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:

Metabolic determinants of tumour initiation

Abstract

Tumours exhibit notable metabolic alterations compared with their corresponding normal tissue counterparts. These metabolic alterations can support anabolic growth, enable survival in hostile environments and regulate gene expression programmes that promote malignant progression. Whether these metabolic changes are selected for during malignant transformation or can themselves be drivers of tumour initiation is unclear. However, intriguingly, many of the major bottlenecks for tumour initiation — control of cell fate, survival and proliferation — are all amenable to metabolic regulation. In this article, we review evidence demonstrating a critical role for metabolic pathways in processes that support the earliest stages of tumour development. We discuss how cell-intrinsic factors, such as the cell of origin or transforming oncogene, and cell-extrinsic factors, such as local nutrient availability, promote or restrain tumour initiation. Deeper insight into how metabolic pathways control tumour initiation will improve our ability to design metabolic interventions to limit tumour incidence.

Key points

  • Metabolism is linked to key processes that are required for tumour initiation: cell fate control, survival, biomass accumulation and proliferation.

  • Through their role as co-substrates for chromatin-modifying enzymes, metabolites have the potential to influence cell fate programmes that control tumour initiation.

  • Hereditary cancer syndromes highlight how mutations in metabolic enzymes can predispose to cancer through the accumulation of the oncometabolites succinate, fumarate and 2-hydroxyglutarate.

  • The intrinsic metabolic configurations of tissues or cells might facilitate or antagonize oncogenic transformation.

  • Environmental factors, including diet, inflammation, hypoxia and nutrient availability, interact to control many processes related to tumour initiation and progression.

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: Challenges in tumour initiation.
Fig. 2: Metabolites have the potential to regulate chromatin-modifying enzymes.
Fig. 3: ROS control processes related to tumour evolution.
Fig. 4: Metabolic pathways support biomass accumulation.
Fig. 5: Metabolic regulation of hypoxic responses.
Fig. 6: Environmental factors influencing tumour initiation.

Similar content being viewed by others

References

  1. Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Frede, J., Adams, D. J. & Jones, P. H. Mutation, clonal fitness and field change in epithelial carcinogenesis. J. Pathol. 234, 296–301 (2014).

    Article  PubMed  Google Scholar 

  3. Haigis, K. M., Cichowski, K. & Elledge, S. J. Tissue-specificity in cancer: the rule, not the exception. Science 363, 1150–1151 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Li, X., Egervari, G., Wang, Y., Berger, S. L. & Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 19, 563–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed Central  Google Scholar 

  6. Martínez-Reyes, I. & Chandel, N. S. Cancer metabolism: looking forward. Nat. Rev. Cancer 21, 669–680 (2021).

    Article  PubMed  Google Scholar 

  7. Ishizawa, K. et al. Tumor-initiating cells are rare in many human tumors. Cell Stem Cell 7, 279–282 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Visvader, J. E. Cells of origin in cancer. Nature 469, 314–322 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Blanpain, C. Tracing the cellular origin of cancer. Nat. Cell Biol. 15, 126–134 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Brown, S. et al. Correction of aberrant growth preserves tissue homeostasis. Nature 548, 334–337 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Dow, L. E. et al. Apc restoration promotes cellular differentiation and reestablishes crypt homeostasis in colorectal cancer. Cell 161, 1539–1552 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ying, Z., Sandoval, M. & Beronja, S. Oncogenic activation of PI3K induces progenitor cell differentiation to suppress epidermal growth. Nat. Cell Biol. 20, 1256–1266 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Warrell, R. P. et al. Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid). N. Engl. J. Med. 324, 1385–1393 (1991).

