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:

Hypoxia signalling through mTOR and the unfolded protein response in cancer

Key Points

  • Cellular responses to hypoxia are mediated by both hypoxia-inducible factor (HIF)-dependent and HIF-independent pathways. Each of these O2-sensitive signalling pathways exhibits unique sensitivity to the severity and duration of O2 deprivation.

  • Hypoxia inhibits signalling downstream of the kinase mammalian target of rapamycin (mTOR) and mRNA translation initiation through multiple independent mechanisms. Signalling through this pathway appears to influence both tumour progression and hypoxia tolerance in advanced tumours.

  • Severe hypoxic exposure causes endoplasmic reticulum (ER) stress and leads to rapid activation of the unfolded protein response (UPR). The UPR regulates several downstream effector pathways that together function to promote hypoxia tolerance.

  • Hypoxic signalling through mTOR and the UPR results in significant changes in mRNA translation that influence gene expression and cellular behaviour in hypoxic cells. Targeting these pathways can reduce or slow tumour growth.

  • Many of the cellular consequences of hypoxia are jointly influenced by overlapping O2-sensitive pathways. HIF, mTOR and UPR signalling during hypoxia influence tumour metabolism, autophagy and ER homeostasis.

  • Many current and experimental anticancer agents cause ER stress and activate the UPR, and may thus show selective toxicity to hypoxic cells. Conversely, hypoxic signalling through the mTOR pathway is likely to influence the efficacy of many of the new drugs targeting this pathway.

Abstract

Hypoxia occurs in the majority of tumours, promoting angiogenesis, metastasis and resistance to therapy. Responses to hypoxia are orchestrated in part through activation of the hypoxia-inducible factor family of transcription factors (HIFs). Recently, two additional O2-sensitive signalling pathways have also been implicated: signalling through the mammalian target of rapamycin (mTOR) kinase and signalling through activation of the unfolded protein response (UPR). Although they are activated independently, growing evidence suggests that HIF-, mTOR- and UPR-dependent responses to hypoxia act in an integrated way, influencing each other and common downstream pathways that affect gene expression, metabolism, cell survival, tumorigenesis and tumour growth.

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: Cellular O2-sensing pathways.
Figure 2: Inhibition of mammalian target of rapamycin (mTOR) signalling by hypoxia.
Figure 3: Hypoxia activates the unfolded protein response (UPR).
Figure 4: Negative feedback loops from hypoxia-inducible factor 1 (HIF1) and unfolded protein response (UPR) to mammalian target of rapamycin (mTOR).

Similar content being viewed by others

References

  1. Dewhirst, M. W., Cao, Y. & Moeller, B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nature Rev. Cancer 8, 425–437 (2008).

    CAS  Google Scholar 

  2. Guertin, D. A. & Sabatini, D. M. Defining the role of mTOR in cancer. Cancer Cell 12, 9–22 (2007).

    CAS  PubMed  Google Scholar 

  3. Browne, G. J. & Proud, C. G. A novel mTOR-regulated phosphorylation site in elongation factor 2 kinase modulates the activity of the kinase and its binding to calmodulin. Mol. Cell. Biol. 24, 2986–2997 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. 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). This was the first study to show that exposure to hypoxia could block activation of mTOR in a HIF-independent manner.

    CAS  PubMed  Google Scholar 

  5. Liu, L. et al. Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol. Cell 21, 521–531 (2006).

    PubMed  PubMed Central  Google Scholar 

  6. Brugarolas, J. et al. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 18, 2893–2904 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Reiling, J. H. & Hafen, E. The hypoxia-induced paralogs Scylla and Charybdis inhibit growth by down-regulating S6K activity upstream of TSC in Drosophila. Genes Dev. 18, 2879–2892 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Sofer, A., Lei, K., Johannessen, C. M. & Ellisen, L. W. Regulation of mTOR and cell growth in response to energy stress by REDD1. Mol. Cell. Biol. 25, 5834–5845 (2005). References 6 and 8 demonstrated that mTOR inhibition during hypoxia is partly dependent on REDD1 signaling to TSC1–TSC2. This also provided a link between HIF and mTOR regulation

    CAS  PubMed  PubMed Central  Google Scholar 

  9. DeYoung, M. P., Horak, P., Sofer, A., Sgroi, D. & Ellisen, L. W. Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling. Genes Dev. 22, 239–251 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Bernardi, R. et al. PML inhibits HIF-1α translation and neoangiogenesis through repression of mTOR. Nature 442, 779–785 (2006).

    CAS  PubMed  Google Scholar 

  11. Li, Y. et al. Bnip3 mediates the hypoxia-induced inhibition on mammalian target of rapamycin by interacting with Rheb. J. Biol. Chem. 282, 35803–35813 (2007).

