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.

  • Letter
  • Published:

Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations

Abstract

Meningiomas are the most common primary nervous system tumor. The tumor suppressor NF2 is disrupted in approximately half of all meningiomas1, but the complete spectrum of genetic changes remains undefined. We performed whole-genome or whole-exome sequencing on 17 meningiomas and focused sequencing on an additional 48 tumors to identify and validate somatic genetic alterations. Most meningiomas had simple genomes, with fewer mutations, rearrangements and copy-number alterations than reported in other tumors in adults. However, several meningiomas harbored more complex patterns of copy-number changes and rearrangements, including one tumor with chromothripsis. We confirmed focal NF2 inactivation in 43% of tumors and found alterations in epigenetic modifiers in an additional 8% of tumors. A subset of meningiomas lacking NF2 alterations harbored recurrent oncogenic mutations in AKT1 (p.Glu17Lys) and SMO (p.Trp535Leu) and exhibited immunohistochemical evidence of activation of these pathways. These mutations were present in therapeutically challenging tumors of the skull base and higher grade. These results begin to define the spectrum of genetic alterations in meningiomas and identify potential therapeutic targets.

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

Figure 1: Landscape of somatic alterations in the grade I meningioma genome.
Figure 2: Somatic rearrangements disrupt tumor suppressors in several meningiomas.
Figure 3: Significant and selected cancer-related somatic mutations, indels and translocations in meningiomas.
Figure 4: Associations between mutations in Hedgehog and AKT-mTOR pathways and histological findings.

Similar content being viewed by others

References

  1. Choy, W. et al. The molecular genetics and tumor pathogenesis of meningiomas and the future directions of meningioma treatments. Neurosurg. Focus 30, E6 (2011).

    Article  PubMed  Google Scholar 

  2. van Alkemade, H. et al. Impaired survival and long-term neurological problems in benign meningioma. Neuro-oncol. 14, 658–666 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Durand, A. et al. WHO grade II and III meningiomas: a study of prognostic factors. J. Neurooncol. 95, 367–375 (2009).

    Article  PubMed  Google Scholar 

  4. Perry, A., Scheithauer, B.W., Stafford, S.L., Lohse, C.M. & Wollan, P.C. “Malignancy” in meningiomas: a clinicopathologic study of 116 patients, with grading implications. Cancer 85, 2046–2056 (1999).

    CAS  PubMed  Google Scholar 

  5. Goutagny, S. et al. Genomic profiling reveals alternative genetic pathways of meningioma malignant progression dependent on the underlying NF2 status. Clin. Cancer Res. 16, 4155–4164 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Krayenbühl, N., Pravdenkova, S. & Al-Mefty, O. De novo versus transformed atypical and anaplastic meningiomas: comparisons of clinical course, cytogenetics, cytokinetics, and outcome. Neurosurgery 61, 495–503, discussion 503–504 (2007).

    Article  PubMed  Google Scholar 

  7. Wen, P.Y., Quant, E., Drappatz, J., Beroukhim, R. & Norden, A.D. Medical therapies for meningiomas. J. Neurooncol. 99, 365–378 (2010).

    Article  CAS  PubMed  Google Scholar 

  8. Bass, A.J. et al. Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTI1A-TCF7L2 fusion. Nat. Genet. 43, 964–968 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Berger, M.F. et al. The genomic complexity of primary human prostate cancer. Nature 470, 214–220 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chapman, M.A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467–472 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Stransky, N. et al. The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157–1160 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Lohr, J.G. et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc. Natl. Acad. Sci. USA 109, 3879–3884 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mawrin, C. & Perry, A. Pathological classification and molecular genetics of meningiomas. J. Neurooncol. 99, 379–391 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Ketter, R. et al. Hyperdiploidy defines a distinct cytogenetic entity of meningiomas. J. Neurooncol. 83, 213–221 (2007).

    Article  PubMed  Google Scholar 

  18. Nunes, F. et al. Inactivation patterns of NF2 and DAL-1/4.1B (EPB41L3) in sporadic meningioma. Cancer Genet. Cytogenet. 162, 135–139 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Stephens, P.J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Takita, J. et al. Aberrations of NEGR1 on 1p31 and MYEOV on 11q13 in neuroblastoma. Cancer Sci. 102, 1645–1650 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Yang, H.W. et al. Alternative splicing of CHEK2 and codeletion with NF2 promote chromosomal instability in meningioma. Neoplasia 14, 20–28 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kooistra, S.M. & Helin, K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol. 13, 297–311 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Hadfield, K.D. et al. Molecular characterisation of SMARCB1 and NF2 in familial and sporadic schwannomatosis. J. Med. Genet. 45, 332–339 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Schmitz, U. et al. INI1 mutations in meningiomas at a potential hotspot in exon 9. Br. J. Cancer 84, 199–201 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tsurusaki, Y. et al. Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome. Nat. Genet. 44, 376–378 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Reifenberger, J. et al. Missense mutations in SMOH in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res. 58, 1798–1803 (1998).

    CAS  PubMed  Google Scholar 

  28. Jones, D.T. et al. Dissecting the genomic complexity underlying medulloblastoma. Nature 488, 100–105 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Laurendeau, I. et al. Gene expression profiling of the hedgehog signaling pathway in human meningiomas. Mol. Med. 16, 262–270 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Aavikko, M. et al. Loss of SUFU function in familial multiple meningioma. Am. J. Hum. Genet. 91, 520–526 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gorlin, R.J. Nevoid basal-cell carcinoma syndrome. Medicine (Baltimore) 66, 98–113 (1987).

