Next Article in Journal
Global Research Trends in the Links between Periodontal Disease and Cancer: A Bibliometric Analysis
Previous Article in Journal
Metagenomics Insight into Veterinary and Zoonotic Pathogens Identified in Urban Wetlands of Los Lagos, Chile
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of the In-Vitro Effects of Albendazole, Mebendazole, and Praziquantel Nanocapsules against Protoscolices of Hydatid Cyst

1
Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad P.O. Box 9177948974, Iran
2
WOAH and NRL for Echinococcosis, Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy
3
Department of Physics, University of Zabol, Zabol P.O. Box 538-98615, Iran
*
Authors to whom correspondence should be addressed.
Pathogens 2024, 13(9), 790; https://doi.org/10.3390/pathogens13090790
Submission received: 24 June 2024 / Revised: 3 September 2024 / Accepted: 10 September 2024 / Published: 12 September 2024
(This article belongs to the Section Parasitic Pathogens)

Abstract

:
Cystic echinococcosis still remains a serious health and economic problem worldwide. The etiologic agent is Echinococcus granulosus sensu lato, giving origin to a fluid-filled cystic lesion. Therapy faces several challenges. Nanodrugs have shown promise as chemotherapeutics against hydatid cysts. The present study evaluated a highly safe lipid nano-polymeric capsule for its superior efficacy and ability to overcome drug resistance. Nanocapsule drugs were formulated into six groups: Albendazole, mebendazole, praziquantel, albendazole + mebendazole, albendazole + praziquantel, and praziquantel + mebendazole. The protoscolicidal effects of these six groups were assessed at 10, 60, and 120 min in three concentrations (1, 0.5, and 0.25 mg/mL). Drug formulations were evaluated via zeta potential, droplet size, solubility, particle size analyzer (PSA), and scanning electron microscopy. According to the PSA results, the mean size of the albendazole nanocapsules was 193.01 nm, mebendazole was 170.40 nm, and praziquantel was 180.44 nm. Albendazole + mebendazole showed the greatest protoscolicidal activity at a concentration of 1 mg/mL after 120 min. In contrast, each drug’s 0.25 mg/mL single-dose times showed the least protoscolicidal activity after 120 min. With the right application of nanotechnology, it is possible to produce safe and effective drugs, such as the polymeric combination of albendazole and mebendazole, which has promising implications.

1. Introduction

The infections caused by metacestodes, the larval stage of Echinococcus species, can be asymptomatic for years or lead to severe illness and even death in animals and humans [1]. The species, representing a serious worldwide health problem and requiring priority veterinary and medical attention, are represented by Echinococcus granulosus sensu lato (s.l.) and Echinococcus multilocularis, the causative agents of cystic echinococcosis (CE) and alveolar echinococcosis (AE). These two taeniid tapeworms have different geographic distributions, pathogenicity, host specificity, and clinical symptoms [2,3].
Their biological cycles involve definitive hosts, particularly canids that may release eggs into the environment by their feces, and intermediate hosts, mainly rodents for E. multilocularis [4] and sheep or other ruminants for E. granulosus s.l. [4,5].
Researchers in the Middle East and Iran focus mainly on researching E. granulosus s.l., since this species most often causes CE [6,7]. This region has a high incidence of E. granulosus s.l. infections due to its widespread presence in livestock and domestic dogs [8]. Understanding and controlling this particular species is crucial for developing public health efforts in these regions.
In detail, E. granulosus s.l.’s life cycle includes different stages in different hosts. This parasite first lives as an adult in the intestines of the definitive host, carnivores such as dogs [6]. Parasite eggs are excreted through the feces of these animals and enter the environment. Intermediate hosts, usually herbivorous animals, such as sheep and goats, contract hydatid cysts by eating grass contaminated with parasite eggs [7]. Cystic echinococcosis infections in humans are uncommon and can occur after eating infected raw food not properly washed [8,9,10] and contaminated by the eggs previously released into the environment. The eggs hatch in the intestines of these animals, and the larvae pass through the intestines [11]. After diffusion through the intestinal mucosa into the lymphatic system or the bloodstream, the parasite can spread into the organs, mainly the liver (65%) and lungs (25%), and seldom (10%) in other tissues (spleen, kidneys, heart, bones, and central nervous system), and give origin to a fluid-filled cystic lesion [12]. The larvae develop into hydatid cysts in these organs that can remain in the intermediate host’s body for years [13]. The parasite’s life cycle is completed when a carnivore consumes the infected flesh or viscera of the intermediate host, and the adult parasites settle in their intestine [14].
Cystic echinococcosis can be diagnosed and followed-up using imaging techniques, abdominal ultrasound (US), and chest radiography for an initial screening, in addition to magnetic resonance imaging (MRI) and computed tomography (CT) to detect more details of the lesion, such as vascularization and organization of adjacent organs, and to obtain a deeper diagnostic picture mostly necessary before surgical removal of the lesion [15]. Radiological investigations need to be supported by immunological analyses and useful tools capable of detecting the IgG antibodies anti-Echinococcus, such as immunochromatographic tests (ICTs), enzyme-linked immunosorbent assays (ELISAs), and immunoblotting (IB) [16,17].
The World Health Organization—Informal Working Group on Echinococcosis (WHO-IWGE) guidelines describe several medical treatments for managing patients affected by CE [18]. Surgery is the most effective treatment [17], but at the same time, it is very invasive, and if it is not properly performed, a new infection may arise as a major surgical complication [19]. Percutaneous therapy (PAIR) is a less deep procedure, but it does not comprise the removal of the echinococcal cyst. Moreover, the watch-and-wait (W/W) protocol is less invasive but requires strict follow-up. All treatments must be chosen according to the lesion’s characteristics (number, stadium, localization, etc.) [18]. However, all treatments described above must be put aside in the patient’s pharmacological management.
There are currently chemotherapeutic drugs that can treat CE with good results, including albendazole (ABZ), mebendazole (MBZ), praziquantel (PZQ), and benzimidazole (BMZ) [20,21]. Usually, the more-indicated drug for the management of CE is ABZ [21], prescribed in association with high-fat meals to increase bioavailability. But, sometimes, if this drug is unavailable, intolerable, or has side effects during the pharmacological treatment of CE patients, it is exchanged with another according to the guidelines [22]. It is known that BMZ side effects are common when high doses are administered for extended periods to human patients [23,24]. ABZ and MBZ have wide-spectrum anthelmintic activities by inhibiting the polymerization of a structural protein, tubulin, in the parasite’s intestinal cells. As a result, the microtubules cannot be constructed, and the life processes of the helminthic cells are interrupted [20]. PZQ acts causing spastic muscle paralysis and Ca2+ influx in the parasite. The worms are thus completely destroyed in the intestine or eliminated through the feces. Some BMZ compounds inhibit the biosynthesis of ergosterol required in the cell membranes of fungi and protozoa [25].
Recently, nanodrugs have emerged as potential pharmaceutical game-changers, significantly transforming therapeutic interventions [24]. It has been proven that nanodrugs can be used successfully to treat hydatid cysts [26,27,28,29]. Nanodrugs have demonstrated great promise as chemotherapeutics in hydatid cysts [30,31,32,33,34,35]. However, these nanomedicines have challenges that must be addressed and resolved before they can revolutionize treatment for hydatid cysts [32]. Nanotechnology must be harnessed for medical advancement as researchers balance therapeutic efficacy, safety profiles, and environmental impact [36,37,38]. The unique properties of nanodrugs make them ideal candidates for improving therapeutic outcomes in various medical conditions since they are small and have specific surface properties [39,40,41]. Nevertheless, there are challenges associated with using nanomedicine to treat hydatid cysts. A prominent concern, for example, is renal toxicity, in which nanoparticles can accumulate in the kidneys and cause adverse effects [42,43]. Moreover, the long-term effects of these nanoparticles on ecosystems are unknown, especially in terms of their composition and excretion [44,45,46]. The complex interactions between nanodrugs’ physicochemical properties, biodistribution patterns, and potential harm to patients and the environment necessitate a comprehensive approach that analyzes their therapeutic potential, biodistribution rates, and potential toxicity issues.
In the present study, nanocapsule systems have been explored as a potential solution to treat hydatid cysts with drug resistance [47,48] and to set new standards for safety, effectiveness, and sustainability. The present study aims to revolutionize how hydatid cysts are treated to bring about a new era of treatment that prioritizes patient well-being and environmental sustainability.

2. Materials and Methods

2.1. Collection of Protoscolices

Hydatid cysts were obtained aseptically from CE-infected sheep, and a 30-min sedimentation time allowed the hydatid fluid to settle in a cylinder for protoscolex isolation. After three rounds of washing with phosphate buffer saline (PBS), the protoscolices were recognizable as the settled bottom layer. Muscle movements and eosin staining were used for the viability assessment. The viability of protoscolices was assured through their movement under a light microscope, using the eosin exclusion test with a concentration of 0.1% eosin stain (1 g of eosin powder in 1000 mL distilled water). After 5 min of exposure, the unstained protoscolices were deemed viable, while stained protoscolices were considered dead. When 90% or more viable protoscolices were recorded in the sediments, the sample was appropriate for the experiments [49].