    CAS  PubMed  Google Scholar 

  12. Hay, N. & Sonenberg, N. Upstream and downstream of mTOR. Genes Dev. 18, 1926–1945 (2004).

    CAS  PubMed  Google Scholar 

  13. Dostie, J., Ferraiuolo, M., Pause, A., Adam, S. A. & Sonenberg, N. A novel shuttling protein, 4E-T, mediates the nuclear import of the mRNA 5′ cap-binding protein, eIF4E. EMBO J. 19, 3142–3156 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Koritzinsky, M. et al. Gene expression during acute and prolonged hypoxia is regulated by distinct mechanisms of translational control. EMBO J. 25, 1114–1125 (2006). This study demonstrated that hypoxia leads to both rapid and sustained inhibition of mRNA translation and that this influences gene expression during hypoxia through both mTOR- and UPR-dependent pathways.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Ferraiuolo, M. A. et al. A role for the eIF4E-binding protein 4E-T in P-body formation and mRNA decay. J. Cell Biol. 170, 913–924 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Freyer, J. P. Rates of oxygen consumption for proliferating and quiescent cells isolated from multicellular tumor spheroids. Adv. Exp. Med. Biol. 345, 335–342 (1994).

    CAS  PubMed  Google Scholar 

  17. Graeber, T. G. et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 379, 88–91 (1996).

    CAS  PubMed  Google Scholar 

  18. Connolly, E., Braunstein, S., Formenti, S. & Schneider, R. J. Hypoxia inhibits protein synthesis through a 4E-BP1 and elongation factor 2 kinase pathway controlled by mTOR and uncoupled in breast cancer cells. Mol. Cell. Biol. 26, 3955–3965 (2006). This study suggests that mTOR activation during moderate hypoxia may act as a barrier to tumorigenesis, which is lost in advanced breast cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Kaper, F., Dornhoefer, N. & Giaccia, A. J. Mutations in the PI3K/PTEN/TSC2 pathway contribute to mammalian target of rapamycin activity and increased translation under hypoxic conditions. Cancer Res. 66, 1561–1569 (2006).

    CAS  PubMed  Google Scholar 

  20. Koritzinsky, M. et al. Phosphorylation of eIF2α is required for mRNA translation inhibition and survival during moderate hypoxia. Radiother. Oncol. 83, 353–361 (2007).

    CAS  PubMed  Google Scholar 

  21. Magagnin, M. G. et al. The mTOR target 4E-BP1 contributes to differential protein expression during normoxia and hypoxia through changes in mRNA translation efficiency. Proteomics (2008).

  22. Richter, J. D. & Sonenberg, N. Regulation of cap-dependent translation by eIF4E inhibitory proteins. Nature 433, 477–480 (2005).

    CAS  PubMed  Google Scholar 

  23. Braunstein, S. et al. A hypoxia-controlled cap-dependent to cap-independent translation switch in breast cancer. Mol. Cell 28, 501–512 (2007). This study indicates that hypoxia contributes to changes in translation, which promote adverse phenotypes in advanced cancers. It also suggests that, in advanced cancer, hypoxic cells need to maintain regulation over cap-dependent translation.

    CAS  PubMed  Google Scholar 

  24. Sowter, H. M., Ratcliffe, P. J., Watson, P., Greenberg, A. H. & Harris, A. L. HIF-1-dependent regulation of hypoxic induction of the cell death factors BNIP3 and NIX in human tumors. Cancer Res. 61, 6669–6673 (2001).

    CAS  PubMed  Google Scholar 

  25. Giatromanolaki, A. et al. BNIP3 expression is linked with hypoxia-regulated protein expression and with poor prognosis in non-small cell lung cancer. Clin. Cancer Res. 10, 5566–5571 (2004).

    CAS  PubMed  Google Scholar 

  26. Giatromanolaki, A., Koukourakis, M. I., Gatter, K. C., Harris, A. L. & Sivridis, E. BNIP3 expression in endometrial cancer relates to active hypoxia inducible factor 1alpha pathway and prognosis. J. Clin. Pathol. 61, 217–220 (2008).

    CAS  PubMed  Google Scholar 

  27. Tan, E. Y. et al. BNIP3 as a progression marker in primary human breast cancer; opposing functions in in situ versus invasive cancer. Clin. Cancer Res. 13, 467–474 (2007).

    CAS  PubMed  Google Scholar 

  28. Erler, J. T. et al. Hypoxia-mediated down-regulation of Bid and Bax in tumors occurs via hypoxia-inducible factor 1-dependent and -independent mechanisms and contributes to drug resistance. Mol. Cell. Biol. 24, 2875–2889 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Mizushima, N. Autophagy: process and function. Genes Dev. 21, 2861–2873 (2007).

