[en] Inevitable tumor recurrence and a poor median survival are frustrating reminders of the inefficacy of our current standard of care for patients with newly diagnosed glioblastoma (GBM), which includes surgery followed by radiotherapy and chemotherapy with the DNA alkylating agent temozolomide. Because resistance to genotoxic damage is achieved mainly through execution of the DNA damage response (DDR) and DNA repair pathways, knowledge of the changes in DNA repair and cell-cycle gene expression that occur during tumor development might help identify new targets and improve treatment. Here, we performed a gene expression analysis targeting components of the DNA repair and cell-cycle machineries in cohorts of paired tumor samples (i.e., biopsies from the same patient obtained at the time of primary tumor operation and at recurrence) from patients treated with radiotherapy or radiotherapy plus temozolomide. We identified and validated a 27-gene signature that resulted in the classification of GBM specimens into three groups, two of which displayed inverse expression profiles. Each group contained primary and recurrent samples, and the tumor at relapse frequently displayed a gene expression profile different from that of the matched primary biopsy. Within the groups that exhibited opposing gene expression profiles, the expression pattern of the gene signature at relapse was linked to progression-free survival. We provide experimental evidence that our signature exposes group-specific vulnerabilities against genotoxicants and inhibitors of the cell cycle and DDR, with the prospect of personalized therapeutic strategies.Significance: These findings suggest that classification of GBM tumors based on a DNA repair and cell-cycle gene expression signature exposes vulnerabilities to standard-of-care therapies and offers the potential for personalized therapeutic strategies.
Disciplines :
Oncology
Author, co-author :
GOBIN, Matthieu ; NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg.
NAZAROV, Petr ; Proteome and Genome Research Unit, LIH, Luxembourg, Luxembourg.
Warta, Rolf; Department of Neurosurgery, University Clinic Heidelberg, Heidelberg, Germany.
Timmer, Marco; Neurosurgery Centre, University Clinic Cologne, Cologne, Germany.
Reifenberger, Guido; Institute for Neuropathology, Düsseldorf, Germany.
Felsberg, Joerg; Institute for Neuropathology, Düsseldorf, Germany.
Vallar, Laurent; Proteome and Genome Research Unit, LIH, Luxembourg, Luxembourg.
Chalmers, Anthony J ; Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom.
Herold-Mende, Christel C; Department of Neurosurgery, University Clinic Heidelberg, Heidelberg, Germany.
Goldbrunner, Roland; Neurosurgery Centre, University Clinic Cologne, Cologne, Germany.
NICLOU, Simone P. ; NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg.
Van Dyck, Eric; NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg. eric.vandyck@lih.lu.
External co-authors :
yes
Language :
English
Title :
A DNA Repair and Cell-Cycle Gene Expression Signature in Primary and Recurrent Glioblastoma: Prognostic Value and Clinical Implications.
Publication date :
15 March 2019
Journal title :
Cancer Research
ISSN :
0008-5472
eISSN :
1538-7445
Publisher :
American Association for Cancer Research, United States - Maryland
Volume :
79
Issue :
6
Pages :
1226-1238
Peer reviewed :
Peer Reviewed verified by ORBi
Funding number :
G0802755/MRC_/Medical Research Council/United Kingdom
Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, et al. Long-term survival with glioblastoma multiforme. Brain 2007;130(Pt 10): 2596–606.
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–96.
Johnson DR, O'Neill BP. Glioblastoma survival in the United States before and during the temozolomide era. J Neurooncol 2012;107: 359–64.
Erasimus H, Gobin M, Niclou S, Van Dyck E. DNA repair mechanisms and their clinical impact in glioblastoma. Mutat Res Rev Mutat Res 2016;769: 19–35.
Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma–are we there yet? Neuro Oncol 2013;15:4–27.
Gallego O. Nonsurgical treatment of recurrent glioblastoma. Curr Oncol 2015;22:e273–81.
Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997–1003.
Brown JS, O'Carrigan B, Jackson SP, Yap TA. Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discov 2017;7:20–37.
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.
Felsberg J, Wolter M, Seul H, Friedensdorf B, Goppert M, Sabel MC, et al. Rapid and sensitive assessment of the IDH1 and IDH2 mutation status in cerebral gliomas based on DNA pyrosequencing. Acta Neuropathol 2010; 119:501–7.
Mikeska T, Bock C, El-Maarri O, Hubner A, Ehrentraut D, Schramm J, et al. Optimization of quantitative MGMT promoter methylation analysis using pyrosequencing and combined bisulfite restriction analysis. J Mol Diagn 2007;9:368–81.
Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL, et al. Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 2008;26:317–25.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.
Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015;43:e47.
Brunet JP, Tamayo P, Golub TR, Mesirov JP. Metagenes and molecular pattern discovery using matrix factorization. Proc Natl Acad Sci U S A 2004; 101:4164–9.
Heinaniemi M, Nykter M, Kramer R, Wienecke-Baldacchino A, Sinkkonen L, Zhou JX, et al. Gene-pair expression signatures reveal lineage control. Nat Methods 2013;10:577–83.
Therneau TG, PM. Modeling Survival Data: Extending the Cox Model. New York, NY: Springer-Verlag; 2000.
Veldman-Jones MH, Brant R, Rooney C, Geh C, Emery H, Harbron CG, et al. Evaluating robustness and sensitivity of the NanoString technologies nCounter platform to enable multiplexed gene expression analysis of clinical samples. Cancer Res 2015;75:2587–93.
Murat A, Migliavacca E, Gorlia T, Lambiv WL, Shay T, Hamou MF, et al. Stem cell-related "self-renewal" signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol 2008;26:3015–24.
Freire P, Vilela M, Deus H, Kim YW, Koul D, Colman H, et al. Exploratory analysis of the copy number alterations in glioblastoma multiforme. PLoS One 2008;3:e4076.
Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell 2013;155: 462–77.
Chen HC, Kodell RL, Cheng KF, Chen JJ. Assessment of performance of survival prediction models for cancer prognosis. BMC Med Res Methodol 2012;12:102.
Gorlia T, van den Bent MJ, Hegi ME, Mirimanoff RO, Weller M, Cairncross JG, et al. Nomograms for predicting survival of patients with newly diagnosed glioblastoma: prognostic factor analysis of EORTC and NCIC trial 26981-22981/CE.3. Lancet Oncol 2008;9:29–38.
Kim H, Zheng S, Amini SS, Virk SM, Mikkelsen T, Brat DJ, et al. Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution. Genome Res 2015;25: 316–27.
Crespo I, Vital AL, Gonzalez-Tablas M, Patino Mdel C, Otero A, Lopes MC, et al. Molecular and genomic alterations in glioblastoma multiforme. Am J Pathol 2015;185:1820–33.
Burgess DJ, Doles J, Zender L, Xue W, Ma B, McCombie WR, et al. Topoisomerase levels determine chemotherapy response in vitro and in vivo. Proc Natl Acad Sci U S A 2008;105:9053–8.
Deans AJ, West SC. DNA interstrand crosslink repair and cancer. Nat Rev Cancer 2011;11:467–80.
Martin LP, Hamilton TC, Schilder RJ. Platinum resistance: the role of DNA repair pathways. Clin Cancer Res 2008;14:1291–5.
Ward A, Khanna KK, Wiegmans AP. Targeting homologous recombination, new pre-clinical and clinical therapeutic combinations inhibiting RAD51. Cancer Treat Rev 2015;41:35–45.
Ledur PF, Onzi GR, Zong H, Lenz G. Culture conditions defining glioblastoma cells behavior: what is the impact for novel discoveries? Oncotarget 2017;8:69185–97.
Fulton B, Short SC, James A, Nowicki S, McBain C, Jefferies S, et al. PARADIGM-2: Two parallel phase I studies of olaparib and radiotherapy or olaparib and radiotherapy plus temozolomide in patients with newly diagnosed glioblastoma, with treatment stratified by MGMT status. Clin Transl Rad Oncol 2017;8:12–6.
Michels J, Vitale I, Saparbaev M, Castedo M, Kroemer G. Predictive biomarkers for cancer therapy with PARP inhibitors. Oncogene 2014; 33:3894–907.
McGrail DJ, Lin CC, Garnett J, Liu Q, Mo W, Dai H, et al. Improved prediction of PARP inhibitor response and identification of synergizing agents through use of a novel gene expression signature generation algorithm. NPJ Syst Biol Appl 2017;3:8.
Koussounadis A, Langdon SP, Harrison DJ, Smith VA. Chemotherapy-induced dynamic gene expression changes in vivo are prognostic in ovarian cancer. Br J Cancer 2014;110:2975–84.
Legrier ME, Bieche I, Gaston J, Beurdeley A, Yvonnet V, Deas O, et al. Activation of IFN/STAT1 signalling predicts response to chemotherapy in oestrogen receptor-negative breast cancer. Br J Cancer 2016;114:177–87.
