1
|
Rostami A, Lundström-Stadelmann B, Frey CF, Beldi G, Lachenmayer A, Chang BCH, Norouzian MM, Hemphill A, Gasser RB. Human Alveolar Echinococcosis-A Neglected Zoonotic Disease Requiring Urgent Attention. Int J Mol Sci 2025; 26:2784. [PMID: 40141427 PMCID: PMC11943292 DOI: 10.3390/ijms26062784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Alveolar echinococcosis (AE) in humans is caused by the larval (metacestode) stage of Echinococcus multilocularis, commonly known as the 'fox tapeworm'. This disease predominantly targets the liver and has an invasive growth pattern, allowing it to spread to adjacent and distant tissues. Due to its gradual progression and tumour-like characteristics, early diagnosis and prompt intervention are crucial, particularly as there are currently no highly effective vaccines or chemotherapeutics against AE. Current estimates suggest that ~10,500 new infections occur annually worldwide; however, more research is required to refine the prevalence and incidence data for both human and animal hosts in endemic areas of the world. This article discusses the biology of E. multilocularis, outlines aspects of the pathogenesis, diagnosis, treatment, and management of AE, reviews its global distribution, annual incidence, and prevalence, highlights the role of molecular parasitology in advancing therapeutic strategies, and presents recommendations for improving the prevention and control of AE in human populations.
Collapse
Affiliation(s)
- Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; (A.R.); (M.M.N.)
| | - Britta Lundström-Stadelmann
- Multidisciplinary Center for Infectious Diseases, University of Bern, Hallerstrasse 6, 3012 Bern, Switzerland; (B.L.-S.); (G.B.)
- Reference Laboratory of Federal Office for Food Safety and Veterinary Affairs, Institute of Parasitology and Echinococcus, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland;
| | - Caroline F. Frey
- Reference Laboratory of Federal Office for Food Safety and Veterinary Affairs, Institute of Parasitology and Echinococcus, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland;
| | - Guido Beldi
- Multidisciplinary Center for Infectious Diseases, University of Bern, Hallerstrasse 6, 3012 Bern, Switzerland; (B.L.-S.); (G.B.)
- Department for Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
- Department for Biomedical Research, Bern University Hospital, University of Bern, 3008 Bern, Switzerland
| | - Anja Lachenmayer
- Department for Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
| | - Bill C. H. Chang
- Department of Veterinary Biosciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Mohammad Mobin Norouzian
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; (A.R.); (M.M.N.)
| | - Andrew Hemphill
- Reference Laboratory of Federal Office for Food Safety and Veterinary Affairs, Institute of Parasitology and Echinococcus, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland;
| | - Robin B. Gasser
- Department of Veterinary Biosciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| |
Collapse
|
2
|
Jing QD, A JD, Liu LX, Fan HN. Current status of drug therapy for alveolar echinococcosis. World J Hepatol 2024; 16:1243-1254. [PMID: 39606163 PMCID: PMC11586754 DOI: 10.4254/wjh.v16.i11.1243] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024] Open
Abstract
Alveolar echinococcosis (AE) is a chronic zoonotic parasitic disease caused by infection with Echinococcus multilocularis. AE is associated with a high mortality rate and poses a significant threat to human health. The primary treatment for AE is surgical resection of the lesions; however, owing to its long incubation period and insidious disease progression, many patients are diagnosed only after the onset of complications such as liver cirrhosis, jaundice, and portal hypertension, which preclude curative surgical intervention. For patients who are unwilling or unable to undergo surgery, lifelong administration of anti-AE medications is necessary. Benzimidazole compounds, such as albendazole and mebendazole, are the current mainstays of treatment, offering good efficacy. Nevertheless, these medications primarily inhibit parasite proliferation rather than eradicate the infection, and their long-term use can lead to significant drug-related toxic effects. Consequently, there is an urgent need to develop new therapeutic strategies that convey better efficacy and reduce the adverse effects associated with current treatments. Recent advancements in AE therapy include novel synthetic compounds such as antiviral agents, antibiotics, antineoplastic agents, immunosuppressants, and antiangiogenic agents, as well as natural compounds derived from traditional Chinese and Tibetan medicine. These new drugs show promising clinical potential because they interfere with parasitic metabolic pathways and cellular structures. This review aims to discuss recent research on AE drug therapy, including mechanisms of action, dosing regimens, signalling pathways, and therapeutic outcomes, with a goal of providing new insights and directions for the development of anti-AE drugs and summarizing current advancements in AE pharmacotherapy.
Collapse
Affiliation(s)
- Qin-Dong Jing
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining 810000, Qinghai Province, China
- School of Clinical Medicine, Qinghai University, Xining 810000, Qinghai Province, China
| | - Ji-De A
- Department of Hepatic Hydatidosis, Qinghai Provincial People's Hospital, Xining 810007, Qinghai Province, China
| | - Lin-Xun Liu
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining 810000, Qinghai Province, China
| | - Hai-Ning Fan
- Department of Hepatobiliary and Pancreatic Surgery, Qinghai Province Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810001, Qinghai Province, China.
| |
Collapse
|
3
|
Wang W, Li J, Qi W, Chen Y, Tian M, Wu C, Zhang Y, Yu Y, Han S, Han X, Duan L, Zhang W. Drug repurposing for hard-to-treat human alveolar echinococcosis: pyronaridine and beyond. Parasitology 2024; 151:1441-1448. [PMID: 39508157 PMCID: PMC12052434 DOI: 10.1017/s0031182024001124] [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: 03/29/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 11/08/2024]
Abstract
Human alveolar echinococcosis is a hard-to-treat and largely untreated parasitic disease with high associated health care costs. The current antiparasitic treatment for alveolar echinococcosis relies exclusively on albendazole, which does not act parasiticidally and can induce severe adverse effects. Alternative, and most importantly, improved treatment options are urgently required. A drug repurposing strategy identified the approved antimalarial pyronaridine as a promising candidate against Echinococcus multilocularis infections. Following a 30-day oral regimen (80 mg kg−1 day−1), pyronaridine achieved an excellent therapeutic outcome in a clinically relevant hepatic alveolar echinococcosis murine model, showing a significant reduction in both metacestode size (72.0%) and counts (85.2%) compared to unmedicated infected mice, which revealed significantly more potent anti-echinococcal potency than albendazole treatment at an equal dose (metacestode size: 42.3%; counts: 4.1%). The strong parasiticidal activity of pyronaridine was further confirmed by the destructive damage to metacestode tissues observed morphologically. In addition, a screening campaign combined with computational similarity searching against an approved drug library led to the identification of pirenzepine, a gastric acid-inhibiting drug, exhibiting potent parasiticidal activity against protoscoleces and in vitro cultured small cysts, which warranted further in vivo investigation as a promising anti-echinococcal lead compound. Pyronaridine has a known drug profile and a long track record of safety, and its repurposing could translate rapidly to clinical use for human patients with alveolar echinococcosis as an alternative or salvage treatment.
Collapse
Affiliation(s)
- Weisi Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, China
- National Center for International Research on Tropical Diseases, Shanghai, China
| | - Jun Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wenjing Qi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ying Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, China
- National Center for International Research on Tropical Diseases, Shanghai, China
| | - Mengxiao Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Chuanchuan Wu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yingfang Yu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, China
- National Center for International Research on Tropical Diseases, Shanghai, China
| | - Shuai Han
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, China
- National Center for International Research on Tropical Diseases, Shanghai, China
| | - Xiumin Han
- Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Liping Duan
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, China
- National Center for International Research on Tropical Diseases, Shanghai, China
| | - Wenbao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
4
|
Autier B, Verger A, Plaisse C, Manuel C, Chollet-Krugler M, Preza M, Lundstroem-Stadelmann B, Amela-Cortes M, Aninat C, Samson M, Brandhonneur N, Dion S. PLGA-PEG-COOH nanoparticles are efficient systems for delivery of mefloquine to Echinococcus multilocularis metacestodes. Exp Parasitol 2024; 265:108811. [PMID: 39111383 DOI: 10.1016/j.exppara.2024.108811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/10/2024] [Accepted: 08/05/2024] [Indexed: 08/11/2024]
Abstract
Alveolar echinococcosis (AE) is a severe disease caused by the infection with the larval stage of Echinococcus multilocularis, the metacestode. As there is no actual curative drug therapy, recommendations to manage AE patients are based on radical surgery and prophylactic administration of albendazole or mebendazole during 2 years to prevent relapses. There is an urgent need for new therapeutic strategies for the management of AE, as the drugs in use are only parasitostatic, and can induce toxicity. This study aimed at developing a drug delivery system for mefloquine, an antiparasitic compound which is highly active against E. multilocularis in vitro and in experimentally infected mice. We formulated mefloquine-loaded PLGA-PEG-COOH (poly-(lactic-co-glycolic acid)) nanoparticles that exhibit stable physical properties and mefloquine content. These nanoparticles crossed the outer acellular laminated layer of metacestodes in vitro and delivered their content to the inner germinal layer within less than 5 min. The in vitro anti-echinococcal activity of mefloquine was not altered during the formulation process. However, toxicity against hepatocytes was not reduced when compared to free mefloquine. Altogether, this study shows that mefloquine-loaded PLGA-PEG-COOH nanoparticles are promising candidates for drug delivery during AE treatment. However, strategies for direct parasite-specific targeting of these particles should be developed.
