Next Article in Journal
Predicting Associations of miRNAs and Candidate Gastric Cancer Genes for Nanomedicine
Previous Article in Journal
Nano-Photocatalytic Materials: Possibilities and Challenges
Previous Article in Special Issue
Sulfur(VI) Fluoride Exchange (SuFEx)-Mediated Synthesis of the Chitosan-PEG Conjugate and Its Supramolecular Hydrogels for Protein Delivery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chitosan-Based Nanomaterials as Valuable Sources of Anti-Leishmanial Agents: A Systematic Review

by
Hamdan I. AlMohammed
1,
Amal Khudair Khalaf
2,
Aishah E. Albalawi
3,
Abdullah D. Alanazi
4,5,
Parastoo Baharvand
6,
Ali Moghaddam
7 and
Hossein Mahmoudvand
8,*
1
Department of Microbiology and Parasitology, Almaarefa University, Riyadh 11597, Saudi Arabia
2
Department of Microbiology, College of Medicine, University of Thiqar, Thiqar 0096442, Iraq
3
Faculty of Science, University of Tabuk, Tabuk 47912, Saudi Arabia
4
Department of Biological Science, Faculty of Science and Humanities, Shaqra University, P.O. Box 1040, Ad-Dawadimi 11911, Saudi Arabia
5
Department of Medical Laboratory, Alghad International Colleges for Applied Medical Science, Tabuk 47913, Saudi Arabia
6
Department of Social Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad 6813833946, Iran
7
Student Research Committee, Lorestan University of Medical Sciences, Khorramabad 6813833946, Iran
8
Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad 6813833946, Iran
*
Author to whom correspondence should be addressed.
Nanomaterials 2021, 11(3), 689; https://doi.org/10.3390/nano11030689
Submission received: 29 January 2021 / Revised: 2 March 2021 / Accepted: 4 March 2021 / Published: 10 March 2021
(This article belongs to the Special Issue Biomedical Applications of Nano-Chitosan)

Abstract

:
Background: The current chemotherapy agents against various forms of leishmaniasis have some problems and side effects, including high toxicity, high cost, and the emergence of resistant strains. Here, we aimed to review the preclinical studies (in vitro and in vivo) on the anti-leishmanial activity of chitosan and chitosan-based particles against Leishmania spp. Methods: This study was conducted based on the 06-PRISMA guidelines and registered in the CAMARADES-NC3Rs Preclinical Systematic Review and Meta-Analysis Facility (SyRF) database. Various English databases such as PubMed, Google Scholar, Web of Science, EBSCO, ScienceDirect, and Scopus were used to find the publications related to the anti-leishmanial effects of chitosan and its derivatives and other pharmaceutical formulations, without a date limitation, to find all the published articles. The keywords included “chitosan”, “chitosan nanoparticles”, “anti-leishmanial”, “Leishmania”, “leishmaniasis”, “cutaneous leishmaniasis”, “visceral leishmaniasis”, “in vitro”, and “in vivo”. The language for data collection were limited to English. Results: Of 2669 papers, 25 papers, including 7 in vitro (28.0%), 7 in vivo (28.0%), and 11 in vitro/in vivo (44.0%) studies conducted up to 2020 met the inclusion criteria for discussion in this systematic review. The most common species of Leishmania used in these studies were L. major (12, 48.0%), L. donovani (7, 28.0%), and L. amazonensis (4, 16.80%). In vivo, the most used animals were BALB/c mice (11, 61.1%) followed by hamsters (6, 33.3%) and Wistar rats (1, 5.5%), respectively. In vitro, the most used Leishmania form was amastigote (8, 44.4%), followed by promastigote (4, 22.2%), and both forms promastigote/amastigote (6, 33.3%). Conclusion: According to the literature, different types of drugs based on chitosan and their derivatives demonstrated considerable in vitro and in vivo anti-leishmanial activity against various Leishmania spp. Based on the findings of this review study, chitosan and its derivatives could be considered as an alternative and complementary source of valuable components against leishmaniasis with a high safety index. Nevertheless, more investigations are required to elaborate on this result, mainly in clinical settings.

1. Introduction

Leishmaniasis is a tropical and subtropical poverty-related disease caused by an intracellular parasite belonging to the genus Leishmania [1]. Humans are generally infected via the bite of a sandfly, mostly Phlebotomus and Lutzomyia, around the world [2]. According to reliable reports, 0.7–1 million new cases of the disease are notified annually, and 12–15 million people are now infected with the disease in different parts of the world [3,4]. Depending on the geographical distribution, various species of Leishmania such as L. tropica, L. major, L. donovani, L. infantum, L. mexicana, L. braziliensis, and L. amazonensis can cause different clinical forms of the disease [5,6]. Considering the classification and clinical picture of leishmaniasis in humans, the diseases are divided into four forms of cutaneous (CL), mucocutaneous (NCL), diffuse cutaneous (DCL), and visceral or kala-azar leishmaniasis (VL) [7,8].
There are a number of systemic and local therapeutic strategies for the treatment of various forms of leishmaniasis, including drugs (e.g., pentavalent antimony derivatives such as meglumine antimoniate (Glucantime®) and sodium stibogluconate (Pentostam®), miltefosine, pentamidine, amphotericin B (Amp B), and paromomycin), as well as physical treatments (e.g., cryotherapy, surgery, thermotherapy, and laser therapy) [9]. Based on recent studies, the current conventional chemotherapeutics generally have difficulty reaching the target tissues at the applied doses and are also linked to adverse side effects on healthy tissues [10], indicating that the drug delivery systems must improve the efficacy, tolerability, specificity, and therapeutic index of anti-leishmanial drugs. Moreover, unresponsiveness to these anti-leishmanial compounds, even to their higher doses, is regularly reported in some parts of the world [10,11]. These limitations motivate researchers to discover an effective alternative agent with low toxicity in natural compounds as a major source of medications with various therapeutics characteristics.
Chitosan (poly-(b-1/4)-2-amino-2-deoxy-D-glucopyranose) is the general name used for a group of natural polysaccharide polymers produced by deacetylation of chitin (Figure 1) [12,13,14]. In recent years, the use of chitosan and its derivatives has attracted the attention of many researchers in medical and pharmaceutical sciences [15] due to its unique properties such as potent biological properties, low toxicity, biocompatibility, biodegradability, immunomodulatory [16], and anti-cancer, anti-nociceptive, anti-oxidant, anti-inflammatory, and anti-microbial properties [17,18].
Recent studies have demonstrated that the preparation of chitosan-based biomedical drugs such as nanoparticles, hydrogels, coatings, suspensions, powders, membranes, and films can impact the pharmaceutical and biomedical effects of these agents [19,20]. Recently, the antimicrobial activities of chitosan and its derivatives have been reported against a wide range of pathogenic viruses, bacteria, filamentous and yeast-like fungi [21,22], and helminthic and protozoan parasites [23,24]. Considering the anti-parasitic properties of chitosan and its derivatives, several investigations have demonstrated their potent anti-parasitic effects against some pathogenic strains such as Cryptosporidium spp. [25], Echinococcus granulosus [26], Leishmania spp. [23,24], and Toxoplasma gondii [27,28]. The present study aimed to review the preclinical studies (in vitro and in vivo) on the anti-leishmanial activity of chitosan and chitosan-based particles against Leishmania spp.

