Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma

Background A high heterogeneity and activation of multiple oncogenic pathways have been implicated in failure of targeted therapies in glioblastoma (GBM). Methods Using The Cancer Genome Atlas data, we identified subtype-specific prognostic core genes by a combined approach of genome-wide Cox regression and Gene Set Enrichment Analysis. The results were validated with 8 combined public datasets containing 608 GBMs. We further examined prognostic chromosome aberrations and mutations. Results In classical and mesenchymal subtypes, 2 receptor tyrosine kinases (RTKs) (MET and IGF1R), and the genes in RTK downstream pathways such as phosphatidylinositol-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), and nuclear factor-kappaB (NF-kB), were commonly detected as prognostic core genes. Classical subtype-specific prognostic core genes included those in cell cycle, DNA repair, and the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway. Immune-related genes were enriched in the prognostic genes showing negative promoter cytosine-phosphate-guanine (CpG) methylation/expression correlations. Mesenchymal subtype-specific prognostic genes were those related to mesenchymal cell movement, PI3K/Akt, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), Wnt/β-catenin, and Wnt/Ca2+ pathways. In copy number alterations and mutations, 6p loss and TP53 mutation were associated with poor and good survival, respectively, in the classical subtype. In the mesenchymal subtype, patients with PIK3R1 or PCLO mutations showed poor prognosis. In the glioma CpG island methylator phenotype (G-CIMP) subtype, patients harboring 10q loss, 12p gain, or 14q loss exhibited poor survival. Furthermore, 10q loss was significantly associated with the recently recognized G-CIMP subclass showing relatively low CpG methylation and poor prognosis. Conclusion These subtype-specific alterations have promising potentials as new prognostic biomarkers and therapeutic targets combined with surrogate markers of GBM subtypes. However, considering the small number of events, the results of copy number alterations and mutations require further validations.

[1]  D. Nam,et al.  WNT signaling in glioblastoma and therapeutic opportunities , 2016, Laboratory Investigation.

[2]  S. Gerson MGMT: its role in cancer aetiology and cancer therapeutics , 2004, Nature Reviews Cancer.

[3]  N. Cruickshanks,et al.  Role and Therapeutic Targeting of the HGF/MET Pathway in Glioblastoma , 2017, Cancers.

[4]  Alona Muzikansky,et al.  The prognostic significance of phosphatidylinositol 3-kinase pathway activation in human gliomas. , 2004, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[5]  B. Yamini,et al.  Nuclear factor-κB in glioblastoma: insights into regulators and targeted therapy. , 2016, Neuro-oncology.

[6]  M. Lingen,et al.  The novel tumor suppressor NOL7 post-transcriptionally regulates thrombospondin-1 expression , 2013, Oncogene.

[7]  Thomas D. Wu,et al.  Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. , 2006, Cancer cell.

[8]  M. Karsy,et al.  Molecular, histopathological, and genomic variants of glioblastoma. , 2014, Frontiers in bioscience.

[9]  Joshua M. Korn,et al.  Comprehensive genomic characterization defines human glioblastoma genes and core pathways , 2008, Nature.

[10]  T. Cloughesy,et al.  mTOR signaling in glioblastoma: lessons learned from bench to bedside , 2010, Neuro-oncology.

[11]  J. Trojan.,et al.  Insulin-like growth factor type I biology and targeting in malignant gliomas , 2007, Neuroscience.

[12]  Liu Cao,et al.  PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma , 2016, Oncotarget.

[13]  Mitchel S Berger,et al.  Contribution of Notch signaling activation to human glioblastoma multiforme. , 2007, Journal of neurosurgery.

[14]  R. Arceci Identification of a CpG Island Methylator Phenotype that Defines a Distinct Subgroup of Glioma , 2010 .

[15]  K. Hoang-Xuan,et al.  Differential gene methylation in paired glioblastomas suggests a role of immune response pathways in tumor progression , 2015, Journal of Neuro-Oncology.

[16]  Stephen R. Piccolo,et al.  Robust meta-analysis shows that glioma transcriptional subtyping complements traditional approaches , 2014, Cellular Oncology.

[17]  Eric S. Lander,et al.  Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing , 2012, Proceedings of the National Academy of Sciences.

[18]  M. Lopes,et al.  Microglia-glioblastoma interactions: New role for Wnt signaling. , 2017, Biochimica et biophysica acta. Reviews on cancer.

