Acessibilidade / Reportar erro

Cytotoxic Effects of Diclofenac and Ibuprofen Zinc (II)-Nicotinamide Ternary Complexes in Breast Cancer Cell Lines

Abstract

Breast cancer is one of the leading types of cancer worldwide, and the search for new treatment options are crucial. Nonsteroidal anti-inflammatory drugs (NSAIDs) -specially ibuprofen and diclofenac-, have shown antitumoral effect against several types of cancer. The synthesis of organometallic compounds has shown significant improvements in pharmacological properties and efficacy of organic molecules. Two zinc II ternary complexes containing the NSAIDs diclofenac and ibuprofen and nicotinamide neutral linker (Nic) were obtained by the two-step solvent metalligand complexation method. The compounds Zn2(Diclof)4(Nic)2 (complex 1) and Zn2(Ibup)4(Nic)2 (complex 2) were tested in breast cancer cell lines (4T1, MCF-7 and MDA-MB-231) to evaluate their cytotoxicity, comparing to ibuprofen and diclofenac as controls. We found that both complex 1 and 2 exerted more than 60% reduction in 4T1 viability at 250µM, and complex 2 decreased cell viability at 250 µM and 137.5 µM in MCF-7 (34.35% and 26.42% reduction, respectively) and in MDA-MB-231 (57.2% and 22.88% reduction, respectively), all compared to controls. Complex 1 was selective only in MCF-7, and complex 2 was selective in both MCF-7 and MDA-MB-231. In summary, our data showed that the cytotoxic effect of complex 1 and 2 is increased comparing to their original NSAID in different breast cancer cell lines, highlighting their potential anti-tumoral activity.

Keywords:
NSAIDs; Zinc complexes; cytotoxicity; coordination compounds; cell viability

HIGHLIGHTS

Diclofenac and ibuprofen-derived complexes decrease viability of breast cancer cell lines

Complex 1 presented antitumor effect in all breast cancer cell lines tested including on triple negative cell line (MDA-MB-231) (IC50 202µM)

Complex 1 was selective for both 4T1 and MDA-MB-231 cells

INTRODUCTION

Cancer is the second most cause of death worldwide, with 9.6 million deaths estimated in 2018. (11 Christopher PW, Bernard WS, Elisabeth W. World Cancer Report 2020. Lyon: WHO International agency for Research on cancer; 2020 p. 613) According to Global Cancer Statistics 2018 (22 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J. for Clin. 2018;68(6):394-424.), for females, breast cancer is the leading type of cancer, with 24.2% of incidence, and leading cause of cancer death, with 15.0% mortality rate. Breast cancer involves an inherited component, with multiple susceptibility genes linked to it, and has a wide variation in tumor morphology and clinical response. Like all cancer types, it implicates in high costs treatments, with several side effects, therefore the search for more specific and efficient compounds against breast cancer is crucial. (22 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J. for Clin. 2018;68(6):394-424.)

Nonsteroidal anti-inflammatory drugs (NSAIDs) are a class of molecules that act on inflammation, vasodilatation, vasoconstriction, and as analgesics and antipyretics. Their mechanism of action is based on the inhibition of COX and LOX enzymes, which are involved in the biosynthesis of prostaglandins (PGs) (33 Boodram JN, Mcgregor IJ, Bruno PM, Cressey PB, Hemann MT, Suntharalingam K. Breast Cancer Stem Cell Potent Copper(II)-Non-Steroidal Anti-Inflammatory Drug Complexes. Angew. Chem. - Int. Ed. 2016;55(8):2845-50.,44 Feng J, Du X, Liu H, Sui X, Zhang C, Tang Y, et al. Manganese-mefenamic acid complexes exhibit high lipoxygenase inhibitory activity. Dalton Transactions. 2014;43(28):10930-9.). Arachidonic acid (AA) is converted by COX and LOX enzymes into lipid mediators known as eicosanoids, which include prostaglandins, thromboxanes, leukotrienes, lipoxins, resolvins, and eoxins, and are involved in inflammation and cancer (55 Ricciotti E, Fitzgerald GA. Prostaglandins and inflammation. Arterioscler, Thromb, and Vasc Biol. 2011;31(5):986-1000.

6 Kobayashi K, Omori K, Murata T. Role of prostaglandins in tumor microenvironment. Cancer Metastasis Rev. 2018;37(2-3):347-54.
-77 Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: Structural, Cellular, and Molecular Biology. Annu. Rev. of Biochem. 2000 Jun;69:145-82.). There have been several studies showing an inverse correlation between the use of NSAIDs and the incidence of several types of cancer, such as breast (88 Sharpe CR, Collet J-P, McNutt M, Belzile E, Boivin J-F, Hanley JA. Nested case-control study of the effects of non-steroidal anti-inflammatory drugs on breast cancer risk and stage. Br J Cancer. 2000 Jul;83(1). 112-20.,99 Moris D, Kontos M, Spartalis E, Fentiman IS. The Role of NSAIDs in Breast Cancer Prevention and Relapse: Current Evidence and Future Perspectives. Breast Care. 2016;11(5). 339-344.), lung (1010 Bittoni MA, Carbone DP, Harris RE. Ibuprofen and fatal lung cancer: A brief report of the prospective results from the Third National Health and Nutrition Examination Survey (NHANES III). Mol. and Clin. Oncol. [Internet]. 2017;6(6):917-20. Available from: https://www.spandidos-publications.com/
https://www.spandidos-publications.com...
,1111 Muscat JE, Chen S-Q, Richie JP, Altorki NK, Citron M, Olson S, et al. Risk of lung carcinoma among users of nonsteroidal antiinflammatory drugs. Cancer. 2003 Apr 1;97(7). 1732-6.), prostate (1212 Doat S, Cénée S, Trétarre B, Rebillard X, Lamy P, Bringer J, et al. Nonsteroidal anti-inflammatory drugs (NSAIDs) and prostate cancer risk: results from the EPICAP study. Cancer Med. 2017;6(10):2461-70.), bladder (1313 Daugherty SE, Pfeiffer RM, Sigurdson AJ, Hayes RB, Leitzmann M, Schatzkin A, et al. Nonsteroidal antiinflammatory drugs and bladder cancer: A pooled analysis. Am. J. of Epidemiol. 2011;173(7):721-30.), ovary (1414 Wernli KJ, Newcomb PA, Hampton JM, Trentham-Dietz A, Egan KM. Inverse association of NSAID use and ovarian cancer in relation to oral contraceptive use and parity. Br J Cancer. 2008;98(11):1781-3.,1515 Baandrup L, Faber MT, Christensen J, Jensen A, Andersen KK, Friis S, et al. Nonsteroidal anti-inflammatory drugs and risk of ovarian cancer: Systematic review and meta-analysis of observational studies. Acta Obstet. Gynecol. Scand. 2013;92:245-55.), esophagus (1616 Corley D, Kerlikowske K, Verma R, Buffler P. Protective association of aspirin/NSAIDs and esophageal cancer: A systematic review and meta-analysis. Gastroenterology. 2003;124(1):47-56.) and stomach (1717 Huang X, Chen Y, Wu J, Zhang X, Wu C, Zhang C, et al. Aspirin and non-steroidal anti-inflammatory drugs use reduce gastric cancer risk: A dose-response meta-analysis. Oncotarget. 2017;8(3):4781-95.). These correlations have prompted studies on antitumoral effects of NSAIDs. (1818 Dell'Omo G, Crescenti D, Vantaggiato C, Parravicini C, Borroni AP, Rizzi N, et al. Inhibition of SIRT1 deacetylase and p53 activation uncouples the anti-inflammatory and chemopreventive actions of NSAIDs. Br J Cancer. 2019;120:537-46.)