2.2. Development of Encapsulated Drugs

At room temperature, a suitably diluted amount of sodium caprylate (fatty acid) and PBS (pH 7.4) were vigorously mixed at a consistent ratio, and drug-to-pluronic and oil-to-surfactant molar ratios were determined in the form of microemulsions containing drugs incorporated in oil-in-water microemulsions which had been developed [50].

2.3. Zeta Potential, Particle Size Analyzer (PSA) Technique, and Scanning Electron Microscopy (SEM)

Twenty-four hours before the device examined the samples, ABZ solutions were made with lipid nano-polymeric capsules according to the instructions. Then, each prepared sample was poured into separate microtubes, mixed with 10 µL of PBS, and transferred to the central laboratory. The particle size and size distribution of the nanoparticles obtained were assessed using a particle size analyzer (PSA; SHIMADZU, SALD-2101, Tokyo, Japan). Additionally, the measurements were reconfirmed at predetermined intervals. Zeta potential was determined using Zeta-Chek (Microtract, ZC007, Haan/Duesseldor, Germany). Furthermore, for conducting scanning electron microscopy (SEM), the samples were first prepared using the Sputter Coater (SC7620) Polaron Range, and finally, SEM was performed by LEO 1450VP, 35 kV, Oberkochen, Germany.

2.4. Scolicidal Assay

A total of six nanocapsules were used in the study, ABZ, MBZ, PZQ, ABZ + MBZ, ABZ + PZQ, and PZQ + MBZ. At three times (10 min, 60 min, and 120 min), three concentrations (1, 0.5, and 0.25 mg/mL) were evaluated for their protoscolicidal effects. The sumac extract solution was prepared by dissolving dried extract in distilled water at 10, 30, and 50 mg/mL concentrations. A drop of protoscolex-rich sediment was combined with 2.5 mL of each sumac solution. Incubation was conducted at 37 °C for 10, 60, and 120 min after the contents had been gently mixed. Protoscolices were stained with 0.1% eosin after each incubation period after the upper phase had been carefully removed. Smeared protoscolices were examined under a light microscope after an incubation period of 15 min. At least 500 specimens were counted in each experiment to determine the percentage of dead protoscolices, with non-treated protoscolices as the control group. To ensure the accuracy of the results, the entire procedure was repeated three times.

2.5. Statistical Analysis

As a result of this analytical approach, significant differences in protoscolicidal effects were identified among the different drug classes and concentrations. R version 4.3.1 (16 June 2023)—“Beagle Scouts” Copyright© 2023 The R Foundation for Statistical Computing Platform: aarch64-apple-darwin20 (64-bit) was used for statistical analysis. The data from this study were compared using the Kruskal–Wallis test. The Mann–Whitney test and t-test were used to compare between groups. Furthermore, post hoc tests included Bonferroni and Tukey–Kramer. The statistical tests were conducted with significance levels of less than 0.05%. The schematic of the methodology in the current study is illustrated in Figure 1.

3. Results

3.1. Zeta Potential, PSA, and SEM Results

The zeta potential spectra of the ABZ, MBZ, and PZQ are shown in Figure 2. It can be concluded in the graphs that the nanocapsules loaded with ABZ are in the range of 5 to −55 millivolts (mean: −35.78 mW), MBZ is in the range of 0 to −48 millivolts (mean: −27.38 mW), and PZQ is in the range of 0 to −43 millivolts (mean: −23.34 mW). A higher percentage of electric charges are present and have more peaks than the graph of free drugs, which shows that the stabilization of the drug on the nanoparticles has been completed well. They have higher stability in the environment than the drug. According to the PSA results, the mean size of the ABZ nanocapsules was 193.01 nm, MBZ was 170.40 nm, and PZQ was 180.44 nm (Figure 3). Figure 4 shows the morphology of the polymeric nanocapsules. The figure shows that the polymeric nanocapsules are sphere-like and of different sizes (Figure 4).

3.2. Protoscolicidal Rate

In 10, 60, and 120 min, the 0.25 mg/mL ABZ concentration exhibited respective protoscolicidal rates of 14.6%, 32%, and 46.6%. For MBZ, the protoscolicidal rate was 19%, 28.6%, and 36.3%. Additionally, the protoscolicidal rate for PZQ was 18%, 32%, and 46.33%, respectively. Furthermore, in 10, 60, and 120 min, at a 0.25 mg/mL concentration for ABZ + MBZ, the protoscolicidal rate was 20.6%, 31.6%, and 47%. For ABZ + PZQ, the protoscolicidal rate was 22.33%, 34.3%, and 41%. Moreover, MBZ + PZQ’s protoscolicidal rate was 21.3%, 32%, and 46.6%. Among these, PZQ exhibited the best performance at 120 min; albeit, all drugs showed significantly higher lethality percentages compared to the control group across all times (p < 0.05) (Figure 5).
Regarding the 0.5 mg/mL concentration, in 10, 60, and 120 min, ABZ exhibited respective protoscolicidal rates of 22.6%, 40%, and 52.3%. For MBZ, the protoscolicidal rate was 27%, 35.6%, and 46.6%. Additionally, the protoscolicidal rate for PZQ was 26.3%, 45%, and 56.3%, respectively. Furthermore, in 10, 60, and 120 min, for the 0.5 mg/mL concentration of ABZ + MBZ, the protoscolicidal rate was 27%, 41%, and 54.3%. For ABZ + PZQ, the protoscolicidal rate was 29.6%, 44.6%, and 50%. Moreover, the MBZ + PZQ protoscolicidal rate was 26%, 39.6%, and 50%. Among these, ABZ exhibited the best performance at 120 min; albeit, all drugs showed significantly higher lethality percentages compared to the control group across all time intervals (p < 0.05) (Figure 6 and Figure 7).
Regarding 1 mg/mL concentration, in 10, 60, and 120 min, ABZ exhibited respective protoscolicidal rates of 29.6%, 53%, and 62%. For MBZ, the protoscolicidal rate was 41%, 54.3%, and 80%. Additionally, the protoscolicidal rate for PZQ was 32.6%, 50.6%, and 63%, respectively. Furthermore, in 10, 60, and 120 min, at a 1 mg/mL concentration for ABZ + MBZ, the protoscolicidal rate was 52.3%, 66%, and 89%. For ABZ + PZQ, the protoscolicidal rate was 42.3%, 61.6%, and 73.3%. Moreover, the MBZ + PZQ protoscolicidal rate was 31.3%, 52.3%, and 57.3%. Among these, ABZ + MBZ exhibited the best performance at 120 min; albeit, all drugs showed significantly higher lethality percentages compared to the control group across all time intervals (p < 0.05) (Figure 8 and Figure 9).