    Article  PubMed  Google Scholar 

  14. Chakrabarty, R. P. & Chandel, N. S. Mitochondria as signaling organelles control mammalian stem cell fate. Cell Stem Cell 28, 394–408 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Intlekofer, A. M. & Finley, L. W. S. Metabolic signatures of cancer cells and stem cells. Nat. Metab. 1, 177–188 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Laugesen, A. & Helin, K. Chromatin repressive complexes in stem cells, development, and cancer. Cell Stem Cell 14, 735–751 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Reid, M. A., Dai, Z. & Locasale, J. W. The impact of cellular metabolism on chromatin dynamics and epigenetics. Nat. Cell Biol. 19, 1298–1306 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Lee, J. V. et al. Akt-dependent metabolic reprogramming regulates tumor cell histone acetylation. Cell Metab. 20, 306–319 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fan, J., Krautkramer, K. A., Feldman, J. L. & Denu, J. M. Metabolic regulation of histone post-translational modifications. ACS Chem. Biol. 10, 95–108 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xiao, M. et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 26, 1326–1338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Schvartzman, J. M., Thompson, C. B. & Finley, L. W. S. Metabolic regulation of chromatin modifications and gene expression. J. Cell Biol. 217, 2247–2259 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Carey, B. W., Finley, L. W. S., Cross, J. R., Allis, C. D. & Thompson, C. B. Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 518, 413–416 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mentch, S. J. et al. Histone methylation dynamics and gene regulation occur through the sensing of one-carbon metabolism. Cell Metab. 22, 861–873 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Su, X., Wellen, K. E. & Rabinowitz, J. D. Metabolic control of methylation and acetylation. Curr. Opin. Chem. Biol. 30, 52–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Martínez-Reyes, I. & Chandel, N. S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 11, 102 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Baksh, S. C. & Finley, L. W. S. Metabolic coordination of cell fate by α-ketoglutarate-dependent dioxygenases. Trends Cell Biol. 31, 24–36 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Pirozzi, C. J. & Yan, H. The implications of IDH mutations for cancer development and therapy. Nat. Rev. Clin. Oncol. 18, 645–661 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lu, C. et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chowdhury, R. et al. The oncometabolite 2‐hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 12, 463–469 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Figueroa, M. E. et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18, 553–567 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Morris, J. P. IV et al. α-Ketoglutarate links p53 to cell fate during tumour suppression. Nature 573, 595–599 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tran, T. Q. et al. α-Ketoglutarate attenuates Wnt signaling and drives differentiation in colorectal cancer. Nat. Cancer 1, 345–358 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gupta, P. B., Pastushenko, I., Skibinski, A., Blanpain, C. & Kuperwasser, C. Phenotypic plasticity: driver of cancer initiation, progression, and therapy resistance. Cell Stem Cell 24, 65–78 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Khacho, M. et al. Mitochondrial dynamics impacts stem cell identity and fate decisions by regulating a nuclear transcriptional program. Cell Stem Cell 19, 232–247 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Cheung, E. C. & Vousden, K. H. The role of ROS in tumour development and progression. Nat. Rev. Cancer 22, 280–297 (2022).

    Article  CAS  PubMed  Google Scholar 

  39. Hamanaka, R. B. et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci. Signal. 6, ra8 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Strasser, A. & Vaux, D. L. Cell death in the origin and treatment of cancer. Mol. Cell 78, 1045–1054 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Harris, I. S. & DeNicola, G. M. The complex interplay between antioxidants and ROS in cancer. Trends Cell Biol. 30, 440–451 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Stockwell, B. R. & Jiang, X. The chemistry and biology of ferroptosis. Cell Chem. Biol. 27, 365–375 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lei, G., Zhuang, L. & Gan, B. Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 22, 381–396 (2022).

    Article  CAS  PubMed  Google Scholar 

  44. Schafer, Z. T. et al. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature 461, 109–113 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Piskounova, E. et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ubellacker, J. M. et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 585, 113–118 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Green, D. R., Galluzzi, L. & Kroemer, G. Metabolic control of cell death. Science 345, 1250256 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Singh, R., Letai, A. & Sarosiek, K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 20, 175–193 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Andersen, J. L. & Kornbluth, S. The tangled circuitry of metabolism and apoptosis. Mol. Cell 49, 399–410 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jiang, X., Overholtzer, M. & Thompson, C. B. Autophagy in cellular metabolism and cancer. J. Clin. Invest. 125, 47–54 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Carneiro, B. A. & El-Deiry, W. S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 17, 395–417 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Santana-Codina, N., Mancias, J. D. & Kimmelman, A. C. The role of autophagy in cancer. Annu. Rev. Cancer Biol. 1, 19–39 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Kim, S. M. et al. PTEN deficiency and AMPK activation promote nutrient scavenging and anabolism in prostate cancer cells. Cancer Discov. 8, 866–883 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Commisso, C. et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 497, 633–637 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kamphorst, J. J. et al. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 75, 544–553 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Garcia-Bermudez, J. et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature 567, 118–122 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zitvogel, L., Tesniere, A. & Kroemer, G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat. Rev. Immunol. 6, 715–727 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Au, E., Wong, G. & Chapman, J. R. Cancer in kidney transplant recipients. Nat. Rev. Nephrol. 14, 508–520 (2018).

