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.

  • Article
  • Published:

Molecular landmarks of tumor hypoxia across cancer types

Abstract

Many primary-tumor subregions have low levels of molecular oxygen, termed hypoxia. Hypoxic tumors are at elevated risk for local failure and distant metastasis, but the molecular hallmarks of tumor hypoxia remain poorly defined. To fill this gap, we quantified hypoxia in 8,006 tumors across 19 tumor types. In ten tumor types, hypoxia was associated with elevated genomic instability. In all 19 tumor types, hypoxic tumors exhibited characteristic driver-mutation signatures. We observed widespread hypoxia-associated dysregulation of microRNAs (miRNAs) across cancers and functionally validated miR-133a-3p as a hypoxia-modulated miRNA. In localized prostate cancer, hypoxia was associated with elevated rates of chromothripsis, allelic loss of PTEN and shorter telomeres. These associations are particularly enriched in polyclonal tumors, representing a constellation of features resembling tumor nimbosus, an aggressive cellular phenotype. Overall, this work establishes that tumor hypoxia may drive aggressive molecular features across cancers and shape the clinical trajectory of individual tumors.

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: The hallmarks of hypoxia in cancer.
Fig. 2: Hypoxia associations with driver aberrations and miRNAs.
Fig. 3: Assessment and validation of hypoxia-associated miRNAs in prostate cancer.
Fig. 4: The landscape of hypoxia in prostate cancer.
Fig. 5: Interactions among hypoxia, HIF1A targets and PTEN in modulating telomere length.
Fig. 6: Subclonal evaluation of hypoxia.

Similar content being viewed by others

Data availability

The raw sequencing data have been deposited in the European Genome-phenome Archive under accession code EGAS00001000900. Processed variant calls are available through the ICGC Data Portal under the project PRAD-CA. TCGA data are available at https://portal.gdc.cancer.gov/projects/TCGA-PRAD. Previously published CPC-GENE data are available at the European Genome-phenome Archive under accession code EGAS00001000900. Previously published CPC-GENE mRNA abundance data are available at the Gene Expression Omnibus under accession code GSE84043.

References

  1. Wilson, W. R. & Hay, M. P. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer 11, 393–410 (2011).

    CAS  PubMed  Google Scholar 

  2. Harris, A. L. Hypoxia: a key regulatory factor in tumour growth. Nat. Rev. Cancer 2, 38–47 (2002).

    CAS  PubMed  Google Scholar 

  3. Bristow, R. G. & Hill, R. P. Hypoxia and metabolism: hypoxia, DNA repair and genetic instability. Nat. Rev. Cancer 8, 180–192 (2008).

    CAS  PubMed  Google Scholar 

  4. Weber, C. E. & Kuo, P. C. The tumor microenvironment. Surg. Oncol. 21, 172–177 (2012).

    PubMed  Google Scholar 

  5. Blagosklonny, M. V. Antiangiogenic therapy and tumor progression. Cancer Cell. 5, 13–17 (2004).

    CAS  PubMed  Google Scholar 

  6. Vaupel, P., Thews, O. & Hoeckel, M. Treatment resistance of solid tumors. Med. Oncol. 18, 243–260 (2001).

    CAS  PubMed  Google Scholar 

  7. Dhani, N., Fyles, A., Hedley, D. & Milosevic, M. The clinical significance of hypoxia in human cancers. Semin. Nucl. Med. 45, 110–121 (2015).

    PubMed  Google Scholar 

  8. Brown, J. M. & Wilson, W. R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 4, 437–447 (2004).

    CAS  PubMed  Google Scholar 

  9. Zannella, V. E. et al. Reprogramming metabolism with metformin improves tumor oxygenation and radiotherapy response. Clin. Cancer Res. 19, 6741–6750 (2013).

    CAS  PubMed  Google Scholar 

  10. Mucaj, V., Shay, J. E. S. & Simon, M. C. Effects of hypoxia and HIFs on cancer metabolism. Int. J. Hematol. 95, 464–470 (2012).

    CAS  PubMed  Google Scholar 

  11. Luoto, K. R., Kumareswaran, R. & Bristow, R. G. Tumor hypoxia as a driving force in genetic instability. Genome Integr. 4, 5 (2013).