4. Discussion

Various parts of the world continue to be affected by cystic echinococcosis. Every single clinical management strategy of a patient, similarly to chemical treatment, is influenced by the hydatid cyst’s structure, localization, and stage. The host’s body produces a fibrous layer surrounding the cyst [51]. A monolayer of germinal cells encircles the parasite’s capsule in a 50 mm thick acellular complex consisting of mucopolysaccharides and proteins [52]. In the cyst fluid or on the germinal layer, these cells differentiate into protoscolices, which stay attached to these germinal layers [53]. During a cyst rupture or surgery, protoscolices can form new cysts if spilled on the intermediate host [19,54]. During cystogenesis, host and parasite proteins are present in a hypotonic fluid surrounding the cyst [55]. Therefore, active transport, selective absorption, and excretion must be possible through the cyst wall [52,56]. To kill the parasite tissues, drugs must penetrate all three layers of the cyst [57].
While chemotherapy has several potential benefits, it also has disadvantages [58,59,60]. There is considerable concern regarding drug resistance among parasites, where they evolve to withstand the effects of drugs to survive [61,62,63]. It is also important to note that the side effects of these chemotherapeutic agents can be highly damaging and significantly impact the patient’s quality of life [64]. There are several benefits associated with this procedure, as well as several drawbacks that necessitate the development of more effective and targeted treatment strategies [65].
The challenge is exacerbated because hydatid cysts are prevalent in many endemic regions where cystic echinococcosis is prominent. As a result, there is a significant barrier to managing chronic conditions due to limited access to healthcare facilities and resources [66]. A further burden created by the time it takes for patients to receive treatment for chronic infections is that it is both costly and challenging for healthcare systems to handle [67,68]. Thus, to address these multifaceted challenges, it is crucial to find innovative solutions that can deal with them. There is no doubt that in recent years, nanotechnology has emerged as one of the most potent forces to deal with hydatid cysts, more so than ever before [69,70]. With its exceptional physical and chemical properties, a nanoparticle can change treatment possibilities by adding a new dimension to them [71]. The ability of nanoparticles to deliver drugs with exceptional precision is one of their significant advantages, one of the most essential advantages of nanoparticles [72].
Albendazole is one of the main chemotherapy drugs used for the treatment of hydatid cysts, and it is used due to its good ability to kill parasites and reduce the size of cysts [73]. By disrupting the parasite’s glucose metabolism, this drug causes its death and destruction, and for this reason, it is very effective and widely used. However, there have been reports of some parasite strains developing resistance to albendazole, reducing its therapeutic efficacy and creating new treatment challenges. In this regard, nanotechnology and nano methods have played a very important role in enhancing the effects of albendazole [74,75]. Nanotechnology has significantly increased the efficiency of albendazole by improving the drug’s absorption capacity, increasing its stability and release time in the body, reducing side effects, and preventing parasite resistance. These achievements show the importance of using nanotechnology in improving chemotherapy treatments and increasing the effectiveness of drugs in dealing with parasitic diseases such as CE [76,77].
Liposomal nanoparticles can encapsulate therapeutic agents within lipid bilayers [78]. Encapsulating the drug in a protective film protects it from degradation so that it can be delivered directly to the cysts without any delays [79,80]. In this way, the therapeutic effect is enhanced and systemic toxicity is reduced, resulting in a better outcome. At the same time, nanotechnology offers extraordinary promise but must be cautiously approached to maximize its potential [81,82]. It is important to remember that nanotoxicity is a potential concern that cannot be ignored [83,84,85]. A lack of thorough study and understanding of nanoparticles can lead to their accumulation in vital organs and trigger unforeseen immune responses. Comprehensive toxicity assessments must be performed before these technologies are incorporated into clinical practice safely and effectively. Lipid microemulsion nanocapsules have emerged as one of the most promising nanotechnological breakthroughs among recent nanotechnology advancements [86]. Developing nanocapsules capable of being solubilized and stable and providing a controlled release of the drug offers an innovative solution to these problems. Researchers are experimenting with the use of nanocapsules to entrap antiparasitic medications to overcome the obstacles posed by drug resistance and the insufficient distribution of these drugs [87,88]. Using nanocapsules, drugs can be released over a prolonged period, preventing pharmaceutical concentrations from falling below therapeutic levels over time and potentially reducing the frequency of drug administration [89].
The presented data on the zeta potential spectrum of ABZ, MBZ, and PZQ nanocapsules reveal intriguing insights into the stability of these drugs when loaded onto nanocapsules. Notably, the higher stability observed in the environment for drug-loaded nanoparticles suggests an effective and improved stabilization process. This finding is crucial as it implies that the nanocapsules could potentially enhance the stability and efficacy of these drugs, offering a promising avenue for drug delivery systems [90,91,92]. Moving on to the protoscolicidal rates at different concentrations and time intervals, the results provide compelling evidence of the efficacy of ABZ, MBZ, and PZQ, individually and in combination. At a 0.25 mg/mL concentration, all three drugs and their combinations exhibited protoscolicidal rates significantly higher than the control group across different time intervals. Interestingly, PZQ demonstrated the best performance at 120 min. The trend continued at higher concentrations (0.5 mg/mL and 1 mg/mL), where the combined formulations, particularly ABZ + MBZ, consistently outperformed individual drugs regarding protoscolicidal rates. These findings underscore the potential synergistic effects of drug combinations and highlight the significance of nanocapsule-based drug delivery systems in enhancing the therapeutic outcomes of these anthelmintic drugs.
Using chitosan–ABZ (ChABZ) and chitosan–PZQ (ChPZQ) nanoparticles in both in vitro and in vivo experiments, researchers assessed the efficacy of ChABZ and ChPZQ in treating hydatid cysts [93]. A wide range of ChABZ and ChPZQ nanoparticle concentrations were used to treat microcysts formed in culture with either ABZ alone or in combination, as well as ABZ + PZQ suspensions and the combination of the two. An in vitro test demonstrated that combining ChABZ and ChPZQ nanoparticles showed significantly higher efficacy than ABZ + PZQ in combination. During prophylactic experiments in which ChABZ + ChPZQ nanoparticles were applied to infected mice, the amount and size of cysts were significantly reduced compared to those treated with the control group.
An investigation conducted by Fateh et al. in 2021 examined whether ABZ nanocrystals impacted the viability of E. granulosus s.s. protoscolices [94]. The nanocrystals had an average size and hydrodynamic diameter of 976 ± 218 and 1334 ± 502 nm, respectively. Using both morphological and molecular techniques, fertile hydatid cysts were obtained from slaughtered sheep’s livers, and those cysts were identified as belonging to E. granulosus s.s. based on their 100% similarity in morphology and genotype. They were tested on a small sample size to ascertain whether the protoscolices were susceptible to ABZ nanocrystals. There was a significant reduction in protoscolex viability due to the ingestion of 1 µg/mL nanocrystals and ABZ within 17 and 23 days, respectively. This indicates ABZ nanocrystals’ effectiveness in eliminating protoscolices.
According to a previous study, nanoformulations of ABZ were studied on protoscolices in vitro and in vivo to assess how they affected them [95]. Solid lipid nanoparticles containing ABZ were prepared using the microemulsion method. As a result of the successful storage of protoscolices in RPMI 1640 for one week, the survival of protoscolices was examined on days 3 and 7 of the experiment by testing the effect of ABZ at both 250 and 500 μg/mL concentrations. Protoscolices were injected into mice on day 3 at 250 μg/mL concentration to ascertain their pathogenicity. After three months, an autopsy was performed on the mice to assess their pathogenicity. Both ABZ and nano-ABZ showed the highest protoscolicidal efficacy on day 7 at both concentrations.
Moreover, researchers developed ABZ-loaded nanostructured lipid carriers (ABZ-NLCs) using a hot high-speed homogenization method [96]. This method increased the therapeutic efficiency of ABZ against E granulosus protoscolices and metacestodes. Methylene blue exclusion testing, scanning, and transmission electron microscopy were used to monitor the in vitro treatment of protoscolices and microcysts with free ABZ and ABZ–NLCs. An intraperitoneal injection of viable protoscolices was performed one day before administering chemoprophylactic treatment to Balb/C mice. After mice were treated with drugs intragastrically every day for 30 days, cyst size and frequency were evaluated to determine how effective the prophylactic treatment was. ABZ-NLCs were added to all protoscolices after 18 days of incubation, and the protoscolices were killed. On the other hand, 51.25% of those incubated in ABZ at 10 g/mL remained viable after 18 days. Between the ABZ-NLC group and the free ABZ group, hydatid cysts recovered from the ABZ–NLC group showed statistically significant differences in mean weight (p < 0.05), which resulted in a prophylactic efficacy of 92.45% and 38.53%, respectively. Germinal layer ultrastructural changes were observed in cysts treated with ABZ-NLCs.
In Table 1 is summarized the in vitro studies on the effects of nanomedicines against hydatid disease in recent years (Table 1).
Although innovative approaches can be successful, implementing them is challenging [110]. Alternative treatments, including surgery and cyst inactivation methods, are important. Surgery, one of the most effective treatments for CE, physically removes the cyst and yields health improvements in the patient, preventing serious complications. However, cyst inactivation methods can help supplement medical and surgical treatments and improve results [111].
It cannot be overstated that the accurate identification of species and genotypes is a cornerstone in treating CE. The potential for different species and genotypes to exhibit varied responses to treatments underscores the importance of this aspect. While this issue was not the focus of our present research, it is a crucial consideration for future studies. Identifying these parameters can significantly enhance treatment outcomes and enable more targeted treatments. Previous studies in this region of Iran [112,113] have indicated a high probability of the G1 genotype, further underscoring the importance of this aspect in our research.
Understanding the complex interactions between drug encapsulation, release kinetics, and immune interactions requires extensive research and development. Rigorous testing is necessary to ensure our nanotechnologies’ safety, effectiveness, and long-lasting benefits. This testing in both in vitro and in vivo models guarantees that our proposed nanotechnologies are thoroughly vetted and ready for implementation.

5. Conclusions

This study showed that nanocapsules of albendazole, mebendazole, and praziquantel have an effect on hydatid cyst protoscolices and are significantly better than the classic formulations of these drugs. Moreover, by controlling the release and increasing the absorption of the drugs, these nanocapsules strengthen their antiparasitic effects and reduce possible side effects. Therefore, nanocapsules can provide a more effective solution for cystic echinococcosis treatment and significantly improve existing treatments’ therapeutic efficiency and safety. Finally, the greatest protoscolicidal activity was evidenced by the albendazole + mebendazole nanocapsules at a concentration of 1 mg/mL after 120 min. The benefits of nanotechnology must be balanced with the risks associated with nanotoxicity, and it is essential to take a cautious approach. Cystic echinococcosis management could be revolutionized by integrating nanocapsules as research and development in this area progress.

Author Contributions

Conceptualization, H.B.; methodology, N.S., S.S., and A.R.; software, N.S.; validation, H.B., C.S., N.S., G.M., and S.S.; formal analysis, N.S.; investigation, N.S.; resources, H.B.; data curation, N.S.; writing—original draft preparation, S.S.; writing—review and editing, S.S., C.S., and N.S.; visualization, H.B., G.M., and C.S.; supervision, H.B.; project administration, H.B.; funding acquisition, H.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ferdowsi University of Mashhad, grant number 24224; charge related to article processing was managed by the Istituto Zooprofilattico Sperimentale della Sardegna.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets generated during the current study are available from the corresponding author upon reasonable request.

