Empagliflozin and Doxorubicin Synergistically Inhibit the Survival of Triple-Negative Breast Cancer Cells via Interfering with the mTOR Pathway and Inhibition of Calmodulin: In Vitro and Molecular Docking Studies

Shenouda G Eliaa, Ahmed A Al-Karmalawy, Rasha M Saleh, Mohamed F Elshal, Shenouda G Eliaa, Ahmed A Al-Karmalawy, Rasha M Saleh, Mohamed F Elshal

Abstract

Triple-negative breast cancers (TNBCs) comprise 10-15% of all breast cancers but with more resistance affinity against chemotherapeutics. Although doxorubicin (DOX) is the recommended first choice, it has observed cardiotoxicity together with apparent drug resistance. The anti-hyperglycemic drug, empagliflozin (EMP), was recently indicated to have in vitro anticancer potential together with its previously reported cardioprotective properties related to calmodulin inhibition. In this study, we carried out molecular docking studies which revealed the potential blocking of the calmodulin receptor by EMP through its binding with similar crucial amino acids compared to its cocrystallized inhibitor (AAA) as a proposed mechanism of action. Moreover, combination of DOX with EMP showed a slightly lower cytotoxic activity against the MDA-MB-231 cell line (IC50 = 1.700 ± 0.121) compared to DOX alone (IC50 = 1.230 ± 0.131), but it achieved a more characteristic arrest in the growth of cells by 4.67-fold more than DOX alone (with only 3.27-fold) in comparison to the control as determined by cell cycle analysis, and at the same time reached an increase in the total apoptosis percentage from 27.05- to 29.22-fold, compared to DOX alone as indicated by Annexin V-FITC apoptosis assay. Briefly, the aforementioned in vitro studies in addition to PCR of pro- and antiapoptotic genes (mTOR, p21, JNK, Bcl2, and MDR1) suggest the chemosensitization effect of EMP combination with DOX which can reduce the required therapeutic dose of DOX in TNBC and eventually will decrease its toxic side effects (especially cardiotoxicity), along with decreasing the chemoresistance of TNBC cells to DOX treatment.

Conflict of interest statement

The authors declare no competing financial interest.

© 2020 American Chemical Society.

Figures

Figure 1
Figure 1
Graphical representation showing that adding EMP enhances the anticancer effects of DOX at a reduced concentration, suggesting that EMP could minimize adverse effects of DOX while preserving its therapeutic value.
Figure 2
Figure 2
2D and 3D representations for the redocked cocrystallized inhibitor (AAA) inside the calmodulin receptor pocket. The cocrystallized inhibitor is represented in green and the redocked one is represented in red.
Figure 3
Figure 3
3D representations and positionings for the docked cocrystallized calmodulin antagonist (AAA) inside A and B subunits of the calmodulin receptor.
Figure 4
Figure 4
3D representations and positionings for empagliflozin inside A and B subunits of the calmodulin receptor.
Figure 5
Figure 5
Graphical representations showing (A) the cytotoxic effects of DOX, EMP, and DOX/EMP combination, (B) IC50 of each treatment, (C) combination index (CI) of EMP and DOX, and (D) dose-reduction index (DRI) of DOX as analyzed with CompuSyn software. Data are average of three independent experiments ± SD.
Figure 6
Figure 6
Impact of conjugates DOX, EMP, and DOX/EMP combination on the cell cycle phases of MDA-MB-231 cells. Data are average of three independent experiments ± SD. *, §, and # represent significance (p < 0.5) compared with control, EMP, or DOX alone, respectively.
Figure 7
Figure 7
Combined effect of DOX and EMP on the apoptosis of MDA-MB-231 cells. Representative dot plot presenting the flow cytometry analysis of cells stained with Annexin V and PI following treatment with vehicle alone (A), 1.3 μM DOX alone (B), 50 μM EMP alone (C), or a combination of 1.3 μM DOX +50 μM EMP (D) for 24 h. Data are representative of three individual experiments.
Figure 8
Figure 8
Distribution of apoptotic cells in the AnnexinV-FITC/PI apoptosis assay in MDA-MB-231 cells after treatment with DOX, EMP, and DOX/EMP combination. Data are average of three independent experiments ± SD. *, §, and # represent significance (p < 0.5) compared with control, EMP, or DOX alone, respectively.
Figure 9
Figure 9
Analysis of gene expression profiles of mTOR (A), Bcl2 (B), JNK (C), and p21 (D), and of MDA-MB-231 cells treated with DOX, EMP, and DOX/EMP combination compared to blank by using real-time PCR. Data are average of three independent experiments ± SD. *, §, and # represent significance (p < 0.5) compared with control, EMP, or DOX alone, respectively.
Figure 10
Figure 10
Analysis of gene expression profiles of the multidrug resistance gene MDR1 in MDA-MB-231 after treatment with DOX, EMP, and DOX/EMP combination compared to blank by using real-time PCR. Data are average of three independent experiments ± SD. * and § represent significance (p < 0.5) compared with control, EMP, or DOX alone, respectively.

