Induction of p21- and p27-mediated cell cycle arrest in Nalm-6 cells using pan-PI3K inhibitor (Buparlisib)

authors:

avatar Ava Safaroghli-Azar ORCID , avatar Mohammadreza Sadri , avatar Davood Bashash , *


how to cite: Safaroghli-Azar A, Sadri M, Bashash D. Induction of p21- and p27-mediated cell cycle arrest in Nalm-6 cells using pan-PI3K inhibitor (Buparlisib). koomesh. 2018;20(4):e153023. 

Abstract

Introduction: In the era of targeted therapies, efforts are being made to identify the novel agents with strong anti-tumor activity in order to improve cure rates of human malignancies while reducing the toxic side effects of intensive regimens. Increasing number of genetic and cancer biology studies indicated a prominent role for the aberrant activation of PI3K/Akt pathway in not only the pathogenesis of acute lymphoblastic leukemia (ALL), but also in acquisition of chemo-resistant phenotype. In this study we aimed to assess the anti-proliferative effect of pan class I PI3K inhibitor, Buparlisib, on Nalm-6 cells. Materials and Methods: To evaluate whether inhibition of PI3K using Buparlisib could exert anti-proliferative effect in leukemic cells, Nalm-6 were subjected to different concentrations of the inhibitor and subsequent cell viability, cell count, cell cycle progression and transcriptional alteration of cell cycle-related genes were investigated. Results: Buparlisib not only decreased the number of viable inhibitor-treated cells in a concentration and time-dependent manner, but also reduced the survival rate of Nalm-6 cells. Furthermore, we found that the anti-proliferative effect of this inhibitor is mediated, at least partially, through induction of G1 arrest as a result of up-regulated p21 and p27 and down-regulated c.Myc expression level. Conclusion: The present study showed that Buparlisib decreased the proliferative capacity of Nalm-6 cells probably via reduction in DNA synthesize rate, induction of cell cycle arrest and alteration in the expression level of proliferative-related genes. The results suggest that PI3K inhibitors may be used as a promising therapeutic strategy for the treatment of ALL patients.

References

  • 1.

    Robak T. Acute lymphoblastic leukaemia in elderly patients. Drugs Aging 2004; 21: 779-791.

  • 2.

    Hoelzer D, Gkbuget N, Ottmann O, Pui C, Relling M, Appelbaum F, et al. Acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2002; 162-192.

  • 3.

    Barrett D, Brown VI, Grupp SA, Teachey DT. Targeting the PI3K/AKT/mTOR signaling axis in children with hematologic malignancies. Pediatr Drugs 2012; 14: 299-316.

  • 4.

    Martelli A, Evangelisti C, Chappell W, Abrams S, Bsecke J, Stivala F, et al. Targeting the translational apparatus to improve leukemia therapy: roles of the PI3K/PTEN/Akt/mTOR pathway. Leukemia 2011; 25: 1064-1079.

  • 5.

    Bressanin D, Evangelisti C, Ricci F, Tabellini G, Chiarini F, Tazzari PL, et al. Harnessing the PI3K/Akt/mTOR pathway in T-cell acute lymphoblastic leukemia: eliminating activity by targeting at different levels. Oncotarget 2012; 3: 811-823.

  • 6.

    Martelli AM, Evangelisti C, Chiarini F, McCubrey JA. The phosphatidylinositol 3-kinase/Akt/mTOR signaling network as a therapeutic target in acute myelogenous leukemia patients. Oncotarget 2010; 1: 89.

  • 7.

    Burris HA. Overcoming acquired resistance to anticancer therapy: focus on the PI3K/AKT/mTOR pathway. Cancer Chemother Pharmacol 2013; 71: 829-842.

  • 8.

    Huang C-H, Mandelker D, Schmidt-Kittler O, Samuels Y, Velculescu VE, Kinzler KW, et al. The structure of a human p110/p85 complex elucidates the effects of oncogenic PI3K mutations. Science 2007; 318: 1744-1748.

  • 9.

    Zhao L, Vogt PK. Helical domain and kinase domain mutations in p110 of phosphatidylinositol 3-kinase induce gain of function by different mechanisms. Proc Natl Acad Sci U S A 2008; 105: 2652-2657.

  • 10.

    Kok K, Geering B, Vanhaesebroeck B. Regulation of phosphoinositide 3-kinase expression in health and disease. Trends Biochem Sci 2009; 34: 115-127.

  • 11.

    Maira S-M, Pecchi S, Huang A, Burger M, Knapp M, Sterker D, et al. Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol Cancer Ther 2012; 11: 317-328.

  • 12.

    Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 2009; 9: 550-562.

  • 13.

    Brachmann SM, Kleylein-Sohn J, Gaulis S, Kauffmann A, Blommers MJ, Kazic-Legueux M, et al. Characterization of the mechanism of action of the pan class I PI3K inhibitor NVP-BKM120 across a broad range of concentrations. Mol Cancer Ther 2012; 11: 1747-1757.

  • 14.

    Burger MT, Pecchi S, Wagman A, Ni ZJ, Knapp M, Hendrickson T, et al. Identification of NVP-BKM120 as a potent, selective, orally bioavailable class I PI3 kinase inhibitor for treating cancer. ACS Med Chem Lett 2011; 2: 774-779.

  • 15.

    Shih KC, Acs P, Burris HA, Hart LL, Kosloff RA, Lamar RE, et al. Phase I study of the combination of BKM120 and bevacizumab in patients with relapsed/refractory glioblastoma multiforme (GBM) or other refractory solid tumors. J Clin Oncol 2018.

  • 16.

    Mayer IA, Abramson VG, Balko JM, Isakoff SJ, Kuba MG, Sanders M, et al. SU2C phase Ib study of pan-PI3K inhibitor BKM120 with letrozole in ER+/HER2-metastatic breast cancer (MBC). J Clin Oncol 2012.

  • 17.

    Tan DS-W, Lim KH, Tai WM, Ahmad A, Pan S, Ng QS, et al. A phase Ib safety and tolerability study of a pan class I PI3K inhibitor buparlisib (BKM120) and gefitinib (gef) in EGFR TKI-resistant NSCLC. J Clin Oncol 2013.

  • 18.

    Rosich L, Saborit-Villarroya I, Lpez-Guerra M, Xargay-Torrent S, Montraveta A, Aymerich M, et al. The phosphatidylinositol-3-kinase inhibitor NVP-BKM120 overcomes resistance signals derived from microenvironment by regulating the Akt/FoxO3a/Bim axis in chronic lymphocytic leukemia cells. Haematologica 2013; 98: 1739-1747.

  • 19.

    Bashash D, Delshad M, Safaroghli-Azar A, Safa M, Momeny M, Ghaffari SH. Novel pan PI3K inhibitor-induced apoptosis in APL cells correlates with suppression of telomerase: An emerging mechanism of action of BKM120. Int J Biochem Cell Biol 2017; 91: 1-8.

  • 20.

    Safa M, Tavasoli B, Manafi R, Kiani F, Kashiri M, Ebrahimi S, et al. Indole-3-carbinol suppresses NF-B activity and stimulates the p53 pathway in pre-B acute lymphoblastic leukemia cells. Tumour Biol 2015; 36: 3919-3930.

  • 21.

    Mitchell KO, El-Deiry WS. Overexpression of c-Myc inhibits p21WAF1/CIP1 expression and induces S-phase entry in 12-O-tetradecanoylphorbol-13-acetate (TPA)-sensitive human cancer cells. Cell Growth Differ 1999; 10: 223-230.

  • 22.

    Bertacchini J, Heidari N, Mediani L, Capitani S, Shahjahani M, Ahmadzadeh A, et al. Targeting PI3K/AKT/mTOR network for treatment of leukemia. Cell Mol Life Sci 2015; 72: 2337-2347.

  • 23.

    Bayati S, Bashash D, Ahmadian S, Safaroghli-Azar A, Alimoghaddam K, Ghavamzadeh A, et al. Inhibition of tachykinin NK 1 receptor using aprepitant induces apoptotic cell death and G1 arrest through Akt/p53 axis in pre-B acute lymphoblastic leukemia cells. Eur J Pharmacol 2016; 791: 274-283.

  • 24.

    Lonetti A, Antunes IL, Chiarini F, Orsini E, Buontempo F, Ricci F, et al. Activity of the pan-class I phosphoinositide 3-kinase inhibitor NVP-BKM120 in T-cell acute lymphoblastic leukemia. Leukemia 2014; 28: 1196-1206.

  • 25.

    Kim A, Park S, Lee JE, Jang WS, Lee SJ, Kang HJ, et al. The dual PI3K and mTOR inhibitor NVP-BEZ235 exhibits anti-proliferative activity and overcomes bortezomib resistance in mantle cell lymphoma cells. Leukemia Res 2012; 36: 912-920.

  • 26.

    Macleod KF, Sherry N, Hannon G, Beach D, Tokino T, Kinzler K, et al. p53-dependent and independent expression of p21 during cell growth, differentiation, and DNA damage. Genes Dev 1995; 9: 935-944.

  • 27.

    Toyoshima H, Hunter T. p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21. Cell 1994; 78: 67-74.

  • 28.

    Zheng Y, Yang J, Qian J, Zhang L, Lu Y, Li H, et al. Novel phosphatidylinositol 3-kinase inhibitor NVP-BKM120 induces apoptosis in myeloma cells and shows synergistic anti-myeloma activity with dexamethasone. J Mol Med 2012; 90: 695-706.