Chronic inflammation can lead to the initiation of cancer (1919 Grivennikov SI, Karin M. Inflammation and oncogenesis: a vicious connection. Vol. 20, Curr. Opin. Genet. and Dev. 2010. p. 65-71.

20 Grivennikov SI, Greten FR, Karin M. Immunity, Inflammation, and Cancer. Cell. 2010.19, 140(6)883-99.
-2121 Bartsch H, Nair J. Chronic inflammation and oxidative stress in the genesis and perpetuation of cancer: Role of lipid peroxidation, DNA damage, and repair. Langenbeck's Arch. Surg. 2006;391:499-510.), and COX enzymes (specially COX-2 (2222 Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: A review. J. Cell. Physiol. 2019.234:5683-99.)) are overexpressed in many malignant lesions (2323 Ohtsuka J, Oshima H, Ezawa I, Abe R, Oshima M, Ohki R. Functional loss of p53 cooperates with the in vivo microenvironment to promote malignant progression of gastric cancers. Sci Rep. 2018;8(1):2291.), such as colon (2424 Tuncer S, Banerjee S. Eicosanoid pathway in colorectal cancer: Recent updates. World J. of Gastroenterol. 2015;7(21):11746-66.), stomach (2525 Echizen K, Hirose O, Maeda Y, Oshima M. Inflammation in gastric cancer: Interplay of the COX-2/prostaglandin E 2 and Toll-like receptor/MyD88 pathways. Cancer Sci. 2016 Apr 18;107(4). 391-7.) and breast cancer (2626 Howe LR. Inflammation and breast cancer. Cyclooxygenase/prostaglandin signaling and breast cancer. Breast Cancer Res. 2007;9(4):210.). This enzyme is also involved in cancer development (2727 Dong XF, Liu TQ, Zhi XT, Zou J, Zhong JT, Li T, et al. COX-2/PGE2 axis regulates HIF2a activity to promote hepatocellular carcinoma hypoxic response and reduce the sensitivity of sorafenib treatment. Clin. Cancer Res. 2018;24(13):3204-16.), poorer patient prognosis (2828 Esbona K, Yi Y, Saha S, Yu M, van Doorn RR, Conklin MW, et al. The Presence of Cyclooxygenase 2, Tumor-Associated Macrophages, and Collagen Alignment as Prognostic Markers for Invasive Breast Carcinoma Patients. Am. J. of Pathol. 2018;188(3):559-73.) and tumor invasion (2929 Greenhough A, Smartt HJM, Moore AE, Roberts HR, Williams AC, Paraskeva C, et al. The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis. 2009 Jan 9;30(3): 377-86). Thus, NSAIDs like ibuprofen and diclofenac can have an antitumoral effect on cancer, as they inhibit both COX-1 and COX-2 enzymes (3030 Bacchi S, Palumbo P, Sponta A, Coppolino MF. Clinical Pharmacology of Non-Steroidal Anti-Inflammatory Drugs : A Review. Antiinflamm Antiallergy Agents Med Chem. 2012;11(1):52-64.).

Organometallic complexes are formed when a metal coordinates with organic compounds, being considered as an intermediate form between organic and inorganic derivatives. Studies have shown that these complexes can be potential anticancer drugs (3131 Smith GS, Therrien B. Targeted and multifunctional arene ruthenium chemotherapeutics. Dalton Trans. 2011;40:10793-800.

32 Zaki M, Hairat S, Aazam ES. Scope of organometallic compounds based on transition metal-arene systems as anticancer agents: Starting from the classical paradigm to targeting multiple strategies. RSC Advances. 2019;9:3239-78.

33 Szczepaniak A, Fichna J. Organometallic compounds and metal complexes in current and future treatments of inflammatory bowel disease and colorectal cancer-a critical review. Biomolecules. 2019;22(9):398.

34 Ronconi L, Sadler PJ. Using coordination chemistry to design new medicines. Coord. Chem. Rev. 2007; 251:1633-48.
-3535 Gianferrara T, Bratsos I, Alessio E. A categorization of metal anticancer compounds based on their mode of action. Dalton Trans. 2009;(37): 7588-98.), since they exhibit a greater effect than the original organic molecule, and higher kinetic stability, structural diversity, ability to bind biological targets, variable oxidation states and the possibility of rational ligand design to control kinetic properties (3636 Allgeier AM, Mirkin CA. Ligand Design for Electrochemically Controlling Stoichiometric and Catalytic Reactivity of Transition Metals. Angew. Chem. Int. Ed. 1998 Apr 20;37(7):894-908.,3737 Gasser G, Metzler-Nolte N. The potential of organometallic complexes in medicinal chemistry. Curr. Opin. Chem. Biol. 2012 Apr;16(1-2):84-91.). Metallodrugs formed using ibuprofen and diclofenac as chelating agents have shown anticancer activity against cancer cell lines (3838 Mandal P, Kundu BK, Vyas K, Sabu V, Helen A, Dhankhar SS, et al. Ruthenium (II) arene NSAID complexes: Inhibition of cyclooxygenase and antiproliferative activity against cancer cell lines. Dalton Trans. 2018;47(2):517-27.

39 Alves Rico SR, Abbasi AZ, Ribeiro G, Ahmed T, Wu XY, de Oliveira Silva D. Diruthenium(II,III) metallodrugs of ibuprofen and naproxen encapsulated in intravenously injectable polymer-lipid nanoparticles exhibit enhanced activity against breast and prostate cancer cells. Nanoscale. 2017;9(30):10701-14.

40 Hanif-Ur-Rehman, Freitas TE, Gomes RN, Colquhoun A, de Oliveira Silva D. Axially-modified paddlewheel diruthenium(II,III)-ibuprofenato metallodrugs and the influence of the structural modification on U87MG and A172 human glioma cell proliferation, apoptosis, mitosis and migration. J. Inorg. Biochem. [Internet]. 2016;165:181-91. Available from: http://dx.doi.org/10.1016/j.jinorgbio.2016.10.003
http://dx.doi.org/10.1016/j.jinorgbio.20...
-4141 Intini FP, Zajac J, Novohradsky V, Saltarella T, Pacifico C, Brabec V, et al. Novel Antitumor Platinum (II) Conjugates Containing the Nonsteroidal Anti-inflammatory Agent Diclofenac: Synthesis and Dual Mechanisms of Antiproliferative Effects. Inorg. Chem. 2017;56(3):1483-97.). Studies using metallodrugs containing ibuprofen showed antiproliferative activity in vitro, and compounds using both diclofenac and ibuprofen also led to inhibition of COX and LOX enzymes (3838 Mandal P, Kundu BK, Vyas K, Sabu V, Helen A, Dhankhar SS, et al. Ruthenium (II) arene NSAID complexes: Inhibition of cyclooxygenase and antiproliferative activity against cancer cell lines. Dalton Trans. 2018;47(2):517-27.,3939 Alves Rico SR, Abbasi AZ, Ribeiro G, Ahmed T, Wu XY, de Oliveira Silva D. Diruthenium(II,III) metallodrugs of ibuprofen and naproxen encapsulated in intravenously injectable polymer-lipid nanoparticles exhibit enhanced activity against breast and prostate cancer cells. Nanoscale. 2017;9(30):10701-14.).