2. Methods

2.1. Database Search

This investigation was performed based on the 06-PRISMA guideline and registered in the CAMARADES-NC3Rs Preclinical Systematic Review and Meta-Analysis Facility (SyRF) database [29]. English databases, including PubMed, Google Scholar, Web of Science, EBSCO, ScienceDirect, and Scopus, were searched for publications related to anti-leishmanial effects of chitosan and its derivatives and other pharmaceutical formulations without a date limitation to identify all published articles. The keywords included “chitosan”, “chitosan nanoparticles”, “anti-leishmanial”, “Leishmania”, “leishmaniasis”, “cutaneous leishmaniasis”, “visceral leishmaniasis”, “in vitro”, and “in vivo”. Moreover, the language for data collection was limited to English.

2.2. Quality Assessment and Article Selection

The studies evaluating the anti-leishmanial effects of chitosan and its derivatives were examined. First, the studies were imported into EndNote X9 (Thomson Reuters, New York, NY, USA) and the duplicates were removed. Afterwards, three authors independently examined the title and abstract of the studies, and the relevant documents about the in vitro and/or in vivo anti-leishmanial effects of chitosan, chitosan derivatives, and chitosan-based nanoparticles were included for further analysis. The same authors carefully read the studies and selected the eligible investigations that adequately met the inclusion criteria. The inclusion criteria were articles evaluating the anti-leishmanial effects of chitosan and its derivatives and emphasizing the design of various forms of nanoparticles containing chitosan and other pharmaceutical formulations against leishmaniasis.
A total of 2669 articles were identified through database searching. Among these articles, 227 articles were removed due to duplication. Of the remaining 2442 articles, 2385 studies were removed due to the inadequate information and the ones in which the abstract was submitted in congresses as preceding papers, conferences, and editorials without full text. One of the main limitations in such studies is the frame of differing access to full texts between different research locations of study, which prevents us from accessing the full-text of some articles. To solve this problem, we searched in various research websites such as ResearchGate and LinkedIn which suggest a choice of direct full-text request from authors as well as exploring archives of wanted journals, or contacting principal investigator to purchase it if available.
From the remaining 57 articles which were assessed for eligibility, 32 articles were excluded due to some reasons such as inconsistency between methods with results, incorrect interpretation of the results, poor methodology, etc.; whereas 32 articles were finally included in this review (Figure 2).

2.3. Data Extraction

Three authors independently extracted information from the selected articles and, if necessary, the differences were resolved upon discussions with the corresponding author. The extracted data included chitosan type, whether it was used in combination or loaded with other drugs, the type of study, animals, doses, time, important results, and references.

3. Results and Discussion

Chitosan as a natural agent with diverse biological activities is generally found in the shells of crustaceans, such as crab, shrimp, squid pen, and crawfish; however, recent investigations have reported that chitosan can be produced from some fungi [15,16,17]. Of 2669 papers, 25 papers, including 7 in vitro (28.0%), 7 in vivo (28.0%), and 11 in vitro/in vivo (44.0%) studies conducted up to 2020 met the inclusion criteria for discussion in this systematic review. Totally, the most common species of Leishmania used in these studies were L. major (12%, 48.0%), L. donovani (7%, 28.0%), and L. amazonensis (4%, 16.80%), respectively.

3.1. Chitosan Treatments In Vitro

In vitro, the most used Leishmania form was amastigotes (8%, 44.4%), followed by promastigotes (4%, 22.2%), and both forms promastigotes/amastigotes (6%, 33.3%). In terms of the concentrations of chitosan and its formulations, the results demonstrated that, in vitro, they were used in the range of 0.03 µg/mL to 20 mg/mL. The findings demonstrated that the most used synthetic drugs for combination therapy in vitro were amphotericin B (14%, 70.0%), followed by miltefosine (3%, 15.0%) and doxorubicin hydrochloride (2%, 10.0%). The most common species of Leishmania used in in vitro studies were L. major (8%, 44.4%), L. donovani (6%, 33.3%), and L. amazonensis (3%, 16.6%), respectively (Table 1).
The results exhibited that the most used in vitro screening strategy to check the anti-leishmanial effects of chitosan and its derivatives were the intracellular living amastigote assay, followed by the extracellular promastigote assay. Now, various in vitro systems are used in the evaluation of agents, which show direct toxic action on the parasite [48]; however, for some agents that act through their metabolites and/or host defense system, it does not seem like a good option [49]. Some in vitro screening studies target the promastigote forms of Leishmania because of the simplicity of the cultivation and handling of parasites, the short time required and the low cost [50]; nevertheless, because of some weaknesses, for example, the lack of host cells in this method and the presence of this parasitic form in the invertebrate vector, it is not considered a reliable target to test anti-leishmanial compounds [51]. The use of intracellular amastigote forms in the in vitro system is also well-known as a more clinically relevant model for the assessment of anti-leishmanial compounds due to having some unique features such as acquiring important cell-health information, low cost, and no need for secondary assays [52,53].
Previously, Mohebali et al. [31] have reported that the chitosan at the doses of 100, 200 and 400 μg/mL completely killed the L. major promastigotes in vitro. In another in vitro study conducted by Feizabadi et al. [37], the results showed that chitosan coupled with L. major secretory and excretory proteins can increase the ability of infected macrophages to remove parasites by reducing apoptosis. Riberio et al. (2014) have reported that chitosan nanoparticles combined with AmpB significantly reduced the lesion size and parasite burden in all the evaluated organs of mice infected with L. amazonensis. They concluded that this compound controls leishmaniasis by increasing the cytokines of IFN-γ, IL-12, IL-4, and IL-10 in the infected mice [54].

3.2. Chitosan Treatments In Vivo

The obtained results showed that the most used animals in vivo were BALB/C mice (11%, 61.1%) followed by hamsters (6%, 33.3%) and Wistar rats (1%, 5.5%). In terms of the concentrations of chitosan and its formulations, the results demonstrated that, in vivo, they were used in the range of 0.005 mg/kg to 500 mg/kg. The most common species of Leishmania used in in vitro studies were L. major (9%, 44.4%), followed by L. donovani (7%, 33.3%) (Table 2).
In vivo assay or using the standardized animal models in laboratories are considered as the best strategies to evaluate the anti-leishmanial drug compounds against various forms of leishmaniasis which has the closest resemblance to the human condition [61]. Although the predictive power of the in vivo assay is not very noticeable, however, the important points in this model are that (i) if a drug is not effective in vivo, there is no need to study further and that (ii) some useful information is obtained about the effective doses as well as the toxicity of the studied drugs [48]. Today, several in vivo experimental models are used to assess and test novel agents against various forms of leishmaniasis, e.g., (i) the BALB/C mice-L. major model considered as a validated model for human CL; (ii) C57BL/6, BALB/C, and CBA/J mice to induce L. amazonensis, L. mexicana infection; (iii) BALB/C mice and Syrian golden hamster as experimental models for L. infantum infection [48,49].
Mohebali et al. [30] have demonstrated that the chitosan at the doses of 100, 200 and 400 μg/mL reduced the size of the lesion from 10.7 ± 3.24 mm in the control group to 1.05 ± 1.02 mm on day 28 at a dose of 400 μg/mL, and also from 6.27 ± 1.23 mm to 2.07 ± 0.87 mm in the 200 μg/mL concentration in the mice infected with L. major. Mohammadi-Samani et al. [56] showed that superoxide dismutase B1 combined with chitosan nanoparticles is able to promote the immunogenicity to cell-mediated immunity (T(H)1 cells producing IgG2a in mice) that is effective in removing of Leishmania parasites and might be considered as a single-dose nanovaccine for leishmaniasis.

