The Genetic Signatures of Pediatric High-Grade Glioma: No Longer a One-Act Play,☆☆

https://doi.org/10.1016/j.semradonc.2014.06.003Get rights and content

Advances in understanding pediatric high-grade glioma (pHGG) genetics have revealed key differences between pHGG and adult HGG and have uncovered unique molecular drivers among subgroups within pHGG. The 3 core adult HGG pathways, the receptor tyrosine kinase-Ras-phosphatidylinositide 3-kinase, p53, and retinoblastoma networks, are also disrupted in pHGG, but they exhibit a different spectrum of effectors targeted by mutation. There are also similarities and differences in the genomic landscape of diffuse intrinsic pontine glioma (DIPG) and pediatric nonbrainstem (pNBS)-HGG. In 2012, histone H3 mutations were identified in nearly 80% of DIPGs and ~35% of pNBS-HGG. These were the first reports of histone mutations in human cancer, implicating novel biology in pediatric gliomagenesis. Additionally, DIPG and midline pNBS-HGG vary in the frequency and specific histone H3 amino acid substitution compared with pNBS-HGGs arising in the cerebral hemispheres, demonstrating a molecular difference among pHGG subgroups. The gene expression signatures as well as DNA methylation signatures of these tumors are also distinctive, reflecting a combination of the driving mutations and the developmental context from which they arise. These data collectively highlight unique selective pressures within the developing brainstem and solidify DIPG as a specific molecular and biological entity among pHGGs. Emerging studies continue to identify novel mutations that distinguish subgroups of pHGG. The molecular heterogeneity among pHGGs will undoubtedly have clinical implications moving forward. The discovery of unique oncogenic drivers is a critical first step in providing patients with appropriate, targeted therapies. Despite these insights, our vantage point has been largely limited to an in-depth analysis of protein coding sequences. Given the clear importance of histone mutations in pHGG, it will be interesting to see how aberrant epigenetic regulation contributes to tumorigenesis in the pediatric context. New mechanistic insights may allow for the identification of distinct vulnerabilities in this devastating spectrum of childhood tumors.

Section snippets

Opening Remarks

The past several years mark a period of tremendous growth in our understanding of pediatric high-grade glioma (pHGG). Advances in genome-wide array-based and sequencing technologies, their precipitous drop in cost, and evaluation of increasingly larger cohorts have all contributed to novel insights into the genetics of these devastating cancers, greatly extending earlier studies that evaluated candidate genes based on their involvement in adult HGG (aHGG). Our aim is to provide context to these

The 3 Core aHGG Pathways Show a Different Spectrum of Alteration in pHGG

As the genomic landscape of aHGG came into view, it shaped initial work into the pediatric disease. The first pHGG studies focused primarily on investigating the involvement of high-frequency recurrent events found in adult tumors. For example, epidermal growth factor receptor (EGFR) is the most commonly altered receptor tyrosine kinase (RTK) in aHGG; with the corresponding gene locus undergoing amplification or intragenic deletion or both in ~50% of cases.7, 8, 9 First identified in adult

Copy Number Imbalances and Gene Expression Profiling

Despite some common copy number imbalances such as 13q and 14q loss in approximately one-third of patients with HGG regardless of age or location, aHGG and pHGG also exhibit a unique constellation of gains and losses that distinguish one from the other, and the same can be said for DIPGs and pNBS-HGGs.15, 16, 17, 18, 19, 23, 24, 25, 26 This suggests that unique combinations of genetic drivers underlie adult and pediatric tumorigenesis, and among childhood HGG, DIPG and pNBS-HGG tumorigenesis.

Histones Make Their Mark

The aforementioned work established the concept that oncogenic events driving pediatric HGG were different from those arising in adults. This appreciation, however, was not fully cemented until early 2012, with the discovery of recurrent histone mutations in pHGG (Fig. 2). As the first reports of histone mutations in human cancer, these mutations implicated novel mechanisms in pHGG tumor biology that are not found to play a significant role in the adult disease.

Whole-genome sequencing of 7

Understanding Mutant Histone Gain-of-Function

All histone H3 mutations in pHGG were heterozygous, and in any individual tumor, only 1 of 16 genes encoding histone H3 was mutated. This pattern clearly indicates a dominant gain-of-function effect.

