We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Nanoparticles for combined photo- and chemo-dynamic therapy of cancer cells involving endogenous glutathione depletion

    Ashi Mittal

    Department of Chemistry, University of Delhi, Delhi, 110007, India

    ,
    Monika Yadav

    Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi, 110007, India

    ,
    Largee Biswas

    Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi, 110007, India

    ,
    Anita Kamra Verma

    **Author for correspondence: Tel.: +91 981 892 1222;

    E-mail Address: akverma@kmc.du.ac.in

    Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi, 110007, India

    Fellow, Delhi School of Public Health, Institution of Eminence, University of Delhi, Delhi, 110007, India

    &
    Indrajit Roy

    *Author for correspondence: Tel.: +91 956 072 1851;

    E-mail Address: indrajitroy11@gmail.com

    Department of Chemistry, University of Delhi, Delhi, 110007, India

    Published Online:https://doi.org/10.2217/nnm-2023-0047

    Background: Reactive oxygen species (ROS) are powerful weapons for various anticancer therapies. However, high glutathione (GSH) levels in cancer cells can significantly reduce the efficacy of such therapies. Methods: In this study, pH-responsive fluorescein-encapsulated zeolitic imidazolate framework-8 nanoparticles were synthesized for ROS-mediated combination therapy. Results: Upon blue light activation, fluorescein displayed a high singlet oxygen photogeneration ability for photodynamic therapy. Concurrently, accumulated Zn2+ from degraded zeolitic imidazolate framework-8 stimulated simultaneous ROS generation and GSH depletion, thereby successfully inducing chemodynamic therapy. This triggered a cascade of photo–physical and chemical processes culminating in the localized generation of ROS, ultimately breaking the intracellular redox equilibrium. Conclusion: This nanoformulation can potentially be used for light-activated ROS-mediated therapy for the management of superficial tumors.