Johnson BE, Mazor T, Hong C, Barnes M, Aihara K, McLean CY, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 2014;343:189–93.
van Thuijl HF, Mazor T, Johnson BE, Fouse SD, Aihara K, Hong C, et al. Evolution of DNA repair defects during malignant progression of low-grade gliomas after temozolomide treatment. Acta Neuropathol 2015;129: 597–607.
Xie C, Sheng H, Zhang N, Li S, Wei X, Zheng X. Association of MSH6 mutation with glioma susceptibility, drug resistance and progression. Mol Clin Oncol 2016;5:236–240.
The Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008;455:1061–8.
Yip S, Miao J, Cahill DP, Iafrate AJ, Aldape K, Nutt CL, et al. MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin Cancer Res 2009;15:4622–9.
Kwon SM, Kang SH, Park CK, Jung S, Park ES, Lee JS, et al. Recurrent glioblastomas reveal molecular subtypes associated with mechanistic implications of drug-resistance. PLoS One 2015;10:e0140528.
Spiegl-Kreinecker S, Pirker C, Marosi C, Buchroithner J, Pichler J, Silye R, et al. Dynamics of chemosensitivity and chromosomal instability in recurrent glioblastoma. Br J Cancer 2007;96:960–9.
Zhang W, Mao JH, Zhu W, Jain AK, Liu K, Brown JB, et al. Centromere and kinetochore gene misexpression predicts cancer patient survival and response to radiotherapy and chemotherapy. Nat Commun 2016;7:12619.
Klughammer J, Kiesel B, Roetzer T, Fortelny N, Nemc A, Nenning KH, et al. The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med 2018;24: 1611–24.
consortium TG. Glioma through the looking GLASS: molecular evolution of diffuse gliomas and the Glioma Longitudinal Analysis Consortium. Neuro Oncol 2018;20:873–84.
Brandes AA, Basso U, Reni M, Vastola F, Tosoni A, Cavallo G, et al. First-line chemotherapy with cisplatin plus fractionated temozolomide in recurrent glioblastoma multiforme: a phase II study of the Gruppo Italiano Coop-erativo di Neuro-Oncologia. J Clin Oncol 2004;22:1598–604.
Buckner JC, Ballman KV, Michalak JC, Burton GV, Cascino TL, Schomberg PJ, et al. Phase III trial of carmustine and cisplatin compared with carmustine alone and standard radiation therapy or accelerated radiation therapy in patients with glioblastoma multiforme: North Central Cancer Treatment Group 93-72-52 and Southwest Oncology Group 9503 Trials. J Clin Oncol 2006;24:3871–9.
Leonard A, Wolff JE. Etoposide improves survival in high-grade glioma: a meta-analysis. Anticancer Res 2013;33:3307–15.
Franceschi E, Cavallo G, Scopece L, Paioli A, Pession A, Magrini E, et al. Phase II trial of carboplatin and etoposide for patients with recurrent high-grade glioma. Br J Cancer 2004;91:1038–44.
Zustovich F, Lombardi G, Della Puppa A, Rotilio A, Scienza R, Pastorelli D. A phase II study of cisplatin and temozolomide in heavily pre-treated patients with temozolomide-refractory high-grade malignant glioma. Anticancer Res 2009;29:4275–9.
Li N, Maly DJ, Chanthery YH, Sirkis DW, Nakamura JL, Berger MS, et al. Radiotherapy followed by aurora kinase inhibition targets tumor-propagating cells in human glioblastoma. Mol Cancer Ther 2015;14:419–28.
Borges KS, Castro-Gamero AM, Moreno DA, da Silva Silveira V, Bras-sesco MS, de Paula Queiroz RG, et al. Inhibition of Aurora kinases enhances chemosensitivity to temozolomide and causes radiosensiti-zation in glioblastoma cells. J Cancer Res Clin Oncol 2012;138: 405–14.
Lee E, Pain M, Wang H, Herman JA, Toledo CM, DeLuca JG, et al. Sensitivity to BUB1B inhibition defines an alternative classification of glioblastoma. Cancer Res 2017;77:5518–29.
Tandle AT, Kramp T, Kil WJ, Halthore A, Gehlhaus K, Shankavaram U, et al. Inhibition of polo-like kinase 1 in glioblastoma multiforme induces mitotic catastrophe and enhances radiosensitisation. Eur J Cancer 2013; 49:3020–8.