1
|
Graeff-Teixeira C, Marcolongo-Pereira C, Kersanach BB, Geiger SM, Negrão-Correa D. Descriptive study on risk of increased morbidity of schistosomiasis and graft loss after liver transplantation. Rev Soc Bras Med Trop 2024; 57:e00201. [PMID: 39082515 PMCID: PMC11290851 DOI: 10.1590/0037-8682-0097-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/29/2024] [Indexed: 08/02/2024] Open
Abstract
Solid-organ transplantation procedures have witnessed a surge in frequency. Consequently, increased attention to associated infections and their impact on graft success is warranted. The liver is the principal target for infection by the flatworm Schistosoma mansoni. Hence, rigorous screening protocols for this parasite should be implemented for liver transplantation donors and recipients. This study investigated the risks posed by schistosomiasis-infected liver tissues for successful liver transplantation (LT), considering donors and recipients, by analyzing reported cases. Among the 43 patients undergoing LT (donors = 19; recipients = 24), 32 were infected with S. mansoni, five were infected with other Schistosoma species, and no identification was made in four patients. Reported follow-up periods ranged from 1 to 132 months, and all patients achieved successful recovery. As these helminths do not replicate in their vertebrate hosts, immunosuppressive treatment is not expected to promote increased morbidity or reactivation. Moreover, suspected or confirmed schistosomiasis infections often have a benign course, and generally, should not prevent LT. The available literature was reviewed and a provisional screening protocol has been proposed.
Collapse
Affiliation(s)
- Carlos Graeff-Teixeira
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia e Núcleo de Doenças Infecciosas, Vitória, ES, Brasil
| | - Clairton Marcolongo-Pereira
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia e Núcleo de Doenças Infecciosas, Vitória, ES, Brasil
- Centro Universitário do Espírito Santo, Faculdade de Medicina, Colatina, ES, Brasil
| | - Betina Bolina Kersanach
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia e Núcleo de Doenças Infecciosas, Vitória, ES, Brasil
| | - Stefan Michael Geiger
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| | - Deborah Negrão-Correa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Laboratório de Esquistossomose e Imuno-helmintologia - Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| |
Collapse
|
2
|
Ekloh W, Asafu-Adjaye A, Tawiah-Mensah CNL, Ayivi-Tosuh SM, Quartey NKA, Aiduenu AF, Gayi BK, Koudonu JAM, Basing LA, Yamoah JAA, Dofuor AK, Osei JHN. A comprehensive exploration of schistosomiasis: Global impact, molecular characterization, drug discovery, artificial intelligence and future prospects. Heliyon 2024; 10:e33070. [PMID: 38988508 PMCID: PMC11234110 DOI: 10.1016/j.heliyon.2024.e33070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
Schistosomiasis, one of the neglected tropical diseases which affects both humans and animals, is caused by trematode worms of the genus Schistosoma. The disease is caused by several species of Schistosoma which affect several organs such as urethra, liver, bladder, intestines, skin and bile ducts. The life cycle of the disease involves an intermediate host (snail) and a mammalian host. It affects people who are in close proximity to water bodies where the intermediate host is abundant. Common clinical manifestations of the disease at various stages include fever, chills, headache, cough, dysuria, hyperplasia and hydronephrosis. To date, most of the control strategies are dependent on effective diagnosis, chemotherapy and public health education on the biology of the vectors and parasites. Microscopy (Kato-Katz) is considered the golden standard for the detection of the parasite, while praziquantel is the drug of choice for the mass treatment of the disease since no vaccines have yet been developed. Most of the previous reviews on schistosomiasis have concentrated on epidemiology, life cycle, diagnosis, control and treatment. Thus, a comprehensive review that is in tune with modern developments is needed. Here, we extend this domain to cover historical perspectives, global impact, symptoms and detection, biochemical and molecular characterization, gene therapy, current drugs and vaccine status. We also discuss the prospects of using plants as potential and alternative sources of novel anti-schistosomal agents. Furthermore, we highlight advanced molecular techniques, imaging and artificial intelligence that may be useful in the future detection and treatment of the disease. Overall, the proper detection of schistosomiasis using state-of-the-art tools and techniques, as well as development of vaccines or new anti-schistosomal drugs may aid in the elimination of the disease.
Collapse
Affiliation(s)
- William Ekloh
- Department of Biochemistry, School of Biological Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Andy Asafu-Adjaye
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Christopher Nii Laryea Tawiah-Mensah
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | | | - Naa Kwarley-Aba Quartey
- Department of Food Science and Technology, Faculty of Biosciences, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Albert Fynn Aiduenu
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Blessing Kwabena Gayi
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | | | - Laud Anthony Basing
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jennifer Afua Afrifa Yamoah
- Animal Health Division, Council for Scientific and Industrial Research-Animal Research Institute, Adenta-Frafraha, Accra, Ghana
| | - Aboagye Kwarteng Dofuor
- Department of Biological Sciences, School of Natural and Environmental Sciences, University of Environment and Sustainable Development, Somanya, Ghana
| | - Joseph Harold Nyarko Osei
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
3
|
Obonyo CO, Rawago FO, Makworo NK, Muok EMO. Efficacy and safety of single-dose artesunate plus sulfalene/pyrimethamine combined with praziquantel for the treatment of children with Schistosoma mansoni or Schistosoma haematobium in western Kenya: a randomised, open-label controlled trial. Parasit Vectors 2024; 17:279. [PMID: 38943214 PMCID: PMC11212220 DOI: 10.1186/s13071-024-06359-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/16/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Reliance on praziquantel for the treatment and control of schistosomiasis is likely to facilitate the emergence of drug resistance. Combination therapy targeting adult and juvenile schistosome worms is urgently needed to improve praziquantel efficacy and delay the potential development of drug resistance. We assessed the efficacy and safety of single-dose praziquantel combined with single-dose artesunate plus sulfalene-pyrimethamine in the treatment of Kenyan children with schistosomiasis. METHODS This was an open-label, randomised clinical trial involving 426 school-aged children (7-15 years old) diagnosed with Schistosoma mansoni (by Kato-Katz) or S. haematobium (by urine filtration). They were randomly assigned (1:1:1) to receive a single dose of praziquantel (40 mg/kg), a single dose of artesunate plus sulfalene-pyrimethamine (12 mg/kg artesunate) or combination therapy using a single dose of praziquantel (40 mg/kg) combined with a single dose of artesunate plus sulfalene-pyrimethamine (12 mg/kg artesunate). The primary outcome was cure and egg reduction rates at 6 weeks post-treatment in the available case population. Adverse events were assessed within 3 h after treatment. RESULTS Of the 426 children enrolled, 135 received praziquantel, 150 received artesunate plus sulfalene-pyrimethamine, and 141 received combination therapy. Outcome data were available for 348 (81.7%) children. For S. mansoni-infected children (n = 335), the cure rates were 75.6%, 60.7%, and 77.8%, and the egg reduction rates were 80.1%, 85.0%, and 88.4% for praziquantel, artesunate plus sulfalene-pyrimethamine, and combination therapy, respectively. For S. haematobium-infected children (n = 145), the corresponding cure rates were 81.4%, 71.1%, and 82.2%, and the egg reduction rates were 95.6%, 97.1%, and 97.7%, respectively. Seventy-one (16.7%) children reported mild-intensity adverse events. The drugs were well tolerated and no serious adverse events were reported. CONCLUSIONS A single oral dose of praziquantel combined with artesunate plus sulfalene-pyrimethamine cured a high proportion of children with S. haematobium but did not significantly improve the treatment efficacy for either urinary or intestinal schistosomiasis. Sequential administration of praziquantel and artesunate plus sulfalene-pyrimethamine may enhance the efficacy and safety outcomes.
Collapse
Affiliation(s)
- Charles O Obonyo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.
| | - Fredrick O Rawago
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Nicholas K Makworo
- Division of Vector-Borne and Neglected Tropical Diseases, County Department of Health, Migori, Kenya
| | - Erick M O Muok
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| |
Collapse
|
4
|
Rakotozandrindrainy R, Rakotoarivelo RA, Kislaya I, Marchese V, Rasamoelina T, Solonirina J, Ratiaharison EF, Razafindrakoto R, Razafindralava NM, Rakotozandrindrainy N, Radomanana M, Andrianarivelo MR, Klein P, Lorenz E, Jaeger A, Hoekstra PT, Corstjens PLAM, Schwarz NG, van Dam GJ, May J, Fusco D. Schistosome infection among pregnant women in the rural highlands of Madagascar: A cross-sectional study calling for public health interventions in vulnerable populations. PLoS Negl Trop Dis 2024; 18:e0011766. [PMID: 38626192 PMCID: PMC11051649 DOI: 10.1371/journal.pntd.0011766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/26/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024] Open
Abstract
INTRODUCTION Schistosomiasis is a parasitic infection highly prevalent in sub-Saharan Africa (SSA) with Madagascar being among the countries with highest burden of the disease worldwide. Despite WHO recommendations, suggesting treatment of pregnant women after the first trimester, this group is still excluded from Mass Drug Administration programs. Our study, had the objective to measure the prevalence of schistosome infection among pregnant women in Madagascar in order to inform public health policies for treatment in this vulnerable population. METHODS Women were recruited for this cross-sectional study between April 2019 and February 2020 when attending Antenatal Care Services (ANCs) at one of 42 included Primary Health Care Centers. The urine-based upconverting reporter particle, lateral flow (UCP-LF) test detecting circulating anodic antigen was used for the detection of schistosome infections. To identify factors associated with the prevalence of schistosome infection crude and adjusted prevalence ratios and 95% CIs were estimated using mixed-effect Poisson regression. RESULTS Among 4,448 participating women aged between 16 and 47 years, the majority (70.4%, 38 n = 3,133) resided in rural settings. Overall, the prevalence of schistosome infection was 55.9% (n = 2486, CI 95%: 53.3-58.5). A statistically significant association was found with age group (increased prevalence in 31-47 years old, compared to 16-20 years old (aPR = 1.15, CI 95%: 1.02-1.29) and with uptake of antimalaria preventive treatment (decreased prevalence, aPR = 0.85, CI 95%: 0.77-0.95). No other associations of any personal characteristics or contextual factors with schistosome infection were found in our multivariate regression analysis. DISCUSSION AND CONCLUSION The high prevalence of schistosome infection in pregnant women supports the consideration of preventive schistosomiasis treatment in ANCs of the Malagasy highlands. We strongly advocate for adapting schistosomiasis programs in highly endemic contexts. This, would contribute to both the WHO and SDGs agendas overall to improving the well-being of women and consequently breaking the vicious cycle of poverty perpetuated by schistosomiasis.
Collapse
Affiliation(s)
| | | | - Irina Kislaya
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Valentina Marchese
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | | | | | | | | | | | | | - Mickael Radomanana
- Infectious diseases service, University Hospital Tambohobe, Fianarantsoa, Madagascar
| | | | - Philipp Klein
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Eva Lorenz
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Anna Jaeger
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | | | | | - Norbert Georg Schwarz
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | | | - Jürgen May
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Daniela Fusco
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | | |
Collapse
|
5
|
Bottieau E, Mbow M, Brosius I, Roucher C, Gueye CT, Mbodj OT, Faye BT, De Hondt A, Smekens B, Arango D, Burm C, Tsoumanis A, Paredis L, Van Herrewege Y, Potters I, Richter J, Rosanas-Urgell A, Cissé B, Mboup S, Polman K. Antimalarial artesunate-mefloquine versus praziquantel in African children with schistosomiasis: an open-label, randomized controlled trial. Nat Med 2024; 30:130-137. [PMID: 38177851 PMCID: PMC10803269 DOI: 10.1038/s41591-023-02719-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024]
Abstract
Schistosomiasis treatment entirely relies on a single drug, praziquantel, prompting research into alternative therapeutics. Here we evaluated the efficacy and safety of the antimalarial combination artesunate-mefloquine for the treatment of schistosomiasis in a proof-of-concept, pragmatic, open-label, randomized controlled trial in primary schools of six villages endemic for schistosomiasis in northern Senegal. Children (6-14 years) were eligible if Schistosoma eggs were detected by microscopy in urine and/or stool. In total, 726 children were randomized 1:1 to praziquantel (standard care: 40 mg kg-1 single dose; n = 364) or to artesunate-mefloquine (antimalarial dosage: artesunate 4 mg kg-1 and mefloquine 8 mg kg-1 daily for three consecutive days; n = 362). Eight children not meeting the inclusion criteria were excluded from efficacy analysis. Median age of the remaining 718 participants was 9 years; 399 (55.6%) were male, and 319 (44.4%) female; 99.3% were infected with Schistosoma haematobium and 15.2% with S. mansoni. Primary outcomes were cure rate, assessed by microscopy, and frequency of drug-related adverse effects of artesunate-mefloquine versus praziquantel at 4 weeks after treatment. Cure rate was 59.6% (208/349) in the artesunate-mefloquine arm versus 62.1% (211/340) in the praziquantel arm. The difference of -2.5% (95% confidence interval (CI) -9.8 to 4.8) met the predefined criteria of noninferiority (margin set at 10%). All drug-related adverse events were mild or moderate, and reported in 28/361 children receiving artesunate-mefloquine (7.8%; 95% CI 5.4 to 11.0) versus 8/363 (2.2%; 95% CI 1.1 to 4.3) receiving praziquantel (P < 0.001). Artesunate-mefloquine at antimalarial dosage was moderately safe and noninferior to standard-care praziquantel for the treatment of schistosomiasis, predominantly due to S. haematobium. Multicentric trials in different populations and epidemiological settings are needed to confirm these findings. ClinicalTrials.gov identifier: NCT03893097 .