Lysine 27 on histone H3 (H3K27) is a residue that can be acetylated or monomethylated, dimethylated, or trimethylated (H3K27me3). Although mutant histone H3.1/3.3 make up a minority of the total cellular histone H3 pool,64 p.K27M mutations led to loss of total H3K27me2/3 of the entire cellular H3

Additional Genetic Associations With pHGG Subgroups

Genome-wide sequencing approaches revealed that 20%-32% of DIPGs harbored somatic missense mutations in ACVR1, also known as ALK2,35, 39, 40, 41 which encodes a receptor serine-threonine kinase mediating bone morphogenetic protein–induced signal transduction.69 These mutations frequently co-occur with histone H3.1 p.K27M substitutions. Both alterations tend to occur in younger patients with DIPG and were not found in pHGG arising outside the brainstem.35, 39, 40, 41 Thus, these mutations

Closing Remarks

Advances in microarrays and next-generation sequencing technologies have provided unprecedented insight into pHGG biology. However, this insight is only a starting point. Researchers and clinicians must now endeavor to not only understand how this unique mutation spectrum contributes to tumorigenesis but also, more importantly, seek to exploit these genetic defects therapeutically. Recent discoveries should inform the generation of improved preclinical models that more faithfully resemble the

Acknowledgments

We thank Dr Zoltan Patay for the MRI used in Figure 3.

References (76)

  • L. Attisano et al.

    Identification of human activin and TGF beta type I receptors that form heteromeric kinase complexes with type II receptors

    Cell

    (1993)
  • T.A. Dolecek et al.

    CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2005-2009

    Neuro Oncol

    (2012)
  • A. Broniscer et al.

    Supratentorial high-grade astrocytoma and diffuse brainstem glioma: Two challenges for the pediatric oncologist

    Oncologist

    (2004)
  • A. Chassot et al.

    Radiotherapy with concurrent and adjuvant temozolomide in children with newly diagnosed diffuse intrinsic pontine glioma

    J Neurooncol

    (2012)
  • K.J. Cohen et al.

    Temozolomide in the treatment of high-grade gliomas in children: A report from the Children׳s Oncology Group

    Neuro Oncol

    (2011)
  • R. Stupp et al.

    Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma

    N Engl J Med

    (2005)
  • A. Broniscer et al.

    Clinical and molecular characteristics of malignant transformation of low-grade glioma in children

    J Clin Oncol

    (2007)
  • D.W. Parsons et al.

    An integrated genomic analysis of human glioblastoma multiforme

    Science

    (2008)
  • Comprehensive genomic characterization defines human glioblastoma genes and core pathways

    Nature

    (2008)
  • P.A. Humphrey et al.

    Anti-synthetic peptide antibody reacting at the fusion junction of deletion-mutant epidermal growth factor receptors in human glioblastoma

    Proc Natl Acad Sci U S A

    (1990)
  • A.J. Wong et al.

    Structural alterations of the epidermal growth factor receptor gene in human gliomas

    Proc Natl Acad Sci U S A

    (1992)
  • A.J. Ekstrand et al.

    Amplified and rearranged epidermal growth factor receptor genes in human glioblastomas reveal deletions of sequences encoding portions of the N- and/or C-terminal tails

    Proc Natl Acad Sci U S A

    (1992)
  • I.F. Pollack et al.

    Rarity of PTEN deletions and EGFR amplification in malignant gliomas of childhood: Results from the Children׳s Cancer Group 945 cohort

    J Neurosurg

    (2006)
  • T. Sung et al.

    Preferential inactivation of the p53 tumor suppressor pathway and lack of EGFR amplification distinguish de novo high grade pediatric astrocytomas from de novo adult astrocytomas

    Brain Pathol

    (2000)
  • D.A. Bax et al.

    A distinct spectrum of copy number aberrations in pediatric high-grade gliomas

    Clin Cancer Res

    (2010)
  • B.S. Paugh 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)
  • B.S. Paugh et al.

    Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease

    J Clin Oncol

    (2010)
  • H.Q. Qu et al.

    Genome-wide profiling using single-nucleotide polymorphism arrays identifies novel chromosomal imbalances in pediatric glioblastomas

    Neuro Oncol

    (2010)
  • M. Zarghooni 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)
  • R.J. Gilbertson et al.

    ERBB1 is amplified and overexpressed in high-grade diffusely infiltrative pediatric brain stem glioma

    Clin Cancer Res

    (2003)
  • D.A. Bax et al.

    EGFRvIII deletion mutations in pediatric high-grade glioma and response to targeted therapy in pediatric glioma cell lines

    Clin Cancer Res

    (2009)
  • G. Li et al.

