Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma

Hamid Nikbakht, Eshini Panditharatna, Leonie G Mikael, Rui Li, Tenzin Gayden, Matthew Osmond, Cheng-Ying Ho, Madhuri Kambhampati, Eugene I Hwang, Damien Faury, Alan Siu, Simon Papillon-Cavanagh, Denise Bechet, Keith L Ligon, Benjamin Ellezam, Wendy J Ingram, Caedyn Stinson, Andrew S Moore, Katherine E Warren, Jason Karamchandani, Roger J Packer, Nada Jabado, Jacek Majewski, Javad Nazarian, Hamid Nikbakht, Eshini Panditharatna, Leonie G Mikael, Rui Li, Tenzin Gayden, Matthew Osmond, Cheng-Ying Ho, Madhuri Kambhampati, Eugene I Hwang, Damien Faury, Alan Siu, Simon Papillon-Cavanagh, Denise Bechet, Keith L Ligon, Benjamin Ellezam, Wendy J Ingram, Caedyn Stinson, Andrew S Moore, Katherine E Warren, Jason Karamchandani, Roger J Packer, Nada Jabado, Jacek Majewski, Javad Nazarian

Abstract

Diffuse Intrinsic Pontine Gliomas (DIPGs) are deadly paediatric brain tumours where needle biopsies help guide diagnosis and targeted therapies. To address spatial heterogeneity, here we analyse 134 specimens from various neuroanatomical structures of whole autopsy brains from nine DIPG patients. Evolutionary reconstruction indicates histone 3 (H3) K27M--including H3.2K27M--mutations potentially arise first and are invariably associated with specific, high-fidelity obligate partners throughout the tumour and its spread, from diagnosis to end-stage disease, suggesting mutual need for tumorigenesis. These H3K27M ubiquitously-associated mutations involve alterations in TP53 cell-cycle (TP53/PPM1D) or specific growth factor pathways (ACVR1/PIK3R1). Later oncogenic alterations arise in sub-clones and often affect the PI3K pathway. Our findings are consistent with early tumour spread outside the brainstem including the cerebrum. The spatial and temporal homogeneity of main driver mutations in DIPG implies they will be captured by limited biopsies and emphasizes the need to develop therapies specifically targeting obligate oncohistone partnerships.

Figures

Figure 1. Oncogenic alterations in 41 sub-regions…
Figure 1. Oncogenic alterations in 41 sub-regions from nine DIPG patients from whole exome sequencing data.
Samples representing different anatomical locations within each patient are represented in columns. The mutations (in rows) were selected based on published datasets in paediatric glioblastoma and specifically DIPG. Mutations were divided into two subgroups; driver mutations which are essential for tumour initiation/maintenance and accessory driver mutations, which can further promote and accelerate tumour growth, but are not absolutely essential for tumour initiation or maintenance.
Figure 2. Selected examples of clonal evolution…
Figure 2. Selected examples of clonal evolution within DIPG tumours.
Left: histograms show the raw allele frequencies (whole exome sequencing data) for each somatic mutation in different autopsy regions within each tumour. Red: ubiquitous mutations across regions; yellow: mutations shared in at least two regions; blue: mutations seen in only one region. Right: phylogenetic trees constructed from the mutation allele frequencies of deep amplicon sequencing data showing the order of evolution along with support probabilities (upper portions of graphs) and clonal mixing proportions within samples (lower portions). For clarity, only mutations selected to be likely oncogenic are shown. (a) DIPG5: a rare case harboring both TP53 and PPM1D mutations, which are generally found to be mutually exclusive. PPM1D and TP53 mutations occur in distinct clones and are both secondary to H3K27M. ATRX is also secondary and subclonal. (b) DIPG6: while it is impossible to resolve the order of H3/TP53/ATRX mutations' appearance, PIK3CA is clearly sub-clonal and appears in the later stages of evolution within this tumour. (c) DIPG2: the H3.1 K27M and ACVR1 main driver mutations are ubiquitous, occur at similar frequencies across all samples, and their mutation order cannot be resolved. Conversely, other accessory driver mutations are clearly secondary in order of appearance, and are present only in distinct subclones.
Figure 3. Tumour spread in DIPG.
Figure 3. Tumour spread in DIPG.
(a) Tumour spread in DIPG2 in the thalamus, cerebellum and brainstem. Tumour extension in the thalamus harbors secondary mutation PIK3CA, MAX and PTEN, which indicates late spread from both Pons 1 and Pons 2. Extension towards the cerebellum is relatively early in the tumour evolution as it lacks secondary mutations found in the primary tumour and other brainstem spread. (b) Evolution of tumour in patient DIPG3. Autopsy revealed two morphologically and histologically distinct regions of the tumour, indicated as DIPG3 Pons 1 (low-grade) and DIPG3 Pons 2 (high-grade). Exome sequencing identified 11 SNVs and several large scale CNAs common to both regions. Shared alterations included H3.2 K27M and ACVR1 G328V mutations that are likely the main driver mutations in this patient. The analysis also indicated a clear clonal substructure of the two regions, with 18 SNVs and 1 CNA found only in DIPG3 Pons 1, and 11 SNVs and 3 CNAs unique to DIPG3 Pons 2. Intriguingly, DIPG3 Pons 2 carries the activating PIK3CA H1047R mutation, which occurs early in the evolution of this sub-clone judging by its high allelic frequency. PIK3CA H1047R is associated with multi-potency and PI3K activation with angiogenesis and growth and this mutation likely contributes to tumour aggressiveness and high-grade features of Pons 2 compared with Pons 1 in DIPG3. Scale bar=500 μm.

