An overview of the mTOR pathway as a target in cancer therapy

Introduction: The mammalian target of rapamycin (mTOR) signaling cascade is a key regulatory pathway controlling initiation of mRNA translation in mammalian cells. The mTOR inhibitor rapamycin and its derivatives have shown potent antineoplastic activities in many preclinical models and clinical trials. First-generation mTOR inhibitors are now FDA-approved for the treatment of renal cell carcinoma. Areas covered: This article reviews the components of the mTOR pathway and their normal functions, highlighting the most common alterations in the pathway, seen in various human malignancies. It also discusses elements and effectors of this signaling cascade and reviews the therapeutic relevance of pharmacological inhibitors of the pathway in several malignancies, including lymphomas, leukemias, sarcomas, renal cell carcinoma, and breast cancer. Expert opinion: mTOR targeting is a highly promising therapeutic approach. First-generation mTOR inhibitors have already shown substantial activity in the treatment of certain tumors, while the emergence of second-generation catalytic mTOR inhibitors provides a better approach to target the pathway in malignant cells and has raised the potential for better clinical outcomes in the future.

[1]  J. Diamond,et al.  [Mechanisms of resistance to BCR-ABL kinase inhibitors]. , 2013, Acta medica portuguesa.

[2]  J. Tamburini,et al.  The dual mTORC1 and mTORC2 inhibitor AZD8055 has anti-tumor activity in acute myeloid leukemia , 2012, Leukemia.

[3]  M. Piccart,et al.  Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. , 2012, The New England journal of medicine.

[4]  K. Flatten,et al.  Dual mTORC1/mTORC2 inhibition diminishes Akt activation and induces Puma-dependent apoptosis in lymphoid malignancies. , 2012, Blood.

[5]  J. Castillo,et al.  CAL-101: a phosphatidylinositol-3-kinase p110-delta inhibitor for the treatment of lymphoid malignancies , 2012, Expert opinion on investigational drugs.

[6]  J. Stockman,et al.  Everolimus for Advanced Pancreatic Neuroendocrine Tumors , 2012 .

[7]  J. Blay,et al.  Phase II study of the mammalian target of rapamycin inhibitor ridaforolimus in patients with advanced bone and soft tissue sarcomas. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  Daniela Cilloni,et al.  Molecular Pathways: BCR-ABL , 2011, Clinical Cancer Research.

[9]  E. Dazert,et al.  mTOR signaling in disease. , 2011, Current opinion in cell biology.

[10]  J. Balko,et al.  Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[11]  V. Rodrik-Outmezguine,et al.  High-dose rapamycin induces apoptosis in human cancer cells by dissociating mTOR complex 1 and suppressing phosphorylation of 4E-BP1 , 2011, Cell cycle.

[12]  L. Platanias,et al.  Emerging roles for mammalian target of rapamycin inhibitors in the treatment of solid tumors and hematological malignancies , 2011, Current opinion in oncology.

[13]  A. Wiestner,et al.  New molecular targets in mantle cell lymphoma. , 2011, Seminars in cancer biology.

[14]  M. Hall,et al.  Rapamycin passes the torch: a new generation of mTOR inhibitors , 2011, Nature Reviews Drug Discovery.

[15]  D. Ettinger,et al.  A phase 2 study of temsirolimus (CCI‐779) in patients with soft tissue sarcomas , 2011, Cancer.

[16]  M. Atkins,et al.  Future directions in renal cell carcinoma: 2011 and beyond. , 2011, Hematology/oncology clinics of North America.

[17]  R. Motzer,et al.  mTOR inhibitors in advanced renal cell carcinoma. , 2011, Hematology/oncology clinics of North America.

[18]  L. Platanias,et al.  Targeting mTOR for the treatment of AML. New agents and new directions , 2011, Oncotarget.

[19]  M. Sanchez-Cespedes,et al.  The role of LKB1 in lung cancer , 2011, Familial Cancer.

[20]  A. Redig,et al.  Dual mTORC2/mTORC1 Targeting Results in Potent Suppressive Effects on Acute Myeloid Leukemia (AML) Progenitors , 2011, Clinical Cancer Research.

[21]  J. Carew,et al.  Mechanisms of mTOR inhibitor resistance in cancer therapy , 2011, Targeted Oncology.

[22]  Michael A. Choti,et al.  DAXX/ATRX, MEN1, and mTOR Pathway Genes Are Frequently Altered in Pancreatic Neuroendocrine Tumors , 2011, Science.

[23]  V. Zinzalla,et al.  Activation of mTORC2 by Association with the Ribosome , 2011, Cell.

[24]  H. Kantarjian,et al.  Front-line and salvage therapies with tyrosine kinase inhibitors and other treatments in chronic myeloid leukemia. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[25]  A. Redig,et al.  Regulation of mammalian target of rapamycin and mitogen activated protein kinase pathways by BCR–ABL , 2011, Leukemia & lymphoma.

[26]  H. Kantarjian,et al.  New developments in the treatment of chronic myeloid leukemia and Philadelphia-positive acute lymphoblastic leukemia , 2011, Leukemia & lymphoma.

[27]  Thomas Ernst,et al.  Impact of BCR-ABL mutations on patients with chronic myeloid leukemia , 2011, Cell cycle.

[28]  M. Talpaz,et al.  Seeking the causes and solutions to imatinib-resistance in chronic myeloid leukemia , 2011, Leukemia.

[29]  R. Ulrich,et al.  Clinical Safety and Activity In a Phase 1 Study of CAL-101, An Isoform-Selective Inhibitor of Phosphatidylinositol 3-Kinase P110δ, In Patients with Relapsed or Refractory Non-Hodgkin Lymphoma , 2010 .

[30]  N. Schmitz,et al.  CNS Disease In Younger Patients ( , 2010 .

[31]  Jiang Li,et al.  mTOR signaling is activated by FLT3 kinase and promotes survival of FLT3-mutated acute myeloid leukemia cells , 2010, Molecular Cancer.

[32]  S. Signoretti,et al.  The Efficacy of the Novel Dual PI3-Kinase/mTOR Inhibitor NVP-BEZ235 Compared with Rapamycin in Renal Cell Carcinoma , 2010, Clinical Cancer Research.

[33]  L. Platanias,et al.  Critical roles for mTORC2- and rapamycin-insensitive mTORC1-complexes in growth and survival of BCR-ABL-expressing leukemic cells , 2010, Proceedings of the National Academy of Sciences.

[34]  A. Nakashima,et al.  Single amino-acid changes that confer constitutive activation of mTOR are discovered in human cancer , 2010, Oncogene.

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

[36]  C. Croce,et al.  Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[37]  L. Platanias,et al.  Induction of autophagy by dual mTORC1-mTORC2 inhibition in BCR-ABL-expressing leukemic cells. , 2010, Autophagy.

[38]  A. Kibel Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial , 2009 .

[39]  B. Coiffier,et al.  Phase III study to evaluate temsirolimus compared with investigator's choice therapy for the treatment of relapsed or refractory mantle cell lymphoma. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[40]  J. McCubrey,et al.  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, Cancer research.

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

[42]  D. A. Foster,et al.  Regulation of mTORC1 and mTORC2 Complex Assembly by Phosphatidic Acid: Competition with Rapamycin , 2008, Molecular and Cellular Biology.

[43]  M. Ohh,et al.  Differential Dependence of Hypoxia-inducible Factors 1α and 2α on mTORC1 and mTORC2* , 2008, Journal of Biological Chemistry.

[44]  B. Manning,et al.  Common corruption of the mTOR signaling network in human tumors , 2008, Oncogene.

[45]  F. Callera,et al.  Lack of antileukemic activity of rapamycin in elderly patients with acute myeloid leukemia evolving from a myelodysplastic syndrome. , 2008, Leukemia research.

[46]  L. Cantley,et al.  PI3K pathway alterations in cancer: variations on a theme , 2008, Oncogene.

[47]  K. Shokat,et al.  Ablation of PI3K blocks BCR-ABL leukemogenesis in mice, and a dual PI3K/mTOR inhibitor prevents expansion of human BCR-ABL+ leukemia cells. , 2008, The Journal of clinical investigation.