    Article  PubMed  Google Scholar 

  59. Jhunjhunwala, S., Hammer, C. & Delamarre, L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 21, 298–312 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Buck, M. D., Sowell, R. T., Kaech, S. M. & Pearce, E. L. Metabolic instruction of immunity. Cell 169, 570–586 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Malladi, S. et al. Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 165, 45–60 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Agudo, J. et al. Quiescent tissue stem cells evade immune surveillance. Immunity 48, 271–285.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Davidson, S. M. et al. Environment impacts the metabolic dependencies of Ras-driven non-small cell lung cancer. Cell Metab. 23, 517–528 (2016). This study shows that the metabolic networks that support proliferation differ when cancer cells are grown in vitro and in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Muir, A. et al. Environmental cystine drives glutamine anaplerosis and sensitizes cancer cells to glutaminase inhibition. eLife 6, e27713 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Cantor, J. R. et al. Physiologic medium rewires cellular metabolism and reveals uric acid as an endogenous inhibitor of UMP synthase. Cell 169, 258–272.e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Vande Voorde, J. et al. Improving the metabolic fidelity of cancer models with a physiological cell culture medium. Sci. Adv. 5, eaau7314 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Rossiter, N. J. et al. CRISPR screens in physiologic medium reveal conditionally essential genes in human cells. Cell Metab. 33, 1248–1263.e9 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Shi, X. et al. Combinatorial GxGxE CRISPR screen identifies SLC25A39 in mitochondrial glutathione transport linking iron homeostasis to OXPHOS. Nat. Commun. 13, 2483 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhu, X. G. et al. Functional genomics in vivo reveal metabolic dependencies of pancreatic cancer cells. Cell Metab. 33, 211–221.e6 (2021).

    Article  CAS  PubMed  Google Scholar 

  70. Biancur, D. E. et al. Functional genomics identifies metabolic vulnerabilities in pancreatic cancer. Cell Metab. 33, 199–210.e8 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Venneti, S. et al. Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo. Sci. Transl Med. 7, 274ra17 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Nelson, D. L. & Cox, M. M. Lehninger Principles of Biochemistry 8th edn (W.H. Freeman, 2021).

  74. Hatzivassiliou, G. et al. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 8, 311–321 (2005).

    Article  CAS  PubMed  Google Scholar 

  75. Pavlova, N. N., Zhu, J. & Thompson, C. B. The hallmarks of cancer metabolism: still emerging. Cell Metab. 34, 355–377 (2022).

    Article  CAS  PubMed  Google Scholar 

  76. Hosios, A. M. & Vander Heiden, M. G. The redox requirements of proliferating mammalian cells. J. Biol. Chem. 293, 7490–7498 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mayers, J. R. & Vander Heiden, M. G. Nature and nurture: what determines tumor metabolic phenotypes? Cancer Res. 77, 3131–3134 (2017).

    Article  CAS  PubMed  Google Scholar 

  78. Kim, J. & DeBerardinis, R. J. Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab. 30, 434–446 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Mayers, J. R. et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 353, 1161–1165 (2016). This study induced pancreatic and lung cancer in mice via Trp53 deletion and Kras activation and found that, despite similar oncogenic lesions, only lung tumours required metabolism of branched-chain amino acids.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Rinaldi, G. et al. In vivo evidence for serine biosynthesis-defined sensitivity of lung metastasis, but not of primary breast tumors, to mTORC1 inhibition. Mol. Cell 81, 386–397.e7 (2021).

    Article  CAS  PubMed  Google Scholar 

  81. Basnet, H. et al. Flura-seq identifies organ-specific metabolic adaptations during early metastatic colonization. eLife 8, e43627 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Yuneva, M. O. et al. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab. 15, 157–170 (2012). Overexpression of either of the oncogenes MYC or MET to induce tumour formation results in differential activation of central metabolic networks depending on both initiating mutation and tissue of origin.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Jun, S. et al. The requirement for pyruvate dehydrogenase in leukemogenesis depends on cell lineage. Cell Metab. 33, 1777–1792 (2021).

    Article  CAS  PubMed  Google Scholar 

  84. Mahendralingam, M. J. et al. Mammary epithelial cells have lineage-rooted metabolic identities. Nat. Metab. 3, 665–681 (2021). This study shows that the metabolic profile of different mamary cell populations is conserved in the resulting breast cancer subtypes.

