–20.4 μg/mL). Using scanning electron microscopy, we determined luteolin as the most efficacious compound against both parasites and additionally was found effective against the schistosomula, the infective stage of S. mansoni (IC50 = 13.3 μg/mL). Luteolin induced tegumental damage to S. mansoni and affected the cuticle, bacillary bands and bacillary glands of T. muris. Our in vivo assessment of luteolin (3) against T. muris infection at a single oral dosing of 100 mg/kg, despite being significantly (27.6%) better than the untreated control group, was markedly weaker than mebendazole (93.1%) in reducing the worm burden in mice. Conclusions/Significance Among the four compounds tested, luteolin demonstrated the best broad-spectrum activity against two different helminths—T. muris and S. mansoni—and was effective against juvenile schistosomes, the stage that is refractory to the current gold standard drug, praziquantel. Medicinal chemistry optimisation including cytotoxicity analysis, analogue development and structure-activity relationship studies are warranted and could lead to the identification of more potent chemical entities for the control of parasitic helminths of humans and animals. Schistosomiasis and trichuriasis affects millions of people worldwide and are caused by blood flukes and whipworms, respectively. Only a handful of anthelmintic drugs exist to treat these infections and the pipeline for the next generation of anthelmintic drugs is sparse, precipitating the need for new drug development. In this context, medicinal plants present a viable source of novel anthelmintic compounds. This inspired us to study the selected naturally occurring compounds derived from a Bhutanese daisy medicinal plant, Ajania nubigena for their anthelmintic activities. Here, using the xWORM motility assay, we demonstrate that two compounds, luteolin (3) and (3R,6R)-linalool oxide acetate (1), display significant broad-spectrum anthelmintic activity against two of the most important genera of human helminth parasites, the nematode whipworm and the platyhelminth blood fluke. Luteolin exhibited the best activities with IC50 values of 5.8 μg/mL against schistosomes and 9.7 μg/mL against whipworms. Using scanning electron microscopy we showed that luteolin damages the tegument of blood flukes and induces abnormalities in the bacillary bands/glands and cuticles of whipworms. Intriguingly, our previous study showed that luteolin (3) was effective against multi-drug resistant Plasmodium falciparum malaria. Due to its broad-spectrum anti-parasitic activities, luteolin (3) is a desirable drug lead scaffold, which could be used for developing effective compounds to control and treat numerous tropical diseases.
Collapse
Affiliation(s)
- Phurpa Wangchuk
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
- * E-mail:
| | - Mark S. Pearson
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| | - Paul R. Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| | - Luke Becker
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| | - Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| | - Darren Pickering
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| | - Michael J. Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, Cairns, Australia
| |
Collapse
|
93
|
N'Diaye M, Dioukhane EM, Ndao B, Diedhiou K, Diawara L, Talla I, Vernet C, Bessin F, Barbier D, Dewavrin P, Klotz F, Georges P. Schistosomiasis Sustained Control Program in Ethnic Groups Around Ninefescha (Eastern Senegal). Am J Trop Med Hyg 2016; 95:614-22. [PMID: 27430549 DOI: 10.4269/ajtmh.15-0125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/25/2016] [Indexed: 11/07/2022] Open
Abstract
Schistosomiasis is the second most significant parasitic disease in children in several African countries. For this purpose, the "Programme National de Lutte contre les Bilharzioses" (PNLB) was developed in partnership with the World Health Organization (WHO) to control this disease in Senegal. However, geographic isolation of Bedik ethnic groups challenged implementation of the key elements of the schistosomiasis program in eastern Senegal, and therefore, a hospital was established in Ninefescha to improve access to health care as well as laboratory support for this population. The program we have implemented from 2008 in partnership with the PNLB/WHO involved campaigns to 1) evaluate schistosomiasis prevalence in children of 53 villages around Ninefescha hospital, 2) perform a mass drug administration following the protocol established by the PNLB in school-aged children, 3) monitor annual prevalence, 4) implement health education campaigns, and 5) oversee the building of latrines. This campaign led to a drop in schistosomiasis prevalence but highlighted that sustainable schistosomiasis control by praziquantel treatment, awareness of the use of latrines, and inhabitants' voluntary commitment to the program are crucial to improve Schistosoma elimination. Moreover, this study revealed that preschool-aged children, for whom praziquantel was not recommended until 2014 in Senegal, constituted a significant reservoir for the parasite.
Collapse
Affiliation(s)
- Monique N'Diaye
- Université de Caen Normandie, UNICAEN, UFR des Sciences Pharmaceutiques, Laboratoire de Parasitologie et Mycologie Médicale, Caen, France.
| | | | | | - Kemo Diedhiou
- Programme National de Lutte contre les Bilharzioses/World Health Organization Neglected Tropical Diseases, Dakar, Senegal
| | - Lamine Diawara
- Inter-country Support Team for West Africa, World Health Organization Neglected Tropical Diseases, Ouagadougou, Burkina Faso
| | - Idrissa Talla
- Programme National de Lutte contre les Bilharzioses/World Health Organization Neglected Tropical Diseases, Dakar, Senegal
| | - Charlotte Vernet
- Université de Caen Normandie, UNICAEN, UFR des Sciences Pharmaceutiques, Laboratoire de Parasitologie et Mycologie Médicale, Caen, France
| | - François Bessin
- Université de Caen Normandie, UNICAEN, UFR des Sciences Pharmaceutiques, Laboratoire de Parasitologie et Mycologie Médicale, Caen, France
| | - Dominique Barbier
- Université de Caen Normandie, UNICAEN, UFR des Sciences Pharmaceutiques, Laboratoire de Parasitologie et Mycologie Médicale, Caen, France
| | | | | | - Pierre Georges
- Université de Caen Normandie, UNICAEN, UFR des Sciences Pharmaceutiques, Laboratoire de Parasitologie et Mycologie Médicale, Caen, France
| |
Collapse
|
94
|
A centenary of Robert T. Leiper's lasting legacy on schistosomiasis and a COUNTDOWN on control of neglected tropical diseases. Parasitology 2016; 144:1602-1612. [PMID: 27363810 PMCID: PMC5964474 DOI: 10.1017/s0031182016000998] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Part of Robert T. Leiper's (1881–1969) lasting legacy in medical helminthology is grounded on his pioneering work on schistosomiasis (Bilharzia). Having undertaken many expeditions to the tropics, his fascination with parasite life cycles typically allowed him to devise simple preventive measures that curtailed transmission. Building on his formative work with others in Africa and Asia, and again in Egypt in 1915, he elucidated the life cycles of African schistosomes. His mandate, then commissioned by the British War Office, was to prevent and break transmission of this disease in British troops. This he did by raising standing orders based on simple water hygiene measures. Whilst feasible in military camp settings, today their routine implementation is sadly out of reach for millions of Africans living in poverty. Whilst we celebrate the centenary of Leiper's research we draw attention to some of his lesser known colleagues, then focus on schistosomiasis in Uganda discussing why expanded access to treatment with praziquantel is needed now. Looking to WHO 2020 targets for neglected tropical diseases, we introduce COUNTDOWN, an implementation research consortium funded by DFID, UK, which fosters the scale-up of interventions and confirm the current relevance of Leiper's original research.
Collapse
|
95
|
Alemu A, Tegegne Y, Damte D, Melku M. Schistosoma mansoni and soil-transmitted helminths among preschool-aged children in Chuahit, Dembia district, Northwest Ethiopia: prevalence, intensity of infection and associated risk factors. BMC Public Health 2016; 16:422. [PMID: 27216255 PMCID: PMC4876558 DOI: 10.1186/s12889-016-2864-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/12/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intestinal schistosomiasis and soil-transmitted helminthiasis are the major public health problems globally. Compared with any other age group, pre-school aged children and school-aged children are the most exposed. There are few studies showing the burden of intestinal schistosomiasis, and soil-transmitted helminthiasis among pre-school aged children in Ethiopia. Hence, this study aimed to assess the prevalence of schistosoma mansoni and soil-transmitted helminths and associated risk factors among preschool aged children of Chuahit and surrounding Kebeles, Northwest Ethiopia. METHODS A community based cross sectional study was conducted from February 2 to March 27 2015. Four hundred one preschool-aged children were included in the study by using two stage cluster sampling technique. Pretested structured questionnaire was employed to collected data via face-to-face interview technique. A single stool specimen was collected, and a portion of the sample was processed by Kato Katz method. RESULTS Of the total children, 141 (35.2 %) harbored one or more intestinal helminthes. Schistosoma mansoni was found in 45 (11.2 %) of preschool age children. Ascaris lumbricoides was the predominant isolate, 77 (19.2 %) followed by S. mansoni, 45 (11.2 %). The least parasites isolated were Tania species, 2 (0.5 %). After adjusting for other variables, being mothers who did not have the habit of washing hands after toilet (AOR = 7.3, 95%CI: 2.97-17.95), being occupationally housewife mothers (AOR = 8.9, 95%CI: 2.27-25.4), using protected spring water as a main family source of water (AOR = 3.9, 95%CI: 1.2-12.3) and child habit of not wearing shoe (AOR = 1.91, 95%CI: 1.01-3.64) were significantly associated with high prevalence of soil-transmitted helminthiasis among preschool-age children in Chuahit. CONCLUSION The current study showed that relatively higher level of STH and S. mansoni among preschool-aged children in Chuahit. This finding calls for a need of public health education, promotion of women education and provision of safe water to reduce the burden of soil-transmitted intestinal helminthiasis and schistosomiaisis.
