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.

Systematic analysis of noncoding somatic mutations and gene expression alterations across 14 tumor types

Subjects

Abstract

Somatic mutations in noncoding sequences are poorly explored in cancer, a rare exception being the recent identification of activating mutations in TERT regulatory DNA. Although this finding is suggestive of a general mechanism for oncogene activation, this hypothesis remains untested. Here we map somatic mutations in 505 tumor genomes across 14 cancer types and systematically screen for associations between mutations in regulatory regions and RNA-level changes. We identify recurrent promoter mutations in several genes but find that TERT mutations are exceptional in showing a strong and genome-wide significant association with increased expression. Detailed analysis of TERT across cancers shows that the strength of this association is highly variable and is strongest in copy number–stable cancers such as thyroid carcinoma. We additionally propose that TERT promoter mutations control expression of the nearby gene CLPTM1L. Our analysis provides a detailed pan-cancer view of TERT transcriptional activation but finds no clear evidence for frequent oncogenic promoter mutations beyond TERT.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Whole-genome mapping of somatic mutations and screening for associations with RNA-level changes across 505 tumors.
Figure 2: Quantile-quantile plots of mutation-to-expression association scores using increasingly stringent search constraints.
Figure 3: Pan-cancer view of TERT expression levels and upstream DNA mutations.
Figure 4: Recurrent somatic mutations near TSSs.

Similar content being viewed by others

References

  1. Yates, L.R. & Campbell, P.J. Evolution of the cancer genome. Nat. Rev. Genet. 13, 795–806 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Watson, I.R., Takahashi, K., Futreal, P.A. & Chin, L. Emerging patterns of somatic mutations in cancer. Nat. Rev. Genet. 14, 703–718 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Garraway, L.A. & Lander, E.S. Lessons from the cancer genome. Cell 153, 17–37 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Li, Q. et al. Integrative eQTL-based analyses reveal the biology of breast cancer risk loci. Cell 152, 633–641 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wittkopp, P.J. & Kalay, G. Cis-regulatory elements: molecular mechanisms and evolutionary processes underlying divergence. Nat. Rev. Genet. 13, 59–69 (2012).

    Article  CAS  Google Scholar 

  6. Huang, F.W. et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Martínez, P. & Blasco, M.A. Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nat. Rev. Cancer 11, 161–176 (2011).

    Article  PubMed  Google Scholar 

  9. Vinagre, J. et al. Frequency of TERT promoter mutations in human cancers. Nat. Commun. 4, 2185 (2013).

    Article  PubMed  Google Scholar 

  10. Killela, P.J. et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc. Natl. Acad. Sci. USA 110, 6021–6026 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lawrence, M.S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kandoth, C. et al. Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013).

    Article  PubMed  Google Scholar 

  13. Alexandrov, L.B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Forbes, S.A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Yamamoto, K., Okamoto, A., Isonishi, S., Ochiai, K. & Ohtake, Y. A novel gene, CRR9, which was up-regulated in CDDP-resistant ovarian tumor cell line, was associated with apoptosis. Biochem. Biophys. Res. Commun. 280, 1148–1154 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Rafnar, T. et al. Sequence variants at the TERT-CLPTM1L locus associate with many cancer types. Nat. Genet. 41, 221–227 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Haiman, C.A. et al. A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor–negative breast cancer. Nat. Genet. 43, 1210–1214 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shete, S. et al. Genome-wide association study identifies five susceptibility loci for glioma. Nat. Genet. 41, 899–904 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kote-Jarai, Z. et al. Fine-mapping identifies multiple prostate cancer risk loci at 5p15, one of which associates with TERT expression. Hum. Mol. Genet. 22, 2520–2528 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bojesen, S.E. et al. Multiple independent variants at the TERT locus are associated with telomere length and risks of breast and ovarian cancer. Nat. Genet. 45, 371–384 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Speedy, H.E. et al. A genome-wide association study identifies multiple susceptibility loci for chronic lymphocytic leukemia. Nat. Genet. 46, 56–60 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Jia, J. et al. CLPTM1L promotes growth and enhances aneuploidy in pancreatic cancer cells. Cancer Res. 74, 2785–2795 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Allory, Y. et al. Telomerase reverse transcriptase promoter mutations in bladder cancer: high frequency across stages, detection in urine, and lack of association with outcome. Eur. Urol. 65, 360–366 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Hurst, C.D., Platt, F.M. & Knowles, M.A. Comprehensive mutation analysis of the TERT promoter in bladder cancer and detection of mutations in voided urine. Eur. Urol. 65, 367–369 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Rachakonda, P.S. et al. TERT promoter mutations in bladder cancer affect patient survival and disease recurrence through modification by a common polymorphism. Proc. Natl. Acad. Sci. USA 110, 17426–17431 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wu, S. et al. Telomerase reverse transcriptase gene promoter mutations help discern the origin of urogenital tumors: a genomic and molecular study. Eur. Urol. 65, 274–277 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Liu, X. et al. Highly prevalent TERT promoter mutations in aggressive thyroid cancers. Endocr. Relat. Cancer 20, 603–610 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Brennan, C.W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wu, G. et al. The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat. Genet. 46, 444–450 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang, K. et al. TERT promoter mutations in renal cell carcinomas and upper tract urothelial carcinomas. Oncotarget 5, 1829–1836 (2014).