3.3. Treatments Using Chitosan as Vehicle

Nowadays, it has been demonstrated that chitosan, its derivatives, and chitosan-based nanomaterials are able to possibly remove barriers in the carrying of drugs, thus improving the efficacy of the drug and subsequently the targeted drug therapy [62]. The findings demonstrated that the most used synthetic drugs for combination therapy in vivo and in vitro were amphotericin B (14%, 70.0%), followed by miltefosine (3%, 15.0%) and doxorubicin hydrochloride (2%, 10.0%). Although most of the studies in this review use chitosan in combination with other drugs, however, chitosan and its derivatives without combination with common drugs have been considered in some studies.
Malli et al. (2019) have also demonstrated that the nanoparticles of poly (isobutyl-cyanoacrylate) coated with chitosan have potent anti-leishmanial effects on L. major promastigotes through morphological changes such as the aberrant shape and swelling of mitochondria and parasitic vacuoles [45]. In a study conducted by Feizabadi et al. (2019), it has been proven that chitosan combined with L. major secretory and excretory proteins can improve the ability of infected macrophages to remove parasites by decreasing apoptosis [37].
For example, Lima et al. [32] have reported that chitosan-silver nanoparticles have more anti-leishmanial activity than chitosan on L. amazonensis promastigotes with the IC50 values of 1.69 and 7.81 µg/mL, respectively. In the study conducted by Seyyed Tabaei et al. [44] have showed that chitosan-polyethylene oxide-berberine nanofibers has potent therapeutic effects on healing of CL induced by L. major in BALAB/C mice through reducing the parasite burden, decreasing the lesion size as well as change in the epidermis and dermis.
In recent years, the anti-parasitic activities of chitosan and its various derivatives/formulations have been studied against several parasitic pathogens such as C. pavum [25], Echinococcus spp. [26], and T. gondii [27,28]. For example, Mammeri et al. (2018) demonstrated that chitosan significantly decreased the viability of Cryptosporidium parvum oocysts by >95% after 24 h of treatment with chitosan mix (C-Mix) and chitosan N-acetyl-D-glucosamine (CNAD). They also reported that C-Mix (34.5%) and CNAD (56%) significantly decreased the oocysts’ shedding by 34.5% and 56% in newborn mice infected with cryptosporidiosis, respectively [25]. Torabi et al. (2018) have demonstrated that chitosan-praziquantel and chitosan-albendazole nanoparticles especially in combination at the doses of 1, 5, and 10 μg/mL significantly reduced the viability of microcysts, weight and number of cysts in vitro and in vivo [26]. In the study conducted by Teimouri et al. (2018), it has been proven that low molecular weight chitosan nanoparticles completely killed the tachyzoites at the concentration of 500 and 1000 ppm in vitro; they also showed that this compound considerably increased the survival time of infected mice with T. gondii RH strain from 6 to 8 days after infection [27].

3.4. Possible Antimicrobial Mechanisms of Chitosan

The precise antimicrobial mechanism of action of chitosan is yet to be fully understood; still, based on the literature, the most likely antimicrobial mechanisms of action of chitosan include the disruption of the cell wall and, consequently, an effect on the membrane’s permeability, inhibition of DNA replication, cell death, and bindings to the trace metal elements resulting in toxin production and microbial growth inhibition [63].
Mohammadi-Samani et al. (2011) have reported that chitosan nanoparticles containing Leishmania superoxide dismutase could be considered a nano-vaccine for leishmaniasis eradication by promoting the immune response toward cell-mediated immunity (TH1 cells producing IgG2a in mice) [56].

3.5. Cytotoxicity Effects of Chitosan

With respect to the cytotoxic effects of chitosan and its various formulations, Karam et al. (2020) found that chitosan nanocapsules containing the essential oil of Matricaria chamomilla have no significant cytotoxicity against macrophage cells with a CC50 (the 50% cytotoxic concentration) value of 207.92 ± 18.53 μg/mL compared to 19.71 ± 1.73 μg/mL for essential oil alone [35]. Another study conducted by Chaubey et al. (2018) indicated that the mannose-conjugated chitosan nanoparticles of curcumin had no significant cytotoxicity against the J774A.1 macrophage cell line with a CC50 value of 26 ± 0.60 mg/mL [41]. Recently, Esfandiari et al. (2019) have reported that paromomycin-loaded mannosylated chitosan nanoparticles had no considerable cytotoxicity against the human monocyte cell line of THP-1 cells with a CC50 value of 3911 μg/mL [43].

4. Conclusions

Studies in recent years revealed that chitosan, its derivatives, and chitosan-based nanomaterials are able possibly remove barriers in the carrying of drugs thus improving the efficacy of the drug and subsequently the targeted drug therapy. In the present review, based on the literature, various forms of drugs based on chitosan and their derivatives exhibited significant antileishmanial activity against various Leishmania spp, in vitro and in vivo. The results showed that chitosan and chitosan-based particles could be considered as an alternative and complementary source of valuable components against leishmaniasis with a high safety index. However, more studies are required to elucidate this finding, particularly in clinical settings.

Author Contributions

Conceptualization, H.I.A. and H.M.; methodology, A.K.K., A.E.A., and A.D.A.; software, P.B. and A.M.; validation, A.K.K., A.E.A., A.D.A., H.I.A., H.M., A.M. and P.B.; formal analysis, A.M. and P.B.; investigation, A.M. and H.M.; data curation, P.B.; writing—original draft preparation, H.M. and P.B.; writing—review and editing A.K.K., A.E.A., A.D.A., H.I.A., and A.M.; supervision, H.M. All authors have read and agreed to the published version of the manuscript.