Acknowledgments

We thank the research deputy of the Ferdowsi University of Mashhad for supporting us. We want to thank Amir Hossein Atazadeh (a graduate of Ferdowsi University of Mashhad) for guiding us in conducting the statistical analysis.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gessese, A.T. Review on epidemiology and public health significance of hydatidosis. Vet. Med. Int. 2020, 2020, 8859116. [Google Scholar] [CrossRef] [PubMed]
  2. Hemphill, A.; Lundström-Stadelmann, B. Echinococcus: The model cestode parasite. Parasitology 2021, 148, 1401–1405. [Google Scholar] [CrossRef] [PubMed]
  3. Sadr, S.; Charbgoo, A.; Borji, H.; Hajjafari, A. Interactions between innate immunity system and Echinococcus granulosus: Permission for vaccine development. Ser. Med. Sci. 2022, 3, 1–18. [Google Scholar]
  4. Thompson, R. Biology and systematics of Echinococcus. Adv. Parasitol. 2017, 95, 65–109. [Google Scholar] [CrossRef]
  5. Woolsey, I.D.; Miller, A.L. Echinococcus granulosus sensu lato and Echinococcus multilocularis: A review. Res. Vet. Sci. 2021, 135, 517–522. [Google Scholar] [CrossRef]
  6. Mandal, S.; Mandal, M.D. Human cystic echinococcosis: Epidemiologic, zoonotic, clinical, diagnostic and therapeutic aspects. Asian Pac. J. Trop. Med. 2012, 5, 253–260. [Google Scholar] [CrossRef]
  7. Pal, M.; Alemu, H.H.; Marami, L.M.; Garedo, D.R.; Bodena, E.B. Cystic Echincoccoosis: A comprehensive review on life cycle, epidemiology, pathogenesis, clinical spectrum, diagnosis, public health and economic implications, treatment, and control. Int. J. Clin. Exp. Med. Res. 2022, 6, 131–141. [Google Scholar] [CrossRef]
  8. Umhang, G.; Bastien, F.; Cartet, A.; Ahmad, H.; van der Ark, K.; Berg, R.; Bonelli, P.; Davidson, R.K.; Deplazes, P.; Deksne, G. Detection of Echinococcus spp. and other taeniid species in lettuces and berries: Two international multicenter studies from the MEmE project. bioRxiv 2024, 189, 598207. [Google Scholar] [CrossRef]
  9. Benelli, G.; Wassermann, M.; Brattig, N.W. Insects dispersing taeniid eggs: Who and how? Vet. Parasitol. 2021, 295, 109450. [Google Scholar] [CrossRef]
  10. Serra, E.; Masu, G.; Chisu, V.; Cappai, S.; Masala, G.; Loi, F.; Piseddu, T. Environmental contamination by Echinococcus spp. eggs as a risk for human health in educational farms of Sardinia, Italy. Vet. Sci. 2022, 9, 143. [Google Scholar] [CrossRef]
  11. Badwaik, N.; Gharde, P.; Shinde, R.K.; Tayade, H.; Navandhar, P.S.; Patil, M. Hydatid cyst or echinococcosis: A comprehensive review of transmission, clinical manifestations, diagnosis, and multidisciplinary treatment. Cureus 2024, 16, e63713. [Google Scholar] [CrossRef] [PubMed]
  12. Moro, P.L.; Schantz, P.M. 281—Echinococcus species (agents of cystic, alveolar, and polycystic echinococcosis). In Principles and Practice of Pediatric Infectious Diseases; Saunders: Philadephia, PA, USA, 2012; pp. 1356–1362.e2. [Google Scholar] [CrossRef]
  13. Mathivathani, C.; Ajaykumar, V.; Bora, C. Epidemiology and public health significance of hydatidosis: A review. Curr. J. Appl. Sci. Technol. 2023, 42, 19–26. [Google Scholar] [CrossRef]
  14. Santucciu, C.; Bonelli, P.; Peruzzu, A.; Fancellu, A.; Marras, V.; Carta, A.; Mastrandrea, S.; Bagella, G.; Piseddu, T.; Profili, S.; et al. Cystic echinococcosis: Clinical, immunological, and biomolecular evaluation of patients from Sardinia (Italy). Pathogens 2020, 9, 907. [Google Scholar] [CrossRef]
  15. Brunetti, E.; Tamarozzi, F.; Macpherson, C.; Filice, C.; Piontek, M.S.; Kabaalioglu, A.; Dong, Y.; Atkinson, N.; Richter, J.; Schreiber-Dietrich, D.; et al. Ultrasound and cystic echinococcosis. Ultrasound Int. Open 2018, 4, E70–E78. [Google Scholar] [CrossRef]
  16. Siles-Lucas, M.; Casulli, A.; Conraths, F.; Müller, N. Laboratory diagnosis of Echinococcus spp. in human patients and infected animals. Adv. Parasitol. 2017, 96, 159–257. [Google Scholar] [CrossRef]
  17. Peruzzu, A.; Mastrandrea, S.; Fancellu, A.; Bonelli, P.; Muehlethaler, K.; Masala, G.; Santucciu, C. Comparison and evaluation of analytic and diagnostic performances of four commercial kits for the detection of antibodies against Echinococcus granulosus and multilocularis in human sera. Comp. Immunol. Microbiol. Infect. Dis. 2022, 86, 101816. [Google Scholar] [CrossRef]
  18. Brunetti, E.; Kern, P.; Vuitton, D.A. Expert consensus for the diagnosis and treatment of cystic and alveolar echinococcosis in humans. Acta Trop. 2010, 114, 1–16. [Google Scholar] [CrossRef]
  19. Santucciu, C.; Bozzi, E.; Profili, S.; Porcu, A.; Masala, G.; Babudieri, S.; Mastrandrea, S. Investigation of patients with new infection of echinococcal cyst in Sardinia, Italy. Diagn. Microbiol. Infect. Dis. 2024, 109, 116224. [Google Scholar] [CrossRef]
  20. Vural, G.; Yardimci, M.; Koçak, M.; Yasar, T.; Kurt, A.; Harem, I.; Carradori, S.; Sciamanna, I.; Siles-Lucas, M.; Fabiani, M.; et al. Efficacy of novel albendazole salt formulations against secondary cystic echinococcosis in experimentally infected mice. Parasitology 2020, 147, 1425–1432. [Google Scholar] [CrossRef]
  21. 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. [Google Scholar] [CrossRef]
  22. El-On, J. Benzimidazole treatment of cystic echinococcosis. Acta Trop. 2003, 85, 243–252. [Google Scholar] [CrossRef] [PubMed]
  23. Walker, M.; Rossignol, J.F.; Torgerson, P.; Hemphill, A. In vitro effects of nitazoxanide on Echinococcus granulosus protoscoleces and metacestodes. J. Antimicrob. Chemother. 2004, 54, 609–616. [Google Scholar] [CrossRef] [PubMed]
  24. Chai, J.-Y.; Jung, B.-K.; Hong, S.-J. Albendazole and mebendazole as anti-parasitic and anti-cancer agents: An update. Korean J. Parasitol. 2021, 59, 189–225. [Google Scholar] [CrossRef]
  25. Ates-Alagoz, Z. Antimicrobial activities of 1-H-benzimidazole-based molecules. Curr. Top. Med. Chem. 2016, 16, 2953–2962. [Google Scholar] [CrossRef]
  26. Shnawa, B.H.; Al-Ali, S.J.; Swar, S.O. Nanoparticles as a new approach for treating hydatid cyst disease. In Veterinary Pathobiology and Public Health; Unique Scientific Publishers: Faisalabad, Pakistan, 2021. [Google Scholar] [CrossRef]
  27. Albalawi, A.E.; Alanazi, A.D.; Baharvand, P.; Sepahvand, M.; Mahmoudvand, H. High potency of organic and inorganic nanoparticles to treat cystic echinococcosis: An evidence-based review. Nanomaterials 2020, 10, 2538. [Google Scholar] [CrossRef]
  28. Shakibaie, M.; Khalaf, A.K.; Rashidipour, M.; Mahmoudvand, H. Effects of green synthesized zinc nanoparticles alone and along with albendazole against hydatid cyst protoscoleces. Ann. Med. Surg. 2022, 78, 103746. [Google Scholar] [CrossRef]
  29. Cheraghipour, K.; Rouzbahani, A.K.; Fallahi, S.; Taherpour, F.; Moradifard, F.; Shakib, P.; Lashgarian, H.E.; Marzban, A. Recent advances in therapeutic strategies against hydatid cysts using nanomaterials: A systematic review. Lett. Drug Des. Discov. 2023, 20, 1185–1193. [Google Scholar] [CrossRef]
  30. Norouzi, R.; Ataei, A.; Hejazy, M.; Noreddin, A.; El Zowalaty, M.E. Scolicidal effects of nanoparticles against hydatid cyst protoscolices in vitro. Int. J. Nanomed. 2020, 15, 1095–1100. [Google Scholar] [CrossRef]
  31. Ezzatkhah, F.; Khalaf, A.K.; Mahmoudvand, H. Copper nanoparticles: Biosynthesis, characterization, and protoscolicidal effects alone and combined with albendazole against hydatid cyst protoscoleces. Biomed. Pharmacother. 2021, 136, 111257. [Google Scholar] [CrossRef]
  32. Bakhtiar, N.M.; Akbarzadeh, A.; Casulli, A.; Mahami-Oskouei, M.; Ahmadpour, E.; Nami, S.; Rostami, A.; Spotin, A. Therapeutic efficacy of nanocompounds in the treatment of cystic and alveolar echinococcoses: Challenges and future prospects. Parasitol. Res. 2019, 118, 2455–2466. [Google Scholar] [CrossRef]
  33. Norouzi, R.; Hejazy, M.; Ataei, A. Scolicidal activity of zinc oxide nanoparticles against hydatid cyst protoscolices in vitro. Nanomed. Res. J. 2019, 4, 23–28. [Google Scholar] [CrossRef]
  34. Cheraghipour, K.; Azarhazine, M.; Zivdari, M.; Beiranvand, M.; Shakib, P.; Rashidipour, M.; Mardanshah, O.; Mohaghegh, M.A.; Marzban, A. Evaluation of scolicidal potential of salicylate coated zinc nanoparticles against Echinococcus granulosus protoscoleces. Exp. Parasitol. 2023, 246, 108456. [Google Scholar] [CrossRef] [PubMed]
  35. Panwar, P.; Pandey, B.; Lakhera, P.; Singh, K. Preparation, characterization, and in vitro release study of albendazole-encapsulated nanosize liposomes. Int. J. Nanomed. 2010, 5, 101–108. [Google Scholar] [CrossRef]