References

    1. Watson I. R.; Takahashi K.; Futreal P. A.; Chin L. (2013) Emerging patterns of somatic mutations in cancer. Nat. Rev. Genet. 14 (10), 703–718. 10.1038/nrg3539.
    1. Tate J. G.; Bamford S.; Jubb H. C.; et al. (2019) COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res. 47 (D1), D941–D947. 10.1093/nar/gky1015.
    1. Heron M.; Anderson R. N. (2017) Changes in the leading cause of death: Recent patterns in heart disease and cancer mortality. NCHS Data Brief 254, 1–8.
    1. Chavez K. J.; Garimella S. V.; Lipkowitz S. (2011) Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer. Breast Dis. 32 (1–2), 35.10.3233/BD-2010-0307.
    1. Ismail-Khan R.; Bui M. M. (2010) A Review of Triple-Negative Breast Cancer. Cancer Control 17, 173–176. 10.1177/107327481001700305.
    1. Yu A. F.; Chan A.; Steingart R. M. (2019) Cardiac Magnetic Resonance and Cardio-oncology–Does T2 Signal the End of Anthracycline Cardiotoxicity?. J. Am. Coll. Cardiol. 73 (7), 792.10.1016/j.jacc.2018.11.045.
    1. Arcamone F. (2012) Doxorubicin: Anticancer Antibiotics, Elsevier.
    1. Tacar O.; Sriamornsak P.; Dass C. R. (2013) Doxorubicin: An update on anticancer molecular action, toxicity and novel drug delivery systems. J. Pharm. Pharmacol. 65 (2), 157–170. 10.1111/j.2042-7158.2012.01567.x.
    1. Ghanem A.; Emara H. A.; Muawia S.; Abd El Maksoud A. I.; Al-Karmalawy A. A.; Elshal M. F. (2020) Tanshinone IIA synergistically enhances the antitumor activity of doxorubicin by interfering with the PI3K/AKT/mTOR pathway and inhibition of topoisomerase II: in vitro and molecular docking studies. New J. Chem. 44, 17374.10.1039/D0NJ04088F.
    1. Thorn C. F.; Oshiro C.; Marsh S.; et al. (2011) Doxorubicin pathways: pharmacodynamics and adverse effects. Pharmacogenet. Genomics 21 (7), 440.10.1097/FPC.0b013e32833ffb56.
    1. Tada Y.; Wada M.; Migita T.; Nagayama J.; Hinoshita E.; Mochida Y.; Maehara Y.; Tsuneyoshi M.; Kuwano M.; Naito S. (2002) Increased Expression of Multidrug Resistance-Associated Proteins in Bladder Cancer during Clinical Course and Drug Resistance to Doxorubicin. Int. J. Cancer 98, 630–635. 10.1002/ijc.10246.
    1. Abolhoda A.; Wilson A. E.; Ross H.; Danenberg P. V.; Burt M.; Scotto K. W. (1999) Rapid activation of MDR1 gene expression in human metastatic sarcoma after in vivo exposure to doxorubicin. Clin. Cancer Res. 5 (11), 3352–3356.
    1. Marquette C.; Nabell L. (2012) Chemotherapy-resistant metastatic breast cancer. Curr. Treat Options Oncol. 13 (2), 263–275. 10.1007/s11864-012-0184-6.
    1. Osman A.-M. M; Al-Harthi S. E; AlArabi O. M; Elshal M. F; Ramadan W. S; Alaama M. N; Al-Kreathy H. M; Damanhouri Z. A; Osman O. H (2013) Chemosensetizing and cardioprotective effects of resveratrol in doxorubicin- treated animals. Cancer Cell Int. 13 (1), 52.10.1186/1475-2867-13-52.
    1. Osman A.-M. M; Telity S. A; Damanhouri Z. A; Al-Harthy S. E; Al-Kreathy H. M; Ramadan W. S; Elshal M. F; Khan L. M; Kamel F. (2015) Chemosensitizing and nephroprotective effect of resveratrol in cisplatin -treated animals. Cancer Cell Int. 15 (1), 6.10.1186/s12935-014-0152-2.
    1. Pushpakom S.; Iorio F.; Eyers P. A.; et al. (2019) Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discovery 18 (1), 41–58. 10.1038/nrd.2018.168.
    1. Park K. (2019) A review of computational drug repurposing. Transl Clin Pharmacol. 27 (2), 59–63. 10.12793/tcp.2019.27.2.59.
    1. Polamreddy P.; Gattu N. (2019) The drug repurposing landscape from 2012 to 2017: evolution, challenges, and possible solutions. Drug Discovery Today 24 (3), 789–795. 10.1016/j.drudis.2018.11.022.
    1. Inzucchi S. E.; Bergenstal R. M.; Buse J. B.; et al. (2012) Management of hyperglycaemia in type 2 diabetes: a patient-centered approach. Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 55 (6), 1577–1596. 10.1007/s00125-012-2534-0.
    1. Committee for Medicinal Products for Human Use (CHMP) . (2017) Guideline on the Evaluation of Anticancer Medicinal Products in Man, European Medicines Agency, London, U.K..
