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:

Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma

Abstract

Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease.

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: Genomic landscape of pediatric midline HGAs.
Figure 2: Increased levels of phosphorylated SMAD1/5/8 in ACVR1-mutant mHGAs.
Figure 4: Mutations identified in ACVR1 are associated with activation of downstream SMAD signaling pathways.
Figure 3: Clustering analysis of global DNA methylation profiles for 98 high-grade astrocytomas.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Khuong-Quang, D.A. et al. K27M mutation in histone H3.3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta Neuropathol. 124, 439–447 (2012).

    Article  CAS  Google Scholar 

  2. Schwartzentruber, J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).

    Article  CAS  Google Scholar 

  3. Wu, G. et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 44, 251–253 (2012).

    Article  CAS  Google Scholar 

  4. Sturm, D. et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 22, 425–437 (2012).

    Article  CAS  Google Scholar 

  5. Fontebasso, A.M. et al. Mutations in SETD2 and genes affecting histone H3K36 methylation target hemispheric high-grade gliomas. Acta Neuropathol. 125, 659–669 (2013).

    Article  CAS  Google Scholar 

  6. Paugh, B.S. et al. Genome-wide analyses identify recurrent amplifications of receptor tyrosine kinases and cell-cycle regulatory genes in diffuse intrinsic pontine glioma. J. Clin. Oncol. 29, 3999–4006 (2011).

    Article  CAS  Google Scholar 

  7. Zarghooni, M. et al. Whole-genome profiling of pediatric diffuse intrinsic pontine gliomas highlights platelet-derived growth factor receptor α and poly (ADP-ribose) polymerase as potential therapeutic targets. J. Clin. Oncol. 28, 1337–1344 (2010).

    Article  CAS  Google Scholar 

  8. Jones, D.T. et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat. Genet. 45, 927–932 (2013).

    Article  CAS  Google Scholar 

  9. Shore, E.M. et al. A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat. Genet. 38, 525–527 (2006).

    Article  CAS  Google Scholar 

  10. Fukuda, T. et al. Generation of a mouse with conditionally activated signaling through the BMP receptor, ALK2. Genesis 44, 159–167 (2006).

    Article  CAS  Google Scholar 

  11. Chaikuad, A. et al. Structure of the bone morphogenetic protein receptor ALK2 and implications for fibrodysplasia ossificans progressiva. J. Biol. Chem. 287, 36990–36998 (2012).

    Article  CAS  Google Scholar 

  12. Fukuda, T. et al. A unique mutation of ALK2, G356D, found in a patient with fibrodysplasia ossificans progressiva is a moderately activated BMP type I receptor. Biochem. Biophys. Res. Commun. 377, 905–909 (2008).

    Article  CAS  Google Scholar 

  13. Mishina, Y., Crombie, R., Bradley, A. & Behringer, R.R. Multiple roles for activin-like kinase-2 signaling during mouse embryogenesis. Dev. Biol. 213, 314–326 (1999).

    Article  CAS  Google Scholar 

  14. Kishigami, S. et al. BMP signaling through ACVRI is required for left-right patterning in the early mouse embryo. Dev. Biol. 276, 185–193 (2004).

    Article  CAS  Google Scholar 

  15. Miyazono, K., Maeda, S. & Imamura, T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev. 16, 251–263 (2005).

    Article  CAS  Google Scholar 

  16. Korchynskyi, O. & ten Dijke, P. Identification and functional characterization of distinct critically important bone morphogenetic protein–specific response elements in the Id1 promoter. J. Biol. Chem. 277, 4883–4891 (2002).

    Article  CAS  Google Scholar 

  17. Singh, D. et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science 337, 1231–1235 (2012).

    Article  CAS  Google Scholar 

  18. Zhang, J. et al. Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas. Nat. Genet. 45, 602–612 (2013).

    Article  CAS  Google Scholar 

  19. Paugh, B.S. et al. Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease. J. Clin. Oncol. 28, 3061–3068 (2010).

    Article  Google Scholar 

  20. Lewis, P.W. et al. Inhibition of PRC2 activity by a gain-of-function H3 H3 mutation found in pediatric glioblastoma. Science 340, 857–861 (2013).