    PubMed  PubMed Central  Google Scholar 

  12. Brizel, D. M., Dodge, R. K., Clough, R. W. & Dewhirst, M. W. Oxygenation of head and neck cancer: changes during radiotherapy and impact on treatment outcome. Radiother. Oncol. 53, 113–117 (1999).

    CAS  PubMed  Google Scholar 

  13. Nordsmark, M. & Overgaard, J. Tumor hypoxia is independent of hemoglobin and prognostic for loco-regional tumor control after primary radiotherapy in advanced head and neck cancer. Acta Oncol. 43, 396–403 (2004).

    PubMed  Google Scholar 

  14. Noman, M. Z. et al. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am. J. Physiol. Cell Physiol. 309, C569–C579 (2015).

    PubMed  PubMed Central  Google Scholar 

  15. Mohyeldin, A., Garzón-Muvdi, T. & Quiñones-Hinojosa, A. Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell 7, 150–161 (2010).

    CAS  PubMed  Google Scholar 

  16. Eliasson, P. & Jönsson, J.-I. The hematopoietic stem cell niche: low in oxygen but a nice place to be. J. Cell Physiol. 222, 17–22 (2010).

    CAS  PubMed  Google Scholar 

  17. Lin, P.-Y. et al. Expression of hypoxia-inducible factor-1α is significantly associated with the progression and prognosis of oral squamous cell carcinomas in Taiwan. J. Oral Pathol. Med. 37, 18–25 (2007).

    Google Scholar 

  18. Rankin, E. B. & Giaccia, A. J. Hypoxic control of metastasis. Science 352, 175–180 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Gilkes, D. M. & Semenza, G. L. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol. 9, 1623–1636 (2013).

    CAS  PubMed  Google Scholar 

  20. Zhong, H. et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 59, 5830–5835 (1999).

    CAS  PubMed  Google Scholar 

  21. Graeber, T. G. et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 379, 88–91 (1996).

    CAS  PubMed  Google Scholar 

  22. Greijer, A. E. & van der Wall, E. The role of hypoxia inducible factor 1 (HIF-1) in hypoxia induced apoptosis. J. Clin. Pathol. 57, 1009–1014 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Bindra, R. S. et al. Down-regulation of rad51 and decreased homologous recombination in hypoxic cancer cells. Mol. Cell Biol. 24, 8504–8518 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Mihaylova, V. T. et al. Decreased expression of the DNA mismatch repair gene Mlh1 under hypoxic stress in mammalian cells. Mol. Cell Biol. 23, 3265–3273 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Koshiji, M. et al. HIf-1α induces genetic instability by transcriptionally downregulating mutsα expression. Mol. Cell 17, 793–803 (2005).

    CAS  PubMed  Google Scholar 

  26. Lalonde, E. et al. Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study. Lancet Oncol. 15, 1521–1532 (2014).

    PubMed  Google Scholar 

  27. Lalonde, E. et al. Translating a prognostic dna genomic classifier into the clinic: retrospective validation in 563 localized prostate tumors. Eur. Urol. 72, 22–31 (2017).

    CAS  PubMed  Google Scholar 

  28. Janssens, G. O. et al. Accelerated radiotherapy with carbogen and nicotinamide for laryngeal cancer: results of a phase III randomized trial. J. Clin. Oncol. 30, 1777–1783 (2012).

    CAS  PubMed  Google Scholar 

  29. Hoskin, P. J., Rojas, A. M., Bentzen, S. M. & Saunders, M. I. Radiotherapy with concurrent carbogen and nicotinamide in bladder carcinoma. J. Clin. Oncol. 28, 4912–4918 (2010).

    PubMed  Google Scholar 

  30. Buffa, F. M., Harris, A. L., West, C. M. & Miller, C. J. Large meta-analysis of multiple cancers reveals a common, compact and highly prognostic hypoxia metagene. Br. J. Cancer 102, 428–435 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Winter, S. C. et al. Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers. Cancer Res. 67, 3441–3449 (2007).