    Plain language summary

    Highly reactive molecules called reactive oxygen species (ROS) are known to be present in excess in cancer cells. As a result, cancer cells are more susceptible to death by any further rise in levels of these species. In the current study, fluorescein-encapsulated zeolitic imidazolate nanoparticles were prepared for blue light-activated ROS-enhancing combination therapy. The nanoparticles displayed significant toxicity against a breast cancer cell line and simultaneously induced glutathione depletion, an antioxidant known to reduce the efficacy of various cancer therapies. Thus, this study reveals the potential of fluorescein-encapsulated zeolitic imidazolate nanoparticles for light-activated ROS-mediated therapy for the treatment of superficial tumors.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. WHO. WHO Cancer. www.who.int/en/news-room/fact-sheets/detail/cancer
    • 2. dos Santos AF, de Almeida DRQ, Terra LF, Baptista MS, Labriola L. Photodynamic therapy in cancer treatment – an update review. J. Cancer Metastasis Treat. 5, 25 (2019).
    • 3. Agostinis P, Berg K, Cengel KA et al. Photodynamic therapy of cancer: an update. CA Cancer J. Clin. 61(4), 250–281 (2011).
    • 4. Chen J, Fan T, Xie Z et al. Advances in nanomaterials for photodynamic therapy applications: status and challenges. Biomaterials 237, 119827 (2020).
    • 5. Perillo B, Di Donato M, Pezone A et al. ROS in cancer therapy: the bright side of the moon. Exp. Mol. Med. 52(2), 192–203 (2020).
    • 6. Yang H, Villani RM, Wang H et al. The role of cellular reactive oxygen species in cancer chemotherapy. J. Exp. Clin. Cancer Res. 37, 266 (2018).
    • 7. Cao C, Wang X, Yang N, Song X, Dong X. Recent advances of cancer chemodynamic therapy based on Fenton/Fenton-like chemistry. Chem. Sci. 13(4), 863–889 (2022).
    • 8. Tang Z, Liu Y, He M, Bu W. Chemodynamic therapy: tumour microenvironment-mediated Fenton and Fenton-like reactions. Angew. Chem. Int. Ed. 58(4), 946–956 (2019).
    • 9. Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 17(11), 657–674 (2020).
    • 10. Li J, Zhuang Z, Zhao Z, Tang BZ. Type I AIE photosensitizers: mechanism and application. View 3(2), 20200121 (2021).
    • 11. Chen D, Liang C, Qu X et al. Metal-free polymer nano-photosensitizer actuates ferroptosis in starved cancer. Biomaterials 292, 121944 (2023).
    • 12. Wang P, Liang C, Zhu J et al. Manganese-based nanoplatform as metal ion-enhanced ROS generator for combined chemodynamic/photodynamic therapy. ACS Appl. Mater. Interfaces 11(44), 41140–41147 (2019).
    • 13. Xie Z, Liang S, Cai X et al. O2-Cu/ZIF-8@Ce6/ZIF-8@F127 composite as a tumor microenvironment-responsive nanoplatform with enhanced photo-/chemodynamic antitumor efficacy. ACS Appl. Mater. Interfaces 11(35), 31671–31680 (2019).
    • 14. Kuppusamy P, Li H, Ilangovan G et al. Noninvasive imaging of tumor redox status and its modification by tissue glutathione levels. Cancer Res. 62(1), 307–312 (2002).
    • 15. Lai X, Geng X, Li M et al. Glutathione-responsive PLGA nanocomplex for dual delivery of doxorubicin and curcumin to overcome tumor multidrug resistance. Nanomedicine 16(16), 1411–1427 (2021).
    • 16. Lin Y, Chen X, Yu C et al. Radiotherapy-mediated redox homeostasis-controllable nanomedicine for enhanced ferroptosis sensitivity in tumor therapy. Acta Biomater. 159, 300–311 (2023).
    • 17. Li X, Wang Q, Yu S et al. Multifunctional MnO2-based nanoplatform-induced ferroptosis and apoptosis for synergetic chemoradiotherapy. Nanomedicine 16(26), 2343–2361 (2021).
    • 18. Keskin S, Kızılel S. Biomedical applications of metal organic frameworks. Ind. Eng. Chem. Res. 50(4), 1799–1812 (2011).
    • 19. Lawson HD, Walton SP, Chan C. Metal-organic frameworks for drug delivery: a design perspective. ACS Appl. Mater. Interfaces 13(6), 7004–7020 (2021).
    • 20. Chen B, Yang Z, Zhu Y, Xia Y. Zeolitic imidazolate framework materials: recent progress in synthesis and applications. J. Mater. Chem. A 2(40), 16811–16831 (2014).
    • 21. Phan A, Doonan CJ, Uribe-Romo FJ, Knobler CB, O'Keeffe M, Yaghi OM. Synthesis, structure, and carbon dioxide capture properties of zeolitic imidazolate frameworks. ACC Chem. Res. 43(1), 58–67 (2010).
    • 22. Mittal A, Gandhi S, Roy I. Mechanistic interaction studies of synthesized ZIF-8 nanoparticles with bovine serum albumin using spectroscopic and molecular docking approaches. Sci. Rep. 12, 10331 (2022).
    • 23. Sun CY, Qin C, Wang XL et al. Zeolitic imidazolate framework-8 as efficient pH-sensitive drug delivery vehicle. Dalton Trans. 41(23), 6906–6909 (2012).