    Article  CAS  Google Scholar 

  32. Santos, D.C. et al. PTCH1 gene mutations in exon 17 and loss of heterozygosity on D9S180 microsatellite in sporadic and inherited human basal cell carcinomas. Int. J. Dermatol. 50, 838–843 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Bleeker, F.E. et al. AKT1E17K in human solid tumours. Oncogene 27, 5648–5650 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Wellenreuther, R. et al. Analysis of the neurofibromatosis 2 gene reveals molecular variants of meningioma. Am. J. Pathol. 146, 827–832 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Mathiesen, T., Lindquist, C., Kihlstrom, L. & Karlsson, B. Recurrence of cranial base meningiomas. Neurosurgery 39, 2–7, discussion 8–9 (1996).

    Article  CAS  PubMed  Google Scholar 

  36. Ellison, D.W. et al. Medulloblastoma: clinicopathological correlates of SHH, WNT, and non-SHH/WNT molecular subgroups. Acta Neuropathol. 121, 381–396 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Karst, A.M. et al. Stathmin 1, a marker of PI3K pathway activation and regulator of microtubule dynamics, is expressed in early pelvic serous carcinomas. Gynecol. Oncol. 123, 5–12 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Riobó, N.A., Lu, K., Ai, X., Haines, G.M. & Emerson, C.P. Jr. Phosphoinositide 3-kinase and Akt are essential for Sonic Hedgehog signaling. Proc. Natl. Acad. Sci. USA 103, 4505–4510 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Von Hoff, D.D. et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N. Engl. J. Med. 361, 1164–1172 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Janku, F. et al. PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J. Clin. Oncol. 30, 777–782 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Parsons, D.W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Samuels, Y. et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 304, 554 (2004).

    CAS  PubMed  Google Scholar 

  43. Demichelis, F. et al. SNP panel identification assay (SPIA): a genetic-based assay for the identification of cell lines. Nucleic Acids Res. 36, 2446–2456 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. DePristo, M.A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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 

  47. Reich, M. et al. GenePattern 2.0. Nat. Genet. 38, 500–501 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Chiang, D.Y. et al. High-resolution mapping of copy-number alterations with massively parallel sequencing. Nat. Methods 6, 99–103 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. 1000 Genomes Project Consortium. A map of human genome variation from population-scale sequencing. Nature 467, 1061–1073 (2010).

Download references

Acknowledgements

The authors would like to thank M. Lawrence, N. Stransky, M. Imielinski, K. Cibulskis, S. Carter, C. Stewart, C.-Z. Zhang, Y. Drier, S. Schumacher and B. Tabak for their assistance with genomic analyses; P. Wen, O. Al-Mefty, T. Batchelor, B. Reisler, M. Johnson, W. Richards and J. Kim for their assistance with tissue acquisition; and A. Pisarek-Horowitz, S.A. Horowitz, G. Bergthold and C.H. Brastianos for critical review of the manuscript. This work was supported by the Brain Science Foundation (I.F.D., P.K.B., R.B. and S.S.), the Pediatric Low-Grade Astrocytoma Foundation (R.B. and W.C.H.), the American Brain Tumor Association (P.K.B.), a Conquer Cancer Foundation Young Investigator Award (P.K.B.), US National Institutes of Health (NIH) grants K08 CA122833 (R.B.), K08 NS064168 (S.S.) and K12 CA090354-11 (P.K.B.), and US Department of Defense grant W81XWH-12-1-0136 (P.M.H.).

Author information

Authors and Affiliations

Authors

Contributions

P.K.B., P.M.H., I.F.D., R.B. and W.C.H. conceived the study, designed the experiments, analyzed the data and wrote the manuscript. P.M.H. performed the bioinformatics analyses, and A.M., G.G., R.B. and M.D.D. provided analytical advice. S.S., A.O.S.-R. and D.N.L. reviewed the histopathology, and S.S. performed the immunohistochemistry. K.L.L. managed the tissue repository. R.T.J., E.P., P.V.H., A.R., A.S. and L.E.M. provided technical support and performed sequencing. W.C.H., I.F.D. and R.B. supervised the study. All authors discussed the results and implications and commented on the manuscript.

Corresponding authors

Correspondence to William C Hahn, Ian F Dunn or Rameen Beroukhim.

Ethics declarations

Competing interests

W.C.H. is a consultant for Blueprint Medicines and Thermo Fisher. R.B. is a shareholder for AstraZeneca. W.C.H. and R.B. are consultants for and receive research support from Novartis.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Tables 1 and 6 and Supplementary Figures 1–5 (PDF 6192 kb)

Supplementary Table 2

Genes sequenced in the Validation set (separate excel file) (XLSX 31 kb)

Supplementary Table 3

Rearrangements in WGS samples (separate excel file) (XLSX 25 kb)

Supplementary Table 4

All mutated genes and significance (separate excel file) (XLSX 29 kb)

Supplementary Table 5

Samples and their individual mutations (separate excel file) (XLSX 16 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Brastianos, P., Horowitz, P., Santagata, S. et al. Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations. Nat Genet 45, 285–289 (2013). https://doi.org/10.1038/ng.2526

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2526

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