Skip to main content

MINI REVIEW article

Front. Microbiol., 08 August 2022
Sec. Infectious Agents and Disease
This article is part of the Research Topic Advances and Challenges in the Detection and Treatment of Pathogenic Microorganisms in Infectious Disease Control View all 25 articles

Advances in the pharmacological treatment of hepatic alveolar echinococcosis: From laboratory to clinic

Xiaolei Xu,,,&#x;Xiaolei Xu1,2,3,4Xinye Qian,&#x;Xinye Qian2,4Cancan Gao&#x;Cancan Gao5Yuan Pang,&#x;Yuan Pang6,7Hu Zhou,,&#x;Hu Zhou1,2,3Lizhen Zhu,Lizhen Zhu1,3Zhan Wang,,Zhan Wang1,2,3Mingquan Pang,Mingquan Pang1,3Defang Wu,Defang Wu1,3Wenhao Yu,Wenhao Yu1,3Fanyu Kong,Fanyu Kong1,3Dalin Shi,Dalin Shi1,3Yuting Guo,,Yuting Guo2,6,7Xiaoxia Su,Xiaoxia Su1,3Wang Hu,Wang Hu2,4Jun YanJun Yan4Xiaobin Feng
Xiaobin Feng4*Haining Fan,
Haining Fan1,3*
  • 1Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qinghai University, Xining, Qinghai, China
  • 2School of Clinical Medicine, Tsinghua University, Beijing, China
  • 3Qinghai Research Key Laboratory for Echinococcosis, Qinghai, China
  • 4Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, Beijing, China
  • 5Department of General Medicine of Air Force Medical Center, Beijing, China
  • 6Department of Mechanical Engineering, Tsinghua University, Beijing, China
  • 7Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China

Hepatic alveolar echinococcosis (HAE) is a zoonotic parasitic disease caused by the larvae of Echinococcus multilocularis. Because of its characteristics of diffuse infiltration and growth similar to tumors, the disability rate and mortality rate are high among patients. Although surgery (including hepatectomy, liver transplantation, and autologous liver transplantation) is the first choice for the treatment of hepatic alveolar echinococcosis in clinic, drug treatment still plays an important and irreplaceable role in patients with end-stage echinococcosis, including patients with multiple organ metastasis, patients with inferior vena cava invasion, or patients with surgical contraindications, etc. However, Albendazole is the only recommended clinical drug which could exhibit a parasitostatic rather than a parasitocidal effect. Novel drugs are needed but few investment was made in the field because the rarity of the cases. Drug repurposing might be a solution. In this review, FDA-approved drugs that have a potential curative effect on hepatic alveolar echinococcosis in animal models are summarized. Further, nano drug delivery systems boosting the therapeutic effect on hepatic alveolar echinococcosis are also reviewed. Taken together, these might contribute to the development of novel strategy for advanced hepatic alveolar echinococcosis.

Introduction

Hepatic echinococcosis (HE) is a zoonotic parasitic disease that seriously endangers people’s health and socio-economic development worldwide, especially in some rural area of China (McManus et al., 2003; Yang et al., 2017). The annual treatment costs and livestock losses associated with the disease are expected to be $3 billion (Kern et al., 2017), which has become a major global public health problem. In 2010, the World Health Organization listed it as one of the 17 neglected diseases that should be controlled or eliminated by 2050. 91% of the new cases in the world come from China each year (Qian et al., 2017).

There are mainly two kinds of echinococcosis: hepatic cystic echinococcosis (HCE) and hepatic alveolar echinococcosis (HAE). HE is caused by the parasite passing through the intestinal mucosa and then entering in the liver through the portal vein system. HAE only accounts for 5–10% of the overall incidence rate of echinococcosis (Akbulut et al., 2014; Akbulut, 2018). Although it is a benign disease, its growth mode is like a malignant disease (Parsak et al., 2007). HAE could not only infiltrate into surrounding tissues such as gallbladder, blood vessels, or biliary system, but also distal organs, such as kidney, lung, brain, bone, etc. (Hatipoglu et al., 2013; Joliat et al., 2015). Although surgery is the ideal treatment for HAE, patients are constantly diagnosed at the advanced stage of the disease as the initial symptoms can be sub-clinical. Also, lack of health knowledge and medical resources makes the problem worse. As a result, many HAE patients lose the opportunity for surgery, leaving medication the only option for these patients.

Mebendazole and Albendazole are mainly used to treat advanced HAE. Study (Davis et al., 1989; El-On, 2003) showed that albendazole is more efficient than mebendazole in the treatment of HAE. Albendazole can inhibit the uptake of glucose, deplete the glycogen of the germinal layer cells, denature the endoplasmic reticulum bodies and mitochondria, increase the lysosomes, and finally lead to the death of the parasite. However, albendazole exhibit a parasitostatic rather than a parasitocidal effect. The plasma concentration of albendazole was only (1.86 ± 0.88) mg/L, while the drug concentration in hydatid lesions was only 1/100 of the plasma concentration, which was far lower than the concentration required to kill multilocular Echinococcus multilocularis. Further, advanced HAE cases must undergo long-term chemotherapy (often life-long), which could result in adverse complications such as abnormal liver function, leukopenia, and hair loss (Meilinger et al., 2013). Therefore, recurrence rates after interruption of therapy are relatively high (Reuter et al., 2004). In addition, long-term drug use is easy to cause drug resistance of parasites (Pawluk et al., 2015), also resulting in the progression of the disease. As a result, effective novel drugs are needed for the disease.

Dilemma

For many years, the pharmaceutical industry has not been compelled to develop novel drugs against HAE due to its relatively rare abundance, investments in the development of new drugs against AE will not result in a high market return, resulting in Low investment of the area. Also, the rarity of the cases would make it almost impossible to conduct clinical trials like randomized control trial, or cohort study, etc. So, even there is a new drug for HAE, it would take years to investigate its efficiency and safety in HAE patients. Up to date, Mebendazole and Albendazole are still the only options for these patients.