Collapse
Affiliation(s)
- Emmanuel Bottieau
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.
| | - Moustapha Mbow
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
- Department of Immunology, Cheikh Anta Diop University, Dakar, Senegal
| | - Isabel Brosius
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Clémentine Roucher
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Cheikh Tidiane Gueye
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Ousmane Thiam Mbodj
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Babacar Thiendella Faye
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Annelies De Hondt
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Bart Smekens
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Diana Arango
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Christophe Burm
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Achilleas Tsoumanis
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Linda Paredis
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Yven Van Herrewege
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Idzi Potters
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Joachim Richter
- Institute of Tropical Medicine and International Health, Charité Universitätsmedizin, Berlin, Germany
| | - Anna Rosanas-Urgell
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Badara Cissé
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Souleymane Mboup
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Katja Polman
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Pfarr KM, Krome AK, Al-Obaidi I, Batchelor H, Vaillant M, Hoerauf A, Opoku NO, Kuesel AC. The pipeline for drugs for control and elimination of neglected tropical diseases: 2. Oral anti-infective drugs and drug combinations for off-label use. Parasit Vectors 2023; 16:394. [PMID: 37907954 PMCID: PMC10619278 DOI: 10.1186/s13071-023-05909-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/31/2023] [Indexed: 11/02/2023] Open
Abstract
In its 'Road map for neglected tropical diseases 2021-2030', the World Health Organization outlined its targets for control and elimination of neglected tropical diseases (NTDs) and research needed to achieve them. For many NTDs, this includes research for new treatment options for case management and/or preventive chemotherapy. Our review of small-molecule anti-infective drugs recently approved by a stringent regulatory authority (SRA) or in at least Phase 2 clinical development for regulatory approval showed that this pipeline cannot deliver all new treatments needed. WHO guidelines and country policies show that drugs may be recommended for control and elimination for NTDs for which they are not SRA approved (i.e. for 'off-label' use) if efficacy and safety data for the relevant NTD are considered sufficient by WHO and country authorities. Here, we are providing an overview of clinical research in the past 10 years evaluating the anti-infective efficacy of oral small-molecule drugs for NTD(s) for which they are neither SRA approved, nor included in current WHO strategies nor, considering the research sponsors, likely to be registered with a SRA for that NTD, if found to be effective and safe. No such research has been done for yaws, guinea worm, Trypanosoma brucei gambiense human African trypanosomiasis (HAT), rabies, trachoma, visceral leishmaniasis, mycetoma, T. b. rhodesiense HAT, echinococcosis, taeniasis/cysticercosis or scabies. Oral drugs evaluated include sparfloxacin and acedapsone for leprosy; rifampicin, rifapentin and moxifloxacin for onchocerciasis; imatinib and levamisole for loiasis; itraconazole, fluconazole, ketoconazole, posaconazole, ravuconazole and disulfiram for Chagas disease, doxycycline and rifampicin for lymphatic filariasis; arterolane, piperaquine, artesunate, artemether, lumefantrine and mefloquine for schistosomiasis; ivermectin, tribendimidine, pyrantel, oxantel and nitazoxanide for soil-transmitted helminths including strongyloidiasis; chloroquine, ivermectin, balapiravir, ribavirin, celgosivir, UV-4B, ivermectin and doxycycline for dengue; streptomycin, amoxicillin, clavulanate for Buruli ulcer; fluconazole and isavuconazonium for mycoses; clarithromycin and dapsone for cutaneous leishmaniasis; and tribendimidine, albendazole, mebendazole and nitazoxanide for foodborne trematodiasis. Additional paths to identification of new treatment options are needed. One promising path is exploitation of the worldwide experience with 'off-label' treatment of diseases with insufficient treatment options as pursued by the 'CURE ID' initiative.
Collapse
Affiliation(s)
- Kenneth M Pfarr
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Anna K Krome
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Bonn, Germany
| | - Issraa Al-Obaidi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Michel Vaillant
- Competence Center for Methodology and Statistics, Luxembourg Institute of Health, Strassen, Grand Duchy of Luxembourg
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Nicholas O Opoku
- Department of Epidemiology and Biostatistics School of Public Health, University of Health and Allied Sciences, Hohoe, Ghana
| | - Annette C Kuesel
- UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases (WHO/TDR), World Health Organization, Geneva, Switzerland.
| |
Collapse
|
7
|
Bocanegra C, Álvarez-Martínez MJ, Arsuaga Vicente M, Belhassen-García M, Chamorro Tojeiro S, Camprubí-Ferrer D, Fernández Soto P, García Vázquez E, Herrador Ortiz Z, Martín O, Muro A, Pérez Arellano JL, Reguera Gómez M, Salas-Coronas J, Salvador F, Sotillo Gallego J, Sulleiro E, Torrús Tendero D, Velasco Arribas M, Rodríguez Guardado A. Executive summary consensus statement of imported diseases group (GEPI) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) and the Spanish Society of Tropical Medicine and International Health (SETMSI), on the diagnostic and treatment of imported schistosomiasis. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2023; 41:505-512. [PMID: 37230838 DOI: 10.1016/j.eimce.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Schistosomiasis is a highly prevalent disease, especially in immigrant populations, and is associated with significant morbidity and diagnostic delays outside endemic areas. For these reasons, the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) and the Spanish Society of Tropical Medicine and International Health (SEMTSI) have developed a joint consensus document to serve as a guide for the screening, diagnosis and treatment of this disease outside endemic areas. A panel of experts from both societies identified the main questions to be answered and developed recommendations based on the scientific evidence available at the time. The document was reviewed by the members from both societies for final approval.
Collapse
Affiliation(s)
- Cristina Bocanegra
- Unidad de Medicina Tropical y Salud Internacional Vall d'Hebron-Drassanes, PROSICS Barcelona, Servicio de Enfermedades Infecciosas Hospital Universitario Vall d'Hebron, Spain
| | - Miriam J Álvarez-Martínez
- Servicio de Microbiología, Hospital Clínic de Barcelona, Departamento de Fundamentos Clínicos, Facultad de Medicina y Ciencias de la Salud, Universidad de Barcelona, ISGlobal, Spain
| | - Marta Arsuaga Vicente
- Unidad de Patología Importada y Salud Internacional (CSUR), Unidad de Alto Aislamiento, CIBERINFEC, Hospital La Paz-Carlos III, Madrid, Spain
| | - Moncef Belhassen-García
- Servicio de Medicina Interna, Unidad de Enfermedades Infecciosas, Hospital Universitario de Salamanca, Centro de Investigación en Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Sandra Chamorro Tojeiro
- Unidad de Referencia Nacional para Enfermedades Tropicales, Servicio de Enfermedades Infecciosas, Hospital Universitario Ramón y Cajal, Madrid, IRYCIS, CIBERINFEC, Spain
| | | | - Pedro Fernández Soto
- Grupo Enfermedades Infecciosas y Tropicales (e-INTRO), Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (IBSAL-CIETUS), Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - Elisa García Vázquez
- Unidad de Enfermedades Infecciosas, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB, Facultad de Medicina, Universidad de Murcia, Spain
| | | | - Oihane Martín
- Servicio de Microbiología y Parasitología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Antonio Muro
- Grupo de Investigación Enfermedades Infecciosas y Tropicales (e-INTRO), IBSAL, CIETUS, Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - José Luis Pérez Arellano
- Universidad de las Palmas de Gran Canaria, Unidad de Enfermedades Infecciosas y Medicina Tropical, Hospital Insular de Gran Canaria, Spain
| | - Marta Reguera Gómez
- Departamento de Parasitología, Facultad de Farmacia, Universidad de Valencia, Valencia, Spain
| | - Joaquín Salas-Coronas
- Unidad de Medicina Tropical, Hospital Universitario Poniente El Ejido, Almería, Spain
| | - Fernando Salvador
- Servicio de Enfermedades Infecciosas, Hospital Universitario Vall d'Hebron, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Sotillo Gallego
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Spain
| | - Elena Sulleiro
- Servicio de Microbiología, Hospital Vall d́Hebron, Barcelona, CIBERINFEC, ISCIII, Universitat Autònoma de Barcelona, Spain
| | - Diego Torrús Tendero
- Unidad de Referencia de Enfermedades Importadas y Salud Internacional, Unidad de Enfermedades Infecciosas, Hospital General Universitario Dr. Balmis, ISABIAL, Alicante, Área de Parasitología, Universidad Miguel Hernández, Spain
| | - María Velasco Arribas
- Sección Infecciosas y Medicina Tropical, Medicina Interna, Hospital Universitario Fundación Alcorcón, Universidad Rey Juan Carlos, Madrid, Spain
| | - Azucena Rodríguez Guardado
- Área de Gestión Clínica Medicina Interna, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.
| |
Collapse
|
8
|
Deebani W, Jan R, Shah Z, Vrinceanu N, Racheriu M. Modeling the transmission phenomena of water-borne disease with non-singular and non-local kernel. Comput Methods Biomech Biomed Engin 2023; 26:1294-1307. [PMID: 36006368 DOI: 10.1080/10255842.2022.2114793] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/16/2022] [Accepted: 08/11/2022] [Indexed: 11/03/2022]
Abstract
Drinking or recreating water that has been polluted with disease-causing organisms or pathogens is what causes waterborne infections. It should be noted that many water-borne infections can also transmit from person to person, by contact with animals or their surroundings, or by ingesting tainted food or beverages. Schistosomiasis is a water-borne infection found in different areas of the globe. Mostly people with this viral infection live in Africa with limited resources and medications. Therefore, investigation of this infection is significant to reduce its economic burden on the society. We formulated a novel epidemic model for schistosomiasis water-borne infection with the help of the Atangana-Baleanu derivative. The rudimentary theory of fractional-calculus has been presented for the analysis of our system. We start by looking at the model solution's non-negativity and uniqueness. The basic reproduction number and equilibria of the hypothesized water-borne infection model are next evaluated. Local stability of the infection-free steady-state has been established through Jacobian matrix method for R 0 < 1 . In addition, the suggested model's solution is calculated using an iterative technique. Finally, we give numerical simulations for various input values to illustrate the impact of memory index and other input factors of the system. Our findings showed the influence of input parameters on the dynamical behaviour of the schistosomiasis infection. The results demonstrate the importance and persuasive behaviour of fractional order, and reveal that fractional memory effects in the model seem to be a good fit for this type of findings.
Collapse
Affiliation(s)
- Wejdan Deebani
- Department of Mathematics, College of Science & Arts, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Rashid Jan
- Department of Mathematics, University of Swabi, Swabi, Pakistan
| | - Zahir Shah
- Department of Mathematical Sciences, University of Lakki Marwat, Lakki Marwat, Pakistan
| | - Narcisa Vrinceanu
- Department of Industrial Machines and Equipments, Lucian Blaga University of Sibiu, Sibiu, Romania
| | - Mihaela Racheriu
- Medicine Faculty, Lucian Blaga University of Sibiu, Sibiu, Romania
- Department of Clinical Surgery, Cty Clin Emergency Hosp, Sibiu, Romania
| |
Collapse
|
9
|
N'Goran EK, Odiere MR, Assandé Aka R, Ouattara M, Aka NAD, Ogutu B, Rawago F, Bagchus WM, Bödding M, Kourany-Lefoll E, Tappert A, Yin X, Bezuidenhout D, Badenhorst H, Huber E, Dälken B, Haj-Ali Saflo O. Efficacy, safety, and palatability of arpraziquantel (L-praziquantel) orodispersible tablets in children aged 3 months to 6 years infected with Schistosoma in Côte d'Ivoire and Kenya: an open-label, partly randomised, phase 3 trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:867-876. [PMID: 36893784 DOI: 10.1016/s1473-3099(23)00048-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/15/2022] [Accepted: 01/17/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND WHO has underlined the need for a child-friendly treatment for schistosomiasis, a prevalent parasitic disease in low-income and middle-income countries. After successful phase 1 and 2 trials, we aimed to evaluate the efficacy, safety, palatability, and pharmacokinetics of arpraziquantel (L-praziquantel) orodispersible tablets for preschool-aged children. METHODS This open-label, partly randomised, phase 3 study was conducted at two hospitals in Côte d'Ivoire and Kenya. Children with a minimum bodyweight of 5 kg in those aged 3 months to 2 years and 8 kg in those aged 2-6 years were eligible. In cohort 1, participants aged 4-6 years infected with Schistosoma mansoni were randomly assigned (2:1) to receive a single dose of oral arpraziquantel 50 mg/kg (cohort 1a) or oral praziquantel 40 mg/kg (cohort 1b) using a computer-generated randomisation list. Cohorts 2 (aged 2-3 years) and 3 (aged 3 months to 2 years) infected with S mansoni, and the first 30 participants in cohort 4a (aged 3 months to 6 years) infected with Schistosoma haematobium, received a single dose of oral arpraziquantel 50 mg/kg. After follow-up assessments, arpraziquantel was increased to 60 mg/kg (cohort 4b). Laboratory personnel were masked to the treatment group, screening, and baseline values. S mansoni was detected using a point-of-care circulating cathodic antigen urine cassette test and confirmed using the Kato-Katz method. The primary efficacy endpoint was clinical cure rate at 17-21 days after treatment in cohorts 1a and 1b, measured in the modified intention-to-treat population and calculated using the Clopper-Pearson method. This study is registered with ClinicalTrials.gov, NCT03845140. FINDINGS Between Sept 2, 2019, and Aug 7, 2021, 2663 participants were prescreened and 326 were diagnosed with S mansoni or S haematobium. 288 were enrolled (n=100 in cohort 1a, n=50 in cohort 1b, n=30 in cohort 2, n=18 in cohort 3, n=30 in cohort 4a, and n=60 in cohort 4b), but eight participants received antimalarial drugs and were excluded from the efficacy analyses. The median age was 5·1 years (IQR 4·1-6·0) and 132 (47%) of 280 participants were female and 148 (53%) were male. Cure rates with arpraziquantel were similar to those with praziquantel (87·8% [95% CI 79·6-93·5] in cohort 1a vs 81·3% [67·4-91·1] in cohort 1b). No safety concerns were identified during the study. The most common drug-related treatment-emergent adverse events were abdominal pain (41 [14%] of 288 participants), diarrhoea (27 [9%]), vomiting (16 [6%]), and somnolence (21 [7%]). INTERPRETATION Arpraziquantel, a first-line orodispersible tablet, showed high efficacy and favourable safety in preschool-aged children with schistosomiasis. FUNDING The Global Health Innovative Technology Fund, the European and Developing Countries Clinical Trials Partnership, and the healthcare business of Merck KGaA, Darmstadt, Germany (CrossRef Funder ID: 10.13039/100009945).
