Activation of mammalian target of rapamycin signaling pathway contributes to tumor cell survival in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma.

Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALCL) frequently carries the t(2;5)(p23;q35) resulting in aberrant expression of chimeric nucleophosmin-ALK. Previously, nucleophosmin-ALK has been shown to activate phosphatidylinositol 3-kinase (PI3K) and its downstream effector, the serine/threonine kinase AKT. In this study, we hypothesized that the mammalian target of rapamycin (mTOR) pathway, which functions downstream of AKT, mediates the oncogenic effects of activated PI3K/AKT in ALK+ ALCL. Here, we provide evidence that mTOR signaling phosphoproteins, including mTOR, eukaryotic initiation factor 4E-binding protein-1, p70S6K, and ribosomal protein S6, are highly phosphorylated in ALK+ ALCL cell lines and tumors. We also show that AKT activation contributes to mTOR phosphorylation, at least in part, as forced expression of constitutively active AKT by myristoylated AKT adenovirus results in increased phosphorylation of mTOR and its downstream effectors. Conversely, inhibition of AKT expression or activity results in decreased mTOR phosphorylation. In addition, pharmacologic inhibition of PI3K/AKT down-regulates the activation of the mTOR signaling pathway. We also show that inhibition of mTOR with rapamycin, as well as silencing mTOR gene product expression using mTOR-specific small interfering RNA, decreased phosphorylation of mTOR signaling proteins and induced cell cycle arrest and apoptosis in ALK+ ALCL cells. Cell cycle arrest was associated with modulation of G(1)-S-phase regulators, including the cyclin-dependent kinase inhibitors p21(waf1) and p27(kip1). Apoptosis following inhibition of mTOR expression or function was associated with down-regulation of antiapoptotic proteins, including c-FLIP, MCL-1, and BCL-2. These findings suggest that the mTOR pathway contributes to nucleophosmin-ALK/PI3K/AKT-mediated tumorigenesis and that inhibition of mTOR represents a potential therapeutic strategy in ALK+ ALCL.

[1]  T. McDonnell,et al.  Apoptotic rate in peripheral T-cell lymphomas. A study using a tissue microarray with validation on full tissue sections. , 2002, American Journal of Clinical Pathology.

[2]  M. Wasik,et al.  Role of phosphatidylinositol 3-kinase-Akt pathway in nucleophosmin/anaplastic lymphoma kinase-mediated lymphomagenesis. , 2001, Cancer research.

[3]  M. Torbenson,et al.  mTOR and P70 S6 Kinase Expression in Primary Liver Neoplasms , 2004, Clinical Cancer Research.

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

[5]  P. Houghton,et al.  Arrest and Determine the Cellular Response to Rapamycin 1 Cooperate in Enforcing Rapamycin-induced G CIP 1 p 53 / p 21 Updated , 2001 .

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

[7]  T. Harris,et al.  TOR Signaling , 2003, Science's STKE.

[8]  Stefano Fumagalli,et al.  Disruption of the p70s6k/p85s6k gene reveals a small mouse phenotype and a new functional S6 kinase , 1998, The EMBO journal.

[9]  Robert T. Abraham,et al.  Phosphorylation of Mammalian Target of Rapamycin (mTOR) at Ser-2448 Is Mediated by p70S6 Kinase* , 2005, Journal of Biological Chemistry.

[10]  M. Mann,et al.  p70S6 kinase signals cell survival as well as growth, inactivating the pro-apoptotic molecule BAD , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[11]  S. Morris,et al.  Nucleophosmin-anaplastic lymphoma kinase associated with anaplastic large-cell lymphoma activates the phosphatidylinositol 3-kinase/Akt antiapoptotic signaling pathway. , 2000, Blood.

[12]  Christine C. Hudson,et al.  A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. , 2000, Cancer research.

[13]  S. Bodine,et al.  Control of Ser2448 Phosphorylation in the Mammalian Target of Rapamycin by Insulin and Skeletal Muscle Load* , 2002, The Journal of Biological Chemistry.

[14]  D N Shapiro,et al.  Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. , 1994, Science.

[15]  J. Blenis,et al.  Identification of S6 Kinase 1 as a Novel Mammalian Target of Rapamycin (mTOR)-phosphorylating Kinase* , 2005, Journal of Biological Chemistry.

[16]  A. Gingras,et al.  The mRNA 5' cap-binding protein eIF4E and control of cell growth. , 1998, Current opinion in cell biology.

