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:

Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism

Abstract

Metabolic transformation is a hallmark of cancer and a critical target for cancer therapy. Cancer metabolism and behaviour are regulated by cell-intrinsic factors as well as metabolite availability in the tumour microenvironment (TME). This metabolic niche within the TME is shaped by four tiers of regulation: (1) intrinsic tumour cell metabolism, (2) interactions between cancer cells and non-cancerous cells, (3) tumour location and heterogeneity and (4) whole-body metabolic homeostasis. Here, we define these modes of metabolic regulation and review how distinct cell types contribute to the metabolite composition of the TME. Finally, we connect these insights to understand how each of these tiers offers unique therapeutic potential to modulate the metabolic profile and function of all cells inhabiting the TME.

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: Schematic representation of the metabolic fluctuations and niches that influence the metabolite composition of the TME.
Fig. 2: Tumour metabolism influences, and is influenced by, the metabolite composition of the TME.
Fig. 3: The metabolic cross-talk between tumour cells and immune or stromal cells within the TME.
Fig. 4: The interorgan and intratumour microenvironments define the metabolic properties of tumour cells.

Similar content being viewed by others

References

  1. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. 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 

  3. Warburg, O. The metabolism of carcinoma cells. J. Cancer Res. 9, 148–163 (1925).

    Article  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  5. Spinelli, J. B. & Haigis, M. C. The multifaceted contributions of mitochondria to cellular metabolism. Nat. Cell Biol. 20, 745–754 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. DeBerardinis, R. J. & Chandel, N. S. We need to talk about the Warburg effect. Nat. Metab. 2, 127–129 (2020).

    Article  PubMed  Google Scholar 

  7. Vyas, S., Zaganjor, E. & Haigis, M. C. Mitochondria and cancer. Cell 166, 555–566 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Elia, I., Schmieder, R., Christen, S. & Fendt, S.M. Organ-specific cancer metabolism and its potential for therapy. in Handbook of Experimental Pharmacology (Springer, 2016).

  9. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mayers, J. R. et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 353, 1161–1165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Christen, S. et al. Breast cancer-derived lung metastases show increased pyruvate carboxylase-dependent anaplerosis. Cell Rep. 17, 837–848 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Hensley, C. T. et al. Metabolic heterogeneity in human lung tumors. Cell 164, 681–694 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gui, D. Y. et al. Environment dictates dependence on mitochondrial complex I for NAD+ and aspartate production and determines cancer cell sensitivity to metformin. Cell Metab. 24, 716–727 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Campbell, S. L. & Wellen, K. E. Metabolic signaling to the nucleus in cancer. Mol. Cell 71, 398–408 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Egeblad, M., Nakasone, E. S. & Werb, Z. Tumors as organs: complex tissues that interface with the entire organism. Dev. Cell 18, 884–901 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nakazawa, M. S., Keith, B. & Simon, M. C. Oxygen availability and metabolic adaptations. Nat. Rev. Cancer 16, 663–673 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Xiao, Z., Dai, Z. & Locasale, J. W. Metabolic landscape of the tumor microenvironment at single cell resolution. Nat. Commun. 10, 3763 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Kumar, S. et al. Intra-tumoral metabolic zonation and resultant phenotypic diversification are dictated by blood vessel proximity. Cell Metab. 30, 201–211.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Harris, I. S. & Brugge, J. S. United they stand, divided they fall. Cell Metab. 30, 624–625 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Lyssiotis, C. A. & Kimmelman, A. C. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 27, 863–875 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Eil, R. et al. Ionic immune suppression within the tumour microenvironment limits T cell effector function. Nature 537, 539–543 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Vodnala, S. K. et al. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science 363, eaau0135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Witney, T. H. et al. PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci. Transl. Med. 7, 310ra169 (2015).

    Article  PubMed  Google Scholar 

  25. Hsu, P. P. & Sabatini, D. M. Cancer cell metabolism: Warburg and beyond. Cell 134, 703–707 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Lunt, S. Y. & Vander Heiden, M. G. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 27, 441–464 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Reid, M. A. et al. Serine synthesis through PHGDH coordinates nucleotide levels by maintaining central carbon metabolism. Nat. Commun. 9, 5442 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Ngo, B. et al. Limited environmental serine and glycine confer brain metastasis sensitivity to PHGDH inhibition. Cancer Discov. http://doi.org/10.1158/2159-8290.cd-19-1228 (2020).