    CAS  PubMed  Google Scholar 

  30. Azad, M. B. et al. Hypoxia induces autophagic cell death in apoptosis-competent cells through a mechanism involving BNIP3. Autophagy 4, 195–204 (2008).

    CAS  PubMed  Google Scholar 

  31. Degenhardt, K. et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10, 51–64 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, H. et al. Mitochondrial autophagy is a HIF-1-dependent adaptive metabolic response to hypoxia. J. Biol. Chem. 283, 10892–10903 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Adhami, F. et al. Cerebral ischemia-hypoxia induces intravascular coagulation and autophagy. Am. J. Pathol. 169, 566–583 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Yan, L. et al. Autophagy in chronically ischemic myocardium. Proc. Natl Acad. Sci. USA 102, 13807–13812 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ravikumar, B. et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nature Genet. 36, 585–595 (2004).

    CAS  PubMed  Google Scholar 

  36. Papandreou, I., Lim, A. L., Laderoute, K. & Denko, N. C. Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L. Cell Death Differ. 15, 1572–1581 (2008).

    CAS  PubMed  Google Scholar 

  37. Maiuri, M. C. et al. Functional and physical interaction between Bcl-XL and a BH3-like domain in Beclin-1. EMBO J. 26, 2527–2539 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nature Rev. Mol. Cell Biol. 8, 519–529 (2007).

    CAS  Google Scholar 

  39. Zhang, K. & Kaufman, R. J. Protein folding in the endoplasmic reticulum and the unfolded protein response. Handb. Exp. Pharmacol. 172, 69–91 (2006).

    CAS  Google Scholar 

  40. Koumenis, C. et al. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2a. Mol. Cell. Biol. 22, 7405–7416 (2002). This study showed that both severe and moderate hypoxia can activate the PERK–EIF2α arm of the UPR and that this is important for hypoxia tolerance.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ma, Y. & Hendershot, L. M. Delineation of a negative feedback regulatory loop that controls protein translation during endoplasmic reticulum stress. J. Biol. Chem. 278, 34864–34873 (2003).

    CAS  PubMed  Google Scholar 

  42. Bi, M. et al. ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth. EMBO J. 24, 3470–3481 (2005). This study demonstrated that activation of the UPR protects cells against hypoxic stress both in vitro and in vivo . Inhibition of PERK reduced tumour hypoxia and prevented growth of tumour xenografts.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol. Cell 7, 1165–1176 (2001).

    CAS  PubMed  Google Scholar 

  44. Romero-Ramirez, L. et al. XBP1 is essential for survival under hypoxic conditions and is required for tumor growth. Cancer Res. 64, 5943–5947 (2004). This paper showed that the IRE1–XBP1 arm of the UPR is also activated during hypoxia and contributes to hypoxia tolerance and tumour growth.

    CAS  PubMed  Google Scholar 

  45. Davies, M. P. et al. Expression and splicing of the unfolded protein response gene XBP-1 are significantly associated with clinical outcome of endocrine-treated breast cancer. Int. J. Cancer 123, 85–88 (2008).

    CAS  PubMed  Google Scholar 

  46. Ma, Y. & Hendershot, L. M. The role of the unfolded protein response in tumour development: friend or foe? Nature Rev. Cancer 4, 966–977 (2004).

    CAS  Google Scholar 

  47. Denoyelle, C. et al. Anti-oncogenic role of the endoplasmic reticulum differentially activated by mutations in the MAPK pathway. Nature Cell Biol. 8, 1053–1063 (2006).

    CAS  PubMed  Google Scholar 

  48. Kraggerud, S. M., Sandvik, J. A. & Pettersen, E. O. Regulation of protein synthesis in human cells exposed to extreme hypoxia. Anticancer Res. 15, 683–686 (1995).

    CAS  PubMed  Google Scholar 

  49. Vattem, K. M. & Wek, R. C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc. Natl Acad. Sci. USA 101, 11269–11274 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Lu, P. D., Harding, H. P. & Ron, D. Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response. J. Cell Biol. 167, 27–33 (2004). This paper demonstrated the mechanism whereby certain mRNA transcripts can be preferentially translated under conditions that activate the UPR, such as hypoxia.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Blais, J. D. et al. Activating transcription factor 4 is translationally regulated by hypoxic stress. Mol. Cell. Biol. 24, 7469–7482 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Ameri, K. et al. Anoxic induction of ATF-4 through HIF-1-independent pathways of protein stabilization in human cancer cells. Blood 103, 1876–1882 (2004).

