Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The tor pathway: a target for cancer therapy

Key Points

  • Target of rapamycin (TOR) — an essential protein that is conserved in eukaryotes — directly or indirectly regulates the translation of ribosomal proteins and, in yeast, regulates ribosome biogenesis.

  • TOR controls cap-dependent translation initiation through phosphorylating and inactivating eukaryotic initiation factor 4E binding proteins, which allows formation of the eIF4F complex that is required for translation initiation of mRNAs that have long structured 5′-untranslated regions.

  • TOR functions as a sensor of mitogen, energy and nutritient levels, acting as a gatekeeper for cell-cycle progression from G1 to S phase.

  • Pathways that regulate TOR signalling are complex and involve positive regulators such as AKT that phosphorylate and inactivate negative regulators such as tuberin (TSC2).

  • Pathways upstream of TOR are frequently activated in cancer. This can be through increased activity of phosphatidylinositol-3-kinase–AKT or kinases that regulate TSC2, or through mutations that inactivate TSC proteins.

  • The TOR pathway is also upregulated in many human cancers and oncogenic transformation might sensitize cells to TOR inhibitors. TOR therefore represents a novel therapeutic target.

  • Rapamycin and its analogues are highly specific inhibitors of TOR and are now in Phase I–III oncology clinical trials.

Abstract

Rapamycin, a macrocyclic lactone, is a highly specific inhibitor of the serine/threonine protein kinase target of rapamycin (TOR). Although it is clear that TOR controls initiation of protein translation, recent results indicate that TOR is a central controller, integrating a plethora of signalling pathways that respond to growth factors and nutritional status. In addition to the role of rapamycin as an immune suppressant, emerging data indicate that genetic and metabolic changes accompanying malignant transformation might cause hypersensitivity to TOR inhibition.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Target of rapamycin is a central regulator of cell growth and proliferation in response to environmental and nutritional conditions.
Figure 2: Domain structure of target of rapamycin.
Figure 3: Regulation of target of rapamycin signalling by raptor.
Figure 4: The target of rapamycin signalling pathway.
Figure 5: Rapamycin can induce apoptosis.
Figure 6: Increased expression of eIF4E is implicated in transformation.

Similar content being viewed by others

References

  1. Sehgal, S. N., Baker, H. & Vezina, C. Rapamycin (AY-22,989), a new antifungal antibiotic. II. Fermentation, isolation and characterization. J. Antibiot. 28, 727–732 (1975).

    Article  CAS  Google Scholar 

  2. Vezina, C., Kudelski, A. & Sehgal, S. N. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J. Antibiot. 28, 721–726 (1975).

    CAS  Google Scholar 

  3. Sehgal, S. N. Rapamune (RAPA, rapamycin, sirolimus): mechanism of action immunosuppressive effect results from blockade of signal transduction and inhibition of cell cycle progression. Clin. Biochem. 31, 335–340 (1998).

    CAS  PubMed  Google Scholar 

  4. Calne, R. Y. et al. Rapamycin for immunosuppression in organ allografting. Lancet 2, 227 (1989).

    CAS  PubMed  Google Scholar 

  5. Schreiber, S. L. Chemistry and biology of the immunophilins and their immunosuppressive ligands. Science 251, 283–287 (1991).

    CAS  PubMed  Google Scholar 

  6. Pohanka, E. New immunosuppressive drugs: an update. Curr. Opin. Urol. 11, 143–151 (2001).

    CAS  PubMed  Google Scholar 

  7. Saunders, R. N., Metcalfe, M. S. & Nicholson, M. L. Rapamycin in transplantation: a review of the evidence. Kidney Int. 59, 3–16 (2001).

    CAS  PubMed  Google Scholar 

  8. Harris, T. E. & Lawrence, J. C. Jr. TOR signaling. Sci. STKE 9 Dec 2003 (doi: 10.1126/stke.2122003re15).

  9. Brown, E. J. et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 369, 756–758 (1994).

    CAS  PubMed  Google Scholar 

  10. Gingras, A. C., Raught, B. & Sonenberg, N. Regulation of translation initiation by FRAP/mTOR. Genes Dev. 15, 807–826 (2001).

    CAS  PubMed  Google Scholar 

  11. Jacinto, E. & Hall, M. N. Tor signalling in bugs, brain and brawn. Nature Rev. Mol. Cell Biol. 4, 117–126 (2003).

    CAS  Google Scholar 

  12. Abraham, R. T. Identification of TOR signaling complexes: more TORC for the cell growth engine. Cell 111, 9–12 (2002).

    CAS  PubMed  Google Scholar 

  13. Hentges, K. E. et al. FRAP/mTOR is required for proliferation and patterning during embryonic development in the mouse. Proc. Natl Acad. Sci. USA 98, 13796–13801 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hentges, K., Thompson, K. & Peterson, A. The flat-top gene is required for the expansion and regionalization of the telencephalic primordium. Development 126, 1601–1609 (1999).

    CAS  PubMed  Google Scholar 

  15. Cardenas, M. E., Cutler, N. S., Lorenz, M. C., Di Como, C. J. & Heitman, J. The TOR signaling cascade regulates gene expression in response to nutrients. Genes Dev. 13, 3271–3279 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hardwick, J. S., Kuruvilla, F. G., Tong, J. K., Shamji, A. F. & Schreiber, S. L. Rapamycin-modulated transcription defines the subset of nutrient-sensitive signaling pathways directly controlled by the Tor proteins. Proc. Natl Acad. Sci. USA 96, 14866–14870 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Powers, T. & Walter, P. Regulation of ribosome biogenesis by the rapamycin-sensitive TOR-signaling pathway in Saccharomyces cerevisiae. Mol. Biol. Cell 10, 987–1000 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Kim, D. H. et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110, 163–175 (2002).

    CAS  PubMed  Google Scholar 

  19. Hara, K. et al. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 110, 177–189 (2002). References 18 and 19 report the identification of raptor as a 150-kDa TOR-binding protein that also binds 4E-BP1 and S6K1. The binding of raptor to TOR is necessary for the TOR-catalysed phosphorylation of 4E-BP1 in vitro and strongly enhances the TOR kinase activity towards p70α. So, raptor is an essential scaffold for the TOR-catalysed phosphorylation of 4E-BP1 and mediates TOR action in vivo.