Funding

Not applicable.to those who have contributed substantially to the work reported.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed during this study are included in this published article.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Torres-Guerrero, E.; Quintanilla-Cedillo, M.R.; Ruiz-Esmenjaud, J.; Arenas, R. Leishmaniasis: A review. F1000Research 2017, 6, 750. [Google Scholar] [CrossRef] [PubMed]
  2. Alvar, J.; Vélez, I.D.; Bern, C.; Herrero, M.; Desjeux, P.; Cano, J.; Jannin, J.; den Boer, M.; WHO Leishmaniasis Control Team. Leishmaniasis Worldwide and Global Estimates of Its Incidence. PLoS ONE 2012, 7, e35671. [Google Scholar] [CrossRef] [PubMed]
  3. World Health Organization. Urbanization: An increasing risk factor for leishmaniasis. Wkly. Epidemiol. Rec. Relev. Épidémiologique Hebd. 2002, 77, 365–370. [Google Scholar]
  4. Pearson, R.D.; Sousa, A.Q. Clinical spectrum of leishmaniasis. Clin Infect Dis. 1996, 22, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hepburn, N.C. Cutaneous leishmaniasis: Clinical dermatology Review article. Clin. Exp. Dermatol. Clin. Dermatol. 2000, 25, 363–370. [Google Scholar] [CrossRef] [PubMed]
  6. Burza, S.; Croft, S.L.; Boelaert, M. Leishmaniasis. Lancet 2018, 392, 951–970. [Google Scholar] [CrossRef]
  7. McGwire, B.S.; Satoskar, A.R. Leishmaniasis: Clinical syndromes and treatment. Qjm: Int. J. Med. 2014, 107, 7–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Scorza, B.M.; Carvalho, E.M.; Wilson, M.E. Cutaneous Manifestations of Human and Murine Leishmaniasis. Int. J. Mol. Sci. 2017, 18, 1296. [Google Scholar] [CrossRef] [Green Version]
  9. Bi, K.; Chen, Y.; Zhao, S.; Kuang, Y.; Wu, C.-H.J. Current Visceral Leishmaniasis Research: A Research Review to Inspire Future Study. BioMed Res. Int. 2018, 2018, 1–13. [Google Scholar] [CrossRef] [Green Version]
  10. WHO Expert Committee on the Control of the Leishmaniases. Meeting, World Health Organization. In Proceedings of the Control of the Leishmaniases: Report of a Meeting of the WHO Expert Committee on the Control of Leishmaniases, Geneva, Switzerland, 22–26 March 2010; World Health Organization: Copenhagen, Denmark, 2010. [Google Scholar]
  11. Monzote, L. Current treatment of leishmaniasis: A review. Open Antimicrob. Agents J. 2009, 1, 9–19. [Google Scholar] [CrossRef]
  12. Santos, D.O.; Coutinho, C.E.R.; Madeira, M.F.; Bottino, C.G.; Vieira, R.T.; Nascimento, S.B.; Bernardino, A.; Bourguignon, S.C.; Corte-Real, S.; Pinho, R.T.; et al. Leishmaniasis treatment—a challenge that remains: A review. Parasitol. Res. 2008, 103, 1–10. [Google Scholar] [CrossRef] [PubMed]
  13. Oliveira, L.F.; Schubach, A.O.; Martins, M.M.; Passos, S.L.; Oliveira, R.V.; Marzochi, M.C.; Andrade, C.A. Systematic review of the adverse effects of cutaneous leishmaniasis treatment in the New World. Acta Trop. 2011, 118, 87–96. [Google Scholar] [CrossRef]
  14. Naveed, M.; Phil, L.; Sohail, M.; Hasnat, M.; Baig, M.M.F.A.; Ihsan, A.U.; Shumzaid, M.; Kakar, M.U.; Khan, T.M.; Akabar, M.D.; et al. Chitosan oligosaccharide (COS): An overview. Int. J. Biol. Macromol. 2019, 129, 827–843. [Google Scholar] [CrossRef] [PubMed]
  15. Younes, I.; Rinaudo, M. Chitin and Chitosan Preparation from Marine Sources. Structure, Properties and Applications. Mar. Drugs 2015, 13, 1133–1174. [Google Scholar] [CrossRef] [Green Version]
  16. Muxika, A.; Etxabide, A.; Uranga, J.; Guerrero, P.; de la Caba, K. Chitosan as a bioactive polymer: Processing, properties and applications. Int. J. Biol. Macromol. 2017, 105, 1358–1368. [Google Scholar] [CrossRef]
  17. Wang, W.; Meng, Q.; Li, Q.; Liu, J.; Zhou, M.; Jin, Z.; Zhao, K. Chitosan Derivatives and Their Application in Biomedicine. Int. J. Mol. Sci. 2020, 21, 487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Ahmed, T.A.; Aljaeid, B.M. Preparation, characterization, and potential application of chitosan, chitosan derivatives, and chitosan metal nanoparticles in pharmaceutical drug delivery. Drug Des. Dev. Ther. 2016, 10, 483–507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Kravanja, G.; Primožič, M.; Knez, Ž.; Leitgeb, M. Chitosan-Based (Nano)Materials for Novel Biomedical Applications. Molecules 2019, 24, 1960. [Google Scholar] [CrossRef] [Green Version]
  20. Rizeq, B.R.; Younes, N.N.; Rasool, K.; Nasrallah, G.K. Synthesis, Bioapplications, and Toxicity Evaluation of Chitosan-Based Nanoparticles. Int. J. Mol. Sci. 2019, 20, 5776. [Google Scholar] [CrossRef] [Green Version]
  21. Guan, G.; Azad, A.K.; Lin, Y.; Kim, S.W.; Tian, Y.; Liu, G.; Wang, H. Biological Effects and Applications of Chitosan and Chito-Oligosaccharides. Front. Physiol. 2019, 10, 516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Rozman, N.A.S.; Tong, W.Y.; Leong, C.R.; Tan, W.N.; Hasanolbasori, M.A.; Abdullah, S.Z. Potential Antimicrobial Applications of Chitosan Nanoparticles (ChNP). J. Microbiol. Biotechnol. 2019, 29, 1009–1013. [Google Scholar] [CrossRef] [PubMed]
  23. Rabea, E.I.; Badawy, M.E.; Stevens, C.V.; Smagghe, G.; Steurbaut, W. Chitosan as antimicrobial agent: Applications and mode of action. Biomacromolecules 2003, 4, 1457–1465. [Google Scholar] [CrossRef]
  24. Varshosaz, J.; Arbabi, B.; Pestehchian, N.; Saberi, S.; Delavari, M. Chitosan-titanium dioxide-glucantime nanoassemblies effects on promastigote and amastigote of Leishmania major. Int. J. Biol. Macromol. 2018, 107, 212–221. [Google Scholar] [CrossRef]
  25. Mammeri, M.; Chevillot, A.; Thomas, M.; Polack, B.; Julien, C.; Marden, J.-P.; Auclair, E.; Vallée, I.; Adjou, K.T. Efficacy of chitosan, a natural polysaccharide, against Cryptosporidium parvum in vitro and in vivo in neonatal mice. Exp. Parasitol. 2018, 194, 1–8. [Google Scholar] [CrossRef]
  26. 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]
  27. Teimouri, A.; Azami, S.J.; Keshavarz, H.; Esmaeili, F.; Alimi, R.; Mavi, S.A.; Shojaee, S. Anti-Toxoplasma activity of various molecular weights and concentrations of chitosan nanoparticles on tachyzoites of RH strain. Int. J. Nanomed. 2018, 13, 1341–1351. [Google Scholar] [CrossRef] [Green Version]
