Review Article

The ever-expanding role of HIF in tumour and stromal biology

  • Nature Cell Biology volume 18, pages 356365 (2016)
  • doi:10.1038/ncb3330
  • Download Citation
Published:

Abstract

Low oxygen tension (hypoxia) is a hallmark of cancer that influences cancer cell function, but is also an important component of the tumour microenvironment as it alters the extracellular matrix, modulates the tumour immune response and increases angiogenesis. Here we discuss the regulation and role of hypoxia and its key transcriptional mediators, the hypoxia-inducible factor (HIF) family of transcription factors, in the tumour microenvironment and stromal compartments.

  • Subscribe to Nature Cell Biology for full access:

    $59

    Subscribe

Additional access options:

Already a subscriber?  Log in  now or  Register  for online access.

References

  1. 1.

    & Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metast. Rev. 26, 225–239 (2007).

  2. 2.

    , , & Why are tumour blood vessels abnormal and why is it important to know? Br. J. Cancer 100, 865–869 (2009).

  3. 3.

    & Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).

  4. 4.

    & Hypoxia, inflammation, and the tumor microenvironment in metastatic disease. Cancer Metast. Rev. 29, 285–293 (2010).

  5. 5.

    Oxygen sensing, hypoxia-inducible factors, and disease pathophysiology. Annu. Rev. Pathol. 9, 47–71 (2014).

  6. 6.

    et al. Widespread hypoxia-inducible expression of HIF-2α in distinct cell populations of different organs. FASEB J. 17, 271–273 (2003).

  7. 7.

    , , & Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl Acad. Sci. USA 92, 5510–5514 (1995).

  8. 8.

    & The multifaceted Von Hippel-Lindau tumour suppressor protein. FEBS Lett. 588, 2704–2711 (2014).

  9. 9.

    , , & Regulation of hypoxia-inducible factor 1alpha is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc. Natl Acad. Sci. USA 95, 7987–7992 (1998).

  10. 10.

    , & VHL, the story of a tumour suppressor gene. Nat. Rev. Cancer 15, 55–64 (2015).

  11. 11.

    & Signalling cross talk of the HIF system: involvement of the FIH protein. Curr. Pharm. Design 15, 3904–3907 (2009).

  12. 12.

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

  13. 13.

    et al. Modulation of hypoxia-inducible factor 1α expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 60, 1541–1545 (2000).

  14. 14.

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

  15. 15.

    , , , & Jr TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell 4, 147–158 (2003).

  16. 16.

    , , , & BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor-1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood 100, 3767–3775 (2002).

  17. 17.

    et al. Loss of the tumor suppressor LKB1 promotes metabolic reprogramming of cancer cells via HIF-1α. Proc. Natl Acad. Sci. USA 111, 2554–2559 (2014).

  18. 18.

    et al. mTOR and HIF-1α-mediated tumor metabolism in an LKB1 mouse model of Peutz-Jeghers syndrome. Proc. Natl Acad. Sci. USA 106, 11137–11142 (2009).

  19. 19.

    et al. Translational activation of HIF1α by YB-1 promotes sarcoma metastasis. Cancer Cell 27, 682–697 (2015).

  20. 20.

    , , , & A novel HIF-1α-integrin-linked kinase regulatory loop that facilitates hypoxia-induced HIF-1α expression and epithelial-mesenchymal transition in cancer cells. Oncotarget 6, 8271–8285 (2015).

  21. 21.

    et al. TCR engagement increases hypoxia-inducible factor-1 alpha protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells. J. Immunol. 174, 7592–7599 (2005).

  22. 22.

    & TCR-signaling events in cellular metabolism and specialization. Front. Immunol. 6, 292 (2015).

  23. 23.

    et al. Control of TH17/Treg balance by hypoxia-inducible factor 1. Cell 146, 772–784 (2011).

  24. 24.

    et al. The cooperative induction of hypoxia-inducible factor-1 alpha and STAT3 during hypoxia induced an impairment of tumor susceptibility to CTL-mediated cell lysis. J. Immunol. 182, 3510–3521 (2009).

  25. 25.

    et al. NF-κB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1α. Nature 453, 807–811 (2008).

  26. 26.