Drug repurposing or studying existing drugs for potential therapeutic utility in newer indications has been identified as an attractive option for treating a number of diseases (Polamreddy and Gattu, 2019), which might be a solution for this problem. Drug repurposing offers manifold advantages over de novo drug discovery and development process such as expedited and economical drug development process (Breckenridge and Jacob, 2019), curtailing the development risks as safety of the compound, a major reason for high attrition rates in drug development process, is already well established (Gupta et al., 2013). Due to huge promise offered by drug repurposing compared to conventional drug development process, this strategy is being adopted by many pharmaceutical companies to redevelop their approved and shelved molecules as novel therapeutic options in a wide range of indications (Neuberger and Oraiopoulos, 2019). Four pillars were identified to assure the success rate in drug repurposing programs (Mittal and Mittal, 2021). Pillar 1: drug pharmacology – sound knowledge of the repurposed drug’s pharmacological characteristics; pillar 2: drug formulation – drug formulation considerations in new indication; pillar 3: evaluation in biological assays – evaluation in representative biological assays with translational potential; pillar 4: clinical evaluation – robust clinical trial methodologies including biomarker-driven approach to provide conclusive evidence of repurposed drug’s efficacy in new indication.

Excitingly, lab experiments have showed that many FDA-approved drugs might have therapeutic effects on HAE. Many of the studies have provided Pillar 1, 2, or 3 evidence for drug repurposing, which might bring new opportunity for advanced HAE patients. In addition, these drugs could be used with Albendazole simultaneously in HAE patients. With these evidence, clinical trials could be enrolling advanced HAE patients for experimental treatment to improve outcome of these patients. In this review, the details of these potential drugs are being reviewed and summarized.

Recent advances in drugs from lab

Antibiotic drugs

Mefloquine, the famous antimalarial drug (Lundström-Stadelmann et al., 2020), showed a significant effect on reduction in parasite growth in mice infected with multilocular Echinococcus intraperitoneally compared with the standard treatment with albendazole (to assess the viability of the parasite, mefloquine-treated metacestodes from in vitro cultures were injected into Balb/c mice. No parasite growth was observed after 5 months of incubation), although its exact role as a drug target remains to be clarified. Consider that HAE is not only a parasitic disease, but also a infectious disease. It is reported that (Mai et al., 2011) broad-spectrum anti-infective drug - nitazonit (nitazoxanide) also showed its effect in killing the parasites. In vitro experiment, no regrowth of cysts was observed even after 3 weeks, 3 months, or 6 months of nitazoxanide and albendazole administration. In vivo test, no metacestodes of the hydatid were found in mice treated by nitazoxanide. In addition, amphotericin B has been proved to effectively inhibit the growth of Echinococcus by using the in vitro culture model. When amphotericin B was added to metacestode tissue after 10 weeks of culture, destruction of vesicles was observed as soon as 1 day after the start of treatment, and all vesicles were disrupted after 8 days (Reuter et al., 2003). The antifungal drug amphotericin B has been applied to patients as a remedial treatment, but it is not an insecticidal drug, and long-term use will cause nephrotoxicity.

Protease inhibitor

Hepatic alveolar echinococcosis, similar to tumors, proliferates by budding or infiltrating into tissues, and constantly promoting angiogenesis. It has been suggested that the unlimited proliferation ability of Echinococcus may be related to some signal transduction protein kinase molecules (Siles-Lucas et al., 2001). As a phosphoserine/phosphothreonine binding module, 14-3-3 protein is involved in the process of DNA damage checkpoint and prevention of apoptosis (Wilker and Yaffe, 2004). In fact, the 14-3-3 cDNA sequence of Echinococcus was compared with the 14-3-3 isomer of other organisms, and the parasite sequence was classified as the isomer related to tumor growth (Spiliotis et al., 2005). Britta Stadelmann et al. screened a library containing 426 FDA-approved drugs (Stadelmann et al., 2014). The study showed that bortezomib (BTZ), a proteasome inhibitor, could cause parasite death in vitro model. The animal model also showed that the parasite weight was reduced by an average of 2 g under a BTZ dosage of 0.5 mg/kg combined with ABZ, proving that bortezomib can be used as a drug target in multilocular Echinococcus. Moreover, GTPases Ras and RAF, and genes encoding epidermal growth factor receptors, were found in Echinococcus Taenia (Spiliotis et al., 2003). RhoA, a protein with GTPase activity in Ras superfamily, activates target proteins by binding to GTP. RhoA could activate RAF, which is an upstream protein of MAPK signaling pathway (Kontaridis et al., 2008). Activated Raf further activates MEK, which is responsible for ERK and JNK phosphorylation (Ke et al., 2019). Activated ERK and JNK induce macrophage polarization by activating certain transcription factors (Weiss et al., 2018). MAPK signaling pathway is crucial for parasitic infection (Zhao et al., 2019). Shigui Chong et al. found that soluble antigen of Echinococcus multilocularis induces macrophage polarization after alveolar echinococcosis infection through RhoA MAPK signal pathway (Chong et al., 2022). Emmpk1, a host extracellular signal regulated kinase (ERK), could activate parasitic MAPK cascade as it could be phosphorylated by multilocular Echinococcus cysts in vitro (Hemphill et al., 2002; Spiliotis et al., 2006). MAPK signaling pathways, including c-Jun N-terminal kinase (JNK), p38 MAPK and ERK, play an important role in signal transduction from cell membrane to nuclear transcription factors, balancing cell survival and death in liver injury (Ballif and Blenis, 2001). Ren Yong Lin et al. reported multilocular echinococcosis could directly affect hepatocyte proliferation via MAPK pathway (Lin et al., 2009). The team also reported that MAPK signaling pathway was upregulated by microarray analysis in mice infected with Echinococcus multilocularis (Lin et al., 2011). According to these findings, MAPK pathway could be a therapeutic target for HAE.

MAPK pathway inhibitors have been applied in clinic for decades. Sorafenib is a small molecule anticancer drug targeting RAF in Raf/ERK signaling pathway to inhibit tumor cell proliferation and angiogenesis by targeting tyrosine kinases, such as intravascular growth factor receptors VEGFR-2 and VEGFR-3 and platelet-derived growth factor receptor (PDGFR). Xiao explored the efficacy of different concentrations of anti-cancer drug sorafenib in multilocular echinococcosis through animal models. The results showed that sorafenib showed strong cyst inhibition effect and was a potential drug for the treatment of echinococcosis (the cyst inhibitory rate of sorafenib was 6.6, 42.4, 68.5, 77.4, 84.0 and 89.5% at 10 μmol/l, while the cyst inhibitory rate of albendazole was 3.8, 12.7, 27.0, 51.4, 54.0, and 73.0% at 30 μmol/l after 48, 72, 96, 120, 144, and 168 h of administration; Xiao, 2021).

