1
|
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: 0] [Impact Index Per Article: 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
|
2
|
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
|
3
|
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
|
4
|
Lu Z, Wang Y, Liu C, Fan H. Efficacy and Safety of Asparagusic Acid against Echinococcus multilocularis In Vitro and in a Murine Infection Model. Trop Med Infect Dis 2024; 9:110. [PMID: 38787043 PMCID: PMC11126102 DOI: 10.3390/tropicalmed9050110] [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: 04/02/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Alveolar echinococcosis (AE) stands as a perilous zoonotic affliction caused by the larvae of Echinococcus multilocularis. There is an imperative need to explore novel therapeutic agents or lead compounds for the treatment of AE. Asparagusic acid, characterized by its low toxicity and possessing antimicrobial, antioxidant, and anti-parasitic attributes, emerges as a promising candidate. The aim of this study was to investigate the in vivo and in vitro efficacy of asparagusic acid against E. multilocularis. Morphological observations, scanning electron microscopy, ROS assays, mitochondrial membrane potential assays, and Western blot were used to evaluate the in vitro effects of asparagusic acid on protoscoleces. The effects of asparagusic acid on vesicles were assessed via PGI release, γ-GGT release, and transmission electron microscopy observations. CellTiter-Glo assays, Caspase3 activity assays, flow cytometry, and Western blot were used for an evaluation of the effect of asparaginic acid on the proliferation and apoptosis of germinal cells. The in vivo efficacy of asparagusic acid was evaluated in a murine AE model. Asparagusic acid exhibited a pronounced killing effect on the protoscoleces post-treatment. Following an intervention with asparagusic acid, there was an increase in ROS levels and a decline in mitochondrial membrane potential in the protoscolex. Moreover, asparagusic acid treatment resulted in the upregulation of PGI and γ-GGT release in metacestode vesicles, concomitant with the inhibition of germinal cell viability. Furthermore, asparagusic acid led to an enhanced relative expression of Caspase3 in the culture supernatant of both the protoscoleces and germinal cells, accompanied by an increase in the proportion of apoptotic germinal cells. Notably, asparagusic acid induced an augmentation in Bax and Caspase3 protein expression while reducing Bcl2 protein expression in both the protoscoleces and germinal cells. In vitro cytotoxicity assessments demonstrated the low toxicity of asparagusic acid towards normal human hepatocytes and HFF cells. Additionally, in vivo experiments revealed that asparagusic acid administration at doses of 10 mg/kg and 40 mg/kg significantly reduced metacestode wet weight. A histopathological analysis displayed the disruption of the germinal layer structure within lesions post-asparagusic acid treatment, alongside the preservation of laminated layer structures. Transmission electron microscopy further revealed mitochondrial swelling and heightened cell necrosis subsequent to the asparagusic acid treatment. Furthermore, asparagusic acid promoted Caspase3 and Bax protein expression while decreasing Bcl2 protein expression in perilesional tissues. Subsequently, it inhibited the expression of Ki67, MMP2, and MMP9 proteins in the perilesional tissues and curbed the activation of the PI3K/Akt signaling pathway within the lesion-host microenvironmental tissues. Asparagusic acid demonstrated a pronounced killing effect on E. multilocularis, suggesting its potential as a promising therapeutic agent for the management of AE.