Collapse
Affiliation(s)
- Brice Autier
- IRSET (UMR_S 1085), INSERM (Institut de Recherche en Santé, Environnement et Travail), EHESP, CHU Rennes, University of Rennes, Rennes, France
| | - Alexis Verger
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes)-UMR 6226, Univ Rennes, F-35000, Rennes, France
| | - Charleen Plaisse
- IRSET (UMR_S 1085), INSERM (Institut de Recherche en Santé, Environnement et Travail), EHESP, University of Rennes, Rennes, France
| | - Christelle Manuel
- IRSET (UMR_S 1085), INSERM (Institut de Recherche en Santé, Environnement et Travail), EHESP, University of Rennes, Rennes, France
| | - Marylène Chollet-Krugler
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes)-UMR 6226, Univ Rennes, F-35000, Rennes, France
| | - Matias Preza
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Britta Lundstroem-Stadelmann
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Multidisciplinary Center of Infectious Diseases, University of Bern, Berne, Switzerland
| | - Marian Amela-Cortes
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes)-UMR 6226, Univ Rennes, F-35000, Rennes, France
| | - Caroline Aninat
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000, Rennes, France
| | - Michel Samson
- IRSET (UMR_S 1085), INSERM (Institut de Recherche en Santé, Environnement et Travail), EHESP, University of Rennes, Rennes, France
| | - Nolwenn Brandhonneur
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes)-UMR 6226, Univ Rennes, F-35000, Rennes, France
| | - Sarah Dion
- IRSET (UMR_S 1085), INSERM (Institut de Recherche en Santé, Environnement et Travail), EHESP, University of Rennes, Rennes, France.
| |
Collapse
|
5
|
Autier B, Robert-Gangneux F, Dion S. Chemotherapy for the treatment of alveolar echinococcosis: Where are we? Parasite 2024; 31:56. [PMID: 39311470 PMCID: PMC11418394 DOI: 10.1051/parasite/2024055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
Alveolar echinococcosis (AE) is a severe liver disease due to infection with the Echinococcus multilocularis larval stage, called the metacestode. Management of AE is based on benzimidazole chemotherapy (albendazole or mebendazole), associated with surgery when possible. Benzimidazoles are the only compounds recommended for the treatment of AE; however, these are parasitostatic, which means that the parasite can resume growth when treatment is interrupted. Also, benzimidazoles can cause liver dysfunction which may prevent their use. Numerous drugs have been reported to have in vitro activity against E. multilocularis, but few had satisfactory in vivo activity, and none were clearly more effective than benzimidazoles. These drugs belong to various therapeutic categories including anti-infective agents (e.g. amphotericin B, mefloquine, pentamidine derivatives), anti-neoplastic compounds (e.g. imatinib, nilotinib, bortezomib), plant-extracted compounds (e.g. thymol, crocin, carvacrol) and others (e.g. metformin, verapamil, thiaclopride). These treatments are generally of limited interest due to their toxicity, their unfavorable pharmacokinetics, or the scarcity of studies involving humans. Apart from benzimidazoles, only amphotericin B, mefloquine and nitazoxanide have been reported to be used for human AE treatment, with unsatisfactory results. Few studies have aimed at developing innovative strategies for AE drug therapy, such as vectorization of drugs using nanoparticles. Altogether, this review emphasizes the urgent need for new therapeutic strategies in AE management, for which there is currently no curative chemotherapy.
Collapse
Affiliation(s)
- Brice Autier
- Université de Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085 Rennes France
| | - Florence Robert-Gangneux
- Université de Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085 Rennes France
| | - Sarah Dion
- Université de Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085 Rennes France
| |
Collapse
|
6
|
Zheng W, Lv G, Li J, Zhang Y, Qi W, Yan M, Wu J, Chan C, Pan X, Zhang W. Repurposing of a library for high-content screening of inhibitors against Echinococcus granulosus. Parasit Vectors 2024; 17:373. [PMID: 39227942 PMCID: PMC11370232 DOI: 10.1186/s13071-024-06456-6] [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: 05/08/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Cystic echinococcosis (CE) is a zoonotic disease caused by the larval stage of the dog tapeworm Echinococcus granulosus sensu lato (E. granulosus), with a worldwide distribution. The current treatment strategy for CE is insufficient. Limited drug screening models severely hamper the discovery of effective anti-echinococcosis drugs. METHODS In the present study, using high-content screening technology, we developed a novel high-throughput screening (HTS) assay by counting the ratio of propidium iodide-stained dead protoscoleces (PSCs) to the total number of PSCs. In vitro and ex vivo cyst viability assays were utilized to determine the effect of drugs on cyst viability. RESULTS Using the newly established HTS assay, we screened approximately 12,000 clinical-stage or The Food and Drug Administration (FDA)-approved small molecules from the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library, as well as the LOPAC1280 and SelleckChem libraries, as a strategic approach to facilitate the drug discovery process. Initial screening yielded 173 compounds with anti-echinococcal properties, 52 of which demonstrated dose-response efficacy against E. granulosus PSCs in vitro. Notably, two agents, omaveloxolone and niclosamide, showed complete inhibition upon further validation in cyst and microcyst viability assays in vitro after incubation for 3 days, and in an ex vivo cyst viability assay using cysts isolated from the livers of mice infected with E. granulosus, as determined by morphological assessment. CONCLUSIONS Through the development of a novel HTS assay and by repurposing libraries, we identified omaveloxolone and niclosamide as potent inhibitors against E. granulosus. These compounds show promise as potential anti-echinococcal drugs, and our strategic approach has the potential to promote drug discovery for parasitic infections.
Collapse
Affiliation(s)
- Weinan Zheng
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China.
| | - Gaofei Lv
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China
| | - Jun Li
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yao Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Wenjing Qi
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Mingzhi Yan
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Jinzhi Wu
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China
| | - Chikin Chan
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China
| | - Xiaoben Pan
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China.
| | - Wenbao Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
7
|
Cardillo NM, Lacy PA, Villarino NF, Doggett JS, Riscoe MK, Bastos RG, Laughery JM, Ueti MW, Suarez CE. Comparative efficacy of buparvaquone and imidocarb in inhibiting the in vitro growth of Babesia bovis. Front Pharmacol 2024; 15:1407548. [PMID: 38751779 PMCID: PMC11094231 DOI: 10.3389/fphar.2024.1407548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction B. bovis is an apicomplexan parasite responsible for bovine babesiosis, a tick-borne disease with a worldwide impact. The disease remains inefficiently controlled, and few effective drugs, including imidocarb dipropionate (ID), are currently available in endemic areas. The objective of this study was to evaluate whether buparvaquone (BPQ), a drug currently used to treat cattle infected with the Babesia-related Theileria spp. parasites, could be active against Babesia parasites. Herein, we compared the effect of ID and BPQ on B. bovis growth in vitro erythrocyte culture. Methods We compared the effect of ID and BPQ on the culture-adapted Texas T2Bo strain of B. bovis. In vitro cultured parasites were incubated with ID and BPQ at two starting parasitemia levels (PPE), 0.2% and 1%. In vitro cultured parasites were treated with ID or BPQ at concentrations ranging from 10 to 300 nM, during 4 consecutive days. Parasitemia levels were daily evaluated using microscopic examination. Data was compared using the independent Student's t-test. Results and discussion Both ID and BPQ significantly inhibited (p < 0.05) the growth of B. bovis, regardless of the initial parasitemia used. At 1% parasitemia, BPQ had lower calculated inhibitory concentration 50 (IC50: 50.01) values than ID (IC50: 117.3). No parasites were found in wells with 0.2% starting parasitemia, treated previously with 50 nM of BPQ or ID, after 2 days of culture without drugs. At 1% parasitemia, no parasite survival was detected at 150 nM of BPQ or 300 nM of ID, suggesting that both drugs acted as babesiacidals. Conclusion Overall, the data suggests that BPQ is effective against B. bovis and shows a residual effect that seems superior to ID, which is currently the first-line drug for treating bovine babesiosis globally.
Collapse
Affiliation(s)
- Natalia M. Cardillo
- Animal Disease Research Unit, United States Department of Agriculture, Agricultural Research Service, WSU, Pullman, WA, United States
- Estación Experimental INTA Paraná Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Parana, Argentina
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | - Paul A. Lacy
- Animal Disease Research Unit, United States Department of Agriculture, Agricultural Research Service, WSU, Pullman, WA, United States
| | - Nicolas F. Villarino
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, United States
| | - J. Stone Doggett
- Oregon Health and Science University, Portland, OR, United States
- VA Portland Healthcare System, Portland, OR, United States
| | - Michael K. Riscoe
- Oregon Health and Science University, Portland, OR, United States
- VA Portland Healthcare System, Portland, OR, United States
| | - Reginaldo G. Bastos
- Animal Disease Research Unit, United States Department of Agriculture, Agricultural Research Service, WSU, Pullman, WA, United States
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | - Jacob M. Laughery
- Animal Disease Research Unit, United States Department of Agriculture, Agricultural Research Service, WSU, Pullman, WA, United States
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | - Massaro W. Ueti
- Animal Disease Research Unit, United States Department of Agriculture, Agricultural Research Service, WSU, Pullman, WA, United States
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | - Carlos E. Suarez
- Animal Disease Research Unit, United States Department of Agriculture, Agricultural Research Service, WSU, Pullman, WA, United States
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| |
Collapse
|
8
|
Tajeri S, Chattopadhyay D, Langsley G, Nijhof AM. A Theileria annulata parasite with a single mutation, methionine 128 to isoleucine (M128I), in cytochrome B is resistant to buparvaquone. PLoS One 2024; 19:e0299002. [PMID: 38626086 PMCID: PMC11020719 DOI: 10.1371/journal.pone.0299002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/04/2024] [Indexed: 04/18/2024] Open
Abstract
Tropical theileriosis is a fatal leukemic-like disease of cattle caused by the tick-transmitted protozoan parasite Theileria annulata. The economics of cattle meat and milk production is severely affected by theileriosis in endemic areas. The hydroxynaphtoquinone buparvaquone (BPQ) is the only available drug currently used to treat clinical theileriosis, whilst BPQ resistance is emerging and spreading in endemic areas. Here, we chronically exposed T. annulata-transformed macrophages in vitro to BPQ and monitored the emergence of drug-resistant parasites. Surviving parasites revealed a significant increase in BPQ IC50 compared to the wild type parasites. Drug resistant parasites from two independent cloned lines had an identical single mutation, M128I, in the gene coding for T. annulata cytochrome B (Tacytb). This in vitro generated mutation has not been reported in resistant field isolates previously, but is reminiscent of the methionine to isoleucine mutation in atovaquone-resistant Plasmodium and Babesia. The M128I mutation did not appear to exert any deleterious effect on parasite fitness (proliferation and differentiation to merozoites). To gain insight into whether drug-resistance could have resulted from altered drug binding to TaCytB we generated in silico a 3D-model of wild type TaCytB and docked BPQ to the predicted 3D-structure. Potential binding sites cluster in four areas of the protein structure including the Q01 site. The bound drug in the Q01 site is expected to pack against an alpha helix, which included M128, suggesting that the change in amino acid in this position may alter drug-binding. The in vitro generated BPQ resistant T. annulata is a useful tool to determine the contribution of the various predicted docking sites to BPQ resistance and will also allow testing novel drugs against theileriosis for their potential to overcome BPQ resistance.