    CAS  PubMed  Google Scholar 

  32. Ragnum, H. B. et al. The tumour hypoxia marker pimonidazole reflects a transcriptional programme associated with aggressive prostate cancer. Br. J. Cancer 112, 382–390 (2015).

    CAS  PubMed  Google Scholar 

  33. Eustace, A. et al. A 26-gene hypoxia signature predicts benefit from hypoxia-modifying therapy in laryngeal cancer but not bladder cancer. Clin. Cancer Res. 19, 4879–4888 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Sørensen, B. S., Toustrup, K., Horsman, M. R., Overgaard, J. & Alsner, J. Identifying pH independent hypoxia induced genes in human squamous cell carcinomas in vitro. Acta Oncol. (Madr.) 49, 895–905 (2010).

    Google Scholar 

  35. Elvidge, G. P. et al. Concordant regulation of gene expression by hypoxia and 2-oxoglutarate-dependent dioxygenase inhibition. J. Biol. Chem. 281, 15215–15226 (2006).

    CAS  PubMed  Google Scholar 

  36. Hu, Z. et al. A compact VEGF signature associated with distant metastases and poor outcomes. BMC Med. 7, 9 (2009).

    PubMed  PubMed Central  Google Scholar 

  37. Seigneuric, R. et al. Impact of supervised gene signatures of early hypoxia on patient survival. Radiother. Oncol. 83, 374–382 (2007).

    CAS  PubMed  Google Scholar 

  38. Hoskin, P. J. et al. Carbogen and nicotinamide in the treatment of bladder cancer with radical radiotherapy. Br. J. Cancer 76, 260–263 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Milholland, B., Auton, A., Suh, Y. & Vijg, J. Age-related somatic mutations in the cancer genome. Oncotarget 6, 24627–24635 (2015).

    PubMed  PubMed Central  Google Scholar 

  40. Yuan, Y. et al. Comprehensive characterization of molecular differences in cancer between male and female patients. Cancer Cell 29, 711–722 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Li, C. H., Haider, S., Shiah, Y.-J., Thai, K. & Boutros, P. C. Sex differences in cancer driver genes and biomarkers. Cancer Res. 78, 5527–5537 (2018).

    CAS  PubMed  Google Scholar 

  42. Hieronymus, H. et al. Copy number alteration burden predicts prostate cancer relapse. Proc. Natl Acad. Sci. USA 111, 11139–11144 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Vollan, H. K. M. et al. A tumor DNA complex aberration index is an independent predictor of survival in breast and ovarian cancer. Mol. Oncol. 9, 115–127 (2015).

    CAS  PubMed  Google Scholar 

  44. Curtis, C. et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486, 346–352 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Pereira, B. et al. The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat. Commun. 7, 11479 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Tan, E. Y. et al. The key hypoxia regulated gene CAIX is upregulated in basal-like breast tumours and is associated with resistance to chemotherapy. Br. J. Cancer 100, 405–411 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Zack, T. I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Huang, X. et al. Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol. Cell 35, 856–867 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Taylor, B. S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Sakamuro, D., Elliott, K. J., Wechsler-Reya, R. & Prendergast, G. C. BIN1 is a novel MYC-interacting protein with features of a tumour suppressor. Nat. Genet. 14, 69–77 (1996).

    CAS  PubMed  Google Scholar 

  52. Edwards, Y. H., Putt, W., Fox, M. & Ives, J. H. A novel human phosphoglucomutase (pgm5) maps to the centromeric region of chromosome 9. Genomics 30, 350–353 (1995).

    CAS  PubMed  Google Scholar 

  53. Milosevic, M. et al. Tumor hypoxia predicts biochemical failure following radiotherapy for clinically localized prostate cancer. Clin. Cancer Res. 18, 2108–2114 (2012).

    CAS  PubMed  Google Scholar 

  54. Fraser, M. et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature 541, 359–364 (2017).

    CAS  PubMed  Google Scholar 

  55. Espiritu, S. M. G. et al. The evolutionary landscape of localized prostate cancers drives clinical aggression. Cell 173, 1003–1013 (2018).

    CAS  PubMed  Google Scholar 

  56. Hopkins, J. F. et al. Mitochondrial mutations drive prostate cancer aggression. Nat. Commun. 8, 656 (2017).

    PubMed  PubMed Central  Google Scholar 

  57. Jackson, W. C. et al. Intermediate endpoints after postprostatectomy radiotherapy: 5-year distant metastasis to predict overall survival. Eur. Urol. https://doi.org/10.1016/j.eururo.2017.12.023 (2018).

    PubMed  Google Scholar 

  58. Chua, M. L. K. et al. A prostate cancer “nimbosus”: genomic instability and schlap1 dysregulation underpin aggression of intraductal and cribriform subpathologies. Eur. Urol. 71, 183–192 (2017).

    Google Scholar 

  59. Chua, M. L. K., van der Kwast, T. H. & Bristow, R. G. Intraductal carcinoma of the prostate: anonymous to ominous. Eur. Urol. 67, 496–503 (2017).

    Google Scholar 

  60. Benita, Y. et al. An integrative genomics approach identifies hypoxia inducible factor-1 (HIF-1)-target genes that form the core response to hypoxia. Nucleic Acids Res. 37, 4587–4602 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Ding, Z. et al. Estimating telomere length from whole genome sequence data. Nucleic Acids Res. 42, e75 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Semenza, G. L. Hypoxia, clonal selection, and the role of hif-1 in tumor progression. Crit. Rev. Biochem. Mol. Biol. 35, 71–103 (2000).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  64. Haider, S. et al. Genomic alterations underlie a pan-cancer metabolic shift associated with tumour hypoxia. Genome. Biol. 17, 140 (2016).

    PubMed  PubMed Central  Google Scholar 

  65. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Preprint at https://www.biorxiv.org/content/early/2017/08/30/161562 (2017).

  66. Zundel, W. et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 14, 391–396 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Hong, M. K. H. et al. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat. Commun. 6, 6605 (2015).

    CAS  PubMed  Google Scholar 

  68. Mertins, P. et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature 534, 55–62 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang, H. et al. Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell 166, 755–765 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Zhang, B. et al. Proteogenomic characterization of human colon and rectal cancer. Nature 513, 382–387 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Wilkerson, M. D. & Hayes, D. N. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics 26, 1572–1573 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Irizarry, R. A. et al. Summaries of affymetrix genechip probe level data. Nucleic Acids Res. 31, e15 (2003).

    PubMed  PubMed Central  Google Scholar 

  73. Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 12, R1 (2011).

    PubMed  PubMed Central  Google Scholar 

  74. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. McKenna, A. et al. The genome analysis toolkit: a mapreduce framework for analyzing next-generation dna sequencing data. Genome Res. 20, 1297–1303 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  77. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    PubMed  PubMed Central  Google Scholar 

  78. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28, i333–i339 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Govind, S. K. et al. ShatterProof: operational detection and quantification of chromothripsis. BMC Bioinformatics 5, 78 (2014).

    Google Scholar 

  80. Masella, A. P. et al. BAMQL: a query language for extracting reads from BAM files. BMC Bioinformatics 17, 305 (2016).

    PubMed  PubMed Central  Google Scholar 

  81. Ha, G. et al. TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 24, 1881–1893 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Deshwar, A. G. et al. PhyloWGS: reconstructing subclonal composition and evolution from whole-genome sequencing of tumors. Genome Biol. 16, 35 (2015).

    PubMed  PubMed Central  Google Scholar 

  83. Parker, C. et al. Polarographic electrode study of tumor oxygenation in clinically localized prostate cancer. Int. J. Radiat. Oncol. 58, 750–757 (2004).

    Google Scholar 

  84. P’ng, C. et al. BPG: seamless, automated and interactive visualization of scientific data. Preprint at https://www.biorxiv.org/content/early/2017/06/26/156067 (2017).

  85. Reimand, J., Kull, M., Peterson, H., Hansen, J. & Vilo, J. g:Profiler: a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res. 35, W193–W200 (2007).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was conducted with the support of Movember funds through Prostate Cancer Canada, and with the additional support of the Ontario Institute for Cancer Research, funded by the Government of Ontario. This work was supported by Prostate Cancer Canada and is funded by the Movember Foundation (grant RS2014-01). P.C.B. was supported by a Terry Fox Research Institute New Investigator Award, a Prostate Cancer Canada Rising Star Fellowship, and a Canadian Institutes of Health Research (CIHR) New Investigator Award. This work has been funded by Fellowships from the CIHR and the Ontario government to V.B. and E.L. S.K.L. is supported as a Movember Rising Star award recipient funded by the Movember Foundation (grants RS2014-03, D2015-12 and D2017-1811), the Telus Motorcycle Ride For Dad (Huronia Branch) and a Ministry of Research and Innovation Early Researcher Award. The authors thank the Princess Margaret Cancer Centre Foundation and Radiation Medicine Program Academic Enrichment Fund for support (to R.G.B.). This work was supported by a Terry Fox Research Institute Program Project Grant. R.G.B. is supported as a recipient of a Canadian Cancer Society Research Scientist Award. Laboratory work for R.G.B is supported by the CRUK Manchester Institute through Cancer Research UK. The authors thank all members of the Boutros and Bristow laboratoriess for helpful suggestions.

Author information

Authors and Affiliations

Authors

Contributions

V.B. conducted bioinformatics and statistical analysis; V.B., L.Y.L., E.L., J.L., R.L., Y.J.S., S.M.G.E., L.E.H., F.Y., V.H., T.N.Y., C.Q.Y. and V.Y.S. performed data processing; C.H., J.R., T.V. and X.H. performed in vitro experiments; V.B. performed data visualization; M.F., M.L.K.C., T.v.d.K., S.K.L., P.C.B. and R.G.B. supervised research; V.B., P.C.B. and R.G.B. initiated the project; V.B. wrote the first draft of the manuscript; all authors approved the manuscript.

Corresponding authors

Correspondence to Paul C. Boutros or Robert G. Bristow.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Pan-cancer hypoxia assessment in primary tumors.

a,b, Tumor hypoxia scores based on the Winter and Ragnum hypoxia signatures for 7,791 independent tumors from 19 tumor types. The median hypoxia score within each cancer type is indicated by the horizontal line. Sample sizes for each tumor type are listed along the bottom (representing independent tumors). Intertumoral variability in hypoxia, measured by interquartile range (IQR), is also shown along the bottom. c, Hypoxia scores from eight independent hypoxia signatures are highly correlated. For each pair of signatures, the Spearman’s ρ for hypoxia scores was calculated for each of the 19 tumor types. The value in each box represents the median ρ for a pair of signatures (ρ = 0.42 ± 0.21, overall mean ± s.d.). d, Comparison of hypoxia rankings across signatures for 19 tumor types from TCGA. Median hypoxia scores for the 19 tumor types were scaled from –1 to 1. Squamous carcinomas of the head and neck, lung and cervix are consistently observed as the most hypoxic. e, Comparison of the distribution of hypoxia scores between the Winter, Buffa and Ragnum hypoxia scores (n = 7,791 independent tumors; Spearman’s ρ, P value calculated using algorithm AS89). Tumor type codes are defined in Supplementary Table 2.

Supplementary Figure 2 Influence of hypoxia on protein abundance.

ac, Comparison of tumor hypoxia scores generated from mRNA abundance and protein abundance data using the Buffa hypoxia signature for BRCA (breast invasive carcinoma) (a), OV (ovarian serous cystadenocarcinoma) (b) and COADREAD (colon adenocarcinoma and rectum adenocarcinoma) (c). Protein-abundance-based hypoxia scores were significantly correlated with mRNA-abundance-based hypoxia scores for all three tumor types. d, The abundance of ten proteins was significantly correlated with protein-based hypoxia scores across all three cancers (FDR < 0.05 in all three cancer types). nBRCA = 77 independent tumors, nOV = 102 independent tumors, nCOADREAD = 86 independent tumors for ad. Spearman’s ρ was used to determine each correlation and P values were calculated using algorithm AS89.