    PubMed  PubMed Central  Google Scholar 

  31. Wu, R.-C. et al. Frequent somatic mutations of the telomerase reverse transcriptase promoter in ovarian clear cell carcinoma but not in other major types of gynaecological malignancy. J. Pathol. 232, 473–481 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cesare, A.J. & Reddel, R.R. Alternative lengthening of telomeres: models, mechanisms and implications. Nat. Rev. Genet. 11, 319–330 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Heidenreich, B. et al. Telomerase reverse transcriptase promoter mutations in primary cutaneous melanoma. Nat. Commun. 5, 3401 (2014).

    Article  PubMed  Google Scholar 

  34. Vivanco, I. & Sawyers, C.L. The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat. Rev. Cancer 2, 489–501 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Weinhold, N., Jacobsen, A., Schultz, N., Sander, C. & Lee, W. Genome-wide analysis of noncoding regulatory mutations in cancer. Nat. Genet. 46, 1160–1165 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, S., Milne, G.T., Kuremsky, J.G., Fink, G.R. & Leppla, S.H. Identification of the proteins required for biosynthesis of diphthamide, the target of bacterial ADP-ribosylating toxins on translation elongation factor 2. Mol. Cell. Biol. 24, 9487–9497 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang, L. et al. Silencing of diphthamide synthesis 3 (Dph3) reduces metastasis of murine melanoma. PLoS ONE 7, e49988 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Nobukuni, Y., Kohno, K. & Miyagawa, K. Gene trap mutagenesis-based forward genetic approach reveals that the tumor suppressor OVCA1 is a component of the biosynthetic pathway of diphthamide on elongation factor 2. J. Biol. Chem. 280, 10572–10577 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Liang, M. et al. Ovca1, a candidate gene of the genetic modifier of Tp53, Mop2, affects mouse embryonic lethality. Genes Chromosom. Cancer 47, 315–325 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Kong, F. et al. OVCA1 inhibits the proliferation of epithelial ovarian cancer cells by decreasing cyclin D1 and increasing p16. Mol. Cell. Biochem. 354, 199–205 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Chen, C.M. & Behringer, R.R. Ovca1 regulates cell proliferation, embryonic development, and tumorigenesis. Genes Dev. 18, 320–332 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bruening, W. et al. Expression of OVCA1, a candidate tumor suppressor, is reduced in tumors and inhibits growth of ovarian cancer cells. Cancer Res. 59, 4973–4983 (1999).

    CAS  PubMed  Google Scholar 

  43. Berger, M.F. et al. Melanoma genome sequencing reveals frequent PREX2 mutations. Nature 485, 502–506 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wu, M.C. et al. Rare-variant association testing for sequencing data with the sequence kernel association test. Am. J. Hum. Genet. 89, 82–93 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li, B. & Leal, S.M. s for detecting associations with rare variants for common diseases: application to analysis of sequence data. Am. J. Hum. Genet. 83, 311–321 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lin, D.Y. & Tang, Z.Z. A general framework for detecting disease associations with rare variants in sequencing studies. Am. J. Hum. Genet. 89, 354–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shtivelman, E. et al. Pathways and therapeutic targets in melanoma. Oncotarget 5, 1701–1752 (2014).