Collapse
Affiliation(s)
- Zhuanhong Lu
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
| | - Yating Wang
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
| | - Chuanchuan Liu
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
- Qinghai Key Laboratory of Echinococcosis Research, Xining 810001, China
| | - Haining Fan
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
- Qinghai Key Laboratory of Echinococcosis Research, Xining 810001, China
| |
Collapse
|
5
|
Yang Y, Wuren T, Wu B, Cheng S, Fan H. The expression of CTLA-4 in hepatic alveolar echinococcosis patients and blocking CTLA-4 to reverse T cell exhaustion in Echinococcus multilocularis-infected mice. Front Immunol 2024; 15:1358361. [PMID: 38605966 PMCID: PMC11007148 DOI: 10.3389/fimmu.2024.1358361] [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: 12/19/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Alveolar echinococcosis (AE) is a zoonotic parasitic disease caused by the infection of Echinococcus multilocularis (E. multilocularis) larvae. Cytotoxic T-lymphocyte antigen 4 (CTLA-4) produces inhibitory signals and induces T cell exhaustion, thereby inhibiting the parasiticidal efficacy of the liver immune system. Therefore, the purpose of this study is to explore how T-cell exhaustion contributes to AE and whether blocking CTLA-4 could reverse T cell exhaustion. Here we discovered that the expression of CTLA-4 was increased in the infiltrating margin around the lesion of the liver from AE patients by using western blot and immunohistochemistry assay. Multiple fluorescence immunohistochemistry identified that CTLA-4 and CD4/CD8 molecules were co-localized. For in vitro experiments, it was found that the sustained stimulation of E. multilocularis antigen could induce T cell exhaustion, blocking CTLA-4-reversed T cell exhaustion. For in vivo experiments, the expression of CTLA-4 was increased in the liver of E. multilocularis-infected mice, and the CTLA-4 and CD4/CD8 molecules were co-localized. Flow cytometry analysis demonstrated that the percentages of both CD4+ T cells and CD8+ T cells in the liver and peripheral blood were significantly increased and induced T exhaustion. When the mice were treated with anti-CTLA-4 antibodies, the number and weight of the lesions decreased significantly. Meanwhile, the flow cytometry results suggested that blocking CTLA-4 could effectively reverse T cell exhaustion and reactivate immune function. Our work reveals that blocking CTLA-4 could effectively reverse the T cell exhaustion caused by E. multilocularis and could be used as a novel target for the treatment of AE.
Collapse
Affiliation(s)
- Yuxuan Yang
- Research Center for High Altitude Medicine, Qinghai University, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Xining, Qinghai, China
- Qinghai Research Key Laboratory for Echinococcosis, Qinghai University, Xining, Qinghai, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Xining, Qinghai, China
| | - Binjie Wu
- Research Center for High Altitude Medicine, Qinghai University, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Xining, Qinghai, China
- Qinghai Research Key Laboratory for Echinococcosis, Qinghai University, Xining, Qinghai, China
| | - Shilei Cheng
- Research Center for High Altitude Medicine, Qinghai University, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Xining, Qinghai, China
- Qinghai Research Key Laboratory for Echinococcosis, Qinghai University, Xining, Qinghai, China
| | - Haining Fan
- Qinghai Research Key Laboratory for Echinococcosis, Qinghai University, Xining, Qinghai, China
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| |
Collapse
|
6
|
Zhou Z, Huayu M, Mu Y, Tang F, Ge RL. Ubenimex combined with Albendazole for the treatment of Echinococcus multilocularis-induced alveolar echinococcosis in mice. Front Vet Sci 2024; 11:1320308. [PMID: 38585297 PMCID: PMC10995866 DOI: 10.3389/fvets.2024.1320308] [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: 10/27/2023] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Alveolar echinococcosis (AE) is a parasitic disease caused by E. multilocularis metacestodes and it is highly prevalent in the northern hemisphere. We have previously found that vaccination with E. multilocularis-Leucine aminopeptidase (EM-LAP) could inhibit the growth and invasion of E. multilocularis in host liver, and Ubenimex, a broad-spectrum inhibitor of LAP, could also inhibit E. multilocularis invasion but had a limited effect on the growth and development of E. multilocularis. Methods In this study, the therapeutic effect of Ubenimex combined with Albendazole on AE was evaluated. Mice were intraperitoneally injected with protoscoleces and imaging examination was performed at week 8 and week 16 to detect cyst change. During this period, mice were intraperitoneally injected with Ubenimex and intragastrically administered with Albendazole suspension. At last, the therapeutic effect was evaluated by morphological and pathological examination and liver function. Results The results revealed that the combined treatment could inhibit the growth and infiltration of cysts in BALB/c mice infected with E. multilocularis protoscoleces. The weight, number, invasion and fibrosis of cysts were reduced in mice treated with Ubenimex in combination with Albendazole. The same effect was achieved by the single Ubenimex treatment because of its inhibitory effect on LAP activity, but it was less effective in inhibiting the growth of cysts. The levels of ALT, AST, TBIL, DBIL, ALP, and γ-GT were reduced after the combined treatment, indicating that treatment with both Ubenimex and Albendazole could alleviate liver damage. Discussion This study suggests that the combined treatment with Ubenimex and Albendazole could be a potential therapeutic strategy for E. multilocularis infections.
