Skip to main content
Log in

Novel Silibinin Loaded Chitosan-Coated PLGA/PCL Nanoparticles Based Inhalation Formulations with Improved Cytotoxicity and Bioavailability for Lung Cancer

  • Published:
BioNanoScience Aims and scope Submit manuscript

Abstract

Silibinin is widely used as an anticancer agent. There are important matters of concern regarding the drug pharmacokinetics related to the conventional formulation due to their low solubility and bioavailability. The aim of the present investigation was to develop, characterize, and evaluate chitosan-coated PLGA/PCL nanoparticles containing Silibinin (SB) intended for pulmonary delivery for treating lung cancer. Nanoparticles were prepared by double emulsion solvent evaporation method followed by lyophilized to obtain inhalation powder form. The prepared nanoparticles (NPs) were evaluated for their physicochemical characteristic along with DSC, FTIR, and SEM. The anticancer activity of SB-loaded NPs was assessed in the human A549 lung cancer cell line utilizing MTT assay and anti-cancer potential assessed by clonogenic assay. The pharmacokinetics and tissue distribution studies of SB-loaded NPs were assessed in comparison with the SB solution. The prepared NPs exhibited particle sizes in the range of 187–284 nm. Zeta potential was altered from negative to positive due to the coating with chitosan. Chitosan-coated NPs showed the sustain release effect up to 48 h with an aerodynamic particle size of 1.82 μm. Moreover, a tremendous increase in cell inhibition was determined by chitosan-coated PLGA NPs. Pharmacokinetics study showed that chitosan coating onto PLGA nanoparticles promoted to release drug in the lungs and increased vivo residence time. Thus, proposed formulations with improved bioavailability can be useful for efficient pulmonary delivery. These nanoparticles coated with chitosan could open a new avenue for effective treatment of lung cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

References

  1. Jemal, A., Siegel, R., Ward, E., et al. (2008). Cancer statistics. CA: a Cancer Journal for Clinicians, 58(2), 71–96.

    Google Scholar 

  2. Rowinsky, E. K., Onetto, N., Canetta, R. M., et al. (1992). Taxol: the first of the taxanes, an important new class of antitumor agents. Seminars in Oncology, 19(6), 646–662.

    Google Scholar 

  3. Ferrari, M. (2005). Cancer nanotechnology: opportunities and challenges. Nature Reviews. Cancer, 5, 161–171.

    Google Scholar 

  4. Nie, S., Xing, Y., Kim, G. J., et al. (2007). Nanotechnology applications in cancer. Annual Review of Biomedical Engineering, 9, 257–288.

    Google Scholar 

  5. Danhier, F., Ansorena, E., Silva, J. M., et al. (2012). PLGA-based nanoparticles: an overview of biomedical applications. Journal of Controlled Release, 161, 505–522.

    Google Scholar 

  6. Varypataki, E. M., Silva, A. L., Barnier-Quer, C., et al. (2016). Synthetic long peptide-based vaccine formulations for induction of cell mediated immunity: a comparative study of cationic liposomes and PLGA nanoparticles. Journal of Controlled Release, 226, 98–106.

    Google Scholar 

  7. Wu, J., Deng, C., Meng, F., et al. (2017). Hyaluronic acid coated PLGA nanoparticulate docetaxel effectively targets and suppresses orthotopic human lung cancer. Journal of Controlled Release, 259, 76–82.

    Google Scholar 

  8. El-Hammadi, M. M., Delgado, A. V., Melguizo, C., et al. (2017). Folic acid-decorated and PEGylated PLGA nanoparticles for improving the antitumour activity of 5-fluorouracil. International Journal of Pharmaceutics, 516, 61–70.

    Google Scholar 

  9. Chen, H., Xie, L. Q., Qin, J., et al. (2016). Surface modification of PLGA nanoparticles with biotinylated chitosan for the sustained in vitro release and the enhanced cytotoxicity of epirubicin. Colloids and Surfaces. B, Biointerfaces, 138, 1–9.

    Google Scholar 

  10. Wang, F., Yuan, J., Zhang, Q., et al. (2018). PTX-loaded three-layer PLGA/CS/ALG nanoparticle based on layer-by-layer method for cancer therapy. Journal of Biomaterials Science. Polymer Edition, 29, 1566–1578.

    Google Scholar 

  11. Nafee, N., Taetz, S., Schneider, M., et al. (2007). Chitosan-coated PLGA nanoparticles for DNA/RNA delivery: effect of the formulation parameters on complexation and transfection of antisense oligonucleotides. Nanomedicine, 1, 173–183.