    Article  CAS  PubMed  Google Scholar 

  85. Chen, P.-H. et al. Metabolic diversity in human non-small cell lung cancer cells. Mol. Cell 76, 838–851.e5 (2019). This comprehensive study reports a detailed metabolic profiling of over 80 NSCLC cell lines.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Sullivan, M. R. et al. Increased serine synthesis provides an advantage for tumors arising in tissues where serine levels are limiting. Cell Metab. 29, 1410–1421.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. DeNicola, G. M. et al. NRF2 regulates serine biosynthesis in non-small cell lung cancer. Nat. Genet. 47, 1475–1481 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Faubert, B., Solmonson, A. & DeBerardinis, R. J. Metabolic reprogramming and cancer progression. Science 368, eaaw5473 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Sciacovelli, M., Schmidt, C., Maher, E. R. & Frezza, C. Metabolic drivers in hereditary cancer syndromes. Annu. Rev. Cancer Biol. 4, 77–97 (2020).

    Article  Google Scholar 

  90. Kaelin, W. G. & McKnight, S. L. Influence of metabolism on epigenetics and disease. Cell 153, 56–69 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Clark, G. R. et al. Germline FH mutations presenting with pheochromocytoma. J. Clin. Endocrinol. Metab. 99, E2046–E2050 (2014).

    Article  CAS  PubMed  Google Scholar 

  92. Baysal, B. E. et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 287, 848–851 (2000).

    Article  CAS  PubMed  Google Scholar 

  93. Vanharanta, S. et al. Early-onset renal cell carcinoma as a novel extraparaganglial component of SDHB-associated heritable paraganglioma. Am. J. Hum. Genet. 74, 153–159 (2004).

    Article  CAS  PubMed  Google Scholar 

  94. Janeway, K. A. et al. Defects in succinate dehydrogenase in gastrointestinal stromal tumors lacking KIT and PDGFRA mutations. Proc. Natl Acad. Sci. USA 108, 314–318 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Tomlinson, I. P. et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat. Genet. 30, 406–410 (2002).

    Article  CAS  PubMed  Google Scholar 

  96. Gimenez-Roqueplo, A.-P. et al. The R22X mutation of the SDHD gene in hereditary paraganglioma abolishes the enzymatic activity of complex II in the mitochondrial respiratory chain and activates the hypoxia pathway. Am. J. Hum. Genet. 69, 1186–1197 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Armstrong, N. et al. SDHB knockout and succinate accumulation are insufficient for tumorigenesis but dual SDHB/NF1 loss yields SDHx-like pheochromocytomas. Cell Rep. 38, 110453 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Selak, M. A. et al. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase. Cancer Cell 7, 77–85 (2005).

    Article  CAS  PubMed  Google Scholar 

  99. Isaacs, J. S. et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell 8, 143–153 (2005).

    Article  CAS  PubMed  Google Scholar 

  100. Sciacovelli, M. et al. Fumarate is an epigenetic modifier that elicits epithelial-to-mesenchymal transition. Nature 537, 544–547 (2016). This is the first report of a causative link between fumarate accumulation and the epithelial-to-mesenchymal transition, a process implicated in cancer initiation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sulkowski, P. L. et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 50, 1086–1092 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Sulkowski, P. L. et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci. Transl Med. 9, eaal2463 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Guzy, R. D., Sharma, B., Bell, E., Chandel, N. S. & Schumacker, P. T. Loss of the SdhB, but not the SdhA, subunit of complex II triggers reactive oxygen species-dependent hypoxia-inducible factor activation and tumorigenesis. Mol. Cell. Biol. 28, 718–731 (2008).

    Article  CAS  PubMed  Google Scholar 

  104. Alderson, N. L. et al. S-(2-Succinyl)cysteine: a novel chemical modification of tissue proteins by a Krebs cycle intermediate. Arch. Biochem. Biophys. 450, 1–8 (2006).

    Article  CAS  PubMed  Google Scholar 

  105. Ooi, A. et al. An antioxidant response phenotype shared between hereditary and sporadic type 2 papillary renal cell carcinoma. Cancer Cell 20, 511–523 (2011).