    CAS  PubMed  Google Scholar 

  20. Loewith, R. et al. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 10, 457–468 (2002).

    CAS  PubMed  Google Scholar 

  21. Kim, D. H. et al. GβL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR. Mol. Cell 11, 895–904 (2003). References 20 and 21 identified GβL (the homologue of yeast Lst8) as a modulator of TOR kinase activity, stabilizing the interaction of raptor with TOR. The binding of GβL to TOR strongly stimulates the kinase activity of TOR towards S6K1 and 4E-BP1, an effect that is reversed by the stable interaction of raptor with TOR.

    CAS  PubMed  Google Scholar 

  22. Schalm, S. S. & Blenis, J. Identification of a conserved motif required for mTOR signaling. Curr. Biol. 12, 632–639 (2002).

    CAS  PubMed  Google Scholar 

  23. Schalm, S. S., Fingar, D. C., Sabatini, D. M. & Blenis, J. TOS Motif-mediated raptor binding regulates 4E-BP1 multisite phosphorylation and function. Curr. Biol. 13, 797–806 (2003).

    CAS  PubMed  Google Scholar 

  24. Rajan, P., Panchision, D. M., Newell, L. F. & McKay, R. D. BMPs signal alternately through a SMAD or FRAP–STAT pathway to regulate fate choice in CNS stem cells. J. Cell Biol. 161, 911–921 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Coolican, S. A., Samuel, D. S., Ewton, D. Z., McWade, F. J. & Florini, J. R. The mitogenic and myogenic actions of insulin-like growth factors utilize distinct signaling pathways. J. Biol. Chem. 272, 6653–6662 (1997).

    CAS  PubMed  Google Scholar 

  26. Shu, L., Zhang, X. & Houghton, P. J. Myogenic differentiation is dependent on both the kinase function and the N-terminal sequence of mammalian target of rapamycin. J. Biol. Chem. 277, 16726–16732 (2002).

    CAS  PubMed  Google Scholar 

  27. Erbay, E. & Chen, J. The mammalian target of rapamycin regulates C2C12 myogenesis via a kinase-independent mechanism. J. Biol. Chem. 276, 36079–36082 (2001).

    CAS  PubMed  Google Scholar 

  28. Martin, K. A. et al. The mTOR/p70 S6K1 pathway regulates vascular smooth muscle cell differentiation. Am. J. Physiol. Cell Physiol. 286, C507–C17 (2004).

    CAS  PubMed  Google Scholar 

  29. Drenan, R. M., Liu, X., Bertram, P. G. & Zheng, X. F. FRAP/mTOR localization in the ER and the golgi apparatus. J. Biol. Chem. 24, 24 (2003).

    Google Scholar 

  30. Kim, J. E. & Chen, J. Cytoplasmic–nuclear shuttling of FKBP12-rapamycin-associated protein is involved in rapamycin-sensitive signaling and translation initiation. Proc. Natl Acad. Sci. USA 97, 14340–14345 (2000). This study reveals a novel regulatory mechanism, which involves cytoplasmic–nuclear shuttling of TOR. Results indicate that TOR is a cytoplasmic–nuclear shuttling protein and uncover a function for the nucleus in the direct regulation of the protein-synthesis machinery.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, X., Shu, L., Hosoi, H., Murti, K. G. & Houghton, P. J. Predominant nuclear localization of mammalian target of rapamycin in normal and malignant cells in culture. J. Biol. Chem. 277, 28127–28134 (2002).

    CAS  PubMed  Google Scholar 

  32. Tirado, O. M., Mateo-Lozano, S., Sanders, S., Dettin, L. E. & Notario, V. The PCPH oncoprotein antagonizes the proapoptotic role of the mammalian target of rapamycin in the response of normal fibroblasts to ionizing radiation. Cancer Res. 63, 6290–6298 (2003).

    CAS  PubMed  Google Scholar 

  33. Sabatini, D. M. et al. Interaction of RAFT1 with gephyrin required for rapamycin-sensitive signaling. Science 284, 1161–1164 (1999).

    CAS  PubMed  Google Scholar 

  34. Tang, S. J. et al. A rapamycin-sensitive signaling pathway contributes to long-term synaptic plasticity in the hippocampus. Proc. Natl Acad. Sci. USA 99, 467–472 (2002).

    CAS  PubMed  Google Scholar 

  35. Lawrence, J. C., Lin, T. A., McMahon, L. P. & Choi, K. M. Modulation of the protein kinase activity of mTOR. Curr. Top. Microbiol. Immunol. 279, 199–213 (2004).

    CAS  PubMed  Google Scholar 

  36. Dan, H. C. et al. Phosphatidylinositol 3-kinase/Akt pathway regulates tuberous sclerosis tumor suppressor complex by phosphorylation of tuberin. J. Biol. Chem. 277, 35364–35370 (2002).

    CAS  PubMed  Google Scholar 

  37. Inoki, K., Li, Y., Zhu, T., Wu, J. & Guan, K. L. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nature Cell Biol. 4, 648–657 (2002).

    CAS  PubMed  Google Scholar 

  38. Potter, C. J., Pedraza, L. G. & Xu, T. Akt regulates growth by directly phosphorylating Tsc2. Nature Cell Biol. 4, 658–665 (2002).

    CAS  PubMed  Google Scholar 

  39. Manning, B. D., Tee, A. R., Logsdon, M. N., Blenis, J. & Cantley, L. C. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol. Cell 10, 151–162 (2002).

    CAS  PubMed  Google Scholar 

  40. Gao, X. et al. Tsc tumour suppressor proteins antagonize amino-acid-TOR signalling. Nature Cell Biol. 4, 699–704 (2002).

    CAS  PubMed  Google Scholar 

  41. Tee, A. R. et al. Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc. Natl Acad. Sci. USA 99, 13571–13576 (2002). References 37–41 show that TSC1–TSC2 inhibits S6K1 and activates 4E-BP1. These functions of TSC1–TSC2 are mediated by inhibiting TOR. Furthermore, TSC2 is directly phosphorylated and inactivated by AKT. Phosphorylation destabilizes TSC2 and disrupts its interaction with TSC1.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang, H. et al. Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3K-Akt signaling through downregulation of PDGFR. J. Clin. Invest. 112, 1223–1233 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Tee, A. R., Anjum, R. & Blenis, J. Inactivation of the tuberous sclerosis complex-1 and-2 gene products occurs by phosphoinositide 3-kinase/Akt-dependent and-independent phosphorylation of tuberin. J. Biol. Chem. 278, 37288–37296 (2003).