    Google Scholar 

  12. Vila, A., Sanchez, A., Tobıo, M., et al. (2002). Design of biodegradable particles for protein delivery. Journal of Controlled Release, 78, 15–24.

    Google Scholar 

  13. Kalam, M. A., Khan, A. A., Khan, S., et al. (2016). Optimizing indomethacin-loaded chitosan nanoparticle size, encapsulation, and release using Box–Behnken experimental design. International Journal of Biological Macromolecules, 87, 329–340.

    Google Scholar 

  14. Zhang, B., Wang, K., Si, J., et al. (2014). Charge-reversal polymers for biodelivery. In Bioinspired and biomimetic polymer systems for drug and gene delivery (Vol. 4, p. 223). Weinheim: Chemical Industry Press and Wiley-VCH Verlag GmbH & Co. KGaA.

    Google Scholar 

  15. Gibot, L., Chabaud, S., Bouhout, et al. (2014). Anticancer properties of chitosan on human melanoma are cell line dependent. International Journal of Biological Macromolecules, 72.

  16. Sun, N. Y., Wei, X. L., Wu, B. J., et al. (2008). Enhanced dissolution of silymarin/ polyvinylpyrrolidone solid dispersion pellets prepared by a one-step fluid-bed coating technique. Powder Technology, 182(1), 72–80.

    Google Scholar 

  17. El-Samaligy, M. S., Afifi, N. N., & Mahmoud, E. A. (2006). Increasing bioavailability of silymarin using a buccal liposomal delivery system: preparation and experimental design investigation. International Journal of Pharmaceutics, 308(1–2), 140–148.

    Google Scholar 

  18. Alipour, S., Montaseri, H., & Tafaghodi, M. (2010). Preparation and characterization of biodegradable paclitaxel loaded alginate microparticles for pulmonary delivery. Colloids and Surfaces. B, Biointerfaces, 81, 521–529.

    Google Scholar 

  19. Maeda, H. (2001). The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Advances in Enzyme Regulation, 41, 189–207.

    Google Scholar 

  20. Ahmad, J., Akhter, S., Rizwanullah, M., et al. (2015). Nanotechnology-based inhalation treatments for lung cancer: state of the art. Nanotechnology, Science and Applications, 19(8), 55–66.

    Google Scholar 

  21. Jiang, L., Li, X., Liu, L., & Zhang, Q. (2013). Thiolated chitosan-modified PLA-PCL-TPGS nanoparticles for oral chemotherapy of lung cancer. Nanoscale Research Letters, 8(1), 66.

    Google Scholar 

  22. Eisenhauer, E. A., & Vermorken, J. B. (1998). The taxoids. Comparative clinical pharmacology and therapeutic potential. Drugs, 55(1), 5–30.

    Google Scholar 

  23. Montero, A., Fossella, F., Hortobagyi, G., et al. (2005). Docetaxel for treatment of solid tumours: a systematic review of clinical data. Lancet Oncology, 6(4), 229–239.

    Google Scholar 

  24. Kraus, L. A., Samuel, S. K., Schmid, S. M., et al. (2003). The mechanism of action of docetaxel (Taxotere) in xenograft models is not limited to bcl-2 phosphorylation. Investigational New Drugs, 21(3), 259–268.

    Google Scholar 

  25. Hernandez-Vargas, H., Palacios, J., & Moreno-Bueno, G. (2007). Telling cells how to die: docetaxel therapy in cancer cell lines. Cell Cycle, 6(7), 780–783.

    Google Scholar 

  26. Makadia, H. K., & Siegel, S. J. (2011). Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers, 3, 1377–1397.

    Google Scholar 

  27. Ha, E.-S., Han, D.-G., Seo, S.-W., et al. (2019). A simple HPLC method for the quantitative determination of silybin in rat plasma: application to a comparative pharmacokinetic study on commercial silymarin products. Molecules, 24, 2180–2188.

    Google Scholar 

  28. Mohammad, M., Badrana, F., Abdullah, H., et al. (2018). Novel docetaxel chitosan-coated PLGA/PCL nanoparticles with magnified cytotoxicity and bioavailability. Biomedicine & Pharmacotherapy, 106, 1461–1468.

    Google Scholar 

  29. Costa, P., & Sousa Lobo, J. M. (2001). Modeling and comparison of dissolution profiles. European Journal of Pharmaceutical Sciences, 13, 123–133.