    Article  CAS  Google Scholar 

  21. Shen, Q. et al. The fibrodysplasia ossificans progressiva R206H ACVR1 mutation activates BMP-independent chondrogenesis and zebrafish embryo ventralization. J. Clin. Invest. 119, 3462–3472 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lahti, L., Peltopuro, P., Piepponen, T.P. & Partanen, J. Cell-autonomous FGF signaling regulates anteroposterior patterning and neuronal differentiation in the mesodiencephalic dopaminergic progenitor domain. Development 139, 894–905 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  24. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  Google Scholar 

  25. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  Google Scholar 

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

    Article  Google Scholar 

  27. McPherson, A. et al. deFuse: an algorithm for gene fusion discovery in tumor RNA-Seq data. PLoS Comput. Biol. 7, e1001138 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Maksimovic, J., Gordon, L. & Oshlack, A. SWAN: subset-quantile within array normalization for Illumina Infinium HumanMethylation450 BeadChips. Genome Biol. 13, R44 (2012).

    Article  Google Scholar 

  30. Suzuki, R. & Shimodaira, H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics 22, 1540–1542 (2006).

    Article  CAS  Google Scholar 

  31. Monje, M. et al. Hedgehog-responsive candidate cell of origin for diffuse intrinsic pontine glioma. Proc. Natl. Acad. Sci. USA 108, 4453–4458 (2011).

    Article  CAS  Google Scholar 

  32. Siegel, P.M., Ryan, E.D., Cardiff, R.D. & Muller, W.J. Elevated expression of activated forms of Neu/ErbB-2 and ErbB-3 are involved in the induction of mammary tumors in transgenic mice: implications for human breast cancer. EMBO J. 18, 2149–2164 (1999).

    Article  CAS  Google Scholar 

  33. Schmierer, B. & Hill, C.S. TGFβ-SMAD signal transduction: molecular specificity and functional flexibility. Nat. Rev. Mol. Cell Biol. 8, 970–982 (2007).

    Article  CAS  Google Scholar 

  34. Kleinman, C.L. et al. Fusion of TTYH1 with the C19MC microRNA cluster drives expression of a brain-specific DNMT3B isoform in the embryonal brain tumor ETMR. Nat. Genet. 46, 39–44 (2014).

    Article  CAS  Google Scholar 

  35. Ramkissoon, L.A. et al. Genomic analysis of diffuse pediatric low-grade gliomas identifies recurrent oncogenic truncating rearrangements in the transcription factor MYBL1. Proc. Natl. Acad. Sci. USA 110, 8188–8193 (2013).

    Article  CAS  Google Scholar 

  36. Firestein, R. et al. CDK8 is a colorectal cancer oncogene that regulates β-catenin activity. Nature 455, 547–551 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to express their sincere gratitude toward all staff at the McGill University and Génome Québec Innovation Centre for excellent technical expertise, library preparation and sequencing. The authors are very grateful to J.-J. Lebrun (McGill University) for primer sequences and materials for SMAD signaling studies. This work was performed within the context of the I-CHANGE (International Childhood Astrocytoma Integrated Genomics and Epigenomics) Consortium and was supported by funding from Genome Canada, Génome Québec, the Institute for Cancer Research of the Canadian Institutes for Health Research (CIHR), McGill University and the Montreal Children's Hospital Foundation. This work was also supported by Hungarian Scientific Research Fund (OTKA) contract T-04639, National Research and Development Fund (NKFP) contract 1A/002/2004 (P.H. and M.G.) and TÁMOP-4.2.2A-11/1/KONV-2012-0025 (A.K. and L.B.). N.J. is a member of the Penny Cole laboratory and the recipient of a Chercheur Clinicien Senior Award. J. Majewski holds a Canada Research Chair (tier 2). L.G., K.L.L. and M.W.K. are supported by NCI P01CA142536. We acknowledge the support of the Zach Carson DIPG Fund at the Dana-Farber Cancer Institute (DFCI), the Ellie Kavalieros Fund (DFCI), the Mikey Czech Foundation, the Prayer From Maria Foundation, the Hope for Caroline Fund (DFCI), the Ryan Harvey DIPG Fund (DFCI), the Stop&Shop Pediatric Brain Tumor Program (DFCI) and the Pediatric Brain Tumor Clinical and Research Fund (DFCI). A.M.F. is supported by a studentship from CIHR, as well as by an award from the CIHR Systems Biology Training Program at McGill University. D.B. is supported by a studentship from the T.D. Trust/Montreal Children's Hospital Foundation, and N. Gerges is supported by a studentship from the Cedars Cancer Institute. N.D.J. is supported by an award from the McGill Integrated Cancer Research Training Program.

Author information

Authors and Affiliations

Authors

Contributions

A.M.F., N. Gerges, P.-O.F., D.B., D.F., L.A.R., A.C., A.H.L., S.A. and Z.D. performed experiments. A.M.F., S.P.-C., J.S., H.N., N.D.J., A.H.L., S.A., Z.D. and P.M.S. analyzed the data and produced figures and tables. D.T.W.J., D.S., P.J., T.T., S.G., M.N., A.B., L.G., D.C.B., J.R.L., J.B.R., T.A., S.B., J.R.G., G.J., K.C., N. Gupta, M.D.P., A.-S.C., B.E., L.C., A.K., L.B., P.H., M.G., J. Myseros, H.M., S.A. and S.M.P. provided tissue samples. K.L.L., J. Majewski, N.J. and M.W.K. provided project leadership and designed the study. All authors contributed to the final manuscript.

Corresponding authors

Correspondence to Keith L Ligon, Jacek Majewski, Nada Jabado or Mark W Kieran.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Patterns of K27M-associated mutations among midline pediatric high-grade astrocytomas.

a, Neuroanatomical distribution of mutations and alterations of interest in pediatric midline high-grade astrocytomas. b, The age of patients harboring H3.3 and H3.1 mutations is significantly different (P = 0.0280; two-tailed t test). c, Mutual exclusivity and associative statistical analyses (represented as P values) of mutations in H3F3A, HIST1H3B/HIST1H3C, TP53, FGFR1, ACVR1, PDGFRA and PI3K pathway genes PIK3CA/PIK3R1/PTEN demonstrates predilection for co-occurrence of and exclusion of particular mutations as unique mutational groups within 39 midline high-grade astrocytomas profiled by whole-exome sequencing. P values were calculated utilizing Fisher's exact test (two-sided) with highlighted values indicating statistically significant (P < 0.05) preferences for co-occurrence or mutual exclusivity.

Supplementary Figure 2 PDGFRA amplification is present in a minority of cells in DIPG.

Representative images from FISH assay for PDGFRA amplification reveal substantial increase in the number of PDFRA-positive nuclei following treatment compared with samples from pretreatment biopsies. Green, PDGFRA; red, Chr4/CEN4.

Supplementary Figure 3 Copy number variant analysis using biopsy material from five different anatomical loci in a single tumor.

a, Tumor biopsies (n = 5) show highly similar CNV patterns across the genome. b, Plots depicting 20-Mb zoom-in of chromosome 4, showing significant PDGFRA, KIT and KDR amplification in only the deep tissue biopsy and not others.

Supplementary Figure 4 H3.1 and H3.3 K27M variants demonstrate similar patterns of global DNA methylation.

Heat map of hierarchical clustering analysis of DNA methylation array data from the top 10,000 most variable β values from 27 K27M-mutant high-grade gliomas demonstrates that H3.3 and H3.1 K27M mutant tumors show similar patterns of global epigenomic dysregulation.

Supplementary Figure 5 Multiscale bootstrapping of the high-grade astrocytoma methylation cluster.

Dendrogram of multiscale bootstrapping of DNA methylation profiles of patient high-grade tumors. P values, represented as the red values at the internal nodes, are the approximately unbiased (AU) P value computed by the R package pvclust.

Supplementary Figure 6 Mutation subgroup-specific overall survival of midline high-grade astrocytomas (mHGAs) and diffuse intrinsic pontine gliomas (DIPGs).

Kaplan-Meier analysis of overall survival (months) of mHGAs with available outcome data, with indicated P values from Mantel-Cox (log-rank) and Gehan-Breslow-Wilcoxon testing for comparison based on a, ACVR1 mutation status (n = 27), b, H3.3/H3.1 K27M mutation status (n = 27) and overall survival restricted to tumors of the pontine area based on c, ACVR1 mutation status (n = 23), d, H3.3/H3.1 K27M mutation status (n = 22).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 2686 kb)

Supplementary Tables 1–8

Supplementary Tables 1–8 (XLSX 130 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fontebasso, A., Papillon-Cavanagh, S., Schwartzentruber, J. et al. Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma. Nat Genet 46, 462–466 (2014). https://doi.org/10.1038/ng.2950

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2950

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