Hemer and Brehm also focused on parasite signaling pathways when looking for new drug targets. It was found that imatinib, a TKI tyrosine kinase inhibitor, has the ability to efficiently kill the protoscolex of Echinococcus in vitro. In addition, imatinib could significantly inhibit the formation of prototapeworm vesicles by parasitic stem cells at concentrations as low as 10 m. After 7 days of treatment, 50% of the vesicles lose activity and induce morphological changes of tapeworm. Therefore, imatinib might become a promising alternative to albendazole in anti-echinococcosis chemotherapy (Hemer and Brehm, 2012). Although little is known about the signal transduction pathway, growth factors and tyrosine kinase signaling molecules might play a vital role in the process of worm differentiation and reproductive activity, which might be potential targets for new drug treatment of echinococcosis. Recent progress in the characterization of TKI and signals confirms the importance of tyrosine kinase activation pathway in larvae (Brehm et al., 2006). Based on these facts, TKI inhibitors could be potential strategies for treating echinococcosis (Dissous et al., 2007).

Immune checkpoint inhibitor

Echinococcus can parasitize in the host for a long time without being cleared by the body’s immune system is because the antigen produced by Echinococcus after entering the host can regulate the body’s immune microenvironment through a variety of ways (Zhang et al., 2016), including escaping the attack of host immune cells, and inducing the immunosuppression environment. It is found that the immune microenvironment is of great significance in the occurrence and development of a variety of diseases, and the change of immune environment also provides a suitable environment for the survival and reproduction of multilocular Echinococcus (Bakhtiar et al., 2020). Multiple evidence show that the hepatic inflammatory microenvironment is crucial in the development of hepatic echinococcosis (Gottstein et al., 2017).

The early stage of animal infection is characterized by Th1/Th2 mixed immune response, which is accompanied with an increase in interferon-γ (IFN-γ), interleukin-4 (IL-4), and related chemokines. In human HAE, CD4+/CD25+ regulatory cells (Tregs) play a major role in regulating immune response and seem to be upregulated in the time course of the disease, which might be related to the suppression of the immune response to specific antigens and promotion of the secretion of anti-inflammatory cytokines like interleukin-10 (IL-10), transforming growth factor β (TGF- β), etc. (Vuitton et al., 2006). Studies showed that prolonged exposure to IL-10 could lead to T cell dysfunction (Monaghan et al., 2012). Similarly, TGF-β also could reduce the intensity of T cell response and induce T cell apoptosis, which is related to PD-1 signaling pathway (Ma et al., 2013). Xiaolin La et al. explored the relationship between PD-1 / PD-L1 pathway and Tregs in different stages of multilocular Echinococcus infection. The expression of PD-1 and PD-L1 increased with the development of HAE erosion. The results also indicated that high expression of PD-1/PD-L1 might play an important role in stimulating CD4+/CD25+ T cells, maintaining peripheral tolerance and immune escape during chronic infection of Echinococcus (La et al., 2015). Junhua Wang et al. evaluated the effect of regulatory T cell (Treg) deficiency on the growth of Taenia saginata. After Treg deficiency was induced by diphtheria toxin (DT), the parasitic lesions (>4 mm) in the liver of mice infected with multilocular bacteria were significantly smaller than those (<2 mm) in the corresponding control group (Wang et al., 2018).

Anne Pauline Bellanger et al. analyzed the cells and serum of 22 healthy blood donors. After blood samples were stimulated with multilocular Echinococcus, a significant increase in PD-L1 and CTLA-4 was observed (Bellanger et al., 2020), suggesting that PD-L1 pathway plays an important role in the infection of multilocular Echinococcus multilocularis (Wang et al., 2018). Fadi jebbawi et al. started immunotherapy 6 weeks after multilocular E. coli infection and maintained it for 8 weeks (Jebbawi et al., 2021). Mice were treated with albendazole (orally for 5 days / week) and PD-L1 (twice a week intraperitoneally). The results showed that PD-L1 blocker can significantly reduce the size and weight of parasitic lesions by increasing CD4/CD8 effector T cells and reducing Treg, which indicates that PD-1/PD-L1 pathway blocking has great potential in the treatment and control of multilocular Echinococcus infection. Chuanshan Zhang et al. found that TIGIT might also be a potential therapeutic target for HAE as liver weight, lesion weight, lesion area, and confluent lesion numbers were significantly lower in TIGIT blocked mice as compared with control mice (Zhang et al., 2020). Natural killer (NK) cells are active members of innate immunity. More and more studies have proved their importance in liver immunity (Kubes and Jenne, 2018). Although the mechanism of NK cell in HAE is not fully understood, Hao Wen and colleagues explored TIGIT as an important potential biomarker in HAE patients and E. multilocular-infected mice. The lack or blocking of TIGIT partially inhibits the growth of E. multilocular by reversing the damaged function of NK cells. The results suggest that targeting TIGIT may be a potential immunotherapeutic strategy for the treatment of patients with AE (Zhang et al., 2021; Table 1).

TABLE 1
www.frontiersin.org

Table 1. Recent advances in drugs for hepatic echinococcosis.

Chinese herbal medicine

Previous studies have reported the efficacy of Chinese herbal medicine in the treatment of echinococcosis. Liu et al. (2021) have tested the anthelmintic effect of saffron on the pathogenic scolex of Echinococcus both in vitro and in vivo. They found that saffron inhibits the expression of matrix metalloproteinases (MMPs), particularly MMP2 and MMP9, in the host tissue around the tapeworm, which could promote the deposition of collagen in the protoscolex. The results showed that saffron could be developed as a new drug for the treatment of HAE.