    CAS  PubMed  Google Scholar 

  44. Potter, C. J., Huang, H. & Xu, T. Drosophila Tsc1 functions with Tsc2 to antagonize insulin signaling in regulating cell growth, cell proliferation, and organ size. Cell 105, 357–368 (2001).

    CAS  PubMed  Google Scholar 

  45. Nellist, M. et al. Identification and characterization of the interaction between tuberin and 14-3-3ζ. J. Biol. Chem. 277, 39417–39424 (2002).

    CAS  PubMed  Google Scholar 

  46. Li, Y., Inoki, K., Yeung, R. & Guan, K. L. Regulation of TSC2 by 14-3-3 binding. J. Biol. Chem. 277, 44593–44596 (2002).

    CAS  PubMed  Google Scholar 

  47. Liu, M. Y., Cai, S., Espejo, A., Bedford, M. T. & Walker, C. L. 14-3-3 interacts with the tumor suppressor tuberin at Akt phosphorylation site(s). Cancer Res. 62, 6475–6480 (2002).

    CAS  PubMed  Google Scholar 

  48. Shumway, S. D., Li, Y. & Xiong, Y. 14-3-3β binds to and negatively regulates the tuberous sclerosis complex 2 (TSC2) tumor suppressor gene product, tuberin. J. Biol. Chem. 278, 2089–2092 (2003).

    CAS  PubMed  Google Scholar 

  49. Radimerski, T., Montagne, J., Hemmings-Mieszczak, M. & Thomas, G. Lethality of Drosophila lacking TSC tumor suppressor function rescued by reducing dS6K signaling. Genes Dev. 16, 2627–2632 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Jaeschke, A. et al. Tuberous sclerosis complex tumor suppressor-mediated S6 kinase inhibition by phosphatidylinositide-3-OH kinase is mTOR independent. J. Cell Biol. 159, 217–224 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Inoki, K., Zhu, T. & Guan, K. L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003).

    CAS  PubMed  Google Scholar 

  52. Wienecke, R., Konig, A. & DeClue, J. E. Identification of tuberin, the tuberous sclerosis-2 product. Tuberin possesses specific Rap1GAP activity. J. Biol. Chem. 270, 16409–16414 (1995).

    CAS  PubMed  Google Scholar 

  53. Xiao, G. H., Shoarinejad, F., Jin, F., Golemis, E. A. & Yeung, R. S. The tuberous sclerosis 2 gene product, tuberin, functions as a Rab5 GTPase activating protein (GAP) in modulating endocytosis. J. Biol. Chem. 272, 6097–6100 (1997).

    CAS  PubMed  Google Scholar 

  54. Astrinidis, A. et al. Tuberin, the tuberous sclerosis complex 2 tumor suppressor gene product, regulates Rho activation, cell adhesion and migration. Oncogene 21, 8470–8476 (2002).

    CAS  PubMed  Google Scholar 

  55. Zhang, Y. et al. Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nature Cell Biol. 5, 578–581 (2003).

    CAS  PubMed  Google Scholar 

  56. Tee, A. R., Manning, B. D., Roux, P. P., Cantley, L. C. & Blenis, J. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr. Biol. 13, 1259–1268 (2003). References 55 and 56 show that the small GTPase RHEB is a direct target of TSC2 GAP activity both in vivo and in vitro . These studies identify RHEB as a molecular target of the TSC tumour suppressors.

    CAS  PubMed  Google Scholar 

  57. Stocker, H. et al. Rheb is an essential regulator of S6K in controlling cell growth in Drosophila. Nature Cell Biol. 5, 559–565 (2003).

    CAS  PubMed  Google Scholar 

  58. Saucedo, L. J. et al. Rheb promotes cell growth as a component of the insulin/TOR signalling network. Nature Cell Biol. 5, 566–571 (2003).

    CAS  PubMed  Google Scholar 

  59. Volarevic, S. & Thomas, G. Role of S6 phosphorylation and S6 kinase in cell growth. Prog. Nucleic Acid Res. Mol. Biol. 65, 101–127 (2001).

    CAS  PubMed  Google Scholar 

  60. Shah, O. J., Anthony, J. C., Kimball, S. R. & Jefferson, L. S. 4E-BP1 and S6K1: translational integration sites for nutritional and hormonal information in muscle. Am. J. Physiol. Endocrinol. Metab. 279, E715–E729 (2000).

    CAS  PubMed  Google Scholar 

  61. Pullen, N. et al. Phosphorylation and activation of p70s6k by PDK1. Science 279, 707–710 (1998).

    CAS  PubMed  Google Scholar 

  62. Dennis, P. B., Pullen, N., Kozma, S. C. & Thomas, G. The principal rapamycin-sensitive p70(s6k) phosphorylation sites, T-229 and T-389, are differentially regulated by rapamycin-insensitive kinase kinases. Mol. Cell Biol. 16, 6242–6251 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Burnett, P. E., Barrow, R. K., Cohen, N. A., Snyder, S. H. & Sabatini, D. M. RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1. Proc. Natl Acad. Sci. USA 95, 1432–1437 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. von Manteuffel, S. R. et al. The insulin-induced signalling pathway leading to S6 and initiation factor 4E binding protein 1 phosphorylation bifurcates at a rapamycin-sensitive point immediately upstream of p70s6k. Mol. Cell Biol. 17, 5426–5436 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Phin, S., Kupferwasser, D., Lam, J. & Lee-Fruman, K. K. Mutational analysis of ribosomal S6 kinase 2 shows differential regulation of its kinase activity from that of ribosomal S6 kinase 1. Biochem. J. 373, 583–591 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Peterson, R. T., Desai, B. N., Hardwick, J. S. & Schreiber, S. L. Protein phosphatase 2A interacts with the 70-kDa S6 kinase and is activated by inhibition of FKBP12-rapamycinassociated protein. Proc. Natl Acad. Sci. USA 96, 4438–4442 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Duvel, K., Santhanam, A., Garrett, S., Schneper, L. & Broach, J. R. Multiple roles of Tap42 in mediating rapamycin-induced transcriptional changes in yeast. Mol. Cell 11, 1467–1478 (2003).