    Google Scholar 

  30. Dash, S., Murthy, P. N., Nath, L., & Chowdhury, P. (2010). Kinetic modelling on drug release from controlled drug delivery systems. Acta Poloniae Pharmaceutica, 67, 217–223.

    Google Scholar 

  31. Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. Journal of Immunological Methods, 65(1–2), 55–63.

    Google Scholar 

  32. Pilicheva, B., Katsarov, P., & Kassarova, M. (2014). Flowability evaluation of dry powder inhalation formulations intended for nasal delivery of betahistine dihydrochloride. Sikk Manipal University Medical Journal, 2(1), 77–90.

    Google Scholar 

  33. Sinha, B., & Mukherjee, B. (2012). Development of an inhalation chamber and a dry powder inhaler device for administration of pulmonary medication in animal model. Drug Development and Industrial Pharmacy, 38, 171–179.

    Google Scholar 

  34. Luo, Y., Zhang, B., Cheng, W., et al. (2010). Preparation, characterization and evaluation of selenite-loaded chitosan / TPP nanoparticles with or without zein coating. Carbohydrate Polymers, 82, 942–951.

    Google Scholar 

  35. Bhattacharya, S. (2020). Fabrication and characterization of chitosan-based polymeric nanoparticles of Imatinib for colorectal cancer targeting application. International Journal of Biological Macromolecules., 151, 104–115.

    Google Scholar 

  36. Fernandes, C. A., & Vanbever, R. (2009). Preclinical models for pulmonary drug delivery. Expert Opinion on Drug Delivery, 6(11), 1231–1245.

    Google Scholar 

  37. NCI. (2012). Equivalent surface area dosage conversion factors, The Frederick National Lab Animal Care and Use Committee. 0, p. 1–2. 36.

  38. Center for Drug Evaluation and Research. Guidance for industry: estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers. US Department of Health and Human Services. 2005. Available from: http://www.fda.gov/downloads/Drugs/Guidance/UCM078932.pdf.

  39. Okuda, T., Suzuki, Y., Kobayashi, Y., et al. (2015). Development of biodegradable polycation-based inhalable dry gene powders by spray freeze drying. Pharmaceutics., 7(3), 233–254.

    Google Scholar 

  40. Xiang, Q.-y., Wang, M.-t., Chen, F., et al. (2007). Lung-targeting delivery of dexamethasone acetate loaded solid lipid nanoparticles. Archives of Pharmacal Research, 30(4), 519–525.

    Google Scholar 

  41. Rizzardini, M., Zappone, M., Villa, P., et al. (1998). Kupffer cell depletion partially prevents hepatic heme oxygenase 1 messenger RNA accumulation in systemic inflammation in mice: role of interleukin 1β. Hepatology., 27(3), 703–710.

    Google Scholar 

  42. Kunii, R., Onishi, H., & Machida, Y. (2007). Preparation and antitumor characteristics of PLA/(PEG-PPG-PEG) nanoparticles loaded with camptothecin. European Journal of Pharmaceutics and Biopharmaceutics, 67, 9–17.

    Google Scholar 

  43. Ocal, H., Arica-Yegin, B., Vural, I., et al. (2014). 5-Fluorouracil-loaded PLA/PLGA PEG-PPG-PEG polymeric nanoparticles: formulation, in vitro characterization and cell culture studies. Drug Development and Industrial Pharmacy, 40, 560–567.

    Google Scholar 

  44. Abouelmagd, S. A., Ku, Y. J., & Yeo. (2015). Low molecular weight chitosan-coated polymeric nanoparticles for sustained and pH-sensitive delivery of paclitaxel. Journal of Drug Targeting, 23, 725–735.

    Google Scholar 

  45. Shariatinia, Z. (2019). Pharmaceutical applications of chitosan. Advances in Colloid and Interface Science, 263, 131–194.

    Google Scholar 

  46. Taghavi, S., Ramezani, M., Alibolandi, M., et al. (2017). Chitosan-modified PLGA nanoparticles tagged with 5TR1 aptamer for in vivo tumor-targeted drug delivery. Cancer Letters, 400, 1–8.

    Google Scholar 

  47. Babu, A., Amreddy, N., Muralidharan, R., et al. (2017). Chemodrug delivery using integrin-targeted PLGA-chitosan nanoparticle for lung cancer therapy. Science Reporter, 7, 14674.