Artemisinins have been found to have antiparasitic effect via various mechanisms, such as ROS-dependent depolarization of the membrane (Antoine et al., 2014), or reactive oxygen species mediated DNA damage (Gopalakrishnan and Kumar, 2015), etc. Artesunate is a semi-synthetic derivative of artemisinin. Its water solubility determines its good absorption, high bioavailability, and long half-life in vivo. Meanwhile, it could be prepared into oral, rectal, intramuscular, and intravenous dosage forms, which is convenient for clinical administration. Xiao et al. (2008) proved the inhibitory effect of artesunate on adult Clonorchis sinensis in vivo through a rat model as significant worm burden reductions was observed after artesunate was administrated. Jiraungkoorskul et al. (Jiraungkoorskul et al., 2005) observed, in vitro, that the surface layer of the artesunate-treated Schistosoma japonicum was seriously damaged and its activity decreased. These findings suggested that artemisinins might have an important role in treating HAE through further validation would be needed.

Jiang C et al. have created a self-made pure traditional Chinese medicine “Xiaobao Decoction” to treat hydatidosis in mice. The inhibition rate of alveolar hydatidosis was 65.7% ~ 80.6% (Jiang, 1995), indicating that the traditional Chinese medicine Xiaobao Decoction might be a promising alternative or adjuvant therapy for HAE (Jiang, 1998). The main ingredients of Xiaobao Decoction are areca, Begonia, snake slough, ground beetle, honeycomb, and scales. In order to achieve better anti-echinococcosis effect, various dosage forms of Xiaobao Decoction were also produced, such as Xiaobao capsule, Xiaobao tablet, etc. Jiang CP et al. further explored the therapeutic effect of Xiaobao pill combined with albendazole in the treatment of hydatidosis, founding that the combination caused more serious damage to the ultrastructure of hepatic multilocular echinococcosis than albendazole alone (Jiang, 1991). Clinical data also showed that Xiaobao pill has a good therapeutic effect on hydatidosis (Jiang, 1986; Jiang and Liu, 1994). However, multi-center clinical study with large sample size is still needed to evaluate its antiparasitic effect.

Nano drug delivery system

Although the traditional tablet albendazole (t-abz) has curative effect on hydatid, its clinical cure rate is low. The reason for this is that ABZ is poorly and erratically absorbed following oral administration resulting in low drug levels in plasma and liver distribution. Therefore, there is an urgent need for designing new formulations of ABZ with increased bioavailability to improve the pharmacokinetic effects of drugs. Liposomal chemotherapy are applied in clinics as they could gather in the targeted tissues and release the drug continuously (Yue and Dai, 2018), which could solve the limitation of traditional tablet albendazole. Thus, drug repositioning combined with nanotechnology to improve drug bioavailability becomes a useful, fast, and inexpensive tool for the treatment of neglected diseases. To elevate the low water solubility, poor absorption, and low bioavailability of Albendazole, Rodrigues et al. (1995) established poly (D, L-lactide) nanoparticles loading with ABZ to investigate this new drug delivery system against E. multilocularis using a mouse model of hepatic alveolar echinococcosis. These treatments significantly reduced parasite node surface size as well as peritoneal metastatic burden compared to untreated mice as the concentration of drug was higher at the focus of infection compared with that of the controlled group. Nayer Mehdizad Bakhtiar et al. also showed that ABZ-loaded polymeric nanoparticles (NPs) had a tendency to increase the mortality rate against protoscoleces and microcysts compared with traditional albendazole (Bakhtiar et al., 2019). Pensel et al. suggest that ABZ nanocrystals (ABZ-NCs) seem to be a useful tool to increase bioavailability (Pensel et al., 2018). The preventive efficacy of the ABZ-NC preparation in mice infected with Echinococcus multilocularis was studied, and it was found that the mean weight of vesicles recovered in the ABZ-NC group was 50% lower than in the untreated mice. The survival rate of protospores isolated from ABZ-NC-treated mice was significantly lower than that of the control group (p < 0.05). After treatment with ABZ-NCs, the cyst weight was reduced by 77%, and the survival rate of its pronuclei was reduced to 34%, which is due to the increased oral bioavailability. Chunhui Hu et al. established a novel nanocrystalline (NC) formulation of ABZ by spray drying ABZ with a triblock copolymer poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (Poloxamer 188), and its physical structure was confirmed by scanning electron microscopy (SEM; Hu et al., 2020). The significant reduction in ABZ crystallite size coupled with the prolonged ABZ supersaturation greatly improved the dissolution properties of the drug compared to the commercial ABZ oral product (Albenda), and in a pharmacokinetic comparison, the NC formulation showed an approximately 4.2-fold higher AUC than Albenda, as measured by the plasma concentration of the active antiparasitic metabolite albendazole. More encouragingly, after 30 days of oral administration of NC and Albenda formulations once a day in SD rats with hepatic alveolar echinococcosis, the NC formulation exhibited 3.7 times more vesicle-inhibitory effect than Albenda. Therefore, NC formulations have the potential to be developed as an improved anti-AE drug therapy (Table 2).

TABLE 2
www.frontiersin.org

Table 2. Nano drug delivery system of Albendazole.

As nano vehicle could increase local drug concentration and bioavailability, other anti-HE might also be loaded in these vehicles to achieve better therapeutic effects. Recently, single-dose liposomal amphotericin B combined with flucytosine and fluconazole was proved to be noninferior to the WHO-recommended treatment for HIV-associated cryptococcal meningitis and was associated with fewer adverse events (Jarvis et al., 2022), showing its merit in regional drug concentration and persistence. As amphotericin B was shown to be effective in inhibiting the growth of Echinococcus, and the antifungal drug amphotericin B was applied to patients as a remedial treatment. The liposomal amphotericin B might be a potential drug for HAE although it needs further validation.

Summary

Albendazole is the only recommended clinical drug for advanced hepatic alveolar echinococcosis. However, its therapeutic efficiency is not optimal. It is reported that antibiotic (Mefloquine, nitazoxanide, and amphotericin B), protease inhibitor (bortezomib, Sorafenib, and imatinib), and immune checkpoint inhibitor (anti-PD-1/PD-L1 drugs, anti-TIGIT drugs) have anti-echinococcosis effect in animal models. Clinical trails investigating these drugs could be considered to achieve a parasitocidal effect rather than a parasitostatic effect. Moreover, nanoparticles loading Albendazole or other potential drugs could increase local drug concentration to treat echinococcosis efficiently. As there are already many nanoparticle could be applied in clinics, such as liposome, it could also be a strategy to develop novel drugs for HAE. Drug repurposing and nano drug delivery system might be promising in novel treatment for advanced alveolar echinococcosis.

Author contributions

XX, XQ, and CG wrote the manuscript. YP, HZ and LZ designed form. ZW, MP, and DW checked out antibiotic drugs. WY, FK, and DS checked out protease inhibitor. YG, XS, and WH checked out immune checkpoint inhibitor. JY checked out immune checkpoint nano drug delivery system. XF and HF reviewed the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This study was supported by the Qinghai Provincial Science and Technology Department Project (No. 2020-ZJ-Y01), Young and Middle-aged Scientific Research Fund Project of the Affiliated Hospital of Qinghai University (No. ASRF-2021-YB-17), and Beijing Natural Science Foundation (Z190024).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Ahmadpour, E., Godrati-Azar, Z., Spotin, A., Norouzi, R., Hamishehkar, H., Nami, S., et al. (2019). Nanostructured lipid carriers of ivermectin as a novel drug delivery system in hydatidosis. Parasit. Vectors 12:469. doi: 10.1186/s13071-019-3719-x

CrossRef Full Text | Google Scholar

Akbulut, S. (2018). Parietal complication of the hydatid disease: comprehensive literature review. Medicine 97:e10671. doi: 10.1097/MD.0000000000010671

PubMed Abstract | CrossRef Full Text | Google Scholar

Akbulut, S., Yavuz, R., Sogutcu, N., Kaya, B., Hatipoglu, S., Senol, A., et al. (2014). Hydatid cyst of the pancreas: report of an undiagnosed case of pancreatic hydatid cyst and brief literature review. World J. Gastrointest Surg. 6, 190–200. doi: 10.4240/wjgs.v6.i10.190

PubMed Abstract | CrossRef Full Text | Google Scholar

Aminpour, S., Rafiei, A., Jelowdar, A., and Kouchak, M. (2019). Evaluation of the protoscolicidal effects of albendazole and albendazole loaded solid lipid nanoparticles. Iran. J. Parasitol. 14, 127–135. doi: 10.18502/ijpa.v14i1.726

PubMed Abstract | CrossRef Full Text | Google Scholar

Antoine, T., Fisher, N., Amewu, R., O'Neill, P. M., Ward, S. A., and Biagini, G. A. (2014). Rapid kill of malaria parasites by artemisinin and semi-synthetic endoperoxides involves ROS-dependent depolarization of the membrane potential. J. Antimicrob. Chemother. 69, 1005–1016. doi: 10.1093/jac/dkt486

PubMed Abstract | CrossRef Full Text | Google Scholar

Bakhtiar, N. M., Akbarzadeh, A., Casulli, A., Mahami-Oskouei, M., Ahmadpour, E., Nami, S., et al. (2019). Therapeutic efficacy of nanocompounds in the treatment of cystic and alveolar echinococcoses: challenges and future prospects. Parasitol. Res. 118, 2455–2466. doi: 10.1007/s00436-019-06416-5.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Bakhtiar, N. M., Spotin, A., Mahami-Oskouei, M., Ahmadpour, E., and Rostami, A. (2020). Recent advances on innate immune pathways related to host-parasite cross-talk in cystic and alveolar echinococcosis. Parasit. Vectors 13:232. doi: 10.1186/s13071-020-04103-4

CrossRef Full Text | Google Scholar

Ballif, B. A., and Blenis, J. (2001). Molecular mechanisms mediating mammalian mitogen-activated protein kinase (MAPK) kinase (MEK)-MAPK cell survival signals. Cell Growth Differ. 12, 397–408. doi: 10.1038/sj.cdd.4400901

CrossRef Full Text | Google Scholar

Bellanger, A. P., Courquet, S., Pallandre, J. R., Godet, Y., and Millon, L. (2020). Echinococcus multilocularis vesicular fluid induces the expression of immune checkpoint proteins in vitro. Parasite Immunol. 42:e12711. doi: 10.1111/pim.12711

PubMed Abstract | CrossRef Full Text | Google Scholar

Breckenridge, A., and Jacob, R. (2019). Overcoming the legal and regulatory barriers to drug repurposing. Nat. Rev. Drug Discov. 18, 1–2. doi: 10.1038/nrd.2018.92

PubMed Abstract | CrossRef Full Text | Google Scholar

Brehm, K., Spiliotis, M., Zavala-Góngora, R., Konrad, C., and Frosch, M. (2006). The molecular mechanisms of larval cestode development: first steps into an unknown world. Parasitol. Int. 55, S15–S21. doi: 10.1016/j.parint.2005.11.003.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Chong, S., Chen, G., Dang, Z., Niu, F., Zhang, L., Ma, H., et al. (2022). Echinococcus multilocularis drives the polarization of macrophages by regulating the RhoA-MAPK signaling pathway and thus affects liver fibrosis. Bioengineered 13, 8747–8758. doi: 10.1080/21655979.2022.2056690

PubMed Abstract | CrossRef Full Text | Google Scholar

Davis, A., Dixon, H., and Pawlowski, Z. S. (1989). Multicentre clinical trials of benzimidazole-carbamates in human cystic echinococcosis (phase 2). Bull. World Health Organ. 67, 503–508.

Google Scholar

Dissous, C., Ahier, A., and Khayath, N. (2007). Protein tyrosine kinases as new potential targets against human schistosomiasis. BioEssays 29, 1281–1288. doi: 10.1002/bies.20662

PubMed Abstract | CrossRef Full Text | Google Scholar

El-On, J. (2003). Benzimidazole treatment of cystic echinococcosis. Acta Trop. 85, 243–252. doi: 10.1016/s0001-706x(02)00217-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Fateh, R., Norouzi, R., Mirzaei, E., Nissapatron, V., Nawaz, M., Khalifeh-Gholi, M., et al. (2021). In vitro evaluation of albendazole nanocrystals against Echinococcus granulosus protoscolices. Ann. Parasitol. 67, 203–212. doi: 10.17420/ap6702.330

PubMed Abstract | CrossRef Full Text | Google Scholar

Gopalakrishnan, A. M., and Kumar, N. (2015). Antimalarial action of artesunate involves DNA damage mediated by reactive oxygen species. Antimicrob. Agents Chemother. 59, 317–325. doi: 10.1128/AAC.03663-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Gottstein, B., Soboslay, P., Ortona, E., Wang, J., Siracusano, A., and Vuitton, D. A. (2017). Immunology of alveolar and cystic echinococcosis (AE and CE). Adv. Parasitol. 96, 1–54. doi: 10.1016/bs.apar.2016.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Gupta, S. C., Sung, B., Prasad, S., Webb, L. J., and Aggarwal, B. B. (2013). Cancer drug discovery by repurposing: teaching new tricks to old dogs. Trends Pharmacol. Sci. 34, 508–517. doi: 10.1016/j.tips.2013.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Hatipoglu, S., Bulbuloglu, B., Piskin, T., Kayaalp, C., and Yilmaz, S. (2013). Living donor liver transplantation for alveolar echinococcus is a difficult procedure. Transplant. Proc. 45, 1028–1030. doi: 10.1016/j.transproceed.2013.02.084

PubMed Abstract | CrossRef Full Text | Google Scholar

Hemer, S., and Brehm, K. (2012). In vitro efficacy of the anticancer drug imatinib on Echinococcus multilocularis larvae. Int. J. Antimicrob. Agents 40, 458–462. doi: 10.1016/j.ijantimicag.2012.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Hemphill, A., Stettler, M., Walker, M., Siles-Lucas, M., Fink, R., and Gottstein, B. (2002). Culture of Echinococcus multilocularis metacestodes: an alternative to animal use. Trends Parasitol. 18, 445–451. doi: 10.1016/s1471-4922(02)02346-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, C., Liu, Z., Liu, C., Zhang, Y., Fan, H., and Qian, F. (2020). Improvement of antialveolar echinococcosis efficacy of albendazole by a novel nanocrystalline formulation with enhanced oral bioavailability. ACS Infect Dis. 6, 802–810. doi: 10.1021/acsinfecdis.9b00231.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Jarvis, J. N., Lawrence, D. S., Meya, D. B., Kagimu, E., Kasibante, J., Mpoza, E., et al. (2022). Ambition study group. Single-dose liposomal amphotericin B treatment for cryptococcal meningitis. N. Engl. J. Med. 386, 1109–1120. doi: 10.1056/NEJMoa2111904

PubMed Abstract | CrossRef Full Text | Google Scholar

Jebbawi, F., Bellanger, A. P., Lunström-Stadelmann, B., Rufener, R., Dosch, M., Goepfert, C., et al. (2021). Innate and adaptive immune responses following PD-L1 blockade in treating chronic murine alveolar echinococcosis. Parasite Immunol. 43:e12834. doi: 10.1111/pim.12834.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, C. P. (1986). Preliminary clinical observation of mebendazole and traditional Chinese medicine treatment in 57 cases with echinococcosis. J. Parasitol. Parasit. Dis. 4, 51–53.

Google Scholar

Jiang, C. P. (1991). An experimental study on albendazole and Chinese herbs anti-Echinococcus powder in treatment of early and anvanced stages of mice intra-peritoneal alveococcus infection. Endem. Dis. Bull. 6, 1–6.

Google Scholar

Jiang, C. P. (1995). Experimental study of xiaobao powder in treating alveolar hydatid disease in mice. China J. Integrat. Tradit. Chin. West. Med. 1, 346–348.

Google Scholar

Jiang, C. (1998). Alveolar echinococcosis in China. Chin. Med. J. 111, 470–475.

Google Scholar

Jiang, C. P., and Liu, Y. H. (1994). A clinicopathological analysis of 70 cases of liver alveolar echinococcosis with a comment of the prognostic factors. Zhonghua Nei Ke Za Zhi 33, 452–454.

Google Scholar

Jiraungkoorskul, W., Sahaphong, S., Sobhon, P., Riengrojpitak, S., and Kangwanrangsan, N. (2005). Effects of praziquantel and artesunate on the tegument of adult Schistosoma mekongi harboured in mice. Parasitol. Int. 54, 177–183. doi: 10.1016/j.parint.2005.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Joliat, G. R., Melloul, E., Petermann, D., Demartines, N., Gillet, M., Uldry, E., et al. (2015). Outcomes after liver resection for hepatic alveolar echinococcosis: a single-center cohort study. World J. Surg. 39, 2529–2534. doi: 10.1007/s00268-015-3109-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ke, X., Do, D. C., Li, C., Zhao, Y., Kollarik, M., Fu, Q., et al. (2019). Ras homolog family member A/Rho-associated protein kinase 1 signaling modulates lineage commitment of mesenchymal stem cells in asthmatic patients through lymphoid enhancer-binding factor 1. J. Allergy Clin. Immunol. 143, 1560.e6–1574.e6. doi: 10.1016/j.jaci.2018.08.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Kern, P., Silva, A., Akhan, O., Müllhaupt, B., Vizcaychipi, K. A., Budke, C., et al. (2017). The echinococcoses: diagnosis, clinical management and burden of disease. Adv. Parasitol. 96, 259–369. doi: 10.1016/bs.apar.2016.09.006

CrossRef Full Text | Google Scholar

Kontaridis, M. I., Yang, W., Bence, K. K., Cullen, D., Wang, B., Bodyak, N., et al. (2008). Deletion of Ptpn 11 (Shp 2) in cardiomyocytes causes dilated cardiomyopathy via effects on the extracellular signal-regulated kinase/mitogen-activated protein kinase and RhoA signaling pathways. Circulation 117, 1423–1435. doi: 10.1161/CIRCULATIONAHA.107.728865

CrossRef Full Text | Google Scholar

Kubes, P., and Jenne, C. (2018). Immune responses in the liver. Annu. Rev. Immunol. 36, 247–277. doi: 10.1146/annurev-immunol-051116-052415.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

La, X., Zhang, F., Li, Y., Li, J., Guo, Y., Zhao, H., et al. (2015). Upregulation of PD-1 on CD4+CD25+ T cells is associated with immunosuppression in liver of mice infected with Echinococcus multilocularis. Int. Immunopharmacol. 26, 357–366. doi: 10.1016/j.intimp.2015.04.013.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, R., Lü, G., Wang, J., Zhang, C., Xie, W., Lu, X., et al. (2011). Time course of gene expression profiling in the liver of experimental mice infected with Echinococcus multilocularis. PLoS One 6:e14557. doi: 10.1371/journal.pone.0014557