    PubMed  Google Scholar 

  68. Schmelzle, T., Beck, T., Martin, D. E. & Hall, M. N. Activation of the RAS/cyclic AMP pathway suppresses a TOR deficiency in yeast. Mol. Cell Biol. 24, 338–351 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Dennis, P. B., Fumagalli, S. & Thomas, G. Target of rapamycin (TOR): balancing the opposing forces of protein synthesis and degradation. Curr. Opin. Genet. Dev. 9, 49–54 (1999).

    CAS  PubMed  Google Scholar 

  70. Murata, K., Wu, J. & Brautigan, D. L. B cell receptor-associated protein α4 displays rapamycin-sensitive binding directly to the catalytic subunit of protein phosphatase 2A. Proc. Natl Acad. Sci. USA 94, 10624–10629 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Inui, S. et al. Ig receptor binding protein 1 (α4) is associated with a rapamycin-sensitive signal transduction in lymphocytes through direct binding to the catalytic subunit of protein phosphatase 2A. Blood 92, 539–546 (1998).

    CAS  PubMed  Google Scholar 

  72. Chen, J., Peterson, R. T. & Schreiber, S. L. α4 associates with protein phosphatases 2A, 4 and 6. Biochim. Biophys. Res. Commun. 247, 827–832 (1998).

    CAS  Google Scholar 

  73. Kloeker, S. et al. Parallel purification of three catalytic subunits of the protein serine/threonine phosphatase 2A family (PP2A(C), PP4(C), and PP6(C)) and analysis of the interaction of PP2A(C) with α4 protein. Protein Expr. Purif. 31, 19–33 (2003).

    CAS  PubMed  Google Scholar 

  74. Jefferies, H. B. et al. Rapamycin suppresses 5′TOP mRNA translation through inhibition of p70s6k. EMBO J. 16, 3693–3704 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Terada, N. et al. Rapamycin selectively inhibits translation of mRNAs encoding elongation factors and ribosomal proteins. Proc. Natl Acad. Sci. USA 91, 11477–11481 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Jefferies, H. B., Reinhard, C., Kozma, S. C. & Thomas, G. Rapamycin selectively represses translation of the 'polypyrimidine tract' mRNA family. Proc. Natl Acad. Sci. USA 91, 4441–4445 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Tang, H. et al. Amino acid-induced translation of TOP mRNAs is fully dependent on phosphatidylinositol 3-kinase-mediated signaling, is partially inhibited by rapamycin, and is independent of S6K1 and rpS6 phosphorylation. Mol. Cell Biol. 21, 8671–8683 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Stolovich, M. et al. Transduction of growth or mitogenic signals into translational activation of TOP mRNAs is fully reliant on the phosphatidylinositol 3-kinase-mediated pathway but requires neither S6K1 nor rpS6 phosphorylation. Mol. Cell Biol. 22, 8101–8113 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Pende, M. et al. Hypoinsulinaemia, glucose intolerance and diminished β-cell size in S6K1-deficient mice. Nature 408, 994–997 (2000).

    CAS  PubMed  Google Scholar 

  80. Wang, X. et al. Regulation of elongation factor 2 kinase by p90RSK1 and p70 S6 kinase. EMBO J. 20, 4370–4379 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Brunn, G. J. et al. Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin. Science 277, 99–101 (1997).

    CAS  PubMed  Google Scholar 

  82. Hara, K. et al. Regulation of eIF-4E BP1 phosphorylation by mTOR. J. Biol. Chem. 272, 26457–26463 (1997).

    CAS  PubMed  Google Scholar 

  83. Gingras, A. C. et al. Regulation of 4E-BP1 phosphorylation: a novel two-step mechanism. Genes Dev. 13, 1422–1437 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Yang, D. Q. & Kastan, M. B. Participation of ATM in insulin signalling through phosphorylation of eIF-4E-binding protein 1. Nature Cell Biol. 2, 893–898 (2000).

    CAS  PubMed  Google Scholar 

  85. Lin, T. A. et al. PHAS-I as a link between mitogen-activated protein kinase and translation initiation. Science 266, 653–656 (1994).

    CAS  PubMed  Google Scholar 

  86. Mothe-Satney, I. et al. Mammalian target of rapamycin-dependent phosphorylation of PHAS-I in four (S/T)P sites detected by phospho-specific antibodies. J. Biol. Chem. 275, 33836–33843 (2000).

    CAS  PubMed  Google Scholar 

  87. Mothe-Satney, I., Yang, D., Fadden, P., Haystead, T. A. & Lawrence, J. C. Jr. Multiple mechanisms control phosphorylation of PHAS-I in five (S/T)P sites that govern translational repression. Mol. Cell. Biol. 20, 3558–3567 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Gingras, A. C. et al. Hierarchical phosphorylation of the translation inhibitor 4E-BP1. Genes Dev. 15, 2852–2864 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Rosenwald, I. B. et al. Eukaryotic translation initiation factor 4E regulates expression of cyclin D1 at transcriptional and post-transcriptional levels. J. Biol. Chem. 270, 21176–21180 (1995).

    CAS  PubMed  Google Scholar 

  90. Hashemolhosseini, S. et al. Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability. J. Biol. Chem. 273, 14424–14429 (1998).

    CAS  PubMed  Google Scholar 

  91. Shantz, L. M. & Pegg, A. E. Overproduction of ornithine decarboxylase caused by relief of translational repression is associated with neoplastic transformation. Cancer Res. 54, 2313–2316 (1994).

    CAS  PubMed  Google Scholar 

  92. Fingar, D. C., Salama, S., Tsou, C., Harlow, E. & Blenis, J. Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev. 16, 1472–1487 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Montagne, J. et al. Drosophila S6 kinase: a regulator of cell size. Science 285, 2126–2129 (1999).

    CAS  PubMed  Google Scholar 

  94. Kozma, S. C. & Thomas, G. Regulation of cell size in growth, development and human disease: PI3K, PKB and S6K. Bioessays 24, 65–71 (2002).

    CAS  PubMed  Google Scholar 

  95. Shima, H. et al. Disruption of the p70s6k/p85s6k gene reveals a small mouse phenotype and a new functional S6 kinase. EMBO J. 17, 6649–6659 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Volarevic, S. et al. Proliferation, but not growth, blocked by conditional deletion of 40S ribosomal protein S6. Science 288, 2045–2047 (2000).

    CAS  PubMed  Google Scholar 

  97. Dilling, M. B. et al. 4E-binding proteins, the suppressors of eukaryotic initiation factor 4E, are down-regulated in cells with acquired or intrinsic resistance to rapamycin. J. Biol. Chem. 277, 13907–13917 (2002).