Based on these studies, our group sought to examine the effect of two organometallic compounds, complex 1 and complex 2, synthesized and previously described by Moura 2020 (4242 Dos Santos PR, Pich CT, Back D, Smiderle F, Dumas F, Moura S. Synthesis, chemical characterization and DNA interaction study of new diclofenac and ibuprofen zinc (II)-nicotinamide ternary complexes as cyclooxygenase inhibitor prototypes. J. Inorg. Biochem. [Internet]. 2020;206(February):111046. Available from: https://doi.org/10.1016/j.jinorgbio.2020.111046
https://doi.org/10.1016/j.jinorgbio.2020...
). The compounds were synthesized using ibuprofen and diclofenac, forming ternary complexes of Zn-Diclof and Zn-Ibup with nicotinamide (Nic) as a nitrogen ligand. Complexes 1 and 2 were tested against three breast cancer (BC) cell lines: MCF-7, a human invasive breast ductal carcinoma, ER+, PR+/-, HER2- luminal and often chemotherapy responsive; MDA-MB-231, also human invasive breast ductal carcinoma, triple negative, claudin-low and intermediate responsive to chemotherapy; and 4T1, a mouse mammary gland that mimics an animal stage IV human breast cancer (4343 Pulaski BA, Ostrand-Rosenberg S. Mouse 4T1 Breast Tumor Model. Curr. Protoc. in Immunol. 2000 Oct;39(1).,4444 Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Research. 2011 Aug 12;13(4). 215.). We aimed to observe the effect of complexes 1 and 2 in inhibiting proliferation of breast cancer cell lines.

MATERIAL AND METHODS

Zinc nitrate, zinc sulphate, nicotinamide, sodium carbonate and ethanol 99% were purchased from Sigma-Aldrich (St. Louis, Missouri, USA). Potassium diclofenac 99% and ibuprofen 99% (pharmaceutical grade) were obtained from Sigma-Aldrich. Ultrapure water was obtained from the Milli-Q (Millipore) apparatus. Ethanol and dimethylformamide (DMF) were purchased form Sigma-Aldrich (grade reagent) and used without further purification.

Physical measurements

1H and 13C NMR analysis was performed on a Fourier 300 spectrometer (Bruker) (300.18 MHz from 1H and 75.49 MHz from 13C with 5 mm probe) with 32 scans for 1H and 5000 scans for 13C, using CDCl3 and DMSO-D6 as solvents. Infrared analysis was performed on a Perkin Elmer Spectrum 400 FTIR spectrometer using solid samples by the KBr pellet technique. Spectral data were obtained in the spectral interval from 400 to 4000 cm-1 (wavenumber) with a resolution of 1.0 cm-1 for 32 scans. High resolution mass spectroscopy was performed according to the following conditions: complex solutions (1.0 μg.mL-1 H2O:MeOH 1:1 v/v) were individually infused directly into the ESI source by means of a syringe pump (Harvard Apparatus) at a flow rate of 150 μL.min-1. ESI (+)-MS and tandem ESI (+)-MS/MS were acquired using a hybrid high-resolution and high accuracy (5 μL.min-1) MicroTof (Q-TOF) mass spectrometer (Bruker Scientific) under the following conditions: capillary and cone voltages were set to +3500 V and +40 V, respectively, with a desolvation temperature of 100 °C. For ESI (+)-MS/MS, the energy for the collision-induced dissociations (CIDs) was optimized for each component. For data acquisition and processing in Q-TOF, control data analysis software (Bruker Scientific) was used. The data were collected in the m/z range of 100-2000 at the speed of two scans/s, providing the resolution of 10,000 (FWHM) at m/z 200.

Chemical synthesis

Figure 1
Ternary complexes 1 and 2.

Complexes 1 and 2 were prepared and analyzed according to Dos Santos and coauthors (4242 Dos Santos PR, Pich CT, Back D, Smiderle F, Dumas F, Moura S. Synthesis, chemical characterization and DNA interaction study of new diclofenac and ibuprofen zinc (II)-nicotinamide ternary complexes as cyclooxygenase inhibitor prototypes. J. Inorg. Biochem. [Internet]. 2020;206(February):111046. Available from: https://doi.org/10.1016/j.jinorgbio.2020.111046
https://doi.org/10.1016/j.jinorgbio.2020...
). For complex 1, 0.655 g (1.0 mmol) of zinc diclofenac salt previously obtained according to Abu Ali and coauthors (4545 Abu Ali H, Jabali B. Synthesis, characterization and biological activity of novel complexes of zinc(II) diclofenac with nitrogen based ligands. Polyhedron. 2016;107:97-106.)was dissolved in 20 mL of ethanol 99% at room temperature and vigorous stir. An ethanolic solution of nicotinamide (0.244 g, 2.0 mmol) was then added dropwise over first solution and keep stirring for two h. The white powder was obtained after one week by filtration and vacuum drier. Yield: 0.70 g, (90%); MP: 180° C; IR (cm-1, KBr pellet): 3310, 3183, 3095, 3072, 3039, 2969, 2925, 1679, 1622, 1607, 1592, 1576, 1564 (νa COO-), 1506, 1415 (νs COO-), 1351, 1303, 1283, 1249, 1199, 1164, 1094, 1058, 869, 837, 774, 747, 720, 698, 656; (Δν COO-) 149; 1HNMR (δ-ppm, DMSO-D6): 3.59 (s-4H, 2CH2), 6.28 (d-2H, 2CH, JH-H = 9Hz), 6.82 (dt-2H, 2CH, JH-H = 7Hz), 7.01 (dt-2H, 2CH, JH-H = 7Hz), 7.08 (dd-2H, 2CH, JH-H = 8Hz), 7.15 (dd-2H, 2CH, JH-H = 7Hz), 7.42 (d-4H, 4CH, JH-H = 8Hz), 7.48 (m-1H, CH(Nic), JH-H = 5Hz), 7.63 (s-1H, NH(Nic)), 8.19 (s-1H, NH(Nic)), 8.21 (m-1H, CH, JH-H = 8Hz), 8.30 (s-2H, 2NH(Diclof)), 8.69 (dd-1H, CH(Nic), JH-H = 5Hz), 9.03 (ds-1H(Nic), CH); 13CNMR (δ?ppm, DMSO-D6): 40.73 (2CH2), 116.34 (2CH), 120.82 (2CH), 123.60 (CH(Nic)), 124.59 (2CH), 126.56 (2CH), 126.85 (2C), 128.97 (2CH), 129.12 (4CH), 129.82 (CH(Nic)), 130.61 (2C), 135.46 (CH(Nic)), 137.57 (2C), 142.86 (4CCl), 148.75 (CH(Nic)), 151.92 (CH(Nic)), 166.45 (CON(Nic)), 177.40 (2COO-(Diclof)); HRMS ESI (+) m/z 1253.9743 (C54H42Cl6N7O8Zn2) [M-diclof]+, 1131,9225 (C48H36Cl6N5O7Zn2) [M-diclof-nic]+, 1009.8843 (C42H30Cl6N3O6Zn2) [Zn2(diclof)3]+, 796.9771 (C34H26Cl4N4NaO5Zn) [Zn(diclof)2Nic+Na]+, 774.9993 (C34H27Cl4N4O5Zn) [Zn(diclof)2Nic+H]+, 674.9318 (C28H20Cl4N2NaO4Zn) [Zn(diclof)2+Na]+, 652.9525 (C28H21Cl4N2O4Zn) [Zn(diclof)2+H]+, 602.0314 (C26H22Cl2N5O4Zn) [Zn(diclof)(nic)2]+, 478.9849 (C20H16Cl2N3O3Zn) [Zn(diclof)(nic)]+, 398.9627 (C14H14Cl2N2NaO2Zn) [Zn(diclof)+NH4+Na]+, 374.9478 (C14H12Cl2NO3Zn) [Zn(diclof)+H2O]+, 357.9379 (C14H10Cl2NO2Zn) [Zn(diclof)]+, 318.0053 (C14H11Cl2NNaO2) [diclof+H+Na]+, 296.0233 (C14H12Cl2NO2) [diclof+2H]+.

