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:

mTOR signalling and cellular metabolism are mutual determinants in cancer

Abstract

Oncogenic signalling and metabolic alterations are interrelated in cancer cells. mTOR, which is frequently activated in cancer, controls cell growth and metabolism. mTOR signalling regulates amino acid, glucose, nucleotide, fatty acid and lipid metabolism. Conversely, metabolic inputs, such as amino acids, activate mTOR. In this Review, we discuss how mTOR signalling rewires cancer cell metabolism and delineate how changes in metabolism, in turn, sustain mTOR signalling and tumorigenicity. Several drugs are being developed to perturb cancer cell metabolism. However, their efficacy as stand-alone therapies, similar to mTOR inhibitors, is limited. Here, we discuss how the interdependence of mTOR signalling and metabolism can be exploited for cancer therapy.

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

Fig. 1: Activation of mTOR signalling.
Fig. 2: mTOR-driven reprogramming of amino acid metabolism.
Fig. 3: mTOR controls glucose metabolism in cancer.
Fig. 4: Rewiring of nucleotide metabolism by mTOR.
Fig. 5: Regulation of fatty acid and lipid metabolism.

Similar content being viewed by others

References

  1. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    PubMed Central  Google Scholar 

  2. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhang, Y. et al. A pan-cancer proteogenomic atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell 31, 820–832 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Alessi, D. R. et al. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Bα. Curr. Biol. 7, 261–269 (1997).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  6. Inoki, K., Li, Y., Xu, T. & Guan, K. L. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 17, 1829–1834 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Long, X., Lin, Y., Ortiz-Vega, S., Yonezawa, K. & Avruch, J. Rheb binds and regulates the mTOR kinase. Curr. Biol. 15, 702–713 (2005).

    CAS  PubMed  Google Scholar 

  8. Yang, H. et al. Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40. Nature 552, 368–373 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Gonzalez, A. & Hall, M. N. Nutrient sensing and TOR signaling in yeast and mammals. EMBO J. 36, 397–408 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Wolfson, R. L. & Sabatini, D. M. The dawn of the age of amino acid sensors for the mTORC1 pathway. Cell Metab. 26, 301–309 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wyant, G. A. et al. mTORC1 activator SLC38A9 is required to efflux essential amino acids from lysosomes and use protein as a nutrient. Cell 171, 642–654 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Duran, R. V. et al. Glutaminolysis activates Rag-mTORC1 signaling. Mol. Cell 47, 349–358 (2012).

    CAS  PubMed  Google Scholar 

  13. Jewell, J. L. et al. Differential regulation of mTORC1 by leucine and glutamine. Science 347, 194–198 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Liu, P. et al. PtdIns(3,4,5)P3-dependent activation of the mTORC2 kinase complex. Cancer Discov. 5, 1194–1209 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Zinzalla, V., Stracka, D., Oppliger, W. & Hall, M. N. Activation of mTORC2 by association with the ribosome. Cell 144, 757–768 (2011).

    CAS  PubMed  Google Scholar 

  16. Thorpe, L. M., Yuzugullu, H. & Zhao, J. J. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 15, 7–24 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Mayer, I. A. & Arteaga, C. L. The PI3K/AKT pathway as a target for cancer treatment. Annu. Rev. Med. 67, 11–28 (2016).

    CAS  PubMed  Google Scholar 

  18. Benjamin, D., Colombi, M., Moroni, C. & Hall, M. N. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat. Rev. Drug Discov. 10, 868–880 (2011).

    CAS  PubMed  Google Scholar 

  19. Wise, D. R. & Thompson, C. B. Glutamine addiction: a new therapeutic target in cancer. Trends Biochem. Sci. 35, 427–433 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Yang, L., Venneti, S. & Nagrath, D. Glutaminolysis: a hallmark of cancer metabolism. Annu. Rev. Biomed. Eng. 19, 163–194 (2017).

    CAS  PubMed  Google Scholar 

  21. Bhutia, Y. D., Babu, E., Ramachandran, S. & Ganapathy, V. Amino acid transporters in cancer and their relevance to “glutamine addiction”: novel targets for the design of a new class of anticancer drugs. Cancer Res. 75, 1782–1788 (2015).

    CAS  PubMed  Google Scholar 

  22. Lu, J. et al. Effects of targeting SLC1A5 on inhibiting gastric cancer growth and tumor development in vitro and in vivo. Oncotarget 8, 76458–76467 (2017).