[17]  J. Blenis,et al.  TOS Motif-Mediated Raptor Binding Regulates 4E-BP1 Multisite Phosphorylation and Function , 2003, Current Biology.

[18]  S. Sehgal,et al.  Activity of rapamycin (AY-22,989) against transplanted tumors. , 1984, The Journal of antibiotics.

[19]  S. Morris,et al.  Translocations involving anaplastic lymphoma kinase (ALK) , 2001, Oncogene.

[20]  J. Douros,et al.  New antitumor substances of natural origin. , 1981, Cancer treatment reviews.

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

[22]  S. Lowe,et al.  Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy , 2004, Nature.

[23]  S. Schreiber,et al.  Erratum: Control of p70 S6 kinase by kinase activity of FRAP in vivo (Nature (1995) 377 (441-446)) , 1995 .

[24]  B. Woda,et al.  Expression of the eukaryotic translation initiation factors 4E and 2alpha in non-Hodgkin's lymphomas. , 1999, The American journal of pathology.

[25]  L. Bowman,et al.  Insulin stimulates the translation of ribosomal proteins and the transcription of rDNA in mouse myoblasts. , 1988, The Journal of biological chemistry.

[26]  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.

[27]  G. Rassidakis,et al.  Inhibition of Akt increases p27Kip1 levels and induces cell cycle arrest in anaplastic large cell lymphoma. , 2005, Blood.

[28]  J. Crespo,et al.  Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. , 2002, Molecular cell.

[29]  T. McDonnell,et al.  Expression of p27Kip1 and c-Jun Activation Binding Protein 1 Are Inversely Correlated in Systemic Anaplastic Large Cell Lymphoma , 2003 .

[30]  N. Sonenberg,et al.  Ras mediates translation initiation factor 4E-induced malignant transformation. , 1992, Genes & development.

[31]  J. Chen,et al.  Cytoplasmic-nuclear shuttling of FKBP12-rapamycin-associated protein is involved in rapamycin-sensitive signaling and translation initiation. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[32]  S. Schreiber,et al.  Control of p70 S6 kinase by kinase activity of FRAP in vivo , 1995, Nature.

[33]  J. Testa,et al.  Cell Cycle Withdrawal Promotes Myogenic Induction of Akt, a Positive Modulator of Myocyte Survival , 1999, Molecular and Cellular Biology.

[34]  M. Polymenis,et al.  An essential E box in the promoter of the gene encoding the mRNA cap-binding protein (eukaryotic initiation factor 4E) is a target for activation by c-myc , 1996, Molecular and cellular biology.

[35]  T. McDonnell,et al.  Expression of p27(Kip1) and c-Jun activation binding protein 1 are inversely correlated in systemic anaplastic large cell lymphoma. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[36]  Tobias Schmelzle,et al.  TOR, a Central Controller of Cell Growth , 2000, Cell.

[37]  R. Fonseca,et al.  Phase II trial of single-agent temsirolimus (CCI-779) for relapsed mantle cell lymphoma. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[38]  Jennifer Skeen,et al.  Dwarfism, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. , 2003, Genes & development.

[39]  N. Sonenberg,et al.  Cell cycle-dependent translational control , 2000 .

[40]  C. Sawyers,et al.  The phosphatidylinositol 3-Kinase–AKT pathway in human cancer , 2002, Nature Reviews Cancer.

[41]  M. Bjornsti,et al.  The tor pathway: a target for cancer therapy , 2004, Nature Reviews Cancer.

[42]  V. Brown,et al.  Rapamycin is active against B-precursor leukemia in vitro and in vivo, an effect that is modulated by IL-7-mediated signaling , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[43]  S. Snyder,et al.  RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[44]  E. Berg,et al.  World Health Organization Classification of Tumours , 2002 .

[45]  J. Blenis,et al.  Identification of a Conserved Motif Required for mTOR Signaling , 2002, Current Biology.

[46]  O. Meyuhas Synthesis of the translational apparatus is regulated at the translational level. , 2000, European journal of biochemistry.

[47]  I. Mellinghoff,et al.  AKT Activity Determines Sensitivity to Mammalian Target of Rapamycin (mTOR) Inhibitors by Regulating Cyclin D1 and c-myc Expression* , 2004, Journal of Biological Chemistry.

[48]  G. Thomas,et al.  The modular phosphorylation and activation of p70s6k , 1997, FEBS letters.