    CAS  PubMed  Google Scholar 

  98. Jiang, H., Coleman, J., Miskimins, R. & Miskimins, W. K. Expression of constitutively active 4EBP-1 enhances p27Kip1 expression and inhibits proliferation of MCF7 breast cancer cells. Cancer Cell Int. 3, 2 (2003).

    PubMed  PubMed Central  Google Scholar 

  99. Law, B. K. et al. Rapamycin potentiates transforming growth factor β-induced growth arrest in nontransformed, oncogene-transformed, and human cancer cells. Mol. Cell. Biol. 22, 8184–8198 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Nourse, J. et al. Interleukin-2-mediated elimination of the p27Kip1 cyclin-dependent kinase inhibitor prevented by rapamycin. Nature 372, 570–573 (1994).

    CAS  PubMed  Google Scholar 

  101. Barata, J. T., Cardoso, A. A., Nadler, L. M. & Boussiotis, V. A. Interleukin-7 promotes survival and cell cycle progression of T-cell acute lymphoblastic leukemia cells by down-regulating the cyclin-dependent kinase inhibitor p27kip1. Blood 98, 1524–1531 (2001).

    CAS  PubMed  Google Scholar 

  102. Kato, J. Y., Matsuoka, M., Polyak, K., Massague, J. & Sherr, C. J. Cyclic AMP-induced G1 phase arrest mediated by an inhibitor (p27Kip1) of cyclin-dependent kinase 4 activation. Cell 79, 487–496 (1994).

    CAS  PubMed  Google Scholar 

  103. Huang, S. et al. p53/p21CIP1 cooperate in enforcing rapamycin-induced G(1) arrest and determine the cellular response to rapamycin. Cancer Res. 61, 3373–3381 (2001).

    CAS  PubMed  Google Scholar 

  104. Lieberthal, W. et al. Rapamycin impairs recovery from acute renal failure: role of cell-cycle arrest and apoptosis of tubular cells. Am. J. Physiol. Renal Physiol. 281, F693–F706 (2001).

    CAS  PubMed  Google Scholar 

  105. Thimmaiah, K. N. et al. Insulin-like growth factor I-mediated protection from rapamycin-induced apoptosis is independent of Ras–Erk1–Erk2 and phosphatidylinositol 3′-kinase–Akt signaling pathways. Cancer Res. 63, 364–374 (2003).

    CAS  PubMed  Google Scholar 

  106. Woltman, A. M. et al. Rapamycin specifically interferes with GM-CSF signaling in human dendritic cells, leading to apoptosis via increased p27KIP1 expression. Blood 101, 1439–1445 (2003).

    CAS  PubMed  Google Scholar 

  107. Kenerson, H. L., Aicher, L. D., True, L. D. & Yeung, R. S. Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors. Cancer Res. 62, 5645–5650 (2002).

    CAS  PubMed  Google Scholar 

  108. Huang, S. et al. Sustained activation of the JNK cascade and rapamycin-induced apoptosis are suppressed by p53/p21Cip1. Mol. Cell 11, 1491–1501 (2003). This work shows that, in the absence of IGF1, prolonged activation of the ASK1–JNK pathway induced apoptosis in response to rapamycin-mediated inhibition of TOR. In the presence of wild-type p53, in a pathway dependent on WAF1, ASK1–JNK activation was transient and apoptosis was suppressed.

    CAS  PubMed  Google Scholar 

  109. Vivanco, I. & Sawyers, C. L. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nature Rev. Cancer 2, 489–501 (2002).

    CAS  Google Scholar 

  110. Luo, J., Manning, B. D. & Cantley, L. C. Targeting the PI3K–Akt pathway in human cancer: rationale and promise. Cancer Cell 4, 257–262 (2003).

    CAS  PubMed  Google Scholar 

  111. Woods, A. et al. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr. Biol. 13, 2004–2008 (2003).

    CAS  PubMed  Google Scholar 

  112. Nakamura, N. et al. Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN. Mol. Cell Biol. 20, 8969–8982 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Neshat, M. S. et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc. Natl Acad. Sci. USA 98, 10314–10319 (2001). This work first identified hypersensitivity to rapamycins in cells lacking the dual phosphatase PTEN. These results provided the initial rationale for testing TOR inhibitors in PTEN -null human cancers.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Podsypanina, K. et al. An inhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinase activity in Pten+/− mice. Proc. Natl Acad. Sci. USA 98, 10320–10325 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Shi, Y. et al. Enhanced sensitivity of multiple myeloma cells containing PTEN mutations to CCI-779. Cancer Res. 62, 5027–5034 (2002).

    CAS  PubMed  Google Scholar 

  116. Mutter, G. L. et al. Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers. J. Natl Cancer Inst. 92, 924–930 (2000).

    CAS  PubMed  Google Scholar 

  117. Yu, K. et al. mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr. Relat. Cancer 8, 249–258 (2001).

    PubMed  Google Scholar 

  118. Zhou, C., Gehrig, P. A., Whang, Y. E. & Boggess, J. F. Rapamycin inhibits telomerase activity by decreasing the hTERT mRNA level in endometrial cancer cells. Mol. Cancer Ther. 2, 789–795 (2003).

    CAS  PubMed  Google Scholar 

  119. Cheadle, J. P., Reeve, M. P., Sampson, J. R. & Kwiatkowski, D. J. Molecular genetic advances in tuberous sclerosis. Hum. Genet. 107, 97–114 (2000).

    CAS  PubMed  Google Scholar 

  120. Brugarolas, J. B., Vazquez, F., Reddy, A., Sellers, W. R. & Kaelin, W. G. Jr. TSC2 regulates VEGF through mTOR-dependent and-independent pathways. Cancer Cell 4, 147–158 (2003). This study found that TSC2 regulates VEGF through TOR-dependent and -independent pathways. Rapamycin normalized hypoxia-inducible factor levels in TSC 2−/− cells, but only partially downregulated VEGF in this setting. The results imply a TOR-independent link between TSC2 loss and VEGF.

    CAS  PubMed  Google Scholar 

  121. Treins, C., Giorgetti-Peraldi, S., Murdaca, J., Semenza, G. L. & Van Obberghen, E. Insulin stimulates hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/target of rapamycin-dependent signaling pathway. J. Biol. Chem. 277, 27975–27981 (2002).

    CAS  PubMed  Google Scholar 

  122. Zhong, H. et al. Modulation of hypoxia-inducible factor 1α expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 60, 1541–1545 (2000).

    CAS  PubMed  Google Scholar 

  123. Zundel, W. et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 14, 391–396 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Jiang, B. H. et al. Phosphatidylinositol 3-kinase signaling controls levels of hypoxia-inducible factor 1. Cell Growth Differ. 12, 363–369 (2001).

    CAS  PubMed  Google Scholar 

  125. Laughner, E., Taghavi, P., Chiles, K., Mahon, P. C. & Semenza, G. L. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1α (HIF-1α) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol. Cell. Biol. 21, 3995–4004 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Mayerhofer, M., Valent, P., Sperr, W. R., Griffin, J. D. & Sillaber, C. BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor-1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood 100, 3767–3775 (2002).

    CAS  PubMed  Google Scholar 

  127. El-Hashemite, N., Walker, V., Zhang, H. & Kwiatkowski, D. J. Loss of Tsc1 or Tsc2 induces vascular endothelial growth factor production through mammalian target of rapamycin. Cancer Res. 63, 5173–5177 (2003).

    CAS  PubMed  Google Scholar 

  128. Guba, M. et al. Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nature Med. 8, 128–135 (2002). It was found that rapamycin inhibited metastatic tumour growth and angiogenesis in in vivo mouse models. Rapamycin showed anti-angiogenic activities linked to a decrease in production of Vegf and to a markedly inhibited response of vascular endothelial cells to stimulation by Vegf.

    CAS  PubMed  Google Scholar 

  129. Humar, R., Kiefer, F. N., Berns, H., Resink, T. J. & Battegay, E. J. Hypoxia enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)-dependent signaling. FASEB J. 16, 771–780 (2002).

    CAS  PubMed  Google Scholar 

  130. Gromov, P. S., Madsen, P., Tomerup, N. & Celis, J. E. A novel approach for expression cloning of small GTPases: identification, tissue distribution and chromosome mapping of the human homolog of rheb. FEBS Lett. 377, 221–226 (1995).

    CAS  PubMed  Google Scholar 

  131. Kwon, H. K. et al. Constitutive activation of p70S6k in cancer cells. Arch. Pharm. Res. 25, 685–690 (2002).

    CAS  PubMed  Google Scholar 

  132. Salh, B., Marotta, A., Wagey, R., Sayed, M. & Pelech, S. Dysregulation of phosphatidylinositol 3-kinase and downstream effectors in human breast cancer. Int. J. Cancer 98, 148–154 (2002).

    CAS  PubMed  Google Scholar 

  133. Wong, A. S. et al. Coexpression of hepatocyte growth factor-Met: an early step in ovarian carcinogenesis? Oncogene 20, 1318–1328 (2001).

    CAS  PubMed  Google Scholar 

  134. Lazaris-Karatzas, A. & Sonenberg, N. The mRNA 5′ cap-binding protein, eIF-4E, cooperates with v-myc or E1A in the transformation of primary rodent fibroblasts. Mol. Cell. Biol. 12, 1234–1238 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Lazaris-Karatzas, A. et al. Ras mediates translation initiation factor 4E-induced malignant transformation. Genes Dev. 6, 1631–1642 (1992). Previous work had shown that overexpression of eIF4E transformed rodent cells. In this paper, it is shown that such overexpression upregulates Ras and that transformation is Ras-dependent. Together, these studies indicate that eIF4E is an oncogene.

    CAS  PubMed  Google Scholar 

  136. Abid, M. R., Li, Y., Anthony, C. & De Benedetti, A. Translational regulation of ribonucleotide reductase by eukaryotic initiation factor 4E links protein synthesis to the control of DNA replication. J. Biol. Chem. 274, 35991–35998 (1999).

    CAS  PubMed  Google Scholar 

  137. Chabes, A. et al. Survival of DNA damage in yeast directly depends on increased dNTP levels allowed by relaxed feedback inhibition of ribonucleotide reductase. Cell 112, 391–401 (2003).

    CAS  PubMed  Google Scholar 

  138. Topisirovic, I. et al. The proline-rich homeodomain protein, PRH, is a tissue-specific inhibitor of eIF4E-dependent cyclin D1 mRNA transport and growth. EMBO J. 22, 689–703 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Rosenwald, I. B. Upregulated expression of the genes encoding translation initiation factors eIF-4E and eIF-2α in transformed cells. Cancer Lett. 102, 113–123 (1996).

    CAS  PubMed  Google Scholar 

  140. Miyagi, Y. et al. Elevated levels of eukaryotic translation initiation factor eIF-4E, mRNA in a broad spectrum of transformed cell lines. Cancer Lett. 91, 247–252 (1995).

    CAS  PubMed  Google Scholar 

  141. Graff, J. R. et al. Reduction of translation initiation factor 4E decreases the malignancy of ras-transformed cloned rat embryo fibroblasts. Int. J. Cancer 60, 255–263 (1995).

    CAS  PubMed  Google Scholar 

  142. Rinker-Schaeffer, C. W., Graff, J. R., De Benedetti, A., Zimmer, S. G. & Rhoads, R. E. Decreasing the level of translation initiation factor 4E with antisense RNA causes reversal of ras-mediated transformation and tumorigenesis of cloned rat embryo fibroblasts. Int. J. Cancer 55, 841–847 (1993).

    CAS  PubMed  Google Scholar 

  143. DeFatta, R. J., Nathan, C. A. & De Benedetti, A. Antisense RNA to eIF4E suppresses oncogenic properties of a head and neck squamous cell carcinoma cell line. Laryngoscope 110, 928–933 (2000).

    CAS  PubMed  Google Scholar 

  144. Wang, S. et al. Expression of eukaryotic translation initiation factors 4E and 2α correlates with the progression of thyroid carcinoma. Thyroid 11, 1101–1107 (2001).

    CAS  PubMed  Google Scholar 

  145. Rosenwald, I. B., Hutzler, M. J., Wang, S., Savas, L. & Fraire, A. E. Expression of eukaryotic translation initiation factors 4E and 2α is increased frequently in bronchioloalveolar but not in squamous cell carcinomas of the lung. Cancer 92, 2164–2171 (2001).