Collapse
Affiliation(s)
- Agersew Alemu
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yalewayker Tegegne
- Gondar University Hospital, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Demekech Damte
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulugeta Melku
- Department of Hematology and Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
96
|
Xu J, Bergquist R, Qian YJ, Wang Q, Yu Q, Peeling R, Croft S, Guo JG, Zhou XN. China-Africa and China-Asia Collaboration on Schistosomiasis Control: A SWOT Analysis. ADVANCES IN PARASITOLOGY 2016; 92:435-66. [PMID: 27137455 DOI: 10.1016/bs.apar.2016.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Schistosomiasis, a disease caused by a trematode, parasitic worm, is a worldwide public health problem. In spite of great progress with regard to morbidity control, even elimination of this infection in recent decades, there are still challenges to overcome in sub-Saharan Africa and endemic areas in Southeast Asia. Regarded as one of the most successful countries with respect to schistosomiasis control, The People's Republic of China has accumulated considerable experience and learnt important lessons in various local settings that could benefit schistosomiasis control in other endemic countries. Based on an analysis of conceived strengths, weaknesses, opportunities and threats (SWOT) of potential collaborative activities with regard to schistosomiasis in Africa and Asia, this article addresses the importance of collaborative efforts and explores the priorities that would be expected to facilitate the transfer of Chinese experience to low- and middle-income countries in Africa and Asia.
Collapse
Affiliation(s)
- J Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, The People's Republic of China; Key Laboratory of Parasite & Vector Biology, Ministry of Public Health, Shanghai, The People's Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, The People's Republic of China
| | - R Bergquist
- Geospatial Health, University of Naples Federico II, Naples, Italy
| | - Y-J Qian
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, The People's Republic of China; Key Laboratory of Parasite & Vector Biology, Ministry of Public Health, Shanghai, The People's Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, The People's Republic of China
| | - Q Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, The People's Republic of China; Key Laboratory of Parasite & Vector Biology, Ministry of Public Health, Shanghai, The People's Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, The People's Republic of China
| | - Q Yu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, The People's Republic of China; Key Laboratory of Parasite & Vector Biology, Ministry of Public Health, Shanghai, The People's Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, The People's Republic of China
| | - R Peeling
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - S Croft
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - J-G Guo
- World Health Organization, Geneva, Switzerland
| | - X-N Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, The People's Republic of China; Key Laboratory of Parasite & Vector Biology, Ministry of Public Health, Shanghai, The People's Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, The People's Republic of China
| |
Collapse
|
97
|
Bélard S, Tamarozzi F, Bustinduy AL, Wallrauch C, Grobusch MP, Kuhn W, Brunetti E, Joekes E, Heller T. Point-of-Care Ultrasound Assessment of Tropical Infectious Diseases--A Review of Applications and Perspectives. Am J Trop Med Hyg 2016; 94:8-21. [PMID: 26416111 PMCID: PMC4710450 DOI: 10.4269/ajtmh.15-0421] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/18/2015] [Indexed: 12/28/2022] Open
Abstract
The development of good quality and affordable ultrasound machines has led to the establishment and implementation of numerous point-of-care ultrasound (POCUS) protocols in various medical disciplines. POCUS for major infectious diseases endemic in tropical regions has received less attention, despite its likely even more pronounced benefit for populations with limited access to imaging infrastructure. Focused assessment with sonography for HIV-associated TB (FASH) and echinococcosis (FASE) are the only two POCUS protocols for tropical infectious diseases, which have been formally investigated and which have been implemented in routine patient care today. This review collates the available evidence for FASH and FASE, and discusses sonographic experiences reported for urinary and intestinal schistosomiasis, lymphatic filariasis, viral hemorrhagic fevers, amebic liver abscess, and visceral leishmaniasis. Potential POCUS protocols are suggested and technical as well as training aspects in the context of resource-limited settings are reviewed. Using the focused approach for tropical infectious diseases will make ultrasound diagnosis available to patients who would otherwise have very limited or no access to medical imaging.