    PubMed  PubMed Central  Google Scholar 

  23. Wang, Q. et al. Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development. J. Pathol. 236, 278–289 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. van Geldermalsen, M. et al. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 35, 3201–3208 (2016).

    PubMed  Google Scholar 

  25. Broer, A., Rahimi, F. & Broer, S. Deletion of amino acid transporter ASCT2 (SLC1A5) reveals an essential role for transporters SNAT1 (SLC38A1) and SNAT2 (SLC38A2) to sustain glutaminolysis in cancer cells. J. Biol. Chem. 291, 13194–13205 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Csibi, A. et al. The mTORC1/S6K1 pathway regulates glutamine metabolism through the eIF4B-dependent control of c-Myc translation. Curr. Biol. 24, 2274–2280 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Gao, P. et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458, 762–765 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Csibi, A. et al. The mTORC1 pathway stimulates glutamine metabolism and cell proliferation by repressing SIRT4. Cell 153, 840–854 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. van der Vos, K. E. et al. Modulation of glutamine metabolism by the PI(3)K-PKB-FOXO network regulates autophagy. Nat. Cell Biol. 14, 829–837 (2012).

    PubMed  Google Scholar 

  30. Momcilovic, M. et al. The GSK3 signaling axis regulates adaptive glutamine metabolism in lung squamous cell carcinoma. Cancer Cell 33, 905–921 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Schulte, M. L. et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat. Med. 24, 194–202 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03057600 (2018).

  33. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02071862 (2018).

  34. Lu, Y. et al. Overexpression of arginine transporter CAT-1 is associated with accumulation of L-arginine and cell growth in human colorectal cancer tissue. PLOS ONE 8, e73866 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Abdelmagid, S. A., Rickard, J. A., McDonald, W. J., Thomas, L. N. & Too, C. K. CAT-1-mediated arginine uptake and regulation of nitric oxide synthases for the survival of human breast cancer cell lines. J. Cell. Biochem. 112, 1084–1092 (2011).

    CAS  PubMed  Google Scholar 

  36. Kishikawa, T. et al. Decreased miR122 in hepatocellular carcinoma leads to chemoresistance with increased arginine. Oncotarget 6, 8339–8352 (2015).

    PubMed  PubMed Central  Google Scholar 

  37. Masuda, M. et al. Alternative mammalian target of rapamycin (mTOR) signal activation in sorafenib-resistant hepatocellular carcinoma cells revealed by array-based pathway profiling. Mol. Cell. Proteomics 13, 1429–1438 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Luengo, A., Gui, D. Y. & Vander Heiden, M. G. Targeting metabolism for cancer therapy. Cell Chem. Biol. 24, 1161–1180 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Dillon, B. J. et al. Incidence and distribution of argininosuccinate synthetase deficiency in human cancers: a method for identifying cancers sensitive to arginine deprivation. Cancer 100, 826–833 (2004).

    CAS  PubMed  Google Scholar 

  40. Delage, B. et al. Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. Int. J. Cancer 126, 2762–2772 (2010).

    CAS  PubMed  Google Scholar 

  41. Rabinovich, S. et al. Diversion of aspartate in ASS1-deficient tumours fosters de novo pyrimidine synthesis. Nature 527, 379–383 (2015). This paper shows that loss of ASS1 expression promotes pyrimidine biosynthesis by aspartate accumulation.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Tsai, W. B. et al. Activation of Ras/PI3K/ERK pathway induces c-Myc stabilization to upregulate argininosuccinate synthetase, leading to arginine deiminase resistance in melanoma cells. Cancer Res. 72, 2622–2633 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Patil, M. D., Bhaumik, J., Babykutty, S., Banerjee, U. C. & Fukumura, D. Arginine dependence of tumor cells: targeting a chink in cancer’s armor. Oncogene 35, 4957–4972 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02899286 (2017).

  45. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02285101 (2018).

  46. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01497925 (2018).

  47. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03254732 (2018).

  48. Shen, W. et al. A novel and promising therapeutic approach for NSCLC: recombinant human arginase alone or combined with autophagy inhibitor. Cell Death Dis. 8, e2720 (2017).

    PubMed  PubMed Central  Google Scholar 

  49. Tsai, W. B. et al. Resistance to arginine deiminase treatment in melanoma cells is associated with induced argininosuccinate synthetase expression involving c-Myc/HIF-1alpha/Sp4. Mol. Cancer Ther. 8, 3223–3233 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Casero, R. A. Jr, Murray Stewart, T. & Pegg, A. E. Polyamine metabolism and cancer: treatments, challenges and opportunities. Nat. Rev. Cancer 18, 681–695 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Bello-Fernandez, C., Packham, G. & Cleveland, J. L. The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proc. Natl Acad. Sci. USA 90, 7804–7808 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Rousseau, D., Kaspar, R., Rosenwald, I., Gehrke, L. & Sonenberg, N. Translation initiation of ornithine decarboxylase and nucleocytoplasmic transport of cyclin D1 mRNA are increased in cells overexpressing eukaryotic initiation factor 4E. Proc. Natl Acad. Sci. USA 93, 1065–1070 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Origanti, S. et al. Ornithine decarboxylase mRNA is stabilized in an mTORC1-dependent manner in Ras-transformed cells. Biochem. J. 442, 199–207 (2012).

    CAS  PubMed  Google Scholar 

  54. Ochocki, J. D. et al. Arginase 2 suppresses renal carcinoma progression via biosynthetic cofactor pyridoxal phosphate depletion and increased polyamine toxicity. Cell Metab. 27, 1263–1280.e6 (2018).

    PubMed  PubMed Central  Google Scholar 

  55. Zabala-Letona, A. et al. mTORC1-dependent AMD1 regulation sustains polyamine metabolism in prostate cancer. Nature 547, 109–113 (2017). This paper reveals that mTORC1 promotes polyamine biosynthesis by stabilization of (pro)AMD1.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang, Q. & Holst, J. L-type amino acid transport and cancer: targeting the mTORC1 pathway to inhibit neoplasia. Am. J. Cancer Res. 5, 1281–1294 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Nicklin, P. et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136, 521–534 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Grzes, K. M. et al. Control of amino acid transport coordinates metabolic reprogramming in T cell malignancy. Leukemia 31, 2771–2779 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Yue, M., Jiang, J., Gao, P., Liu, H. & Qing, G. Oncogenic MYC activates a feedforward regulatory loop promoting essential amino acid metabolism and tumorigenesis. Cell Rep. 21, 3819–3832 (2017).

    CAS  PubMed  Google Scholar 

  60. Elorza, A. et al. HIF2alpha acts as an mTORC1 activator through the amino acid carrier SLC7A5. Mol. Cell 48, 681–691 (2012).

    CAS  PubMed  Google Scholar 

  61. 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 

  62. Xiao, F. et al. Leucine deprivation inhibits proliferation and induces apoptosis of human breast cancer cells via fatty acid synthase. Oncotarget 7, 63679–63689 (2015).

    Google Scholar 

  63. Wang, Q. et al. Androgen receptor and nutrient signaling pathways coordinate the demand for increased amino acid transport during prostate cancer progression. Cancer Res. 71, 7525–7536 (2011).

    CAS  PubMed  Google Scholar 

  64. Zhang, W. et al. Inhibition of tumor growth progression by antiandrogens and mTOR inhibitor in a Pten-deficient mouse model of prostate cancer. Cancer Res. 69, 7466–7472 (2009).

    CAS  PubMed  Google Scholar 

  65. Tonjes, M. et al. BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild-type IDH1. Nat. Med. 19, 901–908 (2013).

    PubMed  PubMed Central  Google Scholar 

  66. Xu, M. et al. BCAT1 promotes tumor cell migration and invasion in hepatocellular carcinoma. Oncol. Lett. 12, 2648–2656 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Zhang, L. & Han, J. Branched-chain amino acid transaminase 1 (BCAT1) promotes the growth of breast cancer cells through improving mTOR-mediated mitochondrial biogenesis and function. Biochem. Biophys. Res. Commun. 486, 224–231 (2017).

    CAS  PubMed  Google Scholar 

  68. Yoshikawa, R. et al. ECA39 is a novel distant metastasis-related biomarker in colorectal cancer. World J. Gastroenterol. 12, 5884–5889 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Hattori, A. et al. Cancer progression by reprogrammed BCAA metabolism in myeloid leukaemia. Nature 545, 500–504 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Google Scholar 

  71. Tran, Q., Lee, H., Park, J., Kim, S. H. & Park, J. Targeting cancer metabolism — revisiting the Warburg effects. Toxicol. Res. 32, 177–193 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Buller, C. L. et al. A GSK-3/TSC2/mTOR pathway regulates glucose uptake and GLUT1 glucose transporter expression. Am. J. Physiol. Cell Physiol. 295, C836–C843 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Masui, K. et al. mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 18, 726–739 (2013). This paper demonstrates how mTORC2 controls glucose metabolism via FOXO and MYC in GBM.

    CAS  PubMed  Google Scholar 

  74. Carvalho, K. C. et al. GLUT1 expression in malignant tumors and its use as an immunodiagnostic marker. Clinics 66, 965–972 (2011).

    PubMed  PubMed Central  Google Scholar 

  75. Wolf, A. et al. Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme. J. Exp. Med. 208, 313–326 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Wang, L. et al. Hexokinase 2-mediated Warburg effect is required for PTEN- and p53-deficiency-driven prostate cancer growth. Cell Rep. 8, 1461–1474 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Betz, C. et al. mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology. Proc. Natl Acad. Sci. USA 110, 12526–12534 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Raez, L. E. et al. A phase I dose-escalation trial of 2-deoxy-D-glucose alone or combined with docetaxel in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 71, 523–530 (2013).

    CAS  PubMed  Google Scholar 

  79. Broecker-Preuss, M., Becher-Boveleth, N., Bockisch, A., Duhrsen, U. & Muller, S. Regulation of glucose uptake in lymphoma cell lines by c-MYC- and PI3K-dependent signaling pathways and impact of glycolytic pathways on cell viability. J. Transl Med. 15, 158 (2017).

    PubMed  PubMed Central  Google Scholar 

  80. Wang, C. et al. PKM2 promotes cell migration and inhibits autophagy by mediating PI3K/AKT activation and contributes to the malignant development of gastric cancer. Sci. Rep. 7, 2886 (2017).

    PubMed  PubMed Central  Google Scholar 

  81. Goldberg, M. S. & Sharp, P. A. Pyruvate kinase M2-specific siRNA induces apoptosis and tumor regression. J. Exp. Med. 209, 217–224 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Sun, Q. et al. Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth. Proc. Natl Acad. Sci. USA 108, 4129–4134 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. He, C. L. et al. Pyruvate kinase M2 activates mTORC1 by phosphorylating AKT1S1. Sci. Rep. 6, 21524 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Ye, J. et al. Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation. Proc. Natl Acad. Sci. USA 109, 6904–6909 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Lynch, T., Moloughney, J. G. & Jacinto, E. in PI3K-mTOR in Cancer and Cancer Therapy — Cancer Drug Discovery and Development (eds Dey, N., De, P. & Leyland-Jones, B.) 29–63 (Humana Press, 2016).

  86. Luo, W. et al. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell 145, 732–744 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Wang, X., Zhang, F. & Wu, X. R. Inhibition of pyruvate kinase M2 markedly reduces chemoresistance of advanced bladder cancer to cisplatin. Sci. Rep. 7, 45983 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Liang, W. et al. Shikonin induces mitochondria-mediated apoptosis and enhances chemotherapeutic sensitivity of gastric cancer through reactive oxygen species. Sci. Rep. 6, 38267 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Hui, S. et al. Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115–118 (2017).

    PubMed  PubMed Central  Google Scholar 

  91. Allen, E. et al. Metabolic symbiosis enables adaptive resistance to anti-angiogenic therapy that is dependent on mTOR signaling. Cell Rep. 15, 1144–1160 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Pisarsky, L. et al. Targeting metabolic symbiosis to overcome resistance to anti-angiogenic therapy. Cell Rep. 15, 1161–1174 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01791595 (2018).

  94. Patra, K. C. & Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 39, 347–354 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Wang, J. et al. Overexpression of G6PD is associated with poor clinical outcome in gastric cancer. Tumour Biol. 33, 95–101 (2012).

    PubMed  Google Scholar 

  96. Evert, M. et al. V-AKT murine thymoma viral oncogene homolog/mammalian target of rapamycin activation induces a module of metabolic changes contributing to growth in insulin-induced hepatocarcinogenesis. Hepatology 55, 1473–1484 (2012).

    CAS  PubMed  Google Scholar 

  97. Duvel, K. et al. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol. Cell 39, 171–183 (2010).

    PubMed  PubMed Central  Google Scholar 

  98. Juvekar, A. et al. Phosphoinositide 3-kinase inhibitors induce DNA damage through nucleoside depletion. Proc. Natl Acad. Sci. USA 113, 4338–4347 (2016).

    Google Scholar 

  99. Cunningham, J. T., Moreno, M. V., Lodi, A., Ronen, S. M. & Ruggero, D. Protein and nucleotide biosynthesis are coupled by a single rate-limiting enzyme, PRPS2, to drive cancer. Cell 157, 1088–1103 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Ben-Sahra, I., Hoxhaj, G., Ricoult, S. J. H., Asara, J. M. & Manning, B. D. mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle. Science 351, 728–733 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Hoxhaj, G. et al. The mTORC1 signaling network senses changes in cellular purine nucleotide levels. Cell Rep. 21, 1331–1346 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Emmanuel, N. et al. Purine nucleotide availability regulates mTORC1 activity through the Rheb GTPase. Cell Rep. 19, 2665–2680 (2017).

    CAS  PubMed  Google Scholar 

  103. Valvezan, A. J. et al. mTORC1 couples nucleotide synthesis to nucleotide demand resulting in a targetable metabolic vulnerability. Cancer Cell 32, 624–638 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Miltenberger, R. J., Sukow, K. A. & Farnham, P. J. An E-box-mediated increase in cad transcription at the G1/S-phase boundary is suppressed by inhibitory c-Myc mutants. Mol. Cell. Biol. 15, 2527–2535 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Ben-Sahra, I., Howell, J. J., Asara, J. M. & Manning, B. D. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 339, 1323–1328 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Robitaille, A. M. et al. Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis. Science 339, 1320–1323 (2013).

    CAS  PubMed  Google Scholar 

  107. Shackelford, D. B. & Shaw, R. J. The LKB1–AMPK pathway: metabolism and growth control in tumour suppression. Nat. Rev. Cancer 9, 563–575 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Kim, J. et al. CPS1 maintains pyrimidine pools and DNA synthesis in KRAS/LKB1-mutant lung cancer cells. Nature 546, 168–172 (2017). This paper reports that CPS1 maintains pyrimidine synthesis in LKB1-mutant lung cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Izzo, F. et al. Pegylated arginine deiminase treatment of patients with unresectable hepatocellular carcinoma: results from phase I/II studies. J. Clin. Oncol. 22, 1815–1822 (2004).

    CAS  PubMed  Google Scholar 

  110. Thongkum, A. et al. The combination of arginine deprivation and 5-fluorouracil improves therapeutic efficacy in argininosuccinate synthetase negative hepatocellular carcinoma. Int. J. Mol. Sci. 18, 1175 (2017).

    PubMed Central  Google Scholar 

  111. Baenke, F., Peck, B., Miess, H. & Schulze, A. Hooked on fat: the role of lipid synthesis in cancer metabolism and tumour development. Dis. Model. Mech. 6, 1353–1363 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Owen, J. L. et al. Insulin stimulation of SREBP-1c processing in transgenic rat hepatocytes requires p70 S6-kinase. Proc. Natl Acad. Sci. USA 109, 16184–16189 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Yecies, J. L. et al. Akt stimulates hepatic SREBP1c and lipogenesis through parallel mTORC1-dependent and independent pathways. Cell Metab. 14, 21–32 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Hagiwara, A. et al. Hepatic mTORC2 activates glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab. 15, 725–738 (2012).

    CAS  PubMed  Google Scholar 

  115. Ricoult, S. J., Yecies, J. L., Ben-Sahra, I. & Manning, B. D. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP. Oncogene 35, 1250–1260 (2016).

    CAS  PubMed  Google Scholar 

  116. Peterson, T. R. et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 146, 408–420 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Lee, G. et al. Post-transcriptional regulation of de novo lipogenesis by mTORC1-S6K1-SRPK2 signaling. Cell 171, 1545–1558 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Guri, Y. et al. mTORC2 promotes tumorigenesis via lipid synthesis. Cancer Cell 32, 807–823 (2017). This paper reveals an essential role of mTORC2 in HCC through lipid synthesis.

    CAS  PubMed  Google Scholar 

  119. Li, S., Oh, Y. T., Yue, P., Khuri, F. R. & Sun, S. Y. Inhibition of mTOR complex 2 induces GSK3/FBXW7-dependent degradation of sterol regulatory element-binding protein 1 (SREBP1) and suppresses lipogenesis in cancer cells. Oncogene 35, 642–650 (2016).