Supplementary Figure 3 Pan-cancer associations of hypoxia with sex, age and ancestry, and power to detect hypoxia-associated SNVs.

ac, Significant differences in tumor hypoxia between females and males were not seen consistently based on all three hypoxia signatures for any tumor type (Mann–Whitney U test). Bonferroni-adjusted P values are shown along the top. d, Younger patients with lung adenocarcinoma had higher hypoxia scores. However, this association was confounded by smoking status. Background color indicates Bonferroni-adjusted P values (algorithm AS89) and dot size indicates the magnitude of the correlation (Spearman’s ρ). eg, Significant differences in tumor hypoxia were consistently seen in patients with breast invasive carcinomas (Kruskal–Wallis test). Bonferroni-adjusted P values are shown along the top. h, Power analysis for a Mann–Whitney U test to detect hypoxia score differences between patients with and without an SNV. The breast invasive carcinoma (BRCA), lung adenocarcinoma (LUAD) and renal clear cell carcinoma (KIRC) cohorts were well powered for this analysis. All tumor type codes are defined in Supplementary Table 2. Tukey box plots are shown in ac, eg. na–c, d, e–g independent tumors = 408, 408, 391 BLCA; NA, 1,093, 997 BRCA; NA, 304, 259 CESC; 377, 377, 342 COADREAD; 159, 159, 158 GBM; 520, 519, 503 HNSC; 533, 533, 526 KIRC; 290, 287, 273 KIRP; 515, 515, 504 LGG; 371, 370, 359 LIHC; 515, 496, 448 LUAD; 501, 492, 388 LUSC; NA, 304, 290 OV; 178, 178, 174 PAAD; 179, NA, 174 PCPG; NA, 333, 143 PRAD; 469, 461, 459 SKCM; 501, 501, 409 THCA; NA, 174, 153 UCEC. All tests were two-sided.

Supplementary Figure 4 Validation of hypoxia-associated alterations in breast cancer, associations with subtypes, pan-cancer signature agreements and miRNA–protein associations.

a, Validation of hypoxia-associated CNAs and SNVs in breast cancer using an independent cohort (Nature 486, 346–352, 2012; n = 1,859 independent tumors; Mann–Whitney U test). bd, Hypoxia scores were significantly different between subtypes of breast cancer in TCGA (b), METABRIC (c) and CPTAC (d) where hypoxia scores were calculated based on protein abundance (n = 817, 1,985 and 77 independent tumors, respectively; Kruskal–Wallis test; Tukey box plots are shown). e, Elevated protein-based hypoxia scores are associated with elevated abundance of TP53 (n = 68 independent tumors; Spearman’s ρ, AS89). fo, Patients with mutations in TP53 had elevated hypoxia scores compared to patients with wild-type TP53 within the different breast cancer subtypes in TGGA and METABRIC (Mann–Whitney U test). pr, We assessed agreement between the Winter, Buffa and Ragnum signatures by comparing whether the different signatures considered a driver gene to be significantly (FDR < 0.05) associated with hypoxia or not. For 98.2 ± 3.1%, 72.4 ± 23.4% and 61.9 ± 16.9% (mean ± s.d.) of driver SNVs (p), CNAs (q) and miRNAs (r), respectively, all three signatures agreed on it being associated or not associated with hypoxia, indicating that the molecular associations we highlight in Fig. 2 are broadly informative about biology across cancer types. np,q and r independent tumors = 388, 405 BLCA; 960, 753 BRCA; 190, 304 CESC; 259, 295 COADREAD; 137 GBM; 497, 478 HNSC; 431, 254 KIRC; 280, 290 KIRP; 513, 512 LGG; 360, 367 LIHC; 475, 447 LUAD; 178, 342 LUSC; 246, 288 OV; 119, 178 PAAD; 162, 179 PCPG; 333, 330 PRAD; 290, 97 SKCM; 486, 500 THCA; 8, 174 UCEC. Correlations between miR-210, a hypoxia-associated miRNA, and protein abundance in BRCA (s; n = 32 independent tumors) and OV (t; n = 139 independent tumors). Results for the top 20 proteins are shown. The protein abundance of lactate dehydrogenase A (LDHA) was significantly positively correlated with the abundance of miR-210 in BRCA (s) and OV (t) (Spearman’s ρ, algorithm AS89). Tumor type codes are defined in Supplementary Table 2. All tests were two-sided.

