Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations

Pawel Buczkowicz, Christine Hoeman, Patricia Rakopoulos, Sanja Pajovic, Louis Letourneau, Misko Dzamba, Andrew Morrison, Peter Lewis, Eric Bouffet, Ute Bartels, Jennifer Zuccaro, Sameer Agnihotri, Scott Ryall, Mark Barszczyk, Yevgen Chornenkyy, Mathieu Bourgey, Guillaume Bourque, Alexandre Montpetit, Francisco Cordero, Pedro Castelo-Branco, Joshua Mangerel, Uri Tabori, King Ching Ho, Annie Huang, Kathryn R Taylor, Alan Mackay, Anne E Bendel, Javad Nazarian, Jason R Fangusaro, Matthias A Karajannis, David Zagzag, Nicholas K Foreman, Andrew Donson, Julia V Hegert, Amy Smith, Jennifer Chan, Lucy Lafay-Cousin, Sandra Dunn, Juliette Hukin, Chris Dunham, Katrin Scheinemann, Jean Michaud, Shayna Zelcer, David Ramsay, Jason Cain, Cameron Brennan, Mark M Souweidane, Chris Jones, C David Allis, Michael Brudno, Oren Becher, Cynthia Hawkins, Pawel Buczkowicz, Christine Hoeman, Patricia Rakopoulos, Sanja Pajovic, Louis Letourneau, Misko Dzamba, Andrew Morrison, Peter Lewis, Eric Bouffet, Ute Bartels, Jennifer Zuccaro, Sameer Agnihotri, Scott Ryall, Mark Barszczyk, Yevgen Chornenkyy, Mathieu Bourgey, Guillaume Bourque, Alexandre Montpetit, Francisco Cordero, Pedro Castelo-Branco, Joshua Mangerel, Uri Tabori, King Ching Ho, Annie Huang, Kathryn R Taylor, Alan Mackay, Anne E Bendel, Javad Nazarian, Jason R Fangusaro, Matthias A Karajannis, David Zagzag, Nicholas K Foreman, Andrew Donson, Julia V Hegert, Amy Smith, Jennifer Chan, Lucy Lafay-Cousin, Sandra Dunn, Juliette Hukin, Chris Dunham, Katrin Scheinemann, Jean Michaud, Shayna Zelcer, David Ramsay, Jason Cain, Cameron Brennan, Mark M Souweidane, Chris Jones, C David Allis, Michael Brudno, Oren Becher, Cynthia Hawkins

Abstract

Diffuse intrinsic pontine glioma (DIPG) is a fatal brain cancer that arises in the brainstem of children, with no effective treatment and near 100% fatality. The failure of most therapies can be attributed to the delicate location of these tumors and to the selection of therapies on the basis of assumptions that DIPGs are molecularly similar to adult disease. Recent studies have unraveled the unique genetic makeup of this brain cancer, with nearly 80% found to harbor a p.Lys27Met histone H3.3 or p.Lys27Met histone H3.1 alteration. However, DIPGs are still thought of as one disease, with limited understanding of the genetic drivers of these tumors. To understand what drives DIPGs, we integrated whole-genome sequencing with methylation, expression and copy number profiling, discovering that DIPGs comprise three molecularly distinct subgroups (H3-K27M, silent and MYCN) and uncovering a new recurrent activating mutation affecting the activin receptor gene ACVR1 in 20% of DIPGs. Mutations in ACVR1 were constitutively activating, leading to SMAD phosphorylation and increased expression of the downstream activin signaling targets ID1 and ID2. Our results highlight distinct molecular subgroups and novel therapeutic targets for this incurable pediatric cancer.

Figures

Figure 1. Methylation profiling reveals three molecular…
Figure 1. Methylation profiling reveals three molecular subgroups of DIPG
(a) Heat map of methylation levels in three DIPG subgroups identified by unsupervised hierarchical clustering and supported by (b) principal components analysis, (c), non-negative matrix factorization (cophenetic coefficient = 0.9934, k=3) and (d) consensus clustering represented by cumulative distribution function and change in Gini.
Figure 2. Molecular subgroups of DIPG share…
Figure 2. Molecular subgroups of DIPG share common clinical features and recurrent genomic events
(a) Clinical and genomic features such as gender, histology, frequency of recurrent mutations, alternative lengthening of telomeres and copy number alterations are represented in a DIPG subgroup specific manner. (b) Probability of two mutational or structural features of DIPG co-occurring based on odds ratio suggests statistically significant association between K27M-H3.3 and PDGFRA amplifications (OR = 8.0, p = 0.0127) and between K27M-H3.1 and ACVR1 mutations (OR = 15.8, p < 0.001). (C) Probability of mutations or structural event of DIPG occurring with a clinical feature such as gender or tumor histology based on odds ratio shows statistically significant correlation between P53 mutations and GBM histology (OR = 10.8, p < 0.005), among others.
Figure 3. ACVR1 mutations constitutively activate BMP…
Figure 3. ACVR1 mutations constitutively activate BMP signaling in vitro and in ACVR1 mutant DIPG
(a) Four mutations (R206H, Q207E, G328E and G328V) were detected in 12/61 DIPG patients. The R206H and Q207E mutations occur in the GS domain and the G328-mutations occur in the protein kinase domain. (b) Human DIPG with ACVR1 mutations have increased pSMAD1/5 expression compared with ACVR1 wild-type DIPG. (c) Western blot showing increased pSMAD1/5 in ACVR1 mutant NHA and DIPG58 cells transfected with G328V-ACVR1 as compared to control cells. (d) Real-time PCR in NHA transfected with empty vector, K27M-H3.3, G328V-ACVR1 or a combination of K27M-H3.3 and G328V-ACVR1 shows increase in ID1 and ID2 gene expression as compared to empty vector control. Error bars represent standard deviation. (e) Mutant G328V-ACVR1 expressing NHA cells have an increased growth rate as compared to empty vector controls (p = 0.0034). (f) Compared to WT-ACVR1 murine brainstem progenitor cultures, mutant ACVR1 has significantly higher BrdU incorporation suggesting increased proliferation (* p<0.05). Error bars represent standard error of the mean.