    • 24. Chen P, He M, Chen B, Hu B. Size- and dose-dependent cytotoxicity of ZIF-8 based on single cell analysis. Ecotoxicol. Environ. Saf. 205, 111110 (2020). •• Increased accumulation of zinc ions due to the high bioavailability of zeolitic imidazolate framework-8 results in enhanced reactive oxygen species generation.
    • 25. Zhao H, Li T, Yao C et al. Dual roles of metal-organic frameworks as nanocarriers for miRNA delivery and adjuvants for chemodynamic therapy. ACS Appl. Mater. Interfaces 13(5), 6034–6042 (2021). • Reports that zinc ions triggered reactive oxygen species generation by zeolitic imidazolate framework-8 nanoparticles for chemodynamic therapy.
    • 26. Hu H, Su M, Ba H et al. ZIF-8 nanoparticles induce neurobehavioral disorders through the regulation of ROS-mediated oxidative stress in zebrafish embryos. Chemosphere 305, 135453 (2022).
    • 27. Garg P, Kaur G, Sharma B, Chaudhary GR. Fluorescein-metal hybrid surfactant conjugates as a smart material for antimicrobial photodynamic therapy against Staphylococcus aureus. ACS Appl. Bio. Mater. 3(7), 4674–4683 (2020).
    • 28. WHO. WHO model list of essential medicines – 22nd list (2021). www.who.int/publications/i/item/WHO-MHP-HPS-EML-2021.02
    • 29. Kim MM, Darafsheh A. Light sources and dosimetry techniques for photodynamic therapy. Photochem. Photobiol. 96(2), 280–294 (2020).
    • 30. Hatakeyama T, Murayama Y, Komatsu S et al. Efficacy of 5-aminolevulinic acid-mediated photodynamic therapy using light-emitting diodes in human colon cancer cells. Oncol. Rep. 29(3), 911–916 (2013).
    • 31. Jamali Z, Hejazi SM, Ebrahimi SM, Moradi-Sardareh H, Paknejad M. Effects of LED-based photodynamic therapy using red and blue lights, with natural hydrophobic photosensitizers on human glioma cell line. Photodiag. Photodyn. Ther. 21, 50–54 (2018).
    • 32. Yoshimoto T, Shimada M, Tokunaga T et al. Blue light irradiation inhibits the growth of colon cancer and activation of cancer-associated fibroblasts. Oncol. Rep. 47(5), 104 (2022). • Study showing antitumor effects of blue light-emitting diode light on colon cancer.
    • 33. Zhuang J, Kuo CH, Chou LY, Liu DY, Weerapana E, Tsung CK. Optimized metal-organic framework nanospheres for drug delivery: evaluation of small-molecule encapsulation. ACS Nano 8(3), 2812–2819 (2014). • Study reporting general synthesis route for encapsulating small molecules such as fluorescein and camptothecin in zeolitic imidazolate framework-8 nanospheres.
    • 34. Hu Y, Kazemian H, Rohani S, Huang Y, Song Y. In situ high pressure study of ZIF-8 by FTIR spectroscopy. Chem. Commun. 47(47), 12694–12696 (2011).
    • 35. dos Santos Ferreira da Silva J, López Malo D, Anceski Bataglion G et al. Adsorption in a fixed-bed column and stability of the antibiotic oxytetracycline supported on Zn(II)-[2-methylimidazolate] frameworks in aqueous media. PLOS ONE 10(6), e0128436 (2015).
    • 36. Zhao B, Yin JJ, Bilski PJ, Chignell CF, Roberts JE, He YY. Enhanced photodynamic efficacy towards melanoma cells by encapsulation of Pc4 in silica nanoparticles. Toxicol. Appl. Pharmacol. 241(2), 163–172 (2009).
    • 37. Yang C, Jiang L, Zhang H, Shimoda LA, DeBerardinis RJ, Semenza GL. Analysis of hypoxia-induced metabolic reprogramming. Methods Enzymol. 542, 425–455 (2014).
    • 38. Kou L, Sun J, Zhai Y, He Z. The endocytosis and intracellular fate of nanomedicines: implication for rational design. Asian J. Pharm. Sci. 8(1), 1–10 (2013).
    • 39. Lajoie P, Nabi IR. Lipid rafts, caveolae, and their endocytosis. Int. Rev. Cell Mol. Biol. 282, 135–163 (2010).
    • 40. Valentovic M. Lovastatin. In: The Comprehensive Pharmacology Reference. Enna SJBylund DB (Eds). Elsevier, Amsterdam, The Netherlands, 1–5 (2007).
    • 41. Kakiesonen M, Roux A. Mechanisms of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 19(5), 313–326 (2018).
    • 42. Gou S, Zhang X, Zheng M et al. Selectivity of commonly used inhibitors of clathrin-mediated and caveolae-dependent endocytosis of G protein-coupled receptors. Biochim. Biophys. Acta Biomembr. 1848(10), 2101–2110 (2015).
    • 43. Chen CL, Hou WH, Liu IH, Hsiao G, Huang SS, Huang JS. Inhibitors of clathrin-dependent endocytosis enhance TGFβ signaling and responses. J. Cell Sci. 122(11), 1863–1871 (2009).
    • 44. Exocytosis and endocytosis. In: Methods in Molecular Biology: Volume 440. Ivanov AI (Ed.). Human Press, NY, USA, 247–257 (2008).
    • 45. Sato K, Nagai J, Mitsui N, Yumoto R, Takano M. Effects of endocytosis inhibitors on internalization of human IgG by Caco-2 human intestinal epithelial cells. Life Sci. 85(23-26), 800–807 (2009).
    • 46. Garrett WS, Mellman I. Studies of endocytosis. In: Dendritic Cells (2nd Edition). Lotze MThomson A (Eds). Academic Press, Amsterdam, The Netherlands, 213–229 (2001).
    • 47. dos Santos T, Varela J, Lynch I, Salvati A, Dawson KA. Effects of transport inhibitors on the cellular uptake of carboxylated polystyrene nanoparticles in different cell lines. PLOS ONE 6(9), e24438 (2011).
    • 48. Chen X, Tong R, Shi Z et al. MOF nanoparticles with encapsulated autophagy inhibitor in controlled drug delivery system for antitumor. ACS Appl. Mater. Interfaces 10(3), 2328–2337 (2018).
    • 49. Xu D, You Y, Zeng F et al. Disassembly of hydrophobic photosensitizer by biodegradable zeolitic imidazolate framework-8 for photodynamic cancer therapy. ACS Appl. Mater. Interfaces 10(18), 15517–15523 (2018).
    • 50. Mourdikoudis S, Pallares RM, Thanh NTK. Characterization techniques for nanoparticles: comparison and complementarity upon studying nanoparticle properties. Nanoscale 10(27), 12871–12934 (2018).
    • 51. Schejn A, Balan L, Falk V, Aranda L, Medjahdi G, Schneider R. Controlling ZIF-8 nano- and microcrystal formation and reactivity through zinc salt variations. CrystEngComm 16(21), 4493–4500 (2014).
    • 52. Gao S, Hou J, Deng Z et al. Improving the acidic stability of zeolitic imidazolate frameworks by biofunctional molecules. Chem 5(6), 1597–1608 (2019).
    • 53. Pelicano H, Carney D, Huang P. ROS stress in cancer cells and therapeutic implications. Drug Resist. Updat. 7(2), 97–110 (2004).
    • 54. Figueroa D, Asaduzzaman M, Young F. Real time monitoring and quantification of reactive oxygen species in breast cancer cell line MCF-7 by 2′,7′-dichlorofluorescein diacetate (DCFDA) assay. J. Pharmacol. Toxicol. Methods 94(Pt 1), 26–33 (2018).
    • 55. Kaja S, Payne AJ, Naumchuk Y, Koulen P. Quantification of lactate dehydrogenase for cell viability testing using cell lines and primary cultured astrocytes. Curr. Protoc. Toxicol. 72, 2.26.1–2.26.10 (2017).
    • 56. Xu W, Liu LZ, Loizidou M, Ahmed M, Charles IG. The role of nitric oxide in cancer. Cell Res. 12(5-6), 311–320 (2002).
    • 57. Zitka O, Skalickova S, Gumulec J et al. Redox status expressed as GSH:GSSG ratio as a marker for oxidative stress in paediatric tumour patients. Oncol. Lett. 4(6), 1247–1253 (2012).
    • 58. Traverso N, Ricciarelli R, Nitti M et al. Role of glutathione in cancer progression and chemoresistance. Oxid. Med. Cell Longev. 2013, 972913 (2013). •• Role of glutathione depletion in sensitizing cancer cells to therapy.
    • 59. Zhang M, Qin X, Zhao Z et al. A self-amplifying nanodrug to manipulate the Janus-faced nature of ferroptosis for tumor therapy. Nanoscale Horiz. 7(2), 198–210 (2022).
    • 60. Krezel A, Bal W. Studies of zinc(II) and nickel(II) complexes of GSH, GSSG and their analogs shed more light on their biological relevance. Bioinorg. Chem. Appl. 2(3-4), 293–305 (2004).
    • 61. Li NC, Gawron O, Bascuas G. Stability of zinc complexes with glutathione and oxidized glutathione. J. Am. Chem. Soc. 76(1), 225–229 (1954).
    • 62. Fanucchi MV. Development of antioxidant and xenobiotic metabolizing enzyme systems. In: The Lung. Harding RPinkerton KEPlopper CG (Eds). Elsevier, Academic Press, CA, USA, 177–185 (2004).
    • 63. Couto N, Wood J, Barber J. The role of glutathione reductase and related enzymes on cellular redox homeostasis network. Free Radic. Biol. Med. 95, 27–42 (2016).
    • 64. Beckett GJ, Chapman BJ, Dyson EH, Hayes JD. Plasma glutathione S-transferase measurements after paracetamol overdose: evidence for early hepatocellular damage. Gut 26(1), 26–31 (1985).
    • 65. Wang F, Zhang YQ. Bioconjugation of silk fibroin nanoparticles with enzyme and peptide and their characterization. In: Advances in Protein Chemistry and Structural Biology: Volume 98. Donev R (Ed.). Elsevier, Academic Press, MA, USA, 263–291 (2015).
    • 66. Shigemitsu H, Ohkubo K, Sato K et al. Fluorescein-based type I supramolecular photosensitizer via induction of charge separation by self-assembly. JACS Au 2(6), 1472–1478 (2022).
    • 67. Kruszewski M. Labile iron pool: the main determinant of cellular response to oxidative stress. Mutat. Res. Fundam. Mol. Mech. Mutagenes 531(1-2), 81–92 (2003). •• Role of ‘free’ cellular iron in inducing oxidative damage by participating in a Fenton reaction.
    • 68. Petronek MS, Spitz DR, Allen BG. Iron-sulfur cluster biogenesis as a critical target in cancer. Antioxidants (Basel) 10(9), 1458 (2021).