    Article  CAS  PubMed  Google Scholar 

  106. Adam, J. et al. Renal cyst formation in Fh1-deficient mice is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2 signaling. Cancer Cell 20, 524–537 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Sciacovelli, M. & Frezza, C. Oncometabolites: unconventional triggers of oncogenic signalling cascades. Free Radic. Biol. Med. 100, 175–181 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Platero-Luengo, A. et al. An O2-sensitive glomus cell-stem cell synapse induces carotid body growth in chronic hypoxia. Cell 156, 291–303 (2014).

    Article  CAS  PubMed  Google Scholar 

  109. Bardella, C., Pollard, P. J. & Tomlinson, I. SDH mutations in cancer. Biochim. Biophys. Acta Bioenerg. 1807, 1432–1443 (2011).

    Article  CAS  Google Scholar 

  110. Medeiros, B. C. et al. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia 31, 272–281 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Yan, H. et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Ward, P. S. et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting α-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17, 225–234 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Watanabe, T., Nobusawa, S., Kleihues, P. & Ohgaki, H. IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am. J. Pathol. 174, 1149–1153 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Lu, C. et al. Induction of sarcomas by mutant IDH2. Genes Dev. 27, 1986–1998 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Amatangelo, M. D. et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood 130, 732–741 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Fukumoto, K., Nguyen, T. B., Chiba, S. & Sakata-Yanagimoto, M. Review of the biologic and clinical significance of genetic mutations in angioimmunoblastic T-cell lymphoma. Cancer Sci. 109, 490–496 (2018).

    Article  CAS  PubMed  Google Scholar 

  117. Butzmann, A. et al. A comprehensive analysis of RHOA mutation positive and negative angioimmunoblastic T-cell lymphomas by targeted deep sequencing, expression profiling and single cell digital image analysis. Int. J. Mol. Med. 46, 1466–1476 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Locasale, J. W. et al. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat. Genet. 43, 869–874 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Lin, A.-P. et al. D2HGDH regulates alpha-ketoglutarate levels and dioxygenase function by modulating IDH2. Nat. Commun. 6, 7768 (2015).

    Article  CAS  PubMed  Google Scholar 

  121. Gorelick, A. N. et al. Respiratory complex and tissue lineage drive recurrent mutations in tumour mtDNA. Nat. Metab. 3, 558–570 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Labuschagne, C. F., Zani, F. & Vousden, K. H. Control of metabolism by p53 – cancer and beyond. Biochim. Biophys. Acta Rev. Cancer 1870, 32–42 (2018).

    Article  CAS  PubMed  Google Scholar 

  124. Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 (2003).

    Article  CAS  PubMed  Google Scholar 

  125. Morris, J. P., Wang, S. C. & Hebrok, M. KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nat. Rev. Cancer 10, 683–695 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Carrer, A. et al. Acetyl-CoA metabolism supports multistep pancreatic tumorigenesis. Cancer Discov. 9, 416–435 (2019). Kras-mutant acinar cells exhibit high levels of acetyl-CoA that fuel cancer cell histone acetylation and acinar-to-ductal metaplasia; accordingly, deletion of Acly, the gene encoding an acetyl-CoA-producing enzyme, reduces tumour incidence and prolongs survival in mouse models of pancreatic cancer.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Agathocleous, M. et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549, 476–481 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  128. Bonnay, F. et al. Oxidative metabolism drives immortalization of neural stem cells during tumorigenesis. Cell 182, 1490–1507.e19 (2020).

    Article  CAS  PubMed  Google Scholar 

  129. Schell, J. C. et al. A role for the mitochondrial pyruvate carrier as a repressor of the Warburg effect and colon cancer cell growth. Mol. Cell 56, 400–413 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Bensard, C. L. et al. Regulation of tumor initiation by the mitochondrial pyruvate carrier. Cell Metab. 31, 284–300.e7 (2020). This study demonstrates that loss of the mitochondrial pyruvate carrier in the intestine induces a metabolic programme that predisposes to cancer initiation.

    Article  CAS  PubMed  Google Scholar 

  131. Perez-Ramirez, C. A. & Christofk, H. R. Challenges in studying stem cell metabolism. Cell Stem Cell 28, 409–423 (2021).

    Article  CAS  PubMed  Google Scholar 

  132. Viale, A. et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 514, 628–632 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Janiszewska, M. et al. Imp2 controls oxidative phosphorylation and is crucial for preserving glioblastoma cancer stem cells. Genes Dev. 26, 1926–1944 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Kuntz, E. M. et al. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nat. Med. 23, 1234–1240 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Vazquez, F. et al. PGC1α expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell 23, 287–301 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Rao, S. et al. AIF-regulated oxidative phosphorylation supports lung cancer development. Cell Res. 29, 579–591 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Raffel, S. et al. BCAT1 restricts αKG levels in AML stem cells leading to IDHmut-like DNA hypermethylation. Nature 551, 384–388 (2017).

    Article  CAS  PubMed  Google Scholar 

  138. Schaaf, M. B., Garg, A. D. & Agostinis, P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 9, 115 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Schito, L. & Semenza, G. L. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer 2, 758–770 (2016).

    Article  PubMed  Google Scholar 

  140. Lee, P., Chandel, N. S. & Simon, M. C. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat. Rev. Mol. Cell Biol. 21, 268–283 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Ito, K. & Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15, 243–256 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Chakraborty, A. A. et al. Histone demethylase KDM6A directly senses oxygen to control chromatin and cell fate. Science 363, 1217–1222 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Intlekofer, A. M. et al. Hypoxia induces production of L-2-hydroxyglutarate. Cell Metab. 22, 304–311 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Oldham, W. M., Clish, C. B., Yang, Y. & Loscalzo, J. Hypoxia-mediated increases in L-2-hydroxyglutarate coordinate the metabolic response to reductive stress. Cell Metab. 22, 291–303 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Shim, E.-H. et al. L-2-Hydroxyglutarate: an epigenetic modifier and putative oncometabolite in renal cancer. Cancer Discov. 4, 1290–1298 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Wahl, D. R. & Venneti, S. 2-Hydoxyglutarate: D/Riving Pathology in gLiomaS. Brain Pathol. 25, 760–768 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Vladoiu, M. C. et al. Childhood cerebellar tumours mirror conserved fetal transcriptional programs. Nature 572, 67–73 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Michealraj, K. A. et al. Metabolic regulation of the epigenome drives lethal infantile ependymoma. Cell 181, 1329–1345.e4 (2020).

    Article  CAS  PubMed  Google Scholar 

  149. Mack, S. C. et al. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature 506, 445–450 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Panwalkar, P. et al. Targeting integrated epigenetic and metabolic pathways in lethal childhood PFA ependymomas. Sci. Transl Med. 13, eabc0497 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Pan, M. et al. Regional glutamine deficiency in tumours promotes dedifferentiation through inhibition of histone demethylation. Nat. Cell Biol. 18, 1090–1101 (2016). This study shows that poorly vascularized areas in the tumour core exhibit low glutamine levels that are associated with cancer cell histone hypermethylation, dedifferentiation and drug resistance.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Ishak Gabra, M. B. et al. Dietary glutamine supplementation suppresses epigenetically-activated oncogenic pathways to inhibit melanoma tumour growth. Nat. Commun. 11, 3326 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Baksh, S. C. et al. Extracellular serine controls epidermal stem cell fate and tumour initiation. Nat. Cell Biol. 22, 779–790 (2020). This study links local nutrient abundance to stem cell differentiation and tumour initiation, showing that serine restriction induces tumour-suppressive differentiation in epidermal stem cells and squamous cell carcinoma by forcing cells to engage in de novo serine synthesis, which produces α-ketoglutarate.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Loenarz, C. & Schofield, C. J. Physiological and biochemical aspects of hydroxylations and demethylations catalyzed by human 2-oxoglutarate oxygenases. Trends Biochem. Sci. 36, 7–18 (2011).

    Article  CAS  PubMed  Google Scholar 

  155. Cimmino, L. et al. Restoration of TET2 function blocks aberrant self-renewal and leukemia progression. Cell 170, 1079–1095.e20 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Rappez, L. et al. SpaceM reveals metabolic states of single cells. Nat. Methods 18, 799–805 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Jang, C. et al. Metabolite exchange between mammalian organs quantified in pigs. Cell Metab. 30, 594–606.e3 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Gopalakrishnan, V., Helmink, B. A., Spencer, C. N., Reuben, A. & Wargo, J. A. The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell 33, 570–580 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Janney, A., Powrie, F. & Mann, E. H. Host-microbiota maladaptation in colorectal cancer. Nature 585, 509–517 (2020).

    Article  CAS  PubMed  Google Scholar 

  160. Louis, P., Hold, G. L. & Flint, H. J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 12, 661–672 (2014).

    Article  CAS  PubMed  Google Scholar 

  161. Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).

    Article  CAS  PubMed  Google Scholar 

  162. Chang, P. V., Hao, L., Offermanns, S. & Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl Acad. Sci. USA 111, 2247–2252 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Bell, H. N. et al. Reuterin in the healthy gut microbiome suppresses colorectal cancer growth through altering redox balance. Cancer Cell 40, 185–200.e6 (2022).

    Article  CAS  PubMed  Google Scholar 

  164. Kadosh, E. et al. The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature 586, 133–138 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Furman, D. et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 25, 1822–1832 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Greten, F. R. & Grivennikov, S. I. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity 51, 27–41 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Lindemans, C. A. et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 528, 560–564 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Gaber, T., Strehl, C. & Buttgereit, F. Metabolic regulation of inflammation. Nat. Rev. Rheumatol. 13, 267–279 (2017).

    Article  PubMed  Google Scholar 

  169. Guerra, C. et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 11, 291–302 (2007).

    Article  CAS  PubMed  Google Scholar 

  170. Alonso-Curbelo, D. et al. A gene–environment-induced epigenetic program initiates tumorigenesis. Nature 590, 642–648 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Morris, J. P. T., Cano, D. A., Sekine, S., Wang, S. C. & Hebrok, M. Beta-catenin blocks Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in mice. J. Clin. Invest. 120, 508–520 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Schwitalla, S. et al. Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell 152, 25–38 (2013).

    Article  CAS  PubMed  Google Scholar 

  173. Li, F. et al. FBP1 loss disrupts liver metabolism and promotes tumorigenesis through a hepatic stellate cell senescence secretome. Nat. Cell Biol. 22, 728–739 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  174. Arnold, M. et al. Obesity and cancer: an update of the global impact. Cancer Epidemiol. 41, 8–15 (2016).

    Article  PubMed  Google Scholar 

  175. Calle, E. E., Rodriguez, C., Walker-Thurmond, K. & Thun, M. J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N. Engl. J. Med. 348, 1625–1638 (2003).

    Article  PubMed  Google Scholar 

  176. Tajan, M. & Vousden, K. H. Dietary approaches to cancer therapy. Cancer Cell 37, 767–785 (2020).

    Article  CAS  PubMed  Google Scholar 

  177. Schulz, M. D. et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature 514, 508–512 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Beyaz, S. et al. Dietary suppression of MHC class II expression in intestinal epithelial cells enhances intestinal tumorigenesis. Cell Stem Cell 28, 1922–1935.e5 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Park, E. J. et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140, 197–208 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Chung, K. M. et al. Endocrine-exocrine signaling drives obesity-associated pancreatic ductal adenocarcinoma. Cell 181, 832–847.e18 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Beyaz, S. et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 531, 53–58 (2016). This study shows that dietary fat ingestion can increase self-renewal capacity in intestinal progenitors and contribute to the development of spontaneous intestinal tumours in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Ringel, A. E. et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell 183, 1848–1866.e26 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Mana, M. D. et al. High-fat diet-activated fatty acid oxidation mediates intestinal stemness and tumorigenicity. Cell Rep. 35, 109212 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Cheng, C.-W. et al. Ketone body signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell 178, 1115–1131.e15 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Dmitrieva-Posocco, O. et al. β-Hydroxybutyrate suppresses colorectal cancer. Nature 605, 160–165 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Geidl-Flueck, B. et al. Fructose- and sucrose- but not glucose-sweetened beverages promote hepatic de novo lipogenesis: a randomized controlled trial. J. Hepatol. 75, 46–54 (2021).

    Article  CAS  PubMed  Google Scholar 

  187. Goncalves, M. D. et al. High-fructose corn syrup enhances intestinal tumor growth in mice. Science 363, 1345–1349 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Bian, Y. et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 585, 277–282 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Lim, A. R., Rathmell, W. K. & Rathmell, J. C. The tumor microenvironment as a metabolic barrier to effector T cells and immunotherapy. eLife 9, e55185 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  190. Courtney, K. D. et al. Isotope tracing of human clear cell renal cell carcinomas demonstrates suppressed glucose oxidation in vivo. Cell Metab. 28, 793–800.e2 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Sack, L. M. et al. Profound tissue specificity in proliferation control underlies cancer drivers and aneuploidy patterns. Cell 173, 499–514.e23 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Hoadley, K. A. et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173, 291–304.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Carmona-Fontaine, C. et al. Metabolic origins of spatial organization in the tumor microenvironment. Proc. Natl Acad. Sci. USA 114, 2934–2939 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Tasdogan, A. et al. Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature 577, 115–120 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  195. Lee, J. S. et al. Urea cycle dysregulation generates clinically relevant genomic and biochemical signatures. Cell 174, 1559–1570.e22 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Reinfeld, B. I. et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 593, 282–288 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Ho, P.-C. et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 162, 1217–1228 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Leone, R. D. & Powell, J. D. Metabolism of immune cells in cancer. Nat. Rev. Cancer 20, 516–531 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Brand, A. et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24, 657–671 (2016).