[48]  R. Motzer,et al.  Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial , 2008, The Lancet.

[49]  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.

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

[51]  Wolfgang Link,et al.  The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications. , 2008, Current cancer drug targets.

[52]  L. Platanias,et al.  Exploiting the mammalian target of rapamycin pathway in hematologic malignancies , 2008, Current opinion in hematology.

[53]  O. Haas,et al.  Evaluation of antileukaemic effects of rapamycin in patients with imatinib‐resistant chronic myeloid leukaemia , 2007, European journal of clinical investigation.

[54]  M. Konopleva,et al.  The dual PI3 kinase/mTOR inhibitor PI-103 prevents p53 induction by Mdm2 inhibition but enhances p53-mediated mitochondrial apoptosis in p53 wild-type AML , 2008, Leukemia.

[55]  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.

[56]  Robert T Abraham,et al.  Targeting the mTOR signaling network in cancer. , 2007, Trends in molecular medicine.

[57]  E. Campo,et al.  Genetic and molecular pathogenesis of mantle cell lymphoma: perspectives for new targeted therapeutics , 2007, Nature Reviews Cancer.

[58]  David McDermott,et al.  Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. , 2007, The New England journal of medicine.

[59]  N. Hay,et al.  The two TORCs and Akt. , 2007, Developmental cell.

[60]  James Brugarolas,et al.  Renal-cell carcinoma--molecular pathways and therapies. , 2007, The New England journal of medicine.

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

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

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

[64]  Gordon B Mills,et al.  mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. , 2006, Cancer research.

[65]  N. Sonenberg,et al.  mTOR signaling: implications for cancer and anticancer therapy , 2005, British Journal of Cancer.

[66]  Steven P. Gygi,et al.  mTOR and S6K1 Mediate Assembly of the Translation Preinitiation Complex through Dynamic Protein Interchange and Ordered Phosphorylation Events , 2005, Cell.

[67]  F. Khuri,et al.  Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. , 2005, Cancer research.

[68]  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.

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

[70]  N. Sonenberg,et al.  Upstream and downstream of mTOR. , 2004, Genes & development.

[71]  T. Golub,et al.  mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways , 2004, Nature Medicine.

[72]  W. Kaelin,et al.  Inhibition of HIF2α Is Sufficient to Suppress pVHL-Defective Tumor Growth , 2003, PLoS biology.

[73]  L. Cantley,et al.  Targeting the PI3K-Akt pathway in human cancer: rationale and promise. , 2003, Cancer cell.

[74]  P. Meltzer,et al.  Mechanisms of sarcoma development , 2003, Nature Reviews Cancer.

[75]  J. Radich,et al.  The role of FLT3 in haematopoietic malignancies , 2003, Nature Reviews Cancer.

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

[77]  D. A. Foster,et al.  Phospholipase D confers rapamycin resistance in human breast cancer cells , 2003, Oncogene.

[78]  Shile Huang,et al.  Rapamycins: Mechanisms of Action and Cellular Resistance , 2003, Cancer biology & therapy.

[79]  Jie Chen,et al.  Phosphatidic Acid-Mediated Mitogenic Activation of mTOR Signaling , 2001, Science.

[80]  J. Melo,et al.  The molecular biology of chronic myeloid leukemia. , 2000, Blood.

[81]  S. Gygi,et al.  Regulation of 4E-BP1 phosphorylation: a novel two-step mechanism. , 1999, Genes & development.

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

[83]  M. Birnbaum,et al.  Insulin Increases the Association of Akt-2 with Glut4-containing Vesicles* , 1998, The Journal of Biological Chemistry.

[84]  S Povey,et al.  Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. , 1997, Science.

[85]  S. Schreiber,et al.  Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[86]  Stuart L. Schreiber,et al.  A mammalian protein targeted by G1-arresting rapamycin–receptor complex , 1994, Nature.

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

[88]  S. Sehgal,et al.  Rapamycin (AY-22,989), a new antifungal antibiotic. II. Fermentation, isolation and characterization. , 1975, The Journal of antibiotics.

[89]  S. Sehgal,et al.  Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. , 1975, The Journal of antibiotics.