  36. Bondarenko, O.; Mortimer, M.; Kahru, A.; Feliu, N.; Javed, I.; Kakinen, A.; Lin, S.; Xia, T.; Song, Y.; Davis, T.P.; et al. Nanotoxicology and nanomedicine: The Yin and Yang of nano-bio interactions for the new decade. Nano Today 2021, 39, 101184. [Google Scholar] [CrossRef]
  37. Vega-Vásquez, P.; Mosier, N.S.; Irudayaraj, J. Nanoscale drug delivery systems: From medicine to agriculture. Front. Bioeng. Biotechnol. 2020, 8, 79. [Google Scholar] [CrossRef]
  38. Inbathamizh, L.; Harsha Varthan, M.; Kumar, R.R.; Rohinth, M.; Tawfeeq Ahmed, Z. Nanotechnology: Ethical impacts, health issues, and safety issues. In Modern Nanotechnology: Green Synthesis, Sustainable Energy and Impacts; Springer Nature: Cham, Switzerland, 2023; Volume 2, pp. 455–477. [Google Scholar] [CrossRef]
  39. Bajwa, H.U.R.; Khan, M.K.; Abbas, Z.; Riaz, R.; Rehman, T.U.; Abbas, R.Z.; Aleem, M.T.; Abbas, A.; Almutairi, M.M.; Alshammari, F.A.; et al. Nanoparticles: Synthesis and their role as potential drug candidates for the treatment of parasitic diseases. Life 2022, 12, 750. [Google Scholar] [CrossRef]
  40. Rajwar, T.K.; Pradhan, D.; Halder, J.; Rai, V.K.; Kar, B.; Ghosh, G.; Rath, G. Opportunity in nanomedicine to counter the challenges of current drug delivery approaches used for the treatment of malaria: A review. J. Drug Target. 2023, 31, 354–368. [Google Scholar] [CrossRef]
  41. Jafari, M.; Abolmaali, S.S.; Tamaddon, A.M.; Zomorodian, K.; Shahriarirad, B. Nanotechnology approaches for delivery and targeting of Amphotericin B in fungal and parasitic diseases. Nanomedicine 2021, 16, 857–877. [Google Scholar] [CrossRef]
  42. Makhdoumi, P.; Karimi, H.; Khazaei, M. Review on metal-based nanoparticles: Role of reactive oxygen species in renal toxicity. Chem. Res. Toxicol. 2020, 33, 2503–2514. [Google Scholar] [CrossRef]
  43. Rana, S.V.S. Recent advances on renal toxicity of engineered nanoparticles-a review. J. Toxicol. Risk Assess. 2021, 7, 36. [Google Scholar] [CrossRef]
  44. Tao, Z.; Zhou, Q.; Zheng, T.; Mo, F.; Ouyang, S. Iron oxide nanoparticles in the soil environment: Adsorption, transformation, and environmental risk. J. Hazard. Mater. 2023, 459, 132107. [Google Scholar] [CrossRef] [PubMed]
  45. Lamas, B.; Martins Breyner, N.; Houdeau, E. Impacts of foodborne inorganic nanoparticles on the gut microbiota-immune axis: Potential consequences for host health. Part. Fibre Toxicol. 2020, 17, 19. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, P.; Guo, Z.; Zhang, Z.; Fu, H.; White, J.C.; Lynch, I. Nanomaterial transformation in the soil—Plant system: Implications for food safety and application in agriculture. Small 2020, 16, 2000705. [Google Scholar] [CrossRef]
  47. Hajjafari, A.; Sadr, S.; Santucciu, C.; Masala, G.; Bayat, M.; Lotfalizadeh, N.; Borji, H.; Partovi Moghaddam, S.; Hajjafari, K. Advances in detecting cystic echinococcosis in intermediate hosts and new diagnostic tools: A literature review. Vet. Sci. 2024, 11, 227. [Google Scholar] [CrossRef]
  48. Sadr, S.; Ahmadi Simab, P.; Niazi, M.; Yousefsani, Z.; Lotfalizadeh, N.; Hajjafari, A.; Borji, H. Anti-inflammatory and immunomodulatory effects of mesenchymal stem cell therapy on parasitic drug resistance. Expert Rev. Anti-Infect. Ther. 2024, 22, 435–451. [Google Scholar] [CrossRef]
  49. Aboelsoued, D.; Toaleb, N.I.; Ibrahim, S.; Ibrahim, S. In vitro and ex vivo protoscolicidal effect of poly(amidoamine) nanoemulsion against Echinococcus granulosus. Sci. Rep. 2024, 14, 11957. [Google Scholar] [CrossRef]
  50. Varshney, M.; Morey, T.E.; Shah, D.O.; Flint, J.A.; Moudgil, B.M.; Seubert, C.N.; Dennis, D.M. Pluronic microemulsions as nanoreservoirs for extraction of bupivacaine from normal saline. J. Am. Chem. Soc. 2004, 126, 5108–5112. [Google Scholar] [CrossRef]
  51. Wen, H.; Vuitton, L.; Tuxun, T.; Li, J.; Vuitton, D.A.; Zhang, W.; McManus, D.P. Echinococcosis: Advances in the 21st century. Clin. Microbiol. Rev. 2019, 32, e00075-18. [Google Scholar] [CrossRef]
  52. Diaz, A.; Casaravilla, C.; Barrios, A.A.; Ferreira, A.M. Parasite molecules and host responses in cystic echinococcosis. Parasite Immunol. 2016, 38, 193–205. [Google Scholar] [CrossRef]
  53. Goussard, P.; Eber, E.; Mfingwana, L.; Nel, P.; Schubert, P.; Janson, J.; Pitcher, R.; le Roux, C. Paediatric pulmonary echinococcosis: A neglected disease. Paediatr. Respir. Rev. 2022, 43, 11–23. [Google Scholar] [CrossRef]
  54. Lupia, T.; Corcione, S.; Guerrera, F.; Costardi, L.; Ruffini, E.; Pinna, S.M.; De Rosa, F.G. Pulmonary echinococcosis or lung hydatidosis: A narrative review. Surg. Infect. 2021, 22, 485–495. [Google Scholar] [CrossRef] [PubMed]
  55. Bakoyiannis, A.; Delis, S.; Triantopoulou, C.; Dervenis, C. Rare cystic liver lesions: A diagnostic and managing challenge. World J. Gastroenterol. 2013, 19, 7603–7619. [Google Scholar] [CrossRef] [PubMed]
  56. Brehm, K.; Koziol, U. Echinococcus—Host interactions at cellular and molecular levels. Adv. Parasitol. 2017, 95, 147–212. [Google Scholar] [CrossRef]
  57. Díaz, Á.; Barrios, A.A.; Grezzi, L.; Mouhape, C.; Jenkins, S.J.; Allen, J.E.; Casaravilla, C. Immunology of a unique biological structure: The Echinococcus laminated layer. Protein Cell 2023, 14, 87–104. [Google Scholar] [CrossRef]
  58. Larrieu, E.; Gavidia, C.M.; Lightowlers, M.W. Control of cystic echinococcosis: Background and prospects. Zoonoses Public Health 2019, 66, 889–899. [Google Scholar] [CrossRef]
  59. Rawla, P.; Sunkara, T.; Muralidharan, P.; Raj, J.P. An updated review of cystic hepatic lesions. Clin. Exp. Hepatol. 2019, 5, 22–29. [Google Scholar] [CrossRef]
  60. Alsharedeh, R.H.; Rezigue, M.; Bashatwah, R.M.; Amawi, H.; Aljabali, A.A.; Obeid, M.A.; Tambuwala, M.M. Nanomaterials as a potential target for infectious parasitic agents. Curr. Drug Deliv. 2024, 21, 828–851. [Google Scholar] [CrossRef]
  61. Kian, M.; Mirzavand, S.; Sharifzadeh, S.; Kalantari, T.; Ashrafmansouri, M.; Nasri, F. Efficacy of mesenchymal stem cells therapy in parasitic infections: Are anti-parasitic drugs combined with mscs more effective? Acta Parasitol. 2022, 67, 1487–1499. [Google Scholar] [CrossRef]
  62. Khezerlou, A.; Alizadeh-Sani, M.; Azizi-Lalabadi, M.; Ehsani, A. Nanoparticles and their antimicrobial properties against pathogens including bacteria, fungi, parasites and viruses. Microb. Pathog. 2018, 123, 505–526. [Google Scholar] [CrossRef]
  63. Idris, O.A.; Wintola, O.A.; Afolayan, A.J. Helminthiases; prevalence, transmission, host-parasite interactions, resistance to common synthetic drugs and treatment. Heliyon 2019, 5, e01161. [Google Scholar] [CrossRef]
  64. Stamatakos, M.; Sargedi, C.; Stefanaki, C.; Safioleas, C.; Matthaiopoulou, I.; Safioleas, M. Anthelminthic treatment: An adjuvant therapeutic strategy against Echinococcus granulosus. Parasitol. Int. 2009, 58, 115–120. [Google Scholar] [CrossRef] [PubMed]
  65. Hemphill, A.; Stadelmann, B.; Rufener, R.; Spiliotis, M.; Boubaker, G.; Müller, J.; Müller, N.; Gorgas, D.; Gottstein, B. Treatment of echinococcosis: Albendazole and mebendazole—What else? Parasite 2014, 21, 70. [Google Scholar] [CrossRef] [PubMed]
  66. Gebreyes, W.A.; Dupouy-Camet, J.; Newport, M.J.; Oliveira, C.J.; Schlesinger, L.S.; Saif, Y.M.; Kariuki, S.; Saif, L.J.; Saville, W.; Wittum, T.; et al. The global one health paradigm: Challenges and opportunities for tackling infectious diseases at the human, animal, and environment interface in low-resource settings. PLoS Negl. Trop. Dis. 2014, 8, e3257. [Google Scholar] [CrossRef] [PubMed]
  67. Lightowlers, M.W.; Gasser, R.B.; Hemphill, A.; Romig, T.; Tamarozzi, F.; Deplazes, P.; Torgerson, P.R.; Garcia, H.H.; Kern, P. Advances in the treatment, diagnosis, control and scientific understanding of taeniid cestode parasite infections over the past 50 years. Int. J. Parasitol. 2021, 51, 1167–1192. [Google Scholar] [CrossRef]
  68. Borhani, M.; Fathi, S.; Darabi, E.; Jalousian, F.; Simsek, S.; Ahmed, H.; Kesik, H.K.; Hosseini, S.H.; Romig, T.; Harandi, M.F.; et al. Echinococcoses in Iran, Turkey, and Pakistan: Old diseases in the new millennium. Clin. Microbiol. Rev. 2021, 34, e0029020. [Google Scholar] [CrossRef]
  69. Barabadi, H.; Honary, S.; Ali Mohammadi, M.; Ahmadpour, E.; Rahimi, M.T.; Alizadeh, A.; Naghibi, F.; Saravanan, M. Green chemical synthesis of gold nanoparticles by using Penicillium aculeatum and their scolicidal activity against hydatid cyst protoscolices of Echinococcus granulosus. Environ. Sci. Pollut. Res. 2017, 24, 5800–5810. [Google Scholar] [CrossRef]
  70. Sun, Y.; Chen, D.; Pan, Y.; Qu, W.; Hao, H.; Wang, X.; Liu, Z.; Xie, S. Nanoparticles for antiparasitic drug delivery. Drug Deliv. 2019, 26, 1206–1221. [Google Scholar] [CrossRef]
  71. Stanicki, D.; Vangijzegem, T.; Ternad, I.; Laurent, S. An update on the applications and characteristics of magnetic iron oxide nanoparticles for drug delivery. Expert Opin. Drug Deliv. 2022, 19, 321–335. [Google Scholar] [CrossRef]
  72. Yohan, D.; Chithrani, B.D. Applications of nanoparticles in nanomedicine. J. Biomed. Nanotechnol. 2014, 10, 2371–2392. [Google Scholar] [CrossRef]
  73. Pensel, P.; Paredes, A.; Albani, C.M.; Allemandi, D.; Bruni, S.S.; Palma, S.D.; Elissondo, M.C. Albendazole nanocrystals in experimental alveolar echinococcosis: Enhanced chemoprophylactic and clinical efficacy in infected mice. Vet. Parasitol. 2018, 251, 78–84. [Google Scholar] [CrossRef]
  74. Gamboa, G.V.U.; Pensel, P.E.; Elissondo, M.C.; Bruni, S.F.S.; Benoit, J.-P.; Palma, S.D.; Allemandi, D.A. Albendazole-lipid nanocapsules: Optimization, characterization and chemoprophylactic efficacy in mice infected with Echinococcus granulosus. Exp. Parasitol. 2019, 198, 79–86. [Google Scholar] [CrossRef] [PubMed]
  75. Fabbri, J.; Pensel, P.E.; Albani, C.M.; Lopez, L.M.; Simonazzi, A.; Bermudez, J.M.; Palma, S.D.; Elissondo, M.C. Albendazole solid dispersions against alveolar echinococcosis: A pharmacotechnical strategy to improve the efficacy of the drug. Parasitology 2020, 147, 1026–1031. [Google Scholar] [CrossRef]
  76. Lopez, L.M.; Pensel, P.E.; Fabbri, J.; Albani, C.M.; Elissondo, N.; Gambino, G.; Elissondo, M.C. The combination of carvacrol and albendazole enhanced the efficacy of monotherapy in experimental alveolar echinococcosis. Acta Trop. 2022, 225, 106198. [Google Scholar] [CrossRef] [PubMed]
  77. Albani, C.; Pensel, P.; Fabbri, J.; Albanese, A.; Paladini, A.; Elissondo, M.C. Experimental treatment of cystic echinococcosis: Combination therapy with carvacrol and thymol versus albendazole. Exp. Parasitol. 2023, 244, 108430. [Google Scholar] [CrossRef] [PubMed]
  78. Leong, E.W.; Ge, R. Lipid nanoparticles as delivery vehicles for inhaled therapeutics. Biomedicines 2022, 10, 2179. [Google Scholar] [CrossRef]
  79. Pavoni, L.; Pavela, R.; Cespi, M.; Bonacucina, G.; Maggi, F.; Zeni, V.; Canale, A.; Lucchi, A.; Bruschi, F.; Benelli, G. Green micro-and nanoemulsions for managing parasites, vectors and pests. Nanomaterials 2019, 9, 1285. [Google Scholar] [CrossRef]
  80. Bhatia, M.; Kumar, S.; Kapoor, A.; Lohan, S. A review on the drug delivery strategies for parasitic infections: Scope and assertion. Drug Deliv. Lett. 2022, 12, 109–121. [Google Scholar] [CrossRef]
  81. Omidian, H.; Mfoafo, K. Exploring the potential of nanotechnology in pediatric healthcare: Advances, challenges, and future directions. Pharmaceutics 2023, 15, 1583. [Google Scholar] [CrossRef]
  82. Sadr, S.; Lotfalizadeh, N.; Abbasi, A.M.; Soleymani, N.; Hajjafari, A.; Roohbaksh Amooli Moghadam, E.; Borji, H. Challenges and prospective of enhancing hydatid cyst chemotherapy by nanotechnology and the future of nanobiosensors for diagnosis. Trop. Med. Infect. Dis. 2023, 8, 494. [Google Scholar] [CrossRef]
  83. Chun, S.; Muthu, M.; Gopal, J. Nanotoxic impacts on staple food crops: There’s plenty of room for the unpredictables. Crit. Rev. Food Sci. Nutr. 2020, 60, 3725–3736. [Google Scholar] [CrossRef]
  84. Ettlinger, R.; Lächelt, U.; Gref, R.; Horcajada, P.; Lammers, T.; Serre, C.; Couvreur, P.; Morris, R.E.; Wuttke, S. Toxicity of metal—Organic framework nanoparticles: From essential analyses to potential applications. Chem. Soc. Rev. 2022, 51, 464–484. [Google Scholar] [CrossRef]
  85. Ren, C.; Hu, X.; Zhou, Q. Influence of environmental factors on nanotoxicity and knowledge gaps thereof. NanoImpact 2016, 2, 82–92. [Google Scholar] [CrossRef]
  86. Wong, H.L.; Wu, X.Y.; Bendayan, R. Nanotechnological advances for the delivery of CNS therapeutics. Adv. Drug Deliv. Rev. 2012, 64, 686–700. [Google Scholar] [CrossRef] [PubMed]
  87. Date, A.A.; Joshi, M.D.; Patravale, V.B. Parasitic diseases: Liposomes and polymeric nanoparticles versus lipid nanoparticles. Adv. Drug Deliv. Rev. 2007, 59, 505–521. [Google Scholar] [CrossRef] [PubMed]
  88. Pandian, S.R.K.; Panneerselvam, T.; Pavadai, P.; Govindaraj, S.; Ravishankar, V.; Palanisamy, P.; Sampath, M.; Sankaranarayanan, M.; Kunjiappan, S. Nano based approach for the treatment of neglected tropical diseases. Front. Nanotechnol. 2021, 3, 665274. [Google Scholar] [CrossRef]
  89. Mengarda, A.C.; Iles, B.; Longo, J.P.F.; de Moraes, J. Recent trends in praziquantel nanoformulations for helminthiasis treatment. Expert Opin. Drug Deliv. 2022, 19, 383–393. [Google Scholar] [CrossRef]
  90. Purohit, D.; Jalwal, P.; Manchanda, D.; Saini, S.; Verma, R.; Kaushik, D.; Mittal, V.; Kumar, M.; Bhattacharya, T.; Rahman, M.H.; et al. Nanocapsules: An emerging drug delivery system. Recent Pat. Nanotechnol. 2023, 17, 190–207. [Google Scholar] [CrossRef]
  91. Mahajan, K.; Bhattacharya, S. The advancement and obstacles in improving the stability of nanocarriers for precision drug delivery in the field of nanomedicine. Curr. Top. Med. Chem. 2024, 24, 686–721. [Google Scholar] [CrossRef]
  92. de Carvalho Moreira, L.M.C.; de Sousa Silva, A.B.A.; de Araújo Medeiros, K.; Oshiro Júnior, J.A.; da Silva, D.T.C.; de Lima Damasceno, B.P.G. Effectiveness in vivo and in vitro of polymeric nanoparticles as a drug release system in the treatment of leishmaniasis. Curr. Med. Chem. 2024, 31, 286–307. [Google Scholar] [CrossRef]
  93. Torabi, N.; Dobakhti, F.; Faghihzadeh, S.; Haniloo, A. In vitro and in vivo effects of chitosan-praziquantel and chitosan-albendazole nanoparticles on Echinococcus granulosus metacestodes. Parasitol. Res. 2018, 117, 2015–2023. [Google Scholar] [CrossRef]
  94. Fateh, R.; Norouzi, R.; Mirzaei, E.; Nissapatron, V.; Nawaz, M.; Khalifeh-Gholi, M.; Hamta, A.; Sadati, S.J.A.; Siyadatpanah, A.; Bafghi, A.F. In vitro evaluation of albendazole nanocrystals against Echinococcus granulosus protoscolices. Ann. Parasitol. 2021, 67, 203–212. [Google Scholar] [CrossRef] [PubMed]
  95. Aminpour, S.; Rafiei, A.; Jelowdar, A.; Kouchak, M. Evaluation of the protoscolicidal effects of albendazole and albendazole loaded solid lipid nanoparticles. Iran. J. Parasitol. 2019, 14, 127–135. [Google Scholar] [CrossRef] [PubMed]
  96. Farhadi, M.; Haniloo, A.; Rostamizadeh, K.; Ahmadi, N. In vitro evaluation of albendazole-loaded nanostructured lipid carriers on Echinococcus granulosus microcysts and their prophylactic efficacy on experimental secondary hydatidosis. Parasitol. Res. 2021, 120, 4049–4060. [Google Scholar] [CrossRef] [PubMed]
  97. Mahmoud, S.M.; Mohemeed, A.A. Effect of cobalt oxide on the vitality of the protoscolex of Echinococcus granulosus isolated from middle hosts. J. Surv. Fish. Sci. 2023, 10 (Suppl. S3), 4608–4613. [Google Scholar]
  98. Raziani, Y.; Cheraghipour, K.; Shakibaie, M.; Yadegari, J.G.; Mahmoudvand, H. High potency of magnetic iron oxide nanoparticles covered by piroctone olamine against cystic echinococcosis. Biomed. Pharmacother. 2023, 161, 114536. [Google Scholar] [CrossRef]
  99. Teimouri, A.; Jafarpour Azami, S.; Hashemi Hafshejani, S.; Ghanimatdan, M.; Bahreini, M.S.; Alimi, R.; Sadjjadi, S.M. Protoscolicidal effects of curcumin nanoemulsion against protoscoleces of Echinococcus granulosus. BMC Complement. Med. Ther. 2023, 23, 124. [Google Scholar] [CrossRef]
  100. Mohammed, S.A.; Ali, A.A. Effect of selenium nanoparticles against protoscoleces of Echinococcus granulosus in vitro and hydatid cysts in mice. Iraqi J. Vet. Sci. 2022, 36, 195–202. [Google Scholar] [CrossRef]
  101. Shnawa, B.H.; Hamad, S.M.; Barzinjy, A.A.; Kareem, P.A.; Ahmed, M.H. Scolicidal activity of biosynthesized zinc oxide nanoparticles by Mentha longifolia L. leaves against Echinococcus granulosus protoscolices. Emergent Mater. 2022, 5, 683–693. [Google Scholar] [CrossRef]
  102. Aljanabi, A.A.; Al-mussawi, A.; Bashi, A.M. Effects of silver—Copper (core-shell) nanoparticles as antiparasitic drug in some blood parameters of mice. Indian J. Forensic Med. Toxicol. 2021, 15, 5279–5285. [Google Scholar] [CrossRef]
  103. Ibrahim, A.A.-J. Scolicidal activity of zirconium oxide (ZrO2) nanoparticles against protoscolices of hydatid cysts. Indian J. Forensic Med. Toxicol. 2020, 14, 469–472. [Google Scholar] [CrossRef]