    Expression of epidermal growth factor variant III (EGFRvIII) in pediatric diffuse intrinsic pontine gliomas

    J Neurooncol

    (2012)
  • K.K. Wong et al.

    Genome-wide allelic imbalance analysis of pediatric gliomas by single nucleotide polymorphic allele array

    Cancer Res

    (2006)
  • S. Puget et al.

    Mesenchymal transition and PDGFRA amplification/mutation are key distinct oncogenic events in pediatric diffuse intrinsic pontine gliomas

    PLoS One

    (2012)
  • J. Grill et al.

    Critical oncogenic mutations in newly diagnosed pediatric diffuse intrinsic pontine glioma

    Pediatr Blood Cancer

    (2012)
  • J. Barrow et al.

    Homozygous loss of ADAM3A revealed by genome-wide analysis of pediatric high-grade glioma and diffuse intrinsic pontine gliomas

    Neuro Oncol

    (2011)
  • B.S. Paugh et al.

    Novel oncogenic PDGFRA mutations in pediatric high-grade gliomas

    Cancer Res

    (2013)
  • K.W. Liu et al.

    SHP-2/PTPN11 mediates gliomagenesis driven by PDGFRA and INK4A/ARF aberrations in mice and humans

    J Clin Invest

    (2011)
  • Cited by (37)

    • Oncohistones

      2023, Epigenetic Cancer Therapy, Second Edition
    • Dual targeting of the epigenome via FACT complex and histone deacetylase is a potent treatment strategy for DIPG

      2021, Cell Reports
      Citation Excerpt :

      Hypophosphorylated Rb interacts with E2F1, resulting in repression of the cell cycle and proliferation arrest. Dysregulation of the Rb pathway is common in pediatric brain cancers (Diaz and Baker, 2014). Similarly, 30% of DIPGs are known to have amplification of genes that regulate G1 to S cell cycle progression, particularly the cyclin D family members CDK4 and CDK6 (Mohammad et al., 2017; Paugh et al., 2011).

    • Pediatric Gliomas: Molecular Landscape and Emerging Targets

      2021, Neurosurgery Clinics of North America
      Citation Excerpt :

      The mutation is found only in a minority of DMGs and likely is not an independent oncogenic driver.10,12,64,65 The ACVR1 mutation is associated with younger age, increased survival, and HIST1H3B, PIK3CA, and PIK3R1 mutations (see Fig. 1).12,13,64 The latter 2, along with mutations in growth factor receptor genes (ie, Platelet Derived Growth Factor Receptor Alpha (PDGFRA), EGFR, FGFR, and ACVR1), altered PTEN promoter methylation, and overexpression of YB1, all are known to act through amplification of cell proliferation via the RTK/Ras/PI3K pathway (see Fig. 2).2,4,6,10,64

    • Clinical and Molecular Characteristics of Thalamic Gliomas: Retrospective Report of 26 Cases

      2019, World Neurosurgery
      Citation Excerpt :

      Approximately 50% of TP53 gene mutations occur in astrocytic gliomas and will be present in ∼30% of primary glioblastoma and more frequent in secondary glioblastomas (65%) than in primary glioblastomas.19-21 Some previous whole genome analysis studies have shown that H3K27M mutations have significant overlap with TP53 mutations.22,23 A total of 11 thalamic glioma cases had TP53 gene mutations, with greater TP53 mutation rates in the H3K27M mutation group (7 of 12; 58.33%) than the wild-type group (4 of 14; 28.57%).

    • Mechanism of cancer: Oncohistones in action

      2018, Journal of Genetics and Genomics
      Citation Excerpt :

      Interestingly, these point mutations are able to “drive” tumorigenesis even if they only occur in one copy of one allele of the genes that encode a particular histone, regardless of many other “good” copies of their genes (Morgan and Shilatifard, 2013). For example, histone H3 mutations in pediatric high-grade glioma (pHGG) were all heterozygous, with only one out of the sixteen H3 genes mutated in any individual tumor, suggesting a dominant gain-of-function effect (Diaz and Baker, 2014). The mutant histones are therefore named as “oncohistones”.

    • Chemotherapy of Pediatric High-Grade Gliomas

      2018, Handbook of Brain Tumor Chemotherapy, Molecular Therapeutics, and Immunotherapy: Second Edition
    View all citing articles on Scopus

    S.J.B. is supported by NIH grant P01 CA096832 and ALSAC of St. Jude Children׳s Research Hospital.

    ☆☆

    The authors declare no conflicts of interest.

    View full text