    PubMed  PubMed Central  Google Scholar 

  48. Koboldt, D.C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Harrow, J. et al. GENCODE: the reference human genome annotation for the ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Anders, S., Pyl, P.T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 10.1093/bioinformatics/btu638 (25 September 2014).

  51. Akrami, R. et al. Comprehensive analysis of long non-coding RNAs in ovarian cancer reveals global patterns and targeted DNA amplification. PLoS ONE 8, e80306 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  53. McLaren, W. et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069–2070 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Cerami, E. et al. The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  56. Sensi, M. et al. Mutually exclusive NRASQ61R and BRAFV600E mutations at the single-cell level in the same human melanoma. Oncogene 25, 3357–3364 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Untergasser, A. et al. Primer3—new capabilities and interfaces. Nucleic Acids Res. 40, e115 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The results published here are based in whole or in part on data generated by the TCGA pilot project established by the National Cancer Institute and National Human Genome Research Institute. Information about TCGA and the investigators and institutions who constitute the TCGA research network can be found at http://cancergenome.nih.gov/. This work was supported by grants from the Swedish Foundation for Strategic Research (E.L. and N.J.F.); the Swedish Medical Research Council (E.L. and J.A.N.); the Swedish Cancer Society (E.L. and J.A.N.); the Magnus Bergvall Foundation (E.L.); the Åke Wiberg foundation (E.L.); the Lars Hierta Memorial Foundation (E.L.); Region Västra Götaland (J.A.N.); BioCARE (J.A.N.); and the Sahlgrenska Academy (J.A.N.). The computations were performed in part on resources provided by the Swedish National Infrastructure on Computing through the Uppsala Multidisciplinary Center for Advanced Computational Science (UPPMAX) under project b2012108. We thank the patients and their families; B. Einarsdottir and L. Nilsson for processing patient samples; R. Olofsson, U. Stierner and J. Mattsson for patient discussions; and members of our laboratory for valuable comments.

Author information

Authors and Affiliations

Authors

Contributions

N.J.F. and E.L. analyzed data. N.J.F. performed experiments. J.A.N. collected and processed clinical material and information. L.N. recruited patients, evaluated clinical data and performed medical record follow up. E.L. and N.J.F. wrote the paper. E.L. conceived the study.

Corresponding author

Correspondence to Erik Larsson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quantile-quantile plots of mutation-to-expression association scores while taking copy-number changes into account.

Rank-based scores (Z-statistic) were used to assess associations between gene expression levels and presence/absence of somatic mutations in 100 bp genomic segments, such that activation gives positive scores and inactivation gives negative scores. Scores obtained in each cancer type were summarized into a pan-cancer score, considering the best-scoring genomic segment for each gene. The analysis is similar to that depicted in the main Fig. 2, but with additional measures taken to compensate for the impact of copy-number changes (see Methods).

Supplementary Figure 2 Positive association between TERT promoter mutations and mRNA levels of the neighboring CLPTM1L revealed by filtering for ETS-site forming mutations.

(a) Q-Q plot of expected vs. observed scores (Z-statistic for mutation-expression association summed across cancer types), when only considering mutations in DNase I hypersensitive regions that also contribute to formation of an ETS factor consensus sequence (GGAA or its reverse complement). A maximum distance of +/- 100 kb to the transcription start site was allowed. Gray lines indicate the 90% confidence interval, determined using 200 simulations (dots above the line are in the upper 5% percentile, and thus significant at the corrected P < 0.05 level). A similar screen for ETS-site disrupting mutations did not produce any significant associations. (b) Box plots showing that expression levels of CLPTML1, which is located close to TERT on chromosome 5, are positively correlated with mutations in the TERT promoter in several cancer types. The effect was strongest in LGG and THCA. The fraction of tumors with TERT promoter mutations is indicated in the figure titles. The central mark is the median and the box edges are the 25th and 75th percentiles, with individual data points indicated in red. P-values were calculated using the two-sided Wilcoxon rank sum test (unadjusted).

Supplementary Figure 3 Statistical power and sparseness of somatic mutations.