  104. Salih, T.A.; Hassan, K.T.; Majeed, S.R.; Ibraheem, I.J.; Hassan, O.M.; Obaid, A. In vitro scolicidal activity of synthesised silver nanoparticles from aqueous plant extract against Echinococcus granulosus. Biotechnol. Rep. 2020, 28, e00545. [Google Scholar] [CrossRef] [PubMed]
  105. Ahmadpour, E.; Godrati-Azar, Z.; Spotin, A.; Norouzi, R.; Hamishehkar, H.; Nami, S.; Heydarian, P.; Rajabi, S.; Mohammadi, M.; Perez-Cordon, G. Nanostructured lipid carriers of ivermectin as a novel drug delivery system in hydatidosis. Parasite Vectors 2019, 12, 469. [Google Scholar] [CrossRef]
  106. Napooni, S.; Arbabi, M.; Delavari, M.; Hooshyar, H.; Rasti, S. Lethal effects of gold nanoparticles on protoscolices of hydatid cyst: In vitro study. Comp. Clin. Pathol. 2019, 28, 143–150. [Google Scholar] [CrossRef]
  107. Naseri, M.; Akbarzadeh, A.; Spotin, A.; Akbari, N.A.R.; Mahami-Oskouei, M.; Ahmadpour, E. Scolicidal and apoptotic activities of albendazole sulfoxide and albendazole sulfoxide-loaded PLGA-PEG as a novel nanopolymeric particle against Echinococcus granulosus protoscoleces. Parasitol. Res. 2016, 115, 4595–4603. [Google Scholar] [CrossRef]
  108. Mahmoudvand, H.; Harandi, M.F.; Shakibaie, M.; Aflatoonian, M.R.; ZiaAli, N.; Makki, M.S.; Jahanbakhsh, S. Scolicidal effects of biogenic selenium nanoparticles against protoscolices of hydatid cysts. Int. J. Surg. 2014, 12, 399–403. [Google Scholar] [CrossRef]
  109. Adas, G.; Arikan, S.; Kemik, O.; Oner, A.; Sahip, N.; Karatepe, O. Use of albendazole sulfoxide, albendazole sulfone, and combined solutions as scolicidal agents on hydatid cysts (in vitro study). World J. Gastroenterol. 2009, 15, 112–116. [Google Scholar] [CrossRef]
  110. Mohammadi, K.H.; Heidarpour, M.; Borji, H. In vivo therapeutic efficacy of the Allium sativum ME in experimentally Echinococcus granulosus infected mice. Comp. Immunol. Microbiol. Infect. Dis. 2018, 60, 23–27. [Google Scholar] [CrossRef]
  111. Mihetiu, A.; Bratu, D.; Sabau, D.; Nastase, O.; Sandu, A.; Tanasescu, C.; Boicean, A.; Ichim, C.; Todor, S.B.; Serban, D.; et al. Optimized strategies for managing abdominal hydatid cysts and their complications. Diagnostics 2024, 14, 1346. [Google Scholar] [CrossRef]
  112. Kamelli, M.; Borji, H.; Naghibi, A. Genetic identification of cattle hydatid cyst isolates from northeast and southwest of Iran. Ann. Parasitol. 2016, 62, 301–305. [Google Scholar] [CrossRef]
  113. Rokni, M.B. Echinococcosis/hydatidosis in Iran. Iran. J. Parasitol. 2009, 4, 1–16. Available online: https://www.sid.ir/EN/VEWSSID/J_pdf/114920090201.pdf (accessed on 17 May 2009).
Figure 1. The schematic of the methodology in the current study.
Figure 1. The schematic of the methodology in the current study.
Pathogens 13 00790 g001
Figure 2. Zeta potential spectra of the nanocapsules: control, loaded with ABZ range of 5 to −55 millivolts (mean: −35.78 mW), MBZ range of 0 to −48 millivolts (mean: −27.38 mW), and PZQ range of 0 to −43 millivolts (mean: −23.34 mW).
Figure 2. Zeta potential spectra of the nanocapsules: control, loaded with ABZ range of 5 to −55 millivolts (mean: −35.78 mW), MBZ range of 0 to −48 millivolts (mean: −27.38 mW), and PZQ range of 0 to −43 millivolts (mean: −23.34 mW).
Pathogens 13 00790 g002
Figure 3. The mean size of ABZ nanocapsules is 193.01 nm, MBZ is 170.40 nm, and PZQ is 180.44 nm.
Figure 3. The mean size of ABZ nanocapsules is 193.01 nm, MBZ is 170.40 nm, and PZQ is 180.44 nm.
Pathogens 13 00790 g003
Figure 4. SEM image of polymeric nanocapsules.
Figure 4. SEM image of polymeric nanocapsules.
Pathogens 13 00790 g004
Figure 5. The protoscolicidal rate of 0.25 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).
Figure 5. The protoscolicidal rate of 0.25 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).
Pathogens 13 00790 g005
Figure 6. The protoscolicidal rate of 0.5 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).
Figure 6. The protoscolicidal rate of 0.5 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).
Pathogens 13 00790 g006
Figure 7. Light microscopy images showing the viable protoscolices of Echinococcus granulosus (stained with 0.1% eosin) (A: ×40). Live and red stained dead protoscolices after exposure to nano-ABZ (0.5 mg/mL) after 60 min of exposure (stained with 0.1% eosin) (B: ×10).
Figure 7. Light microscopy images showing the viable protoscolices of Echinococcus granulosus (stained with 0.1% eosin) (A: ×40). Live and red stained dead protoscolices after exposure to nano-ABZ (0.5 mg/mL) after 60 min of exposure (stained with 0.1% eosin) (B: ×10).
Pathogens 13 00790 g007
Figure 8. The protoscolicidal rate of 1 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).
Figure 8. The protoscolicidal rate of 1 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).
Pathogens 13 00790 g008
Figure 9. Light microscopy images showing the viable protoscolices of Echinococcus granulosus (A: ×40). Live and red stained dead protoscolices after exposure to nano-ABZ (1 mg/mL) after 10 min of exposure (stained with 0.1% eosin) (B: ×40, C: ×10).
Figure 9. Light microscopy images showing the viable protoscolices of Echinococcus granulosus (A: ×40). Live and red stained dead protoscolices after exposure to nano-ABZ (1 mg/mL) after 10 min of exposure (stained with 0.1% eosin) (B: ×40, C: ×10).
Pathogens 13 00790 g009
Table 1. In vitro studies on the effects of nanomedicines against CE in recent years.
Table 1. In vitro studies on the effects of nanomedicines against CE in recent years.
NanomedicineDurationFindingsDosageReferences
Cobalt oxide (CO)5, 10, 15, 30, 60 minCO:
-Killed 100% of protoscolices of sheep liver cyst at 0.50 mg/mL in 60 min
-Killed 100% of protoscolices of cattle liver cyst at 0.100 in 60 min
-Killed 100% of protoscolices in goat liver cyst at 0.100 mg/mL in 30 and 60 min
0.025, 0.050, 0.100
(mg/mL)
Mahmoud, M.S. et al., 2023
[97]
Iron oxide nanoparticles coated with PO (FOMNPsP)10, 20, 30, 60 minFOMNPsP:
Killed 100% of protoscolices
-at 400 µg/mL after 10 min
-at 200 and 300 µg/mL after 20 min
-at 100 µg/mL after 30 min
100–400 μg/mLRaziani, Y. et al., 2023
[98]
Curcumin nanoemulsion
(CUR-NE)
10, 20, 30, 60, 120 minCUR-NE:
Killed 94% and 73.33% of protoscolices at 1250 and 625 (µg/mL) in 60 min
156, 312, 625, 1250
(µg/mL)
Teimouri, A. et al.,
2023
[99]
Selenium nanoparticles
(Se-NPs)
10, 20, 30, 60 minSe-NPs:
Killed 100% of protoscolices at 500 µg/mL after 20 min
50,100, 150, 200, 250, 350, 500 (µg/mL)Mohammed, M.A. et al., 2022
[100]
Zinc nanoparticles (Zn-NPs)5–60 minZn-NPs:
Killed 81.6% of protoscolices at 200 μg/mL
Zn-NPs + ALZ:
Killed 100% of protoscolices at 200 μg/mL after 10 min
50, 100, 200 (μg/mL) Zn-NPs alone and combined with albendazole
(100 μg/mL)
Shakibaie, M. et al., 2022
[28]
Biosynthesized zinc oxide nanoparticles (ZnO NPs)
by Mentha longifolia L. leaves
15, 30 min
and
1, 1.5,
2, 2.5 h
ZnO NPs:
Killed 100% of protoscolices at 400 ppm in 150 min
100, 200, 400 ppmShnawa., B.H. et al., 2022
[101]
Copper nanoparticles
(CuNPs)
5–60 minCuNPs:
Killed 73.3% of protoscolices at 750 mg/mL after 60 min
250, 500, 750 mg/mLEzzatkhah, F. et al., 2021
[31]
Silver–copper (core-shell) nanoparticles10, 30, 60 minConcentrations 250 and 500 mg/mL are the most effective in the
vitality of the protoscolices compared to 50 and 125 mg/mL
50, 125, 250, 500 mg/mLAljanabi, A.A. et al., 2021
[102]
Zirconium oxide (ZrO2)15,
30, 60 min.
ZrO2:
Killed 53.1% of protoscolices at 4000 μg/mL in 60 min
250, 500, 1000, 2000, 4000 μg/mLAbdul, A. and Ibrahim, J., 2020
[103]
-Piper nigrum (PN),
-Ziziphus Spina-Christi (ZSC),
-Eucalyptus globulus (EUCGLO)
+ Ag-NPs.
15, 30, 45 minKilled more than 90% of protoscolices in
minimum time.
1:10
2:10
3:10
(plants: Ag-NPs)
Salih, T.A. et al., 2020
[104]
Nanostructured lipid carriers of ivermectin15, 30, 60, 120, 150
min
NLC-loaded IVM:
Killed 100% of protoscolices at 400 and 800 μg/mL after 60 and 120 min
50, 100, 200, 400, 800 μg/mLAhmadpour, E. et al., 2019
[105]
Chitosan–praziquantel
(ChPZQ)
and
chitosan–albendazole (ChABZ)
Microcyst culture
flasks
for 16 days
There were no viable microcysts
observed on day 10 post-incubation with either 5 or 10 μg/mL
doses of ChABZ + ChPZQ combination
1, 5, 10 μg/mLTorabi, N. et al., 2018
[93]
Gold nanoparticles5, 10, 20, 30, 60 minGold nanoparticles: Killed 76% of protoscolices at 4000 μg/mL in 60 min250, 500, 1000, 2000, 4000 μg/mLNapooni, S. et al., 2018
[106]
-Albendazole sulfoxide