  30. 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 

  31. Comerford, S. A. et al. Acetate dependence of tumors. Cell 159, 1591–1602 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. DeBerardinis, R. J. et al. Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl Acad. Sci. USA 104, 19345–19350 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Weinberg, F., Ramnath, N. & Nagrath, D. Reactive oxygen species in the tumor microenvironment: an overview. Cancers (Basel) 11, E1191 (2019).

    Article  Google Scholar 

  34. Sullivan, L. B. & Chandel, N. S. Mitochondrial reactive oxygen species and cancer. Cancer Metab. 2, 17 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. García-Cañaveras, J. C., Chen, L. & Rabinowitz, J. D. The tumor metabolic microenvironment: lessons from lactate. Cancer Res. 79, 3155–3162 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Spinelli, J. B. et al. Metabolic recycling of ammonia via glutamate dehydrogenase supports breast cancer biomass. Science 358, 941–946 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cluntun, A. A., Lukey, M. J., Cerione, R. A. & Locasale, J. W. Glutamine metabolism in cancer: understanding the heterogeneity. Trends Cancer 3, 169–180 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Garcia-Bermudez, J. et al. Aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours. Nat. Cell Biol. 20, 775–781 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kurmi, K. & Haigis, M. C. Nitrogen metabolism in cancer and immunity. Trends Cell Biol. 30, 408–424 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhuang, Y., Chan, D. K., Haugrud, A. B. & Miskimins, W. K. Mechanisms by which low glucose enhances the cytotoxicity of metformin to cancer cells both in vitro and in vivo. PLoS ONE 9, e108444 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Birsoy, K. et al. Metabolic determinants of cancer cell sensitivity to glucose limitation and biguanides. Nature 508, 108–112 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Jiang, J., Srivastava, S. & Zhang, J. Starve cancer cells of glutamine: break the spell or make a hungry monster? Cancers (Basel) 11, E804 (2019).

    Article  Google Scholar 

  49. 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 

  50. Jeong, S. M. et al. SIRT4 has tumor-suppressive activity and regulates the cellular metabolic response to DNA damage by inhibiting mitochondrial glutamine metabolism. Cancer Cell 23, 450–463 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cantor, J. R. The rise of physiologic media. Trends Cell Biol. 29, 854–861 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Ackermann, T. & Tardito, S. Cell culture medium formulation and its implications in cancer metabolism. Trends Cancer 5, 329–332 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Vande Voorde, J. et al. Improving the metabolic fidelity of cancer models with a physiological cell culture medium. Sci. Adv. 5, u7314 (2019).

    Article  Google Scholar 

  54. Boon, R. et al. Amino acid levels determine metabolism and CYP450 function of hepatocytes and hepatoma cell lines. Nat. Commun. 11, 1393 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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 

  56. Elia, I. et al. Proline metabolism supports metastasis formation and could be inhibited to selectively target metastasizing cancer cells. Nat. Commun. 8, 15267 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Coloff, J. L. et al. Differential glutamate metabolism in proliferating and quiescent mammary epithelial cells. Cell Metab. 23, 867–880 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. Simian, M. & Bissell, M. J. Organoids: a historical perspective of thinking in three dimensions. J. Cell Biol. 216, 31–40 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).

    Article  CAS  PubMed  Google Scholar 

  60. Elia, I. et al. Breast cancer cells rely on environmental pyruvate to shape the metastatic niche. Nature 568, 117–121 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. De Wever, O., Demetter, P., Mareel, M. & Bracke, M. Stromal myofibroblasts are drivers of invasive cancer growth. Int. J. Cancer 123, 2229–2238 (2008).

