1932

Abstract

Cell-intrinsic mechanisms of nutrient sensing are intimately linked to adaptive metabolic responses, and these pathways play critical roles in the complex and dynamic nutrient environment of a growing tumor. Nutrient-responsive transcription factors (e.g., HIF, SREBP, ATF4) and signaling pathways (e.g., mTORC1, AMPK) allow tumor cells to tune their metabolic output and strategies to fluctuations in nutrient availability, thus balancing tumor cell proliferation and survival with a combination of anabolic and adaptive responses. Coupling these nutrient-sensing mechanisms to the control of recycling and scavenging processes, such as autophagy and macropinocytosis, further enhances the adaptability to nutrients within tumors. Here, we discuss the key nutrient-sensing pathways active in cancer cells, how oncogenic events influence these pathways, and their likely contributions to tumor growth and survival. A better understanding of nutrient-sensing strategies and metabolic adaptations within the tumor microenvironment is critical to defining and targeting metabolic vulnerabilities in cancer.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-cancerbio-030617-050329
2018-03-04
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/cancerbio/2/1/annurev-cancerbio-030617-050329.html?itemId=/content/journals/10.1146/annurev-cancerbio-030617-050329&mimeType=html&fmt=ahah

Literature Cited

  1. Babu E, Bhutia YD, Ramachandran S, Gnanaprakasam JP, Prasad PD. et al. 2015. Deletion of the amino acid transporter Slc6a14 suppresses tumour growth in spontaneous mouse models of breast cancer. Biochem. J. 469:17–23 [Google Scholar]
  2. Bar-Peled L, Chantranupong L, Cherniack AD, Chen WW, Ottina KA. et al. 2013. A tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 340:1100–6 [Google Scholar]
  3. Ben-Sahra I, Hoxhaj G, Ricoult SJH, Asara JM, Manning BD. 2016. mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle. Science 351:728–33 [Google Scholar]
  4. Ben-Sahra I, Manning BD. 2017. mTORC1 signaling and the metabolic control of cell growth. Curr. Opin. Cell Biol. 45:72–82 [Google Scholar]
  5. Blais JD, Filipenko V, Bi M, Harding HP, Ron D. et al. 2004. Activating transcription factor 4 is translationally regulated by hypoxic stress. Mol. Cell. Biol. 24:7469–82 [Google Scholar]
  6. Brugarolas J, Lei K, Hurley RL, Manning BD, Reiling JH. et al. 2004. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev 18:2893–904 [Google Scholar]
  7. Castellano BM, Thelen AM, Moldavski O, Feltes M, van der Welle REN. et al. 2017. Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science 355:1306–11 [Google Scholar]
  8. Chandel NS, McClintock DS, Feliciano CE, Wood TM, Melendez JA. et al. 2000. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1α during hypoxia: a mechanism of O2 sensing. J. Biol. Chem. 275:25130–38 [Google Scholar]
  9. Chantranupong L, Scaria SM, Saxton RA, Gygi MP, Shen K. et al. 2016. The CASTOR proteins are arginine sensors for the mTORC1 pathway. Cell 165:153–64 [Google Scholar]
  10. Clendening JW, Penn LZ. 2012. Targeting tumor cell metabolism with statins. Oncogene 31:4967–78 [Google Scholar]
  11. Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ. et al. 2013. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 497:633–37 [Google Scholar]
  12. Cormerais Y, Giuliano S, LeFloch R, Front B, Durivault J. et al. 2016. Genetic disruption of the multifunctional CD98/LAT1 complex demonstrates the key role of essential amino acid transport in the control of mTORC1 and tumor growth. Cancer Res 76:4481–92 [Google Scholar]