-ABZ-loaded PLGA-PEG
5, 10, 20, 30,
60 min
-ABZs:
Killed 100% of protoscolices at
200 μg/mL in 20, 30, and 60 min
-ABZ-loaded PLGA-PEG:
Killed 100% of protoscolices at 150
and 200 μg/mL in all
exposure times (5 to 60 min)
50, 100, 150, 200 (μg/mL)Naseri, M. et al., 2016
[107]
Selenium nanoparticles
(Se-NPs)
10, 20, 30, 60 minSe-NPs: Killed 100% of protoscolices at 250 mg/mL in 20 min50,100.150, 200, 250, 300, 350, 400, 450, 500 mg/mLMahmoudvand, H. et al., 2014
[108]
Albendazole sulfoxide, albendazole sulfone, and
combined solutions
5 and 10 minABZ sulfone: 97.3%;
ABZ sulfoxide: 98.4%;
the combined solution: 98.6% were effective in 5 min
50 μg/mLAdas, G. et al., 2009
[109]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Soleymani, N.; Sadr, S.; Santucciu, C.; Rahdar, A.; Masala, G.; Borji, H. Evaluation of the In-Vitro Effects of Albendazole, Mebendazole, and Praziquantel Nanocapsules against Protoscolices of Hydatid Cyst. Pathogens 2024, 13, 790. https://doi.org/10.3390/pathogens13090790

AMA Style

Soleymani N, Sadr S, Santucciu C, Rahdar A, Masala G, Borji H. Evaluation of the In-Vitro Effects of Albendazole, Mebendazole, and Praziquantel Nanocapsules against Protoscolices of Hydatid Cyst. Pathogens. 2024; 13(9):790. https://doi.org/10.3390/pathogens13090790

Chicago/Turabian Style

Soleymani, Nooshinmehr, Soheil Sadr, Cinzia Santucciu, Abbas Rahdar, Giovanna Masala, and Hassan Borji. 2024. "Evaluation of the In-Vitro Effects of Albendazole, Mebendazole, and Praziquantel Nanocapsules against Protoscolices of Hydatid Cyst" Pathogens 13, no. 9: 790. https://doi.org/10.3390/pathogens13090790

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop