Protective Effect of Indomethacin-loaded Polymeric Nanoparticles Against Oxidative Stress-Induced Cytotoxicity in Human Breast Adenocarcinoma Cell Model

Authors

DOI:

https://doi.org/10.32635/2176-9745.RBC.2022v68n4.2545

Keywords:

indomethacin/pharmacology, antioxidants, nanocapsules, neoplasms

Abstract

Introduction: Anti-inflammatory drugs are being utilized to treat cancer because of its inflammatory microenvironment. Objective: The objective of this study is to investigate the antioxidant potential of indomethacin and its genotoxicity, since free or loaded in polymeric nanocapsules using MCF-7 (human breast cancer) cells as an in vitro model. Method: Development of indomethacin-loaded polyepsiloncaprolactone (PCL) nanocapsules by interfacial deposition method. It is characterized by pH determination by potentiometer, mean diameter and polydispersity index by dynamic light scattering; zeta potential by electrophoretic mobility; encapsulation efficacy by high performance liquid chromatography method; corona effect formation; 2ʹ,7ʹ-dichlorofluorescin diacetate (DCFH-DA) method by spectrofluorimetric assay; nitric oxide (NO) determination by spectrophotometric and genotoxicity assay by plasmid DNA cleavage method. Results: The results showed a mild acidic pH (4.78 ± 0.10), sizes around 200 nm and PDI<0.2 with a zeta potential around -20 mV and encapsulation efficiency of 99% (1 mg mL-1), showing a dose-dependent corona formation profile in 24h incubation. Conclusion: DCFH-DA assay showed no production of reactive oxygen species (ROS) while NO determination showed that Ind-OH-NC from 26.7 to 100 μM increased reactive nitrogen species (RNS), demonstrating antioxidant potential against MCF-7 cells. No sample at the concentrations evaluated induced DNA cleavage, being considered a safe treatment.

Downloads

Download data is not yet available.

References

Rahme E, Ghosn J, Dasgupta K, et al. Association between frequent use of nonsteroidal anti-inflammatory drugs and breast cancer. BMC Cancer. 2005;5:159. doi: https://doi.org/10.1186/1471-2407-5-159

Ackerstaff E, Gimi B, Artemov D, et al. Anti-inflammatory agent indomethacin reduces invasion and alters metabolism in a human breast cancer cell line. Neoplasia. 2007;9(3):222-35. doi: https://doi.org/10.1593/neo.06673

Zappavigna S, Cossu AM, Grimaldi A, et al. Anti-inflammatory drugs as anticancer agents. In J Mol Sci. 2020;21(7):2605. doi: https://doi.org/10.3390/ijms21072605

Halliwell B, Whiteman M. Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol. 2004;142(2):231-55. doi: https://doi.org/10.1038/sj.bjp.0705776

Sosa V, Moliné T, Somoza R, et al. Oxidative stress and cancer: an overview. Ageing Res Rev. 2013;12(1):376-90. doi: https://doi.org/10.1016/j.arr.2012.10.004

Crawford S. Anti-inflammatory/antioxidant use in long-term maintenance cancer therapy: a new therapeutic approach to disease progression and recurrence. Ther Adv Med Oncol. 2014;6(2):52-68. doi: https://doi.org/10.1177/1758834014521111

Oberley TD. Oxidative damage and cancer. Am J Pathol. 2002;160(2):403-8. doi: https://doi.org/10.1016/S0002-9440(10)64857-2

Kennedy RK, Veena V, Naik PR, et al. Phenazine-1-carboxamide (PCN) from pseudomonas sp. strain PUP6 selectively induced apoptosis in lung (A549) and breast (MDA MB-231) cancer cells by inhibition of antiapoptotic Bcl-2 family proteins. Apoptosis. 2015;20(6):858-68. doi: https://doi.org/10.1007/s10495-015-1118-0

Huerta S, Chilka S, Bonavida B. Nitric oxide donors: novel cancer therapeutics (review). Int J Oncol. 2008;33(5):909-27. doi: https://doi.org/10.3892/ijo_00000079

Galadari S, Rahman A, Pallichankandy S, et al. Reactive oxygen species and cancer paradox: to promote or to suppress? Free Radic Biol Med. 2017;04:144-64. doi: https://doi.org/10.1016/j.freeradbiomed.2017.01.004

Chinery R, Beauchamp RD, Shyr Y, et al. Antioxidants reduce cyclooxygenase-2 expression, prostaglandin production, and proliferation in colorectal cancer cells. Cancer Res [Internet]. 1998 [cited 2021 Dec 14];58(11):2323-7. Available from: https://aacrjournals.org/cancerres/article-pdf/58/11/2323/2466382/cr0580112323.pdf

Kummer CL, Coelho TCRB. Antiinflamatórios não esteróides inibidores da ciclooxigenase-2 (COX-2): aspectos atuais. Rev Bras Anestesiol. 2002;52(4):498-512. doi: https://doi.org/10.1590/S0034-70942002000400014

Farrugia G, Balzan R. The proapoptotic effect of traditional and novel nonsteroidal anti-inflammatory drugs in mammalian and yeast cells. Oxid Med Cell Longev. 2013;2013:504230. doi: https://doi.org/10.1155/2013/504230

Pantziarka P, Sukhatme V, Bouche G, et al. Repurposing drugs in oncology (ReDO) – diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi: https://doi.org/10.3332/ecancer.2016.610

Bernardi A, Jacques-Silva MC, Delgado-Cañedo A, et al. Nonsteroidal anti-inflammatory drugs inhibit the growth of C6 and U138-MG glioma cell lines. Eur J Pharmacol. 2006;532(3):214-22. doi: https://doi.org/10.1016/j.ejphar.2006.01.008

Franco C, Silva ML, Viana AR, et al. Cytotoxicity evaluation of indomethacin-loaded polymeric nanoparticles in a human breast adenocarcinoma cell model. Braz J Dev. 2021;7(7):67004-14. doi: https://doi.org/10.34117/bjdv7n7-124

Yoshitomi T, Sha S, Vong LB, et al. Indomethacin-loaded redox nanoparticles improve oral bioavailability of indomethacin and suppress its small intestinal inflammation. Ther Deliv. 2014;5(1):29-38. doi: https://doi.org/10.4155/tde.13.133

Riasat R, Guangjun N, Riasat Z, et al. Effects of nanoparticles on gastrointestinal disorders and therapy. J Clin Toxicol. 2016;6(4):1000313. doi: https://doi.org/10.4172/2161-0495.1000313

Sukul A, Das SC, Saha JK, et al. Screening of analgesic, antimicrobial, cytotoxic and antioxidant activities of metal complexes of indomethacin. J Pharm Sci. 2014;13(2):175-80. doi: https://doi.org/10.3329/dujps.v13i2.21895

Pohlmann AR, Weiss V, Mertins O, et al. Spray-dried indomethacin-loaded polyester nanocapsules and nanospheres: development, stability evaluation and nanostructure models. Eur J Pharm Sci. 2002;16(4-5):305-12. doi: https://doi.org/10.1016/s0928-0987(02)00127-6

Bernardi A, Braganhol E, Jäger E, et al. Indomethacin-loaded nanocapsules treatment reduces in vivo glioblastoma growth in a rat glioma model. Cancer Lett. 2009;281(1):53-63. doi: https://doi.org/10.1016/j.canlet.2009.02.018

Esposti MD. Measuring mitochondrial reactive oxygen species. Methods. 2002;26(4):335-40. doi: https://doi.org/10.1016/S1046-2023(02)00039-7

Vizzotto BS, Dias RS, Iglesias BA, et al. DNA photocleavage and melanoma cells cytotoxicity induced by a meso-tetra-ruthenated porphyrin under visible light irradiation. J Photobiol B. 2020;209:111922. doi: https://doi.org/10.1016/j.jphotobiol.2020.111922

Choi WS, Shin PG, Lee JH, et al. The regulatory effect of veratric acid on NO production in LPS-stimulated RAW264.7 macrophage cells. Cell Immunol. 2012;280(2):164-70. doi: https://doi.org/10.1016/j.cellimm.2012.12.007

International Conference on Harmonization. Validation of analytical procedures: text and methodology Q2(R1) [Internet]. Current Step 4 version. Genebra: ICH; 2005 [cited 2021 Dec 14]. Available from: https://www.gmp-compliance.org/files/guidemgr/Q2(R1).pdf

Iavicoli I, Fontana L, Leso V, et al. Hormetic dose-responses in nanotechnology studies. Sci Total Environ. 2014;487:361-74. doi: https://doi.org/10.1016/j.scitotenv.2014.04.023

Remant-Bahadur KC, Thapa B, Xu P. pH and redox dual responsive nanoparticle for nuclear targeted drug delivery. Mol Pharm. 2012;9(9):2719-29. doi: https://doi.org/10.1021/mp300274g

Rota C, Chignell CF, Mason RP. Evidence for free radical formation during the oxidation of 2’-7’-dichlorofluorescin to the fluorescent dye 2’-7’-dichlorofluorescein by horseradish peroxidase: possible implications for oxidative stress measurements. Free Radic Biol Med. 1999;27(7-8):873-81. doi: https://doi.org/10.1016/s0891-5849(99)00137-9