For complex 2, 0.475 g (1.0 mmol) of zinc ibuprofen salt previously obtained according to Abu Ali and coauthors (46) was dissolved in 20 mL of dimethylformamide (DMF) at room temperature and vigorous stir. Nicotinamide (0.244 g, 2.0 mmol) was then added dropwise over first solution and keep stirring for two h. The clear solution was concentered by vacuum evaporation at 20% of start volume and keeped in dark flask at room temperature for 30 days. The clear crystals were then obtained by filtration, washed with cold DMF and dried over freeze drier by 24 h. Yield: 0.42g (70%); MP:148 °C; IR (cm-1, KBr pellet): 3440, 3318, 3283, 3201, 3182, 2959, 2930, 2867, 1689, 1636 (νa COO-), 1604, 1575, 1512, 1458 (νs COO-), 1412, 1372, 1287, 1199, 1056, 793, 699, 601, (Δν COO-) 178; 1HNMR (δ-ppm, CDCl3): 0.82 (d-12H, 4CH3, JH-H = 6Hz), 1.35 (d-6H, 2CH3, JH-H = 7Hz), 1.74 (sep-2H, 2CH, JH-H = 7Hz), 2.27 (d-4H, 2CH2, JH-H = 7Hz), 3.62 (q-2H, 2CH, JH-H = 7), 4,98 (s-1H, 1NH(Nic)), 6.83 (d-4H, 4CH, JH-H = 8Hz), 7,00 (d-4H, 4CH, JH-H = 8Hz), 7.43 (dd-1H, 1CH(Nic), JH-H = 8Hz), 8,26 (s-1H, 1NH(Nic)), 8.43 (dt-2H, 2CH(Nic), JH-H = 8Hz), 8.59 (dd-2H, 2CH(Nic),JH-H = 5Hz), 8.85 (ds-2H, 2CH(Nic), JH-H = 2Hz); 13CNMR (δ-ppm, CDCl3): 19.54 (2CH3), 22.33 (4CH3), 30.09 (2CH), 44.86 (2CH), 46.24 (2CH2), 124.57 (CH(Nic)), 127.09 (4CH), 129.07 (4CH), 130.70 (CH(Nic)), 137.01 (2C), 139.76 (CH(Nic)), 140.06 (2C), 146.83 (CH(Nic)), 150.26 (C(Nic)), 164.39 (CON(Nic))183.07 (2CO2-); HRMS ESI (+): m/z 1093.3807 (C58H74N2NaO9Zn2) [M-Nic+Na]+, 1071.3983 (C58H75N2O9Zn2) [M-Nic+H]+, 987.3219 (C51H63N4O8Zn2) [M-ibup]+, 865.2731 (C45H57N2O7Zn2) [Zn2(ibup)3(Nic)]+, 883.2853 (C45H59N2O8Zn2) [Zn2(ibup)3(Nic)+H2O]+, 619.2110 (C32H40N2NaO5Zn) [Zn(ibup)2(Nic)+Na]+, 597.2296 (C32H41N2O5Zn) [Zn(ibup)2(Nic)+H]+, 513.1469 (C25H29N4O4Zn) [Zn(ibup)(Nic)2]+, 475.1813 (C26H35O4Zn) [Zn(ibup)2+H]+, 328.0889 (C13H23NNaO3Zn) [Zn(ibup)+H2O+NH4+Na]+, 287.0620 (C13H19O3Zn) [Zn(ibup)+H2O]+, 229.1201 (C13H18NaO2) [ibup+Na+H]+.

Chemical synthesis

The compounds were synthesized according to Moura 2020 [42].

Cell lines

The cell lines MCF-7 (human mammary gland adenocarcinoma), 4T1 (Mus musculus mammary gland), MDA-MB-231 (human mammary gland adenocarcinoma) and Vero (Cercopithecus aethiops kidney normal) were purchased from the American Type Culture Collection (ATCC). MCF-7 cells were cultured in DMEM Low glucose, MDA-MB-231 and Vero in DMEM High glucose, and 4T1 in RPMI, all supplemented with 10% Fetal bovine serum (FBS). All cells were incubated in a humidified incubator at 5% CO2 and 37 °C, and experiments were performed using freshly thawed cells after three passages. Cell lines were tested for Mycoplasma contamination.

Cytotoxicity assay

Cells were seeded in 96-well flat-bottom plates at concentrations of 5x103 cells/well for MCF-7, MDA-MB-231 and 4T1 cells and 2x103 cells/well for Vero cells. After 24 h of incubation, 250 μM, 137.5 μM, 25 μM, 13.75 μM, or 2.5 μM of complex 1 and complex 2 was added and the plates were incubated for an additional 24 h at 37°C in a humidified incubator with 5% CO2. For the treatment curve, 4T1 cells were incubated with 250 µM of compounds and controls for 6h, 12h, 24h and 48h. For the vehicle control, DMSO was added in the highest concentration that was used to dilute the compounds. As an additional negative control (NC), untreated cells were also assayed. Cell viability was assessed using a colorimetric assay based on the reduction of 3-[4,5-dimethylthiazol-2-y1]-2,-diphenyltetrazolium bromide (MTT) by mitochondrial enzymes (Molecular Probes™, Thermo Fisher Scientific, Waltham, MA, USA). Briefly, 100μL of medium was removed and 40μL of MTT reagent (5-mg/mL) was added into each well. Cells were incubated for 4h and the precipitated formazan crystals were dissolved in dimethyl sulfoxide (DMSO). Finally, the optic density (OD) was analyzed at 570/620 nm using a micro-plate reader (EZ Read 400, Biochrom). Assays were performed in triplicate. For cytotoxicity calculation, the OD of the treated cells was multiplied by the percentage equivalent to the cells treated with DMSO, and then divided by the OD of the cells treated with DMSO, as the following:

OD of NC - 100%

OD of DMSO treated cells - x

Viability = (OD of treated cells x % equivalent to DMSO) / (OD of DMSO treated cells)

Data analysis

The half maximal inhibitory concentrations (IC50) of compounds and controls were calculated using linear and polynomial regression analyses using Microsoft Excel 2009, with consequent resolution of the line equation of the graph with Wolfram Alpha. The IC50 values were reported as a mean of two independent experiments. The selectivity index (SI) was calculated according to the following equation:

S I = ( I C 50 o f V e r o c e l l s ) / ( I C 50 o f t u m o r c e l l s )

where a SI >1 indicates that the compound is more cytotoxic to tumor cell, and a SI<1 indicates the opposite (4545 Abu Ali H, Jabali B. Synthesis, characterization and biological activity of novel complexes of zinc(II) diclofenac with nitrogen based ligands. Polyhedron. 2016;107:97-106.,4646 Abu Ali H, Omar SN, Darawsheh MD, Fares H. Synthesis, characterization and antimicrobial activity of zinc(II) ibuprofen complexes with nitrogen-based ligands. J. Coord. Chem. 2016;69(6):1110-22.).