References

    1. Stiller CA. Population based survival rates for childhood cancer in Britain, 1980–91. BMJ. 1994;309:1612–6.
    1. Freeman CR, Perilongo G. Chemotherapy for brain stem gliomas. Childs Nerv Syst. 1999;15:545–53.
    1. Maria BL, et al. Brainstem glioma: I. Pathology, clinical features, and therapy. J Child Neurol. 1993;8:112–28.
    1. Hargrave D, Bartels U, Bouffet E. Diffuse brainstem glioma in children: critical review of clinical trials. Lancet Oncol. 2006;7:241–8.
    1. Donaldson SS, Laningham F, Fisher PG. Advances toward an understanding of brainstem gliomas. J Clin Oncol. 2006;24:1266–72.
    1. Buczkowicz P, et al. Aurora Kinase B Is a Potential Therapeutic Target in Pediatric Diffuse Intrinsic Pontine Glioma. Brain Pathol. 2012
    1. Paugh BS, 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. 2011;29:3999–4006.
    1. Paugh BS, et al. Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease. J Clin Oncol. 2010;28:3061–8.
    1. Zarghooni M, et al. Whole-genome profiling of pediatric diffuse intrinsic pontine gliomas highlights platelet-derived growth factor receptor alpha and poly (ADP-ribose) polymerase as potential therapeutic targets. J Clin Oncol. 2010;28:1337–44.
    1. Khuong-Quang DA, et al. K27M mutation in histone H3.3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta Neuropathol. 2012;124:439–47.
    1. Schwartzentruber J, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–31.
    1. Wu G, et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet. 2012;44:251–3.
    1. Herbert BS, Shay JW, Wright WE. Analysis of telomeres and telomerase. Curr Protoc Cell Biol. 2003;Chapter 18(Unit 18):6.
    1. Henson JD, et al. DNA C-circles are specific and quantifiable markers of alternative-lengthening-of-telomeres activity. Nat Biotechnol. 2009;27:1181–5.
    1. Kowanetz M, Valcourt U, Bergstrom R, Heldin CH, Moustakas A. Id2 and Id3 define the potency of cell proliferation and differentiation responses to transforming growth factor beta and bone morphogenetic protein. Mol Cell Biol. 2004;24:4241–54.
    1. Kurooka H, Nakahiro T, Mori K, Sano K, Yokota Y. BMP signaling is responsible for serum-induced Id2 expression. Biochem Biophys Res Commun. 2012;420:281–7.
    1. Shin M, et al. Identification of a novel bone morphogenetic protein (BMP)-inducible transcript, BMP-inducible transcript-1, by utilizing the conserved BMP-responsive elements in the Id genes. J Bone Miner Metab. 2013;31:34–43.
    1. Song GA, et al. Molecular consequences of the ACVR1(R206H) mutation of fibrodysplasia ossificans progressiva. J Biol Chem. 2010;285:22542–53.
    1. Kaplan FS, et al. Classic and atypical fibrodysplasia ossificans progressiva (FOP) phenotypes are caused by mutations in the bone morphogenetic protein (BMP) type I receptor ACVR1. Hum Mutat. 2009;30:379–90.
    1. Petrie KA, et al. Novel mutations in ACVR1 result in atypical features in two fibrodysplasia ossificans progressiva patients. PLoS One. 2009;4:e5005.
    1. Chen XS, Zhang YH, Cai QY, Yao ZX. ID2: A negative transcription factor regulating oligodendroglia differentiation. J Neurosci Res. 2012;90:925–32.
    1. Liu Z, et al. Overexpressed DNA-binding protein inhibitor 2 as an unfavorable prognosis factor promotes cell proliferation in nasopharyngeal carcinoma. Acta Biochim Biophys Sin (Shanghai) 2012;44:503–12.
    1. Rollin J, et al. The intracellular localization of ID2 expression has a predictive value in non small cell lung cancer. PLoS One. 2009;4:e4158.
    1. Wazir U, Jiang WG, Sharma AK, Newbold RF, Mokbel K. The mRNA expression of inhibitors of DNA binding-1 and -2 is associated with advanced tumour stage and adverse clinical outcome in human breast cancer. Anticancer Res. 2013;33:2179–83.
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26:589–95.
    1. Jiang Y, Wang Y, Brudno M. PRISM: pair-read informed split-read mapping for base-pair level detection of insertion, deletion and structural variants. Bioinformatics. 2012;28:2576–83.
    1. Mills RE, et al. Natural genetic variation caused by small insertions and deletions in the human genome. Genome Res. 2011;21:830–9.
    1. Fiume M, et al. Savant Genome Browser 2: visualization and analysis for population-scale genomics. Nucleic Acids Res. 2012;40:W615–21.
    1. Medvedev P, Fiume M, Dzamba M, Smith T, Brudno M. Detecting copy number variation with mated short reads. Genome Res. 2010;20:1613–22.
    1. Sasai K, et al. O6-methylguanine-DNA methyltransferase is downregulated in transformed astrocyte cells: implications for anti-glioma therapies. Mol Cancer. 2007;6:36.
    1. Bland JM, Altman DG. Statistics notes. The odds ratio. BMJ. 2000;320:1468.

Source: PubMed

3
Abonnere