    CAS  PubMed  Google Scholar 

  120. Chen, Y. et al. mTOR complex-2 stimulates acetyl-CoA and de novo lipogenesis through ATP citrate lyase in HER2/PIK3CA-hyperactive breast cancer. Oncotarget 7, 25224–25240 (2016).

    PubMed  PubMed Central  Google Scholar 

  121. Stine, Z. E., Walton, Z. E., Altman, B. J., Hsieh, A. L. & Dang, C. V. MYC, metabolism, and cancer. Cancer Discov. 5, 1024–1039 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Dang, C. V. MYC on the path to cancer. Cell 149, 22–35 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02980029 (2018).

  124. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02223247 (2017).

  125. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03179904 (2018).

  126. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03032484 (2017).

  127. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02891538 (2018).

  128. White, P. J. et al. The BCKDH kinase and phosphatase integrate BCAA and lipid metabolism via regulation of ATP-citrate lyase. Cell Metab. 27, 1281–1293 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Abdul-Wahed, A., Guilmeau, S. & Postic, C. Sweet sixteenth for ChREBP: established roles and future goals. Cell Metab. 26, 324–341 (2017).

    CAS  PubMed  Google Scholar 

  130. Yan, C. et al. The mTOR inhibitor rapamycin synergizes with a fatty acid synthase inhibitor to induce cytotoxicity in ER/HER2-positive breast cancer cells. PLOS ONE 9, e97697 (2014).

    PubMed  PubMed Central  Google Scholar 

  131. Takeuchi, K. & Reue, K. Biochemistry, physiology, and genetics of GPAT, AGPAT, and lipin enzymes in triglyceride synthesis. Am. J. Physiol. Endocrinol. Metab. 296, E1195–E1209 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Quinn, W. J. III et al. mTORC1 stimulates phosphatidylcholine synthesis to promote triglyceride secretion J. Clin. Invest. 127, 4207–4215 (2017).

    PubMed  PubMed Central  Google Scholar 

  133. Kim, S. W., Kim, H. J., Chun, Y. J. & Kim, M. Y. Ceramide produces apoptosis through induction of p27(kip1) by protein phosphatase 2A-dependent Akt dephosphorylation in PC-3 prostate cancer cells. J. Toxicol. Environ. Health A 73, 1465–1476 (2010).

    CAS  PubMed  Google Scholar 

  134. Morad, S. A. & Cabot, M. C. Ceramide-orchestrated signalling in cancer cells. Nat. Rev. Cancer 13, 51–65 (2013).

    CAS  PubMed  Google Scholar 

  135. Ader, I., Brizuela, L., Bouquerel, P., Malavaud, B. & Cuvillier, O. Sphingosine kinase 1: a new modulator of hypoxia inducible factor 1alpha during hypoxia in human cancer cells. Cancer Res. 68, 8635–8642 (2008).

    CAS  PubMed  Google Scholar 

  136. Bouquerel, P. et al. Essential role for SphK1/S1P signaling to regulate hypoxia-inducible factor 2alpha expression and activity in cancer. Oncogenesis 5, e209 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Sano, O. et al. Using a biologically annotated library to analyze the anticancer mechanism of serine palmitoyl transferase (SPT) inhibitors. FEBS Open Bio 7, 495–503 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03377179 (2018).

  139. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02757326 (2018).

  140. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02939807 (2018).

  141. Xun, C. et al. Targeting sphingosine kinase 2 (SphK2) by ABC294640 inhibits colorectal cancer cell growth in vitro and in vivo. J. Exp. Clin. Cancer Res. 34, 94 (2015).

    PubMed  PubMed Central  Google Scholar 

  142. Mattaini, K. R., Sullivan, M. R. & Vander Heiden, M. G. The importance of serine metabolism in cancer. J. Cell Biol. 214, 249–257 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Adams, C. M. Role of the transcription factor ATF4 in the anabolic actions of insulin and the anti-anabolic actions of glucocorticoids. J. Biol. Chem. 282, 16744–16753 (2007).

    CAS  PubMed  Google Scholar 

  144. Gu, Y. et al. mTORC2 regulates amino acid metabolism in cancer by phosphorylation of the cystine-glutamate antiporter xCT. Mol. Cell 67, 128–138 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Sahu, N. et al. Proline starvation induces unresolved ER stress and hinders mTORC1-dependent tumorigenesis. Cell Metab. 24, 753–761 (2016). This paper demonstrates the dependence on proline for mTORC1-dependent tumorigenesis in a subset of cancer cells.