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, R. Y., Wang, J. H., Lu, X. M., Zhou, X. T., Mantion, G., Wen, H., et al. (2009). Components of the mitogen-activated protein kinase cascade are activated in hepatic cells by Echinococcus multilocularis metacestode. World J. Gastroenterol. 15, 2116–2124. doi: 10.3748/wjg.15.2116

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, C., Fan, H., Guan, L., Ge, R. L., and Ma, L. (2021). In vivo and in vitro efficacy of crocin against Echinococcus multilocularis. Parasit. Vectors 14:364. doi: 10.1186/s13071-021-04866-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Wang, X. Q., Ren, W. X., Chen, Y. L., Yu, Y., Zhang, J. K., et al. (2013). Novel albendazole-chitosan nanoparticles for intestinal absorption enhancement and hepatic targeting improvement in rats. J. Biomed. Mater Res. B Appl. Biomater 101, 998–1005. doi: 10.1002/jbm.b.32908.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Lundström-Stadelmann, B., Rufener, R., and Hemphill, A. (2020). Drug repurposing applied: activity of the anti-malarial mefloquine against Echinococcus multilocularis. Int. J. Parasitol. Drugs Drug Resist. 13, 121–129. doi: 10.1016/j.ijpddr.2020.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, H., Zhang, H. H., and Wei, L. (2013). Frequency of T-cell Fox P 3+ Treg and CD4+/CD8+ PD-1 expression is related to HBeAg seroconversion in hepatitis B patients on pegylated interferon. Chin. Med. J. 126, 267–273.

Google Scholar

Mai, H. A., Wang, Y. H., Zhao, J. M., and Bai, L. (2011). Effect of different drugs on Echinococcus multilocularis metacestodes in vitro. Zhongguo Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi 29, 419–424. doi: 10.1007/s10008-010-1224-4

CrossRef Full Text | Google Scholar

McManus, D. P., Zhang, W., Li, J., and Bartley, P. B. (2003). Echinococcosis. Lancet 362, 1295–1304. doi: 10.1016/S0140-6736(03)14573-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Meilinger, M., Stoeckl, C., Pollheimer, M., Kern, P., Reisinger, E. C., Seeber, K., et al. (2013). Progressive alveolar echinococcosis after discontinuation of anthelmintic therapy. Parasit. Vectors 6:287. doi: 10.1186/1756-3305-6-287

PubMed Abstract | CrossRef Full Text | Google Scholar

Mittal, N., and Mittal, R. (2021). Repurposing old molecules for new indications: defining pillars of success from lessons in the past. Eur. J. Pharmacol. 912:174569. doi: 10.1016/j.ejphar.2021.174569.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Monaghan, S. F., Thakkar, R. K., Tran, M. L., Huang, X., Cioffi, W. G., Ayala, A., et al. (2012). Programmed death 1 expression as a marker for immune and physiological dysfunction in the critically ill surgical patient. Shock 38, 117–122. doi: 10.1097/SHK.0b013e31825de6a3

PubMed Abstract | CrossRef Full Text | Google Scholar

Neuberger, A., and Oraiopoulos, N. (2019). Drakeman DL. Renovation as innovation: is repurposing the future of drug discovery research? Drug Discov. Today 24, 1–3. doi: 10.1016/j.drudis.2018.06.012.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Parsak, C. K., Demiryurek, H. H., Inal, M., Sakman, G., Koltas, I. S., Erkocak, E. U., et al. (2007). Alveolar hydatid disease: imaging findings and surgical approach. Acta Chir. Belg. 107, 572–577. doi: 10.1080/00015458.2007.11680128

PubMed Abstract | CrossRef Full Text | Google Scholar

Pawluk, S. A., Roels, C. A., Wilby, K. J., and Ensom, M. H. (2015). A review of pharmacokinetic drug-drug interactions with the anthelmintic medications albendazole and mebendazole. Clin. Pharmacokinet. 54, 371–383. doi: 10.1007/s40262-015-0243-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Pensel, P., Paredes, A., Albani, C. M., Allemandi, D., Sanchez Bruni, S., Palma, S. D., et al. (2018). Albendazole nanocrystals in experimental alveolar echinococcosis: enhanced chemoprophylactic and clinical efficacy in infected mice. Vet. Parasitol. 251, 78–84. doi: 10.1016/j.vetpar.2017.12.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Polamreddy, P., and Gattu, N. (2019). The drug repurposing landscape from 2012 to 2017: evolution, challenges, and possible solutions. Drug Discov. Today 24, 789–795. doi: 10.1016/j.drudis.2018.11.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, M. B., Abela-Ridder, B., Wu, W. P., and Zhou, X. N. (2017). Combating echinococcosis in China: strengthening the research and development. Infect. Dis. Poverty 6:161. doi: 10.1186/s40249-017-0374-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Reuter, S., Buck, A., Manfras, B., Kratzer, W., Seitz, H. M., Darge, K., et al. (2004). Structured treatment interruption in patients with alveolar echinococcosis. Hepatology 39, 509–517. doi: 10.1002/hep.20078

PubMed Abstract | CrossRef Full Text | Google Scholar

Reuter, S., Merkle, M., Brehm, K., Kern, P., and Manfras, B. (2003). Effect of amphotericin B on larval growth of Echinococcus multilocularis. Antimicrob. Agents Chemother. 47, 620–625. doi: 10.1128/AAC.47.2.620-625.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodrigues, J. M. Jr., Bories, C., Emery, I., Fessi, H., Devissaguet, J. P., and Liance, M. (1995). Development of an injectable formulation of albendazole and in vivo evaluation of its efficacy against Echinococcus multilocularis metacestode. Int. J. Parasitol. 25, 1437–1441. doi: 10.1016/0020-7519(95)00055-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Siles-Lucas, M., Nunes, C. P., and Zaha, A. (2001). Comparative analysis of the 14-3-3 gene and its expression in Echinococcus granulosus and Echinococcus multilocularis metacestodes. Parasitology 122, 281–287. doi: 10.1017/s0031182001007405

PubMed Abstract | CrossRef Full Text | Google Scholar

Spiliotis, M., Konrad, C., Gelmedin, V., Tappe, D., Brückner, S., Mösch, H. U., et al. (2006). Characterisation of EmMPK1, an ERK-like MAP kinase from Echinococcus multilocularis which is activated in response to human epidermal growth factor. Int. J. Parasitol. 36, 112–1097. doi: 10.1016/j.ijpara.2006.05.008.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Spiliotis, M., Kroner, A., and Brehm, K. (2003). Identification, molecular characterization and expression of the gene encoding the epidermal growth factor receptor orthologue from the fox-tapeworm Echinococcus multilocularis. Gene 323, 57–65. doi: 10.1016/j.gene.2003.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Spiliotis, M., Tappe, D., Brückner, S., Mösch, H. U., and Brehm, K. (2005). Molecular cloning and characterization of Ras-and Raf-homologues from the fox-tapeworm Echinococcus multilocularis. Mol. Biochem. Parasitol. 139, 225–237. doi: 10.1016/j.molbiopara.2004.11.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Stadelmann, B., Aeschbacher, D., Huber, C., Spiliotis, M., Müller, J., and Hemphill, A. (2014). Profound activity of the anti-cancer drug bortezomib against Echinococcus multilocularis metacestodes identifies the proteasome as a novel drug target for cestodes. PLoS Negl. Trop. Dis. 8:e3352. doi: 10.1371/journal.pntd.0003352

PubMed Abstract | CrossRef Full Text | Google Scholar

Vuitton, D. A., Zhang, S. L., Yang, Y., Godot, V., Beurton, I., Mantion, G., et al. (2006). Survival strategy of Echinococcus multilocularis in the human host. Parasitol. Int. 55, S51–S55. doi: 10.1016/j.parint.2005.11.007.Epub

CrossRef Full Text | Google Scholar

Wang, J., Cardoso, R., Marreros, N., Müller, N., Lundström-Stadelmann, B., Siffert, M., et al. (2018). Foxp 3+ T regulatory cells as a potential target for immunotherapy against primary infection with Echinococcus multilocularis eggs. Infect. Immun. 86, e00542–e00518. doi: 10.1128/IAI.00542-18

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Jebbawi, F., Bellanger, A. P., Beldi, G., Millon, L., and Gottstein, B. (2018). Immunotherapy of alveolar echinococcosis via PD-1/PD-L1 immune checkpoint blockade in mice. Parasite Immunol. 40:e12596. doi: 10.1111/pim.12596

PubMed Abstract | CrossRef Full Text | Google Scholar

Weiss, J. M., Davies, L. C., Karwan, M., Ileva, L., Ozaki, M. K., Cheng, R. Y., et al. (2018). Itaconic acid mediates crosstalk between macrophage metabolism and peritoneal tumors. J. Clin. Invest. 128, 3794–3805. doi: 10.1172/JCI99169

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilker, E., and Yaffe, M. B. (2004). 14-3-3 proteins--a focus on cancer and human disease. J. Mol. Cell. Cardiol. 37, 633–642. doi: 10.1016/j.yjmcc.2004.04.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, S. H., Jian, X., Tanner, M., Yong-Nian, Z., Keiser, J., Utzinger, J., et al. (2008). Artemether, artesunate, praziquantel and tribendimidine administered singly at different dosages against Clonorchis sinensis: a comparative in vivo study. Acta Trop. 106, 54–59. doi: 10.1016/j.actatropica.2008.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, N. (2021). Concept and mechanism innovations provide a sustainable driver for echinococcosis control in China[J]. Chin. J. Schistosomiasis Control 33, 329–333. doi: 10.16250/j.32.1374.2021188

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, S., Wu, J., Ding, C., Cui, Y., Zhou, Y., Li, Y., et al. (2017). Epidemiological features of and changes in incidence of infectious diseases in China in the first decade after the SARS outbreak: an observational trend study. Lancet Infect. Dis. 17, 716–725. doi: 10.1016/S1473-3099(17)30227-X

CrossRef Full Text | Google Scholar

Yue, X., and Dai, Z. (2018). Liposomal nanotechnology for cancer theranostics. Curr. Med. Chem. 25, 1397–1408. doi: 10.2174/0929867324666170306105350

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, C., Lin, R., Li, Z., Yang, S., Bi, X., Wang, H., et al. (2020). Immune exhaustion of T cells in alveolar echinococcosis patients and its reversal by blocking checkpoint receptor TIGIT in a murine model. Hepatology 71, 1297–1315. doi: 10.1002/hep.30896

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, C., Wang, H., Li, J., Hou, X., Li, L., Wang, W., et al. (2021). Involvement of TIGIT in natural killer cell exhaustion and immune escape in patients and mouse model With liver Echinococcus multilocularis infection. Hepatology 74, 3376–3393. doi: 10.1002/hep.32035.Epub

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Q., Ye, J. R., Ma, H. M., Wu, J. J., Jiang, T., and Zheng, H. (2016). Role of immune tolerance in BALB/c mice with anaphylactic shock after Echinococcus granulosus infection. Immunol. Res. 64, 233–241. doi: 10.1007/s12026-015-8741-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, Y., Gui, W., Niu, F., and Chong, S. (2019). The MAPK signaling pathways as a novel way in regulation and treatment of parasitic diseases. Diseases 7:9. doi: 10.3390/diseases7010009

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: hepatic alveolar echinococcosis, drug repurposing, antibiotic drug, protease inhibitor, immune checkpoint inhibitor

Citation: Xu X, Qian X, Gao C, Pang Y, Zhou H, Zhu L, Wang Z, Pang M, Wu D, Yu W, Kong F, Shi D, Guo Y, Su X, Hu W, Yan J, Feng X and Fan H (2022) Advances in the pharmacological treatment of hepatic alveolar echinococcosis: From laboratory to clinic. Front. Microbiol. 13:953846. doi: 10.3389/fmicb.2022.953846

Received: 26 May 2022; Accepted: 21 July 2022;
Published: 08 August 2022.

Edited by:

Xiaoxiao Wang, Zhejiang Center for Disease Control and Prevention, China

Reviewed by:

Wentao Wang, Sichuan University, China
Sami Akbulut, İnönü University, Turkey
Tevfik Tolga Sahin, İnönü University, Turkey

Copyright © 2022 Xu, Qian, Gao, Pang, Zhou, Zhu, Wang, Pang, Wu, Yu, Kong, Shi, Guo, Su, Hu, Yan, Feng and Fan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xiaobin Feng, fengxiaobin200708@aliyun.com; Haining Fan, fanhaining@medmail.com.cn

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.