    Article  PubMed  Google Scholar 

  62. Pavlides, S. et al. The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle 8, 3984–4001 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Geeraerts, X., Bolli, E., Fendt, S. M. & Van Ginderachter, J. A. Macrophage metabolism as therapeutic target for cancer, atherosclerosis, and obesity. Front. Immunol. 8, 289 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    Article  PubMed  Google Scholar 

  65. Buck, M. D., O’Sullivan, D. & Pearce, E. L. T cell metabolism drives immunity. J. Exp. Med. 212, 1345–1360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Pollizzi, K. N. & Powell, J. D. Integrating canonical and metabolic signalling programmes in the regulation of T cell responses. Nat. Rev. Immunol. 14, 435–446 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Delgoffe, G. M. & Powell, J. D. Feeding an army: the metabolism of T cells in activation, anergy, and exhaustion. Mol. Immunol. 68, 492–496 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Singer, K. et al. Warburg phenotype in renal cell carcinoma: high expression of glucose-transporter 1 (GLUT-1) correlates with low CD8+ T-cell infiltration in the tumor. Int. J. Cancer 128, 2085–2095 (2011).

    Article  CAS  PubMed  Google Scholar 

  69. Cascone, T. et al. Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab. 27, 977–987.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chang, C. H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Peng, M. et al. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 354, 481–484 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Cham, C. M. & Gajewski, T. F. Glucose availability regulates IFN-γ production and p70S6 kinase activation in CD8+ effector T cells. J. Immunol. 174, 4670–4677 (2005).

    Article  CAS  PubMed  Google Scholar 

  73. Cham, C. M., Driessens, G., O’Keefe, J. P. & Gajewski, T. F. Glucose deprivation inhibits multiple key gene expression events and effector functions in CD8+ T cells. Eur. J. Immunol. 38, 2438–2450 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. 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 

  75. Fischer, K. et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 109, 3812–3819 (2007).

    Article  CAS  PubMed  Google Scholar 

  76. 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 

  77. Zhang, Y. et al. Enhancing CD8+ T cell fatty acid catabolism within a metabolically challenging tumor microenvironment increases the efficacy of melanoma immunotherapy. Cancer Cell 32, 377–391.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Qiu, J. et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep. 27, 2063–2074.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang, R. et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871–882 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Carr, E. L. et al. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J. Immunol. 185, 1037–1044 (2010).

    Article  CAS  PubMed  Google Scholar 

  81. Klysz, D. et al. Glutamine-dependent α-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Sci. Signal. 8, ra97 (2015).

    Article  PubMed  Google Scholar 

  82. Johnson, M. O. et al. Distinct regulation of Th17 and Th1 cell differentiation by glutaminase-dependent metabolism. Cell 175, 1780–1795.e19 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Geiger, R. et al. L-Arginine Modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167, 829–842.e13 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Roy, D. G. et al. Methionine metabolism shapes T helper cell responses through regulation of epigenetic reprogramming. Cell Metab. 31, 250–266.e9 (2020).

    Article  CAS  PubMed  Google Scholar 

  85. Mocellin, S., Bronte, V. & Nitti, D. Nitric oxide, a double edged sword in cancer biology: searching for therapeutic opportunities. Med. Res. Rev. 27, 317–352 (2007).

    Article  CAS  PubMed  Google Scholar 

  86. Sinclair, L. V. et al. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 14, 500–508 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Klein Geltink, R. I. & Pearce, E. L. The importance of methionine metabolism. eLife 8, e47221 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Brody, J. R. et al. Expression of indoleamine 2,3-dioxygenase in metastatic malignant melanoma recruits regulatory T cells to avoid immune detection and affects survival. Cell Cycle 8, 1930–1934 (2009).

    Article  CAS  PubMed  Google Scholar 

  89. Witkiewicz, A. K. et al. Genotyping and expression analysis of IDO2 in human pancreatic cancer: a novel, active target. J. Am. Coll. Surg. 208, 781–789 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Inaba, T. et al. Role of the immunosuppressive enzyme indoleamine 2,3-dioxygenase in the progression of ovarian carcinoma. Gynecol. Oncol. 115, 185–192 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Mezrich, J. D. et al. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 185, 3190–3198 (2010).

    Article  CAS  PubMed  Google Scholar 

  92. Mellor, A. L. & Munn, D. H. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat. Rev. Immunol. 4, 762–774 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Leone, R. D. et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 366, 1013–1021 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Cassetta, L. & Pollard, J. W. Targeting macrophages: therapeutic approaches in cancer. Nat. Rev. Drug Discov. 17, 887–904 (2018).

    Article  CAS  PubMed  Google Scholar 

  95. Qian, B. Z. & Pollard, J. W. Macrophage diversity enhances tumor progression and metastasis. Cell 141, 39–51 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Blagih, J. et al. Cancer-specific loss of p53 leads to a modulation of myeloid and T cell responses. Cell Rep. 30, 481–496.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Lujambio, A. et al. Non-cell-autonomous tumor suppression by p53. Cell 153, 449–460 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Miller, A. et al. Exploring metabolic configurations of single cells within complex tissue microenvironments. Cell Metab. 26, 788–800.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  99. Wenes, M. et al. Macrophage metabolism controls tumor blood vessel morphogenesis and metastasis. Cell Metab. 24, 701–715 (2016).

    Article  CAS  PubMed  Google Scholar 

  100. Penny, H. L. et al. Warburg metabolism in tumor-conditioned macrophages promotes metastasis in human pancreatic ductal adenocarcinoma. OncoImmunology 5, e1191731 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  101. Colegio, O. R. et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513, 559–563 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Chen, P. et al. Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc. Natl Acad. Sci. USA 114, 580–585 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 575–580 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Liu, P. S. et al. α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    Article  CAS  PubMed  Google Scholar 

  105. Palmieri, E. M. et al. Pharmacologic or genetic targeting of glutamine synthetase skews macrophages toward an M1-like phenotype and inhibits tumor metastasis. Cell Rep. 20, 1654–1666 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wang, X. F. et al. The role of indoleamine 2,3-dioxygenase (IDO) in immune tolerance: focus on macrophage polarization of THP-1 cells. Cell. Immunol. 289, 42–48 (2014).

    Article  CAS  PubMed  Google Scholar 

  107. Halbrook, C. J. et al. Macrophage-released pyrimidines inhibit gemcitabine therapy in pancreatic cancer. Cell Metab. 29, 1390–1399.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Bussard, K. M., Mutkus, L., Stumpf, K., Gomez-Manzano, C. & Marini, F. C. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 18, 84 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  109. Hanahan, D. & Coussens, L. M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).

    Article  CAS  PubMed  Google Scholar 

  110. Gouirand, V., Guillaumond, F. & Vasseur, S. Influence of the tumor microenvironment on cancer cells metabolic reprogramming. Front. Oncol. 8, 117 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Schwörer, S., Vardhana, S. A. & Thompson, C. B. Cancer metabolism drives a stromal regenerative response. Cell Metab. 29, 576–591 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  112. Zhao, X., Kwan, J. Y. Y., Yip, K., Liu, P. P. & Liu, F. F. Targeting metabolic dysregulation for fibrosis therapy. Nat. Rev. Drug Discov. 19, 57–75 (2020).

    Article  CAS  PubMed  Google Scholar 

  113. Leca, J. et al. Cancer-associated fibroblast-derived annexin A6+ extracellular vesicles support pancreatic cancer aggressiveness. J. Clin. Invest. 126, 4140–4156 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Yang, L. et al. Targeting stromal glutamine synthetase in tumors disrupts tumor microenvironment-regulated cancer cell growth. Cell Metab. 24, 685–700 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Sousa, C. M. et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 536, 479–483 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Auciello, F. R. et al. A stromal lysolipid–autotaxin signaling axis promotes pancreatic tumor progression. Cancer Discov. 9, 617–627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Bertero, T. et al. Tumor-stroma mechanics coordinate amino acid availability to sustain tumor growth and malignancy. Cell Metab. 29, 124–140.e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  118. Salimian Rizi, B. et al. Nitric oxide mediates metabolic coupling of omentum-derived adipose stroma to ovarian and endometrial cancer cells. Cancer Res. 75, 456–471 (2015).

    Article  CAS  PubMed  Google Scholar 

  119. Zhao, H. et al. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. eLife 5, e10250 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Olivares, O. et al. Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat. Commun. 8, 16031 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wang, W. et al. Effector T cells abrogate stroma-mediated chemoresistance in ovarian cancer. Cell 165, 1092–1105 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. 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 

  123. Mootha, V. K. et al. Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria. Cell 115, 629–640 (2003).

    Article  CAS  PubMed  Google Scholar 

  124. Nusinow, D. P. et al. Quantitative proteomics of the cancer cell line encyclopedia. Cell 180, 387–402.e16 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Hu, J. et al. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat. Biotechnol. 31, 522–529 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Gaude, E. & Frezza, C. Tissue-specific and convergent metabolic transformation of cancer correlates with metastatic potential and patient survival. Nat. Commun. 7, 13041 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Mayers, J. R. et al. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat. Med. 20, 1193–1198 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. 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 

  129. Schild, T., Low, V., Blenis, J. & Gomes, A. P. Unique metabolic adaptations dictate distal organ-specific metastatic colonization. Cancer Cell 33, 347–354 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Maher, E. A. et al. Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR Biomed. 25, 1234–1244 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Chen, J. et al. Gain of glucose-independent growth upon metastasis of breast cancer cells to the brain. Cancer Res. 75, 554–565 (2015).

    Article  CAS  PubMed  Google Scholar 

  132. Loo, J. M. et al. Extracellular metabolic energetics can promote cancer progression. Cell 160, 393–406 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Yamaguchi, N. et al. PCK1 and DHODH drive colorectal cancer liver metastatic colonization and hypoxic growth by promoting nucleotide synthesis. eLife 8, e52135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Nguyen, A. et al. PKLR promotes colorectal cancer liver colonization through induction of glutathione synthesis. J. Clin. Invest. 126, 681–694 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  135. Elia, I., Doglioni, G. & Fendt, S. M. Metabolic hallmarks of metastasis formation. Trends Cell Biol. 28, 673–684 (2018).

    Article  CAS  PubMed  Google Scholar 

  136. Pan, M. et al. Regional glutamine deficiency in tumours promotes dedifferentiation through inhibition of histone demethylation. Nat. Cell Biol. 18, 1090–1101 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Okegawa, T. et al. Intratumor heterogeneity in primary kidney cancer revealed by metabolic profiling of multiple spatially separated samples within tumors. EBioMedicine 19, 31–38 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Lien, E. C. & Vander Heiden, M. G. A framework for examining how diet impacts tumour metabolism. Nat. Rev. Cancer 19, 651–661 (2019).

    Article  CAS  PubMed  Google Scholar 

  139. Humpton, T. J., Hock, A. K., Maddocks, O. D. K. & Vousden, K. H. p53-mediated adaptation to serine starvation is retained by a common tumour-derived mutant. Cancer Metab. 6, 18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  140. Maddocks, O. D. K. et al. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 544, 372–376 (2017).

    Article  CAS  PubMed  Google Scholar 

  141. Ma, E. H. et al. Serine is an essential metabolite for effector T cell expansion. Cell Metab. 25, 345–357 (2017).

    Article  CAS  PubMed  Google Scholar 

  142. Kurniawan, H. et al. Glutathione restricts serine metabolism to preserve regulatory T cell function. Cell Metab. 31, 920–936.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. 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 

  144. Knott, S. R. V. et al. Asparagine bioavailability governs metastasis in a model of breast cancer. Nature 554, 378–381 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Lien, E. C., Westermark, A. M., Li, Z., Sapp, K. M. & Heiden, M. Vander. Caloric restriction alters lipid metabolism to contribute to tumor growth inhibition. Preprint at bioRxiv https://doi.org/10.1101/2020.03.09.984302 (2020).

  146. Hopkins, B. D. et al. Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 560, 499–503 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Kanarek, N. et al. Histidine catabolism is a major determinant of methotrexate sensitivity. Nature 559, 632–636 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. David, L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563 (2014).

    Article  CAS  PubMed  Google Scholar 

  149. Schmidt, T. S. B., Raes, J. & Bork, P. The human gut microbiome: from association to modulation. Cell 172, 1198–1215 (2018).

    Article  CAS  PubMed  Google Scholar 

  150. Shats, I. et al. Bacteria boost mammalian host NAD metabolism by engaging the deamidated biosynthesis pathway. Cell Metab. 31, 564–579.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Cogdill, A. P., Gaudreau, P. O., Arora, R., Gopalakrishnan, V. & Wargo, J. A. The impact of intratumoral and gastrointestinal microbiota on systemic cancer therapy. Trends Immunol. 39, 900–920 (2018).

    Article  CAS  PubMed  Google Scholar 

  152. Farber, S. & Diamond, L. K. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid. N. Engl. J. Med. 238, 787–793 (1948).