Statistical analyses were performed with GraphPad Prism 5 (GraphPad Software Inc., CA) using two-way ANOVA followed by Bonferroni correction.

RESULTS

Cytotoxic effects

Cytotoxic effects of complex 1 and complex 2 were tested against breast cancer cell lines MCF-7, MDA-MB-231 and 4T1, and Vero cells were used as non-tumorigenic cell line. Different concentrations were tested (250 µM, 137.5 µM, 25 µM, 13.75 µM and 2.5 µM), and ibuprofen and diclofenac were used in the same concentrations in cell lines as a control.

Both complex 1 and complex 2 exhibited more than 60% reduction in 4T1 cell viability at the concentrations of 250 and 137.5 µM, when compared to controls (Figure 2A and 2E). Complex 1 also decreased viability at 25 µM (22.88% reduction). Only complex 1 decreased cell viability at the concentrations of 250 µM and 137.5 µM in MCF-7 (34.35% and 26.42% reduction, respectively) (Figure 2B) and in MDA-MB-231 (57.2% and 22.88% reduction, respectively) (Figure 2C). Complex 2 showed no significant reduction in MCF-7 and MDA-MB-231, compared to ibuprofen (Figure 2F and 2G). Using non-tumorigenic cell lines (Vero), we observe that both compounds decreased viability at 250 µM (29.96% for complex 1 and 29.97% for complex 2), and at 137,5 µM, only complex 1 caused reduction (30.58%), showing a cytotoxic and non-selective effect (Figure 2D and 2H).

Figure 2
Cytotoxicity of complex 1, complex 2 and controls (ibuprofen and diclofenac). (A) Viability of 4T1 cells treated with complex 1 and diclofenac. (B) Viability of MCF-7 cells treated with complex 1 and diclofenac. (C) Viability of MDA-MB-231 cells treated with complex 1 and diclofenac. (D) Viability of Vero cells treated with complex 1 and diclofenac. (E) Viability of 4T1 cells treated with complex 2 and ibuprofen. (F) Viability of MCF-7 cells treated with complex 2 and ibuprofen. (G) Viability of MDA-MB-231 cells treated with complex 2 and ibuprofen. (H) Viability of Vero cells treated with complex 2 and ibuprofen. Cell viability was measured by MTT assay. Dose response curves represent viability 24h after treatment with 2.5 - 250 μM of each compound performed in triplicates. DMSO was used as a vehicle control. Values represent the mean and standard deviation of two independent experiments. * indicates p-values lower than 0.05, ** indicates p-values lower than 0.01 and *** indicates p-values lower than 0.001.

IC50 values

The IC50 values (the concentration of a drug that is required for 50% inhibition in vitro) were calculated for each complex and controls (Table 1). We found that for all cell lines tested, complex 1 exhibited the lowest IC50 compared to controls and complex 2 in all cell lines, indicating that it had the greatest cytotoxic effects. Although complex 1 was effective at inhibiting tumor cell lines growth, it also inhibited Vero cells. Complex 2 presented lower IC50 value for 4T1 cells comparing to the other cell lines, so 4T1 was the cell line that was more affected by the compounds’ activity. When controls were tested, diclofenac showed lower IC50 values than ibuprofen, because it is more cytotoxic.

Table 1
Cytotoxic activity (IC50 μM) of compounds (complex 1 and complex 2) and controls (ibuprofen and diclofenac).

Selectivity index

Selectivity index (SI) (Table 2) is a ratio that measures the window between cytotoxic and antitumoral activity. We used the IC50 found in Vero cells and divided by IC50 of tumoral cells. For both 4T1 and MDA-MB-231 cells, complex 1 was selective, as its SI was >1. Complex 2 presented selective effects only on 4T1 cells. For MCF-7, neither the compounds nor the controls exhibited a SI >1, showing that they are all not selective for this cell line. Diclofenac had a SI >1 for 4T1, and ibuprofen showed a SI <1 for all cell lines tested, meaning that it is not selective.

Table 2
Selectivity index (SI) of compounds (complex 1 and complex 2) and controls (ibuprofen and diclofenac).

Since the compounds exhibited the greatest effects on tumor cell growth of 4T1 cell line, we sought to examine the effect of the complexes at viability through different time points (6h, 12h, 24h and 48h) in order to observe when the cytotoxic effect starts (Figure 3). Both complex 1 and 2 had a time-dependent effect on cell viability, after 6h of the treatment we found a significant reduction on viability comparing to controls (Figure 3A and 3B). Diclofenac decreased around 30% cell viability after 48h, and in contrast to ibuprofen that did not affect cell viability.

Figure 3
Cytotoxicity of 4T1 cells after 6h, 12h, 24h and 48h of treatment with compounds and controls. (A) 4T1 cells treated with complex 1 and diclofenac. (B) Cells treated with complex 2 and ibuprofen. Cell viability was measured by MTT assay. Cells were treated with 250 µM of compounds and controls. Values represent the mean and standard deviation of three replicates of one experiment. * indicates p-values lower than 0.05, ** indicates p-values lower than 0.01 and *** indicates p-values lower than 0.001.

DISCUSSION

Breast cancer is one of the leading causes of death in the developed world, especially among women, implying in high costs treatments, which sometimes causes major side effects. For this reason, several studies have been performed to better understand the mechanisms of compounds candidates to be used as treatment of cancer.

Our study and others showed that organometallic compounds derived from ibuprofen and diclofenac presented inhibition of tumor cell growth in vitro (3838 Mandal P, Kundu BK, Vyas K, Sabu V, Helen A, Dhankhar SS, et al. Ruthenium (II) arene NSAID complexes: Inhibition of cyclooxygenase and antiproliferative activity against cancer cell lines. Dalton Trans. 2018;47(2):517-27.,3939 Alves Rico SR, Abbasi AZ, Ribeiro G, Ahmed T, Wu XY, de Oliveira Silva D. Diruthenium(II,III) metallodrugs of ibuprofen and naproxen encapsulated in intravenously injectable polymer-lipid nanoparticles exhibit enhanced activity against breast and prostate cancer cells. Nanoscale. 2017;9(30):10701-14.,4141 Intini FP, Zajac J, Novohradsky V, Saltarella T, Pacifico C, Brabec V, et al. Novel Antitumor Platinum (II) Conjugates Containing the Nonsteroidal Anti-inflammatory Agent Diclofenac: Synthesis and Dual Mechanisms of Antiproliferative Effects. Inorg. Chem. 2017;56(3):1483-97.). We found that complex 1, derived from diclofenac, and complex 2, derived from ibuprofen, decreased cell viability in breast cancer cell lines when compared to controls, especially after 48h of treatment, which may be associated with their high stability (4242 Dos Santos PR, Pich CT, Back D, Smiderle F, Dumas F, Moura S. Synthesis, chemical characterization and DNA interaction study of new diclofenac and ibuprofen zinc (II)-nicotinamide ternary complexes as cyclooxygenase inhibitor prototypes. J. Inorg. Biochem. [Internet]. 2020;206(February):111046. Available from: https://doi.org/10.1016/j.jinorgbio.2020.111046
https://doi.org/10.1016/j.jinorgbio.2020...
). Since the addition of metals to an organic molecule can augment its pharmacological properties, possibly the coordination to Zn+ caused this effect on these complexes. When compounds were tested against 4T1 cells in different time points, we observed that cytotoxicity effects started at 6h and increased over time, suggesting that the compounds might affect cell death. Furthermore, the presence of nicotinamide increases the solubility of compounds (4242 Dos Santos PR, Pich CT, Back D, Smiderle F, Dumas F, Moura S. Synthesis, chemical characterization and DNA interaction study of new diclofenac and ibuprofen zinc (II)-nicotinamide ternary complexes as cyclooxygenase inhibitor prototypes. J. Inorg. Biochem. [Internet]. 2020;206(February):111046. Available from: https://doi.org/10.1016/j.jinorgbio.2020.111046
https://doi.org/10.1016/j.jinorgbio.2020...
), which may be related to the cytotoxicity of both complexes (3838 Mandal P, Kundu BK, Vyas K, Sabu V, Helen A, Dhankhar SS, et al. Ruthenium (II) arene NSAID complexes: Inhibition of cyclooxygenase and antiproliferative activity against cancer cell lines. Dalton Trans. 2018;47(2):517-27.,4747 Quispe M A, Zavala C D, Rojas C J, Posso R M, Vaisberg W A. Efecto citotóxico selectivo in vitro de muricin H (acetogenina de Annona muricata) en cultivos celulares de cáncer de pulmón. Rev peru med exp salud publica. 2006;23(4):256-69.,4848 Benadiba M, dos Santos RRP, Silva D de O, Colquhoun A. Inhibition of C6 rat glioma proliferation by [Ru2Cl(Ibp)4] depends on changes in p21, p27, Bax/Bcl2 ratio and mitochondrial membrane potential. J. Inorg. Biochem. [Internet]. 2010;104(9):928-35. Available from: http://dx.doi.org/10.1016/j.jinorgbio.2010.04.011
http://dx.doi.org/10.1016/j.jinorgbio.20...
). It was previously demonstrated that the complexed form of the Zn II NSAIDs and the Nic binder produces molecules of low toxicity, no DNA cleavage activity (instead of its ability to interact with the molecule) and low chemical lability in polar solvent (4242 Dos Santos PR, Pich CT, Back D, Smiderle F, Dumas F, Moura S. Synthesis, chemical characterization and DNA interaction study of new diclofenac and ibuprofen zinc (II)-nicotinamide ternary complexes as cyclooxygenase inhibitor prototypes. J. Inorg. Biochem. [Internet]. 2020;206(February):111046. Available from: https://doi.org/10.1016/j.jinorgbio.2020.111046
https://doi.org/10.1016/j.jinorgbio.2020...
).

The mechanism used by the compounds to reduce cell viability remains unknown, however we hypothesize that it is related to COX inhibition, as other groups found with other NSAIDs-derived compounds (3030 Bacchi S, Palumbo P, Sponta A, Coppolino MF. Clinical Pharmacology of Non-Steroidal Anti-Inflammatory Drugs : A Review. Antiinflamm Antiallergy Agents Med Chem. 2012;11(1):52-64.,4949 Tabrizi L, Olasunkanmi LO, Fadare OA. Experimental and theoretical investigations of cyclometalated ruthenium(ii) complex containing CCC-pincer and anti-inflammatory drugs as ligands: synthesis, characterization, inhibition of cyclooxygenase and in vitro cytotoxicity activities in various can. Dalton Trans. 2019;48:728-40.). Both ibuprofen and diclofenac are non-selective COX inhibitors, meaning they inhibit both COX-1 and 2, although diclofenac has a higher selectivity for COX-2 (5050 Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-ß through a PGE2-dependent mechanisms. Carcinogenesis. 2008;29(11):2227-35.,5151 Grosser T. Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities. J. Clin. Investig. 2005 Dec 8;116(1):4-15.). MDA-MB-231 and 4T1 were previously shown to have high expression of COX-2 and MCF-7 showed low expression of COX-2 (5252 Majumder M, Dunn L, Liu L, Hasan A, Vincent K, Brackstone M, et al. COX-2 induces oncogenic micro RNA miR655 in human breast cancer. Sci Rep. 2018;8(1):327.). Thus, we hypothesize that complex 1 had more cytotoxic effect on MDA-MB-231 and 4T1 cells because since it is derived from diclofenac, it preferably inhibits COX-2. Specially for breast cancer, COX-2 was sufficient to promote tumorigenesis in transgenic mice (5353 Liu CH, Chang SH, Narko K, Trifan OC, Wu MT, Smith E, et al. Overexpression of Cyclooxygenase-2 is Sufficient to Induce Tumorigenesis in Transgenic Mice. J. Biol. Chem. 2001;276(21):18563-9.) and was also related to higher histological grade, tumors with larger sizes, high Ki-67 and p53 expression, negative hormone receptor status, HER-2 amplification and presence of axillary lymph node metastases (1818 Dell'Omo G, Crescenti D, Vantaggiato C, Parravicini C, Borroni AP, Rizzi N, et al. Inhibition of SIRT1 deacetylase and p53 activation uncouples the anti-inflammatory and chemopreventive actions of NSAIDs. Br J Cancer. 2019;120:537-46.).

Both compounds did not show greater cytotoxic effects in MCF-7 cells - which may be due to low COX-2 expression- and presented more cytotoxic effects against 4T1 cell lines. Interestingly the complex 1 presented a cytotoxic effect on MDA-MB-231. The distinct effects of compounds on BC cell lines are probably due to different phenotype and origin of the cells, as 4T1 cells are originated from mice, and MCF-7 and MDA-MB-231 from human invasive breast ductal carcinoma. Additionally, molecular subtypes of each cell are individual, as MCF-7 is classified as luminal (less aggressive, better prognosis) and MDA-MB-231 as claudin-low (more aggressive, worse prognosis) (4444 Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Research. 2011 Aug 12;13(4). 215.,5454 Ristimäki A, Sivula A, Lundin J, Lundin M, Salminen T, Haglund C, et al. Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Res. 2002;62(3):632-5.). Other study presented alternative pathways that show how NSAIDs, including ibuprofen and diclofenac, can act on MDA-MB-231 cell lines, through inhibiting the sirtuin 1 (SIRT1) deacetylase activity, increasing acetylation and activity of the tumor suppressor p53 and the expression of the antiproliferative gene p21 (5555 Dai X, Cheng H, Bai Z, Li J. Breast cancer cell line classification and Its relevance with breast tumor subtyping. J. Cancer. 2017;8:3131-41.). Moreover, studies with diclofenac showed COX-independent mechanisms, such as modulation of MYC expression and glucose metabolism, resulting in impaired carcinoma cell line proliferation in murine melanoma cell line B16F10 (5656 Gottfried E, Lang SA, Renner K, Bosserhoff A, Gronwald W, Rehli M, et al. New Aspects of an Old Drug - Diclofenac Targets. Plos One, 2013;(7):e66987.).

In summary, our results demonstrated that complexes 1 and 2 are more effective than the NSAIDs ibuprofen and diclofenac in reducing cellular growth in three different breast cancer cell lines, highlighting the potential antitumoral activity of Zinc-NSAIDs and Nic complexes. In this context, future studies should be conducted to better understand the mechanism by which the synthesized compounds, complex 1 and complex 2, act on cell viability.