    CAS  PubMed  Google Scholar 

  146. Wang, S. et al. Expression of the eukaryotic translation initiation factors 4E and 2α in non-Hodgkin's lymphomas. Am. J. Pathol. 155, 247–255 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Bauer, C. et al. Overexpression of the eukaryotic translation initiation factor 4G (eIF4G-1) in squamous cell lung carcinoma. Int. J. Cancer 98, 181–185 (2002).

    CAS  PubMed  Google Scholar 

  148. Fukuchi-Shimogori, T. et al. Malignant transformation by overproduction of translation initiation factor eIF4G. Cancer Res. 57, 5041–5044 (1997).

    CAS  PubMed  Google Scholar 

  149. Martin, M. E. et al. 4E binding protein 1 expression is inversely correlated to the progression of gastrointestinal cancers. Int. J. Biochem. Cell Biol. 32, 633–642 (2000).

    CAS  PubMed  Google Scholar 

  150. Nomura, M. et al. Involvement of the Akt/mTOR pathway on EGF-induced cell transformation. Mol. Carcinog. 38, 25–32 (2003).

    CAS  PubMed  Google Scholar 

  151. Skorski, T. et al. Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway. EMBO J. 16, 6151–6161 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Ly, C., Arechiga, A. F., Melo, J. V., Walsh, C. M. & Ong, S. T. Bcr–Abl kinase modulates the translation regulators ribosomal protein S6 and 4E-BP1 in chronic myelogenous leukemia cells via the mammalian target of rapamycin. Cancer Res. 63, 5716–5722 (2003).

    CAS  PubMed  Google Scholar 

  153. Gera, J. F. et al. AKT activity determines sensitivity to mTOR inhibitors by regulating cyclin D1 and c-myc expression. J. Biol. Chem. 279, 2737–2746 (2003).

    PubMed  Google Scholar 

  154. Louro, I. D. et al. The zinc finger protein GLI induces cellular sensitivity to the mTOR inhibitor rapamycin. Cell Growth Differ. 10, 503–516 (1999).

    CAS  PubMed  Google Scholar 

  155. Pietenpol, J. A. et al. TGF-β 1 inhibition of c-myc transcription and growth in keratinocytes is abrogated by viral transforming proteins with pRB binding domains. Cell 61, 777–785 (1990).

    CAS  PubMed  Google Scholar 

  156. Song, K., Cornelius, S. C., Reiss, M. & Danielpour, D. Insulin-like growth factor-I inhibits transcriptional responses of transforming growth factor-β by phosphatidylinositol 3-kinase/Akt-dependent suppression of the activation of Smad3 but not Smad2. J. Biol. Chem. 278, 38342–38351 (2003).

    CAS  PubMed  Google Scholar 

  157. Douros, J. & Suffness, M. New antitumor substances of natural origin. Cancer Treat. Rev. 8, 63–87 (1981).

    CAS  PubMed  Google Scholar 

  158. Houchens, D. P., Ovejera, A. A., Riblet, S. M. & Slagel, D. E. Human brain tumor xenografts in nude mice as a chemotherapy model. Eur. J. Cancer Clin. Oncol. 19, 799–805 (1983).

    CAS  PubMed  Google Scholar 

  159. Eng, C. P., Sehgal, S. N. & Vezina, C. Activity of rapamycin (AY-22,989) against transplanted tumors. J. Antibiot. 37, 1231–1237 (1984).

    CAS  Google Scholar 

  160. Dancey, J. E. Clinical development of mammalian target of rapamycin inhibitors. Hematol. Oncol. Clin. North Am. 16, 1101–1114 (2002).

    PubMed  Google Scholar 

  161. Atkins, M. B. et al. Randomized phase II study of multiple dose levels of CCI–779, a novel mTOR kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J. Clin. Oncol. 22, 909–918 (2004).

    CAS  PubMed  Google Scholar 

  162. Boulay, A. et al. Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells. Cancer Res. 64, 252–261 (2004).

    CAS  PubMed  Google Scholar 

  163. Peralba, J. M. et al. Pharmacodynamic evaluation of CCI-779, an inhibitor of mTOR, in cancer patients. Clin. Cancer Res. 9, 2887–2892 (2003).

    CAS  PubMed  Google Scholar 

  164. Huang, S., Bjornsti, M. A. & Houghton, P. J. Rapamycins: mechanism of action and cellular resistance. Cancer Biol. Ther. 2, 222–232 (2003).

    CAS  PubMed  Google Scholar 

  165. Peng, T., Golub, T. R. & Sabatini, D. M. The immunosuppressant rapamycin mimics a starvation-like signal distinct from amino acid and glucose deprivation. Mol. Cell. Biol. 22, 5575–5584 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Rosenwald, A. et al. The proliferation gene expression signature is a quantitative integrator of oncogenic events that predicts survival in mantle cell lymphoma. Cancer Cell 3, 185–197 (2003).

    CAS  PubMed  Google Scholar 

  167. Liang, A., Lei, T., LuYing, Z. & YuPing, G. The expression of proto-oncogene eIF4E in laryngeal squamous cell carcinoma. Laryngoscope 113, 1238–1243 (2003).

    PubMed  Google Scholar 

  168. Nathan, C. A. et al. Molecular analysis of surgical margins in head and neck squamous cell carcinoma patients. Laryngoscope 112, 2129–2140 (2002).

    CAS  PubMed  Google Scholar 

  169. Haydon, M. S., Googe, J. D., Sorrells, D. S., Ghali, G. E. & Li, B. D. Progression of eIF4e gene amplification and overexpression in benign and malignant tumors of the head and neck. Cancer 88, 2803–2810 (2000).

    CAS  PubMed  Google Scholar 

  170. Sorrells, D. L., Meschonat, C., Black, D. & Li, B. D. Pattern of amplification and overexpression of the eukaryotic initiation factor 4E gene in solid tumor. J. Surg. Res. 85, 37–42 (1999).

    CAS  PubMed  Google Scholar 

  171. Seki, N. et al. Expression of eukaryotic initiation factor 4E in atypical adenomatous hyperplasia and adenocarcinoma of the human peripheral lung. Clin. Cancer Res. 8, 3046–3053 (2002).

    CAS  PubMed  Google Scholar 

  172. Kerekatte, V. et al. The proto-oncogene/translation factor eIF4E: a survey of its expression in breast carcinomas. Int. J. Cancer 64, 27–31 (1995).

    CAS  PubMed  Google Scholar 

  173. Li, B. D. et al. Prospective study of eukaryotic initiation factor 4E protein elevation and breast cancer outcome. Ann. Surg. 235, 732–738 (2002).