Zhang X, Lin Y, Gillies RJ. Tumor pH and its measurement. J Nucl Med. 2010;51(8):1167-70. doi: https://doi.org/10.2967/jnumed.109.068981

Mahalingaiah PKS, Singh KP. Chronic oxidative stress increases growth and tumorigenic potential of MCF-7 breast cancer cells. PLoS ONE. 2014;9(1):e87371. doi: https://doi.org/10.1371/journal.pone.0087371

Bryan NS, Grisham MB. Methods to detect nitric oxide and its metabolites in biological samples. Free Radic Biol Med. 2007;43(5):645-57. doi: https://doi.org/10.1016/j.freeradbiomed.2007.04.026

Sreenivasulua R, Reddya KT, Sujithab P, et al. Synthesis, antiproliferative and apoptosis induction potential activities of novel bis(indolyl)hydrazide-hydrazone derivatives. Bioorg Med Chem. 2019;27(6):1043-55. doi: https://doi.org/10.1016/j.bmc.2019.02.002

Tor YS, Yazan LS, Foo JB, et al. Induction of apoptosis in MCF-7 cells via oxidative stress generation, mitochondria-dependent and caspase-independent pathway by ethyl acetate extract of Dillenia suffruticosa and Its chemical profile. PLoS ONE. 2015;10(6):e0127441. doi: https://doi.org/10.1371/journal.pone.0127441

Szwed M, Torgersen ML, Kumari RV, et al. Biological response and cytotoxicity induced by lipid nanocapsules. J Nanobiotechnology. 2020;18(1):5. doi: https://doi.org/10.1186/s12951-019-0567-y

Park HB, Kim YJ, Lee SM, et al. Dual drug-loaded liposomes for synergistic efficacy in MCF-7 breast cancer cells and cancer stem cells. Biomed Sci Letters. 2019;25(2):159-69. doi: https://doi.org/10.15616/BSL.2019.25.2.159

Dror Y, Sorkin R, Brand G, et al. The effect of the serum corona on interactions between a single nano-object and a living cell. Sci Rep. 2017;7:45758. doi: https://doi.org/10.1038/srep45758

Westmeier D, Chen C, Stauber RH, et al. The bio-corona and its impact on nanomaterial toxicity. Eur J Nanomed. 2015;7(3):153-68. doi: https://doi.org/10.1515/ejnm-2015-0018

Barbosa KBF, Costa NMB, Alfenas RCG, et al. Estresse oxidativo: conceito, implicações e fatores modulatórios. Rev Nutr. 2010;23(4):629-43. doi: https://doi.org/10.1590/S1415-52732010000400013

Santos MC. Estudo do efeito do potencial de superfície na internalização de nanopartículas de magnetita em células cultivadas [trabalho de conclusão de curso na Internet]. Goiás: Universidade Federal de Goiás; 2007 [acesso 2021 dez 14]. Disponível em: https://www.prpg.ufg.br/up/85/o/modelo1.pdf

Froder JG, Dupeyrón D, Carvalho JCT, et al. In vitro study of the cytotoxic and genotoxic effects of indomethacin-loaded Eudragit(®) L 100 nanocapsules. Genet Mol Res. 2016;15(3). doi: https://doi.org/10.4238/gmr.15038727

Husain MA, Ishqi HM, Sarwar T, et al. Interaction of indomethacin with calf thymus DNA: a multi-spectroscopic, thermodynamic and molecular modelling approach. Medchemcomm. 2017;8(6):1283-96. doi: https://doi.org/10.1039/c7md00094d

Nagai N, Yoshioka C, Yoshimasa I. Topical therapies for rheumatoid arthritis by gel ointments containing indomethacin nanoparticles in adjuvant-induced arthritis rat. J Oleo Sci. 2015;64(3):337-46. doi: https://doi.org/10.5650/jos.ess14170

Downloads

Published

2022-09-13

How to Cite

1.
Franco C, Viana AR, Ourique AF, Vizzotto BS, Krause LMF. Protective Effect of Indomethacin-loaded Polymeric Nanoparticles Against Oxidative Stress-Induced Cytotoxicity in Human Breast Adenocarcinoma Cell Model. Rev. Bras. Cancerol. [Internet]. 2022 Sep. 13 [cited 2024 Jul. 22];68(4):e-012545. Available from: https://rbc.inca.gov.br/index.php/revista/article/view/2545

Issue

Section

ORIGINAL ARTICLE