  28. Cheraghipour, K.; Masoori, L.; Ezzatkhah, F.; Salimikia, I.; Amiri, S.; Makenali, A.S.; Taherpour, F.; Mahmoudvand, H. Effect of chitosan on Toxoplasma gondii infection: A systematic review. Parasite Epidemiol. Control. 2020, 11, e00189. [Google Scholar] [CrossRef] [PubMed]
  29. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; Prisma Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Ann. Intern. Med. 2009, 151, 264–269. [Google Scholar] [CrossRef] [Green Version]
  30. Singh, P.K.; Pawar, V.K.; Jaiswal, A.K.; Singh, Y.; Srikanth, C.H.; Chaurasia, M.; Bora, H.K.; Raval, K.; Meher, J.G.; Gayen, J.R.; et al. Chitosan coated PluronicF127 micelles for effective delivery of Amphotericin B in experimental visceral leishmaniasis. Int. J. Biol. Macromol. 2017, 105, 1220–1231. [Google Scholar] [CrossRef] [PubMed]
  31. Mohebali, M.; Esboei, B.R.; Mousavi, P.; Fakhar, M.; Akhoundi, B. Potent antileishmanial activity of chitosan against Iranian strain of Leishmania major (MRHO/IR/75/ER): In vitro and in vivo assay. J. Vector Borne Dis. 2018, 55, 111. [Google Scholar] [CrossRef]
  32. Lima, D.D.S.; Gullon, B.; Cardelle-Cobas, A.; Brito, L.M.; Rodrigues, K.A.F.; Quelemes, P.V.; Ramos-Jesus, J.; Arcanjo, D.D.R.; Plácido, A.; Batziou, K.; et al. Chitosan-based silver nanoparticles: A study of the antibacterial, antileishmanial and cytotoxic effects. J. Bioact. Compat. Polym. 2017, 32, 397–410. [Google Scholar] [CrossRef] [Green Version]
  33. Kunjachan, S.; Gupta, S.; Dwivedi, A.K.; Dube, A.; Chourasia, M.K. Chitosan-based macrophage-mediated drug targeting for the treatment of experimental visceral leishmaniasis. J. Microencapsul. 2011, 28, 301–310. [Google Scholar] [CrossRef]
  34. Tripathi, P.; Jaiswal, A.K.; Dube, A.; Mishra, P.R. Hexadecylphosphocholine (Miltefosine) stabilized chitosan modified Ampholipospheres as prototype co-delivery vehicle for enhanced killing of L. donovani. Int. J. Biol. Macromol. 2017, 105, 625–637. [Google Scholar] [CrossRef]
  35. Karam, T.K.; Ortega, S.; Nakamura, T.U.; Auzély-Velty, R.; Nakamura, C.V. Development of chitosan nanocapsules containing essential oil of Matricaria chamomilla L. for the treatment of cutaneous leishmaniasis. Int. J. Biol. Macromol. 2020, 162, 199–208. [Google Scholar] [CrossRef] [PubMed]
  36. Tripathi, P.; Dwivedi, P.; Khatik, R.; Jaiswal, A.K.; Dube, A.; Shukla, P.; Mishra, P.R. Development of 4-sulfated N -acetyl galactosamine anchored chitosan nanoparticles: A dual strategy for effective management of Leishmaniasis. Colloids Surf. B Biointerfaces 2015, 136, 150–159. [Google Scholar] [CrossRef] [PubMed]
  37. Feizabadi, E.; Zavaran Hosseini, A.; Soudi Sara, K. Studying the role of chitosan nanoparticle loaded with Leishmania major Secretory and excretory antigens on the number of apoptotic macrophages in parasite sensitive mouse. Daneshvar Med. Basic Clin. Res. J. 2020, 26, 9–18. [Google Scholar]
  38. Khan, M.; Shereen, M.A.; Khokhar, M.; Kamil, A.; Rahman, H. A novel effective therapeutic approach for treatment of Leishmania tropica through Miltefosine Loaded Chitosan Nanoparticles. Res. Sq. 2020. [Google Scholar] [CrossRef] [Green Version]
  39. Riezk, A.; Van Bocxlaer, K.; Yardley, V.; Murdan, S.; Croft, S.L. Activity of Amphotericin B-Loaded Chitosan Nanoparticles against Experimental Cutaneous Leishmaniasis. Molecules 2020, 25, 4002. [Google Scholar] [CrossRef]
  40. Rahimi, M.; Tabaei, S.J.S.; Ziai, S.A.; Sadri, M. Anti-Leishmanial Effects of Chitosan-Polyethylene Oxide Nanofibers Containing Berberine: An Applied Model for Leishmania Wound Dressing. Iran. J. Med. Sci. 2020, 45, 286–297. [Google Scholar]
  41. Chaubey, P.; Mishra, B.; Mudavath, S.L.; Patel, R.R.; Chaurasia, S.; Sundar, S.; Suvarna, V.; Monteiro, M. Mannose-conjugated curcumin-chitosan nanoparticles: Efficacy and toxicity assessments against Leishmania donovani. Int. J. Biol. Macromol. 2018, 111, 109–120. [Google Scholar] [CrossRef]
  42. Cabral, F.V.; Pelegrino, M.T.; Sauter, I.P.; Seabra, A.B.; Cortez, M.; Ribeiro, M.S. Nitric oxide-loaded chitosan nanoparticles as an innovative antileishmanial platform. Nitric Oxide-Biol. Chem. 2019, 93, 25–33. [Google Scholar] [CrossRef] [PubMed]
  43. Esfandiari, F.; Motazedian, M.H.; Asgari, Q.; Morowvat, M.H.; Molaei, M.; Heli, H. Paromomycin-loaded mannosylated chitosan nanoparticles: Synthesis, characterization and targeted drug delivery against leishmaniasis. Acta Trop. 2019, 197, 105072. [Google Scholar] [CrossRef]
  44. Seyyed Tabaei, S.J.; Rahimi, M.; Akbaribazm, M.; Ziai, S.A.; Sadri, M.; Shahrokhi, S.R.; Rezaei, M.S. Chitosan-based nano-scaffolds as antileishmanial wound dressing in BALB/c mice treatment: Characterization and design of tissue regeneration. Iran J. Basic Med. Sci. 2020, 23, 788–799. [Google Scholar]
  45. Malli, S.; Pomel, S.; Dennemont, I.; Loiseau, P.M.; Bouchemal, K. Combination of amphotericin B and chitosan platelets for the treatment of experimental cutaneous leishmaniasis: Histological and immunohistochemical examinations. J. Drug Deliv. Sci. Technol. 2019, 50, 34–41. [Google Scholar] [CrossRef]
  46. Malli, S.; Pomel, S.; Ayadi, Y.; Deloménie, C.; Da Costa, A.; Loiseau, P.M.; Bouchemal, K. Topically Applied Chitosan-Coated Poly(isobutylcyanoacrylate) Nanoparticles Are Active Against Cutaneous Leishmaniasis by Accelerating Lesion Healing and Reducing the Parasitic Load. ACS Appl. Bio Mater. 2019, 2, 2573–2586. [Google Scholar] [CrossRef]
  47. Gupta, P.K.; Jaiswal, A.K.; Asthana, S.; Verma, A.; Kumar, V.; Shukla, P.; Dwivedi, P.; Dube, A.; Mishra, P.R. Self Assembled Ionically Sodium Alginate Cross-Linked Amphotericin B Encapsulated Glycol Chitosan Stearate Nanoparticles: Applicability in Better Chemotherapy and Non-Toxic Delivery in Visceral Leishmaniasis. Pharm. Res. 2015, 32, 1727–1740. [Google Scholar] [CrossRef]
  48. Siqueira-Neto, J.L.; Song, O.-R.; Oh, H.; Sohn, J.-H.; Yang, G.; Nam, J.; Jang, J.; Cechetto, J.; Lee, C.B.; Moon, S.; et al. Antileishmanial High-Throughput Drug Screening Reveals Drug Candidates with New Scaffolds. PLoS Negl. Trop. Dis. 2010, 4, e675. [Google Scholar] [CrossRef] [Green Version]
  49. Sharlow, E.R.; Close, D.; Shun, T.; Leimgruber, S.; Reed, R.; Mustata, G.; Wipf, P.; Johnson, J.; O’Neil, M.; Grogl, M.; et al. Identification of Potent Chemotypes Targeting Leishmania major Using a High-Throughput, Low-Stringency, Computationally Enhanced, Small Molecule Screen. PLoS Negl. Trop. Dis. 2009, 3, e540. [Google Scholar] [CrossRef] [Green Version]
  50. St George, S.; Bishop, J.V.; Titus, R.G.; Selitrennikoff, C.P. Novel compounds active against Leishmania major. Antimicrob. Agents Chemother. 2006, 50, 474–479. [Google Scholar] [CrossRef] [Green Version]
  51. Fumarola, L.; Spinelli, R.; Brandonisio, O. In vitro assays for evaluation of drug activity against Leishmania spp. Res. Microbiol. 2004, 155, 224–230. [Google Scholar] [CrossRef]
  52. De Muylder, G.; Ang, K.K.; Chen, S.; Arkin, M.R.; Engel, J.C.; McKerrow, J.H. A screen against Leishmania intracellular amastigotes: Comparison to a promastigote screen and identification of a host cell-specific hit. PLoS Negl. Trop. Dis. 2011, 5, e1253. [Google Scholar] [CrossRef] [Green Version]
  53. Vermeersch, M.; Da Luz, R.I.; Toté, K.; Timmermans, J.-P.; Cos, P.; Maes, L. In Vitro Susceptibilities of Leishmania donovani Promastigote and Amastigote Stages to Antileishmanial Reference Drugs: Practical Relevance of Stage-Specific Differences. Antimicrob. Agents Chemother. 2009, 53, 3855–3859. [Google Scholar] [CrossRef] [Green Version]
  54. Ribeiro, T.G.; Franca, J.R.; Fuscaldi, L.L.; Santos, M.L.; Duarte, M.C.; Lage, P.S.; Martins, V.T.; Costa, L.E.; Fernandes, S.O.; Cardoso, V.N.; et al. An optimized nanoparticle delivery system based on chitosan and chondroitin sulfate molecules reduces the toxicity of amphotericin B and is effective in treating tegumentary leishmaniasis. Int. J. Nanomed. 2014, 9, 5341–5353. [Google Scholar]
  55. Asthana, S.; Jaiswal, A.K.; Gupta, P.K.; Pawar, V.K.; Dube, A.; Chourasia, M.K. Immunoadjuvant Chemotherapy of Visceral Leishmaniasis in Hamsters Using Amphotericin B-Encapsulated Nanoemulsion Template-Based Chitosan Nanocapsules. Antimicrob. Agents Chemother. 2013, 57, 1714–1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Mohammadi-Samani, S.; Bahraini, D.; Shokri, J.; Kamali-Sarvestani, E.; Baezegar-Jalali, M.; Samiei, A.; Danesh-Bahreini, M.A.; Barzegar-Jalali, M. Nanovaccine for leishmaniasis: Preparation of chitosan nanoparticles containing Leishmania superoxide dismutase and evaluation of its immunogenicity in BALB/c mice. Int. J. Nanomed. 2011, 6, 835–842. [Google Scholar] [CrossRef] [Green Version]
  57. Mehrizi, T.Z.; Ardestani, M.S.; Hoseini, M.H.M.; Khamesipour, A.; Mosaffa, N.; Ramezani, A. Novel nano-sized chitosan amphotericin B formulation with considerable improvement against Leishmania major. Nanomedicine 2018, 13, 3129–3147. [Google Scholar] [CrossRef]
  58. Moreno, E.; Schwartz, J.; Larrea, E.; Conde, I.; Font, M.; Sanmartín, C.; Irache, J.M.; Espuelas, S. Assessment of β-lapachone loaded in lecithin-chitosan nanoparticles for the topical treatment of cutaneous leishmaniasis in L. major infected BALB/c mice. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 2003–2012. [Google Scholar] [CrossRef]
  59. Mehrizi, T.Z.; Ardestani, M.S.; Hoseini, M.H.M.; Khamesipour, A.; Mosaffa, N.; Ramezani, A. Novel Nanosized Chitosan-Betulinic Acid Against Resistant Leishmania Major and First Clinical Observation of such parasite in Kidney. Sci. Rep. 2018, 8, 11759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Serrano, D.R.; Lalatsa, A.; Dea-Ayuela, M.A.; Bilbao-Ramos, P.E.; Garrett, N.L.; Moger, J.; Guarro, J.; Capilla, J.; Ballesteros, M.P.; Schätzlein, A.G.; et al. Oral Particle Uptake and Organ Targeting Drives the Activity of Amphotericin B Nanoparticles. Mol. Pharm. 2015, 12, 420–431. [Google Scholar] [CrossRef]
  61. Nagy, A.; Harrison, A.; Sabbani, S.; Munson Jr, R.S.; Dutta, P.K.; Waldman, W.J. Silver nanoparticles embedded in zeolite membranes: Release of silver ions and mechanism of antibacterial action. Int. J. Nanomed. 2011, 6, 1833. [Google Scholar]
  62. Suman Gupta, N. Visceral leishmaniasis: Experimental models for drug discovery. Indian J. Med. Res. 2011, 133, 27–39. [Google Scholar] [PubMed]
  63. Li, J.; Cai, C.; Li, J.; Li, J.; Li, J.; Sun, T.; Wang, L.; Wu, H.; Yu, G. Chitosan-based nanomaterials for drug delivery. Molecules 2018, 23, 2661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The chemical structure of chitin and chitosan.
Figure 1. The chemical structure of chitin and chitosan.
Nanomaterials 11 00689 g001
Figure 2. Flowchart describing the study design process.
Figure 2. Flowchart describing the study design process.
Nanomaterials 11 00689 g002
Table 1. A list of in vitro studies of chitosan and its derivatives as resource for anti-leishmanial agents.
Table 1. A list of in vitro studies of chitosan and its derivatives as resource for anti-leishmanial agents.
Type of ChitosanCombined with the DrugYear of PublicationParasite FormLeishmania spp.ConcentrationTimeOutcomeRef.
AmB-loaded pluronic F127 (PF 127) micelles coated with chitosan (Cs-PF-AmB-M)Amphotericin B (AmpB)2017PromastigotesLeishmania donovani0.03, 0.05,
0.1, 0.2, 0.4, 0.8 µg/mL
72 hExperiments have shown that Cs-PF-AmB-M at a dose of 0.049 µg/mL may reduce parasitic load by 50%; whereas PF-AmB-M at a dose of 0.08 µg/mL reduced parasitic load by 50%.[30]
Chitosan -2018Promastigotes L. major50, 100, 200, 400 μg/mL30, 60, 120 and 180 minThe results showed that chitosan at the concentrations of 200 and 400 μg/mL after 180 min killed 100% of promastigote. [31]
Chitosan-based silver nanoparticles-2017Promastigotes and amastigotesL. amazonensis0.42 to 27µg48 hThe results showed that this compound has potent anti-leishmanial effects against promastigote and amastigote stages of L. amazonensis after 48 h exposure, with IC50 values ranging from 0.422 to 2120 μg/mL.[32]
Chitosan microparticlesDoxorubicin hydrochloride (DOX)2011Promastigotes L. donovani0.03, 0.08, 0.13 and 0.2 mg/mL20 hThe results showed that the greatest effect of these microparticles was in the first 60 min and caused nonspecific activation of phagocytosis in macrophages.[33]
Chitosan anchored nanostructured lipid carriers (NLC)Miltefosine (HePC-
hexadecyl
phosphocholine)
and amphotericin B (AmB)
2017Amastigotes L. donovani50, 100, 250, 500, 1000 ng/mL4 h The results showed that the highest effect of these nanoparticles was at a concentration of 1000 ng/mL, which killed more than 80% of amastigotes, while AmB alone reduced the parasitic load by about 60%.[34]
Chitosan nanocapsules containing essential oil of Matricaria chamomilla
(NCEO)
-2020Promastigotes L. amazonensis0.1–1000 μg/mL48 hThe results showed that IC50 NCEO was 7.18 ± 0.7 μg/mL against promastigotes and 14.29 ± 1.01 μg/mL against amastigotes.[35]
Chitosan nanoparticles
(CNPs) and, 4-SO4GalNAc modified chitosan nanoparticles
(SCNP)
Amphotericin B (AmpB)2015Amastigotes L. donovani0.05, 0.1, 0.2. 0.4. 0.8 (µg/mL)24 hThe results showed that AmB-SCNPs and AmB–CNPs had a better effect in comparison with amphotericin B and more than 80% of their lethality was recorded, while for amphotericin B 70% lethality have been recorded.[36]
Chitosan nanoparticles-2019Amastigotes L. major5–250 µL/mL48 hChitosan coupled with L. major secretory and excretory proteins can increase the ability of infected macrophages to remove parasites by reducing apoptosis.[37]
Chitosan nanoparticlesMiltefosine2020Promastigotes and amastigotes L. tropica100 µL/mL72 hThe results showed that IC50 value for promastigote and amastigote forms of L. tropica was 0.07 ± 0.05 µL/mL and 0.09 ± 0.02 µL/mL, respectively,[38]
Chitosan nanoparticles and
sodium tripolyphosphate (TPP)
Amphotericin B (AmB)2020Amastigotes L. major and L. mexicana1 mg/mL7 daysThe results showed that EC50 value of AmB-CH-TPP for L. major and L. mexicana amastigotes was 0.14 ± 0.09 µg/mL and 0.5 ± 0.01 µg/mL, respectively.[39]
Chitosan-polyethylene
oxide nanofibers containing berberine
-2020Promastigotes and amastigote L. major0.01–50 μg/mL24, 48, 72 hThe results showed that this compound has potent anti-leishmanial effects against promastigotes and amastigotes of L. major with IC50 values ranging from 0.197 to 1.023 μg/mL.[40]
Curcumin-loaded mannose-functionalized chitosan nanoparticles
(Cur-MCN)
-2018Amastigotes
L. donovani0.05–2.0 mg/L72 hThe results showed that Cur-MCN at the concentration of 0.518 ± 0.01 mg/L reduced 50% of amastigotes; also, no toxic effect on macrophages was observed in the use of Cur-MCN.[41]
Encapsulate S-nitroso-mercaptosuccinic acid into chitosan nanoparticles (NONPs)-2019Promastigotes and amastigotesL. amazonensis25, 50, 75, 100, 200, 400 µM24 hExperiments on amastigotes and promastigotes of L. amazonensis showed that NONPs reduced 65% of the parasitic load at a dose of 200 µM and killed 85% of promastigotes at a dose of 75 µM. These nanoparticles also reduced the number of amastigotes from 8.5 ± 1.2 in the control group to 4.5 ± 0.4 per 300 macrophages and reduced the infection rate from 76.2 ± 7.1 to 63.7 ± 5.4.[42]
Mannosylated chitosan (MCS)
with dextran
(dex)
Paromomycin (PM)2019Amastigotes L. major5, 10, 20, 40, 80, 160, 320 μg/mL24, 48 hThe results showed that this compound has no cytotoxicity on macrophages and at a dose of 5 μg/mL reduced more than 60% of the parasitic load inside macrophages.[43]
Nanosized chitosan-betulinic acid-2020Promastigotes and amastigoteL. major20 μg/mL48 hThe results showed that BK20 (20 μg/mL) was effective to kill the parasite by 86% compared to negative control group. The infection rate and the mean number of amastigotes per each macrophage were found to be 73% and 7%, respectively.[44]
Oleoyl chitosan and α-cyclodextrin
(α-CD)
-2019Amastigotes L. major100 μL4 daysThe results showed that the use of oleoyl chitosan/α-CD platelets at a dose of 60.24 ± 4.42 μg/mL killed 50% of amastigotes.[45]
Poly
(isobutylcyano acrylate) nanoparticles coated with chitosan (Cs-NPs)
Amphotericin B-deoxycholate (AmB-DOC)2019Promastigotes and amastigoteL. major20 mg/mL10 min, 20 min, 30 min, 1 h, or 2 hThe IC50 values for L. major promastigote and axenic amastigote forms were 1.14 ± 0.11 μg/mL and 0.53 ± 0.07 μg/mL, respectively.[46]
Sodium alginate-glycol chitosan stearate nanoparticles
(SA-GCS-NP)
Amphotericin B (AmB)2015Amastigotes L. donovani10 ng/mL48 hThe IC50 values of AmB-SAGCS-NP and AmB for amastigotes of L. donovani were 0.128 ± 0.024 μg/mL and 0.214 ± 0.06 μg/mL, respectively.[47]
Table 2. A list of in vivo studies of chitosan and its derivatives as resources for anti-leishmanial agents.
Table 2. A list of in vivo studies of chitosan and its derivatives as resources for anti-leishmanial agents.
Type of ChitosanCombined with the DrugMethodAdministration AnimalLeishmania spp.DoseTimeOutcomeYear of PublicationRef.
AmB-loaded pluronic F127 (PF 127) micelles coated with chitosan (Cs-PF-AmB-M)Amphotericin B (AmB)Film hydration methodIntraperitonealSyrian golden hamsterLeishmania donovani1 mg/kg5 daysThe results showed that Cs-PF-AmB-M and PF-AmB-M significantly reduced the parasite load; also, the number of amastigotes was significantly reduced by 52.67 ± 17.24.2017[30]
Chitosan--Topically BALB/c miceLeishmania major200, 400 μg/mL28 daysChitosan reduced the size of the lesion from 10.7 ± 3.24 mm in the control group to 1.05 ± 1.02 mm on day 28 at a dose of 400 μg/mL.2018[31]
Chitosan microparticlesDoxorubicin hydrochloride (DOX) -IntraperitonealGolden hamstersLeishmania donovani500 mg/kg7 daysThe results showed that this compound killed 78.2 ± 10.4% of amastigotes.2011[33]
Chitosan anchored nanostructured lipid carriers (NLC)Miltefosine (HePC-
hexadecyl
phosphocholine)
and amphotericin B (AmB)
-IntravenousNaive hamstersLeishmania donovani1 mg/kg5 daysThe results showed that HePC-AmB-CNLCs could reduce the parasitic load by 88.14 ± 4.12%, while tween 80-AmB-CNLCs and AmB reduced the parasite load by 70.91 ± 3.5% and 53.26 ± 2.5%, respectively.2017[34]
