scholarly journals Dual Inhibition of Class IA Phosphatidylinositol 3-Kinase and Mammalian Target of Rapamycin as a New Therapeutic Option for T-Cell Acute Lymphoblastic Leukemia

2009 ◽  
Vol 69 (8) ◽  
pp. 3520-3528 ◽  
Author(s):  
Francesca Chiarini ◽  
Federica Falà ◽  
Pier Luigi Tazzari ◽  
Francesca Ricci ◽  
Annalisa Astolfi ◽  
...  
2017 ◽  
Vol 233 (3) ◽  
pp. 1796-1811 ◽  
Author(s):  
Cecilia Evangelisti ◽  
Alessandra Cappellini ◽  
Mariana Oliveira ◽  
Rita Fragoso ◽  
João T. Barata ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2025-2025
Author(s):  
Francesca Chiarini ◽  
Cecilia Grimaldi ◽  
Francesca Ricci ◽  
Pierluigi Tazzari ◽  
Camilla Evangelisti ◽  
...  

Abstract Abstract 2025 Poster Board II-2 Introduction: Recent findings have highlighted that constitutively active phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) signaling is a common feature of T-cell acute lymphoblastic leukemia (T-ALL) where it strongly influences cell proliferation and survival. Pathway activation could be due to several reasons which include Notch1 activation leading to HES1-mediated transcriptional suppression of PTEN gene, PTEN phosphorylation or oxidation, and inactivation of SHIP1 phosphatase. These findings lend compelling weight for the application of PI3K/Akt/mTOR inhibitors in T-ALL. Rapamycin and its analogues have shown some promising effects in pre-clinical models of T-ALL. However, mTOR inhibitors are mainly cytostatic and could hyperactivate Akt due to the existence of feedback loops between mTOR, p70 S6 kinase, PI3K, and Akt. Recently, dual PI3K/mTOR inhibitors have been synthesized. Here, we have analyzed the therapeutic potential of the novel, dual PI3K/mTOR inhibitor, NVP-BEZ235, an orally bioavailable imidazoquinoline derivative, which has entered clinical trials for solid tumors, on both T-ALL cell lines and patient samples. Methods and Patients: We employed a panel of cell lines with up-regulated PI3K/Akt/mTOR signaling, including CEM-R cells [which overexpress high levels of the membrane transporter, 170-kDa P-glycoprotein (P-gp)], MOLT-4 and CEM-S cells (which lack PTEN expression), Jurkat cells (which do not express both PTEN and SHIP1), and RPMI-8402 and BE-13 cells. MOLT-4, CEM, and Jurkat cells have a non-functional p53 pathway. Moreover, both Jurkat and MOLT-4 cells have aberrant Notch1 signaling. Patients samples displayed pathway activation as documented by increased levels of p-Akt, p-4E-BP1, and p-S6 ribosomal protein, as well as low/absent PTEN expression. Results: NVP-BEZ235 was cytotoxic to the panel of cell lines as documented by MTT assays. NVP-BEZ235 IC50 ranged from 80 to 280 nM at 24 h. A comparison between NVP-BEZ235 and the dual PI3K/mTOR inhibitor PI-103, a small synthetic molecule of the pyridofuropyrimidine class with the same targets, demonstrated that NVP-BEZ235 was more effective than PI-103 when employed at equimolar concentrations. NVP-BEZ235 did not significantly affect the proliferation of peripheral blood T-lymphocytes from healthy donors stimulated with phytohemagglutinin and interleukin-2, whereas it blocked leukemic cells in the G1 phase of the cell cycle, and this was accompanied by decreased levels of phosphorylated Retinoblastoma protein. NVP-BEZ235 treatment also resulted in apoptotic cell death (about 20-30% at 6 h of exposure, when employed at 200 nM), as documented by Annexin V/propidium iodide staining and cytofluorimetric analysis. Moreover, NVP-BEZ235 activated caspase-8 and caspase-3, as demonstrated by western blot. Western blot documented a dose- and time-dependent dephosphorylation of Akt and its downstream target, GSK-3β, in response to NVP-BEZ235. mTOR downstream targets were also efficiently dephosphorylated, including p70S6 kinase, S6 ribosomal protein, and 4E-BP1. Remarkably, NVP-BEZ235 targeted the side population (SP, identified by Hoechst 33342 staining and ABCG2 expression) of T-ALL cell lines, which might correspond to leukemia initiating cells, and synergized with several chemotherapeutic agents (dexamethasone, vincristine, cyclophosphamide, Ara-C) currently employed for treating T-ALL patients. NVP-BEZ235 reduced chemoresistance to vincristine induced in Jurkat cells by co-culturing with MS-5 stromal cells which mimic the bone marrow microenvironment. NVP-BEZ235 was cytotoxic (IC50: 10-15 nM at 96 h) to primary lymphoblasts from patients with T-ALL, where the drug dephosphorylated 4E-BP1, at variance with rapamycin. Of note, NVP-BEZ235 targeted the SP also in T-ALL patient samples. Conclusions: NVP-BEZ235 was cytotoxic to T-ALL cell lines and patient lymphoblasts (including SP cells) at concentrations that have been previously reported to be achievable in vivo. Taken together, our findings indicate that longitudinal inhibition at two nodes of the PI3K/Akt/mTOR network with NVP-BEZ235, either alone or in combination with other drugs, may serve as an efficient treatment towards T-ALL cells (including those overexpressing P-gp and independently from p53 status) which require upregulation of this signaling pathway for their survival and growth. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1921-1921 ◽  
Author(s):  
Francesca Chiarini ◽  
Federica Fala ◽  
Francesca Ricci ◽  
PierLuigi Tazzari ◽  
Annalisa Astolfi ◽  
...  

Abstract Constitutively activated phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) signaling is a common feature of T-cell acute lymphoblastic leukaemia (T-ALL). Recently, it was demonstrated that activated Notch-1 leads to constitutive activation of the PI3K/Akt/mTOR pathway by HES1-mediated transcriptional suppression of the PTEN gene. In addition, PTEN is mutated in about 20% of T-ALL patients, and virtually all T-ALL cell lines that are resistant to Notch-1 inhibition with γ-secretase inhibitors, contain mutations leading to either no or low PTEN expression. These findings lend compelling weight for the application of PI3K/Akt/mTOR inhibitors in T-ALL. However, our knowledge of PI3K/Akt/mTOR signalling in T-ALL is still limited and it is not clear whether it could be an effective target for innovative therapeutic strategies. Here, we have characterized PI3K/Akt/mTOR signalling in T-ALL cell lines (Jurkat, MOLT-4, CEM) lacking PTEN expression, including one (CEM-R) which over expresses high levels of the membrane transporter, 170-kDa P-glycoprotein (P-gp), one of the main determinants of multidrug-resistance. While MOLT-4 cells display wild-type p53, both Jurkat and CEM have a non-functional p53 pathway. Moreover, we have analyzed the therapeutic potential of the dual PI3K/mTOR inhibitor, PI-103, a small synthetic molecule of the pyridofuropyrimidine class, on both T-ALL cell lines and patient samples. T-ALL cell lines expressed p110α, p110β, p110γ, and p110α PI3K. Moreover, they expressed Akt1 and Akt2, both of which were found to be constitutively phosphorylated on Ser 473 and Ser 474, respectively, by immunoprecipitation experiments. Treatment of T-ALL cell lines with selective pharmacological inhibitors of p110 PI3K isoforms, demonstrated that only a p110α PI3K inhibitor (PIK75) was cytotoxic, resulting in a 40–50% reduction of cell growth when used at 2 μM. Consistently, only PIK75 induced Akt1 and Akt2 dephosphorylation on Ser 473 or Ser 474, respectively, hinting that p110α is the most important isoform for the activation of downstream signalling events. PI-103 was cytotoxic to all T-ALL cell lines including P-gp overexpressing cells, as it reduced cell growth by approximately 70% when employed at 2 μM for 24 h. PI-103 IC50 ranged from 0.5 to 1.0 μM at 24 h. PI-103 treatment resulted in apoptotic cell death (about 30% at 6 h of exposure, when employed at 0.75 μM), as demonstrated by Annexin V/propidium iodide staining and cytofluorimetric analysis. PI-103 caused both Akt1 and Akt2 dephosphorylation, accompanied by dephosphorylation of the Akt downstream target, glycogen synthase kinase (GSK) -3β. Also mTOR downstream targets were dephosphorylated in response to PI-103, including p70S6 kinase, ribosomal S6 protein, and 4E-BP1. Moreover, PI-103 resulted in lower levels of c-Myc expression. PI-103 activated caspase-3, -8, and -9. In contrast, an mTOR inhibitor (rapamycin) was less cytotoxic than PI-103 (25–30% reduction of cell growth at 100 nM after 24 h), blocked cells in the G1 phase of the cell cycle, and was much less effective in inducing apoptosis (about 5% at 6 h of treatment). Remarkably, rapamycin was almost completely ineffective against CEM-R cells. A combination consisting of PIK75 and rapamycin was less cytotoxic to T-ALL cell lines than PI-103 alone. Furthermore, rapamycin treatment, at variance with PI-103, resulted in an overactivation of the Akt/ GSK-3β axis, as documented by increased phosphorylation levels of both Akt and GSK- 3β. PI-103 was also cytotoxic to primary lymphoblasts from patients with T-ALL (IC50: 0.15 nM at 96 h), displaying constitutive phosphorylation of Akt and 4E-BP1, as well as low/absent PTEN expression. These data indicate that multi-targeted therapy towards PI3K and mTOR, may serve as an efficient treatment towards T-ALL cells (including those over expressing P-gp and independently from p53 state) which require upregulation of PI3K/Akt/mTOR signaling for their survival and growth.


Blood ◽  
2009 ◽  
Vol 113 (26) ◽  
pp. 6695-6698 ◽  
Author(s):  
Jean-Michel Terme ◽  
Ludovic Lhermitte ◽  
Vahid Asnafi ◽  
Pierre Jalinot

Abstract T-cell acute lymphoblastic leukemia 1 (TAL1), also known as stem cell leukemia (SCL), plays important roles in differentiation of hematopoietic and endothelial cells and is deregulated in a high percentage of T-cell acute lymphoblastic leukemia (T-ALL). In this report we show that the intracellular concentration of TAL1 is regulated by transforming growth factor β (TGF-β), which triggers its polyubiquitylation and degradation by the proteasome. This effect is mediated by AKT1, which phosphorylates TAL1 at threonine 90. Immunoprecipitation experiments showed that this event increases association of TAL1 with the E3 ubiquitin ligase CHIP. The E47 heterodimerization partner of TAL1 hinders this association. Our observations indicate that activation of the TGF-β and phosphatidylinositol 3-kinase/AKT pathways might reverse overexpression of TAL1 in leukemic cells by inducing proteolysis of this important oncogene.


Sign in / Sign up

Export Citation Format

Share Document