[19]  D. Maric,et al.  Tumor-infiltrating myeloid cells activate Dll4/Notch/TGF-β signaling to drive malignant progression. , 2014, Cancer research.

[20]  A. Merlo,et al.  Notch signaling in glioblastoma: a developmental drug target? , 2010, BMC medicine.

[21]  Steven J. M. Jones,et al.  Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma , 2016, Cell.

[22]  R. Mirimanoff,et al.  MGMT gene silencing and benefit from temozolomide in glioblastoma. , 2005, The New England journal of medicine.

[23]  B. Carneiro,et al.  Wnt/beta-catenin pathway: modulating anticancer immune response , 2017, Journal of Hematology & Oncology.

[24]  Rafael A Irizarry,et al.  Exploration, normalization, and summaries of high density oligonucleotide array probe level data. , 2003, Biostatistics.

[25]  R. Bourgon,et al.  Patients With Proneural Glioblastoma May Derive Overall Survival Benefit From the Addition of Bevacizumab to First-Line Radiotherapy and Temozolomide: Retrospective Analysis of the AVAglio Trial. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[26]  Andrew P. Stubbs,et al.  Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology. , 2009, Cancer research.

[27]  S. Horvath,et al.  Gene Expression Profiling of Gliomas Strongly Predicts Survival , 2004, Cancer Research.

[28]  E. Domany,et al.  Stem cell-related "self-renewal" signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[29]  S. Horvath,et al.  Gene expression analysis of glioblastomas identifies the major molecular basis for the prognostic benefit of younger age , 2008, BMC Medical Genomics.

[30]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[31]  D. Haussler,et al.  The Somatic Genomic Landscape of Glioblastoma , 2013, Cell.

[32]  M. West,et al.  Gene expression profiling and genetic markers in glioblastoma survival. , 2005, Cancer research.

[33]  L. Chin,et al.  Malignant astrocytic glioma: genetics, biology, and paths to treatment. , 2007, Genes & development.

[34]  G. Getz,et al.  GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers , 2011, Genome Biology.

[35]  R. Wilson,et al.  Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. , 2010, Cancer cell.

[36]  C. James,et al.  Proteoglycans and their roles in brain cancer , 2013, The FEBS journal.

[37]  Li Ding,et al.  Somatic Mutations of PIK3R1 Promote Gliomagenesis , 2012, PloS one.

[38]  Zhongming Zhao,et al.  Uncovering MicroRNA and Transcription Factor Mediated Regulatory Networks in Glioblastoma , 2012, PLoS Comput. Biol..

[39]  S. Gabriel,et al.  Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. , 2010, Cancer cell.

[40]  Annette Lee,et al.  Genome-Wide Methylation Analyses in Glioblastoma Multiforme , 2014, PloS one.

[41]  C. Sander,et al.  Automated Network Analysis Identifies Core Pathways in Glioblastoma , 2010, PloS one.

[42]  B. Kálmán,et al.  Molecular Heterogeneity of Glioblastoma and its Clinical Relevance , 2014, Pathology & Oncology Research.

[43]  Jean-Yves Delattre,et al.  An ANOCEF genomic and transcriptomic microarray study of the response to radiotherapy or to alkylating first-line chemotherapy in glioblastoma patients , 2010, Molecular Cancer.

[44]  Shawn M. Gillespie,et al.  Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma , 2014, Science.

[45]  Edward F. Chang,et al.  Erratum: Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment (Cancer Cell (2017) 32(1) (42–56.e6)(S1535610817302532)(10.1016/j.ccell.2017.06.003)) , 2018 .

[46]  Jingxuan Yang,et al.  Deregulated Signaling Pathways in Glioblastoma Multiforme: Molecular Mechanisms and Therapeutic Targets , 2012, Cancer investigation.

[47]  Subha Madhavan,et al.  Rembrandt: Helping Personalized Medicine Become a Reality through Integrative Translational Research , 2009, Molecular Cancer Research.

[48]  F. Marincola,et al.  Identification of genetic determinants of breast cancer immune phenotypes by integrative genome-scale analysis , 2017, Oncoimmunology.

[49]  S. Nozell,et al.  NF-κB and STAT3 signaling in glioma: targets for future therapies , 2010, Expert review of neurotherapeutics.

[50]  T. Jiang,et al.  Prognostic value of MGMT promoter methylation and TP53 mutation in glioblastomas depends on IDH1 mutation. , 2015, Asian Pacific journal of cancer prevention : APJCP.