  13. Curry NL, Mino-Kenudson M, Oliver TG, Yilmaz ÖH, Yilmaz VO. et al. 2013. Pten-null tumors cohabiting the same lung display differential AKT activation and sensitivity to dietary restriction. Cancer Discov 3:908–21 [Google Scholar]
  14. Dang CV, Kim J-W, Gao P, Yustein J. 2008. The interplay between MYC and HIF in cancer. Nat. Rev. Cancer 8:51–56 [Google Scholar]
  15. Davidson SM, Jonas O, Keibler MA, Hou HW, Luengo A. et al. 2017. Direct evidence for cancer-cell-autonomous extracellular protein catabolism in pancreatic tumors. Nat. Med. 23:235–41 [Google Scholar]
  16. DeBerardinis RJ, Chandel NS. 2016. Fundamentals of cancer metabolism. Sci. Adv. 2:e1600200 [Google Scholar]
  17. DeNicola GM, Chen P-H, Mullarky E, Sudderth JA, Hu Z. et al. 2015. NRF2 regulates serine biosynthesis in non–small cell lung cancer. Nat. Genet. 47:1475–81 [Google Scholar]
  18. Dibble CC, Manning BD. 2013. Signal integration by mTORC1 coordinates nutrient input with biosynthetic output. Nat. Cell Biol. 15:555–64 [Google Scholar]
  19. Dong J, Qiu H, Garcia-Barrio M, Anderson J, Hinnebusch AG. 2000. Uncharged tRNA activates GCN2 by displacing the protein kinase moiety from a bipartite tRNA-binding domain. Mol. Cell 6:269–79 [Google Scholar]
  20. Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI. et al. 2010. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol. Cell 39:171–83 [Google Scholar]
  21. Efeyan A, Comb WC, Sabatini DM. 2015. Nutrient-sensing mechanisms and pathways. Nature 517:302–10 [Google Scholar]
  22. Egan DF, Kim J, Shaw RJ, Guan K-L. 2011. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy 7:645–46 [Google Scholar]
  23. Eng CH, Wang Z, Tkach D, Toral-Barza L, Ugwonali S. et al. 2016. Macroautophagy is dispensable for growth of KRAS mutant tumors and chloroquine efficacy. PNAS 113:182–87 [Google Scholar]
  24. Faubert B, Boily G, Izreig S, Griss T, Samborska B. et al. 2013. AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo. Cell Metab 17:113–24 [Google Scholar]
  25. Faubert B, DeBerardinis RJ. 2017. Analyzing tumor metabolism in vivo. Annu. Rev. Cancer Biol. 1:99–117 [Google Scholar]
  26. Ganapathy V, Thangaraju M, Prasad PD. 2009. Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond. Pharmacol. Ther. 121:29–40 [Google Scholar]
  27. Ganley IG, Lam DH, Wang J, Ding X, Chen S, Jiang X. 2009. ULK1·ATG13·FIP200 complex mediates mTOR signaling and is essential for autophagy. J. Biol. Chem. 284:12297–305 [Google Scholar]
  28. Goel S, Duda DG, Xu L, Munn LL, Boucher Y. et al. 2011. Normalization of the vasculature for treatment of cancer and other diseases. Physiol. Rev. 91:1071–121 [Google Scholar]
  29. Griffiths B, Lewis CA, Bensaad K, Ros S, Zhang Q. et al. 2013. Sterol regulatory element binding protein-dependent regulation of lipid synthesis supports cell survival and tumor growth. Cancer Metab 1:3 [Google Scholar]
  30. Guo D, Prins RM, Dang J, Kuga D, Iwanami A. et al. 2009. EGFR signaling through an Akt-SREBP-1–dependent, rapamycin-resistant pathway sensitizes glioblastomas to antilipogenic therapy. Sci. Signal. 2:ra82 [Google Scholar]
  31. Guo D, Reinitz F, Youssef M, Hong C, Nathanson D. et al. 2011. An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLR-dependent pathway. Cancer Discov 1:442–56 [Google Scholar]
  32. Guo JY, Chen H-Y, Mathew R, Fan J, Strohecker AM. et al. 2011. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev 25:460–70 [Google Scholar]
  33. Guo JY, Teng X, Laddha SV, Ma S, Van Nostrand SC. et al. 2016. Autophagy provides metabolic substrates to maintain energy charge and nucleotide pools in Ras-driven lung cancer cells. Genes Dev 30:1704–17 [Google Scholar]