    CAS  PubMed  Google Scholar 

  53. Blais, J. D. et al. Perk-dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress. Mol. Cell. Biol. 26, 9517–9532 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Murphy, B. J., Laderoute, K. R., Short, S. M. & Sutherland, R. M. The identification of heme oxygenase as a major hypoxic stress protein in Chinese hamster ovary cells. Br. J. Cancer 64, 69–73 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Roll, D. E., Murphy, B. J., Laderoute, K. R., Sutherland, R. M. & Smith, H. C. Oxygen regulated 80 kDa protein and glucose regulated 78 kDa protein are identical. Mol. Cell. Biochem. 103, 141–148 (1991).

    CAS  PubMed  Google Scholar 

  56. Wilson, R. E. & Sutherland, R. M. Enhanced synthesis of specific proteins, RNA, and DNA caused by hypoxia and reoxygenation. Int. J. Radiat. Oncol. Biol. Phys. 16, 957–961 (1989).

    CAS  PubMed  Google Scholar 

  57. Gess, B. et al. The cellular oxygen tension regulates expression of the endoplasmic oxidoreductase ERO1-Lα. Eur. J. Biochem. 270, 2228–2235 (2003).

    CAS  PubMed  Google Scholar 

  58. Tanaka, S., Uehara, T. & Nomura, Y. Up-regulation of protein-disulfide isomerase in response to hypoxia/brain ischemia and its protective effect against apoptotic cell death. J. Biol. Chem. 275, 10388–10393 (2000).

    CAS  PubMed  Google Scholar 

  59. Drogat, B. et al. IRE1 signaling is essential for ischemia-induced vascular endothelial growth factor-A expression and contributes to angiogenesis and tumor growth in vivo. Cancer Res. 67, 6700–6707 (2007).

    CAS  PubMed  Google Scholar 

  60. May, D. et al. Ero1-L α plays a key role in a HIF-1-mediated pathway to improve disulfide bond formation and VEGF secretion under hypoxia: implication for cancer. Oncogene 24, 1011–1020 (2005).

    CAS  PubMed  Google Scholar 

  61. Anelli, T. & Sitia, R. Protein quality control in the early secretory pathway. EMBO J. 27, 315–327 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Qi, X., Okuma, Y., Hosoi, T., Kaneko, M. & Nomura, Y. Induction of murine HRD1 in experimental cerebral ischemia. Brain Res. Mol. Brain Res. 130, 30–38 (2004).

    CAS  PubMed  Google Scholar 

  63. Magagnin, M. G. et al. Proteomic analysis of gene expression following hypoxia and reoxygenation reveals proteins involved in the recovery from endoplasmic reticulum and oxidative stress. Radiother. Oncol. 83, 340–345 (2007).

    CAS  PubMed  Google Scholar 

  64. Kouroku, Y. et al. ER stress (PERK/eIF2α phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 14, 230–239 (2007).

    CAS  PubMed  Google Scholar 

  65. Ogata, M. et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol. Cell. Biol. 26, 9220–9231 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Bernales, S., McDonald, K. L. & Walter, P. Autophagy counterbalances endoplasmic reticulum expansion during the unfolded protein response. PLoS Biol. 4, e423 (2006).

    PubMed  PubMed Central  Google Scholar 

  67. Leber, J. H., Bernales, S. & Walter, P. IRE1-independent gain control of the unfolded protein response. PLoS Biol. 2, E235 (2004).

    PubMed  PubMed Central  Google Scholar 

  68. Hetz, C. et al. Proapoptotic BAX and BAK modulate the unfolded protein response by a direct interaction with IRE1α. Science 312, 572–576 (2006).

    CAS  PubMed  Google Scholar 

  69. Zinszner, H. et al. CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev. 12, 982–995 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Marciniak, S. J. et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 18, 3066–3077 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. 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 

  72. 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).

    CAS  PubMed  Google Scholar 

  73. Mazure, N. M., Chen, E. Y., Laderoute, K. R. & Giaccia, A. J. Induction of vascular endothelial growth factor by hypoxia is modulated by a phosphatidylinositol 3-kinase/Akt signaling pathway in Ha-ras-transformed cells through a hypoxia inducible factor-1 transcriptional element. Blood 90, 3322–3331 (1997).

    CAS  PubMed  Google Scholar 

  74. Zhong, H. et al. Modulation of hypoxia-inducible factor 1a 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 

  75. 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). This study was one of the first to provide a link between receptor tyrosine kinase signalling through mTOR to regulate HIF.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Hudson, C. C. et al. Regulation of hypoxia-inducible factor 1α expression and function by the mammalian target of rapamycin. Mol. Cell. Biol. 22, 7004–7014 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Thomas, G. V. et al. Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nature Med. 12, 122–127 (2006).

    CAS  PubMed  Google Scholar 

  78. Land, S. C. & Tee, A. R. Hypoxia-inducible factor 1α is regulated by the mammalian target of rapamycin (mTOR) via an mTOR signaling motif. J. Biol. Chem. 282, 20534–20543 (2007).