Acknowledgments

We thank Dr. Jarbas Rodrigues de Oliveira and Dr. Cristiano Valim Bizarro for the contribution on the project.

REFERENCES

  • 1
    Christopher PW, Bernard WS, Elisabeth W. World Cancer Report 2020. Lyon: WHO International agency for Research on cancer; 2020 p. 613
  • 2
    Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J. for Clin. 2018;68(6):394-424.
  • 3
    Boodram JN, Mcgregor IJ, Bruno PM, Cressey PB, Hemann MT, Suntharalingam K. Breast Cancer Stem Cell Potent Copper(II)-Non-Steroidal Anti-Inflammatory Drug Complexes. Angew. Chem. - Int. Ed. 2016;55(8):2845-50.
  • 4
    Feng J, Du X, Liu H, Sui X, Zhang C, Tang Y, et al. Manganese-mefenamic acid complexes exhibit high lipoxygenase inhibitory activity. Dalton Transactions. 2014;43(28):10930-9.
  • 5
    Ricciotti E, Fitzgerald GA. Prostaglandins and inflammation. Arterioscler, Thromb, and Vasc Biol. 2011;31(5):986-1000.
  • 6
    Kobayashi K, Omori K, Murata T. Role of prostaglandins in tumor microenvironment. Cancer Metastasis Rev. 2018;37(2-3):347-54.
  • 7
    Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: Structural, Cellular, and Molecular Biology. Annu. Rev. of Biochem. 2000 Jun;69:145-82.
  • 8
    Sharpe CR, Collet J-P, McNutt M, Belzile E, Boivin J-F, Hanley JA. Nested case-control study of the effects of non-steroidal anti-inflammatory drugs on breast cancer risk and stage. Br J Cancer. 2000 Jul;83(1). 112-20.
  • 9
    Moris D, Kontos M, Spartalis E, Fentiman IS. The Role of NSAIDs in Breast Cancer Prevention and Relapse: Current Evidence and Future Perspectives. Breast Care. 2016;11(5). 339-344.
  • 10
    Bittoni MA, Carbone DP, Harris RE. Ibuprofen and fatal lung cancer: A brief report of the prospective results from the Third National Health and Nutrition Examination Survey (NHANES III). Mol. and Clin. Oncol. [Internet]. 2017;6(6):917-20. Available from: https://www.spandidos-publications.com/
    » https://www.spandidos-publications.com
  • 11
    Muscat JE, Chen S-Q, Richie JP, Altorki NK, Citron M, Olson S, et al. Risk of lung carcinoma among users of nonsteroidal antiinflammatory drugs. Cancer. 2003 Apr 1;97(7). 1732-6.
  • 12
    Doat S, Cénée S, Trétarre B, Rebillard X, Lamy P, Bringer J, et al. Nonsteroidal anti-inflammatory drugs (NSAIDs) and prostate cancer risk: results from the EPICAP study. Cancer Med. 2017;6(10):2461-70.
  • 13
    Daugherty SE, Pfeiffer RM, Sigurdson AJ, Hayes RB, Leitzmann M, Schatzkin A, et al. Nonsteroidal antiinflammatory drugs and bladder cancer: A pooled analysis. Am. J. of Epidemiol. 2011;173(7):721-30.
  • 14
    Wernli KJ, Newcomb PA, Hampton JM, Trentham-Dietz A, Egan KM. Inverse association of NSAID use and ovarian cancer in relation to oral contraceptive use and parity. Br J Cancer. 2008;98(11):1781-3.
  • 15
    Baandrup L, Faber MT, Christensen J, Jensen A, Andersen KK, Friis S, et al. Nonsteroidal anti-inflammatory drugs and risk of ovarian cancer: Systematic review and meta-analysis of observational studies. Acta Obstet. Gynecol. Scand. 2013;92:245-55.
  • 16
    Corley D, Kerlikowske K, Verma R, Buffler P. Protective association of aspirin/NSAIDs and esophageal cancer: A systematic review and meta-analysis. Gastroenterology. 2003;124(1):47-56.
  • 17
    Huang X, Chen Y, Wu J, Zhang X, Wu C, Zhang C, et al. Aspirin and non-steroidal anti-inflammatory drugs use reduce gastric cancer risk: A dose-response meta-analysis. Oncotarget. 2017;8(3):4781-95.
  • 18
    Dell'Omo G, Crescenti D, Vantaggiato C, Parravicini C, Borroni AP, Rizzi N, et al. Inhibition of SIRT1 deacetylase and p53 activation uncouples the anti-inflammatory and chemopreventive actions of NSAIDs. Br J Cancer. 2019;120:537-46.
  • 19
    Grivennikov SI, Karin M. Inflammation and oncogenesis: a vicious connection. Vol. 20, Curr. Opin. Genet. and Dev. 2010. p. 65-71.
  • 20
    Grivennikov SI, Greten FR, Karin M. Immunity, Inflammation, and Cancer. Cell. 2010.19, 140(6)883-99.
  • 21
    Bartsch H, Nair J. Chronic inflammation and oxidative stress in the genesis and perpetuation of cancer: Role of lipid peroxidation, DNA damage, and repair. Langenbeck's Arch. Surg. 2006;391:499-510.
  • 22
    Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: A review. J. Cell. Physiol. 2019.234:5683-99.
  • 23
    Ohtsuka J, Oshima H, Ezawa I, Abe R, Oshima M, Ohki R. Functional loss of p53 cooperates with the in vivo microenvironment to promote malignant progression of gastric cancers. Sci Rep. 2018;8(1):2291.
  • 24
    Tuncer S, Banerjee S. Eicosanoid pathway in colorectal cancer: Recent updates. World J. of Gastroenterol. 2015;7(21):11746-66.
  • 25
    Echizen K, Hirose O, Maeda Y, Oshima M. Inflammation in gastric cancer: Interplay of the COX-2/prostaglandin E 2 and Toll-like receptor/MyD88 pathways. Cancer Sci. 2016 Apr 18;107(4). 391-7.
  • 26
    Howe LR. Inflammation and breast cancer. Cyclooxygenase/prostaglandin signaling and breast cancer. Breast Cancer Res. 2007;9(4):210.
  • 27
    Dong XF, Liu TQ, Zhi XT, Zou J, Zhong JT, Li T, et al. COX-2/PGE2 axis regulates HIF2a activity to promote hepatocellular carcinoma hypoxic response and reduce the sensitivity of sorafenib treatment. Clin. Cancer Res. 2018;24(13):3204-16.
  • 28
    Esbona K, Yi Y, Saha S, Yu M, van Doorn RR, Conklin MW, et al. The Presence of Cyclooxygenase 2, Tumor-Associated Macrophages, and Collagen Alignment as Prognostic Markers for Invasive Breast Carcinoma Patients. Am. J. of Pathol. 2018;188(3):559-73.
  • 29
    Greenhough A, Smartt HJM, Moore AE, Roberts HR, Williams AC, Paraskeva C, et al. The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis. 2009 Jan 9;30(3): 377-86
  • 30
    Bacchi S, Palumbo P, Sponta A, Coppolino MF. Clinical Pharmacology of Non-Steroidal Anti-Inflammatory Drugs : A Review. Antiinflamm Antiallergy Agents Med Chem. 2012;11(1):52-64.
  • 31
    Smith GS, Therrien B. Targeted and multifunctional arene ruthenium chemotherapeutics. Dalton Trans. 2011;40:10793-800.
  • 32