    1. Uzma F.; Al-Mutairi F.; Humaira P.; Sahar K. (2020) An in-vitro and in Silico Anticancer Study of FDA Approved Antidiabetic Drugs Glimepiride and Empagliflozin. Int. J. Pharma Bio Sci. 10 (2), 52–57. 10.22376/ijpbs/lpr.2020.10.2.L52-57.
    1. Mustroph J.; Wagemann O.; Lücht C. M.; et al. (2018) Empagliflozin reduces Ca/calmodulin-dependent kinase II activity in isolated ventricular cardiomyocytes. ESC Hear Fail. 5 (4), 642–648. 10.1002/ehf2.12336.
    1. Anker S. D.; Butler J. (2018) Empagliflozin, calcium, and SGLT1/2 receptor affinity: another piece of the puzzle. ESC Heart Fail. 5, 549–551. 10.1002/ehf2.12345.
    1. Connelly K. A.; Zhang Y.; Visram A.; Advani A.; Batchu S. N.; Desjardins J.-F.; Thai K.; Gilbert R. E. (2019) Empagliflozin Improves Diastolic Function in a Nondiabetic Rodent Model of Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci. 4 (1), 27–37. 10.1016/j.jacbts.2018.11.010.
    1. Hait W. N.; Lazo J. S. (1986) Calmodulin: A potential target for cancer chemotherapeutic agents. J. Clin. Oncol. 4 (6), 994–1012. 10.1200/JCO.1986.4.6.994.
    1. Brzozowski J. S.; Skelding K. A. (2019) The multi-functional calcium/calmodulin stimulated protein kinase (CaMK) family: Emerging targets for anti-cancer therapeutic intervention. Pharmaceuticals 12 (1), 8.10.3390/ph12010008.
    1. Chemical Computing Group, Inc. (2016) Molecular operating environment (MOE), Montreal.
    1. Harmat V.; Böcskei Z.; Náray-Szabó G.; et al. (2000) A new potent calmodulin antagonist with arylalkylamine structure: Crystallographic, spectroscopic and functional studies. J. Mol. Biol. 297 (3), 747–755. 10.1006/jmbi.2000.3607.
    1. Chou T.-C. (2006) Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol. Rev. 58 (3), 621–681. 10.1124/pr.58.3.10.
    1. Osman A.-M. M.; Bayoumi H. M.; Al-Harthi S. E.; Damanhouri Z. A.; ElShal M. F. (2012) Modulation of doxorubicin cytotoxicity by resveratrol in a human breast cancer cell line. Cancer Cell Int. 12, 47.10.1186/1475-2867-12-47.
    1. Mohamed S.; Elshal M.; Kumosani T.; Aldahlawi A.; Basbrain T.; Alshehri F.; Choudhry H. (2016) L-asparaginase isolated from Phaseolus vulgaris seeds exhibited potent anti-acute lymphoblastic leukemia effects in-vitro and low immunogenic properties in-vivo. Int. J. Environ. Res. Public Health 13 (10), 1008.10.3390/ijerph13101008.
    1. Livak K. J.; Schmittgen T. D. (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods 25 (4), 402–408. 10.1006/meth.2001.1262.
    1. Davis I. W.; Baker D. (2009) RosettaLigand Docking with Full Ligand and Receptor Flexibility. J. Mol. Biol. 385 (2), 381–392. 10.1016/j.jmb.2008.11.010.
    1. McConkey B. J.; Sobolev V.; Edelman M. (2002) The performance of current methods in ligand-protein docking. Curr. Sci. 83 (7), 845–856.
    1. Chou T.-C. (2010) Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 70 (2), 440–446. 10.1158/0008-5472.CAN-09-1947.
    1. Advani S. H. (2010) Targeting mTOR pathway: A new concept in cancer therapy. Indian J. Med. Paediatr. Oncol. 31 (4), 132.10.4103/0971-5851.76197.
    1. Asnaghi L.; Calastretti A.; Bevilacqua A.; D'Agnano I.; Gatti G.; Canti G.; Delia D.; Capaccioli S.; Nicolin A. (2004) Bcl-2 phosphorylation and apoptosis activated by damaged microtubules require mTOR and are regulated by Akt. Oncogene 23, 5781–5791. 10.1038/sj.onc.1207698.
    1. Eom Y. H.; Kim H. S.; Lee A.; Song B. J.; Chae B. J. (2016) Breast Cancer BCL2 as a Subtype-Specific Prognostic Marker for Breast Cancer 19 (3), 252–260. 10.4048/jbc.2016.19.3.252.
    1. Wood R. A.; Barbour M. J.; Gould G. W.; Cunningham M. R.; Plevin R. J. (2018) Conflicting evidence for the role of JNK as a target in breast cancer cell proliferation: Comparisons between pharmacological inhibition and selective shRNA knockdown approaches. Pharmacol. Res. Perspect. 6, e00376.10.1002/prp2.376.
    1. Tournier C. (2013) The 2 Faces of JNK Signaling in Cancer. Genes Cancer 4, 397–400. 10.1177/1947601913486349.
    1. Shamloo B.; Usluer S. (2019) p21 in Cancer Research. Cancers 11, 1178.10.3390/cancers11081178.

Source: PubMed

3
Se inscrever