    Article  CAS  PubMed  Google Scholar 

  201. Angelin, A. et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 25, 1282–1293.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Possemato, R. et al. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature 476, 346–350 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Maddocks, O. D. K. et al. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 493, 542–546 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  205. Gao, X. et al. Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature 572, 397–401 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank S. Baksh (Sloan Kettering Institute, USA) for critical reading of the manuscript and members of the Finley laboratory for helpful discussion. L.W.S.F. is a Searle Scholar. The authors acknowledge the support of a Human Frontier Science Program Fellowship LT000200/2021-L (to J.S.B.), grants to L.W.S.F. from the Pershing Square Sohn Prize for Cancer Research and the NIH/NCI (R37CA252305), and the Memorial Sloan Kettering Cancer Center Support Grant P30CA008748.

Author information

Authors and Affiliations

Authors

Contributions

J.S.B. and L.W.S.F. contributed equally to all aspects of the article.

Corresponding author

Correspondence to Lydia W. S. Finley.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Endocrinology thanks Christian Frezza and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Glossary

α-ketoglutarate-dependent dioxygenases

A family of iron-containing enzymes that use molecular oxygen and α-ketoglutarate to oxidize a substrate, producing succinate and carbon dioxide as by-products.

Anaplerosis

Influx of intermediates into the tricarboxylic acid cycle.

Autophagy

The process by which cellular components are delivered to the lysosome for digestion.

Cataplerosis

Efflux of intermediates from the tricarboxylic acid cycle to other metabolic pathways.

Conditional essentiality

While essential genes are broadly considered to be required for cell viability under a wide array of conditions, genes exhibiting conditional essentiality are required for cell viability under specific circumstances, such as loss of another gene, activation of a signalling pathway or restriction of extracellular nutrients.

Epithelial–mesenchymal transition

A process in which epithelial cells change fate towards mesenchymal lineages, thereby gaining migratory capacity, invasiveness and often stem-like features.

Homeostatic stem cells

Undifferentiated progenitor cells that maintain tissue integrity by balancing self-renewal and differentiation into mature cells.

Isotope-tracing studies

Experiments in which heavy-labelled atoms are followed through metabolic networks, often by incubating cells with labelled forms of nutrients, such as glucose or glutamine, and then assessing labelling of downstream metabolic intermediates.

Jumonji-domain-containing histone demethylases

Members of the family of α-ketoglutarate-dependent dioxygenases that contain a common structural domain that facilitates protein demethylation.

Macropinocytosis

A non-selective form of endocytosis in which cells engulf extracellular fluid and other macromolecular cargo.

Myeloid cells

Bone-marrow-derived cells of a common lineage, including monocytes, macrophages, dendritic cells and granulocytes.

Receptor-mediated endocytosis

A process by which cell surface receptors and their ligands are internalized in a clathrin-dependent fashion.

Stromal cells

Cells that functionally or architecturally support tissues throughout the body.

Tumour initiation

The process in which a cell (‘cell of origin’) acquires the ability to self-renew, propagate and withstand elimination to eventually give rise to a tumour.

Tumour microenvironment

(TME). The cells and molecules that comprise the local ecosystem of a tumour, including the cancer cells, stromal and immune cells and blood vessels, as well as the interstitial fluid and any proteins, metabolites or other molecules therein.

Tumour progression

The evolution of a tumour towards increased malignancy; can refer to increases in tumour size, stage or dissemination to new sites for metastatic growth.

Tumour-associated stem cells

A subset of cancer cells within a tumour that display capacity for self-renewal and differentiation; these cells are often associated with therapy resistance and tumour aggressiveness.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brunner, J.S., Finley, L.W.S. Metabolic determinants of tumour initiation. Nat Rev Endocrinol 19, 134–150 (2023). https://doi.org/10.1038/s41574-022-00773-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-022-00773-5

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