    Zaki M, Hairat S, Aazam ES. Scope of organometallic compounds based on transition metal-arene systems as anticancer agents: Starting from the classical paradigm to targeting multiple strategies. RSC Advances. 2019;9:3239-78.
  • 33
    Szczepaniak A, Fichna J. Organometallic compounds and metal complexes in current and future treatments of inflammatory bowel disease and colorectal cancer-a critical review. Biomolecules. 2019;22(9):398.
  • 34
    Ronconi L, Sadler PJ. Using coordination chemistry to design new medicines. Coord. Chem. Rev. 2007; 251:1633-48.
  • 35
    Gianferrara T, Bratsos I, Alessio E. A categorization of metal anticancer compounds based on their mode of action. Dalton Trans. 2009;(37): 7588-98.
  • 36
    Allgeier AM, Mirkin CA. Ligand Design for Electrochemically Controlling Stoichiometric and Catalytic Reactivity of Transition Metals. Angew. Chem. Int. Ed. 1998 Apr 20;37(7):894-908.
  • 37
    Gasser G, Metzler-Nolte N. The potential of organometallic complexes in medicinal chemistry. Curr. Opin. Chem. Biol. 2012 Apr;16(1-2):84-91.
  • 38
    Mandal P, Kundu BK, Vyas K, Sabu V, Helen A, Dhankhar SS, et al. Ruthenium (II) arene NSAID complexes: Inhibition of cyclooxygenase and antiproliferative activity against cancer cell lines. Dalton Trans. 2018;47(2):517-27.
  • 39
    Alves Rico SR, Abbasi AZ, Ribeiro G, Ahmed T, Wu XY, de Oliveira Silva D. Diruthenium(II,III) metallodrugs of ibuprofen and naproxen encapsulated in intravenously injectable polymer-lipid nanoparticles exhibit enhanced activity against breast and prostate cancer cells. Nanoscale. 2017;9(30):10701-14.
  • 40
    Hanif-Ur-Rehman, Freitas TE, Gomes RN, Colquhoun A, de Oliveira Silva D. Axially-modified paddlewheel diruthenium(II,III)-ibuprofenato metallodrugs and the influence of the structural modification on U87MG and A172 human glioma cell proliferation, apoptosis, mitosis and migration. J. Inorg. Biochem. [Internet]. 2016;165:181-91. Available from: http://dx.doi.org/10.1016/j.jinorgbio.2016.10.003
    » http://dx.doi.org/10.1016/j.jinorgbio.2016.10.003
  • 41
    Intini FP, Zajac J, Novohradsky V, Saltarella T, Pacifico C, Brabec V, et al. Novel Antitumor Platinum (II) Conjugates Containing the Nonsteroidal Anti-inflammatory Agent Diclofenac: Synthesis and Dual Mechanisms of Antiproliferative Effects. Inorg. Chem. 2017;56(3):1483-97.
  • 42
    Dos Santos PR, Pich CT, Back D, Smiderle F, Dumas F, Moura S. Synthesis, chemical characterization and DNA interaction study of new diclofenac and ibuprofen zinc (II)-nicotinamide ternary complexes as cyclooxygenase inhibitor prototypes. J. Inorg. Biochem. [Internet]. 2020;206(February):111046. Available from: https://doi.org/10.1016/j.jinorgbio.2020.111046
    » https://doi.org/10.1016/j.jinorgbio.2020.111046
  • 43
    Pulaski BA, Ostrand-Rosenberg S. Mouse 4T1 Breast Tumor Model. Curr. Protoc. in Immunol. 2000 Oct;39(1).
  • 44
    Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Research. 2011 Aug 12;13(4). 215.
  • 45
    Abu Ali H, Jabali B. Synthesis, characterization and biological activity of novel complexes of zinc(II) diclofenac with nitrogen based ligands. Polyhedron. 2016;107:97-106.
  • 46
    Abu Ali H, Omar SN, Darawsheh MD, Fares H. Synthesis, characterization and antimicrobial activity of zinc(II) ibuprofen complexes with nitrogen-based ligands. J. Coord. Chem. 2016;69(6):1110-22.
  • 47
    Quispe M A, Zavala C D, Rojas C J, Posso R M, Vaisberg W A. Efecto citotóxico selectivo in vitro de muricin H (acetogenina de Annona muricata) en cultivos celulares de cáncer de pulmón. Rev peru med exp salud publica. 2006;23(4):256-69.
  • 48
    Benadiba M, dos Santos RRP, Silva D de O, Colquhoun A. Inhibition of C6 rat glioma proliferation by [Ru2Cl(Ibp)4] depends on changes in p21, p27, Bax/Bcl2 ratio and mitochondrial membrane potential. J. Inorg. Biochem. [Internet]. 2010;104(9):928-35. Available from: http://dx.doi.org/10.1016/j.jinorgbio.2010.04.011
    » http://dx.doi.org/10.1016/j.jinorgbio.2010.04.011
  • 49
    Tabrizi L, Olasunkanmi LO, Fadare OA. Experimental and theoretical investigations of cyclometalated ruthenium(ii) complex containing CCC-pincer and anti-inflammatory drugs as ligands: synthesis, characterization, inhibition of cyclooxygenase and in vitro cytotoxicity activities in various can. Dalton Trans. 2019;48:728-40.
  • 50
    Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-ß through a PGE2-dependent mechanisms. Carcinogenesis. 2008;29(11):2227-35.
  • 51
    Grosser T. Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities. J. Clin. Investig. 2005 Dec 8;116(1):4-15.
  • 52
    Majumder M, Dunn L, Liu L, Hasan A, Vincent K, Brackstone M, et al. COX-2 induces oncogenic micro RNA miR655 in human breast cancer. Sci Rep. 2018;8(1):327.
  • 53
    Liu CH, Chang SH, Narko K, Trifan OC, Wu MT, Smith E, et al. Overexpression of Cyclooxygenase-2 is Sufficient to Induce Tumorigenesis in Transgenic Mice. J. Biol. Chem. 2001;276(21):18563-9.
  • 54
    Ristimäki A, Sivula A, Lundin J, Lundin M, Salminen T, Haglund C, et al. Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Res. 2002;62(3):632-5.
  • 55
    Dai X, Cheng H, Bai Z, Li J. Breast cancer cell line classification and Its relevance with breast tumor subtyping. J. Cancer. 2017;8:3131-41.
  • 56
    Gottfried E, Lang SA, Renner K, Bosserhoff A, Gronwald W, Rehli M, et al. New Aspects of an Old Drug - Diclofenac Targets. Plos One, 2013;(7):e66987.
  • Funding:

    This research was funded by FAPERGS 16/2551-0000523-0 and CAPES financial code 1.

Edited by

Editor-in-Chief:

Alexandre Rasi Aoki

Associate Editor:

Daniel Fernandes

Publication Dates

  • Publication in this collection
    10 Dec 2021
  • Date of issue
    2021

History

  • Received
    12 Jan 2021
  • Accepted
    09 May 2021
Instituto de Tecnologia do Paraná - Tecpar Rua Prof. Algacyr Munhoz Mader, 3775 - CIC, 81350-010 Curitiba PR Brazil, Tel.: +55 41 3316-3052/3054, Fax: +55 41 3346-2872 - Curitiba - PR - Brazil
E-mail: babt@tecpar.br