Collapse
Affiliation(s)
- Eliézer K N'Goran
- Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire, Kisumu, Kenya
| | - Maurice R Odiere
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | | | - Mamadou Ouattara
- Université Félix Houphouët-Boigny, Abidjan, Côte d'Ivoire, Kisumu, Kenya
| | - N A David Aka
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Bernhards Ogutu
- Kenya Medical Research Institute, Centre for Clinical Research, Nairobi, Kenya
| | - Fredrick Rawago
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Wilhelmina M Bagchus
- Merck Institute of Pharmacometrics, Merck Serono SA (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | | - Elly Kourany-Lefoll
- Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Aliona Tappert
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | - Xiaoyan Yin
- EMD Serono Research and Development Institute Inc (an affiliate of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| | - Deon Bezuidenhout
- Merck Pty Ltd (an affiliate of Merck KGaA, Darmstadt, Germany), Modderfontein, South Africa
| | - Henk Badenhorst
- Merck Pty Ltd (an affiliate of Merck KGaA, Darmstadt, Germany), Modderfontein, South Africa
| | - Eric Huber
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | | | - Okba Haj-Ali Saflo
- Merck SLU (an affiliate of Merck KGaA, Darmstadt, Germany), Madrid, Spain
| |
Collapse
|
10
|
Cherkaoui D, Mesquita SG, Huang D, Lugli EB, Webster BL, McKendry RA. CRISPR-assisted test for Schistosoma haematobium. Sci Rep 2023; 13:4990. [PMID: 36973334 PMCID: PMC10042105 DOI: 10.1038/s41598-023-31238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
Schistosomiasis is a major neglected tropical disease targeted for elimination as a public health issue by 2030, however there is an urgent need for more sensitive and specific diagnostic tests suitable to resource-limited settings. Here we developed CATSH, a CRISPR-assisted diagnostic test for Schistosoma haematobium, utilising recombinase polymerase amplification, Cas12a-targeted cleavage and portable real-time fluorescence detection. CATSH showed high analytical sensitivity, consistent detection of a single parasitic egg and specificity for urogenital Schistosoma species. Thanks to a novel CRISPR-compatible sample preparation developed using simulated urine samples containing parasitic eggs, CATSH had a sample-to-result within 2 h. The components of CATSH can be lyophilised, reducing cold chain dependence and widening access to lower and middle-income countries. This work presents a new application of CRISPR diagnostics for highly sensitive and specific detection of parasitic pathogens in remote areas and could have a significant impact on the elimination of neglected tropical diseases.
Collapse
Affiliation(s)
- Dounia Cherkaoui
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK.
- Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Silvia G Mesquita
- Wolfson Wellcome Biomedical Laboratories, Department of Science, Natural History Museum, Cromwell Road, London, SW7 5BD, UK
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
- London Centre for Neglected Tropical Disease Research (LCNTDR), London, W21 PG, UK
| | - Da Huang
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK
| | - Elena B Lugli
- Wolfson Wellcome Biomedical Laboratories, Department of Science, Natural History Museum, Cromwell Road, London, SW7 5BD, UK
- London Centre for Neglected Tropical Disease Research (LCNTDR), London, W21 PG, UK
| | - Bonnie L Webster
- Wolfson Wellcome Biomedical Laboratories, Department of Science, Natural History Museum, Cromwell Road, London, SW7 5BD, UK.
- London Centre for Neglected Tropical Disease Research (LCNTDR), London, W21 PG, UK.
| | - Rachel A McKendry
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK.
- Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
11
|
Comparative proteomic profiles of Schistosoma japonicum male worms derived from single-sex and bisexual infections. Int J Parasitol 2022; 52:815-828. [PMID: 36265673 DOI: 10.1016/j.ijpara.2022.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022]
Abstract
Schistosomiasis, which is caused by parasitic schistosomes, remains the second most prevalent parasitic disease of mammals worldwide. To successfully maintain fecundity, schistosomes have evolved a lifecycle that involves the cooperation of morphologically distinct male and female forms. Eggs produced by worm pairs are vital to the lifecycle of the parasite and are responsible for pathogenesis. Understanding the reproductive mechanism of schistosomes will help to control infection. In this study, the proteomic profiles of single-sex infected male (SM) worms and bisexual infected mated male (MM) worms of Schistosoma japonicum at 18, 21, 23, and 25 days p.i. were identified through data-independent acquisition. In total, 674 differentially expressed proteins (DEPs) were identified for the SM and MM worms at all four timepoints. Bioinformatic analysis demonstrated that most of the DEPs were involved in biosynthetic processes including locomotion, cell growth and death, cell motility, and metabolic processes such as protein metabolism and glucose metabolism. Schistosoma japonicum glycosyltransferase (SjGT) and S. japonicum nicastrin protein (SjNCSTN) were selected for quantitative real‑time PCR analysis and long-term interference with small interfering RNA (siRNA) to further explore the functions of the DEPs. Sjgt mRNA expression was mainly enriched in male worms, while Sjncstn was enriched in both sexes. siRNA against SjGT and SjNCSTN resulted in minor morphological changes in the testes of male worms and significant decreased vitality and fertility. The present study provides comprehensive proteomic profiles of S. japonicum SM and MM worms at 18, 21, 23, and 25 days p.i. and offers insights into the mechanisms underlying the growth and maturation of schistosomes.
Collapse
|
12
|
Fogarty CE, Suwansa-ard S, Phan P, McManus DP, Duke MG, Wyeth RC, Cummins SF, Wang T. Identification of Putative Neuropeptides That Alter the Behaviour of Schistosoma mansoni Cercariae. BIOLOGY 2022; 11:biology11091344. [PMID: 36138823 PMCID: PMC9495596 DOI: 10.3390/biology11091344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022]
Abstract
Elucidating the infectivity of Schistosoma mansoni, one of the main etiological agents of human schistosomiasis, requires an improved understanding of the behavioural mechanisms of cercariae, the non-feeding mammalian infective stage. This study investigated the presence and effect of cercariae-derived putative neuropeptides on cercarial behaviour when applied externally. Cercariae were peptidomically analysed and 11 neuropeptide precursor proteins, all of which were specific to the Schistosoma genus and most of which highly expressed in the cercarial stage, were identified in cercariae for the first time. Protein–protein interaction analysis predicted the interaction of various neuropeptide precursors (e.g., Sm-npp-30, Sm-npp-33, Sm-npp-35) with cercarial structural proteins (e.g., myosin heavy chain and titin). In total, nine putative neuropeptides, selected based on their high hydrophobicity and small size (~1 kilodalton), were tested on cercariae (3 mg/mL) in acute exposure (1 min) and prolonged exposure (360 min) behavioural bioassays. The peptides AAYMDLPW-NH2, NRKIDQSFYSYY-NH2, FLLALPSP-OH, and NYLWDTRL-NH2 stimulated acute increases in cercarial spinning, stopping, and directional change during active states. However, only NRKIDQSFYSYY-NH2 caused the same behavioural changes at a lower concentration (0.1 mg/mL). After prolonged exposure, AAYMDLPW-NH2 and NYLWDTRL-NH2 caused increasing passive behaviour and NRKIDQSFYSYY-NH2 caused increasing body-first and head-pulling movements. These findings characterise behaviour-altering novel putative neuropeptides, which may inform future biocontrol innovations to prevent human schistosomiasis.
Collapse
Affiliation(s)
- Conor E. Fogarty
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
| | - Saowaros Suwansa-ard
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
| | - Phong Phan
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
| | - Donald P. McManus
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Mary G. Duke
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Russell C. Wyeth
- Department of Biology, St. Francis Xavier University, Antigonish, NS B2G 2W5, Canada
| | - Scott F. Cummins
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4556, Australia
- Correspondence:
| |
Collapse
|
13
|
Marquez J, Dinguirard N, Gonzalez A, Kane A, Joffe N, Yoshino T, Castillo M. Molecular characterization of thioester-containing proteins in Biomphalaria glabrata and their differential gene expression upon Schistosoma mansoni exposure. Front Immunol 2022; 13:903158. [PMID: 35967434 PMCID: PMC9363628 DOI: 10.3389/fimmu.2022.903158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Schistosomiasis is a disease caused by trematode parasites of the genus Schistosoma that affects approximately 200 million people worldwide. Schistosomiasis has been a persistent problem in endemic areas as there is no vaccine available, currently used anti-helmintic medications do not prevent reinfection, and most concerning, drug resistance has been documented in laboratory and field isolates. Thus, alternative approaches to curtail this human disease are warranted. Understanding the immunobiology of the obligate intermediate host of these parasites, which include the freshwater snail Biomphalaria glabrata, may facilitate the development of novel methods to stop or reduce transmission to humans. Molecules from the thioester-containing protein (TEP) superfamily have been shown to be involved in immunological functions in many animals including corals and humans. In this study we identified, characterized, and compared TEP transcripts and their expression upon S. mansoni exposure in resistant and susceptible strains of B. glabrata snails. Results showed the expression of 11 unique TEPs in B. glabrata snails. These transcripts present high sequence identity at the nucleotide and putative amino acid levels between susceptible and resistant strains. Further analysis revealed differences in several TEPs’ constitutive expression levels between resistant and susceptible snail strains, with C3-1, C3-3, and CD109 having higher constitutive expression levels in the resistant (BS90) strain, whereas C3-2 and TEP-1 showed higher constitutive expression levels in the susceptible (NMRI) strain. Furthermore, TEP-specific response to S. mansoni miracidia exposure reiterated their differential expression, with resistant snails upregulating the expression of both TEP-4 and TEP-3 at 2 h and 48 h post-exposure, respectively. Further understanding the diverse TEP genes and their functions in invertebrate animal vectors will not only expand our knowledge in regard to this ancient family of immune proteins, but also offer the opportunity to identify novel molecular targets that could aid in the efforts to develop control methods to reduce schistosomiasis transmission.
Collapse
Affiliation(s)
- J. Marquez
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - N. Dinguirard
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - A. Gonzalez
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - A.E. Kane
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - N.R. Joffe
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - T.P. Yoshino
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - M.G. Castillo
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
- *Correspondence: M.G. Castillo,
| |
Collapse
|
14
|
Kadaleka S, Abelman S, Tchuenche JM. A Mathematical Model of the Transmission Dynamics of Bovine Schistosomiasis with Contaminated Environment. Acta Biotheor 2022; 70:9. [PMID: 35020068 DOI: 10.1007/s10441-021-09434-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/28/2021] [Indexed: 11/01/2022]
Abstract
Schistosomiasis, a vector-borne chronically debilitating infectious disease, is a serious public health concern for humans and animals in the affected tropical and sub-tropical regions. We formulate and theoretically analyze a deterministic mathematical model with snail and bovine hosts. The basic reproduction number [Formula: see text] is computed and used to investigate the local stability of the model's steady states. Global stability of the endemic equilibrium is carried out by constructing a suitable Lyapunov function. Sensitivity analysis shows that the basic reproduction number is most sensitive to the model parameters related to the contaminated environment, namely: shedding rate of cercariae by snails, cercariae to miracidia survival probability, snails-miracidia effective contact rate and natural death rate of miracidia and cercariae. Numerical results show that when no intervention measures are implemented, there is an increase of the infected classes, and a rapid decline of the number of susceptible and exposed bovines and snails. Effects of the variation of some of the key sensitive model parameters on the schistosomiasis dynamics as well as on the initial disease transmission threshold parameter [Formula: see text] are graphically depicted.
Collapse
|
15
|
Comparative characterization of microRNAs of Schistosoma japonicum from SCID mice and BALB/c mice: Clues to the regulation of parasite growth and development. Acta Trop 2022; 225:106200. [PMID: 34740636 DOI: 10.1016/j.actatropica.2021.106200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
Schistosomiasis, caused by a parasite with a wide range of mammalian hosts, remains one of the most prevailing parasitic diseases in the world. While numerous studies have reported that the growth and reproduction of schistosomes in immunodeficient mice was significantly retarded, the underlying molecular mechanisms have yet to be revealed. In this study, we comparatively analyzed the microRNA expression of Schistosoma japonicum derived from SCID and BALB/c mice on the 35th day post-infection by high-throughput RNA sequencing as prominent morphological abnormalities had been observed in schistosomes from SCID mice when compared with those from BALB/c mice. The results revealed that more than 72% and 61% of clean reads in the small RNA libraries of female and male schistosomes, respectively, could be mapped to the selected miRs in the miRBase or the sequences of species-specific genomes. Further analysis identified 122 miRNAs using TPM >0.01 as the threshold value, including 75 known and 47 novel miRNAs, 96 of which were commonly expressed across all the four tested schistosome libraries. Comparative analysis of the libraries of schistosomes from SCID and BALB/c mice identified 15 differentially expressed miRNAs (5 up-regulated and 10 down-regulated) among females and 16 among males (9 up-regulated and 7 down-regulated). Integrated analysis of the two sets of differentially expressed miRNAs of female and male worms identified 2 miRNAs (sja-miR-3488 and sja-miR-novel_29) that overlapped between female and male datasets. Prediction of miRNA targets and Gene Ontology (GO) term enrichment analysis of the predicted target genes revealed that these genes were involved in some important biological processes, such as nucleic acid metabolic process, macromolecule modification, and cellular aromatic compound metabolic process. The predicted target genes were further matched to the differentially expressed genes in male and female schistosomes from the above two hosts, obtaining 7 genes that may be responsible for regulating the growth, development and sex maturation of schistosomes. Taken together, this study provides the first identification of differentially expressed miRNAs in schistosomes from SCID and BALB/c mice. These miRNAs and their predicted target mRNAs are probably involved in the regulation of development, growth, and maturation of schistosomes. Therefore, this study expands our understanding of schistosome development regulation and host-parasite relationship, and also provides a valuable set of potential anti-schistosomal targets for prevention and control of schistosomiasis.
Collapse
|
16
|
Drug associations as alternative and complementary therapy for neglected tropical diseases. Acta Trop 2022; 225:106210. [PMID: 34687644 DOI: 10.1016/j.actatropica.2021.106210] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/02/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022]
Abstract
The present paper aims to establish different treatments for neglected tropical disease by a survey on drug conjugations and possible fixed-dose combinations (FDC) used to obtain alternative, safer and more effective treatments. The source databases used were Science Direct and PubMed/Medline, in the intervals between 2015 and 2021 with the drugs key-words or diseases, like "schistosomiasis", "praziquantel", "malaria", "artesunate", "Chagas' disease", "benznidazole", "filariasis", diethylcarbamazine", "ivermectin", " albendazole". 118 works were the object of intense analysis, other articles and documents were used to increase the quality of the studies, such as consensuses for harmonizing therapeutics and historical articles. As a result, an effective NTD control can be achieved when different public health approaches are combined with interventions guided by the epidemiology of each location and the availability of appropriate measures to detect, prevent and control disease. It was also possible to verify that the FDCs promote a simplification of the therapeutic regimen, which promotes better patient compliance and enables a reduction in the development of parasitic resistance, requiring further studies aimed at resistant strains, since the combined APIs usually act by different mechanisms or at different target sites. In addition to eliminating the process of developing a new drug based on the identification and validation of active compounds, which is a complex, long process and requires a strong long-term investment, other advantages that FDCs have are related to productive gain and gain from the industrial plant, which can favor and encourage the R&D of new FDCs not only for NTDs but also for other diseases that require the use of more than one drug.
Collapse
|
17
|
Hailegebriel T, Nibret E, Munshea A. Efficacy of Praziquantel for the Treatment of Human Schistosomiasis in Ethiopia: A Systematic Review and Meta-Analysis. J Trop Med 2021; 2021:2625255. [PMID: 34966433 PMCID: PMC8712180 DOI: 10.1155/2021/2625255] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/04/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Schistosomiasis is one of the neglected tropical diseases causing a serious human health problem in Ethiopia. Praziquantel is the only drug that has been used for the treatment of human schistosomiasis in the country. In line with this, the efficacy of praziquantel has been evaluated in a few interventional studies in the country, but there is a lack in systematically gathered and analyzed information for policymakers. The aim of this systematic review and meta-analysis was to provide a summary of the efficacy of praziquantel for the treatment of human schistosomiasis in Ethiopia. METHODS We conducted a literature search from ScienceDirect, PubMed/Medlin, and Google Scholar databases. A total of 140 articles published in English from 1980 to June 2021 were accessed and 15 of them were eligible for this meta-analysis. The meta-analysis was conducted using Stata 14 software, "metan command." The heterogeneities among studies were evaluated using I 2 test. RESULTS A total of 140 articles were reviewed, but only 15 of them fulfilled the inclusion criteria. The polled cure rate of 40 mg/kg praziquantel was 89.2% (95% CI: 85.4-93.1) and 93.6% (95% CI: 80.6-106) among Schistosoma mansoni and S. haematobium, respectively. Similarly, the mean egg reduction rates of 40 mg/kg praziquantel were 90.2% and 85% among S. mansoni and S. haematobium infected subjects, respectively. The common adverse events observed after receiving praziquantel include abdominal pain, vomiting, headache, diarrhea, and bloody stool. CONCLUSION This systematic review and meta-analysis has indicated that praziquantel is still an appropriate drug for the treatment of human schistosomiasis in Ethiopia.
Collapse
Affiliation(s)
- Tamirat Hailegebriel
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
- Biotechnology Research Institute (BRI), Bahir Dar University, Bahir Dar, Ethiopia
| | - Endalkachew Nibret
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
- Biotechnology Research Institute (BRI), Bahir Dar University, Bahir Dar, Ethiopia
| | - Abaineh Munshea
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
- Biotechnology Research Institute (BRI), Bahir Dar University, Bahir Dar, Ethiopia
| |
Collapse
|
18
|
From the One Health Perspective: Schistosomiasis Japonica and Flooding. Pathogens 2021; 10:pathogens10121538. [PMID: 34959493 PMCID: PMC8709050 DOI: 10.3390/pathogens10121538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 01/09/2023] Open
Abstract
Schistosomiasis is a water-borne parasitic disease distributed worldwide, while schistosomiasis japonica localizes in the People’s Republic of China, the Philippines, and a few regions of Indonesia. Although significant achievements have been obtained in these endemic countries, great challenges still exist to reach the elimination of schistosomiasis japonica, as the occurrence of flooding can lead to several adverse consequences on the prevalence of schistosomiasis. This review summarizes the influence of flooding on the transmission of schistosomiasis japonica and interventions responding to the adverse impacts from the One Health perspective in human beings, animals, and the environment. For human and animals, behavioral changes and the damage of water conservancy and sanitary facilities will increase the intensity of water contact. For the environment, the density of Oncomelania snails significantly increases from the third year after flooding, and the snail habitats can be enlarged due to active and passive diffusion. With more water contact of human and other reservoir hosts, and larger snail habitats with higher density of living snails, the transmission risk of schistosomiasis increases under the influence of flooding. With the agenda set for global schistosomiasis elimination, interventions from the One Health perspective are put forward to respond to the impacts of increased flooding. For human beings, conducting health education to increase the consciousness of self-protection, preventive chemotherapy for high-risk populations, supply of safe water, early case finding, timely reporting, and treating cases will protect people from infection and prevent the outbreak of schistosomiasis. For animals, culling susceptible domestic animals, herding livestock in snail-free areas, treating livestock with infection or at high risk of infection, harmless treatment of animal feces to avoid water contamination, and monitoring the infection status of wild animals in flooding areas are important to cut off the transmission chain from the resources. For the environment, early warning of flooding, setting up warning signs and killing cercaria in risk areas during and post flooding, reconstructing damaged water conservancy facilities, developing hygiene and sanitary facilities, conducting snail surveys, using molluscicide, and predicting areas with high risk of schistosomiasis transmission after flooding all contribute to reducing the transmission risk of schistosomiasis. These strategies need the cooperation of the ministry of health, meteorological administration, water resources, agriculture, and forestry to achieve the goal of minimizing the impact of flooding on the transmission of schistosomiasis. In conclusion, flooding is one of the important factors affecting the transmission of schistosomiasis japonica. Multi-sectoral cooperation is needed to effectively prevent and control the adverse impacts of flooding on human beings, animals, and the environment.
Collapse
|
19
|
Lund AJ, Sokolow SH, Jones IJ, Wood CL, Ali S, Chamberlin A, Sy AB, Sam MM, Jouanard N, Schacht AM, Senghor S, Fall A, Ndione R, Riveau G, De Leo GA, López-Carr D. Exposure, hazard, and vulnerability all contribute to Schistosoma haematobium re-infection in northern Senegal. PLoS Negl Trop Dis 2021; 15:e0009806. [PMID: 34610025 PMCID: PMC8525765 DOI: 10.1371/journal.pntd.0009806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 10/19/2021] [Accepted: 09/10/2021] [Indexed: 11/19/2022] Open
Abstract
Background Infectious disease risk is driven by three interrelated components: exposure, hazard, and vulnerability. For schistosomiasis, exposure occurs through contact with water, which is often tied to daily activities. Water contact, however, does not imply risk unless the environmental hazard of snails and parasites is also present in the water. By increasing reliance on hazardous activities and environments, socio-economic vulnerability can hinder reductions in exposure to a hazard. We aimed to quantify the contributions of exposure, hazard, and vulnerability to the presence and intensity of Schistosoma haematobium re-infection. Methodology/Principal findings In 13 villages along the Senegal River, we collected parasitological data from 821 school-aged children, survey data from 411 households where those children resided, and ecological data from all 24 village water access sites. We fit mixed-effects logistic and negative binomial regressions with indices of exposure, hazard, and vulnerability as explanatory variables of Schistosoma haematobium presence and intensity, respectively, controlling for demographic variables. Using multi-model inference to calculate the relative importance of each component of risk, we found that hazard (Ʃwi = 0.95) was the most important component of S. haematobium presence, followed by vulnerability (Ʃwi = 0.91). Exposure (Ʃwi = 1.00) was the most important component of S. haematobium intensity, followed by hazard (Ʃwi = 0.77). Model averaging quantified associations between each infection outcome and indices of exposure, hazard, and vulnerability, revealing a positive association between hazard and infection presence (OR = 1.49, 95% CI 1.12, 1.97), and a positive association between exposure and infection intensity (RR 2.59–3.86, depending on the category; all 95% CIs above 1) Conclusions/Significance Our findings underscore the linkages between social (exposure and vulnerability) and environmental (hazard) processes in the acquisition and accumulation of S. haematobium infection. This approach highlights the importance of implementing both social and environmental interventions to complement mass drug administration. While the impacts of natural hazards tend to be described in terms of social determinants such as exposure and vulnerability, the risk for infectious disease is often expressed in terms of environmental determinants without fully considering the socio-ecological processes that put people in contact with infective agents of disease. In the case of schistosomiasis, risk is determined by human interactions with freshwater environments where schistosome parasites circulate between people and aquatic snails. In this study, we quantified the relative contributions of exposure, hazard, and vulnerability to schistosome re-infection among schoolchildren in an endemic region of northern Senegal. We find that hazard and vulnerability influence whether a child becomes infected, while exposure and hazard influence the burden of worms once infection is acquired. Increasing numbers of worms is known to be positively associated with increasing severity of disease. Our findings underscore the importance of evaluating social and environmental determinants of disease simultaneously; omitting measures of exposure, hazard or vulnerability may limit our understanding of risk.
Collapse
Affiliation(s)
- Andrea J. Lund
- Emmett Interdisciplinary Program in Environment and Resources, Stanford University, Stanford, California, United States of America
- * E-mail:
| | - Susanne H. Sokolow
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America
- Woods Institute for the Environment, Stanford University, Stanford, California, United States of America
| | - Isabel J. Jones
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America
| | - Chelsea L. Wood
- School of Aquatic and Fishery Sciences, University of Washington, Seattle, Washington, United States of America
| | - Sofia Ali
- Stanford University, Stanford, California, United States of America
| | - Andrew Chamberlin
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America
| | - Alioune Badara Sy
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
| | - M. Moustapha Sam
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
| | - Nicolas Jouanard
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
- Station d’Innovation Aquacole, Saint Louis, Sénégal
| | - Anne-Marie Schacht
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
- University of Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Simon Senghor
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
| | - Assane Fall
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
| | - Raphael Ndione
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
| | - Gilles Riveau
- Centre de Recherche Biomédicale–Espoir Pour La Sante, Saint Louis, Sénégal
- University of Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Giulio A. De Leo
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America
- Woods Institute for the Environment, Stanford University, Stanford, California, United States of America
| | - David López-Carr
- Department of Geography, University of California, Santa Barbara, CA, United States of America
| |
Collapse
|
20
|
Jones IJ, Sokolow SH, Chamberlin AJ, Lund AJ, Jouanard N, Bandagny L, Ndione R, Senghor S, Schacht AM, Riveau G, Hopkins SR, Rohr JR, Remais JV, Lafferty KD, Kuris AM, Wood CL, De Leo G. Schistosome infection in Senegal is associated with different spatial extents of risk and ecological drivers for Schistosoma haematobium and S. mansoni. PLoS Negl Trop Dis 2021; 15:e0009712. [PMID: 34570777 PMCID: PMC8476036 DOI: 10.1371/journal.pntd.0009712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022] Open
Abstract
Schistosome parasites infect more than 200 million people annually, mostly in sub-Saharan Africa, where people may be co-infected with more than one species of the parasite. Infection risk for any single species is determined, in part, by the distribution of its obligate intermediate host snail. As the World Health Organization reprioritizes snail control to reduce the global burden of schistosomiasis, there is renewed importance in knowing when and where to target those efforts, which could vary by schistosome species. This study estimates factors associated with schistosomiasis risk in 16 villages located in the Senegal River Basin, a region hyperendemic for Schistosoma haematobium and S. mansoni. We first analyzed the spatial distributions of the two schistosomes’ intermediate host snails (Bulinus spp. and Biomphalaria pfeifferi, respectively) at village water access sites. Then, we separately evaluated the relationships between human S. haematobium and S. mansoni infections and (i) the area of remotely-sensed snail habitat across spatial extents ranging from 1 to 120 m from shorelines, and (ii) water access site size and shape characteristics. We compared the influence of snail habitat across spatial extents because, while snail sampling is traditionally done near shorelines, we hypothesized that snails further from shore also contribute to infection risk. We found that, controlling for demographic variables, human risk for S. haematobium infection was positively correlated with snail habitat when snail habitat was measured over a much greater radius from shore (45 m to 120 m) than usual. S. haematobium risk was also associated with large, open water access sites. However, S. mansoni infection risk was associated with small, sheltered water access sites, and was not positively correlated with snail habitat at any spatial sampling radius. Our findings highlight the need to consider different ecological and environmental factors driving the transmission of each schistosome species in co-endemic landscapes. Schistosome parasites infect more than 200 million people worldwide, mainly in sub-Saharan Africa, where many people are at-risk for infection by multiple schistosome species simultaneously. To reduce the global burden of schistosomiasis, control of the parasites’ intermediate host–specific species of freshwater snails–has been elevated in priority to complement mass drug administration campaigns in endemic areas. To maximize the efficacy and efficiency of snail control efforts, a better understanding of where to target intermediate host snails is badly needed. This includes a better understanding of the spatial scale at which snails in the environment contribute to human infection risk, and, in co-endemic settings, how ecological determinants of infection risk vary by schistosome species. We used quantitative snail sampling and remotely-sensed data at 16 villages in the Senegal River Basin to compare and contrast ecological correlates and spatial scales of infection risk from freshwater snails that transmit Schistosoma haematobium versus S. mansoni. We found that infection risk for S. haematobium was associated with snail habitat at a larger spatial radius than is typically considered for schistosomiasis monitoring and control, whereas infection risk for S. mansoni was not positively correlated with snail habitat at any spatial sampling radius, but was associated with small water access sites enclosed by emergent vegetation. Our findings highlight the need to consider the different ecological and environmental factors driving the transmission of each schistosome species in co-endemic landscapes.