    PubMed  PubMed Central  Google Scholar 

  174. Scott, P. A. et al. Differential expression of vascular endothelial growth factor mRNA vs protein isoform expression in human breast cancer and relationship to eIF-4E. Br. J. Cancer 77, 2120–2128 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Nathan, C. A. et al. Elevated expression of eIF4E and FGF-2 isoforms during vascularization of breast carcinomas. Oncogene 15, 1087–1094 (1997).

    CAS  PubMed  Google Scholar 

  176. Berkel, H. J., Turbat-Herrera, E. A., Shi, R. & de Benedetti, A. Expression of the translation initiation factor eIF4E in the polyp-cancer sequence in the colon. Cancer Epidemiol. Biomarkers Prev. 10, 663–666 (2001).

    CAS  PubMed  Google Scholar 

  177. Rosenwald, I. B. et al. Upregulation of protein synthesis initiation factor eIF-4E is an early event during colon carcinogenesis. Oncogene 18, 2507–2517 (1999).

    CAS  PubMed  Google Scholar 

  178. Crew, J. P. et al. Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression. Br. J. Cancer 82, 161–166 (2000).

    CAS  PubMed  Google Scholar 

  179. Chen, Y., Zheng, Y. & Foster, D. A. Phospholipase D confers rapamycin resistance in human breast cancer cells. Oncogene 22, 3937–3942 (2003).

    CAS  PubMed  Google Scholar 

  180. Wang, L., Rolfe, M. & Proud, C. G. Ca2+-independent protein kinase C activity is required for α1-adrenergic-receptor-mediated regulation of ribosomal protein S6 kinases in adult cardiomyocytes. Biochem. J. 373, 603–611 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Lim, H. K. et al. Phosphatidic acid regulates systemic inflammatory responses by modulating the Akt-mammalian target of rapamycin–p70 S6 kinase 1 pathway. J. Biol. Chem. 5, 5 (2003).

    Google Scholar 

  182. Dennis, P. B. et al. Mammalian TOR: a homeostatic ATP sensor. Science 294, 1102–1105 (2001).

    CAS  PubMed  Google Scholar 

  183. Meijer, A. J. Amino acids as regulators and components of nonproteinogenic pathways. J. Nutr. 133, 2057S–2062S (2003).

    CAS  PubMed  Google Scholar 

  184. Wang, L., Fraley, C. D., Faridi, J., Kornberg, A. & Roth, R. A. Inorganic polyphosphate stimulates mammalian TOR, a kinase involved in the proliferation of mammary cancer cells. Proc. Natl Acad. Sci. USA 100, 11249–11254 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Arsham, A. M., Howell, J. J. & Simon, M. C. A novel hypoxia-inducible factor-independent hypoxic response regulating mammalian target of rapamycin and its targets. J. Biol. Chem. 278, 29655–29660 (2003). It is shown that hypoxia rapidly and reversibly triggers hypophosphorylation of TOR and its downstream effectors. Hypoxic regulation of TOR is dominant to activation through several distinct signalling pathways and implicates the role of TOR in sensing O 2 status of cells.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are supported by grants from the United States Public Health Service and the American, Lebanese, Syrian Associated Charities.

Correction: The DOI number given for this article in the May 2004 print issue of Nature Reviews Cancer was wrong. The correct DOI number is: doi:10.1038/nrc1362.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Peter J. Houghton.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

Cancer.gov

breast cancer

endometrial cancer

LocusLink

4E-BP1

ASK1

BRCA1

cyclin D1

eIF2α

eIF4E

FKBP12

KIP1

LKB1

p53

PDK1

PP2A

PP4

PP6

PTEN

RAC1

RAP1A

RAP1-GAP

RAB5

RHEB

S6K1

TERT

TOR

TSC1

TSC2

Glossary

WD REPEAT

A protein-binding and small-ligand-binding motif that is present in the amino-acid sequence of several proteins.

SCAFFOLD PROTEIN

A protein with the primary function of bringing together proteins that share a common signalling pathway to allow for interaction between those proteins. Also referred to as docking proteins.

STAT3

A member of the STAT family of signalling proteins. Following activation, STAT3 forms a heterodimer with other STAT family members. This complex migrates to the nucleus to act as a transcription factor, activating genes that are involved in cell division and differentiation.

14-3-3 PROTEINS

This family of proteins bind to a phosphorylated serine residue within the 14-3-3 domain of target proteins. 14-3-3 proteins can induce several effects — altered interactions with other binding partners, changes in the localization within the cell, altered catalytic activity and altered stability of the target protein. 14-3-3 proteins have been implicated in several biological functions, including cell-cycle progression, signal transduction and apoptosis.

SMALL G PROTEINS

This family of small proteins mediate signalling by binding and hydrolysing GTP. The GTP-bound form is active. It interacts with and activates several effector proteins that mediate downstream signalling events. G proteins are regulated by proteins that accelerate GTP hydrolysis to GDP (GAPs), increase the rate of GTP binding (GEFs) or inhibit the release of GDP (GDIs).

ORNITHINE DECARBOXYLASE

Rate-limiting enzyme in the conversion of ornithine to putrescine. Putrescine and other polyamines are required for the division of normal cells.

LOSS OF HETEROZYGOSITY

(LOH). In cells that carry a mutated allele of a tumour-suppressor gene, the gene becomes fully inactivated when the cell loses a large part of the chromosome carrying the wild-type allele. Regions with high frequency of LOH are believed to harbour tumour-suppressor genes.

BCR–ABL

A fusion protein resulting from the translocation between a gene in the breakpoint cluster region (BCR) and the gene coding for the ABL tyrosine kinase. This fusion protein is responsible for some types of leukaemia.

HYPOCALCAEMIA

Low levels of calcium within the body.

THROMBOCYTOPAENIA

Low levels platelets in the blood.

MUCOSITIS

Inflammation of mucosa often seen as sores in the mouth.

LEUCOPAENIA

Low white-blood-cell count.

HYPERLIPIDAEMIA

Excess levels of lipids in the blood.

PHARMACODYNAMIC

The effects on the biochemistry of the body resulting from treatment with a drug or combination of drugs.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bjornsti, MA., Houghton, P. The tor pathway: a target for cancer therapy. Nat Rev Cancer 4, 335–348 (2004). https://doi.org/10.1038/nrc1362

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1362

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing