Beyond rapalog therapy: preclinical pharmacology and antitumor activity of WYE-125132, an ATP-competitive and specific inhibitor of mTORC1 and mTORC2.

The mammalian target of rapamycin (mTOR) is a major component of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway that is dysregulated in 50% of all human malignancies. Rapamycin and its analogues (rapalogs) partially inhibit mTOR through allosteric binding to mTOR complex 1 (mTORC1) but not mTOR complex 2 (mTORC2), an emerging player in cancer. Here, we report WYE-125132 (WYE-132), a highly potent, ATP-competitive, and specific mTOR kinase inhibitor (IC(50): 0.19 +/- 0.07 nmol/L; >5,000-fold selective versus PI3Ks). WYE-132 inhibited mTORC1 and mTORC2 in diverse cancer models in vitro and in vivo. Importantly, consistent with genetic ablation of mTORC2, WYE-132 targeted P-AKT(S473) and AKT function without significantly reducing the steady-state level of the PI3K/PDK1 activity biomarker P-AKT(T308), highlighting a prominent and direct regulation of AKT by mTORC2 in cancer cells. Compared with the rapalog temsirolimus/CCI-779, WYE-132 elicited a substantially stronger inhibition of cancer cell growth and survival, protein synthesis, cell size, bioenergetic metabolism, and adaptation to hypoxia. Oral administration of WYE-132 to tumor-bearing mice showed potent single-agent antitumor activity against MDA361 breast, U87MG glioma, A549 and H1975 lung, as well as A498 and 786-O renal tumors. An optimal dose of WYE-132 achieved a substantial regression of MDA361 and A549 large tumors and caused complete regression of A498 large tumors when coadministered with bevacizumab. Our results further validate mTOR as a critical driver for tumor growth, establish WYE-132 as a potent and profound anticancer agent, and provide a strong rationale for clinical development of specific mTOR kinase inhibitors as new cancer therapy.

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

[2]  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.

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

[4]  D. Guertin,et al.  The Pharmacology of mTOR Inhibition , 2009, Science Signaling.

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

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

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

[8]  데이빗 제임스 리차드,et al.  Thienopyrimidine and pyrazolopyrimidine compounds and their use as mtor kinase and pi3 kinase inhibitors , 2008 .

[9]  W. Sellers,et al.  Drug discovery approaches targeting the PI3K/Akt pathway in cancer , 2008, Oncogene.

[10]  Pier Paolo Pandolfi,et al.  The PTEN–PI3K pathway: of feedbacks and cross-talks , 2008, Oncogene.

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

[12]  P. Pandolfi,et al.  Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. , 2008, The Journal of clinical investigation.

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

[14]  Guido Kroemer,et al.  Tumor cell metabolism: cancer's Achilles' heel. , 2008, Cancer cell.

[15]  R. Abraham,et al.  A new pharmacologic action of CCI-779 involves FKBP12-independent inhibition of mTOR kinase activity and profound repression of global protein synthesis. , 2008, Cancer research.

[16]  L. Toral-Barza,et al.  Synthesis and structure-activity relationships of ring-opened 17-hydroxywortmannins: potent phosphoinositide 3-kinase inhibitors with improved properties and anticancer efficacy. , 2008, Journal of medicinal chemistry.

[17]  W. Rathmell,et al.  VHL inactivation in renal cell carcinoma: implications for diagnosis, prognosis and treatment , 2008, Expert review of anticancer therapy.

[18]  R. Deberardinis,et al.  The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. , 2008, Cell metabolism.

[19]  R. Abraham,et al.  Discovery of lactoquinomycin and related pyranonaphthoquinones as potent and allosteric inhibitors of AKT/PKB: mechanistic involvement of AKT catalytic activation loop cysteines , 2007, Molecular Cancer Therapeutics.

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

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

[22]  R. Abraham,et al.  The Mammalian Target of Rapamycin Signaling Pathway: Twists and Turns in the Road to Cancer Therapy , 2007, Clinical Cancer Research.

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

[24]  S. Signoretti,et al.  The Role of Mammalian Target of Rapamycin Inhibitors in the Treatment of Advanced Renal Cancer , 2007, Clinical Cancer Research.

[25]  D. Guertin,et al.  Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. , 2006, Developmental cell.

[26]  P. Houghton,et al.  mTOR and cancer therapy , 2006, Oncogene.

[27]  J. Qin,et al.  SIN1/MIP1 Maintains rictor-mTOR Complex Integrity and Regulates Akt Phosphorylation and Substrate Specificity , 2006, Cell.

[28]  J. Dancey Therapeutic targets: MTOR and related pathways , 2006, Cancer biology & therapy.

[29]  William A Weiss,et al.  A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. , 2006, Cancer cell.

[30]  J. Shabanowitz,et al.  mTOR‐dependent stimulation of the association of eIF4G and eIF3 by insulin , 2006, The EMBO journal.

[31]  M. Hall,et al.  TOR Signaling in Growth and Metabolism , 2006, Cell.

[32]  I. Mellinghoff,et al.  Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer , 2006, Nature Medicine.

[33]  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.

[34]  J. Gibbons,et al.  Characterization of the cloned full-length and a truncated human target of rapamycin: activity, specificity, and enzyme inhibition as studied by a high capacity assay. , 2005, Biochemical and biophysical research communications.

[35]  C. Hauser,et al.  Antitumor activity of rapamycin in a transgenic mouse model of ErbB2-dependent human breast cancer. , 2005, Cancer research.

[36]  P. Frost,et al.  PWT-458, a novel pegylated-17-hydroxywortmannin, inhibits phosphatidylinositol 3-kinase signaling and suppresses growth of solid tumors , 2005, Cancer biology & therapy.

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

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

[39]  M. Lazar,et al.  Mitochondrial remodeling in adipose tissue associated with obesity and treatment with rosiglitazone. , 2004, The Journal of clinical investigation.

[40]  Robert T Abraham,et al.  PI 3-kinase related kinases: 'big' players in stress-induced signaling pathways. , 2004, DNA repair.

[41]  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.

[42]  Kun-Liang Guan,et al.  Dysregulation of the TSC-mTOR pathway in human disease , 2004, Nature Genetics.

[43]  T. Hunter,et al.  The Protein Kinase Complement of the Human Genome , 2002, Science.

[44]  R. Abraham,et al.  The Rapamycin-Binding Domain Governs Substrate Selectivity by the Mammalian Target of Rapamycin , 2002, Molecular and Cellular Biology.

[45]  J. Blenis,et al.  Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. , 2002, Genes & development.

[46]  G. Koehl,et al.  Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor , 2002, Nature Medicine.

[47]  G. Semenza,et al.  Advances in Brief Modulation of Hypoxia-inducible Factor 1 a Expression by the Epidermal Growth Factor / Phosphatidylinositol 3-Kinase / PTEN / AKT / FRAP Pathway in Human Prostate Cancer Cells : Implications for Tumor Angiogenesis and Therapeutics 1 , 2000 .

[48]  J. Clardy,et al.  Refined structure of the FKBP12-rapamycin-FRB ternary complex at 2.2 A resolution. , 1999, Acta crystallographica. Section D, Biological crystallography.

[49]  Christine C. Hudson,et al.  Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin. , 1997, Science.

[50]  P. Cohen,et al.  Mechanism of activation of protein kinase B by insulin and IGF‐1. , 1996, The EMBO journal.

[51]  Jan E. Schnitzer,et al.  Role of GTP Hydrolysis in Fission of Caveolae Directly from Plasma Membranes , 1996, Science.

[52]  Stuart L. Schreiber,et al.  Structure of the FKBP12-Rapamycin Complex Interacting with Binding Domain of Human FRAP , 1996, Science.