    Article  CAS  PubMed  Google Scholar 

  153. Locasale, J. W. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat. Rev. Cancer 13, 572–583 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Ladanyi, A. et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 37, 2285–2301 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Huang, S. C. C. et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat. Immunol. 15, 846–855 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. O’Sullivan, D., Sanin, D. E., Pearce, E. J. & Pearce, E. L. Metabolic interventions in the immune response to cancer. Nat. Rev. Immunol. 19, 324–335 (2019).

    Article  PubMed  Google Scholar 

  157. Li, X. et al. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 16, 425–441 (2019).

    Article  CAS  PubMed  Google Scholar 

  158. Renner, K. et al. Metabolic plasticity of human T cells: preserved cytokine production under glucose deprivation or mitochondrial restriction, but 2-deoxy-glucose affects effector functions. Eur. J. Immunol. 45, 2504–2516 (2015).

    Article  CAS  PubMed  Google Scholar 

  159. Renner, K. et al. Metabolic hallmarks of tumor and immune cells in the tumor microenvironment. Front. Immunol. 8, 248 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Murray, C. M. et al. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat. Chem. Biol. 1, 371–376 (2005).

    Article  CAS  PubMed  Google Scholar 

  161. Ron-Harel, N. et al. Mitochondrial biogenesis and proteome remodeling promote one-carbon metabolism for T cell activation. Cell Metab. 24, 104–117 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Palsson-McDermott, E. M. et al. Pyruvate kinase M2 is required for the expression of the immune checkpoint PD-L1 in immune cells and tumors. Front. Immunol. 8, 1300 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  163. Chesney, J. A., Telang, S., Yaddanapudi, K. & Grewal, J. S. Targeting 6-phosphofructo-2-kinase (PFKFB3) as an immunotherapeutic strategy. J. Clin. Oncol. 12, 1461–1470 (2016).

    Google Scholar 

  164. Kung, C. et al. Small molecule activation of PKM2 in cancer cells induces serine auxotrophy. Chem. Biol. 19, 1187–1198 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Battaglia, M., Stabilini, A. & Roncarolo, M. G. Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. Blood 105, 4743–4748 (2005).

    Article  CAS  PubMed  Google Scholar 

  166. Battaglia, M. et al. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J. Immunol. 177, 8338–8347 (2006).

    Article  CAS  PubMed  Google Scholar 

  167. Araki, K. et al. mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Sugiura, A. & Rathmell, J. C. Metabolic barriers to T cell function in tumors. J. Immunol. 200, 400–407 (2018).

    Article  CAS  PubMed  Google Scholar 

  169. Eikawa, S. et al. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc. Natl Acad. Sci. USA 112, 1809–1814 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Scharping, N. E., Menk, A. V., Whetstone, R. D., Zeng, X. & Delgoffe, G. M. Efficacy of PD-1 blockade is potentiated by metformin-induced reduction of tumor hypoxia. Cancer Immunol. Res. 5, 9–16 (2017).

    Article  CAS  PubMed  Google Scholar 

  171. Schurich, A., Magalhaes, I. & Mattsson, J. Metabolic regulation of CAR T cell function by the hypoxic microenvironment in solid tumors. Immunotherapy 11, 335–345 (2019).

    Article  CAS  PubMed  Google Scholar 

  172. Sadelain, M., Brentjens, R. & Rivière, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 3, 388–398 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Jaspers, J. E. & Brentjens, R. J. Development of CAR T cells designed to improve antitumor efficacy and safety. Pharmacol. Ther. 178, 83–91 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Caruana, I. et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 21, 524–529 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. Boon, J. van Der Reest and J. Drijvers for valuable feedback on the manuscript. I.E. acknowledges funding from the European Molecular Biology Organization (EMBO) and the Cancer Research Institute (CRI). M.C.H. is supported by the Ludwig Center at Harvard, the Glenn Foundation for Medical Research and US National Institutes of Health grant RO1CA213062.

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote, edited and approved the final manuscript.

Corresponding author

Correspondence to Marcia C. Haigis.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt.

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

Elia, I., Haigis, M.C. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab 3, 21–32 (2021). https://doi.org/10.1038/s42255-020-00317-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00317-z

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