Integrative bioinformatics analysis of the correlation between CDKN2A homozygous deletion, RB1 loss, and PDGFRA amplification in glioma

Introduction: Malignant gliomas constitute a heterogeneous group of primary central nervous system tumors originating from glial cells or their precursors. They display aggressive characteristics and unfavorable clinical outcomes. The 2021 World Health Organization classification emphasizes molecular markers as essential elements for glioma categorization, therapeutic planning, and prognostic assessment. Specific genetic alterations, including CDKN2A homozygous deletion, RB1 loss, and PDGFRA amplification, emerge as potentially crucial determinants of tumor behavior and patient outcomes.
Objectives: This investigation examines the relationship between these genetic modifications and survival outcomes while evaluating their influence on the tumor immune landscape.
Methods: We utilized the cBioPortal database to analyze the mutation status of CDKN2A homozygous deletion, RB1 loss, and PDGFRA amplification in gliomas, and further investigated the correlation between these mutations and glioma prognosis. Next, we evaluated the expression levels of these genes in gliomas by analyzing data from the Cancer Genome Atlas (TCGA) database. In addition, we explored the potential prognostic value of these genes in gliomas using information from the Chinese Glioma Genome Atlas and TCGA databases. Finally, immunohistochemistry was performed to detect the protein expression of cyclin-dependent kinase inhibitor 2A (CDKN2A), retinoblastoma 1 (RB1), and platelet-derived growth factor receptor alpha (PDGFRA) in gliomas.
Results: Our analysis revealed that the homozygous deletion of CDKN2A and the amplification of PDGFRA are significantly correlated with poor overall survival and reduced progression-free survival in glioma patients (p<0.05), while the loss of RB1 showed no significant association with prognosis. Additionally, the expression levels of PDGFRA and RB1 were closely related to immune cell infiltration.
Conclusion: High expression of CDKN2A and RB1 generally indicated poorer survival outcomes, whereas elevated expression of PDGFRA exhibited an opposite trend, potentially influencing glioma immune response and therapeutic efficacy by modulating the tumor microenvironment. Immunohistochemical analysis further confirmed the high expression of CDKN2A, RB1, and PDGFRA in gliomas.
- Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: A summary. Neuro Oncol. 2021;23(8):1231-1251. doi: 10.1093/neuonc/noab106
- Eckel-Passow JE, Lachance DH, Molinaro AM, et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med. 2015;372(26):2499-24508. doi: 10.1056/nejmoa1407279
- Wang LM, Englander ZK, Miller ML, Bruce JN. Malignant glioma. Adv Exp Med Biol. 2023;1405:1-30. doi: 10.1007/978-3-031-23705-8_1
- Wu W, Klockow JL, Zhang M, et al. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol Res. 2021;171:105780. doi: 10.1016/j.phrs.2021.105780
- Oszvald A, Güresir E, Setzer M, et al. Glioblastoma therapy in the elderly and the importance of the extent of resection regardless of age. J Neurosurg. 2012;116(2):357-364. doi: 10.3171/2011.8.Jns102114
- Le Rhun E, Preusser M, Roth P, et al. Molecular targeted therapy of glioblastoma. Cancer Treat Rev. 2019;80:101896. doi: 10.1016/j.ctrv.2019.101896
- Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807-1812. doi: 10.1126/science.1164382
- Bleeker FE, Atai NA, Lamba S, et al. The prognostic IDH1(R132) mutation is associated with reduced NADP+- dependent IDH activity in glioblastoma. Acta Neuropathol. 2010;119(4):487-494. doi: 10.1007/s00401-010-0645-6
- Khuong-Quang DA, Buczkowicz P, Rakopoulos P, et al. K27M mutation in histone H3.3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta Neuropathol. 2012;124(3):439-447. doi: 10.1007/s00401-012-0998-0
- Pekmezci M, Rice T, Molinaro AM, et al. Adult infiltrating gliomas with WHO 2016 integrated diagnosis: Additional prognostic roles of ATRX and TERT. Acta Neuropathol. 2017;133(6):1001-1016. doi: 10.1007/s00401-017-1690-1
- Bäcklund LM, Nilsson BR, Liu L, Ichimura K, Collins VP. Mutations in Rb1 pathway-related genes are associated with poor prognosis in anaplastic astrocytomas. Br J Cancer. 2005;93(1):124-130. doi: 10.1038/sj.bjc.6602661
- Reis GF, Pekmezci M, Hansen HM, et al. CDKN2A loss is associated with shortened overall survival in lower-grade (World Health Organization Grades II-III) astrocytomas. J Neuropathol Exp Neurol. 2015;74(5):442-452. doi: 10.1097/nen.0000000000000188
- Śledzińska P, Bebyn MG, Furtak J, Kowalewski J, Lewandowska MA. Prognostic and predictive biomarkers in gliomas. Int J Mole Sci. 2021;22(19):10373. doi: 10.3390/ijms221910373
- Schwartzbaum JA, Fisher JL, Aldape KD, Wrensch M. Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol. 2006;2(9):494-503; quiz 1. doi: 10.1038/ncpneuro0289
- Ostrom QT, Bauchet L, Davis FG, et al. The epidemiology of glioma in adults: A “state of the science” review. Neuro Oncol. 2014;16(7):896-913. doi: 10.1093/neuonc/nou087
- Molinaro AM, Taylor JW, Wiencke JK, Wrensch MR. Genetic and molecular epidemiology of adult diffuse glioma. Nat Rev Neurol. 2019;15(7):405-417. doi: 10.1038/s41582-019-0220-2
- Ostrom QT, Cioffi G, Gittleman H, et al. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2012-2016. Neuro Oncol. 2019;21(Suppl 5):v1-v100. doi: 10.1093/neuonc/noz150
- Sant M, Minicozzi P, Lagorio S, Børge Johannesen T, Marcos-Gragera R, Francisci S. Survival of European patients with central nervous system tumors. Int J Cancer. 2012;131(1):173-815. doi: 10.1002/ijc.26335
- Chiang J, Harreld JH, Tinkle CL, et al. A single-center study of the clinicopathologic correlates of gliomas with a MYB or MYBL1 alteration. Acta Neuropathol. 2019;138(6):1091-1092. doi: 10.1007/s00401-019-02081-1
- Hao Z, Guo D. EGFR mutation: Novel prognostic factor associated with immune infiltration in lower-grade glioma; an exploratory study. BMC Cancer. 2019;19(1):1184. doi: 10.1186/s12885-019-6384-8
- Appay R, Dehais C, Maurage CA, et al. CDKN2A homozygous deletion is a strong adverse prognosis factor in diffuse malignant IDH-mutant gliomas. Neuro Oncol. 2019;21(12):1519-1528. doi: 10.1093/neuonc/noz124
- Lu VM, O’Connor KP, Shah AH, et al. The prognostic significance of CDKN2A homozygous deletion in IDH-mutant lower-grade glioma and glioblastoma: A systematic review of the contemporary literature. J Neurooncol. 2020;148(2):221-229. doi: 10.1007/s11060-020-03528-2
- Carlotto BS, Trevisan P, Provenzi VO, et al. PDGFRA, KIT, and KDR gene amplification in glioblastoma: Heterogeneity and clinical significance. Neuromolecul Med. 2023;25(3):441-450. doi: 10.1007/s12017-023-08749-y
- Sullivan JP, Nahed BV, Madden MW, et al. Brain tumor cells in circulation are enriched for mesenchymal gene expression. Cancer Discov. 2014;4(11):1299-1309. doi: 10.1158/2159-8290.Cd-14-0471