Collapse
Affiliation(s)
- Isabel J Jones
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America
| | - Susanne H Sokolow
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America.,Stanford Woods Institute for the Environment, Stanford University, Stanford, California, United States of America
| | - Andrew J Chamberlin
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America
| | - Andrea J Lund
- Emmett Interdisciplinary Program in Environment and Resources, Stanford University, Stanford, California, United States of America
| | - Nicolas Jouanard
- Biomedical Research Center EPLS, Saint-Louis, Senegal.,Station d'Innovation Aquacole, Saint-Louis, Senegal
| | | | | | - Simon Senghor
- Biomedical Research Center EPLS, Saint-Louis, Senegal
| | - Anne-Marie Schacht
- Biomedical Research Center EPLS, Saint-Louis, Senegal.,Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Gilles Riveau
- Biomedical Research Center EPLS, Saint-Louis, Senegal.,Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Skylar R Hopkins
- National Center for Ecological Analysis and Synthesis, Santa Barbara, California, United States of America.,Department of Applied Ecology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jason R Rohr
- Department of Biological Science, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Justin V Remais
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Kevin D Lafferty
- Western Ecological Research Center, United States Geological Survey at Marine Science Institute, University of California, Santa Barbara, California, United States of America
| | - Armand M Kuris
- Department of Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, California, United States of America
| | - Chelsea L Wood
- School of Aquatic and Fishery Sciences, University of Washington, Seattle, Washington, United States of America
| | - Giulio De Leo
- Hopkins Marine Station, Stanford University, Pacific Grove, California, United States of America.,Stanford Woods Institute for the Environment, Stanford University, Stanford, California, United States of America
| |
Collapse
|
21
|
Cheuka PM. Drug Discovery and Target Identification against Schistosomiasis: a Reality Check on Progress and Future Prospects. Curr Top Med Chem 2021; 22:1595-1610. [PMID: 34565320 DOI: 10.2174/1568026621666210924101805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
Schistosomiasis ranks among the most important infectious diseases, with over 200 million people currently being infected and > 280,000 deaths reported annually. Chemotherapeutic treatment has relied on one drug, praziquantel, for four decades, while other drugs, such as oxamniquine and metrifonate, are no longer preferred for clinical use due to their narrow spectrum of activity - these are only active against S. mansoni and S. haematobium, respectively. Despite being cheap, safe, and effective against all schistosome species, praziquantel is ineffective against immature worms, which may lead to reinfections and treatment failure in endemic areas; a situation that necessitates repeated administration besides other limitations. Therefore, novel drugs are urgently needed to overcome this situation. In this paper, an up to date review of drug targets identified and validated against schistosomiasis while also encompassing promising clinical and preclinical candidate drugs is presented. While there are considerable efforts aimed at identifying and validating drug targets, the pipeline for new antischistosomals is dry. Moreover, the majority of compounds evaluated preclinically are not really advanced because most of them were evaluated in very small preclinical species such as mice alone. Overall, it appears that although a lot of research is going on at discovery phases, unfortunately, it does not translate to advanced preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Peter Mubanga Cheuka
- Department of Chemistry, School of Natural Sciences, University of Zambia, Lusaka. Zambia
| |
Collapse
|
22
|
Ma Y, Xu D, Yan X, Wu Z, Kayani SI, Shen Q, Fu X, Xie L, Hao X, Hassani D, Li L, Liu H, Pan Q, Lv Z, Liu P, Sun X, Tang K. Jasmonate- and abscisic acid-activated AaGSW1-AaTCP15/AaORA transcriptional cascade promotes artemisinin biosynthesis in Artemisia annua. PLANT BIOTECHNOLOGY JOURNAL 2021; 19:1412-1428. [PMID: 33539631 PMCID: PMC8313134 DOI: 10.1111/pbi.13561] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 05/12/2023]
Abstract
Artemisinin, a sesquiterpene lactone widely used in malaria treatment, was discovered in the medicinal plant Artemisia annua. The biosynthesis of artemisinin is efficiently regulated by jasmonate (JA) and abscisic acid (ABA) via regulatory factors. However, the mechanisms linking JA and ABA signalling with artemisinin biosynthesis through an associated regulatory network of downstream transcription factors (TFs) remain enigmatic. Here we report AaTCP15, a JA and ABA dual-responsive teosinte branched1/cycloidea/proliferating (TCP) TF, which is essential for JA and ABA-induced artemisinin biosynthesis by directly binding to and activating the promoters of DBR2 and ALDH1, two genes encoding enzymes for artemisinin biosynthesis. Furthermore, AaORA, another positive regulator of artemisinin biosynthesis responds to JA and ABA, interacts with and enhances the transactivation activity of AaTCP15 and simultaneously activates AaTCP15 transcripts. Hence, they form an AaORA-AaTCP15 module to synergistically activate DBR2, a crucial gene for artemisinin biosynthesis. More importantly, AaTCP15 expression is activated by the multiple reported JA and ABA-responsive TFs that promote artemisinin biosynthesis. Among them, AaGSW1 acts at the nexus of JA and ABA signalling to activate the artemisinin biosynthetic pathway and directly binds to and activates the AaTCP15 promoter apart from the AaORA promoter, which further facilitates formation of the AaGSW1-AaTCP15/AaORA regulatory module to integrate JA and ABA-mediated artemisinin biosynthesis. Our results establish a multilayer regulatory network of the AaGSW1-AaTCP15/AaORA module to regulate artemisinin biosynthesis through JA and ABA signalling, and provide an interesting avenue for future research exploring the special transcriptional regulation module of TCP genes associated with specialized metabolites in plants.
Collapse
Affiliation(s)
- Ya‐Nan Ma
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Dong‐Bei Xu
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
- Institute of Ecological AgricultureSichuan Agricultural UniversityChengduChina
| | - Xin Yan
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Zhang‐Kuanyu Wu
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Sadaf Ilyas Kayani
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Qian Shen
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Xue‐Qing Fu
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Li‐Hui Xie
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Xiao‐Long Hao
- Laboratory of Medicinal Plant BiotechnologyCollege of PharmacyZhejiang Chinese Medical UniversityHangzhouChina
| | - Danial Hassani
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Ling Li
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Hang Liu
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Qi‐Fang Pan
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Zong‐You Lv
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Pin Liu
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Xiao‐Fen Sun
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| | - Ke‐Xuan Tang
- Joint International Research Laboratory of Metabolic and Developmental SciencesKey Laboratory of Urban Agriculture (South) Ministry of AgriculturePlant Biotechnology Research CenterFudan‐SJTU‐Nottingham Plant Biotechnology R&D CenterSchool of Agriculture and BiologyShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
23
|
Roucher C, Brosius I, Mbow M, Faye BT, De Hondt A, Smekens B, Arango D, Burm C, Tsoumanis A, Paredis L, van Herrewege Y, Potters I, Cisse B, Mboup S, Polman K, Bottieau E. Evaluation of Artesunate-mefloquine as a Novel Alternative Treatment for Schistosomiasis in African Children (SchistoSAM): protocol of a proof-of-concept, open-label, two-arm, individually-randomised controlled trial. BMJ Open 2021; 11:e047147. [PMID: 34168029 PMCID: PMC8231067 DOI: 10.1136/bmjopen-2020-047147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
INTRODUCTION Alternative drugs and diagnostics are needed for the treatment and control of schistosomiasis. The exclusive use of praziquantel (PZQ) in mass drug administration programmes may result in the emergence of drug resistance. PZQ has little activity against Schistosoma larvae, thus reinfection remains a problem in high-risk communities. Furthermore, the insufficient sensitivity of conventional microscopy hinders therapeutic response assessment. Evaluation of artesunate-mefloquine (AM) as a Novel Alternative Treatment for Schistosomiasis in African Children (SchistoSAM) aims to evaluate the safety and efficacy of the antimalarial combination artesunate-mefloquine, re-purposed for the treatment of schistosomiasis, and to assess the performance of highly sensitive novel antigen-based and DNA-based assays as tools for monitoring treatment response. METHODS AND ANALYSIS The SchistoSAM study is an open-label, two-arm, individually randomised controlled non-inferiority trial, with a follow-up of 48 weeks. Primary school-aged children from the Richard Toll district in northern Senegal, an area endemic for Schistosoma mansoni and Schistosoma haematobium, are allocated to the AM intervention arm (3-day courses at 6-week intervals) or the PZQ control arm (single dose of 40 mg/kg). The trial's primary endpoints are the efficacy (cure rate (CR), assessed by microscopy) and safety (frequency and pattern of drug-related adverse events) of one AM course versus PZQ at 4 weeks after treatment. Secondary endpoints include (1) cumulative CR, egg reduction rate and safety after each additional course of AM, and at weeks 24 and 48, (2) prevalence and severity of schistosomiasis-related morbidity and (3) malaria prevalence, incidence and morbidity, both after 24 and 48 weeks. CRs and intensity reduction rates are also assessed by antigen-based and DNA-based diagnostic assays, for which performance for treatment monitoring is evaluated. ETHICS AND DISSEMINATION Ethics approval was obtained both in Belgium and Senegal. Oral assent from the children and signed informed consent from their legal representatives was obtained, prior to enrolment. The results will be disseminated in peer-reviewed journals and at international conferences. TRIAL REGISTRATION NUMBER NCT03893097; pre-results.
Collapse
Affiliation(s)
- Clémentine Roucher
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Isabel Brosius
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Moustapha Mbow
- Department of Immunology, Cheikh Anta Diop University, Dakar, Senegal
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | | | - Annelies De Hondt
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Bart Smekens
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Diana Arango
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Christophe Burm
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Achilleas Tsoumanis
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Linda Paredis
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Yven van Herrewege
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Idzi Potters
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| | - Badara Cisse
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Souleymane Mboup
- Institute for Health Research, Epidemiological Surveillance and Training (IRESSEF), Dakar, Senegal
| | - Katja Polman
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Emmanuel Bottieau
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerpen, Belgium
| |
Collapse
|
24
|
Minzi OM, Mnkugwe RH, Ngaimisi E, Kinung’hi S, Hansson A, Pohanka A, Kamuhabwa A, Aklillu E. Effect of Dihydroartemisinin-Piperaquine on the Pharmacokinetics of Praziquantel for Treatment of Schistosoma mansoni Infection. Pharmaceuticals (Basel) 2021; 14:ph14050400. [PMID: 33922522 PMCID: PMC8145331 DOI: 10.3390/ph14050400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Praziquantel (PZQ) and dihydroartemisinin-piperaquine (DHP) combination recently showed superior effectiveness than PZQ alone to treat intestinal schistosomiasis. In this follow-up study, we investigated the effect of DHP co-administration on the pharmacokinetics of PZQ and its enantiomers among 64 Schistosoma mansoni infected children treated with PZQ alone (n = 32) or PZQ + DHP combination (n = 32). Plasma samples collected at 0, 1, 2, 4, 6, and 8 h post-dose were quantified using UPLCMS/MS. The geometric mean (GM) of AUCs for total PZQ, R-PZQ and S-PZQ were significantly higher among children who received PZQ + DHP than PZQ alone. The geometric mean ratio (GMR) and (90% CI) of AUC0–∞ for PZQ + DHP to PZQ for total PZQ, R-PZQ, and S-PZQ were 2.18 (1.27, 3.76), 3.98 (2.27, 7.0) and 1.86 (1.06, 3.28), respectively. The GMR and (90% CI) of AUC0–8 for total PZQ, R-PZQ, and S-PZQ were 1.73 (1.12, 2.69), 2.94 (1.75, 4.92), and 1.50 (0.97, 2.31), respectively. The GM of Cmax for total PZQ, R-PZQ and S-PZQ were significantly higher among those who received PZQ + DHP than PZQ alone. The GMR (90% CI) of Cmax of PZQ + DHP to PZQ for total PZQ, R-PZQ, and S-PZQ were 1.75 (1.15, 2.65), 3.08 (1.91, 4.96), and 1.50 (1.0, 2.25%), respectively. The 90% CI of the GMRs for both AUCs and Cmax for total PZQ, R-PZQ, and S-PZQ were outside the acceptable 0.80–1.25 range, indicating that the two treatment arms were not bioequivalent. DHP co-administration significantly increases systemic PZQ exposure, and this may contribute to increased effectiveness of PZQ + DHP combination therapy than PZQ alone to treat schistosomiasis.
Collapse
Affiliation(s)
- Omary Mashiku Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, 11103 Dar es Salaam, Tanzania; (O.M.M.); (A.K.)
| | - Rajabu Hussein Mnkugwe
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden; (R.H.M.); (A.P.)
- Department of Clinical Pharmacology, School of Medicine, Muhimbili University of Health and Allied Sciences, 11103 Dar es Salaam, Tanzania
| | - Eliford Ngaimisi
- Office of Clinical Pharmacology, Division of Pharmacometrics, Food and Drugs Administration, Silver Spring, MD 20993, USA;
| | - Safari Kinung’hi
- National Institute for Medical Research, Mwanza Research Centre, 33104 Mwanza, Tanzania;
| | - Anna Hansson
- Department of Clinical Pharmacology, Karolinska University Hospital-Huddinge, 141 86 Stockholm, Sweden;
| | - Anton Pohanka
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden; (R.H.M.); (A.P.)
- Department of Clinical Pharmacology, Karolinska University Hospital-Huddinge, 141 86 Stockholm, Sweden;
| | - Appolinary Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, 11103 Dar es Salaam, Tanzania; (O.M.M.); (A.K.)
| | - Eleni Aklillu
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden; (R.H.M.); (A.P.)
- Correspondence: ; Tel.: +46-73-511-6131
| |
Collapse
|
25
|
Fukushige M, Chase-Topping M, Woolhouse MEJ, Mutapi F. Efficacy of praziquantel has been maintained over four decades (from 1977 to 2018): A systematic review and meta-analysis of factors influence its efficacy. PLoS Negl Trop Dis 2021; 15:e0009189. [PMID: 33730095 PMCID: PMC7968639 DOI: 10.1371/journal.pntd.0009189] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/01/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The antihelminthic drug praziquantel has been used as the drug of choice for treating schistosome infection for more than 40 years. Although some epidemiological studies have reported low praziquantel efficacy in cure rate (CR) and/or egg reduction rate (ERR), there is no consistent robust evidence of the development of schistosome resistance to praziquantel (PZQ). There is need to determine factors that lead to variable treatment CR and/or ERR. Therefore, we conducted a systematic review and meta-analysis to review CR and ERR as well as identify their predictors. METHODOLOGY/PRINCIPAL FINDINGS In this systematic review and meta-analysis, a literature review was conducted using Biosis Citation Index, Data Citation Index, MEDLINE, and Web of Science Core Collection all of which were provided through Web of Science. Alongside these, EMBASE, and CAB abstracts were searched to identify relevant articles. Random effect meta-regression models were used to identify the factors that influence CR and/or ERR by considering differences in host characteristics and drug dose. In total, 12,127 potential articles were screened and 146 eligible articles (published from 1979 to 2020) were identified and included for the meta-analysis. We found that there has been no significant reduction in CR or ERR over the study period. The results showed more variability in CR, compared with ERR which was more consistent and remained high. The results showed a positive effect of "PZQ treatment dose" with the current recommended dose of 40 mg/kg body weight achieving 57% to 88% CR depending on schistosome species, age of participants, and number of parasitological samples used for diagnosis, and ERR of 95%. CONCLUSIONS/SIGNIFICANCE Based on a review of over 40 years of research there is no evidence to support concerns about schistosomes developing resistance to PZQ. These results indicate that PZQ remains effective in treating schistosomiasis.
Collapse
Affiliation(s)
- Mizuho Fukushige
- Institute of Immunology and Infection Research, Centre for Immunity, Infection & Evolution, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Margo Chase-Topping
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Francisca Mutapi
- Institute of Immunology and Infection Research, Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
26
|
Mnkugwe RH, Minzi O, Kinung’hi S, Kamuhabwa A, Aklillu E. Efficacy and safety of praziquantel and dihydroartemisinin piperaquine combination for treatment and control of intestinal schistosomiasis: A randomized, non-inferiority clinical trial. PLoS Negl Trop Dis 2020; 14:e0008619. [PMID: 32966290 PMCID: PMC7510991 DOI: 10.1371/journal.pntd.0008619] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 07/20/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Despite the reported success in reducing morbidity, praziquantel alone is insufficient for the control and elimination of schistosomiasis, partly due to its poor efficacy against the juvenile worms. Artemisinin derivatives are effective against juvenile worms but are less effective against adult worms. We compared the safety and efficacy of praziquantel and Dihydroartemisinin-piperaquine combination against the standard praziquantel alone for treatment of intestinal schistosomiasis. METHODS In this randomized, open-label, non-inferiority trial, 639 Schistosoma mansoni infected children were enrolled and randomized to receive either praziquantel alone or praziquantel plus Dihydroartemisinin-piperaquine combination. Two stool samples were collected on consecutive days at baseline, 3 and 8 weeks post-treatment and analyzed using thick smear Kato Katz method. Efficacy was assessed by cure and egg reduction rates at 3 and 8 weeks post-treatment. Adverse events were assessed within four hours of drugs intake. The primary outcome was cure rates at 8 weeks of post-treatment. Secondary outcomes were egg reduction rates at 8 weeks of post-treatment and treatment-associated adverse events. RESULTS At 3 weeks of post-treatment, cure rates were 88.3% (263/298, 95% CI = 84.1%- 91.4%) and 81.2% (277/341, 95% CI = 76.7%- 85.0%) for the combination therapy and praziquantel alone, respectively (p < 0.01, odds ratio (OR) = 1.74, 95% CI of OR = 1.11 to 2.69). At 8 weeks, there was a significant drop in the cure rates in praziquantel alone group to 63.9% (218/341, 95% CI = 58.7%- 68.8%) compared to 81.9% (244/298, 95% CI = 77.1%- 85.8%) in the combination therapy group (p < 0.0001, OR = 2.55, 95%CI of OR = 1.75 to 3.69). Egg reduction rates at 8 weeks post-treatment were significantly higher in the combination therapy group 93.6% (95% CI = 90.8%- 96.4%) compared to 87.9% (95% CI = 84.4%- 91.4%) in the praziquantel only group (p = 0.01). On both Univariate and Multivariate regression analysis, type of treatment received was a significant predictor of cure at week 8 post-treatment. Overall, 30.8% (95% CI = 27.2%- 34.4%) of the study participants experienced mild and transient treatment-associated adverse events, post-treatment abdominal pain (27.1%) being the most common adverse event observed. There was no significant difference in the overall occurrence of adverse events between the two treatment groups. CONCLUSION Praziquantel and Dihydroartemisinin piperaquine combination therapy is safe, and more efficacious compared to praziquantel alone for the treatment of intestinal schistosomiasis. Further studies are needed to explore if the combination therapy can be considered as an option for mass drug administration to control and eventually eliminate schistosomiasis.
Collapse
Affiliation(s)
- Rajabu Hussein Mnkugwe
- Department of Clinical Pharmacology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Omary Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Safari Kinung’hi
- National Institute for Medical Research (NIMR), Mwanza Research Centre, Mwanza, Tanzania
| | - Appolinary Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Eleni Aklillu
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Global research on artemisinin and its derivatives: Perspectives from patents. Pharmacol Res 2020; 159:105048. [PMID: 32590098 PMCID: PMC7309871 DOI: 10.1016/j.phrs.2020.105048] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/18/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022]
Abstract
Artemisinin patents firstly appeared in 1986, and have rapidly increased since 2010. Universities and research centers are collaborating intensively with industry. New indications worth exploring for artemisinin are neoplasms, diseases affecting the immune and nervous systems. Artemisinin-based combination therapy is currently receiving much research attention.
Background The isolation of artemisinin in 1971 heralded the beginning of a new era in antimalarial drug therapy, and artemisinin-based combination therapies are currently the mainstay of malaria treatment worldwide. Artemisinin-related studies have been extensively and intensively executed in the last few decades. However, although many purely technological reviews have been completed in this field, studies on artemisinin from the perspective of patents are still very limited. In terms of the importance of patents for academic research and commercial development, this study aims to reveal the overall patent landscape of artemisinin in the temporal, spatial, and technological dimensions. This work may provide a useful reference for relevant decision-making by researchers, investors, and policymakers. Methods All available patent data relevant to artemisinin derivatives and artemisinin-based drug combinations developed for use in various therapeutic areas were collected from the Derwent Innovation database. Descriptive statistics and citation analyses were used to analyze the patent landscape. Results A total of 4594 patent documents and 1450 simple patent families from 1986 to 2019 were analyzed. A comprehensive patent landscape of artemisinin is presented from the aspects of time trends, filing countries, patent ownership, co-patents, technological categories, therapeutic areas, and citation networks and pathways. Conclusions China and the United States are mainly responsible for the dramatic increase of artemisinin patents over the last three decades. From the point of view of patents, notable technological issues on artemisinin are chemical and biological synthesis, novel combinations, new formulations and administration routes, drug repositioning, and minimizing the resistance. Furthermore, a critical challenge lies in how to stimulate the industry to develop artemisinin-related drugs by government regulation and public-private partnership.
Collapse
|
28
|
Dai F, Song JH, Hong YP, Bai X, Sohn WM, Hong SJ. Dopaminergic antagonists inhibit bile chemotaxis of adult Clonorchis sinensis and its egg production. PLoS Negl Trop Dis 2020; 14:e0008220. [PMID: 32226018 PMCID: PMC7145267 DOI: 10.1371/journal.pntd.0008220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/09/2020] [Accepted: 03/13/2020] [Indexed: 12/15/2022] Open
Abstract
Human clonorchiasis, caused by Clonorchis sinensis, is endemic in East Asian countries. C. sinensis metacercariae excyst in the duodenum of mammalian hosts, migrate to the intrahepatic bile duct, and mature into adults in the milieu of bile. We have previously shown that newly excysted juvenile C. sinensis move chemotactically toward bile and bile acids. Here, the chemotactic behavior of adult C. sinensis (CsAd) toward bile and bile acids was investigated. CsAds moved toward 0.05-5% bile and were most attracted to 0.5% bile but moved away from 10% bile. Upon exposure to 1-10% bile, CsAds eventually stopped moving and then died quickly. Among bile acids, CsAds showed strong chemotaxis toward cholic acid (CA) and deoxycholic acid. On the contrary, CsAds repelled from lithocholic acid (LCA). Moreover, at higher than 10 mM LCA, CsAds became sluggish and eventually died. Dopamine D1 receptor antagonists (LE-300 and SKF-83566), D2/3 receptor antagonists (raclopride and its derivative CS-49612), and a dopamine re-uptake inhibitor inhibited CA-induced chemotaxis of CsAds almost completely. Clinically used antipsychotic drugs, namely chlorpromazine, haloperidol, and clozapine, are dopaminergic antagonists and are secreted into bile. They completely inhibited chemotaxis of CsAds toward CA. At the maximum doses used to treat patients, the three tested medicines only expelled 2-12% of CsAds from the experimentally infected rabbits, but reduced egg production by 64-79%. Thus, antipsychotic medicines with dopaminergic antagonism could be considered as new anthelmintic candidates for human C. sinensis infections.
Collapse
Affiliation(s)
- Fuhong Dai
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
- Department of Parasitology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, PR China
| | - Jin-Ho Song
- Department of Pharmacology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Yeon Pyo Hong
- Department of Preventive Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Xuelian Bai
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
- Clinical Medicine Laboratory, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, PR China
| | - Woon-Mok Sohn
- Department of Parasitology and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Sung-Jong Hong
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
29
|
Affiliation(s)
- Stewart Sell
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY, USA
| |
Collapse
|
30
|
Comparative study of excretory-secretory proteins released by Schistosoma mansoni-resistant, susceptible and naïve Biomphalaria glabrata. Parasit Vectors 2019; 12:452. [PMID: 31521183 PMCID: PMC6744689 DOI: 10.1186/s13071-019-3708-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/05/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Schistosomiasis is a harmful neglected tropical disease caused by infection with Schistosoma spp., such as Schistosoma mansoni. Schistosoma must transition within a molluscan host to survive. Chemical analyses of schistosome-molluscan interactions indicate that host identification involves chemosensation, including naïve host preference. Proteomic technique advances enable sophisticated comparative analyses between infected and naïve snail host proteins. This study aimed to compare resistant, susceptible and naïve Biomphalaria glabrata snail-conditioned water (SCW) to identify potential attractants and deterrents. METHODS Behavioural bioassays were performed on S. mansoni miracidia to compare the effects of susceptible, F1 resistant and naïve B. glabrata SCW. The F1 resistant and susceptible B. glabrata SCW excretory-secretory proteins (ESPs) were fractionated using SDS-PAGE, identified with LC-MS/MS and compared to naïve snail ESPs. Protein-protein interaction (PPI) analyses based on published studies (including experiments, co-expression, text-mining and gene fusion) identified S. mansoni and B. glabrata protein interaction. Data are available via ProteomeXchange with identifier PXD015129. RESULTS A total of 291, 410 and 597 ESPs were detected in the susceptible, F1 resistant and naïve SCW, respectively. Less overlap in ESPs was identified between susceptible and naïve snails than F1 resistant and naïve snails. F1 resistant B. glabrata ESPs were predominately associated with anti-pathogen activity and detoxification, such as leukocyte elastase and peroxiredoxin. Susceptible B. glabrata several proteins correlated with immunity and anti-inflammation, such as glutathione S-transferase and zinc metalloproteinase, and S. mansoni sporocyst presence. PPI analyses found that uncharacterised S. mansoni protein Smp_142140.1 potentially interacts with numerous B. glabrata proteins. CONCLUSIONS This study identified ESPs released by F1 resistant, susceptible and naïve B. glabrata to explain S. mansoni miracidia interplay. Susceptible B. glabrata ESPs shed light on potential S. mansoni miracidia deterrents. Further targeted research on specific ESPs identified in this study could help inhibit B. glabrata and S. mansoni interactions and stop human schistosomiasis.
Collapse
|
31
|
Gu X, Peng Y, Zhao Y, Liang X, Tang Y, Liu J. A novel derivative of artemisinin inhibits cell proliferation and metastasis via down-regulation of cathepsin K in breast cancer. Eur J Pharmacol 2019; 858:172382. [DOI: 10.1016/j.ejphar.2019.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 02/04/2023]
|
32
|
Vil’ VA, Terent’ev AO, Mulina OM. Bioactive Natural and Synthetic Peroxides for the Treatment of Helminth and Protozoan Pathogens: Synthesis and Properties. Curr Top Med Chem 2019; 19:1201-1225. [DOI: 10.2174/1568026619666190620143848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
The significant spread of helminth and protozoan infections, the uncontrolled intake of the
known drugs by a large population, the emergence of resistant forms of pathogens have prompted people
to search for alternative drugs. In this review, we have focused attention on structures and synthesis of
peroxides active against parasites causing neglected tropical diseases and toxoplasmosis. To date, promising
active natural, semi-synthetic and synthetic peroxides compounds have been found.
Collapse
Affiliation(s)
- Vera A. Vil’
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospekt 47, Moscow, 119991, Russian Federation
| | - Alexander O. Terent’ev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospekt 47, Moscow, 119991, Russian Federation
| | - Olga M. Mulina
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospekt 47, Moscow, 119991, Russian Federation
| |
Collapse
|
33
|
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: 1.0] [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
|
34
|
Houmsou RS, Wama BE, Agere H, Uniga JA, Amuta EU, Kela SL. High Efficacy of Praziquantel in Schistosoma haematobium-Infected Children in Taraba State, Northeast Nigeria: A follow-up study. Sultan Qaboos Univ Med J 2018; 18:e304-e310. [PMID: 30607270 DOI: 10.18295/squmj.2018.18.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/12/2017] [Accepted: 09/30/2018] [Indexed: 11/16/2022] Open
Abstract
Objectives This study aimed to assess the efficacy of praziquantel in reducing urinary schistosomiasis prevalence, parasite burden and morbidity rates among a previously reported sample of Schistosoma haematobium-infected children. In addition, predisposing factors for reinfection one year post-treatment were also determined. Methods This prospective follow-up study was conducted between March 2014 and February 2015 among 675 previously reported children with urinary schistosomiasis in the Murbai and Surbai communities of Ardo Kola, Taraba State, Nigeria. A single dose of 40 mg/kg of praziquantel was administered to each infected child, with a second dose administered one month later if necessary. The number of S. haematobium eggs in urine samples was calculated at baseline and post-treatment. Results At four weeks post-treatment, the overall cure rate was 98.1%. Among children with low and heavy parasite burdens at baseline, egg reduction rates (ERRs) were 100% and 96.5%, respectively. The vast majority of children with microhaematuria (98.7%) and proteinuria (98.6%) at baseline were cured at follow-up. Following a second dose, the ERR, overall and morbidity cure rates increased to 100%. At one year post-treatment, 272 infected children (40.3%) were re-assessed; of these, 51 children (18.8%) were reinfected. Close proximity to bodies of water (odds ratio [OR] = 1.23, 95% confidence interval [CI]: 0.998-1.530; P = 0.05) and fishing (OR = 2.23, 95% CI: 0.828-6.040; P = 0.01) were significant factors that predisposed children to reinfection. Conclusion A moderate rate of reinfection was noted. Governmental and nongovernmental organisations in Nigeria should collaborate on mass treatment and health education campaigns to reduce the incidence of urinary schistosomiasis reinfections.
Collapse
Affiliation(s)
- Robert S Houmsou
- Department of Biological Sciences, Taraba State University, Jalingo, Taraba State, Nigeria
| | - Binga E Wama
- Department of Biological Sciences, Taraba State University, Jalingo, Taraba State, Nigeria
| | - Hemen Agere
- Department of Biological Sciences, Faculty of Pure & Applied Sciences, Federal University Wukari, Taraba State, Nigeria
| | - John A Uniga
- Paediatrics Unit, Federal Medical Centre, Jalingo, Taraba State, Nigeria
| | - Elizabeth U Amuta
- Department of Biological Sciences, University of Agriculture, Makurdi, Benue State, Nigeria
| | - Santaya L Kela
- Department of Biological Sciences, Federal University of Kachere, Kashere, Gombe State, Nigeria
| |
Collapse
|
35
|
Bergquist R, Elmorshedy H. Artemether and Praziquantel: Origin, Mode of Action, Impact, and Suggested Application for Effective Control of Human Schistosomiasis. Trop Med Infect Dis 2018; 3:tropicalmed3040125. [PMID: 30572592 PMCID: PMC6306701 DOI: 10.3390/tropicalmed3040125] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 02/02/2023] Open
Abstract
The stumbling block for the continued, single-drug use of praziquantel (PZQ) against schistosomiasis is less justified by the risk of drug resistance than by the fact that this drug is inactive against juvenile parasites, which will mature and start egg production after chemotherapy. Artemisinin derivatives, currently used against malaria in the form of artemisinin-based combination therapy (ACT), provide an opportunity as these drugs are not only active against malaria plasmodia, but surprisingly also against juvenile schistosomes. An artemisinin/PZQ combination would be complementary, and potentially additive, as it would kill two schistosome life cycle stages and thus confer a transmission-blocking modality to current chemotherapy. We focus here on single versus combined regimens in endemic settings. Although the risk of artemisinin resistance, already emerging with respect to malaria therapy in Southeast Asia, prevents use in countries where ACT is needed for malaria care, an artemisinin-enforced praziquantel treatment (APT) should be acceptable in regions of North Africa (including Egypt), the Middle East, China, and Brazil that are not endemic for malaria. Thanks to recent progress with respect to high-resolution diagnostics, based on circulating schistosome antigens in humans and molecular approaches for snail surveys, it should be possible to keep areas scheduled for schistosomiasis elimination under surveillance, bringing rapid response to bear on problems arising. The next steps would be to investigate where and for how long APT should be applied to make a lasting impact. A large-scale field trial in an area with modest transmission should tell how apt this approach is.
Collapse
Affiliation(s)
| | - Hala Elmorshedy
- College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia.
- Department of Tropical Health, High Institute of Public Health, Alexandria University, Alexandria 21561, Egypt.
| |
Collapse
|
36
|
Qian C, Zhang Y, Zhang X, Yuan C, Gao Z, Yuan H, Zhong J. Effectiveness of the new integrated strategy to control the transmission of Schistosoma japonicum in China: a systematic review and meta-analysis. ACTA ACUST UNITED AC 2018; 25:54. [PMID: 30444486 PMCID: PMC6238655 DOI: 10.1051/parasite/2018058] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/04/2018] [Indexed: 12/30/2022]
Abstract
Since 2004, the national schistosomiasis control strategy in China has shifted from the morbidity control strategy (conventional strategy) to an integrated strategy (new strategy). We investigated the effectiveness of the new strategy and compared it against the conventional strategy. We retrieved from electronic databases the literature regarding the new strategy published from 2000 to 2017. The effect of the new or conventional strategy on infection by Schistosoma japonicum of humans and snails (Oncomelania hupensis) was evaluated with pooled log relative risk (logRR). A total of only eight eligible publications were included in the final meta-analysis. The results showed that implementation of the new strategy reduced the infection risk by 3–4 times relative to the conventional strategy. More specifically, the conventional strategy caused a reduction in both human (logRR = 0.56, 95% CI: 0.12–0.99) and snail infections (logRR = 0.34, 95% CI: −0.69–1.37), while the new strategy also significantly reduced both human (logRR = 1.89, 95% CI: 1.33–2.46) and snail infections (logRR = 1.61, 95% CI: 1.06–2.15). In contrast to the conventional strategy, the new strategy appeared more effective to control both human (logRR difference = 1.32, 95% CI: 0.78–1.86) and snail infections (logRR difference = 1.53, 95% CI: 0.76–2.31). Our data demonstrate that the new integrated strategy is highly effective to control the transmission of S. japonicum in China, and this strategy is recommended for schistosomiasis elimination in other affected regions across the world, with adaptation to local conditions.
Collapse
Affiliation(s)
- Chunyan Qian
- Yuhang Branch, The Second Affiliated Hospital of Zhejiang University, Hangzhou 311100, Zhejiang Province, PR China - School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | - Yuefeng Zhang
- Yuhang Branch, The Second Affiliated Hospital of Zhejiang University, Hangzhou 311100, Zhejiang Province, PR China
| | - Xinyan Zhang
- Department of Clinical Laboratory, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200001, PR China
| | - Chao Yuan
- Shanghai Skin Disease Hospital, Shanghai 200443, PR China
| | - Zhichao Gao
- Yuhang Branch, The Second Affiliated Hospital of Zhejiang University, Hangzhou 311100, Zhejiang Province, PR China
| | - Hong Yuan
- Yuhang Branch, The Second Affiliated Hospital of Zhejiang University, Hangzhou 311100, Zhejiang Province, PR China
| | - Jiang Zhong
- School of Life Sciences, Fudan University, Shanghai 200433, PR China
| |
Collapse
|
37
|
Rezende CMF, Coitinho JB, Costa M, Silva MR, Giusta M, Oliveira-Prado R, Corrêa-Oliveira R, Nagem R, Goes AM. Biochemical analysis and identification of linear B-cell epitopes from recombinant Sm21.7 antigen from Schistosoma mansoni. Mol Immunol 2018; 101:29-37. [PMID: 29857222 DOI: 10.1016/j.molimm.2018.05.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/14/2018] [Accepted: 05/23/2018] [Indexed: 12/28/2022]
Abstract
Schistosoma mansoni tegument is a dynamic host-interactive layer that is an essential source of parasite antigens and a relevant field for schistosome vaccine research. Sm21.7 is a cytoskeleton antigen found in S. mansoni tegument that engenders protection in experimental challenge infection. Because of its crucial role in the parasite tegument and its promising protective capability, Sm21.7 is an exciting target for the development of therapeutic strategies. The present study describes Sm21.7 structural and biophysical features using circular dichroism spectroscopy and identifies linear B-cell epitopes of Sm21.7 using in-silico methods and immunoassay. The Sm21.7 gene was cloned into the pETDEST42 vector, and the recombinant protein was overexpressed in Escherichia coli DE3. The soluble protein was purified by affinity chromatography followed by ion-exchange chromatography. Purified recombinant Sm21.7 was analyzed by circular dichroism spectroscopy which demonstrated that the rSm21.7 structure was comprised of approximately 38% α-helices and its conformation remains stable at temperatures of up to 60 °C. Prediction of rSm21.7 B-cell epitopes was based on amino acid physicochemical properties. Sixteen peptides corresponding to predicted epitopes were synthesized and immunoreactivity assessed by spot peptide array using pooled rSm21.7-immunized mice sera or patients' sera with different clinical forms of S. mansoni infection. Immunoassays revealed that sera from rSm21.7-immunized mice reacted predominantly with peptides located in the dynein-light chain domain (DLC) at the C-terminal region of rSm21.7. Comparative analysis of the antibody response of acute, intestinal and hepatosplenic patients' sera to the Sm21.7 peptides showed that a differential recognition pattern of Sm21.7-derived peptides by intestinal patients' sera might contribute to down-regulate the immune response in chronic intestinal patients. Together, the results may help the development of S. mansoni vaccine strategies based on the rSm21.7 antigen.
Collapse
Affiliation(s)
- Cíntia M F Rezende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil.
| | - Juliana B Coitinho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Mariana Costa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Marina Rodrigues Silva
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Mário Giusta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Roberta Oliveira-Prado
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Corrêa-Oliveira
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Ronaldo Nagem
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Alfredo M Goes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
38
|
Means AR, van Lieshout L, Brienen E, Yuhas K, Hughes JP, Ndungu P, Singa B, Walson JL. Combined effectiveness of anthelmintic chemotherapy and WASH among HIV-infected adults. PLoS Negl Trop Dis 2018; 12:e0005955. [PMID: 29346385 PMCID: PMC5773082 DOI: 10.1371/journal.pntd.0005955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/12/2017] [Indexed: 11/18/2022] Open
Abstract
Introduction Current global helminth control guidelines focus on regular deworming of targeted populations for morbidity control. However, water, sanitation, and hygiene (WASH) interventions may also be important for reducing helminth transmission. We evaluated the impact of different potential helminth protective packages on infection prevalence, including repeated treatment with albendazole and praziquantel with and without WASH access. Methodology/Principal findings We conducted a cohort study nested within a randomized trial of empiric deworming of HIV-infected adults in Kenya. Helminth infections and infection intensity were diagnosed using semi-quantitative real-time PCR. We conducted a manual forward stepwise model building approach to identify if there are packages of interventions that may be protective against an STH infection of any species (combined outcome) and each helminth species individually. We conducted secondary analyses using the same approach only amongst individuals with no anthelmintis exposure. We used interaction terms to test for potential intervention synergy. Approximately 22% of the 701 stool samples provided were helminth-infected, most of which were of low to moderate intensity. The odds of infection with any STH species were lower for individuals who were treated with albendazole (aOR:0.11, 95%CI: 0.05, 0.20, p<0.001), adjusting for age and sex. Although most WASH conditions demonstrated minimal additional benefit in reducing the probability of infection with any STH species, access to safe flooring did appear to offer some additional protection (aOR:0.34, 95%CI: 0.20, 0.56, p<0.001). For schistosomiasis, only treatment with praziquantel was protective (aOR:0.30 95%CI: 0.14, 0.60, p = 0.001). Amongst individuals who were not treated with albendazole or praziquantel, the most protective intervention package to reduce probability of STH infections included safe flooring (aOR:0.34, 95%CI: 0.20, 0.59, p<0.001) and latrine access (aOR:0.59, 95%CI: 0.35, 0.99, p = 0.05). Across all species, there was no evidence of synergy or antagonism between anthelmintic chemotherapy with albendazole or praziquantel and WASH resources. Conclusions/Significance Deworming is effective in reducing the probability of helminth infections amongst HIV-infected adults. With the exception of safe flooring, WASH offers minimal additional benefit. However, WASH does appear to significantly reduce infection prevalence in adults who are not treated with chemotherapy. Trial registration ClinicalTrials.gov, NCT00507221. More than 2 billion people are infected with soil transmitted helminthiasis (STH) and schistosomiasis globally. In addition to deworming with albendazole and praziquantel, water, sanitation, and hygiene (WASH) may be needed to reduce the risk of helminth reinfection and ultimately to break transmission. However evidence on the effectiveness of integrated deworming and WASH programs are mixed. Using data from a randomized controlled trial of deworming among HIV-infected adults in Kenya, we evaluated the associations between different potential packages of deworming and WASH interventions with helminth prevalence. We also aimed to understand if there was a benefit to accessing both WASH and deworming (i.e. synergy). Our findings indicate that deworming treatments were effective in reducing the probability of infection with most helminth species. Of the WASH conditions assessed, safe flooring and latrine access significantly reduced the probability of helminth infections among adults with access to treatment. However WASH may play a more prominent protective role among individuals without access to deworming treatments. These findings also suggest minimal synergistic benefit of coordinated anthelmintic chemotherapy and WASH in this population. These findings have important implications for adult populations who are currently not targeted by deworming campaigns.
Collapse
Affiliation(s)
- Arianna R. Means
- Department of Global Health, University of Washington, Seattle, WA, United States
- DeWorm3, Natural History Museum, London, United Kingdom
- * E-mail:
| | - Lisette van Lieshout
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Eric Brienen
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Krista Yuhas
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - James P. Hughes
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Paul Ndungu
- Centre for Clinical Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Benson Singa
- Centre for Clinical Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Judd L. Walson
- Department of Global Health, University of Washington, Seattle, WA, United States
- DeWorm3, Natural History Museum, London, United Kingdom
- Departments of Medicine, Pediatrics, and Epidemiology, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
39
|
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: 5.3] [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
|
40
|
Abstract
Praziquantel has remained the drug of choice for schistosomiasis chemotherapy for almost 40 years. The pressing need to develop a new antischistosomal drug may necessitate exploring and filtering chemotherapeutic history to search for the most promising ones. In this context, this review attempts to summarize all progress made in schistosomiasis chemotherapy from the early 20th century (mid-1910s) to 2016. We gathered almost 100 compounds providing information on therapeutic action, specifically covering at least first in vivo studies in animal model and in vitro. Pharmacokinetic and toxicity profiles of antischistosomal agents were also described. Preclinical studies indicate a handful of promising future candidates.
Collapse
|
41
|
Vil' VA, Yaremenko IA, Ilovaisky AI, Terent'ev AO. Peroxides with Anthelmintic, Antiprotozoal, Fungicidal and Antiviral Bioactivity: Properties, Synthesis and Reactions. Molecules 2017; 22:E1881. [PMID: 29099089 PMCID: PMC6150334 DOI: 10.3390/molecules22111881] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/30/2017] [Indexed: 11/23/2022] Open
Abstract
The biological activity of organic peroxides is usually associated with the antimalarial properties of artemisinin and its derivatives. However, the analysis of published data indicates that organic peroxides exhibit a variety of biological activity, which is still being given insufficient attention. In the present review, we deal with natural, semi-synthetic and synthetic peroxides exhibiting anthelmintic, antiprotozoal, fungicidal, antiviral and other activities that have not been described in detail earlier. The review is mainly concerned with the development of methods for the synthesis of biologically active natural peroxides, as well as its isolation from natural sources and the modification of natural peroxides. In addition, much attention is paid to the substantially cheaper biologically active synthetic peroxides. The present review summarizes 217 publications mainly from 2000 onwards.
Collapse
Affiliation(s)
- Vera A Vil'
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991 Moscow, Russia.
- Faculty of Chemical and Pharmaceutical Technology and Biomedical Products, D. I. Mendeleev University of Chemical Technology of Russia, 9 Miusskaya Square, 125047 Moscow, Russia.
- All-Russian Research Institute for Phytopathology, B. Vyazyomy, 143050 Moscow, Russia.
| | - Ivan A Yaremenko
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991 Moscow, Russia.
- Faculty of Chemical and Pharmaceutical Technology and Biomedical Products, D. I. Mendeleev University of Chemical Technology of Russia, 9 Miusskaya Square, 125047 Moscow, Russia.
- All-Russian Research Institute for Phytopathology, B. Vyazyomy, 143050 Moscow, Russia.
| | - Alexey I Ilovaisky
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991 Moscow, Russia.
| | - Alexander O Terent'ev
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991 Moscow, Russia.
- Faculty of Chemical and Pharmaceutical Technology and Biomedical Products, D. I. Mendeleev University of Chemical Technology of Russia, 9 Miusskaya Square, 125047 Moscow, Russia.
- All-Russian Research Institute for Phytopathology, B. Vyazyomy, 143050 Moscow, Russia.
| |
Collapse
|
42
|
From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin Cancer Biol 2017; 46:65-83. [DOI: 10.1016/j.semcancer.2017.02.009] [Citation(s) in RCA: 354] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/15/2017] [Accepted: 02/24/2017] [Indexed: 12/24/2022]
|
43
|
Baekby M, Glerup H, Stribolt K, Tarp B. Hepatosplenic schistosomiasis: playing hide-and-seek with an elusive parasite. BMJ Case Rep 2017; 2017:bcr-2017-219437. [PMID: 28814579 DOI: 10.1136/bcr-2017-219437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
A 27-year-old man of Eritrean origin presented with persistent left-sided abdominal pain. Initial investigation showed signs of liver fibrosis, portal hypertension and splenomegaly. A diagnosis of hepatosplenic schistosomiasis was suspected on grounds of elevated total IgE, grey area antischistosomiasis antibodies and the high endemic status of his native country. However, repeated microscopy of faecal and urine samples, as well as rectal biopsies, failed to demonstrate schistosomal eggs. Finally, the diagnosis of hepatosplenic schistosomiasis was established through demonstration of a Schistosoma mansoni egg in a liver biopsy taken in an attempt to clarify the cause of the above findings. The patient had recently been treated for uncomplicated malaria. Lowered schistosomiasis worm/egg burden and hence reduced sensitivity of classic microscopy-based schistosomiasis testing was attributed to the antischistosomal activity of the antimalarial chemotherapy.
Collapse
Affiliation(s)
- Martin Baekby
- Department of Clinical Microbiology, Aarhus Universitetshospital, Aarhus N, Denmark
| | - Henning Glerup
- Department of Internal Medicine, Regionshospitalet Silkeborg, Silkeborg, Denmark
| | - Katrine Stribolt
- Department of Pathology, Regionshospitalet Randers, Randers, Midtjylland, Denmark
| | - Britta Tarp
- Department of Internal Medicine, Regionshospitalet Silkeborg, Silkeborg, Denmark
| |
Collapse
|
44
|
LOW PREVALENCE OF INTESTINAL SCHISTOSOMIASIS AMONG FISHERFOLK LIVING ALONG THE RIVER NILE IN NORTH-WESTERN UGANDA: A BIOSOCIAL INVESTIGATION. J Biosoc Sci 2017; 48 Suppl 1:S74-91. [PMID: 27428067 DOI: 10.1017/s0021932016000237] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Mass drug administration has been less successful as a technique for controlling intestinal schistosomiasis (S. mansoni) than anticipated. In Uganda, the mass distribution of praziquantel has been provided to populations at risk of infection since the early 2000s, but prevalence mostly remains high. This is the case, for example, at locations in north-western and south-eastern Uganda. However, there is a remarkable exception. Among Madi fishing populations and their immediate neighbours, living close to the border with South Sudan, the rate of infection has dropped dramatically. A parasitological survey carried out at twelve fishing sites in 2013 identified only three cases of S. mansoni among 383 adults tested. This article asks: why is the prevalence of S. mansoni so low among fisherfolk in northern Uganda? Taking a biosocial approach, it suggests that the mass distribution of drugs, free of charge, has had an impact. However, the low prevalence of infection cannot be attributed to this alone. Other important factors may also have contributed to the decline in infection. These include changing fishing livelihoods, local attitudes to public health interventions, access to water and sanitation facilities, hygiene practices and the use of anti-malarial treatments. Above all, the article highlights the importance of investigating both social and biological dimensions of infection simultaneously, and of recognizing the local complexities of sustainably treating this debilitating parasitic disease.
Collapse
|
45
|
The Potential Therapeutic Effects of Artesunate on Stroke and Other Central Nervous System Diseases. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1489050. [PMID: 28116289 PMCID: PMC5223005 DOI: 10.1155/2016/1489050] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/20/2016] [Indexed: 12/31/2022]
Abstract
Artesunate is an important agent for cerebral malaria and all kinds of other severe malaria because it is highly efficient, lowly toxic, and well-tolerated. Loads of research pointed out that it had widespread pharmacological activities such as antiparasites, antitumor, anti-inflammation, antimicrobes activities. As we know, the occurrence and development of neurological disorders usually refer to intricate pathophysiologic mechanisms and multiple etiopathogenesis. Recent progress has also demonstrated that drugs with single mechanism and serious side-effects are not likely the candidates for treatment of the neurological disorders. Therefore, the pluripotent action of artesunate may result in it playing an important role in the prevention and treatment of these neurological disorders. This review provides an overview of primary pharmacological mechanism of artesunate and its potential therapeutic effects on neurological disorders. Meanwhile, we also briefly summarize the primary mechanisms of artemisinin and its derivatives. We hope that, with the evidence presented in this review, the effect of artesunate in prevention and curing for neurological disorders can be further explored and studied in the foreseeable future.
Collapse
|
46
|
Ross AGP, Chau TN, Inobaya MT, Olveda RM, Li Y, Harn DA. A new global strategy for the elimination of schistosomiasis. Int J Infect Dis 2016; 54:130-137. [PMID: 27939558 DOI: 10.1016/j.ijid.2016.09.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 09/18/2016] [Indexed: 01/17/2023] Open
Abstract
Mass drug administration utilising a single oral dose of 40mg/kg of praziquantel (PZQ) has been endorsed and advocated by the World Health Organisation (WHO) for the global control and elimination of schistosomiasis. However, this strategy is failing primarily because the drugs are not getting to the people who need them the most. The current global coverage is 20%, the drug compliance rate is less than 50%, and the drug efficacy is approximately 50%. Thus in reality, only about 5% of the reservoir human population is actually receiving intermittent chemotherapy. Despite claims that more of the drug will soon be made available the current strategy is inherently flawed and will not lead to disease elimination. We discuss the many practical issues related to this global strategy, and advocate for an integrated control strategy targeting the life cycle and the most at-risk. Moreover, we discuss how an integrated control package for schistosomiasis should fit within a larger integrated health package for rural and remote villages in the developing world. A holistic health system approach is required to achieve sustainable control and ultimately disease elimination.
Collapse
Affiliation(s)
- Allen G P Ross
- Menzies Health Institute Queensland, Griffith University, Australia.
| | - Thao N Chau
- Discipline of Public Health, Flinders University, Australia
| | | | - Remigio M Olveda
- Research Institute for Tropical Medicine, Department of Health, the Philippines
| | - Yuesheng Li
- QIMR Berghofer Medical Research Institute, Australia; Hunan Institute of Parasitic Diseases, Peoples Republic of China
| | - Donald A Harn
- Centre for Tropical and Emerging Global Health Diseases, University of Georgia, USA
| |
Collapse
|
47
|
Yamabe M, Kumagai T, Shimogawara R, Blay EA, Hino A, Ichimura K, Sato A, Kim HS, Ohta N. Novel synthetic compounds with endoperoxide structure damage juvenile stage of Schistosoma mansoni by targeting lysosome-like organelles. Parasitol Int 2016; 66:917-924. [PMID: 27771462 DOI: 10.1016/j.parint.2016.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/13/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
Abstract
The new synthetic compound 1,2,6,7-tetraoxaspiro[7.11]nonadecan (N-89), a novel anti-malaria drug candidate, is also a promising drug candidate against schistosomiasis with killing effects against juvenile stage of S. mansoni. In order to investigate how N-89 kills schistosomes, we used a derivative of N-89, 6-(1,2,6,7-tetraoxaspiro[7.11] nonadec-4-yl)hexan-1-ol (N-251), which enables us to conjugate with fluorescent reagents. Firstly, N-251 showed strong killing effects to larvae of S. mansoni in vitro. Ultrastructural analysis showed the disruptions of the lysosome-like organelles or the acetabular glands, followed by cytoplasmic lysis inside the worm body in N-251-treated group under electron microscopy. For rhodamine-conjugated N-251 and organelle markers, we observed that N-251 accumulated in acidic organelle. In addition, LysoTracker signals in these acidic organelles disappeared in N-251-treated group over time. Finally, we observed that the activity of cathepsin B, a lysosome-specific enzyme, was also decreased together with alternation of acidic organelle marker signal by N-251-treated group. These results suggested that our synthesized compounds induced the dysfunction or the disruption of acidic lysosome-like organelles and finally led to worm death.
Collapse
Affiliation(s)
- Masafumi Yamabe
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takashi Kumagai
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Rieko Shimogawara
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Emmanuel Awusah Blay
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Akina Hino
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Akira Sato
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Hye-Sook Kim
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Nobuo Ohta
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
48
|
Barda B, Coulibaly JT, Puchkov M, Huwyler J, Hattendorf J, Keiser J. Efficacy and Safety of Moxidectin, Synriam, Synriam-Praziquantel versus Praziquantel against Schistosoma haematobium and S. mansoni Infections: A Randomized, Exploratory Phase 2 Trial. PLoS Negl Trop Dis 2016; 10:e0005008. [PMID: 27636542 PMCID: PMC5026339 DOI: 10.1371/journal.pntd.0005008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/28/2016] [Indexed: 12/30/2022] Open
Abstract
Background Schistosomiasis affects millions of people, yet treatment options are limited. The antimalarial Synriam (piperaquine 150 mg/arterolane 750 mg) and the anthelminthic moxidectin revealed promising antischistosomal properties in preclinical or clinical studies. Methodology We conducted two single-blind, randomized exploratory Phase 2 trials in Schistosoma mansoni and S. haematobium-infected adolescents in northern and central Côte d’Ivoire. Our primary endpoints were cure rates (CRs) and egg reduction rates (ERRs) based on geometric mean and safety. Each subject was asked to provide two stool samples (S. mansoni trial) for Kato-Katz analysis or three urine samples (S. haematobium trial) for urine filtration and one finger prick for malaria screening at baseline and follow-up. Participants were randomly assigned to either moxidectin, Synriam, Synriam plus praziquantel or praziquantel. Principal Findings 128 adolescents (age: 12–17 years) were included in each study. Against S. haematobium moxidectin and Synriam revealed low efficacy. On the other hand, Synriam plus praziquantel and praziquantel yielded CRs of 60.0% and 38.5% and ERRs of 96.0% and 93.5%, respectively. CRs observed in the treatment of S. mansoni were 13.0%, 6.7%, 27.0%, and 27.6% for moxidectin, Synriam, Synriam plus praziquantel and praziquantel, respectively. ERRs ranged from 64.9% (Synriam) to 87.5% (praziquantel). Conclusion/Significance Synriam and moxidectin show low efficacy against S. haematobium, hence an ancillary benefit is not expected when these drugs are used for treating onchocerciasis and malaria in co-endemic settings. Further studies are needed to corroborate our findings that moxidectin and Synriam show moderate ERRs against S. mansoni. Schistosomiasis is a parasitic infection that affects millions of people all over the world and it is due to schistosomes, helminths (worms) that infect the intestine and the urinary bladder. Treatment options are limited, with praziquantel being the only used drug. The antimalarial Synriam and the anthelminthic moxidectin revealed good action against this worm in previous studies. We conducted two studies in Schistosoma mansoni and S. haematobium-infected adolescents in Côte d’Ivoire. Subjects positive for the infection were allocated by chance to the four groups of treatment (moxidectin, Synriam, Synriam plus praziquantel or praziquantel); participants did not know which drug they took. Our aim was to calculate how many participants were negative after the treatment and how did the intensity of infection change before and after treatment. Each subject provided stools and urines for examination. 128 adolescents were included in each study. Moxidectin and Synriam did not work well against S. haematobium. Against S. mansoni, only a small part of the participants were negative after treatment in all treatment groups, but the intensity of infections were reduced. Further studies are needed to better understand this result.
Collapse
Affiliation(s)
- Beatrice Barda
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Jean T. Coulibaly
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- Unite´ de Formation et de Recherche Biosciences, Universite´ Felix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Maxim Puchkov
- Department of Pharmaceutical Technology, University of Basel, Basel, Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Technology, 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, Basel, Switzerland
- University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Prophylactic effect of artemether on human schistosomiasis mansoni among Egyptian children: A randomized controlled trial. Acta Trop 2016; 158:52-58. [PMID: 26921676 DOI: 10.1016/j.actatropica.2016.02.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 02/15/2016] [Accepted: 02/21/2016] [Indexed: 11/23/2022]
Abstract
A double-blind, randomized controlled trial was conducted in an endemic focus for Schistosoma mansoni in Kafr El-Sheikh Governorate, Northern Nile Delta, Egypt, to evaluate the prophylactic effect of artemether (ART) given in conjunction with praziquantel (PZQ). The study encompassed 913 primary school children randomly assigned to two treatment groups PZQ/ART and PZQ/ART-placebo. At baseline, both groups received 40 mg/kg body weight of PZQ twice four weeks apart, after which one group received 6 mg/kg body weight of ART every 3 weeks in 5 cycles during the transmission season and the other group received ART-placebo. At the end of the study, prevalence of infection among the PZQ/ART was approximately half that of the PZQ/ART-placebo group, i.e. 6.7% versus 11.6%, and incidence of new infections for the PZQ/ART was 2.7% versus 6.5% for the PZQ/ART-placebo. In conclusion, PZQ/ART combined therapy might be considered as an adjunct measure against human schistosomiasis, by specifically reducing transmission and therefore contribute to disease elimination.
Collapse
|
50
|
Cheng PC, Lin CN, Peng SY, Kang TF, Lee KM. Combined IL-12 Plasmid and Recombinant SjGST Enhance the Protective and Anti-pathology Effect of SjGST DNA Vaccine Against Schistosoma japonicum. PLoS Negl Trop Dis 2016; 10:e0004459. [PMID: 26891172 PMCID: PMC4758724 DOI: 10.1371/journal.pntd.0004459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/23/2016] [Indexed: 01/10/2023] Open
Abstract
Schistosomiasis is listed as one of most important tropical diseases and more than 200 million people are estimated to be infected. Development of a vaccine is thought to be the most effective way to control this disease. Recombinant 26-kDa glutathione S-transferase (rSjGST) has previously been reported to achieve a worm reduction rate of 42-44%. To improve the efficiency of the vaccine against Schistosoma japonicum, we immunized mice with a combination of pcDNA vector-encoded 26-kDa SjGST (pcDNA/SjGST), IL-12 expressing-plasmid (pIL-12), and rSjGST. Co-vaccination with pcDNA/SjGST, pIL-12, and rSjGST led to a reduction in worm burden, hepatic egg burden, and the size of liver tissue granulomas than that in the untreated infection controls. In addition, we detected high levels of specific IgG, IgG1, and IgG2a against the rSjGST antigen in infected mice vaccinated with this combination of pcDNA/SjGST, pIL-12, and rSjGST. Moreover, high expression levels of Th2 cytokines, including IL-4 and IL-10, were also detected in this group, without diminished levels of IL-12, INF-γ, and TNF-α cytokines that are related to parasite killing. In conclusion, we have developed a new vaccination regimen against S. japonicum infection and shown that co-immunization with pcDNA/SjGST vaccine, pIL-12, and rSjGST has significant anti-parasite, anti-hepatic egg and anti-pathology effects in mice. The efficacy of this vaccination method should be further validated in large animals such as water buffalo. This method may help to reduce the transmission of zoonotic schistosomiasis japonica.
Collapse
Affiliation(s)
- Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, Taipei Medical University, Taipei, Taiwan
- * E-mail: (PCC); (KML)
| | - Ching-Nan Lin
- Institute of Microbiology and Immunology, National Yang-Mng University, Taipei, Taiwan
- Institute of Tropical Medicine, National Yang-Mng University, Taipei, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Fu Kang
- Institute of Tropical Medicine, National Yang-Mng University, Taipei, Taiwan
| | - Kin-Mu Lee
- Institute of Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan
- * E-mail: (PCC); (KML)
| |
Collapse
|