    , , & Hypoxic gene activation by lipopolysaccharide in macrophages: implication of hypoxia-inducible factor 1α. Blood 103, 1124–1130 (2004).

  27. 27.

    et al. Toll-like receptor activation and hypoxia use distinct signaling pathways to stabilize hypoxia-inducible factor 1α (HIF1A) and result in differential HIF1A-dependent gene expression. J. Leukocyte Biol. 90, 551–562 (2011).

  28. 28.

    et al. High resolution imaging of intracellular oxygen concentration by phosphorescence lifetime. Sci. Rep. 5, 10657 (2015).

  29. 29.

    & Oligomycin inhibits HIF-1α expression in hypoxic tumor cells. Am. J. Physiol. Cell Physiol. 288, C1023–C1029 (2005).

  30. 30.

    et al. PGC-1α is coupled to HIF-1α-dependent gene expression by increasing mitochondrial oxygen consumption in skeletal muscle cells. Proc. Natl Acad. Sci. USA 106, 2188–2193 (2009).

  31. 31.

    et al. The tumor suppressor folliculin regulates AMPK-dependent metabolic transformation. J. Clin. Invest. 124, 2640–2650 (2014).

  32. 32.

    et al. Suppression of PGC-1α is critical for reprogramming oxidative metabolism in renal cell carcinoma. Cell Rep. 12, 116–127 (2015).

  33. 33.

    , & Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat. Rev. Drug Discov. 8, 579–591 (2009).

  34. 34.

    et al. Role of reactive oxygen species in the regulation of HIF-1 by prolyl hydroxylase 2 under mild hypoxia. Free Radical Res. 46, 705–717 (2012).

  35. 35.

    & Mitochondrial oxygen sensing: regulation of hypoxia-inducible factor by mitochondrial generated reactive oxygen species. Essays Biochem. 43, 17–27 (2007).

  36. 36.

    et al. Mitochondrial dysfunction resulting from loss of cytochrome c impairs cellular oxygen sensing and hypoxic HIF-α activation. Cell Metab. 1, 393–399 (2005).

  37. 37.

    et al. Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc. Natl Acad. Sci. USA 95, 11715–11720 (1998).

  38. 38.

    et al. The FIH hydroxylase is a cellular peroxide sensor that modulates HIF transcriptional activity. EMBO Rep. 13, 251–257 (2012).

  39. 39.

    Oxygen versus reactive oxygen in the regulation of HIF-1α: the balance tips. Biochem. Res. Int. 2012, 436981 (2012).

  40. 40.

    et al. Accumulation of Krebs cycle intermediates and over-expression of HIF1α in tumours which result from germline FH and SDH mutations. Hum. Mol. Genet. 14, 2231–2239 (2005).

  41. 41.

    et al. Reversible inactivation of HIF-1 prolyl hydroxylases allows cell metabolism to control basal HIF-1. J. Biol. Chem. 280, 41928–41939 (2005).

  42. 42.

    & Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J. Natl Cancer Inst. 102, 932–941 (2010).

  43. 43.

    , , & R-2-hydroxyglutarate as the key effector of IDH mutations promoting oncogenesis. Cancer Cell 23, 274–276 (2013).

  44. 44.

    , , & Emerging novel functions of the oxygen-sensing prolyl hydroxylase domain enzymes. Trends Biochem. Sci. 38, 3–11 (2013).

  45. 45.

    et al. Metabolic reprogramming of cancer-associated fibroblasts by IDH3α downregulation. Cell Rep. 10, 1335–1348 (2015).

  46. 46.

    et al. Aberrant IDH3α expression promotes malignant tumor growth by inducing HIF-1-mediated metabolic reprogramming and angiogenesis. Oncogene 34, 4758–4766 (2015).

  47. 47.

    et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 513, 251–255 (2014).

  48. 48.

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

  49. 49.

    , , , & Pyruvate kinase M2 regulates gene transcription by acting as a protein kinase. Mol. Cell 45, 598–609 (2012).

  50. 50.

    & Metabolic symbiosis in cancer: refocusing the Warburg lens. Mol. Carcinogen. 52, 329–337 (2013).

  51. 51.

    , & Extracellular acidity, a “reappreciated” trait of tumor environment driving malignancy: perspectives in diagnosis and therapy. Cancer Metast. Rev. 33, 823–832 (2014).

  52. 52.

    , & The intercellular metabolic interplay between tumor and immune cells. Front. Immunol. 5, 358 (2014).

  53. 53.

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

  54. 54.

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

  55. 55.

    Proceedings: tumor angiogenesis factor. Cancer Res. 34, 2109–2113 (1974).

  56. 56.

    , , & Stromal endothelial cells directly influence cancer progression. Sci. Transl. Med. 3, 66ra5 (2011).

  57. 57.

    et al. From vessel sprouting to normalization: role of the prolyl hydroxylase domain protein/hypoxia-inducible factor oxygen-sensing machinery. Arterioscl. Throm. Vas. Biol. 30, 2331–2336 (2010).

  58. 58.

    & Endothelial cell metabolism and tumour angiogenesis: glucose and glutamine as essential fuels and lactate as the driving force. J. Intern. Med. 273, 156–165 (2013).

  59. 59.

    et al. Fatty acid carbon is essential for dNTP synthesis in endothelial cells. Nature 520, 192–197 (2015).

  60. 60.

    et al. HIF-1-mediated suppression of acyl-CoA dehydrogenases and fatty acid oxidation is critical for cancer progression. Cell Rep. 8, 1930–1942 (2014).

  61. 61.

    et al. The hypoxia-inducible microRNA cluster miR-199a214 targets myocardial PPARδ and impairs mitochondrial fatty acid oxidation. Cell. Metab. 18, 341–354 (2013).

  62. 62.

    et al. Dietary obesity-associated HIF1α activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2-NAD+ system. Gene. Dev. 26, 259–270 (2012).

  63. 63.

    et al. Hypoxia-inducible factor 2 regulates hepatic lipid metabolism. Mol. Cell. Biol. 29, 4527–4538 (2009).

  64. 64.

    et al. Loss of HIF-1α in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell 6, 485–495 (2004).

  65. 65.

    et al. Endothelial deletion of hypoxia-inducible factor-2α (HIF-2α) alters vascular function and tumor angiogenesis. Blood 114, 469–477 (2009).

  66. 66.

    et al. Endothelial HIF-2α regulates murine pathological angiogenesis and revascularization processes. J. Clin. Invest. 122, 1427–1443 (2012).

  67. 67.

    et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 136, 839–851 (2009).

  68. 68.

    et al. Endothelial cell HIF-1α and HIF-2α differentially regulate metastatic success. Cancer Cell 21, 52–65 (2012).

  69. 69.

    & Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 10, 417–427 (2011).

  70. 70.

    et al. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am. J. Pathol. 160, 985–1000 (2002).

  71. 71.

    et al. A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature 456, 809–813 (2008).

  72. 72.

    et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell 21, 66–81 (2012).

  73. 73.

    et al. Targeting vascular pericytes in hypoxic tumors increases lung metastasis via angiopoietin-2. Cell Rep. 10, 1066–1081 (2015).

  74. 74.

    & Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 17, 471–494 (2014).

  75. 75.

    et al. Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J. Clin. Invest. 122, 1832–1848 (2012).

  76. 76.

    et al. Blocking lipid synthesis overcomes tumor regrowth and metastasis after antiangiogenic therapy withdrawal. Cell Metab. 20, 280–294 (2014).

  77. 77.

    et al. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 9, 349–365 (2014).

  78. 78.

    et al. Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Res. 64, 3731–3736 (2004).

  79. 79.

    et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 6, 553–563 (2004).

  80. 80.

    & Fibroblasts in cancer. Nat. Rev. Cancer 6, 392–401 (2006).

  81. 81.

    et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Cancer Res. 73, 5657–5668 (2013).

  82. 82.

    et al. The myofibroblast: one function, multiple origins. Am. J. Pathol. 170, 1807–1816 (2007).

  83. 83.

    et al. Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res. 64, 8492–8495 (2004).

  84. 84.

    , & Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat. Rev. Cancer 14, 430–439 (2014).

  85. 85.

    et al. Loss of fibroblast HIF-1α accelerates tumorigenesis. Cancer Res. 72, 3187–3195 (2012).

  86. 86.

    et al. Hypoxia and loss of PHD2 inactivate stromal fibroblasts to decrease tumour stiffness and metastasis. EMBO Rep. 16, 1394–1408 (2015).