Chitosan nanocapsule (CNC)Amphotericin BEmulsification n-solvent evaporationIntraperitonealSyrian golden
hamsters
Leishmania donovani1 mg of drug/kg30 daysThe results showed that this compound killed 86.1 ± 2.08% of Leishmania amastigotes.2013[55]
Chitosan nanoparticles-Ionotropic gelation processSubcutaneouslyBALB/c miceLeishmania major5 μg/50 μL3 weeksThe results showed that injection of this compound in BALB/c mice could activate TH1 cells and IgG2a and eradicate Leishmania with cell-mediated immunity.2011[56]
Chitosan nanoparticlesAmphotericin B (AmpB)Polyelectrolyte complexes techniqueIntravenousBALB/c miceLeishmania amazonensis100 μL/kg10 daysThe results showed that the combined use of chitosan and chondroitin sulfate nanoparticles with amphotericin B can significantly reduce the lesion size and parasitic load and also provide higher levels of IFN-γ and IL-12.2014[54]
Chitosan nanoparticles
(CNPs) and, 4-SO4GalNAc modified chitosan nanoparticles
(SCNP)
Amphotericin B (AmpB)Ionic gelationIntravenousWistar ratsLeishmania donovani1 mg/kg0.5,
1, 2, 4, 6 and 24 h
The results showed that the use of AmB-SCNPs reduced the load of parasites in the spleen by 75.30 ± 3.76%, but the use of AmB-CNPs and amphotericin B alone kills 63.89 ± 3.44% and 47.56 ± 2.37% of parasites.2015[36]
Chitosan nanoparticles and
sodium tripolyphosphate (TPP)
Amphotericin B (AmB)Dextran sulphate aqueous solution IntravenousBALB/c miceLeishmania major1.25, 2.5, 5 mg/kg10 daysAmB-CH-TPP at a dose of 5 mg/kg reduced the size of the lesion by 83% and also reduce the parasitic load by 99%, but CH-TPP only reduced 35% of the lesion size and 65% of parasitic load.2020[39]
Chitosan plateletsAmphotericin B-deoxycholate-Intralesional
BALB/c miceLeishmania major100 μL/kg13 daysThe results showed that the use of AmB-DOC and the chitosan platelets caused thickening and dry scales on the lesion, which indicated improvement; granuloma spread in these mice is more limited and the number of infected macrophages is less than the use of AmB-DOC.2019[45]
Curcumin-loaded mannose-functionalized chitosan nanoparticles
(Cur-MCN)
--IntraperitonealHamsterLeishmania donovani50 mg/kg5 daysThe results showed that Cur-MCN have more anti-leishmaniasis properties than curcumin alone and are also more efficient at drug delivery than Cur-CN (curcumin-loaded unconjugated chitosan nanoparticles). Cur-MCN were able to reduce the parasitic load in the spleen by 94.20% and the number of amastigotes from 1647 ± 125.2 in the control group to 112 ± 32.2 per 500 macrophages.2018[41]
Nano chitosan Amphotericin B-IntralesionalBALB/c miceLeishmania major5, 7, 10 mg/kg3 weeksThe results showed that this compound improved the lesion and reduce its diameter to 0 mm and killed 81% of amastigotes; additionally, no mortality was reported in mice after using this compound; while using amphotericin B alone, 10% of mice died, and no toxicity or side effects were reported.2018[57]
Poly
(isobutylcyano acrylate) nanoparticles coated with chitosan (Cs-NPs)
Amphotericin B-deoxycholate (AmB-DOC)-Topically BALB/c miceLeishmania major100 μL/kg13 daysThe results showed that topical application of this compound with or without AmB-DOC on the skin of L. major mice could cause a slight improvement of the CL lesion; also, the collected skin samples showed that this combination reduces the parasitic load.2019[46]
Sodium alginate-glycol chitosan stearate nanoparticles
(SA-GCS-NP)
Amphotericin B (AmB)Ionotropic complexation methodIntraperitoneallySyrian golden
hamsters
Leishmania donovani5, 10, 20 mg/kg5 daysThe results showed that AmB-SAGCS-NP reduced 70.21 ± 3.46% of the parasitic load, while AmB kills only 53.24 ± 2.84% of amastigotes. 2015[47]
β-lapachone (βLP) in lecithin-chitosan nanoparticles (NP)--Topically BALB/c miceLeishmania major20 mg/kg21 daysThe use of these nanoparticles in CL reduced the number of amastigotes from 46 to 11 per 100 macrophages; also, these nanoparticles reduced the size of the lesion from 61.2 ± 21.2 mm2 to 35.7 ± 29.4 mm2.2015[58]
Nanosized chitosan-betulinic acid-Drug adsorption and phase separationIntraperitoneally BALB/c miceLeishmania major20 mg/kg28 daysThe lesion size in positive control group (GUL200) was negligibly decreased to 1.2 mm; also, in B20 mg/kg and K12.5 mg/kg receiver mice, the lesion size was slightly decreased, while in the group of BK20 mg/kg, the lesion size was considerably decreased and reached to zero (p < 0.001)2018[59]
Chitosan-based nano-scaffolds-Electrospinning methodTopicallyBALB/c miceLeishmania major20 wt%28 daysThis compound significantly reduced skin ulcer diameter (p = 0.000), parasite burden (p = 0.003), changes in the epidermis (p = 0.023), and dermis (p = 0.032); indicated significantly strong effectiveness of the produced nano-scaffolds against Leishmania ulcers.2020[44]
N-Palmitoyl-N-monomethyl-N,N-dimethyl-N,N,N-trimethyl6-O-glycol chitosan
nanoparticles (GCPQ)
Amphotericin B-Orally BALB/c miceLeishmania
infantum
5 mg/kg10 daysAmB-GCPQ nanoparticles demonstrated higher efficacy compared with parenteral liposomal AmB.2015[60]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

AlMohammed, H.I.; Khudair Khalaf, A.; E. Albalawi, A.; Alanazi, A.D.; Baharvand, P.; Moghaddam, A.; Mahmoudvand, H. Chitosan-Based Nanomaterials as Valuable Sources of Anti-Leishmanial Agents: A Systematic Review. Nanomaterials 2021, 11, 689. https://doi.org/10.3390/nano11030689

AMA Style

AlMohammed HI, Khudair Khalaf A, E. Albalawi A, Alanazi AD, Baharvand P, Moghaddam A, Mahmoudvand H. Chitosan-Based Nanomaterials as Valuable Sources of Anti-Leishmanial Agents: A Systematic Review. Nanomaterials. 2021; 11(3):689. https://doi.org/10.3390/nano11030689

Chicago/Turabian Style

AlMohammed, Hamdan I., Amal Khudair Khalaf, Aishah E. Albalawi, Abdullah D. Alanazi, Parastoo Baharvand, Ali Moghaddam, and Hossein Mahmoudvand. 2021. "Chitosan-Based Nanomaterials as Valuable Sources of Anti-Leishmanial Agents: A Systematic Review" Nanomaterials 11, no. 3: 689. https://doi.org/10.3390/nano11030689

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