Collapse
Affiliation(s)
- Shahin Tajeri
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research, Freie Universität Berlin, Berlin, Germany
| | - Debasish Chattopadhyay
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Gordon Langsley
- Inserm U1016-CNRS UMR8104, Institut Cochin, Paris, France
- Laboratoire de Biologie Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
| | - Ard M. Nijhof
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
9
|
Kaethner M, Rennar G, Gallinger T, Kämpfer T, Hemphill A, Mäder P, Luque-Gómez A, Schlitzer M, Lundström-Stadelmann B. In Vitro Activities of Dithiocarbamate Derivatives against Echinococcus multilocularis Metacestode Vesicles. Trop Med Infect Dis 2023; 8:517. [PMID: 38133449 PMCID: PMC10747736 DOI: 10.3390/tropicalmed8120517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
The metacestode stage of the fox tapeworm Echinococcus multilocularis causes the severe zoonotic disease alveolar echinococcosis. New treatment options are urgently needed. Disulfiram and dithiocarbamates were previously shown to exhibit activity against the trematode Schistosoma mansoni. As both parasites belong to the platyhelminths, here we investigated whether these compounds were also active against E. multilocularis metacestode vesicles in vitro. We used an in vitro drug-screening cascade for the identification of novel compounds against E. multilocularis metacestode vesicles with disulfiram and 51 dithiocarbamates. Five compounds showed activity against E. multilocularis metacestode vesicles after five days of drug incubation in a damage marker release assay. Structure-activity relationship analyses revealed that a S-2-hydroxy-5-nitro benzyl moiety was necessary for anti-echinococcal activity, as derivatives without this group had no effect on E. multilocularis metacestode vesicles. The five active compounds were further tested for potential cytotoxicity in mammalian cells. For two compounds with low toxicity (Schl-32.315 and Schl-33.652), IC50 values in metacestode vesicles and IC50 values in germinal layer cells were calculated. The compounds were not highly active on isolated GL cells with IC50 values of 27.0 ± 4.2 µM for Schl-32.315 and 24.7 ± 11.5 µM for Schl-33.652, respectively. Against metacestode vesicles, Schl-32.315 was not very active either with an IC50 value of 41.6 ± 3.2 µM, while Schl-33.652 showed a low IC50 of 4.3 ± 1 µM and should be further investigated in the future for its activity against alveolar echinococcosis.
Collapse
Affiliation(s)
- Marc Kaethner
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.K.); (T.K.); (A.H.)
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Georg Rennar
- Institute of Pharmaceutical Chemistry, Philipps-Universität Marburg, 35037 Marburg, Germany (M.S.)
| | - Tom Gallinger
- Institute of Pharmaceutical Chemistry, Philipps-Universität Marburg, 35037 Marburg, Germany (M.S.)
| | - Tobias Kämpfer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.K.); (T.K.); (A.H.)
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.K.); (T.K.); (A.H.)
| | - Patrick Mäder
- Institute of Pharmaceutical Chemistry, Philipps-Universität Marburg, 35037 Marburg, Germany (M.S.)
| | - Ana Luque-Gómez
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Facultad de Ciencias, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - Martin Schlitzer
- Institute of Pharmaceutical Chemistry, Philipps-Universität Marburg, 35037 Marburg, Germany (M.S.)
| | - Britta Lundström-Stadelmann
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.K.); (T.K.); (A.H.)
- Multidisciplinary Center for Infectious Diseases, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
10
|
Jiang H, Wang X, Guo L, Tan X, Gui X, Liao Z, Li Z, Chen X, Wu X. Effect of sunitinib against Echinococcus multilocularis through inhibition of VEGFA-induced angiogenesis. Parasit Vectors 2023; 16:407. [PMID: 37936208 PMCID: PMC10631006 DOI: 10.1186/s13071-023-05999-4] [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: 03/07/2023] [Accepted: 10/04/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Alveolar echinococcosis (AE) is a lethal zoonosis caused by the fox tapeworm Echinococcus multilocularis. The disease is difficult to treat, and an effective therapeutic drug is urgently needed. Echinococcus multilocularis-associated angiogenesis is required by the parasite for growth and metastasis; however, whether antiangiogenic therapy is effective for treating AE is unclear. METHODS The in vivo efficacy of sunitinib malate (SU11248) was evaluated in mice by secondary infection with E. multilocularis. Enzyme-linked immunosorbent assays (ELISAs) were used to evaluate treatment effects on serum IL-4 and vascular endothelial growth factor A (VEGFA) levels after SU11248 treatment. Gross morphological observations and immunohistochemical staining were used to evaluate the impact of SU11248 on angiogenesis and the expression of pro-angiogenic factors VEGFA and VEGF receptor 2 (VEGFR2) in the metacestode tissues. Furthermore, the anthelmintic effects of SU11248 were tested on E. multilocularis metacestodes in vitro. The effect of SU11248 on the expression of VEGFA, VEGFR2, and phosphorylated VEGFR2 (p-VEGFR2) in liver cells infected with protoscoleces in vitro was detected by western blotting, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assay (ELISA). The influence of SU11248 on endothelial progenitor cell (EPC) proliferation and migration was determined using CCK8 and transwell assays. RESULTS In vivo, SU11248 treatment markedly reduced neovascular lesion formation and substantially inhibited E. multilocularis metacestode growth in mice. Further, it exhibited high anti-hydatid activity as efficiently as albendazole (ABZ), and the treatment resulted in reduced protoscolex development. In addition, VEGFA, VEGFR2, and p-VEGFR2 expression was significantly decreased in the metacestode tissues after SU11248 treatment. However, no effect of SU11248 on serum IL-4 levels was observed. In vitro, SU11248 exhibited some anthelmintic effects and damaged the cellular structure in the germinal layer of metacestodes at concentrations below those generally considered acceptable for treatment (0.12-0.5 μM). Western blotting, RT-qPCR, and ELISA showed that in co-cultured systems, only p-VEGFR2 levels tended to decrease with increasing SU11248 concentrations. Furthermore, SU11248 was less toxic to Reuber rat hepatoma (RH) cells and metacestodes than to EPCs, and 0.1 μM SU11248 completely inhibited EPC migration to the supernatants of liver cell and protoscolex co-cultures. CONCLUSIONS SU11248 is a potential candidate drug for the treatment of AE, which predominantly inhibits parasite-induced angiogenesis. Host-targeted anti-angiogenesis treatment strategies constitute a new avenue for the treatment of AE.
Collapse
Affiliation(s)
- Huijiao Jiang
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xiaoyi Wang
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Lijiao Guo
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xiaowu Tan
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xianwei Gui
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Zhenyu Liao
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
- Department of Experimental Medicine, Jintang First People's Hospital West China Hospital Sichuan University Jintang Hospital, Chengdu, 610400, Sichuan, China
| | - Zhiwei Li
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xueling Chen
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China.
| | - Xiangwei Wu
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
11
|
Loos JA, Franco M, Chop M, Rodriguez Rodrigues C, Cumino AC. Resveratrol against Echinococcus sp.: Discrepancies between In Vitro and In Vivo Responses. Trop Med Infect Dis 2023; 8:460. [PMID: 37888588 PMCID: PMC10610609 DOI: 10.3390/tropicalmed8100460] [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: 07/21/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
In an attempt to find new anti-echinococcal drugs, resveratrol (Rsv) effectiveness against the larval stages of Echinococcus granulosus and E. multilocularis was evaluated. The in vitro effect of Rsv on parasites was assessed via optical and electron microscopy, RT-qPCR and immunohistochemistry. In vivo efficacy was evaluated in murine models of cystic (CE) and alveolar echinococcosis (AE). The impact of infection and drug treatment on the mouse bone marrow hematopoietic stem cell (HSC) population and its differentiation into dendritic cells (BMDCs) was investigated via flow cytometry and RT-qPCR. In vitro treatment with Rsv reduced E. granulosus metacestode and protoscolex viability in a concentration-dependent manner, caused ultrastructural damage, increased autophagy gene transcription, and raised Eg-Atg8 expression while suppressing Eg-TOR. However, the intraperitoneal administration of Rsv was not only ineffective, but also promoted parasite development in mice with CE and AE. In the early infection model of AE treated with Rsv, an expansion of HSCs was observed followed by their differentiation towards BMCDs. The latter showed an anti-inflammatory phenotype and reduced LPS-stimulated activation compared to control BMDCs. We suggest that Rsv ineffectiveness could have been caused by the low intracystic concentration achieved in vivo and the drug's hormetic effect, with opposite anti-parasitic and immunomodulatory responses in different doses.
Collapse
Affiliation(s)
- Julia A. Loos
- Instituto de Investigaciones en Producción, Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, Mar del Plata 7600, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
| | - Micaela Franco
- Hospital Interzonal General de Agudos “Dr. Oscar E Alende”, Mar del Plata 7600, Argentina;
| | - Maia Chop
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, Mar del Plata 7600, Argentina
| | - Christian Rodriguez Rodrigues
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, Mar del Plata 7600, Argentina
| | - Andrea C. Cumino
- Instituto de Investigaciones en Producción, Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, Mar del Plata 7600, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, Mar del Plata 7600, Argentina
| |
Collapse
|
12
|
Kaethner M, Preza M, Kaempfer T, Zumstein P, Tamponi C, Varcasia A, Hemphill A, Brehm K, Lundström-Stadelmann B. Establishment and application of unbiased in vitro drug screening assays for the identification of compounds against Echinococcus granulosus sensu stricto. PLoS Negl Trop Dis 2023; 17:e0011343. [PMID: 37540716 PMCID: PMC10431624 DOI: 10.1371/journal.pntd.0011343] [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: 05/01/2023] [Revised: 08/16/2023] [Accepted: 07/13/2023] [Indexed: 08/06/2023] Open
Abstract
Echinococcus multilocularis and E. granulosus s.l. are the causative agents of alveolar and cystic echinococcosis, respectively. Drug treatment options for these severe and neglected diseases are limited to benzimidazoles, which are not always efficacious, and adverse side effects are reported. Thus, novel and improved treatments are needed. In this study, the previously established platform for E. multilocularis in vitro drug assessment was adapted to E. granulosus s.s. In a first step, in vitro culture protocols for E. granulosus s.s. were established. This resulted in the generation of large amounts of E. granulosus s.s. metacestode vesicles as well as germinal layer (GL) cells. In vitro culture of these cells formed metacestode vesicles displaying structural characteristics of metacestode cysts generated in vivo. Next, drug susceptibilities of E. multilocularis and E. granulosus s.s. protoscoleces, metacestode vesicles and GL cells were comparatively assessed employing established assays including (i) metacestode vesicle damage marker release assay, (ii) metacestode vesicle viability assay, (iii) GL cell viability assay, and (iv) protoscolex motility assay. The standard drugs albendazole, buparvaquone, mefloquine, MMV665807, monepantel, niclosamide and nitazoxanide were included. MMV665807, niclosamide and nitazoxanide were active against the parasite in all four assays against both species. MMV665807 and monepantel were significantly more active against E. multilocularis metacestode vesicles, while albendazole and nitazoxanide were significantly more active against E. multilocularis GL cells. Albendazole displayed activity against E. multilocularis GL cells, but no effects were seen in albendazole-treated E. granulosus s.s. GL cells within five days. Treatment of protoscoleces with albendazole and monepantel had no impact on motility. Similar results were observed for both species with praziquantel and its enantiomers against protoscoleces. In conclusion, in vitro culture techniques and drug screening methods previously established for E. multilocularis were successfully implemented for E. granulosus s.s., allowing comparisons of drug efficacy between the two species. This study provides in vitro culture techniques for the reliable generation of E. granulosus s.s. metacestode vesicles and GL cell cultures and describes the validation of standardized in vitro drug screening methods for E. granulosus s.s.
Collapse
Affiliation(s)
- Marc Kaethner
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Matías Preza
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Tobias Kaempfer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Pascal Zumstein
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Claudia Tamponi
- Department of Veterinary Medicine, University of Sassari, Italy
| | | | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Klaus Brehm
- Institute of Hygiene and Microbiology, University of Würzburg, Germany
| | - Britta Lundström-Stadelmann
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Li J, Yang Y, Han X, Li J, Tian M, Qi W, An H, Wu C, Zhang Y, Han S, Duan L, Wang W, Zhang W. Oral Delivery of Anti-Parasitic Agent-Loaded PLGA Nanoparticles: Enhanced Liver Targeting and Improved Therapeutic Effect on Hepatic Alveolar Echinococcosis. Int J Nanomedicine 2023; 18:3069-3085. [PMID: 37312930 PMCID: PMC10259527 DOI: 10.2147/ijn.s397526] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/26/2023] [Indexed: 06/15/2023] Open
Abstract
Background Alveolar echinococcosis (AE) is a lethal parasitic disease caused by infection with the metacestode of the dog/fox tapeworm Echinococcus multilocularis, which primarily affects the liver. Although continued efforts have been made to find new drugs against this orphan and neglected disease, the current treatment options remain limited, with drug delivery considered a likely barrier for successful treatment. Methods Nanoparticles (NPs) have gained much attention in the field of drug delivery due to their potential to improve delivery efficiency and targetability. In this study, biocompatible PLGA nanoparticles encapsulating a novel carbazole aminoalcohol anti-AE agent (H1402) were prepared to promote the delivery of the parent drug to liver tissue for treating hepatic AE. Results H1402-loaded nanoparticles (H1402-NPs) had a uniform spherical shape and a mean particle size of 55 nm. Compound H1402 was efficiently encapsulated into PLGA NPs with a maximal encapsulation efficiency of 82.1% and drug loading content of 8.2%. An in vitro uptake assay demonstrated that H1402-NPs rapidly penetrated the in vitro cultured pre-cyst wall and extensively accumulated in the pre-cysts of E. multilocularis within only 1 h. The biodistribution profile of H1402-NPs determined through ex vivo fluorescence imaging revealed significantly enhanced liver distribution compared to unencapsulated H1402, which translated to improved therapeutic efficacy and reduced systemic toxicity (especially hepatotoxicity and cytotoxicity) in a hepatic AE murine model. Following a 30-day oral regimen (100 mg/kg/day), H1402-NPs significantly reduced the parasitic burden in both the parasite mass (liver and metacestode total weight, 8.8%) and average metacestode size (89.9%) compared to unmedicated infected mice (both p-values < 0.05); the treatment outcome was more effective than those of albendazole- and free H1402-treated individuals. Conclusion Our findings demonstrate the advantages of encapsulating H1402 into PLGA nanoparticles and highlight the potential of H1402-NPs as a promising liver-targeting therapeutic strategy for hepatic AE.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yangyang Yang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Xiumin Han
- Qinghai Provincial People’s Hospital, Xining, Qinghai, People’s Republic of China
| | - Jing Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Mengxiao Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Wenjing Qi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Huniu An
- Qinghai Provincial People’s Hospital, Xining, Qinghai, People’s Republic of China
| | - Chuanchuan Wu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Shuai Han
- NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, People’s Republic of China
| | - Liping Duan
- NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, People’s Republic of China
| | - Weisi Wang
- NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, People’s Republic of China
| | - Wenbao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| |
Collapse
|
14
|
Autier B, Manuel C, Lundstroem-Stadelmann B, Girard JP, Gottstein B, Gangneux JP, Samson M, Robert-Gangneux F, Dion S. Endogenous IL-33 Accelerates Metacestode Growth during Late-Stage Alveolar Echinococcosis. Microbiol Spectr 2023; 11:e0423922. [PMID: 36786637 PMCID: PMC10101030 DOI: 10.1128/spectrum.04239-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/28/2023] [Indexed: 02/15/2023] Open
Abstract
During the course of the infectious disease alveolar echinococcosis (AE), the larval stage of Echinococcus multilocularis develops in the liver, where an initial Th1/Th17 immune response may allow its elimination in resistant individuals. In patients susceptible to infection and disease, the Th2 response initiates later, inducing tolerance to the parasite. The role of interleukin 33 (IL-33), an alarmin released during necrosis and known to drive a Th2 immune response, has not yet been described during AE. Wild-type (WT) and IL-33-/- C57BL/6J mice were infected by peritoneal inoculation with E. multilocularis metacestodes and euthanized 4 months later, and their immune response were analyzed. Immunofluorescence staining and IL-33 enzyme-linked immunosorbent assay (ELISA) were also performed on liver samples from human patients with AE. Overall, metacestode lesions were smaller in IL-33-/- mice than in WT mice. IL-33 was detected in periparasitic tissues, but not in mouse or human serum. In infected mice, endogenous IL-33 modified peritoneal macrophage polarization and cytokine profiles. Th2 cytokine concentrations were positively correlated with parasite mass in WT mice, but not in IL-33-/- mice. In human AE patients, IL-33 concentrations were higher in parasitic tissues than in distant liver parenchyma. The main sources of IL-33 were CD31+ endothelial cells of the neovasculature, present within lymphoid periparasitic infiltrates together with FOXP3+ Tregs. In the murine model, periparasitic IL-33 correlated with accelerated parasite growth putatively through the polarization of M2-like macrophages and release of immunosuppressive cytokines IL-10 and transforming growth factor β1 (TGF-β1). We concluded that IL-33 is a key alarmin in AE that contributes to the tolerogenic effect of systemic Th2 cytokines. IMPORTANCE Infection with the metacestode stage of Echinococcus multilocularis, known as alveolar echinococcosis, is the most severe cestodosis worldwide. However, less than 1% of exposed individuals, in which the immune system is unable to control the parasite, develop the disease. The factors responsible for this interindividual variability are not fully understood. In this in vivo study comparing wild-type and IL-33-/- infected mice, together with data from human clinical samples, we determined that IL-33, an alarmin released following tissue injury and involved in the pathogenesis of cancer and asthma, accelerates the progression of the disease by modulating the periparasitic microenvironment. This suggests that targeting IL-33 could be of interest for the management of patients with AE, and that IL-33 polymorphisms could be responsible for increased susceptibility to AE.
Collapse
Affiliation(s)
- Brice Autier
- IRSET (UMR_S 1085), INSERM (Institut de recherche en santé, environnement et travail), EHESP, CHU Rennes, University of Rennes, Rennes, France
| | - Christelle Manuel
- IRSET (UMR_S 1085), INSERM (Institut de recherche en santé, environnement et travail), EHESP, University of Rennes, Rennes, France
| | - Britta Lundstroem-Stadelmann
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, Toulouse, France
| | - Bruno Gottstein
- Institute of Infectious Diseases, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Jean-Pierre Gangneux
- IRSET (UMR_S 1085), INSERM (Institut de recherche en santé, environnement et travail), EHESP, CHU Rennes, University of Rennes, Rennes, France
| | - Michel Samson
- IRSET (UMR_S 1085), INSERM (Institut de recherche en santé, environnement et travail), EHESP, University of Rennes, Rennes, France
| | - Florence Robert-Gangneux
- IRSET (UMR_S 1085), INSERM (Institut de recherche en santé, environnement et travail), EHESP, CHU Rennes, University of Rennes, Rennes, France
| | - Sarah Dion
- IRSET (UMR_S 1085), INSERM (Institut de recherche en santé, environnement et travail), EHESP, University of Rennes, Rennes, France
| |
Collapse
|
15
|
Tyagi R, Paul A, Raj VS, Ojha KK, Kumar S, Panda AK, Chaurasia A, Yadav MK. A Drug Repurposing Approach to Identify Therapeutics by Screening Pathogen Box Exploiting SARS-CoV-2 Main Protease. Chem Biodivers 2023; 20:e202200600. [PMID: 36597267 DOI: 10.1002/cbdv.202200600] [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: 06/22/2022] [Revised: 12/03/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
Coronavirus disease-19 (COVID-19) is caused by severe acute respiratory syndrome coronavirus -2 (SARS-CoV-2) and is responsible for a higher degree of morbidity and mortality worldwide. There is a smaller number of approved therapeutics available to target the SARS-CoV-2 virus, and the virus is evolving at a fast pace. So, there is a continuous need for new therapeutics to combat COVID-19. The main protease (Mpro ) enzyme of SARS-CoV-2 is essential for replication and transcription of the viral genome, thus could be a potent target for the treatment of COVID-19. In the present study, we performed an in-silico screening analysis of 400 diverse bioactive inhibitors with proven antibacterial and antiviral properties against Mpro drug target. Ten compounds showed a higher binding affinity for Mpro than the reference compound (N3), with desired physicochemical properties. Furthermore, in-depth docking and superimposition revealed that three compounds (MMV1782211, MMV1782220, and MMV1578574) are actively interacting with the catalytic domain of Mpro . In addition, the molecular dynamics simulation study showed a solid and stable interaction of MMV178221-Mpro complex compared to the other two molecules (MMV1782220, and MMV1578574). In line with this observation, MM/PBSA free energy calculation also demonstrated the highest binding free energy of -115.8 kJ/mol for MMV178221-Mpro compound. In conclusion, the present in silico analysis revealed MMV1782211 as a possible and potent molecule to target the Mpro and must be explored in vitro and in vivo to combat the COVID-19.
Collapse
Affiliation(s)
- Rashmi Tyagi
- Center for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Sonepat, 131 029, Haryana, India
| | - Anubrat Paul
- Center for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Sonepat, 131 029, Haryana, India
| | - V Samuel Raj
- Center for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Sonepat, 131 029, Haryana, India
| | - Krishna Kumar Ojha
- Department of Bioinformatics, Central University of South Bihar, Gaya, 824 236, Bihar, India
| | - Sunil Kumar
- ICAR-Indian Agriculture Statistical Research Institute, New Delhi, India, 110012
| | - Aditya K Panda
- Department of Biosciences and Bioinformatics, Khallikote University, Berhampur, 761008, Odisha, India
| | - Anurag Chaurasia
- ICAR-Indian Institute of Vegetable Research, Varanasi, 221305, UP, India
| | - Manoj Kumar Yadav
- Center for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Sonepat, 131 029, Haryana, India
- Department of Biomedical Engineering, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, 131 029, Haryana, India
| |
Collapse
|
16
|
Preliminary Structure-Activity Relationship Study of the MMV Pathogen Box Compound MMV675968 (2,4-Diaminoquinazoline) Unveils Novel Inhibitors of Trypanosoma brucei brucei. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196574. [PMID: 36235118 PMCID: PMC9571290 DOI: 10.3390/molecules27196574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
New drugs are urgently needed for the treatment of human African trypanosomiasis (HAT). In line with our quest for novel inhibitors of trypanosomes, a small library of analogs of the antitrypanosomal hit (MMV675968) available at MMV as solid materials was screened for antitrypanosomal activity. In silico exploration of two potent antitrypanosomal structural analogs (7-MMV1578647 and 10-MMV1578445) as inhibitors of dihydrofolate reductase (DHFR) was achieved, together with elucidation of other antitrypanosomal modes of action. In addition, they were assessed in vitro for tentative inhibition of DHFR in a crude trypanosome extract. Their ADMET properties were also predicted using dedicated software. Overall, the two diaminoquinazoline analogs displayed approximately 40-fold and 60-fold more potency and selectivity in vitro than the parent hit, respectively (MMV1578445 (10): IC50 = 0.045 µM, SI = 1737; MMV1578467 (7): IC50 = 0.06 µM; SI = 412). Analogs 7 and 10 were also strong binders of the DHFR enzyme in silico, in all their accessible protonation states, and interacted with key DHFR ligand recognition residues Val32, Asp54, and Ile160. They also exhibited significant activity against trypanosome protein isolate. MMV1578445 (10) portrayed fast and irreversible trypanosome growth arrest between 4–72 h at IC99. Analogs 7 and 10 induced in vitro ferric iron reduction and DNA fragmentation or apoptosis induction, respectively. The two potent analogs endowed with predicted suitable physicochemical and ADMET properties are good candidates for further deciphering their potential as starting points for new drug development for HAT.
Collapse
|
17
|
Chaudhry S, Zurbriggen R, Preza M, Kämpfer T, Kaethner M, Memedovski R, Scorrano N, Hemphill A, Doggett JS, Lundström-Stadelmann B. Dual inhibition of the Echinococcus multilocularis energy metabolism. Front Vet Sci 2022; 9:981664. [PMID: 35990276 PMCID: PMC9388906 DOI: 10.3389/fvets.2022.981664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Alveolar echinococcosis is caused by the metacestode stage of the zoonotic parasite Echinococcus multilocularis. Current chemotherapeutic treatment options rely on benzimidazoles, which have limited curative capabilities and can cause severe side effects. Thus, novel treatment options are urgently needed. In search for novel targetable pathways we focused on the mitochondrial energy metabolism of E. multilocularis. The parasite relies hereby on two pathways: The classical oxidative phosphorylation including the electron transfer chain (ETC), and the anaerobic malate dismutation (MD). We screened 13 endochin-like quinolones (ELQs) in vitro for their activities against two isolates of E. multilocularis metacestodes and isolated germinal layer cells by the phosphoglucose isomerase (PGI) assay and the CellTiter Glo assay. For the five most active ELQs (ELQ-121, ELQ-136, ELQ-271, ELQ-400, and ELQ-437), EC50 values against metacestodes were assessed by PGI assay, and IC50 values against mammalian cells were measured by Alamar Blue assay. Further, the gene sequence of the proposed target, the mitochondrial cytochrome b, was analyzed. This allowed for a limited structure activity relationship study of ELQs against E. multilocularis, including analyses of the inhibition of the two functional sites of the cytochrome b. By applying the Seahorse XFp Extracellular Flux Analyzer, oxygen consumption assays showed that ELQ-400 inhibits the E. multilocularis cytochrome bc1 complex under normoxic conditions. When tested under anaerobic conditions, ELQ-400 was hardly active against E. multilocularis metacestodes. These results were confirmed by transmission electron microscopy. ELQ-400 treatment increased levels of parasite-released succinate, the final electron acceptor of the MD. This suggests that the parasite switched to MD for energy generation. Therefore, MD was inhibited with quinazoline, which did not induce damage to metacestodes under anaerobic conditions. However, it reduced the production of succinate compared to control treated parasites (i.e., inhibited the MD). The combination treatment with quinazoline strongly improved the activity of the bc1 inhibitor ELQ-400 against E. multilocularis metacestodes under anaerobic conditions. We conclude that simultaneous targeting of the ETC and the MD of E. multilocularis is a possible novel treatment approach for alveolar echinococcosis, and possibly also other foodborne diseases inflicted by platyhelminths, which cause substantial economic losses in livestock industry.
Collapse
Affiliation(s)
- Sheena Chaudhry
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Raphael Zurbriggen
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Matías Preza
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tobias Kämpfer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Marc Kaethner
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Roman Memedovski
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nathalie Scorrano
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Joseph Stone Doggett
- Department of Infectious Diseases and Pathobiology, Oregon Health and Science University, Portland, OR, United States
- Department of Infectious Diseases and Pathobiology, Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Britta Lundström-Stadelmann
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- *Correspondence: Britta Lundström-Stadelmann
| |
Collapse
|
18
|
Wang S, Ma Y, Wang W, Dai Y, Sun H, Li J, Wang S, Li F. Status and prospect of novel treatment options toward alveolar and cystic echinococcosis. Acta Trop 2022; 226:106252. [PMID: 34808118 DOI: 10.1016/j.actatropica.2021.106252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/19/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023]
Abstract
Cystic echinococcosis (CE) and alveolar echinococcosis (AE) are the two most important global parasitic infectious diseases caused by species of Echinococcus granulosus and E. multilocularis, respectively. Although numerous trials have been performed in search of novel therapeutic options to curb the neglected zoonosis, no other nonsurgical options are currently available to replace the licensed anti echinococcal drugs albendazole (ABZ) and mebendazole (MBZ). A safer and more effective treatment plan for echinococcosis is therefore urgently needed to compensate for this therapeutic shortfall. Here, we present a review of the literature for state-of-the-art valuable anti-parasitic compounds and novel strategies that have proved effective against CE and AE, which includes details about the pharmaceutical type, practical approach, experimental plan, model application and protoscolecidal effects in vivo and in vitro. The content includes the current application of traditional clinical chemicals, the preparation of new compounds with various drug loadings, repurposing findings, combined programs, the prospects for Chinese herbal medicines, non-drug administrations and the exploration of target inhibitors based on open-source information for parasitic genes. Next the conventional experimental projects and pharmacodynamic evaluation methods are systematically summarized and evaluated. The demands to optimize the construction of the echinococcosis model and improve the dynamic monitoring method in vivo are also discussed given the shortcomings of in vivo models and monitoring methods.
Collapse
Affiliation(s)
- Sibo Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yibo Ma
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi 832000, Xinjiang, China
| | - Weishan Wang
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi 832000, Xinjiang, China
| | - Yi Dai
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi 832000, Xinjiang, China
| | - Haohao Sun
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi 832000, Xinjiang, China
| | - Jing Li
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi 832000, Xinjiang, China
| | - Shan Wang
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi 832000, Xinjiang, China.
| | - Feng Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
19
|
Maca against Echinococcosis?-A Reverse Approach from Patient to In Vitro Testing. Pathogens 2021; 10:pathogens10101335. [PMID: 34684284 PMCID: PMC8537204 DOI: 10.3390/pathogens10101335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023] Open
Abstract
Drug-based treatment of alveolar echinococcosis (AE) with benzimidazoles is in most cases non-curative, thus has to be taken lifelong. Here, we report on a 56-year-old male AE patient who received standard benzimidazole treatment and biliary plastic stents, and additionally self-medicated himself with the Peruvian plant extract Maca (Lepidium meyenii). After 42 months, viable parasite tissue had disappeared. Based on this striking observation, the anti-echinococcal activity of Maca was investigated in vitro and in mice experimentally infected with Echinococcus multilocularis metacestodes. Albendazole (ABZ)-treated mice and mice treated with an ABZ+Maca combination exhibited a significantly reduced parasite burden compared to untreated or Maca-treated mice. As shown by a newly established UHPLC-MS/MS-based measurement of ABZ-metabolites, the presence of Maca during the treatment did not alter ABZ plasma levels. In vitro assays corroborated these findings, as exposure to Maca had no notable effect on E. multilocularis metacestodes, and in cultures of germinal layer cells, possibly unspecific, cytotoxic effects of Maca were observed. However, in the combined treatments, Maca inhibited the activity of ABZ in vitro. While Maca had no direct anti-parasitic activity, it induced in vitro proliferation of murine spleen cells, suggesting that immunomodulatory properties could have contributed to the curative effect seen in the patient.
Collapse
|
20
|
Liu C, Fan H, Ma J, Ma L, Ge RL. In vitro and in vivo efficacy of thiacloprid against Echinococcus multilocularis. Parasit Vectors 2021; 14:450. [PMID: 34488852 PMCID: PMC8419995 DOI: 10.1186/s13071-021-04952-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Alveolar echinococcosis (AE) is a chronic zoonosis caused by the larval form of Echinococcus multilocularis (E. multilocularis). Current chemotherapy against AE has relied on albendazole and mebendazole, which only exhibit parasitostatic and not parasiticidal efficacy. Therefore, novel compounds for the treatment of this disease are needed. METHODS Phosphoglucose isomerase (PGI) assays were used for compound screening of seven neonicotinoids. The anti-parasitic effects of thiacloprid were then evaluated on E. multilocularis metacestode vesicles, germinal cells and protoscoleces in vitro. Human foreskin fibroblasts (HFF) and Reuber rat hepatoma (RH) cells were used to assess cytotoxicity. Glucose consumption in E. multilocularis protoscoleces and germinal cells was assessed by measuring uptake of 2-deoxyglucose (2-DG). Molecular docking was used to evaluate the potential binding sites of thiacloprid to acetylcholine receptors. In vivo efficacy of thiacloprid was evaluated in mice by secondary infection with E. multilocularis. In addition, ELISA and flow cytometry were used to evaluate the effects of cytokines and T lymphocyte subsets after thiacloprid treatment. Furthermore, collagen deposition and degradation in the host lesion microenvironment were evaluated. RESULTS We found that thiacloprid is the most promising compound, with an IC50 of 4.54 ± 1.10 μM and 2.89 ± 0.34 μM, respectively, against in vitro-cultured E. multilocularis metacestodes and germinal cells. Thiacloprid was less toxic for HFF and RH mammalian cell lines than for metacestodes. In addition, thiacloprid inhibited the acetylcholinesterase activity in protoscoleces, metacestodes and germinal cells. Thiacloprid inhibited glucose consumption by protoscoleces and germinal cells. Subsequently, transmission electron microscopy revealed that treatment with thiacloprid damaged the germinal layer. In vivo, metacestode weight was significantly reduced following oral administration of thiacloprid at 15 and 30 mg/kg. The level of CD4+ T lymphocytes in metacestodes and spleen increased after thiacloprid treatment. Anti-echinococcosis-related cytokines (IL-2, IL-4, IL-10) were significantly increased. Furthermore, thiacloprid inhibited the expression of matrix metalloproteinases (MMPs 1, 3, 9, 13) and promoted collagen deposition in the host lesion microenvironment. CONCLUSIONS The results demonstrated that thiacloprid had parasiticidal activity against E. multilocularis in vitro and in vivo, and could be used as a novel lead compound for the treatment of AE.
Collapse
Affiliation(s)
- Chuanchuan Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining, 810001 Qinghai China
- Qinghai University Affiliated Hospital, Xining, 810001 Qinghai China
- Qinghai Key Laboratory for Echinococcosis, Xining, 810001 Qinghai China
| | - Haining Fan
- Qinghai University Affiliated Hospital, Xining, 810001 Qinghai China
- Qinghai Key Laboratory for Echinococcosis, Xining, 810001 Qinghai China
| | - Jie Ma
- Qinghai University Affiliated Hospital, Xining, 810001 Qinghai China
| | - Lan Ma
- Research Center for High Altitude Medicine, Qinghai University, Xining, 810001 Qinghai China
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, 810001 Qinghai China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, 810001 Qinghai China
| | - Ri-li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, 810001 Qinghai China
- Qinghai Key Laboratory for Echinococcosis, Xining, 810001 Qinghai China
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, 810001 Qinghai China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, 810001 Qinghai China
| |
Collapse
|
21
|
Pou S, Dodean RA, Frueh L, Liebman KM, Gallagher RT, Jin H, Jacobs RT, Nilsen A, Stuart DR, Doggett JS, Riscoe MK, Winter RW. A New Scalable Synthesis of ELQ-300, ELQ-316, and other Antiparasitic Quinolones. Org Process Res Dev 2021; 25:1841-1852. [PMID: 35110959 DOI: 10.1021/acs.oprd.1c00099] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Endochin-Like Quinolone (ELQ) compound class may yield effective, safe treatments for a range of important human and animal afflictions. However, to access the public health potential of this compound series, a synthetic route needed to be devised that lowers costs and is amenable to large scale production. In the new synthetic route described here, a substituted β-keto ester, formed by an Ullmann reaction and subsequent acylation, is reacted with an aniline via a Conrad-Limpach reaction to produce 3-substituted 4(1H)-quinolones such as ELQ-300 and ELQ-316. This synthetic route, the first described to be truly amenable to industrial scale production, is relatively short (5 reaction steps), does not require palladium, chromatographic separation or protecting group chemistry, and may be performed without high vacuum distillation.
Collapse
Affiliation(s)
- Sovitj Pou
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rozalia A Dodean
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Lisa Frueh
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Katherine M Liebman
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rory T Gallagher
- Department of Chemistry, Portland State University, 1719 SW 10 Avenue, Portland, Oregon 97201, United States
| | - Haihong Jin
- Medicinal Chemistry Core, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Robert T Jacobs
- Medicines for Malaria Venture, ICC, route de Pré-Bois 20, P.O. Box 1826, 1215 Geneva 15, Switzerland
| | - Aaron Nilsen
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States.,Medicinal Chemistry Core, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - David R Stuart
- Department of Chemistry, Portland State University, 1719 SW 10 Avenue, Portland, Oregon 97201, United States
| | - J Stone Doggett
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States.,School of Medicine Division of Infectious Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Michael K Riscoe
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States.,Department of Microbiology and Molecular Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Rolf W Winter
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| |
Collapse
|
22
|
Bioinformatic prediction and identification of immunogenic epitopes of the antigenic 14-3-3 protein of Echinococcus multilocularis. Acta Trop 2021; 220:105955. [PMID: 33979643 DOI: 10.1016/j.actatropica.2021.105955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Alveolar echinococcosis is a high-risk parasitic disease caused by the larval stage of Echinococcus multilocularis. The study aimed to predict and identify the dominant Th1/Th2 and B cell epitopes of the antigen protein 14-3-3 beta:alpha from Echinococcus multilocularis. METHODS A comparison of the four amino acid sequences of 14-3-3 beta:alpha was respectively derived from Echinococcus multilocularis, Rattus norvegicus, Canis lupus familiaris, and Homo sapiens was carried out by CLUSTALW to provide a basis for excluding similar epitopes. The amino acid sequence information was analyzed by SOPMA and the homology model was established by Swiss-Model. IEDB and SYFPEITHI were used to predict T cell epitopes. According to the Bcepred and ABCpred, the B cell epitopes were comprehensively predicted and analyzed. The dominant epitopes were validated by Lymphocyte Proliferation, ELISA, ELISpot, and Flow cytometry. RESULTS Eight potential epitopes of 14-3-3 from Echinococcus multilocularis were screened according to the results of prediction and analysis: 14-3-31-15, 14-3-36-21, 14-3-371-86, 14-3-3144-157, 14-3-3145-166, 14-3-3146-160, 14-3-3153-161, and 14-3-3164-177. The 3D structure model of the protein was constructed and the location distribution of potential epitope was ascertained. Respectively, the epitopes of the dominant antigen of B cells were validated as 14-3-3145-166 and 14-3-3164-177; the Th1 dominant antigen epitopes were 14-3-36-21, 14-3-3145-166; and the Th2 dominant epitopes was 14-3-3145-166. CONCLUSION In this study, two dominant antigen epitopes of B cells, two Th1 dominant antigen epitopes, and one Th2 dominant antigen epitope were validated. Our work provides a basis for the subsequent development of efficient and safe vaccines targeting epitopes of Echinococcus multilocularis.
Collapse
|
23
|
Verbaanderd C, Rooman I, Huys I. Exploring new uses for existing drugs: innovative mechanisms to fund independent clinical research. Trials 2021; 22:322. [PMID: 33947441 PMCID: PMC8093905 DOI: 10.1186/s13063-021-05273-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 04/15/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Finding new therapeutic uses for existing medicines could lead to safe, affordable and timely new treatment options for patients with high medical needs. However, due to a lack of economic incentives, pharmaceutical developers are rarely interested to invest in research with approved medicines, especially when they are out of basic patent or regulatory protection. Consequently, potential new uses for these medicines are mainly studied in independent clinical trials initiated and led by researchers from academia, research institutes, or collaborative groups. Yet, additional financial support is needed to conduct expensive phase III clinical trials to confirm the results from exploratory research. METHODS In this study, scientific and grey literature was searched to identify and evaluate new mechanisms for funding clinical trials with repurposed medicines. Semi-structured interviews were conducted with 16 European stakeholders with expertise in clinical research, funding mechanisms and/or drug repurposing between November 2018 and February 2019 to consider the future perspectives of applying new funding mechanisms. RESULTS Traditional grant funding awarded by government and philanthropic organisations or companies is well known and widely implemented in all research fields. In contrast, only little research has focused on the application potential of newer mechanisms to fund independent clinical research, such as social impact bonds, crowdfunding or public-private partnerships. Interviewees stated that there is a substantial need for additional financial support in health research, especially in areas where there is limited commercial interest. However, the implementation of new funding mechanisms is facing several practical and financial challenges, such as a lack of expertise and guidelines, high transaction costs and difficulties to measure health outcomes. Furthermore, interviewees highlighted the need for increased collaboration and centralisation at a European and international level to make clinical research more efficient and reduce the need for additional funding. CONCLUSIONS New funding mechanisms to support clinical research may become more important in the future but the unresolved issues identified in the current study warrant further exploration.
Collapse
Affiliation(s)
- Ciska Verbaanderd
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
- Anticancer Fund, Strombeek-Bever, Belgium.
| | - Ilse Rooman
- Anticancer Fund, Strombeek-Bever, Belgium
- Oncology Research Centre, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Huys
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Wang J, Marreros N, Rufener R, Hemphill A, Gottstein B, Lundström-Stadelmann B. Short communication: Efficacy of albendazole in Echinococcus multilocularis-infected mice depends on the functional immunity of the host. Exp Parasitol 2020; 219:108013. [PMID: 33010287 DOI: 10.1016/j.exppara.2020.108013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/08/2020] [Accepted: 09/29/2020] [Indexed: 01/10/2023]
Abstract
Alveolar echinococcosis (AE) is a deadly parasitic disease that requires lifelong treatment with albendazole. Development of host immunity is pivotal with regard to the clinical outcome of AE, but its influence on conventional albendazole treatment is unknown. Using T-cell deficient athymic nude mice, we demonstrated that functional immunity is required for albendazole to be efficacious against murine AE. These results call for attention given the increasing number of immunocompromised patients with AE.
Collapse
Affiliation(s)
- Junhua Wang
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Berne, Längassstrasse 122, 3012, Berne, Switzerland; Institute for Infectious Diseases, Faculty of Medicine, University of Berne, Friedbühlstrasse 51, 3010, Berne, Switzerland
| | - Nelson Marreros
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Berne, Längassstrasse 122, 3012, Berne, Switzerland
| | - Reto Rufener
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Berne, Längassstrasse 122, 3012, Berne, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Berne, Längassstrasse 122, 3012, Berne, Switzerland
| | - Bruno Gottstein
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Berne, Längassstrasse 122, 3012, Berne, Switzerland; Institute for Infectious Diseases, Faculty of Medicine, University of Berne, Friedbühlstrasse 51, 3010, Berne, Switzerland
| | - Britta Lundström-Stadelmann
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Berne, Längassstrasse 122, 3012, Berne, Switzerland.
| |
Collapse
|
25
|
Metformin Suppresses Development of the Echinococcus multilocularis Larval Stage by Targeting the TOR Pathway. Antimicrob Agents Chemother 2020; 64:AAC.01808-19. [PMID: 32540980 DOI: 10.1128/aac.01808-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Alveolar echinococcosis (AE) is a severe disease caused by the larval stage of the tapeworm Echinococcus multilocularis Current chemotherapeutic treatment options based on benzimidazoles are of limited effectiveness, which underlines the need to find new antiechinococcosis drugs. Metformin is an antihyperglycemic and antiproliferative agent that shows activity against the related parasite Echinococcus granulosus Hence, we assessed the in vitro and in vivo effects of the drug on E. multilocularis Metformin exerted significant dose-dependent killing effects on in vitro cultured parasite stem cells and protoscoleces and significantly reduced the dedifferentiation of protoscoleces into metacestodes. Likewise, oral administration of metformin (50 mg/kg of body weight/day for 8 weeks) was effective in achieving a significant reduction of parasite weight in a secondary murine AE model. Our results revealed mitochondrial membrane depolarization, activation of Em-AMPK, suppression of Em-TOR, and overexpression of Em-Atg8 in the germinal layer of metformin-treated metacestode vesicles. The opposite effects on the level of active Em-TOR in response to exogenous insulin and rapamycin suggest that Em-TOR is part of the parasite's insulin signaling pathway. Finally, the presence of the key lysosomal pathway components, through which metformin reportedly acts, was confirmed in the parasite by in silico assays. Taken together, these results introduce metformin as a promising candidate for AE treatment. Although our study highlights the importance of those direct mechanisms by which metformin reduces parasite viability, it does not necessarily preclude any additional systemic effects of the drug that might reduce parasite growth in vivo.
Collapse
|
26
|
In vivo effect of magnetic microspheres loaded with E2-a in the treatment of alveolar echinococcosis. Sci Rep 2020; 10:12589. [PMID: 32724060 PMCID: PMC7387340 DOI: 10.1038/s41598-020-69484-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/09/2020] [Indexed: 11/16/2022] Open
Abstract
The alveolar echinococcosis of human is a severe helminthic disease caused by the larva of Echinococcus multilocularis tapeworms. Novel compounds or therapy strategies for the treatment of alveolar echinococcosis are urgently needed due to the limitation of the widely used albendazole. Magnetic microspheres as drug carriers in magnetically targeted therapy of tumor have gained growing interests advantaged by delivering the drug to the aimed site, achieving localized therapeutic effect effectively under the influence of an external magnetic field. In this study, we formulated magnetic microspheres loaded with E2-a (PLGA-Fe-E2-a) and identified the activity in E. multilocularis-infected mice which infected with 3,000 protoscoleces intraperitoneally. Compared with the untreated control, with the help of a magnet, there was a significant reduction in parasite burden with PLGA-Fe-E2-a treatment and similar reduction observed with albendazole. PLGA-Fe-E2-a treatment group also showed a significant increase in the IFN-γ level and impaired morphological and ultrastructural alterations. Most importantly, one-third concentrations of E2-a from PLGA-Fe-E2 based on the release profile of E2-a was equally effective in inhibiting metacestode growth as E2-a treated group, supporting efficacy and bioavailability of a drug. It will be an alternative treatment for alveolar echinococcosis using magnetic microspheres as drug carriers.
Collapse
|
27
|
Lundström-Stadelmann B, Rufener R, Hemphill A. Drug repurposing applied: Activity of the anti-malarial mefloquine against Echinococcus multilocularis. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 13:121-129. [PMID: 32636148 PMCID: PMC7389337 DOI: 10.1016/j.ijpddr.2020.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
The current chemotherapeutical treatment against alveolar echinococcosis relies exclusively on benzimidazoles, which are not parasiticidal and can induce severe toxicity. There are no alternative treatment options. To identify novel drugs with activity against Echinococcus multilocularis metacestodes, researchers have studied potentially interesting drug targets (e.g. the parasite's energy metabolism), and/or adopted drug repurposing approaches by undertaking whole organism screenings. We here focus on drug screening approaches, which utilize an in vitro screening cascade that includes assessment of the drug-induced physical damage of metacestodes, the impact on metacestode viability and the viability of isolated parasite stem cells, structure-activity relationship (SAR) analysis of compound derivatives, and the mode of action. Finally, once in vitro data are indicative for a therapeutic window, the efficacy of selected compounds is assessed in experimentally infected mice. Using this screening cascade, we found that the anti-malarial mefloquine was active against E. multilocularis metacestodes in vitro and in vivo. To shed more light into the mode of action of mefloquine, SAR analysis on mefloquine analogues was performed. E. multilocularis ferritin was identified as a mefloquine-binding protein, but its precise role as a drug target remains to be elucidated. In mice that were infected either intraperitoneally with metacestodes or orally with eggs, oral treatment with mefloquine led to a significant reduction of parasite growth compared to the standard treatment with albendazole. However, mefloquine was not acting parasiticidally. Assessment of mefloquine plasma concentrations in treated mice showed that levels were reached which are close to serum concentrations that are achieved in humans during long-term malaria prophylaxis. Mefloquine might be applied in human AE patients as a salvage treatment. Future studies should focus on other repurposed anti-infective compounds (MMV665807, niclosamide, atovaquone), which showed stronger in vitro activity against E. multilocularis than mefloquine.
Collapse
Affiliation(s)
- Britta Lundström-Stadelmann
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Längggassstrasse 122, 3012, Bern, Switzerland.
| | - Reto Rufener
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Längggassstrasse 122, 3012, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Längggassstrasse 122, 3012, Bern, Switzerland
| |
Collapse
|
28
|
Bhattacharya A, Corbeil A, do Monte-Neto RL, Fernandez-Prada C. Of Drugs and Trypanosomatids: New Tools and Knowledge to Reduce Bottlenecks in Drug Discovery. Genes (Basel) 2020; 11:genes11070722. [PMID: 32610603 PMCID: PMC7397081 DOI: 10.3390/genes11070722] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Leishmaniasis (Leishmania species), sleeping sickness (Trypanosoma brucei), and Chagas disease (Trypanosoma cruzi) are devastating and globally spread diseases caused by trypanosomatid parasites. At present, drugs for treating trypanosomatid diseases are far from ideal due to host toxicity, elevated cost, limited access, and increasing rates of drug resistance. Technological advances in parasitology, chemistry, and genomics have unlocked new possibilities for novel drug concepts and compound screening technologies that were previously inaccessible. In this perspective, we discuss current models used in drug-discovery cascades targeting trypanosomatids (from in vitro to in vivo approaches), their use and limitations in a biological context, as well as different examples of recently discovered lead compounds.
Collapse
Affiliation(s)
- Arijit Bhattacharya
- Department of Microbiology, Adamas University, Kolkata, West Bengal 700 126, India;
| | - Audrey Corbeil
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | | | - Christopher Fernandez-Prada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Correspondence: ; Tel.: +1-450-773-8521 (ext. 32802)
| |
Collapse
|
29
|
Partridge FA, Forman R, Bataille CJR, Wynne GM, Nick M, Russell AJ, Else KJ, Sattelle DB. Anthelmintic drug discovery: target identification, screening methods and the role of open science. Beilstein J Org Chem 2020; 16:1203-1224. [PMID: 32550933 PMCID: PMC7277699 DOI: 10.3762/bjoc.16.105] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Helminths, including cestodes, nematodes and trematodes, are a huge global health burden, infecting hundreds of millions of people. In many cases, existing drugs such as benzimidazoles, diethylcarbamazine, ivermectin and praziquantel are insufficiently efficacious, contraindicated in some populations, or at risk of the development of resistance, thereby impeding progress towards World Health Organization goals to control or eliminate these neglected tropical diseases. However, there has been limited recent progress in developing new drugs for these diseases due to lack of commercial attractiveness, leading to the introduction of novel, more efficient models for drug innovation that attempt to reduce the cost of research and development. Open science aims to achieve this by encouraging collaboration and the sharing of data and resources between organisations. In this review we discuss how open science has been applied to anthelmintic drug discovery. Open resources, including genomic information from many parasites, are enabling the identification of targets for new antiparasitic agents. Phenotypic screening remains important, and there has been much progress in open-source systems for compound screening with parasites, including motility assays but also high content assays with more detailed investigation of helminth physiology. Distributed open science compound screening programs, such as the Medicines for Malaria Venture Pathogen Box, have been successful at facilitating screening in diverse assays against many different parasite pathogens and models. Of the compounds identified so far in these screens, tolfenpyrad, a repurposed insecticide, shows significant promise and there has been much progress in creating more potent and selective derivatives. This work exemplifies how open science approaches can catalyse drug discovery against neglected diseases.
Collapse
Affiliation(s)
- Frederick A Partridge
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Ruth Forman
- The Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - Carole J R Bataille
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA United Kingdom
| | - Graham M Wynne
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA United Kingdom
| | - Marina Nick
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA United Kingdom
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| | - Kathryn J Else
- The Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - David B Sattelle
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| |
Collapse
|
30
|
Whole-Cell Phenotypic Screening of Medicines for Malaria Venture Pathogen Box Identifies Specific Inhibitors of Plasmodium falciparum Late-Stage Development and Egress. Antimicrob Agents Chemother 2020; 64:AAC.01802-19. [PMID: 32071059 DOI: 10.1128/aac.01802-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
We report a systematic, cellular phenotype-based antimalarial screening of the Medicines for Malaria Venture Pathogen Box collection, which facilitated the identification of specific blockers of late-stage intraerythrocytic development of Plasmodium falciparum First, from standard growth inhibition assays, we identified 173 molecules with antimalarial activity (50% effective concentration [EC50] ≤ 10 μM), which included 62 additional molecules over previously known antimalarial candidates from the Pathogen Box. We identified 90 molecules with EC50 of ≤1 μM, which had significant effect on the ring-trophozoite transition, while 9 molecules inhibited the trophozoite-schizont transition and 21 molecules inhibited the schizont-ring transition (with ≥50% parasites failing to proceed to the next stage) at 1 μM. We therefore rescreened all 173 molecules and validated hits in microscopy to prioritize 12 hits as selective blockers of the schizont-ring transition. Seven of these molecules inhibited the calcium ionophore-induced egress of Toxoplasma gondii, a related apicomplexan parasite, suggesting that the inhibitors may be acting via a conserved mechanism which could be further exploited for target identification studies. We demonstrate that two molecules, MMV020670 and MMV026356, identified as schizont inhibitors in our screens, induce the fragmentation of DNA in merozoites, thereby impairing their ability to egress and invade. Further mechanistic studies would facilitate the therapeutic exploitation of these molecules as broadly active inhibitors targeting late-stage development and egress of apicomplexan parasites relevant to human health.
Collapse
|
31
|
Li J, Wang W, Yao J, Wang T, Li S, Qi W, Han S, Ren Y, Dang Z, Han X, Guo G, Guo B, Wang L, Duan L, Zhang W. Old drug repurposing for neglected disease: Pyronaridine as a promising candidate for the treatment of Echinococcus granulosus infections. EBioMedicine 2020; 54:102711. [PMID: 32279056 PMCID: PMC7152711 DOI: 10.1016/j.ebiom.2020.102711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/17/2020] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cystic echinococcosis (CE), a condition caused by the larval stage of the dog tapeworm Echinococcus granulosus sensu stricto, is a globally distributed zoonotic disease. Current treatment options for CE are limited, and an effective and safe anti-echinococcal drug is urgently required. METHODS Drug repurposing strategy was employed to identify new therapeutic agents against echinococcal cysts. An in vitro protoscolicidal assay along with in vivo murine models was applied in the drug screening. A microinjection procedure was employed to mimic the clinical PAIR (puncture, aspiration, injection and reaspiration) technique to evaluate the potential application of the candidate drug in clinical practice. FINDINGS We repurposed pyronaridine, an approved antimalarial drug, for the treatment of CE. Following a three-dose intraperitoneal regimen (57 mg/kg, q.d. for 3 days), pyronaridine caused 100% cyst mortality. Oral administration of pyronaridine at 57 mg/kg, q.d. for 30 days significantly reduced the parasitic burden in the pre-infected mice compared with albendazole group (p < 0.001). Using a microinjection of drug into cysts, pyronaridine (200 μM) showed highly effective in term of inhibition of cyst growth (p < 0.05, compared with saline group). Pharmacokinetic analysis revealed that pyronaridine was highly distributed in the liver and lungs, the most affected organs of CE. Function analysis showed that pyronaridine inhibited the activity of topoisomerase I (IC50 = 209.7 ± 1.1 μM). In addition, classical apoptotic hallmarks, including DNA fragmentation and caspase activation, were triggered. INTERPRETATION Given its approved clinical safety, the repurposing of pyronaridine offers a rapidly translational option for treating CE including PAIR. FUND: National Natural Science Foundation of China and International Cooperation Project of the Qinghai Science and Technology Department.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Weisi Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Key Laboratory of Parasitology and Vector Biology of the Chinese Ministry of Health, Shanghai 200025, China
| | - Junmin Yao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Key Laboratory of Parasitology and Vector Biology of the Chinese Ministry of Health, Shanghai 200025, China
| | - Tian Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Shizhu Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Key Laboratory of Parasitology and Vector Biology of the Chinese Ministry of Health, Shanghai 200025, China
| | - Wenjing Qi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Shuai Han
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Key Laboratory of Parasitology and Vector Biology of the Chinese Ministry of Health, Shanghai 200025, China
| | - Yuan Ren
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Zhisheng Dang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Key Laboratory of Parasitology and Vector Biology of the Chinese Ministry of Health, Shanghai 200025, China
| | - Xiumin Han
- Qinghai Provincial People's Hospital, Xining 810007, China
| | - Gang Guo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Baoping Guo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Liqin Wang
- Biotechnology Research Institute, Xinjiang Academy of Animal Science, Urumqi 830000, China
| | - Liping Duan
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Key Laboratory of Parasitology and Vector Biology of the Chinese Ministry of Health, Shanghai 200025, China; Qinghai Provincial People's Hospital, Xining 810007, China.
| | - Wenbao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China.
| |
Collapse
|
32
|
Sepúlveda-Crespo D, Reguera RM, Rojo-Vázquez F, Balaña-Fouce R, Martínez-Valladares M. Drug discovery technologies: Caenorhabditis elegans as a model for anthelmintic therapeutics. Med Res Rev 2020; 40:1715-1753. [PMID: 32166776 DOI: 10.1002/med.21668] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/10/2019] [Accepted: 02/26/2020] [Indexed: 12/16/2022]
Abstract
Helminthiasis is one of the gravest problems worldwide. There is a growing concern on less available anthelmintics and the emergence of resistance creating a major threat to human and livestock health resources. Novel and broad-spectrum anthelmintics are urgently needed. The free-living nematode Caenorhabditis elegans could address this issue through automated high-throughput technologies for the screening of large chemical libraries. This review discusses the strong advantages and limitations for using C elegans as a screening method for anthelmintic drug discovery. C elegans is the best model available for the validation of novel effective drugs in treating most, if not all, helminth infections, and for the elucidation the mode of action of anthelmintic candidates. This review also focuses on available technologies in the discovery of anthelmintics published over the last 15 years with particular attention to high-throughput technologies over conventional screens. On the other hand, this review highlights how combinatorial and nanomedicine strategies could prolong the use of anthelmintics and control resistance problems.
Collapse
Affiliation(s)
- Daniel Sepúlveda-Crespo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Francisco Rojo-Vázquez
- Instituto de Ganadería de Montaña (CSIC-Universidad de León), León, Spain.,Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, León, Spain
| | - María Martínez-Valladares
- Instituto de Ganadería de Montaña (CSIC-Universidad de León), León, Spain.,Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| |
Collapse
|
33
|
Ritler D, Rufener R, Li JV, Kämpfer U, Müller J, Bühr C, Schürch S, Lundström-Stadelmann B. In vitro metabolomic footprint of the Echinococcus multilocularis metacestode. Sci Rep 2019; 9:19438. [PMID: 31857639 PMCID: PMC6923418 DOI: 10.1038/s41598-019-56073-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/04/2019] [Indexed: 12/17/2022] Open
Abstract
Alveolar echinococcosis (AE) is a zoonotic disease that is deadly if left untreated. AE is caused by the larval metacestode stage of the cestode Echinococcus multilocularis. Better knowledge on the host-parasite interface could yield novel targets for improvement of the treatment against AE. We analyzed culture media incubated with in vitro grown E. multilocularis metacestodes by 1H nuclear magnetic resonance spectroscopy to identify the unknown metabolic footprint of the parasite. Moreover, we quantitatively analyzed all amino acids, acetate, glucose, lactate, and succinate in time-course experiments using liquid chromatography and enzymatic assays. The E. multilocularis metacestodes consumed glucose and, surprisingly, threonine and produced succinate, acetate, and alanine as major fermentation products. The metabolic composition of vesicle fluid (VF) from in vitro grown E. multilocularis metacestodes was different from parasite-incubated culture medium with respect to the abundance, but not the spectrum, of metabolites, and some metabolites, in particular amino acids, accumulated in the VF. Overall, this study presents the first characterization of the in vitro metabolic footprint of E. multilocularis metacestodes and VF composition, and it provides the basis for analyses of potentially targetable pathways for future drug development.
Collapse
Affiliation(s)
- Dominic Ritler
- Institute of Parasitology, Department of Infectious Disease and Pathobiology, Vetsuisse Bern, University of Bern, Bern, Switzerland
| | - Reto Rufener
- Institute of Parasitology, Department of Infectious Disease and Pathobiology, Vetsuisse Bern, University of Bern, Bern, Switzerland
| | - Jia V Li
- Division of Systems and Digestive Medicine, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Urs Kämpfer
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Joachim Müller
- Institute of Parasitology, Department of Infectious Disease and Pathobiology, Vetsuisse Bern, University of Bern, Bern, Switzerland
| | - Claudia Bühr
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Stefan Schürch
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Britta Lundström-Stadelmann
- Institute of Parasitology, Department of Infectious Disease and Pathobiology, Vetsuisse Bern, University of Bern, Bern, Switzerland.
| |
Collapse
|
34
|
Enkai S, Inaoka DK, Kouguchi H, Irie T, Yagi K, Kita K. Mitochondrial complex III in larval stage of Echinococcus multilocularis as a potential chemotherapeutic target and in vivo efficacy of atovaquone against primary hydatid cysts. Parasitol Int 2019; 75:102004. [PMID: 31678356 DOI: 10.1016/j.parint.2019.102004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022]
Abstract
Echinococcus multilocularis employs aerobic and anaerobic respiration pathways for its survival in the specialized environment of the host. Under anaerobic conditions, fumarate respiration has been identified as a promising target for drug development against E. multilocularis larvae, although the relevance of oxidative phosphorylation in its survival remains unclear. Here, we focused on the inhibition of mitochondrial cytochrome bc1 complex (complex III) and evaluated aerobic respiratory activity using mitochondrial fractions from E. multilocularis protoscoleces. An enzymatic assay revealed that the mitochondrial fractions possessed NADH-cytochrome c reductase (mitochondrial complexes I and III) and succinate-cytochrome c reductase (mitochondrial complexes II and III) activities in the aerobic pathway. Enzymatic analysis showed that atovaquone, a commercially available anti-malarial drug, inhibited mitochondrial complex III at 1.5 nM (IC50). In addition, culture experiments revealed the ability of atovaquone to kill protoscoleces under aerobic conditions, but not under anaerobic conditions, indicating that protoscoleces altered their respiration system to oxidative phosphorylation or fumarate respiration depending on the oxygen supply. Furthermore, combined administration of atovaquone with atpenin A5, a quinone binding site inhibitor of complex II, completely killed protoscoleces in the culture. Thus, inhibition of both complex II and complex III was essential for strong antiparasitic effect on E. multilocularis. Additionally, we demonstrated that oral administration of atovaquone significantly reduced primary alveolar hydatid cyst development in the mouse liver, compared with the untreated control, indicating that complex III is a promising target for development of anti-echinococcal drug.
Collapse
Affiliation(s)
- Shigehiro Enkai
- School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Daniel Ken Inaoka
- School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Host-Defense Biochemistry, Institute of tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Hirokazu Kouguchi
- Department of Infectious Diseases, Hokkaido Institute of Public Health, N19 W12, Kita-Ku, Sapporo, Hokkaido 060-0819, Japan
| | - Takao Irie
- Department of Infectious Diseases, Hokkaido Institute of Public Health, N19 W12, Kita-Ku, Sapporo, Hokkaido 060-0819, Japan
| | - Kinpei Yagi
- Department of Infectious Diseases, Hokkaido Institute of Public Health, N19 W12, Kita-Ku, Sapporo, Hokkaido 060-0819, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Host-Defense Biochemistry, Institute of tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
35
|
Lundström-Stadelmann B, Rufener R, Ritler D, Zurbriggen R, Hemphill A. The importance of being parasiticidal… an update on drug development for the treatment of alveolar echinococcosis. Food Waterborne Parasitol 2019; 15:e00040. [PMID: 32095613 PMCID: PMC7034016 DOI: 10.1016/j.fawpar.2019.e00040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/06/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023] Open
Abstract
The lethal disease alveolar echinococcosis (AE) is caused by the metacestode stage of the fox tapeworm Echinococcus multilocularis. Current chemotherapeutical treatment of AE relies on albendazole and mebendazole, with the caveat that these compounds are not parasiticidal. Drugs have to be taken for a prolonged period of time, often life-long, which can cause adverse effects and reduces the patients' quality of life. In some individuals, benzimidazoles are inactive or cause toxicity, leading to treatment discontinuation. Alternatives to benzimidazoles are urgently needed. Over the recent years, in vivo and in vitro models for low-to-medium throughput drug discovery against AE have been set in place. In vitro drug tests include the phosphoglucose-isomerase (PGI) assay to measure physical damage induced to metacestodes, and viability assays to assess parasiticidal activity against metacestodes and stem cells. In vitro models are also employed for studies on mechanisms of action. In vivo models are thus far based on rodents, mainly mice, and benefits could be gained in future by comparative approaches in naturally infected dogs or captive monkeys. For the identification of novel drugs against AE, a rare disease with a low expected market return, drug-repurposing is the most promising strategy. A variety of chemically synthesized compounds as well as natural products have been analyzed with respect to in vitro and/or in vivo activities against AE. We here review and discuss the most active of these compounds including anti-infective compounds (benzimidazoles, nitazoxanide, amphotericin B, itraconazole, clarithromycin, DB1127, and buparvaquone), the anti-infective anti-malarials (artemisinin, ozonids, mefloquine, and MMV665807) and anti-cancer drugs (isoflavones, 2-methoxyestradiol, methotrexate, navelbine, vincristine, kinase inhibitors, metallo-organic ruthenium complexes, bortezomib, and taxanes). Taking into account the efficacy as well as the potential availability for patients, the most promising candidates are new formulations of benzimidazoles and mefloquine. Future drug-repurposing approaches should also target the energy metabolism of E. multilocularis, in particular the understudied malate dismutation pathway, as this offers an essential target in the parasite, which is not present in mammals. Benzimidazoles are used to treat AE, but new drugs are needed. New drugs against AE can be identified by drug repurposing. Drugs against other infectious diseases and cancer can be repurposed against AE. Most promising are new formulations of benzimidazoles and mefloquine. Future approaches should include targeting the energy metabolism of the parasite.
Collapse
Affiliation(s)
- Britta Lundström-Stadelmann
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Reto Rufener
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Dominic Ritler
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Raphael Zurbriggen
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| |
Collapse
|