Collapse
Affiliation(s)
- Sabine Bélard
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Francesca Tamarozzi
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Amaya L Bustinduy
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Claudia Wallrauch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Martin P Grobusch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Walter Kuhn
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Enrico Brunetti
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Elizabeth Joekes
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Tom Heller
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Pneumology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, World Health Organization Collaborating Centre for Clinical Management of Cystic Echinococcosis, University of Pavia, Pavia, Italy; Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom; Department of Medicine, Klinikum Muenchen-Perlach, Munich, Germany; Center for Operational Medicine, Medical College Georgia, Georgia Regents University, Augusta, Georgia; Division of Infectious and Tropical Diseases, University of Pavia/IRCCS San Matteo Hospital Foundation, Pavia, Italy; Department of Radiology, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| |
Collapse
|
98
|
Tambo E, Khater EIM, Chen JH, Bergquist R, Zhou XN. Nobel prize for the artemisinin and ivermectin discoveries: a great boost towards elimination of the global infectious diseases of poverty. Infect Dis Poverty 2015; 4:58. [PMID: 26708575 PMCID: PMC4692067 DOI: 10.1186/s40249-015-0091-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/10/2015] [Indexed: 11/15/2022] Open
Abstract
The Millennium Development Goals (MDGs) made a marked transformation for neglected and vulnerable communities in the developing countries from the start, but infectious diseases of poverty (IDoPs) continue to inflict a disproportionate global public health burden with associated consequences, thereby contributing to the vicious cycle of poverty and inequity. However, the effectiveness and large-scale coverage of artemisinin combination therapy (ACT) have revolutionized malaria treatment just as the control of lymphatic filariasis (LF) and onchocerciasis have benefitted from harnessing the broad-spectrum effect of avermectin-based derivatives. The paradigm shift in therapeutic approach, effected by these two drugs and their impact on community-based interventions of parasitic diseases plaguing the endemic low- and middle-income countries (LIMCs), led to the Nobel Prize in Physiology or Medicine in 2015. However, the story would not be complete without mentioning praziquantel. The huge contribution of this drug in modernizing the control of schistosomiasis and also some intestinal helminth infections had already shifted the focus from control to potential elimination of this disease. Together, these new drugs have provided humankind with powerful new tools for the alleviation of infectious diseases that humans have lived with since time immemorial. These drugs all have broad-spectrum effects, yet they are very safe and can even be packaged together in various combinations. The strong effect on so many of the great infectious scourges in the developing countries has not only had a remarkable influence on many endemic diseases, but also contributed to improving the cost structure of healthcare. Significant benefits include improved quality of preventive and curative medicine, promotion of community-based interventions, universal health coverage and the fostering of global partnerships. The laudable progress and benefits achieved are indispensable in championing, strengthening and moving forward elimination of the IDoPs. However, there is an urgent need for further innovative, contextual and integrated approaches along with the advent of the Sustainable Development Goals (SDGs), replacing the MDGs in ensuring global health security, well-being and economic prosperity for all.
Collapse
Affiliation(s)
- Ernest Tambo
- Department of Biochemistry and Pharmaceutical Sciences, Higher Institute of Health Sciences, Université des Montagnes, Bangangté, Cameroon. .,Sydney Brenner Institute for Molecular Biosciences, University of the Witwatersrand, Johannesburg, South Africa. .,Africa Disease Intelligence and Surveillance, Communication and Response Foundation (Africa DISCoR), Yaoundé, Cameroon. .,Center for Sustainable Malaria Control, Department of Biochemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa.
| | - Emad I M Khater
- Public Health Pests Laboratory, Jeddah Municipality, Jeddah, Saudi Arabia.,Department of Entomology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, P.R. China. .,Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, 200025, P.R. China. .,WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025, P.R. China.
| | | | - Xiao-Nong Zhou
- Public Health Pests Laboratory, Jeddah Municipality, Jeddah, Saudi Arabia. .,Department of Entomology, Faculty of Science, Ain Shams University, Cairo, Egypt. .,National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, P.R. China.
| |
Collapse
|
99
|
King CH, Sutherland LJ, Bertsch D. Systematic Review and Meta-analysis of the Impact of Chemical-Based Mollusciciding for Control of Schistosoma mansoni and S. haematobium Transmission. PLoS Negl Trop Dis 2015; 9:e0004290. [PMID: 26709922 PMCID: PMC4692485 DOI: 10.1371/journal.pntd.0004290] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022] Open
Abstract
Background Programs for schistosomiasis control are advancing worldwide, with many benefits noted in terms of disease reduction. Yet risk of reinfection and recurrent disease remain, even in areas with high treatment coverage. In the search for means to better prevent new Schistosoma infections, attention has returned to an older strategy for transmission control, i.e., chemical mollusciciding, to suppress intermediate host snail species responsible for S. mansoni and S. haematobium transmission. The objective of this systematic review and meta-analysis was to summarize prior experience in molluscicide-based control of Bulinus and Biomphalaria spp. snails, and estimate its impact on local human Schistosoma infection. Methodology/Principal Findings The review was registered at inception with PROSPERO (CRD42013006869). Studies were identified by online database searches and hand searches of private archives. Eligible studies included published or unpublished mollusciciding field trials performed before January 2014 involving host snails for S. mansoni or S. haematobium, with a primary focus on the use of niclosamide. Among 63 included papers, there was large variability in terms of molluscicide dosing, and treatment intervals varied from 3–52 weeks depending on location, water source, and type of application. Among 35 studies reporting on prevalence, random effects meta-analysis indicated that, on average, odds of infection were reduced 77% (OR 0.23, CI95% 0.17, 0.31) during the course of mollusciciding, with increased impact if combined with drug therapy, and progressively greater impact over time. In 17 studies reporting local incidence, risk of new infection was reduced 64% (RR 0.36 CI95% 0.25, 0.5), but additional drug treatment did not appear to influence incidence effects. Conclusion/Significance While there are hurdles to implementing molluscicide control, its impact on local transmission is typically strong, albeit incomplete. Based on past experience, regular focal mollusciciding is likely to contribute significantly to the move toward elimination of schistosomiasis in high risk areas. Infection with Schistosoma blood flukes is a leading cause of chronic parasitic disease in at-risk areas of Africa, South America, Asia, and the Philippines. Over past decades, many national programs have implemented regular drug treatment to control or prevent the advanced complications of Schistosoma infection. However, these periodic treatments do not stop transmission of the parasite, which occurs when human sewage contaminates local water bodies and parasite eggs infect intermediate host snails. In this systematic review, we collated past experience of using chemically-mediated snail control for prevention of schistosomiasis. This approach, used in many Schistosoma-affected countries before the advent of the current oral drug regimens, has the potential to significantly reduce transmission if properly applied. Our meta-analysis of 63 studies (performed 1953–1981) catalogued a wide variety of water treatments and schedules employed. Among studies reporting on human infection, we found that snail control reduced local human prevalence and incidence of infection in most, but not all locations. Estimates from the aggregated studies indicate that snail control (alone) typically reduced new infections by 64% and local prevalence declined over a period of years. This decline was accelerated and more profound (84% reduction) if drug treatment was also made available.
Collapse
Affiliation(s)
- 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
- * E-mail:
| | - Laura J. Sutherland
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David Bertsch
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
100
|
Abstract
Mass drug administration using praziquantel is the backbone of the current strategy for the control of schistosomiasis. As the theoretical plans have moved into practical application, certain challenges with this approach have surfaced, and it is likely that annual mass drug administration alone may not be sufficient to achieve program goals. However, mass drug administration is still the only available intervention that can be readily used in the wide variety of settings where schistosomiasis is endemic. The task then becomes how to improve this approach and identify what adjuncts to mass drug administration are effective, as programs move from morbidity control to elimination goals. Other aspects worthy of consideration include how best to employ new diagnostic tools to more easily identify where treatment is needed, and new formulations of praziquantel to extend the availability of treatment to all age groups. The aim of this review is to highlight both areas of challenge and of opportunity to improve the public health impact of schistosomiasis control programs.
Collapse
Affiliation(s)
- W Evan Secor
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, 30329, USA
| |
Collapse
|