    CAS  PubMed  Google Scholar 

  146. Gu, X. et al. SAMTOR is an S-adenosylmethionine sensor for the mTORC1 pathway. Science 358, 813–818 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Castellano, B. M. et al. Lysosomal cholesterolactivates mTORC1 via an SLC38A9–Niemann-Pick C1signaling complex. Science 335, 1306–1311 (2017).

    Google Scholar 

  148. Mayers, J. R. et al. Tissue of origin dictates BCAA metabolism in mutant Kras-driven cancers. Science 353, 1161–1165 (2016). This paper reports that cancer-specific changes in BCAA metabolism are driven by the tissue of origin rather than underlying mutations.

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Kankotia, S. & Stacpoole, P. W. Dichloroacetate and cancer: new home for an orphan drug? Biochim. Biophys. Acta 1846, 617–629 (2014).

    CAS  PubMed  Google Scholar 

  150. Skrott, Z. et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature 552, 194–199 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Howell, J. J. et al. Metformin inhibits hepatic mTORC1 signaling via dose-dependent mechanisms involving AMPK and the TSC complex. Cell Metab. 25, 463–471 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Jara, J. A. & Lopez-Munoz, R. Metformin and cancer: between the bioenergetic disturbances and the antifolate activity. Pharmacol. Res. 101, 102–108 (2015).

    CAS  PubMed  Google Scholar 

  153. Pollak, M. N. Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discov. 2, 778–790 (2012).

    CAS  PubMed  Google Scholar 

  154. Morales, D. R. & Morris, A. D. Metformin in cancer treatment and prevention. Annu. Rev. Med. 66, 17–29 (2015).

    CAS  PubMed  Google Scholar 

  155. Benjamin, D. et al. Syrosingopine sensitizes cancer cells to killing by metformin. Sci. Adv. 2, e1601756 (2016). This paper demonstrates synergistic effects of metformin and syrosingopine in several cancer models.

    Google Scholar 

  156. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01529593 (2018).

  157. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02145559 (2018).

  158. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02048384 (2017).

  159. Kim, L. C., Cook, R. S. & Chen, J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 36, 2191–2201 (2017).

    CAS  PubMed  Google Scholar 

  160. Sousa, C. M. et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 536, 479–483 (2016). This paper shows that cancer-associated fibroblasts support tumour cells by secreting non-essential amino acids.

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Duluc, C. et al. Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Mol. Med. 7, 735–753 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Timosenko, E., Hadjinicolaou, A. V. & Cerundolo, V. Modulation of cancer-specific immune responses by amino acid degrading enzymes. Immunotherapy 9, 83–97 (2017).

    CAS  PubMed  Google Scholar 

  163. Pavlova, N. N. & Thompson, C. B. The emerging hallmarks of cancer metabolism. Cell Metab. 23, 27–47 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Chang, C. H. et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162, 1229–1241 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Bar-Peled, L., Schweitzer, L. D., Zoncu, R. & Sabatini, D. M. Ragulator is a GEF for the rag GTPases that signal amino acid levels to mTORC1. Cell 150, 1196–1208 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Shen, K. et al. Architecture of the human GATOR1 and GATOR1-Rag GTPases complexes. Nature 556, 64–69 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Wolfson, R. L. et al. KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1. Nature 543, 438–442 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Tsun, Z. Y. et al. The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. Mol. Cell 52, 495–505 (2013).

    CAS  PubMed  Google Scholar 

  169. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03163667 (2017).

Download references

Acknowledgements

D.M. acknowledges support from the German Research Foundation (DFG). M.N.H. acknowledges support from the European Research Council (ERC) (Mechanisms of Evasive Resistance in Cancer (MERiC)), SystemsX.CH, the Louis Jeantet Foundation and the Swiss National Science Foundation.

Reviewer information

Nature Reviews Cancer thanks the anonymous reviewers for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

D.M. and S.P. researched data for the article, substantially contributed to the discussion of content, wrote the article and edited the manuscript before submission. M.N.H. substantially contributed to the discussion of content, wrote the article and edited the manuscript before submission.

Corresponding author

Correspondence to Michael N. Hall.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mossmann, D., Park, S. & Hall, M.N. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer 18, 744–757 (2018). https://doi.org/10.1038/s41568-018-0074-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-018-0074-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer