Glioblastoma TCGA Mesenchymal and IGS 23 Tumors are Identifiable by IHC and have an Immune-phenotype Indicating a Potential Benefit from Immunotherapy.
Biomarkers, Tumor
/ genetics
Brain Neoplasms
/ classification
Computational Biology
Follow-Up Studies
Glioblastoma
/ classification
Humans
Immunohistochemistry
/ methods
Immunophenotyping
/ methods
Mesoderm
/ pathology
Oncogene Proteins, Fusion
/ genetics
Prognosis
RNA-Seq
Retrospective Studies
Tissue Array Analysis
Journal
Clinical cancer research : an official journal of the American Association for Cancer Research
ISSN: 1557-3265
Titre abrégé: Clin Cancer Res
Pays: United States
ID NLM: 9502500
Informations de publication
Date de publication:
15 12 2020
15 12 2020
Historique:
received:
16
06
2020
revised:
29
07
2020
accepted:
24
09
2020
pubmed:
2
10
2020
medline:
15
12
2021
entrez:
1
10
2020
Statut:
ppublish
Résumé
Molecular subtype classifications in glioblastoma may detect therapy sensitivities. IHC would potentially allow the identification of molecular subtypes in routine clinical practice. Formalin-fixed, paraffin-embedded tumor samples of 124 uniformly treated, newly diagnosed patients with glioblastoma were submitted to RNA sequencing, IHC, and immune-phenotyping to identify differences in molecular subtypes associated with treatment sensitivities. We detected high molecular and IHC overlapping of the The Cancer Genome Atlas (TCGA) mesenchymal subtype with instrinsic glioma subtypes (IGS) cluster 23 and of the TCGA classical subtype with IGS cluster 18. IHC patterns, gene fusion profiles, and immune-phenotypes varied across subtypes. IHC revealed that the TCGA classical subtype was identified by high expression of EGFR and low expression of PTEN, while the mesenchymal subtype was identified by low expression of SOX2 and high expression of two antibodies, SHC1 and TCIRG1, selected on the basis of RNA differential transcriptomic expression. The proneural subtype was identified by frequent positive IDH1 expression and high Olig2 and Ki67 expression. Immune-phenotyping showed that mesenchymal and IGS 23 tumors exhibited a higher positive effector cell score, a higher negative suppressor cell score, and lower levels of immune checkpoint molecules. The cell-type deconvolution analysis revealed that these tumors are highly enriched in M2 macrophages, resting memory CD4 There is a subset of tumors, frequently classified as mesenchymal or IGS cluster 23, that may be identified with IHC and could well be optimal candidates for immunotherapy.
Identifiants
pubmed: 32998960
pii: 1078-0432.CCR-20-2171
doi: 10.1158/1078-0432.CCR-20-2171
doi:
Substances chimiques
Biomarkers, Tumor
0
Oncogene Proteins, Fusion
0
Types de publication
Journal Article
Research Support, Non-U.S. Gov't
Langues
eng
Sous-ensembles de citation
IM
Pagination
6600-6609Informations de copyright
©2020 American Association for Cancer Research.
Références
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.
Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, Lhermitte B, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA. 2017;318:2306–16.
Hegi ME, Diserens A-C, Gorlia T, Hamou M-F, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997–1003.
Bleeker FE, Atai NA, Lamba S, Jonker A, Rijkeboer D, Bosch KS, et al. The prognostic IDH1(R132) mutation is associated with reduced NADP+-dependent IDH activity in glioblastoma. Acta Neuropathol. 2010;119:487–94.
Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–52.
Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
Li A, Walling J, Ahn S, Kotliarov Y, Su Q, Quezado M, et al. Unsupervised analysis of transcriptomic profiles reveals six glioma subtypes. Cancer Res. 2009;69:2091–9.
Freije WA, Castro-Vargas FE, Fang Z, Horvath S, Cloughesy T, Liau LM, et al. Gene expression profiling of gliomas strongly predicts survival. Cancer Res. 2004;64:6503–10.
Nigro JM, Misra A, Zhang Li, Smirnov I, Colman H, Griffin C, et al. Integrated array-comparative genomic hybridization and expression array profiles identify clinically relevant molecular subtypes of glioblastoma. Cancer Res. 2005;65:1678–86.
Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157–73.
Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110.
Brennan CW, Verhaak RGW, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155:462–77.
Wang Q, Hu B, Hu X, Kim H, Squatrito M, Scarpace L, et al. Tumor evolution of glioma-intrinsic gene expression subtypes associates with immunological changes in the microenvironment. Cancer Cell. 2017;32:42–56.
Nutt CL, Mani DR, Betensky RA, Tamayo P, Cairncross JG, Ladd C, et al. Gene expression-based classification of malignant gliomas correlates better with survival than histological classification. Cancer Res. 2003;63:1602–7.
Godard S, Getz G, Delorenzi M, Farmer P, Kobayashi H, Desbaillets I, et al. Classification of human astrocytic gliomas on the basis of gene expression: a correlated group of genes with angiogenic activity emerges as a strong predictor of subtypes. Cancer Res. 2003;63:6613–25.
Gravendeel LAM, Kouwenhoven MCM, Gevaert O, de Rooi JJ, Stubbs AP, Duijm JE, et al. Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology. Cancer Res. 2009;69:9065–72.
Karcher S, Steiner H-H, Ahmadi R, Zoubaa S, Vasvari G, Bauer H, et al. Different angiogenic phenotypes in primary and secondary glioblastomas. Int J Cancer. 2006;118:2182–9.
Liang Y, Diehn M, Watson N, Bollen AW, Aldape KD, Nicholas MK, et al. Gene expression profiling reveals molecularly and clinically distinct subtypes of glioblastoma multiforme. Proc Natl Acad Sci U S A. 2005;102:5814–9.
Rickman DS, Bobek MP, Misek DE, Kuick R, Blaivas M, Kurnit DM, et al. Distinctive molecular profiles of high-grade and low-grade gliomas based on oligonucleotide microarray analysis. Cancer Res. 2001;61:6885–91.
Sallinen SL, Sallinen PK, Haapasalo HK, Helin HJ, Helén PT, Schraml P, et al. Identification of differentially expressed genes in human gliomas by DNA microarray and tissue chip techniques. Cancer Res. 2000;60:6617–22.
Cloughesy T, Finocchiaro G, Belda-Iniesta C, Recht L, Brandes AA, Pineda E, et al. Randomized, double-blind, placebo-controlled, multicenter phase II study of onartuzumab plus bevacizumab versus placebo plus bevacizumab in patients with recurrent glioblastoma: efficacy, safety, and hepatocyte growth factor and O-6-methylguanine-DNA methyltransferase biomarker analyses. J Clin Oncol. 2017;35:343–51.
Sandmann T, Bourgon R, Garcia J, Li C, Cloughesy T, Chinot OL, 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. J Clin Oncol. 2015;33:2735–44.
Erdem-Eraslan L, Gravendeel LA, de Rooi J, Eilers PHC, Idbaih A, Splie WGM, et al. Intrinsic molecular subtypes of glioma are prognostic and predict benefit from adjuvant procarbazine, lomustine, and vincristine chemotherapy in combination with other prognostic factors in anaplastic oligodendroglial brain tumors: a report from EORTC study 26951. J Clin Oncol. 2013;31:328–36.
Erdem-Eraslan L, van den Bent MJ, Hoogstrate Y, Naz-Khan H, Stubbs A, van der Spek P, et al. Identification of patients with recurrent glioblastoma who may benefit from combined bevacizumab and CCNU therapy: a report from the BELOB trial. Cancer Res. 2016;76:525–34.
Gao Y, Weenink B, van den Bent MJ, Erdem-Eraslan L, Kros JM, Sillevis Smitt P, et al. Expression-based intrinsic glioma subtypes are prognostic in low-grade gliomas of the EORTC22033–26033 clinical trial. Eur J Cancer. 2018;94:168–78.
Gravendeel LA, de Rooi JJ, Eilers PH, van den Bent MJ, Sillevis Smitt PA, French PJ. Gene expression profiles of gliomas in formalin-fixed paraffin-embedded material. Br J Cancer. 2012;106:538–45.
Conroy S, Kruyt FAE, Joseph JV, Balasubramaniyan V, Bhat KP, Wagemakers M, et al. Subclassification of newly diagnosed glioblastomas through an immunohistochemical approach. PLoS One. 2014;9:e115687.
Popova SN, Bergqvist M, Dimberg A, Edqvist P-H, Ekman S, Hesselager G, et al. Subtyping of gliomas of various WHO grades by the application of immunohistochemistry. Histopathology. 2014;64:365–79.
Nagy Á, Garzuly F, Padányi G, Szűcs I, Feldmann Á, Murnyák B, et al. Molecular subgroups of glioblastoma- an assessment by immunohistochemical markers. Pathol Oncol Res. 2019;25:21–31.
Motomura K, Natsume A, Watanabe R, Ito I, Kato Y, Momota H, et al. Immunohistochemical analysis-based proteomic subclassification of newly diagnosed glioblastomas. Cancer Sci. 2012;103:1871–9.
Le Mercier M, Hastir D, Moles Lopez X, De Nève N, Maris C, Trepant A-L, et al. A simplified approach for the molecular classification of glioblastomas. PLoS One. 2012;7:e45475.
Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18:248–62.
Nikanjam M, Okamura R, Barkauskas DA, Kurzrock R. Targeting fusions for improved outcomes in oncology treatment. Cancer. 2020;126:1315–21.
Esteve-Codina A, Arpi O, Martinez-García M, Pineda E, Mallo M, Gut M, et al. A comparison of RNA-seq results from paired formalin-fixed paraffin-embedded and fresh-frozen glioblastoma tissue samples. PLoS One. 2017;12:e0170632.
Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 1999;59:793–7.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323.
Wang L, Wang S, Li W. RSeQC: quality control of RNA-seq experiments. Bioinformatics. 2012;28:2184–5.
Bowman RL, Wang Q, Carro A, Verhaak RG, Squatrito M. GlioVis data portal for visualization and analysis of brain tumor expression datasets. Neuro-oncology. 2017;19:139–41.
Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12:453–7.
Stransky N, Cerami E, Schalm S, Kim JL, Lengauer C. The landscape of kinase fusions in cancer. Nat Commun. 2014;5:4846.
Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP, et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell. 2010;17:510–22.
Bulwin G-C, Heinemann T, Bugge V, Winter M, Lohan A, Schlawinsky M, et al. TIRC7 inhibits T cell proliferation by modulation of CTLA-4 expression. J Immunol. 2006;177:6833–41.
Grégoire H, Roncali L, Rousseau A, Chérel M, Delneste Y, Jeannin P, et al. Targeting tumor associated macrophages to overcome conventional treatment resistance in glioblastoma. Front Pharmacol. 2020;11:368.
Wen PY, Reardon DA, Armstrong TS, Phuphanich S, Aiken RD, Landolfi JC, et al. A randomized double-blind placebo-controlled phase II trial of dendritic cell vaccine ICT-107 in newly diagnosed patients with glioblastoma. Clin Cancer Res. 2019;25:5799–807.
Pombo Antunes AR, Scheyltjens I, Duerinck J, Neyns B, Movahedi K, Van Ginderachter JA. Understanding the glioblastoma immune microenvironment as basis for the development of new immunotherapeutic strategies. Elife. 2020;9:e52176.
Prins RM, Soto H, Konkankit V, Odesa SK, Eskin A, Yong WH, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17:1603–15.