  34. Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A. et al. 2008. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 30:214–26 [Google Scholar]
  35. Hamann JC, Surcel A, Chen R, Teragawa C, Albeck JG. et al. 2017. Entosis is induced by glucose starvation. Cell Rep 20:201–10 [Google Scholar]
  36. Han J, Back SH, Hur J, Lin Y-H, Gildersleeve R. et al. 2013. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat. Cell Biol. 15:481–90 [Google Scholar]
  37. Hara K, Yonezawa K, Weng Q-P, Kozlowski MT, Belham C, Avruch J. 1998. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J. Biol. Chem. 273:14484–94 [Google Scholar]
  38. Hardie DG. 2014. AMP-activated protein kinase: maintaining energy homeostasis at the cellular and whole-body levels. Annu. Rev. Nutr. 34:31–55 [Google Scholar]
  39. Hardie DG, Alessi DR. 2013. LKB1 and AMPK and the cancer-metabolism link—ten years after. BMC Biol 11:36 [Google Scholar]
  40. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD. et al. 2003. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 11:619–33 [Google Scholar]
  41. Hinnebusch AG, Ivanov IP, Sonenberg N. 2016. Translational control by 5′-untranslated regions of eukaryotic mRNAs. Science 352:1413–16 [Google Scholar]
  42. Horton JD, Goldstein JL, Brown MS. 2002. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Investig. 109:1125–31 [Google Scholar]
  43. Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR. et al. 2012. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 485:55–61 [Google Scholar]
  44. Hu C-J, Wang L-Y, Chodosh LA, Keith B, Simon MC. 2003. Differential roles of hypoxia-inducible factor 1α (HIF-1α) and HIF-2α in hypoxic gene regulation. Mol. Cell. Biol. 23:9361–74 [Google Scholar]
  45. Hu J, Locasale JW, Bielas JH, O'Sullivan J, Sheahan K. et al. 2013. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat. Biotechnol. 31:522–29 [Google Scholar]
  46. Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC. et al. 2002. Regulation of hypoxia-inducible factor 1α expression and function by the mammalian target of rapamycin. Mol. Cell. Biol. 22:7004–14 [Google Scholar]
  47. Ilagan E, Manning BD. 2016. Emerging role of mTOR in the response to cancer therapeutics. Trends Cancer 2:241–51 [Google Scholar]
  48. Inoki K, Zhu T, Guan K-L. 2003. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115:577–90 [Google Scholar]
  49. Iurlaro R, Püschel F, León-Annicchiarico CL, O'Connor H, Martin SJ. et al. 2017. Glucose deprivation induces ATF4-mediated apoptosis through TRAIL death receptors. Mol. Cell. Biol. 37:e00479–16 [Google Scholar]
  50. Ivan M, Kondo K, Yang H, Kim W, Valiando J. et al. 2001. HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292:464–68 [Google Scholar]
  51. Iyer NV, Kotch LE, Agani F, Leung SW, Laughner E. et al. 1998. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor1α. Genes Dev 12:149–62 [Google Scholar]
  52. Jaakkola P, Mole DR, Tian Y-M, Wilson MI, Gielbert J. et al. 2001. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292:468–72 [Google Scholar]
  53. Jefferies HB, Reinhard C, Kozma SC, Thomas G. 1994. Rapamycin selectively represses translation of the “polypyrimidine tract” mRNA family. PNAS 91:4441–45 [Google Scholar]
  54. Jeon S-M, Chandel NS, Hay N. 2012. AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature 485:661–65 [Google Scholar]
  55. Jung CH, Jun CB, Ro S-H, Kim Y-M, Otto NM. et al. 2009. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol. Biol. Cell 20:1992–2003 [Google Scholar]
  56. Kalaany NY, Sabatini DM. 2009. Tumours with PI3K activation are resistant to dietary restriction. Nature 458:725–31 [Google Scholar]
  57. Kamphorst JJ, Cross JR, Fan J, de Stanchina E, Mathew R. et al. 2013. Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. PNAS 110:8882–87 [Google Scholar]
  58. Kamphorst JJ, Nofal M, Commisso C, Hackett SR, Lu W. et al. 2015. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res 75:544–53 [Google Scholar]
  59. Karsli-Uzunbas G, Guo JY, Price S, Teng X, Laddha SV. et al. 2014. Autophagy is required for glucose homeostasis and lung tumor maintenance. Cancer Discov 4:914–27 [Google Scholar]
  60. Karunakaran S, Ramachandran S, Coothankandaswamy V, Elangovan S, Babu E. et al. 2011. SLC6A14 (ATB0,+) protein, a highly concentrative and broad specific amino acid transporter, is a novel and effective drug target for treatment of estrogen receptor-positive breast cancer. J. Biol. Chem. 286:31830–38 [Google Scholar]
  61. Kaur J, Debnath J. 2015. Autophagy at the crossroads of catabolism and anabolism. Nat. Rev. Mol. Cell. Biol. 16:461–72 [Google Scholar]
  62. Keith B, Johnson RS, Simon MC. 2012. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 12:9–22 [Google Scholar]
  63. Kim J, Kundu M, Viollet B, Guan K-L. 2011. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell. Biol. 13:132–41 [Google Scholar]
  64. Kim SM, Roy SG, Chen B, Nguyen TM, McMonigle RJ. et al. 2016. Targeting cancer metabolism by simultaneously disrupting parallel nutrient access pathways. J. Clin. Investig. 126:4088–102 [Google Scholar]
  65. Kim WY, Kaelin WG. 2004. Role of VHL gene mutation in human cancer. J. Clin. Oncol. 22:4991–5004 [Google Scholar]
  66. Kondo K, Kim WY, Lechpammer M, Kaelin WG Jr. 2003. Inhibition of HIF2α is sufficient to suppress pVHL-defective tumor growth. PLOS Biol 1:e83 [Google Scholar]
  67. Kondo K, Klco J, Nakamura E, Lechpammer M, Kaelin WG Jr. 2002. Inhibition of HIF is necessary for tumor suppression by the von Hippel-Lindau protein. Cancer Cell 1:237–46 [Google Scholar]
  68. Koumenis C, Naczki C, Koritzinsky M, Rastani S, Diehl A. et al. 2002. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2α. Mol. Cell. Biol. 22:7405–16 [Google Scholar]
  69. Krajcovic M, Krishna S, Akkari L, Joyce JA, Overholtzer M. 2013. mTOR regulates phagosome and entotic vacuole fission. Mol. Biol. Cell 24:3736–45 [Google Scholar]
  70. Krall AS, Xu S, Graeber TG, Braas D, Christofk HR. 2016. Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor. Nat. Commun. 7:11457 [Google Scholar]
  71. Krishna S, Palm W, Lee Y, Yang W, Bandyopadhyay U. et al. 2016. PIKfyve regulates vacuole maturation and nutrient recovery following engulfment. Dev. Cell 38:536–47 [Google Scholar]
  72. Kritchevsky D. 2001. Caloric restriction and cancer. J. Nutr. Sci. Vitaminol. 47:13–19 [Google Scholar]
  73. Laderoute KR, Amin K, Calaoagan JM, Knapp M, Le T. et al. 2006. 5′-AMP-activated protein kinase (AMPK) is induced by low-oxygen and glucose deprivation conditions found in solid-tumor microenvironments. Mol. Cell Biol. 26:5336–47 [Google Scholar]
  74. Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL. 2001. 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 [Google Scholar]
  75. Leprivier G, Remke M, Rotblat B, Dubuc A, Mateo AR. et al. 2013. The eEF2 kinase confers resistance to nutrient deprivation by blocking translation elongation. Cell 153:1064–79 [Google Scholar]
  76. Lewis CA, Brault C, Peck B, Bensaad K, Griffiths B. et al. 2015. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene 34:5128–40 [Google Scholar]
  77. Lin L, Yee SW, Kim RB, Giacomini KM. 2015. SLC transporters as therapeutic targets: emerging opportunities. Nat. Rev. Drug Discov. 14:543–60 [Google Scholar]
  78. Liu L, Wise DR, Diehl JA, Simon MC. 2008. Hypoxic reactive oxygen species regulate the integrated stress response and cell survival. J. Biol. Chem. 283:31153–62 [Google Scholar]
  79. Liu M, Xia Y, Ding J, Ye B, Zhao E. et al. 2016. Transcriptional profiling reveals a common metabolic program in high-risk human neuroblastoma and mouse neuroblastoma sphere-forming cells. Cell Rep 17:609–23 [Google Scholar]
  80. Lock R, Roy S, Kenific CM, Su JS, Salas E. et al. 2011. Autophagy facilitates glycolysis during Ras-mediated oncogenic transformation. Mol. Biol. Cell 22:165–78 [Google Scholar]
  81. Ma XM, Blenis J. 2009. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell Biol. 10:307–18 [Google Scholar]
  82. Maddocks ODK, Athineos D, Cheung EC, Lee P, Zhang T. et al. 2017. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 544:372–76 [Google Scholar]
  83. Maddocks ODK, Berkers CR, Mason SM, Zheng L, Blyth K. et al. 2013. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 493:542–46 [Google Scholar]
  84. Magnuson B, Ekim B, Fingar DC. 2012. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem. J. 441:1–21 [Google Scholar]
  85. Majumder PK, Febbo PG, Bikoff R, Berger R, Xue Q. et al. 2004. mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways. Nat. Med. 10:594–601 [Google Scholar]
  86. Manning BD, Toker A. 2017. AKT/PKB signaling: navigating the network. Cell 169:381–405 [Google Scholar]
  87. Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. 2001. Independent function of two destruction domains in hypoxia‐inducible factor‐α chains activated by prolyl hydroxylation. EMBO J 20:5197–206 [Google Scholar]
  88. Maxwell PH, Wiesener MS, Chang G-W, Clifford SC, Vaux EC. et al. 1999. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399:271–75 [Google Scholar]
  89. Mayers JR, Torrence ME, Danai LV, Papagiannakopoulos T, Davidson SM. et al. 2016. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 353:1161–65 [Google Scholar]
  90. Medes G, Thomas A, Weinhouse S. 1953. Metabolism of neoplastic tissue. IV. A study of lipid synthesis in neoplastic tissue slices. Cancer Res 13:27–29 [Google Scholar]
  91. Menendez JA, Lupu R. 2007. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer 7:763–77 [Google Scholar]
  92. Menon S, Dibble CC, Talbott G, Hoxhaj G, Valvezan AJ. et al. 2014. Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome. Cell 156:771–85 [Google Scholar]
  93. Muranen T, Iwanicki MP, Curry NL, Hwang J, DuBois CD. et al. 2017. Starved epithelial cells uptake extracellular matrix for survival. Nat. Commun. 8:13989 [Google Scholar]
  94. Nakazawa MS, Keith B, Simon MC. 2016. Oxygen availability and metabolic adaptations. Nat. Rev. Cancer 16:663–73 [Google Scholar]
  95. Okosun J, Wolfson RL, Wang J, Araf S, Wilkins L. et al. 2016. Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma. Nat. Genet. 48:183–88 [Google Scholar]
  96. Otto C, Kaemmerer U, Illert B, Muehling B, Pfetzer N. et al. 2008. Growth of human gastric cancer cells in nude mice is delayed by a ketogenic diet supplemented with omega-3 fatty acids and medium-chain triglycerides. BMC Cancer 8:122 [Google Scholar]
  97. Overholtzer M, Mailleux AA, Mouneimne G, Normand G, Schnitt SJ. et al. 2007. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. Cell 131:966–79 [Google Scholar]
  98. Palm W, Park Y, Wright K, Pavlova NN, Tuveson DA, Thompson CB. 2015. The utilization of extracellular proteins as nutrients is suppressed by mTORC1. Cell 162:259–70 [Google Scholar]
  99. Park Y, Reyna-Neyra A, Philippe L, Thoreen CC. 2017. mTORC1 balances cellular amino acid supply with demand for protein synthesis through post-transcriptional control of ATF4. Cell Rep 19:1083–90 [Google Scholar]
  100. Pascual G, Avgustinova A, Mejetta S, Martín M, Castellanos A. et al. 2017. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 541:41–45 [Google Scholar]
  101. Peck B, Schug ZT, Zhang Q, Dankworth B, Jones DT. et al. 2016. Inhibition of fatty acid desaturation is detrimental to cancer cell survival in metabolically compromised environments. Cancer Metab 4:6 [Google Scholar]
  102. Porstmann T, Santos CR, Griffiths B, Cully M, Wu M. et al. 2008. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab 8:224–36 [Google Scholar]
  103. Potente M, Gerhardt H, Carmeliet P. 2011. Basic and therapeutic aspects of angiogenesis. Cell 146:873–87 [Google Scholar]
  104. Qing G, Li B, Vu A, Skuli N, Walton ZE. et al. 2012. ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation. Cancer Cell 22:631–44 [Google Scholar]
  105. Qiu B, Simon MC. 2015. Oncogenes strike a balance between cellular growth and homeostasis. Semin. Cell Dev. Biol. 43:3–10 [Google Scholar]
  106. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H. et al. 2003. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 112:1809–20 [Google Scholar]
  107. Rao S, Tortola L, Perlot T, Wirnsberger G, Novatchkova M. et al. 2014. A dual role for autophagy in a murine model of lung cancer. Nat. Commun. 5:3056 [Google Scholar]
  108. Raval RR, Lau KW, Tran MGB, Sowter HM, Mandriota SJ. et al. 2005. Contrasting properties of hypoxia-inducible factor 1 (HIF-1) and HIF-2 in von Hippel-Lindau-associated renal cell carcinoma. Mol. Cell. Biol. 25:5675–86 [Google Scholar]
  109. Ricoult SJH, Yecies JL, Ben-Sahra I, Manning BD. 2016. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP. Oncogene 35:1250–60 [Google Scholar]
  110. Rosenfeldt MT, O'Prey J, Morton JP, Nixon C, MacKay G. et al. 2013. p53 status determines the role of autophagy in pancreatic tumour development. Nature 504:296–300 [Google Scholar]
  111. Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM. 2010. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141:290–303 [Google Scholar]
  112. Santana-Codina N, Mancias JD, Kimmelman AC. 2017. The role of autophagy in cancer. Annu. Rev. Cancer Biol. 1:19–39 [Google Scholar]
  113. Santanam U, Banach-Petrosky W, Abate-Shen C, Shen MM, White E, DiPaola RS. 2016. Atg7 cooperates with Pten loss to drive prostate cancer tumor growth. Genes Dev 30:399–407 [Google Scholar]
  114. Saxton RA, Sabatini DM. 2017. mTOR signaling in growth, metabolism, and disease. Cell 169:361–71 [Google Scholar]
  115. Scheuner D, Song B, McEwen E, Liu C, Laybutt R. et al. 2001. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol. Cell 7:1165–76 [Google Scholar]
  116. Semenza GL. 2003. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3:721–32 [Google Scholar]
  117. Semenza GL, Jiang B-H, Leung SW, Passantino R, Concordet J-P. et al. 1996. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J. Biol. Chem. 271:32529–37 [Google Scholar]
  118. Semenza GL, Roth PH, Fang HM, Wang GL. 1994. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J. Biol. Chem. 269:23757–63 [Google Scholar]
  119. Shackelford DB, Shaw RJ. 2009. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat. Rev. Cancer 9:563–75 [Google Scholar]
  120. Sousa CM, Biancur DE, Wang X, Halbrook CJ, Sherman MH. et al. 2016. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 536:479–83 [Google Scholar]
  121. Strohecker AM, White E. 2014. Autophagy promotes BrafV600E-driven lung tumorigenesis by preserving mitochondrial metabolism. Autophagy 10:384–85 [Google Scholar]
  122. Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C. et al. 2011. Autophagy-deficient mice develop multiple liver tumors. Genes Dev 25:795–800 [Google Scholar]
  123. Thomas GV, Tran C, Mellinghoff IK, Welsbie DS, Chan E. et al. 2006. Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nat. Med. 12:122–27 [Google Scholar]
  124. Thomlinson RH, Gray LH. 1955. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br. J. Cancer 9:539–49 [Google Scholar]
  125. Thoreen CC, Chantranupong L, Keys HR, Wang T, Gray NS, Sabatini DM. 2012. A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 485:109–13 [Google Scholar]
  126. van Geldermalsen M, Wang Q, Nagarajah R, Marshall AD, Thoeng A. et al. 2016. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 35:3201–8 [Google Scholar]
  127. Wang GL, Jiang BH, Rue EA, Semenza GL. 1995. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. PNAS 92:5510–14 [Google Scholar]
  128. Wang Q, Hardie R-A, Hoy AJ, van Geldermalsen M, Gao D. et al. 2015. Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development. J. Pathol. 236:278–89 [Google Scholar]
  129. Wellberg EA, Johnson S, Finlay-Schultz J, Lewis AS, Terrell KL. et al. 2016. The glucose transporter GLUT1 is required for ErbB2-induced mammary tumorigenesis. Breast Cancer Res 18:131 [Google Scholar]
  130. Williams KJ, Argus JP, Zhu Y, Wilks MQ, Marbois BN. et al. 2013. An essential requirement for the SCAP/SREBP signaling axis to protect cancer cells from lipotoxicity. Cancer Res 73:2850–62 [Google Scholar]
  131. Wolfson RL, Chantranupong L, Saxton RA, Shen K, Scaria SM. et al. 2016. Sestrin2 is a leucine sensor for the mTORC1 pathway. Science 351:43–48 [Google Scholar]
  132. Xia S, Lin R, Jin L, Zhao L, Kang H-B. et al. 2017. Prevention of dietary-fat-fueled ketogenesis attenuates BRAF V600E tumor growth. Cell Metab 25:358–73 [Google Scholar]
  133. Yang A, Rajeshkumar NV, Wang X, Yabuuchi S, Alexander BM. et al. 2014. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov 4:905–13 [Google Scholar]
  134. Yang S, Wang X, Contino G, Liesa M, Sahin E. et al. 2011. Pancreatic cancers require autophagy for tumor growth. Genes Dev 25:717–29 [Google Scholar]
  135. Ye J, Kumanova M, Hart LS, Sloane K, Zhang H. et al. 2010. The GCN2‐ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation. EMBO J 29:2082–96 [Google Scholar]
  136. Ye J, Mancuso A, Tong X, Ward PS, Fan J. et al. 2012. Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation. PNAS 109:6904–9 [Google Scholar]
  137. Young RM, Ackerman D, Quinn ZL, Mancuso A, Gruber M. et al. 2013. Dysregulated mTORC1 renders cells critically dependent on desaturated lipids for survival under tumor-like stress. Genes Dev 27:1115–31 [Google Scholar]
  138. Yu F, White SB, Zhao Q, Lee FS. 2001. HIF-1α binding to VHL is regulated by stimulus-sensitive proline hydroxylation. PNAS 98:9630–35 [Google Scholar]
  139. Yue Z, Jin S, Yang C, Levine AJ, Heintz N. 2003. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. PNAS 100:15077–82 [Google Scholar]
  140. Yuneva MO, Fan TWM, Allen TD, Higashi RM, Ferraris DV. et al. 2012. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab 15:157–70 [Google Scholar]
  141. Zhou W, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, Seyfried TN. 2007. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr. Metab. 4:5 [Google Scholar]
  142. Zimmer M, Doucette D, Siddiqui N, Iliopoulos O. 2004. Inhibition of hypoxia-inducible factor is sufficient for growth suppression of VHL−/− tumors. Mol. Cancer Res. 2:89–95 [Google Scholar]
/content/journals/10.1146/annurev-cancerbio-030617-050329
Loading
/content/journals/10.1146/annurev-cancerbio-030617-050329
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error