    Google Scholar 

  48. Ibrahim, M. M., Abdelgawad, H. A., Osama, A. S., et al. (2013). Nanoparticle-based topical ophthalmic formulations for sustained celecoxib release. Journal of Pharmaceutical Sciences, 102, 1036–1053.

    Google Scholar 

  49. Coffin, M. D., & McGinity, J. W. (1992). Biodegradable pseudolatexes: the chemical stability of poly (D,L-lactide) and poly (ε-caprolactone) nanoparticles in aqueous media. Pharmaceutical Research, 9, 200–205.

    Google Scholar 

  50. Lemoine, D., Francois, C., Kedzierewicz, F., et al. (1996). Stability study of nanoparticles of poly (ε-caprolactone), poly (D,L-lactide) and poly(D,L-lactide-co-glycolide). Biomaterials, 17, 2191–2197.

    Google Scholar 

  51. Lamprecht, A., Ubrich, N., Perez, H., et al. (2000). Influences of process parameters on nanoparticle preparation performed by a double emulsion pressure homogenization technique. International Journal of Pharmaceutics, 196, 177–182.

    Google Scholar 

  52. M. Malhotra, D.K. Majumdar (2001), Permeation through cornea, Indian J. Experimental Biology,39:11–24.

  53. Robinson, J. R., & Mlynek, G. M. (1995). Bioadhesive and phase-change polymers for ocular drug delivery. Advanced Drug Delivery Reviews, 16, 45–50.

    Google Scholar 

  54. Collado-González, M., González Espinosa, Y., & Goycoolea, F. M. (2019). Interaction between chitosan and mucin: fundamentals and applications. Biomimetics, 4.

  55. Ways, T. M. M., Lau, W. M., & Khutoryanskiy, V. V. (2018). Chitosan and its derivatives for application in mucoadhesive drug delivery systems. Polymers, 10, 24–30.

    Google Scholar 

  56. Gentile, P., Chiono, V., Carmagnola, I., et al. (2014). An overview of poly(lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. International Journal of Molecular Sciences, 15(3), 3640–3659.

    Google Scholar 

  57. Seju, U., Kumar, A., & Sawant, K. K. (2011). Development and evaluation of olanzapine-loaded PLGA nanoparticles for nose-to-brain delivery: in vitro and in vivo studies. Acta Biomaterialia, 7(12), 4169–4176.

    Google Scholar 

  58. Kalimouttou, S., Skiba, M., Bon, P., et al. (2009). PLGA nanoparticles: drug loading, characterization, in vitro drug release and in vivo studies. Journal of Nanoscience and Nanotechnology, 9(1), 150–158.

    Google Scholar 

  59. Patel, J., Dhingani, A., Garala, K., Raval, M., & Sheth, N. (2014). Quality by design approach for oral bioavailability enhancement of Irbesartan by self-nanoemulsifying tablets. Drug Delivery, 21(6), 412–435.

    Google Scholar 

  60. Musumeci, Teresa & Ventura, Cinzia & Giannone, et al (2006) PLA/PLGA nanoparticles for sustained release of docetaxel. International Journal of Pharmaceutics, 325:172–179.

  61. Bartsch, S. E., & Griesser, U. J. (2004). Physicochemical properties of the binary system glibenclamide and polyethylene glycol 4000. Journal of Thermal Analysis and Calorimetry, 77, 555–569.

    Google Scholar 

  62. Zhang, J. Q., Liu, J., Li, X. L., & Jasti, B. R. (2007). Preparation and characterization of solid lipid nanoparticles containing Silibinin. Drug Delivery, 14(6), 381–387.

    Google Scholar 

  63. Shah, P., Sarolia, J., Vyas, B., et al. (2020). PLGA nanoparticles for nose to brain delivery of clonazepam: formulation, optimization by 32 factorial design, in vitro and in vivo evaluation. Current Drug Delivery. https://doi.org/10.2174/1567201817666200708115627.

  64. Chronopoulou, L., Massimi, M., & Federica, M. (2013). Chitosan-coated PLGA nanoparticles: a sustained drug release strategy for cell cultures. Colloids and Surfaces. B, Biointerfaces, 103, 310–317.

    Google Scholar 

  65. Dhand, C., Prabhakaran, M., Beuerman, R., et al. (2014). Role of size of drug delivery carriers for pulmonary and intravenous administration with emphasis on cancer therapeutics and lung-targeted drug delivery. RSC Advances, 62.

  66. Al, Q., Grenha, A., Carrion, D. R., et al. (2012). Microencapsulated chitosan nanoparticles for pulmonary protein delivery: in vivo evaluation of insulin-loaded formulations. Journal of Controlled Release, 157(3), 383–390.

    Google Scholar 

  67. Suarez, S., & Hickey, A. J. (2000). Drug properties affecting aerosol behaviour. Respiratory Care, 45, 652–666.

    Google Scholar 

  68. Paul, P., Sengupta, S., Mukherjee, B., et al. (2018). Chitosan-coated nanoparticles enhanced lung pharmacokinetic profile of voriconazole upon pulmonary delivery in mice. Nanomedicine, 13(6), 1–13.

    Google Scholar 

  69. Yang, S. C., Lu, L. F., Cai, Y., et al. (1999). Body distribution in mice of intravenously injected camptothecin solid lipid nanoparticles and targeting effect on brain. Journal of Controlled Release, 59, 299–307.

    Google Scholar 

  70. Serrano, A. G., & Perez-Gil, J. (2006). Protein-lipid interactions and surface activity in the pulmonary surfactant system. Chemistry and Physics of Lipids, 141, 105–118.

    Google Scholar 

  71. Nguyen, J., Xie, X., Neu, M., et al. (2008). Effects of cell-penetrating peptides and pegylation on transfection efficiency of polyethylenimine in mouse lungs. The Journal of Gene Medicine, 10(11), 1236–1246.

    Google Scholar 

  72. Mohammed, M. A., Syeda, J. T. M., Wasan, K. M., et al. (2017). An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics., 9(4), 53.

    Google Scholar 

  73. Jeans, A. R., Howard, S. J., Al-Nakeeb, Z., et al. (2012). Combination of voriconazole and anidulafungin for treatment of triazole-resistant aspergillus fumigatus in an in vitro model of invasive pulmonary aspergillosis. Antimicrobial Agents and Chemotherapy, 56(10), 5180–5185.

    Google Scholar 

  74. Sangwan, S., Agosti, J. M., Bauer, L. A., et al. (2001). Aerosolized protein delivery in asthma: gamma camera analysis of regional deposition and perfusion. Journal of Aerosol Medicine, 14, 185–195.

    Google Scholar 

  75. Townsley, M. I. (2012). Structure and composition of pulmonary arteries, capillaries and veins. Comprehensive Physiology, 2, 675–709.

    Google Scholar 

  76. Dyawanapelly, S., Koli, U., Dharamdasani, V., et al. (2016). Improved mucoadhesion and cell uptake of chitosan and chitosan oligosaccharide surface-modified polymer nanoparticles for mucosal delivery of proteins. Drug Delivery and Translational Research, 6(4), 365–379.

    Google Scholar 

  77. Parveen, S., & Sahoo, S. K. (2011). Long circulating chitosan/PEG blended PLGA nanoparticle for tumor drug delivery. European Journal of Pharmacology, 670(2–3), 372–383.

    Google Scholar 

  78. Chen, H., Xie, L. Q., Qin, J., et al. (2016). Surface modification of PLGA nanoparticles with biotinylated chitosan for the sustained in vitro release and the enhanced cytotoxicity of epirubicin. Colloids and Surfaces. B, Biointerfaces, 138, 1–9.

    Google Scholar 

  79. Mangal, S., Gao, W., Li, T., et al. (2017). Pulmonary delivery of nanoparticle chemotherapy for the treatment of lung cancers: challenges and opportunities. Acta Pharmacologica Sinica, 38(6), 782–797.

    Google Scholar 

Download references

Funding

This work was financially supported by the Gujarat council on science and technology (GUJCOST), Department of Science & Technology, Government of Gujarat, Gandhinagar (grant numbers GUJCOST/MRP/16-17/306).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Priya Patel.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Research Involving Humans and Animals Statement

Human-NA. Animal-All experimental protocols were reviewed and approved by the Institutional Animal Ethics Committee (IAEC), constituted as per guidelines of the Committee for Purpose of Control and Supervision of Experiments on Animals Government of India (IAEC/DPS/SU/1705; dated 12th December 2016) prior to initiation of the experiment.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

ESM 1

(DOCX 131 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Raval, M., Patel, P., Airao, V. et al. Novel Silibinin Loaded Chitosan-Coated PLGA/PCL Nanoparticles Based Inhalation Formulations with Improved Cytotoxicity and Bioavailability for Lung Cancer. BioNanoSci. 11, 67–83 (2021). https://doi.org/10.1007/s12668-020-00797-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12668-020-00797-z

Keywords

Navigation