Collapse
Affiliation(s)
- Zhen Zhou
- Research Center for High Altitude Medicine of Qinghai University, Xining, Qinghai, China
- Key Laboratory of High Altitude Medicine in Qinghai Provincial, Qinghai University, Xining, Qinghai, China
| | - Meiduo Huayu
- Research Center for High Altitude Medicine of Qinghai University, Xining, Qinghai, China
- Key Laboratory of High Altitude Medicine in Qinghai Provincial, Qinghai University, Xining, Qinghai, China
| | - Yalin Mu
- Department of Medical Imaging Center, Qinghai University Affiliated Hospital, Xining, Qinghai, China
| | - Feng Tang
- Research Center for High Altitude Medicine of Qinghai University, Xining, Qinghai, China
- Key Laboratory of High Altitude Medicine in Qinghai Provincial, Qinghai University, Xining, Qinghai, China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine of Qinghai University, Xining, Qinghai, China
- Key Laboratory of High Altitude Medicine in Qinghai Provincial, Qinghai University, Xining, Qinghai, China
| |
Collapse
|
7
|
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
|
8
|
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: 0] [Impact Index Per Article: 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
|
9
|
Zhou Z, Zhou P, Mu Y, Wang L, Cao Z, Dong S, Bao H, Yang B, Xin M, Li R, Ge RL, Tang F. Therapeutic effect on Alveolar echinococcosis by targeting EM-Leucine aminopeptidase. Front Immunol 2022; 13:1027500. [PMID: 36311709 PMCID: PMC9614657 DOI: 10.3389/fimmu.2022.1027500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Alveolar echinococcosis (AE) is a parasitic disease caused by E. multilocularis metacestodes and it is highly prevalent in the northern hemisphere. We have previously found that vaccination with E. multilocularis Leucine aminopeptidase (EM-LAP) induced specific immune response and had an inhibiting effect on the parasites. In this study, the therapeutic effect of recombinant EM-LAP (rEM-LAP) on AE was evaluated and verified using Ubenimex, a broad-spectrum inhibitor of LAP. The results reveal that rEM-LAP could inhibit cyst growth and invasion and induce specific immunity response in BALB/c mice infected with E. multilocularis protoscoleces. The ultrasonic, MRI, and morphological results show that treatment with rEM-LAP inhibits E. multilocularis infection and reduces cyst weight, number, fibrosis and invasion. The same effect is observed for the treatment with Ubenimex by inhibiting LAP activity. The indirect ELISA shows that rEM-LAP could induce specific immunity response and produce high levels of IgG, IgG1, IgG2a, IgM, and IgA, and the serum levels of IFN-γ and IL-4 are significantly increased compared to the control groups, indicating that treatment with rEM-LAP leads to a Th1 and Th2 mixed-type immune response. This study suggests that EM-LAP could be a potential therapeutic target of E. multilocularis infection.
Collapse
Affiliation(s)
- Zhen Zhou
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Plateau Medical Application, Key Laboratory of Ministry of Education, Qinghai University, Xining, China
| | - Pei Zhou
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Plateau Medical Application, Key Laboratory of Ministry of Education, Qinghai University, Xining, China
| | - Yalin Mu
- Department of Medical Imaging Center, Qinghai University Affiliated Hospital, Xining, China
| | - Lei Wang
- Department of Pathology, The Second Xiangya Hospital DE Central South University, Changsha, China
| | - Zhenjin Cao
- Department of Medical Imaging Center, Qinghai University Affiliated Hospital, Xining, China
| | - Shizhong Dong
- Department of Medical Imaging Center, Qinghai University Affiliated Hospital, Xining, China
| | - Haihua Bao
- Department of Medical Imaging Center, Qinghai University Affiliated Hospital, Xining, China
| | - Baoliang Yang
- Department of ENT, Qinghai Red Cross Hospital, Xining, China
| | - Minyuan Xin
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Plateau Medical Application, Key Laboratory of Ministry of Education, Qinghai University, Xining, China
| | - Runle Li
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Plateau Medical Application, Key Laboratory of Ministry of Education, Qinghai University, Xining, China
- *Correspondence: Runle Li, ; Ri-Li Ge, ; Feng Tang,
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Plateau Medical Application, Key Laboratory of Ministry of Education, Qinghai University, Xining, China
- *Correspondence: Runle Li, ; Ri-Li Ge, ; Feng Tang,
| | - Feng Tang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Plateau Medical Application, Key Laboratory of Ministry of Education, Qinghai University, Xining, China
- *Correspondence: Runle Li, ; Ri-Li Ge, ; Feng Tang,
| |
Collapse
|