    CAS  PubMed  Google Scholar 

  79. Gorlach, A. et al. Efficient translation of mouse hypoxia-inducible factor-1α under normoxic and hypoxic conditions. Biochim. Biophys. Acta 1493, 125–134 (2000).

    CAS  PubMed  Google Scholar 

  80. Stein, I. et al. Translation of vascular endothelial growth factor mRNA by internal ribosome entry: implications for translation under hypoxia. Mol. Cell. Biol. 18, 3112–3119 (1998). This study identified an internal ribosome entry site in VEGFA, which can promote its selective synthesis under conditions where mTOR is inhibited.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Lang, K. J., Kappel, A. & Goodall, G. J. Hypoxia-inducible factor-1α mRNA contains an internal ribosome entry site that allows efficient translation during normoxia and hypoxia. Mol. Biol. Cell 13, 1792–1801 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bert, A. G., Grepin, R., Vadas, M. A. & Goodall, G. J. Assessing IRES activity in the HIF-1α and other cellular 5′ UTRs. RNA 12, 1074–1083 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Young, R. M. et al. Hypoxia-mediated selective mRNA translation by an internal ribosome entry site independent mechanism. J. Biol. Chem. 283, 16309–16319 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. 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 

  85. 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 

  86. Edinger, A. L., Linardic, C. M., Chiang, G. G., Thompson, C. B. & Abraham, R. T. Differential effects of rapamycin on mammalian target of rapamycin signaling functions in mammalian cells. Cancer Res. 63, 8451–8460 (2003).

    CAS  PubMed  Google Scholar 

  87. Semenza, G. L. Targeting HIF-1 for cancer therapy. Nature Rev. Cancer 3, 721–732 (2003).

    CAS  Google Scholar 

  88. Zhang, H. et al. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell 11, 407–420 (2007).

    CAS  PubMed  Google Scholar 

  89. DeBerardinis, R. J., Lum, J. J., Hatzivassiliou, G. & Thompson, C. B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7, 11–20 (2008).

    CAS  PubMed  Google Scholar 

  90. Dang, C. V., Kim, J. W., Gao, P. & Yustein, J. The interplay between MYC and HIF in cancer. Nature Rev. Cancer 8, 51–56 (2008).

    CAS  Google Scholar 

  91. Gordan, J. D., Thompson, C. B. & Simon, M. C. HIF and c-Myc: sibling rivals for control of cancer cell metabolism and proliferation. Cancer Cell 12, 108–113 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Corn, P. G. et al. Mxi1 is induced by hypoxia in a HIF-1-dependent manner and protects cells from c-Myc-induced apoptosis. Cancer Biol. Ther. 4, 1285–1294 (2005).

    CAS  PubMed  Google Scholar 

  93. Gordan, J. D., Bertout, J. A., Hu, C. J., Diehl, J. A. & Simon, M. C. HIF-2α promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell 11, 335–347 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Karantza-Wadsworth, V. et al. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 21, 1621–1635 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Mathew, R., Karantza-Wadsworth, V. & White, E. Role of autophagy in cancer. Nature Rev. Cancer 7, 961–967 (2007).

    CAS  Google Scholar 

  96. Toescu, E. C. Hypoxia sensing and pathways of cytosolic Ca2+ increases. Cell Calcium 36, 187–199 (2004).

    CAS  PubMed  Google Scholar 

  97. Hoyer-Hansen, M. & Jaattela, M. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ. 14, 1576–1582 (2007).

    CAS  PubMed  Google Scholar 

  98. Sakaki, K., Wu, J. & Kaufman, R. J. Protein kinase C-θ is required for autophagy in response to stress in the endoplasmic reticulum. J. Biol. Chem. 283, 15370–15380 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Ozcan, U. et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306, 457–461 (2004).

    PubMed  Google Scholar 

  100. Ozcan, U. et al. Loss of the tuberous sclerosis complex tumor suppressors triggers the unfolded protein response to regulate insulin signaling and apoptosis. Mol. Cell 29, 541–551 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Harding, H. P. et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 11, 619–633 (2003). This study provides evidence to suggest that UPR activation and upregulation of ATF4 is an important mediator of sensitivity to oxidative stress.

    CAS  PubMed  Google Scholar 

  102. Koditz, J. et al. Oxygen-dependent ATF-4 stability is mediated by the PHD3 oxygen sensor. Blood 110, 3610–3617 (2007).

    PubMed  Google Scholar 

  103. Maxwell, P. H. The HIF pathway in cancer. Semin. Cell Dev. Biol. 16, 523–530 (2005).

    CAS  PubMed  Google Scholar 

  104. Koritzinsky, M. & Wouters, B. G. Hypoxia and regulation of messenger RNA translation. Methods Enzymol. 435, 247–273 (2007).

    CAS  PubMed  Google Scholar 

  105. Kulshreshtha, R. et al. A microRNA signature of hypoxia. Mol. Cell. Biol. 27, 1859–1867 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Hebert, C., Norris, K., Scheper, M. A., Nikitakis, N. & Sauk, J. J. High mobility group A2 is a target for miRNA-98 in head and neck squamous cell carcinoma. Mol. Cancer 6, 5 (2007).

    PubMed  PubMed Central  Google Scholar 

  107. Fasanaro, P. et al. MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine-kinase ligand Ephrin-A3. J. Biol. Chem. 283, 15878–15883 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Hua, Z. et al. MiRNA-directed regulation of VEGF and other angiogenic factors under hypoxia. PLoS ONE 1, e116 (2006).

    PubMed  PubMed Central  Google Scholar 

  109. Giannakakis, A. et al. miR-210 links hypoxia with cell cycle regulation and is deleted in hum an epithelial ovarian cancer. Cancer Biol. Ther. 7, 255–264 (2007).

    PubMed  Google Scholar 

  110. Camps, C. et al. hsa-miR-210 is induced by hypoxia and is an independent prognostic factor in breast cancer. Clin. Cancer Res. 14, 1340–1348 (2008).

    CAS  PubMed  Google Scholar 

  111. Tu, B. P. & Weissman, J. S. The FAD- and O2-dependent reaction cycle of Ero1-mediated oxidative protein folding in the endoplasmic reticulum. Mol. Cell 10, 983–994 (2002). This in vitro study indicates that O 2 may serve as the final electron acceptor to drive disulfide bond formation in the ER.

    CAS  PubMed  Google Scholar 

  112. Gray, L. H., Conger, A. D., Ebert, M., Hornsey, S. & Scott, O. C. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br. J. Radiol. 26, 638–648 (1953).

    CAS  PubMed  Google Scholar 

  113. Brown, J. M. & Wilson, W. R. Exploiting tumour hypoxia in cancer treatment. Nature Rev. Cancer 4, 437–447 (2004).

    CAS  Google Scholar 

  114. Brown, J. M. SR 4233 (tirapazamine): a new anticancer drug exploiting hypoxia in solid tumours. Br. J. Cancer 67, 1163–1170 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Melillo, G. Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev. 26, 341–352 (2007).

    CAS  PubMed  Google Scholar 

  116. Weppler, S. A. et al. Response of U87 glioma xenografts treated with concurrent rapamycin and fractionated radiotherapy: possible role for thrombosis. Radiother. Oncol. 82, 96–104 (2007).

    CAS  PubMed  Google Scholar 

  117. Guba, M. et al. Rapamycin induces tumor-specific thrombosis via tissue factor in the presence of VEGF. Blood 105, 4463–4469 (2005).

    CAS  PubMed  Google Scholar 

  118. Jain, R. K. Molecular regulation of vessel maturation. Nature Med. 9, 685–693 (2003).

    CAS  PubMed  Google Scholar 

  119. Gupta, A. K. et al. The HIV protease inhibitor nelfinavir downregulates Akt phosphorylation by inhibiting proteasomal activity and inducing the unfolded protein response. Neoplasia 9, 271–278 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Pyrko, P. et al. HIV-1 protease inhibitors nelfinavir and atazanavir induce malignant glioma death by triggering endoplasmic reticulum stress. Cancer Res. 67, 10920–10928 (2007).

    CAS  PubMed  Google Scholar 

  121. Gupta, A. K., Cerniglia, G. J., Mick, R., McKenna, W. G. & Muschel, R. J. HIV protease inhibitors block Akt signaling and radiosensitize tumor cells both in vitro and in vivo. Cancer Res. 65, 8256–8265 (2005).

    CAS  PubMed  Google Scholar 

  122. Pore, N. et al. Nelfinavir down-regulates hypoxia-inducible factor 1α and VEGF expression and increases tumor oxygenation: implications for radiotherapy. Cancer Res. 66, 9252–9259 (2006).

    CAS  PubMed  Google Scholar 

  123. Gills, J. J. et al. Nelfinavir, A lead HIV protease inhibitor, is a broad-spectrum, anticancer agent that induces endoplasmic reticulum stress, autophagy, and apoptosis in vitro and in vivo. Clin. Cancer Res. 13, 5183–5194 (2007).

    CAS  PubMed  Google Scholar 

  124. Luwor, R. B., Lu, Y., Li, X., Mendelsohn, J. & Fan, Z. The antiepidermal growth factor receptor monoclonal antibody cetuximab/C225 reduces hypoxia-inducible factor-1α, leading to transcriptional inhibition of vascular endothelial growth factor expression. Oncogene 24, 4433–4441 (2005).

    CAS  PubMed  Google Scholar 

  125. Huang, S. M. & Harari, P. M. Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: inhibition of damage repair, cell cycle kinetics, and tumor angiogenesis. Clin. Cancer Res. 6, 2166–2174 (2000).

    CAS  PubMed  Google Scholar 

  126. Hardee, M. E. et al. Her2/neu signaling blockade improves tumor oxygenation in a multifactorial fashion in Her2/neu+ tumors. Cancer Chemother. Pharmacol. 26 Mar 2008 (doi:10.1007/s00280-008-0729-3).

    PubMed  PubMed Central  Google Scholar 

  127. Guan, H., Jia, S. F., Zhou, Z., Stewart, J. & Kleinerman, E. S. Herceptin down-regulates HER-2/neu and vascular endothelial growth factor expression and enhances taxol-induced cytotoxicity of human Ewing's sarcoma cells in vitro and in vivo. Clin. Cancer Res. 11, 2008–2017 (2005).

    CAS  PubMed  Google Scholar 

  128. Lu, Y., Liang, K., Li, X. & Fan, Z. Responses of cancer cells with wild-type or tyrosine kinase domain-mutated epidermal growth factor receptor (EGFR) to EGFR-targeted therapy are linked to downregulation of hypoxia-inducible factor-1α. Mol. Cancer 6, 63 (2007).

    PubMed  PubMed Central  Google Scholar 

  129. Warburton, C. et al. Treatment of HER-2/neu overexpressing breast cancer xenograft models with trastuzumab (Herceptin) and gefitinib (ZD1839): drug combination effects on tumor growth, HER-2/neu and epidermal growth factor receptor expression, and viable hypoxic cell fraction. Clin. Cancer Res. 10, 2512–2524 (2004).

    CAS  PubMed  Google Scholar 

  130. Solomon, B. et al. Modulation of intratumoral hypoxia by the epidermal growth factor receptor inhibitor gefitinib detected using small animal PET imaging. Mol. Cancer Ther. 4, 1417–1422 (2005).

    CAS  PubMed  Google Scholar 

  131. Hirata, A. et al. ZD1839 (Iressa) induces antiangiogenic effects through inhibition of epidermal growth factor receptor tyrosine kinase. Cancer Res. 62, 2554–2560 (2002).

    CAS  PubMed  Google Scholar 

  132. Vlahovic, G. et al. Treatment with imatinib improves drug delivery and efficacy in NSCLC xenografts. Br. J. Cancer 97, 735–740 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Murphy, D. A. et al. Inhibition of tumor endothelial ERK activation, angiogenesis, and tumor growth by sorafenib (BAY43–9006). Am. J. Pathol. 169, 1875–1885 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Del Bufalo, D. et al. Antiangiogenic potential of the mammalian target of rapamycin inhibitor temsirolimus. Cancer Res. 66, 5549–5554 (2006).

    CAS  PubMed  Google Scholar 

  135. Wan, X., Shen, N., Mendoza, A., Khanna, C. & Helman, L. J. CCI-779 inhibits rhabdomyosarcoma xenograft growth by an antiangiogenic mechanism linked to the targeting of mTOR/Hif-1α/VEGF signaling. Neoplasia 8, 394–401 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Huynh, H., Teo, C. C. & Soo, K. C. Bevacizumab and rapamycin inhibit tumor growth in peritoneal model of human ovarian cancer. Mol. Cancer Ther. 6, 2959–2966 (2007).

    CAS  PubMed  Google Scholar 

  137. Shi, Y. et al. Farnesyltransferase inhibitor effects on prostate tumor micro-environment and radiation survival. Prostate 62, 69–82 (2005).

    CAS  PubMed  Google Scholar 

  138. Delmas, C. et al. The farnesyltransferase inhibitor R115777 reduces hypoxia and matrix metalloproteinase 2 expression in human glioma xenograft. Clin. Cancer Res. 9, 6062–6068 (2003).

    CAS  PubMed  Google Scholar 

  139. Birle, D. C. & Hedley, D. W. Suppression of the hypoxia-inducible factor-1 response in cervical carcinoma xenografts by proteasome inhibitors. Cancer Res. 67, 1735–1743 (2007).

    CAS  PubMed  Google Scholar 

  140. Kaluz, S., Kaluzova, M. & Stanbridge, E. J. Proteasomal inhibition attenuates transcriptional activity of hypoxia-inducible factor 1 (HIF-1) via specific effect on the HIF-1α C-terminal activation domain. Mol. Cell. Biol. 26, 5895–5907 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Roccaro, A. M. et al. Bortezomib mediates antiangiogenesis in multiple myeloma via direct and indirect effects on endothelial cells. Cancer Res. 66, 184–191 (2006).

    CAS  PubMed  Google Scholar 

  142. Brignole, C. et al. Effect of bortezomib on human neuroblastoma cell growth, apoptosis, and angiogenesis. J. Natl Cancer Inst. 98, 1142–1157 (2006).

    CAS  PubMed  Google Scholar 

  143. Pore, N., Gupta, A. K., Cerniglia, G. J. & Maity, A. HIV protease inhibitors decrease VEGF/HIF-1α expression and angiogenesis in glioblastoma cells. Neoplasia 8, 889–895 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Isaacs, J. S. et al. Hsp90 regulates a von Hippel Lindau-independent hypoxia-inducible factor-1α -degradative pathway. J. Biol. Chem. 277, 29936–29944 (2002).

    CAS  PubMed  Google Scholar 

  145. Mabjeesh, N. J. et al. Geldanamycin induces degradation of hypoxia-inducible factor 1α protein via the proteosome pathway in prostate cancer cells. Cancer Res. 62, 2478–2482 (2002).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We wish to acknowledge the excellent work and contributions from all members of our teams in both Toronto and Maastricht. We also wish to thank C. Koumenis for our ongoing productive and stimulating collaboration. These studies received financial support from the Dutch Science Organization (ZonMW-NWO Top grant 912-03-047 to BW and VENI grant 916.56.015 to M.K.), the Dutch Cancer Society (KWF grant UM 2003-2821 to B.W.), and the EU 6th framework programme (Euroxy programme to B.G.W.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bradly G. Wouters.

Related links

Related links

DATABASES

National Cancer Institute

breast cancer

National Cancer Institute Drug Dictionary

nelfinavir

rapamycin

tirapazamine

OMIM

Huntington disease

Peutz–Jeghers syndrome

tuberous sclerosis syndrome

Glossary

Rapamycin

An antifungal macrolide produced by bacteria, commonly used to prevent post-transplantation organ rejection owing to its immunosuppressive action. Rapamycin binds FKBP12, which in turn binds and inhibits mTORC1.

Autophagy

Literally, “self eating”, autophagy is a process by which cellular components are captured, degraded and recycled using the lysosomal machinery. It is the only cellular process that can degrade covalently linked protein complexes and organelles.

Protein maturation

Maturation of secreted or membrane-destined proteins typically involves N-linked glycosylation during co-translational import to the ER, as well as glycan modification, oxidative folding and isomerization due to disulphide bond formation within the ER lumen. Chaperones accompany the proteins to aid folding and prevent aggregation. Strict quality control is applied before protein release from the folding machinery and export to the Golgi.

Hypoxic phenotype

Characterized by adaptive responses such as decreased proliferation and protein synthesis, high rates of glycolysis and anaerobic metabolism, increased secretion of angiogenic factors and increased metastatic potential. These, together with susceptibility to death pathways, ultimately determine hypoxia tolerance.

mTOR complex 1

(mTORC1). One of two protein complexes (along with mTORC2) known to harbour mTOR. mTORC1 contains mTOR, mLST8, PRAS40 and raptor. Its activation stimulates protein synthesis and cell growth through phosphorylation of p70S6K, 4E-BP and EEF2K.

TSC1–TSC2 complex

This complex of the tumour suppressors TSC1 and TSC2, also known as hamartin and tuberin, respectively, displays GTPase-activating protein activity towards RHEB, resulting in negative regulation of mTORC1. Its activity is negatively regulated by Akt and Erk-mediated phosphorylation of TSC2, and positively regulated by AMPK-dependent TSC2 phosphorylation.

Cap-dependent translation

The main mode of mRNA translation initiation that relies on recruitment of the small ribosomal subunit and associated factors via the 5′ cap structure of the mRNA. This requires binding of the EIF4F complex at the cap, which acts as a scaffold for the ribosomal complex.

Hamartomas

Benign tumours that may cause symptoms due to their location and size.

Tunicamycin

A mixture of homologous antibiotics that prevent N-linked glycosylation of proteins in the ER lumen by inhibiting the enzyme N-acetylglucosamine transferase. As a result, it is a potent inducer of the UPR.

Thapsigargin

An inhibitor of Ca2+ pumps located in the sarcoplasmic reticulum and ER membranes. Depletion of ER calcium stores results in the induction of the UPR.

5′ Terminal oligopyrimidine

A stretch of nucleotides at the 5′ end of the mRNA, consisting of the pyrimidine derivatives cytosine and uracil. These sequences are present in the mRNA of some ribosomal proteins and translation factors and confer extreme responsiveness to positive and negative growth signalling in terms of translation efficiency. The sequence requirements and molecular mechanism are not well understood.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wouters, B., Koritzinsky, M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nat Rev Cancer 8, 851–864 (2008). https://doi.org/10.1038/nrc2501

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc2501

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