  87. 87.

    et al. The cancer cell oxygen sensor PHD2 promotes metastasis via activation of cancer-associated fibroblasts. Cell Rep. 12, 992–1005 (2015).

  88. 88.

    & Cancer associated fibroblasts: the dark side of the coin. Am. J. Cancer Res. 1, 482–497 (2011).

  89. 89.

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

  90. 90.

    , & Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin. Cancer Biol. 25, 47–60 (2014).

  91. 91.

    et al. Metabolic reprogramming and two-compartment tumor metabolism: opposing role(s) of HIF1α and HIF2α in tumor-associated fibroblasts and human breast cancer cells. Cell Cycle 11, 3280–3289 (2012).

  92. 92.

    et al. Targeting the lactate transporter MCT1 in endothelial cells inhibits lactate-induced HIF-1 activation and tumor angiogenesis. PLoS ONE 7, e33418 (2012).

  93. 93.

    et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Invest. 118, 3930–3942 (2008).

  94. 94.

    et al. Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 72, 5130–5140 (2012).

  95. 95.

    , & The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin. Cancer Biol. 25, 23–32 (2014).

  96. 96.

    et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121, 335–348 (2005).

  97. 97.

    et al. Regulation of the chemokine receptor CXCR4 by hypoxia. J. Exp. Med. 198, 1391–1402 (2003).

  98. 98.

    , , , & Tissue injury and hypoxia promote malignant progression of prostate cancer by inducing CXCL13 expression in tumor myofibroblasts. Proc. Natl Acad. Sci. USA 111, 14776–14781 (2014).

  99. 99.

    et al. Hypoxia-inducible factor 1-dependent expression of platelet-derived growth factor B promotes lymphatic metastasis of hypoxic breast cancer cells. Proc. Natl Acad. Sci. USA 109, E2707–E2716 (2012).

  100. 100.

    et al. Hypoxia-inducible factor-1 mediates the biological effects of oxygen on human trophoblast differentiation through TGFβ3. J. Clin. Invest. 105, 577–587 (2000).

  101. 101.

    , , & Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell 5, 429–441 (2004).

  102. 102.

    & Regulation of immune responses by L-arginine metabolism. Nat. Rev. Immunol. 5, 641–654 (2005).

  103. 103.

    et al. Arginase II expressed in cancer-associated fibroblasts indicates tissue hypoxia and predicts poor outcome in patients with pancreatic cancer. PLoS ONE 8, e55146 (2013).

  104. 104.

    et al. Granzyme B degradation by autophagy decreases tumor cell susceptibility to natural killer-mediated lysis under hypoxia. Proc. Natl Acad. Sci. USA 110, 17450–17455 (2013).

  105. 105.

    et al. HIF1α induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 13, 206–220 (2008).

  106. 106.

    & Regulation of immune cells by local-tissue oxygen tension: HIF1α and adenosine receptors. Nat. Rev. Immunol. 5, 712–721 (2005).

  107. 107.

    et al. Loss of prolyl hydroxylase-2 in myeloid cells and T-lymphocytes impairs tumor development. Int. J. Cancer 134, 849–858 (2014).

  108. 108.

    , & The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J. Pathol. 196, 254–265 (2002).

  109. 109.

    & Macrophage diversity enhances tumor progression and metastasis. Cell 141, 39–51 (2010).

  110. 110.

    et al. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 24, 695–709 (2013).

  111. 111.

    , & Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 104, 2224–2234 (2004).

  112. 112.

    et al. Regulation of EMAP II by hypoxia. Am. J. Pathol. 162, 93–103 (2003).

  113. 113.

    et al. Increased metabolites of 5-lipoxygenase from hypoxic ovarian cancer cells promote tumor-associated macrophage infiltration. Oncogene 34, 1241–1252 (2015).

  114. 114.

    et al. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF. Cancer Cell 19, 31–44 (2011).

  115. 115.

    et al. Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24, 589–602 (2013).

  116. 116.

    et al. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res. 74, 24–30 (2014).

  117. 117.

    et al. Differential activation and antagonistic function of HIF-α isoforms in macrophages are essential for NO homeostasis. Gene. Dev. 24, 491–501 (2010).

  118. 118.

    et al. HIF-1α is essential for myeloid cell-mediated inflammation. Cell 112, 645–657 (2003).

  119. 119.

    et al. Hypoxia-inducible factor 2α regulates macrophage function in mouse models of acute and tumor inflammation. J. Clin. Invest. 120, 2699–2714 (2010).

  120. 120.

    et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 70, 7465–7475 (2010).

  121. 121.

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

  122. 122.

    , & Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J. Biol. Chem. 277, 23111–23115 (2002).

  123. 123.

    et al. Modeling alveolar soft part sarcomagenesis in the mouse: a role for lactate in the tumor microenvironment. Cancer Cell 26, 851–862 (2014).

  124. 124.

    et al. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature 456, 814–818 (2008).

  125. 125.

    et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 207, 2439–2453 (2010).

  126. 126.

    et al. Tumor promoting effects of myeloid derived suppressor cells are potentiated by hypoxia-induced expression of miR-210. Cancer Res. 75, 3771–3787 (2015).

  127. 127.

    et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 72, 3906–3911 (2012).

  128. 128.

    et al. The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature 522, 106–110 (2015).

  129. 129.

    et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 440, 1222–1226 (2006).

  130. 130.

    , , & Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 11, 519–531 (2011).

  131. 131.

    et al. Anoxia/reoxygenation-induced neutrophil adherence to cultured endothelial cells. Am. J. Physiol. 262, H1891–H1898 (1992).

  132. 132.

    et al. Neutrophils oppose uterine epithelial carcinogenesis via debridement of hypoxic tumor cells. Cancer Cell 28, 785–799 (2015).

  133. 133.

    et al. Hypoxia-induced neutrophil survival is mediated by HIF-1α-dependent NF-κB activity. J. Exp. Med. 201, 105–115 (2005).

  134. 134.

    & The complex role of neutrophils in tumor angiogenesis and metastasis. Cancer Immunol. Res. 4, 83–91 (2016).

  135. 135.

    et al. Polarization of tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 16, 183–194 (2009).

  136. 136.

    et al. Galectin-1: a link between tumor hypoxia and tumor immune privilege. J. Clin. Oncol. 23, 8932–8941 (2005).

  137. 137.

    et al. Foxp3 expressing CD4+CD25high regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J. Immunol. 173, 1444–1453 (2004).

  138. 138.

    et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 475, 226–230 (2011).

  139. 139.

    et al. Ecto-5-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia. J. Clin. Invest. 110, 993–1002 (2002).

  140. 140.

    et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl Acad. Sci. USA 103, 13132–13137 (2006).

  141. 141.

    , , & E3 ubiquitin ligase VHL regulates hypoxia-inducible factor-1α to maintain regulatory T cell stability and suppressive capacity. Immunity 42, 1062–1074 (2015).

  142. 142.

    et al. Th17 cells in cancer: the ultimate identity crisis. Front. Immunol. 5, 276 (2014).

  143. 143.

    , , , & Hypoxia controls CD4+CD25+ regulatory T-cell homeostasis via hypoxia-inducible factor-1α. Eur. J. Immunol. 38, 2412–2418 (2008).

  144. 144.

    et al. Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc. Natl Acad. Sci. USA 109, E2784–E2793 (2012).

  145. 145.

    et al. HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 208, 1367–1376 (2011).

  146. 146.

    The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

  147. 147.

    et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 211, 781–790 (2014).

  148. 148.

    , , & A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 74, 665–674 (2014).

  149. 149.

    , , , & Structural integration in hypoxia-inducible factors. Nature 524, 303–308 (2015).

  150. 150.

    Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732 (2003).

Download references

Acknowledgements

We thank those whose work informed the writing of this manuscript and apologize to those authors whose elegant studies we were unable to acknowledge in this Review. This work was supported by NIH grants CA-67166 and CA-197713, the Silicon Valley Foundation, the Sydney Frank Foundation and the Kimmelman Fund (A.J.G.).

Author information

Affiliations

  1. Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, California 94305, USA

    • Edward L. LaGory
    •  & Amato J. Giaccia

Authors

  1. Search for Edward L. LaGory in:

  2. Search for Amato J. Giaccia in:

Competing interests

The authors declare no competing financial interests.

Corresponding author

Correspondence to Amato J. Giaccia.