References

    1. Buczkowicz P., Bartels U., Bouffet E., Becher O. & Hawkins C. Histopathological spectrum of paediatric diffuse intrinsic pontine glioma: diagnostic and therapeutic implications. Acta. Neuropathol. 128, 573–581 (2014).
    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).
    1. Schwartzentruber J. et al.. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).
    1. Wu G. et al.. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 44, 251–253 (2012).
    1. Buczkowicz P. et al.. Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat. Genet. 46, 451–456 (2014).
    1. Fontebasso A. M. et al.. Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma. Nat. Genet. 46, 462–466 (2014).
    1. Taylor K. R. et al.. Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma. Nat. Genet. 46, 457–461 (2014).
    1. 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).
    1. Kumar A. et al.. Deep sequencing of multiple regions of glial tumors reveals spatial heterogeneity for mutations in clinically relevant genes. Genome Biol. 15, 530 (2014).
    1. Sottoriva A. et al.. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl Acad. Sci. USA 110, 4009–4014 (2013).
    1. Szerlip N. J. et al.. Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc. Natl Acad. Sci. USA 109, 3041–3046 (2012).
    1. Bechet D. et al.. Specific detection of methionine 27 mutation in histone 3 variants (H3K27M) in fixed tissue from high-grade astrocytomas. Acta Neuropathol. 128, 733–741 (2014).
    1. Caretti V. et al.. Subventricular spread of diffuse intrinsic pontine glioma. Acta Neuropathol. 128, 605–607 (2014).
    1. Gerlinger M. et al.. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).
    1. Eleveld T. F. et al.. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat. Genet. 47, 864–871 (2015).
    1. Wu X. et al.. Clonal selection drives genetic divergence of metastatic medulloblastoma. Nature 482, 529–533 (2012).
    1. Johnson B. E. et al.. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343, 189–193 (2014).
    1. Walker D. A. et al.. A multi-disciplinary consensus statement concerning surgical approaches to low-grade, high-grade astrocytomas and diffuse intrinsic pontine gliomas in childhood (CPN Paris 2011) using the Delphi method. Neuro. Oncol. 15, 462–468 (2013).
    1. Kambhampati M. et al.. A standardized autopsy procurement allows for the comprehensive study of DIPG biology. Oncotarget 6, 12740–12747 (2015).
    1. Castel D. et al.. Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes. Acta Neuropathol. 130, 815–827 (2015).
    1. Nik-Zainal S. et al.. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).
    1. Popic V. et al.. Fast and scalable inference of multi-sample cancer lineages. Genome. Biol. 16, 91 (2015).
    1. Zhang L. et al.. Exome sequencing identifies somatic gain-of-function PPM1D mutations in brainstem gliomas. Nat. Genet. 46, 726–730 (2014).
    1. Koren S. et al.. PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature 525, 114–118 (2015).
    1. Karar J. & Maity A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 4, 51 (2011).
    1. Lewis P. W. et al.. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340, 857–861 (2013).
    1. Funato K., Major T., Lewis P. W., Allis C. D. & Tabar V. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science 346, 1529–1533 (2014).
    1. Hindson C. M. et al.. Absolute quantification by droplet digital PCR versus analog real-time PCR. Nat. Methods 10, 1003–1005 (2013).
    1. McKenna A. et al.. The genome analysis toolkit: a mapreduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).
    1. Li H. & Durbin R. Fast and accurate short read alignment with burrows-wheeler transform. Bioinformatics 25, 1754–1760 (2009).
    1. Li H. et al.. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).
    1. Wang K., Li M. & Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).
    1. Shi Y. & Majewski J. FishingCNV: a graphical software package for detecting rare copy number variations in exome-sequencing data. Bioinformatics 29, 1461–1462 (2013).
    1. Nadaf J., Majewski J. & Fahiminiya S. ExomeAI: detection of recurrent allelic imbalance in tumors using whole-exome sequencing data. Bioinformatics 31, 429–431 (2015).
    1. Deshwar A. G. et al.. PhyloWGS: reconstructing subclonal composition and evolution from whole-genome sequencing of tumors. Genome Biol. 16, 35 (2015).
    1. Love M. I., Huber W. & Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome. Biol. 15, 550 (2014).
    1. Dobin A. et al.. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
    1. Carbon S. et al.. AmiGO: online access to ontology and annotation data. Bioinformatics 25, 288–289 (2009).
    1. Huang, da W., Sherman B. T. & Lempicki R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

Source: PubMed

3
Sottoscrivi