(a) Histogram showing the percentage of genomic tiles (100 bp genomic segments) that were somatically mutated in >1, >2 or more tumors, counted across all 505 analyzed tumor genomes. In the right panel, the percentages refer to tiles that, in addition to being mutated, also overlap with DNase I hypersensitive regions. (b) Theoretical power analysis using simulated mutations. Tumors were randomly selected to be targeted by artificial mutations at one specific tile in the ACTB promoter. The endogenous ACTB mRNA level was elevated by a factor 1.5 or 2 in mutated tumors, while otherwise retaining the normal gene expression profile. Mutations were spread randomly across all tumors and cancer types, to mimic a pan-cancer mutational pattern. The analysis was performed using DNase I HS filtering while including all genes (no COSMIC filter), with variable settings for the maximum TSS distance (y-axis). The number of introduced mutations was varied (x-axis), and the experiment was repeated 100 times for each set of parameters. Apart from the simulated mutations, the analysis was identical to the main screen, including correction for multiple testing using random permutations. Red tiles indicate conditions in which the compound score would, on average, reach significance at the corrected P < 0.05 level.

Supplementary Figure 4 No reproducible association with expression levels for PLEKHS1 or DPH3 noncoding mutations.

(a) PLEKHS1 mRNA levels were similar in 6 PLEKHS1 mutated vs. 15 non-mutated bladder carcinomas (BLCA; this cancer type had the highest fraction of PLEKHS1 mutated samples). (b) A positive association observed for DPH3 in 6 mutated vs. 32 non-mutated melanomas (SKCM) was too weak too reach genome-wide significance in the screen (main Fig. 2), and the effect was not replicated in 25/148 mutated/non-mutated melanomas genotyped using exome sequencing data (WXS) (Supplementary Table 7/Supplementary Fig. 6). No mRNA structural changes were revealed by inspection of RNA-seq alignments in mutated and non-mutated samples for these genes (based on WGS samples). P-values were calculated using the two-sided Wilcoxon rank sum test.

Supplementary Figure 5 Regulatory and sequence context of recurrently mutated noncoding positions in PLEKHS1 and DPH3.

(a) Somatically mutated positions in PLEKHS1, 70 and 73 bp into the first intron, are flanked by sequences of perfect reverse complementarity. Complementary sequences are 11 bp long including the mutated positions themselves. (b) Recurrent somatic mutations upstream of DPH3, located in a region of strong regulatory potential with multiple overlapping ChIP-seq peaks including ETS factors ELK4/ELF1 and ELK1. The mutations flank an ETS core motif sequence. In both panels ChIP-seq data for 161 transcription factors (TF) from ENCODE (Gerstein et al. Nature 2012, 489:91-100), visualized using the UCSC browser ENCODE Regulation track, is indicated at the bottom, with predicted TF binding sites from FactorBook (Wang et al. Nucleic Acids Res 2013, 41:D171-176) shown in green.

Supplementary Figure 6 Exome-based genotyping of DPH3 promoter mutations.

Exome data from TCGA was used to genotype the DPH3 promoter for mutations in the positions highlighted in main Fig. 4b. Data was obtained for 176 independent SKCM tumors (TCGA sample type 6, metastatis, which is the main type considered by TCGA for melanoma and the only type in our WGS analysis), and 242 SKCM normals (TCGA sample type 10). All included samples had at least 2 exome reads covering the positions of interest. Mutations were detected in 25/176 tumors and 0/242 normals. The histograms indicate the minimum read coverage in the three relevant positions (-25, -26 and -29) in each tumor. For validation we used a set of 25 tumors with both exome and WGS data, and found that the exome-based method could rediscover 4/5 mutations detected by WGS with no additional positives.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6, Supplementary Tables 3, 5, 6 and 8–11 and Supplementary Note (PDF 1321 kb)

Supplementary Table 1

Detailed information regarding included TCGA WGS and RNA-seq samples including UUID codes for cgHub retrevial. (XLSX 114 kb)

Supplementary Table 2

WGS read coverage information for the positions of recurrent mutations in the promoter regions of DPH3, TERT and PLEKHS1. (XLSX 131 kb)

Supplementary Table 4

Somatic mutations detected upstream of the TERT TSS. (XLSX 52 kb)

Supplementary Table 7

Genotyping of DPH3 promoter mutations using TCGA exome (WXS) data. (XLSX 76 kb)

Supplementary Data Set 1

Supplementary Data Set 1 (ZIP 6010 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fredriksson, N., Ny, L., Nilsson, J. et al. Systematic analysis of noncoding somatic mutations and gene expression alterations across 14 tumor types. Nat Genet 46, 1258–1263 (2014). https://doi.org/10.1038/ng.3141

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3141

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