Supplementary Figure 5 Hypoxia associations with clinicopathological features, TERT mRNA abundance, PTEN mRNA abundance and mitochondrial mutations.

a, Tumor hypoxia increases with T-category (n = 405 independent tumors). b, Tumor hypoxia significantly differs by Gleason score (n = 479 independent tumors). c, Mutations in the origin of heavy strand replication (OHR) and MYC have previously been associated with poor prognosis in localized prostate cancer (Nat. Commun. 8, 656, 2017). In line with this prognostic association, a significant difference in hypoxia score was noted based on OHR and MYC mutation status (n = 152 independent tumors). Patients in the CPC-GENE (d; n = 191 independent tumors) and TCGA (e; n = 308 independent tumors) cohorts with the aggressive IDC-CA pathological feature have significantly higher tumor hypoxia scores compared to patients without IDC-CA. f, PTEN mRNA abundance is negatively correlated with the mRNA abundance of TERT, a HIF-1A target, in the TCGA cohort (n = 333 independent tumors; Spearman’s ρ, algorithm AS89). g, PTEN mRNA abundance levels differ based on hypoxia and TERT mRNA abundance (n = 333 independent tumors). h, Monoclonal tumors with high hypoxia scores have higher TERT mRNA abundance than other subgroups of tumors based on clonality and hypoxia (n = 125 independent tumors). i, The mRNA abundance of PTEN is modulated by both PTEN copy number status and hypoxia (n = 60 independent tumors). Tukey box plots are shown in ae, gi. Kruskal–Wallis tests used for ac, gi. Mann–Whitney U test used for d and e. All tests were two-sided.

Supplementary Figure 6 Enrichment, mRNA confirmation and pathway analysis of hypoxia-associated CNAs, hypoxia-associated events in monoclonal tumors and a model for hypoxia as a driver of aggressive prostate cancer.

a, Hypoxia-associated CNA hits were enriched on chromosomes 7 and 10. Numbers along the top are Bonferroni-adjusted P values (n = 360 independent tumors; hypergeometric test). Comparison of mRNA abundance in TCGA (b; n = 148 independent tumors) and CPC-GENE (c; n = 210 independent tumors) between tumors that have a copy number loss or gain to tumors that are neutral for hypoxia-associated genes at the CNA level (Mann–Whitney U test). FDR-adjusted P values are shown along the top (Mann–Whitney U test). Fold changes are shown along the bottom. d, Pathways related to the 20 genes associated with hypoxia at the CNA level and functionally confirmed at the mRNA level. Dot size indicates the number of genes in the data set that are in the gene set. Dot color indicates the q value. The color of the lines connecting the nodes indicates the overlap between connected gene sets. e,f, PTEN mRNA abundance is influenced by hypoxia together with PTEN copy number status (e; n = 60 independent tumors; Kruskal–Wallis test) and IDC-CA (f; n = 57 independent tumors; Kruskal–Wallis test). g, Allelic loss of PTEN and IDC/CA are often observed in the same monoclonal tumors (Fisher’s exact test). h, Hypoxia applies a selective pressure within prostate tumors, driving the selection of aggressive tumor subclones. Driver mutations are shown as yellow, orange and red stars. The onset of hypoxia is shown as a gradient from white to blue, with subsequent reoxygenation shown as a gradient from blue to white. Once normal levels of oxygen are reestablished, aggressive subclones that survived the hypoxic microenvironment can rapidly expand. Tukey box plots are shown in b, c, e and f. All tests were two-sided.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Note

Reporting Summary

Supplementary Table 1

Pancancer hypoxia scores

Supplementary Table 2

TCGA tumor type descriptions

Supplementary Table 3

Molecular correlates of hypoxia in breast, lung and kidney cancer

Supplementary Table 4

CPC-GENE data

Supplementary Table 5

Hypoxia-associated CNAs in localized prostate cancer

Supplementary Table 6

miRNA sequence information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bhandari, V., Hoey, C., Liu, L.Y. et al. Molecular landmarks of tumor hypoxia across cancer types. Nat Genet 51, 308–318 (2019). https://doi.org/10.1038/s41588-018-0318-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0318-2

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