Dual Inhibition of PI3K and mTORC1/2 Signaling by NVP-BEZ235 as a New Therapeutic Strategy for Acute Myeloid Leukemia

Purpose: The growth and survival of acute myeloid leukemia (AML) cells are enhanced by the deregulation of signaling pathways such as phosphoinositide 3-kinase (PI3K)/Akt and mammalian target of rapamycin (mTOR). Major efforts have thus been made to develop molecules targeting these activated pathways. The mTOR serine/threonine kinase belongs to two separate complexes: mTORC1 and mTORC2. The mTORC1 pathway is rapamycin sensitive and controls protein translation through the phosphorylation of 4E-BP1 in most models. In AML, however, the translation process is deregulated and rapamycin resistant. Furthermore, the activity of PI3K/Akt and mTOR is closely related, as mTORC2 activates the oncogenic kinase Akt. We therefore tested, in this study, the antileukemic activity of the dual PI3K/mTOR ATP-competitive inhibitor NVP-BEZ235 compound (Novartis). Experimental Design: The activity of NVP-BEZ235 was tested in primary AML samples (n = 21) and human leukemic cell lines. The different signaling pathways were analyzed by Western blotting. The cap-dependent mRNA translation was studied by 7-methyl-GTP pull-down experiments, polysomal analysis, and [3H]leucine incorporation assays. The antileukemic activity of NVP-BEZ235 was tested by analyzing its effects on leukemic progenitor clonogenicity, blast cell proliferation, and survival. Results: The NVP-BEZ235 compound was found to inhibit PI3K and mTORC1 signaling and also mTORC2 activity. Furthermore, NVP-BEZ235 fully inhibits the rapamycin-resistant phosphorylation of 4E-BP1, resulting in a marked inhibition of protein translation in AML cells. Hence, NVP-BEZ235 reduces the proliferation rate and induces an important apoptotic response in AML cells without affecting normal CD34+ survival. Conclusions: Our results clearly show the antileukemic efficiency of the NVP-BEZ235 compound, which therefore represents a promising option for future AML therapies. Clin Cancer Res; 16(22); 5424–35. ©2010 AACR.

[1]  D. Dittmer,et al.  Dual inhibition of PI3K and mTOR inhibits autocrine and paracrine proliferative loops in PI3K/Akt/mTOR-addicted lymphomas. , 2010, Blood.

[2]  J. Tamburini,et al.  Autocrine IGF-1/IGF-1R signaling is responsible for constitutive PI3K/Akt activation in acute myeloid leukemia: therapeutic value of neutralizing anti-IGF-1R antibody , 2010, Haematologica.

[3]  Jing Chen,et al.  Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor , 2010, Nature Medicine.

[4]  Charles P. Lin,et al.  Dual targeting of the PI3K/Akt/mTOR pathway as an antitumor strategy in Waldenstrom macroglobulinemia. , 2010, Blood.

[5]  P. Lollini,et al.  NVP-BEZ235 as a New Therapeutic Option for Sarcomas , 2010, Clinical Cancer Research.

[6]  D. Fingar,et al.  mTOR Ser-2481 Autophosphorylation Monitors mTORC-specific Catalytic Activity and Clarifies Rapamycin Mechanism of Action* , 2009, The Journal of Biological Chemistry.

[7]  J. Tamburini,et al.  Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia. , 2009, Blood.

[8]  Kevin Curran,et al.  Biochemical, cellular, and in vivo activity of novel ATP-competitive and selective inhibitors of the mammalian target of rapamycin. , 2009, Cancer research.

[9]  Pixu Liu,et al.  Targeting the phosphoinositide 3-kinase pathway in cancer , 2009, Nature Reviews Drug Discovery.

[10]  N. Munshi,et al.  Antimyeloma activity of the orally bioavailable dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235. , 2009, Cancer research.

[11]  C. Chresta,et al.  Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTOR) , 2009, The Biochemical journal.

[12]  D. Hedley,et al.  Activity of a novel, dual PI3-kinase/mTor inhibitor NVP-BEZ235 against primary human pancreatic cancers grown as orthotopic xenografts , 2009, British Journal of Cancer.

[13]  D. Sabatini,et al.  An ATP-competitive Mammalian Target of Rapamycin Inhibitor Reveals Rapamycin-resistant Functions of mTORC1* , 2009, Journal of Biological Chemistry.

[14]  Gerard Manning,et al.  TORC-specific phosphorylation of mammalian target of rapamycin (mTOR): phospho-Ser2481 is a marker for intact mTOR signaling complex 2. , 2009, Cancer research.

[15]  D. Guertin,et al.  mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. , 2009, Cancer cell.

[16]  Robbie Loewith,et al.  Active-Site Inhibitors of mTOR Target Rapamycin-Resistant Outputs of mTORC1 and mTORC2 , 2009, PLoS biology.

[17]  D. Alessi,et al.  mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1). , 2008, The Biochemical journal.

[18]  T. Maeda,et al.  Isolation of Hyperactive Mutants of Mammalian Target of Rapamycin* , 2008, Journal of Biological Chemistry.

[19]  E. Estey,et al.  Therapeutic advances in leukemia and myelodysplastic syndrome over the past 40 years , 2008, Cancer.

[20]  S. Fröhling,et al.  Deregulation of signaling pathways in acute myeloid leukemia. , 2008, Seminars in oncology.

[21]  Daniela Gabriel,et al.  Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity , 2008, Molecular Cancer Therapeutics.

[22]  K. Shokat,et al.  PI-103, a dual inhibitor of Class IA phosphatidylinositide 3-kinase and mTOR, has antileukemic activity in AML , 2008, Leukemia.

[23]  J. Tamburini,et al.  Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. , 2008, Blood.

[24]  X. Bai,et al.  Rheb Activates mTOR by Antagonizing Its Endogenous Inhibitor, FKBP38 , 2007, Science.

[25]  F. Khuri,et al.  Inhibition of Mammalian Target of Rapamycin Induces Phosphatidylinositol 3-Kinase-Dependent and Mnk-Mediated Eukaryotic Translation Initiation Factor 4E Phosphorylation , 2007, Molecular and Cellular Biology.

[26]  Philippe Broët,et al.  Constitutive phosphoinositide 3-kinase/Akt activation represents a favorable prognostic factor in de novo acute myelogenous leukemia patients. , 2007, Blood.

[27]  David M Sabatini,et al.  Defining the role of mTOR in cancer. , 2007, Cancer cell.

[28]  D. Sabatini,et al.  Rapamycin derivatives reduce mTORC2 signaling and inhibit AKT activation in AML. , 2007, Blood.

[29]  C. Billottet,et al.  A selective inhibitor of the p110delta isoform of PI 3-kinase inhibits AML cell proliferation and survival and increases the cytotoxic effects of VP16. , 2006, Oncogene.

[30]  N. Sonenberg,et al.  mTOR, translation initiation and cancer , 2006, Oncogene.

[31]  D. Sabatini mTOR and cancer: insights into a complex relationship , 2006, Nature Reviews Cancer.

[32]  D. Sabatini,et al.  Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. , 2006, Molecular cell.

[33]  Steven M Kornblau,et al.  Simultaneous activation of multiple signal transduction pathways confers poor prognosis in acute myelogenous leukemia. , 2004, Blood.

[34]  E. Solary,et al.  Essential role for the p110δ isoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia , 2005 .

[35]  G. Laurent,et al.  Antileukemic activity of rapamycin in acute myeloid leukemia. , 2005, Blood.

[36]  D. Guertin,et al.  Phosphorylation and Regulation of Akt/PKB by the Rictor-mTOR Complex , 2005, Science.

[37]  E. Solary,et al.  Essential role for the p110delta isoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia. , 2005, Blood.

[38]  R. Loewith,et al.  Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive , 2004, Nature Cell Biology.

[39]  D. Guertin,et al.  Rictor, a Novel Binding Partner of mTOR, Defines a Rapamycin-Insensitive and Raptor-Independent Pathway that Regulates the Cytoskeleton , 2004, Current Biology.

[40]  M. Clemens,et al.  Targets and mechanisms for the regulation of translation in malignant transformation , 2004, Oncogene.

[41]  J. Avruch,et al.  Dissociation of raptor from mTOR is a mechanism of rapamycin‐induced inhibition of mTOR function , 2004, Genes to cells : devoted to molecular & cellular mechanisms.

[42]  J. Arthur,et al.  In vivo role of the PIF‐binding docking site of PDK1 defined by knock‐in mutation , 2003, The EMBO journal.

[43]  M. Carroll,et al.  Survival of acute myeloid leukemia cells requires PI3 kinase activation. , 2003, Blood.

[44]  J. Avruch,et al.  Raptor, a Binding Partner of Target of Rapamycin (TOR), Mediates TOR Action , 2002, Cell.

[45]  D. Sabatini,et al.  mTOR Interacts with Raptor to Form a Nutrient-Sensitive Complex that Signals to the Cell Growth Machinery , 2002, Cell.

[46]  S K Burley,et al.  Hierarchical phosphorylation of the translation inhibitor 4E-BP1. , 2001, Genes & development.

[47]  A. Gingras,et al.  Regulation of translation initiation by FRAP/mTOR. , 2001, Genes & development.

[48]  A. C. Maiyar,et al.  Serum and glucocorticoid‐inducible kinase (SGK) is a target of the PI 3‐kinase‐stimulated signaling pathway , 1999, The EMBO journal.

[49]  P. Cohen,et al.  Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. , 1999, The Biochemical journal.

[50]  A. Gingras,et al.  eIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation. , 1999, Annual review of biochemistry.