Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas

Patrick J Killela, Christopher J Pirozzi, Patrick Healy, Zachary J Reitman, Eric Lipp, B Ahmed Rasheed, Rui Yang, Bill H Diplas, Zhaohui Wang, Paula K Greer, Huishan Zhu, Catherine Y Wang, Austin B Carpenter, Henry Friedman, Allan H Friedman, Stephen T Keir, Jie He, Yiping He, Roger E McLendon, James E Herndon 2nd, Hai Yan, Darell D Bigner, Patrick J Killela, Christopher J Pirozzi, Patrick Healy, Zachary J Reitman, Eric Lipp, B Ahmed Rasheed, Rui Yang, Bill H Diplas, Zhaohui Wang, Paula K Greer, Huishan Zhu, Catherine Y Wang, Austin B Carpenter, Henry Friedman, Allan H Friedman, Stephen T Keir, Jie He, Yiping He, Roger E McLendon, James E Herndon 2nd, Hai Yan, Darell D Bigner

Abstract

Frequent mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) and the promoter of telomerase reverse transcriptase (TERT) represent two significant discoveries in glioma genomics. Understanding the degree to which these two mutations co-occur or occur exclusively of one another in glioma subtypes presents a unique opportunity to guide glioma classification and prognosis. We analyzed the relationship between overall survival (OS) and the presence of IDH1/2 and TERT promoter mutations in a panel of 473 adult gliomas. We hypothesized and show that genetic signatures capable of distinguishing among several types of gliomas could be established providing clinically relevant information that can serve as an adjunct to histopathological diagnosis. We found that mutations in the TERT promoter occurred in 74.2% of glioblastomas (GBM), but occurred in a minority of Grade II-III astrocytomas (18.2%). In contrast, IDH1/2 mutations were observed in 78.4% of Grade II-III astrocytomas, but were uncommon in primary GBM. In oligodendrogliomas, TERT promoter and IDH1/2 mutations co-occurred in 79% of cases. Patients whose Grade III-IV gliomas exhibit TERT promoter mutations alone predominately have primary GBMs associated with poor median OS (11.5 months). Patients whose Grade III-IV gliomas exhibit IDH1/2 mutations alone predominately have astrocytic morphologies and exhibit a median OS of 57 months while patients whose tumors exhibit both TERT promoter and IDH1/2 mutations predominately exhibit oligodendroglial morphologies and exhibit median OS of 125 months. Analyzing gliomas based on their genetic signatures allows for the stratification of these patients into distinct cohorts, with unique prognosis and survival.

Figures

Fig 1. Distribution of TERT promoter and…
Fig 1. Distribution of TERT promoter and IDH1/2 mutations in a panel of 473 adult gliomas
Mutational analysis of 473 adult gliomas for TERT promoter and IDH1/2 mutations. Data are from 240 Grade IV GBM (A), 88 Grade II-III astrocytomas (B), 58 Grade II-III oligoastrocytomas (C), and, 87 Grade II-III oligodendrogliomas (D). Mutation status is indicated by color shading, with gray coloring indicating wild type sequence, red indicating mutations in the TERT promoter, and green indicating mutations in IDH1/2.
Fig 2. Overall Survival stratified by TERT…
Fig 2. Overall Survival stratified by TERT promoter and IDH1/2 mutational status and histology within each tumor grade
Overall survival was represented by Kaplan Meier plots for individual WHO tumor grade: a) Grade II (n=103), b) Grade III (n=121), c) Grade IV (n=218). Only subgroups with at least 10 patients were included in the analyses. Tumors were represented by mutations status on the left (TERT promoter status / IDH1/2 status) and histology on the right (A represents Astrocytomas, O represents Oligodendrogliomas, and OA represents Oligoastrocytomas).
Fig 2. Overall Survival stratified by TERT…
Fig 2. Overall Survival stratified by TERT promoter and IDH1/2 mutational status and histology within each tumor grade
Overall survival was represented by Kaplan Meier plots for individual WHO tumor grade: a) Grade II (n=103), b) Grade III (n=121), c) Grade IV (n=218). Only subgroups with at least 10 patients were included in the analyses. Tumors were represented by mutations status on the left (TERT promoter status / IDH1/2 status) and histology on the right (A represents Astrocytomas, O represents Oligodendrogliomas, and OA represents Oligoastrocytomas).
Fig 2. Overall Survival stratified by TERT…
Fig 2. Overall Survival stratified by TERT promoter and IDH1/2 mutational status and histology within each tumor grade
Overall survival was represented by Kaplan Meier plots for individual WHO tumor grade: a) Grade II (n=103), b) Grade III (n=121), c) Grade IV (n=218). Only subgroups with at least 10 patients were included in the analyses. Tumors were represented by mutations status on the left (TERT promoter status / IDH1/2 status) and histology on the right (A represents Astrocytomas, O represents Oligodendrogliomas, and OA represents Oligoastrocytomas).
Fig 3. Overall Survival stratified by TERT…
Fig 3. Overall Survival stratified by TERT promoter and IDH1/2 mutational status and histology among Grade III and IV patients
Overall survival was represented by Kaplan Meier plots stratified by a) histology (A represents Astrocytomas, O represents Oligodendrogliomas, OA represents Oligoastrocytomas, and GBM represents Glioblastoma) and b) TERT promoter / IDH1/2 mutation status for all Grade III and Grade IV gliomas analyzed in this study.

References

    1. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro-oncology. 2012;14(Suppl 5):v1–49.
    1. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta neuropathologica. 2007;114(2):97–109.
    1. Scherer HJ. Cerebral Astrocytomas and Their Derivatives. The American Journal of Cancer. 1940;40(2):159–198.
    1. Wen PY, Kesari S. Malignant gliomas in adults. The New England journal of medicine. 2008;359(5):492–507.
    1. Cancer Genome Atlas Research N. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–1068.
    1. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807–1812.
    1. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, Beroukhim R, Bernard B, Wu CJ, Genovese G, Shmulevich I, Barnholtz-Sloan J, et al. The Somatic Genomic Landscape of Glioblastoma. Cell. 2013;155(2):462–477.
    1. Jiao Y, Killela PJ, Reitman ZJ, Rasheed AB, Heaphy CM, de Wilde RF, Rodriguez FJ, Rosemberg S, Oba-Shinjo SM, Nagahashi Marie SK, Bettegowda C, Agrawal N, Lipp E, Pirozzi C, Lopez G, He Y, et al. Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas. Oncotarget. 2012;3(7):709–722.
    1. Johnson BE, Mazor T, Hong C, Barnes M, Aihara K, McLean CY, Fouse SD, Yamamoto S, Ueda H, Tatsuno K, Asthana S, Jalbert LE, Nelson SJ, Bollen AW, Gustafson WC, Charron E, et al. Mutational Analysis Reveals the Origin and Therapy-Driven Evolution of Recurrent Glioma. Science. 2013
    1. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, Misra A, Nigro JM, Colman H, Soroceanu L, Williams PM, Modrusan Z, Feuerstein BG, Aldape K. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer cell. 2006;9(3):157–173.
    1. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O'Kelly M, Tamayo P, Weir BA, Gabriel S, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer cell. 2010;17(1):98–110.
    1. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, Friedman H, Friedman A, Reardon D, Herndon J, Kinzler KW, Velculescu VE, et al. IDH1 and IDH2 mutations in gliomas. The New England journal of medicine. 2009;360(8):765–773.
    1. Griewank KG, Murali R, Schilling B, Scholz S, Sucker A, Song M, Susskind D, Grabellus F, Zimmer L, Hillen U, Steuhl KP, Schadendorf D, Westekemper H, Zeschnigk M. TERT promoter mutations in ocular melanoma distinguish between conjunctival and uveal tumours. British journal of cancer. 2013;109(2):497–501.
    1. Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, Kadel S, Moll I, Nagore E, Hemminki K, Schadendorf D, Kumar R. TERT promoter mutations in familial and sporadic melanoma. Science. 2013;339(6122):959–961.
    1. Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339(6122):957–959.
    1. Killela PJ, Reitman ZJ, Jiao Y, Bettegowda C, Agrawal N, Diaz LA, Jr, Friedman AH, Friedman H, Gallia GL, Giovanella BC, Grollman AP, He TC, He Y, Hruban RH, Jallo GI, Mandahl N, et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(15):6021–6026.
    1. Koelsche C, Sahm F, Capper D, Reuss D, Sturm D, Jones DT, Kool M, Northcott PA, Wiestler B, Bohmer K, Meyer J, Mawrin C, Hartmann C, Mittelbronn M, Platten M, Brokinkel B, et al. Distribution of TERT promoter mutations in pediatric and adult tumors of the nervous system. Acta neuropathologica. 2013
    1. Liu X, Wu G, Shan Y, Hartmann C, von Deimling A, Xing M. Highly prevalent TERT promoter mutations in bladder cancer and glioblastoma. Cell cycle. 2013;12(10):1637–1638.
    1. Vinagre J, Almeida A, Populo H, Batista R, Lyra J, Pinto V, Coelho R, Celestino R, Prazeres H, Lima L, Melo M, da Rocha AG, Preto A, Castro P, Castro L, Pardal F, et al. Frequency of TERT promoter mutations in human cancers. Nature communications. 2013;4:2185.
    1. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL, Shay JW. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266(5193):2011–2015.
    1. Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. European journal of cancer. 1997;33(5):787–791.
    1. Arita H, Narita Y, Fukushima S, Tateishi K, Matsushita Y, Yoshida A, Miyakita Y, Ohno M, Collins VP, Kawahara N, Shibui S, Ichimura K. Upregulating mutations in the TERT promoter commonly occur in adult malignant gliomas and are strongly associated with total 1p19q loss. Acta neuropathologica. 2013;126(2):267–276.
    1. Nonoguchi N, Ohta T, Oh JE, Kim YH, Kleihues P, Ohgaki H. TERT promoter mutations in primary and secondary glioblastomas. Acta neuropathologica. 2013
    1. Marian CO, Cho SK, McEllin BM, Maher EA, Hatanpaa KJ, Madden CJ, Mickey BE, Wright WE, Shay JW, Bachoo RM. The telomerase antagonist, imetelstat, efficiently targets glioblastoma tumor-initiating cells leading to decreased proliferation and tumor growth. Clinical cancer research: an official journal of the American Association for Cancer Research. 2010;16(1):154–163.
    1. Ruden M, Puri N. Novel anticancer therapeutics targeting telomerase. Cancer treatment reviews. 2013;39(5):444–456.
    1. Xia W, Wang P, Lin C, Li Z, Gao X, Wang G, Zhao X. Bioreducible polyethylenimine-delivered siRNA targeting human telomerase reverse transcriptase inhibits HepG2 cell growth in vitro and in vivo. Journal of controlled release: official journal of the Controlled Release Society. 2012;157(3):427–436.
    1. Zhang PH, Zou L, Tu ZG. RNAi-hTERT inhibition hepatocellular carcinoma cell proliferation via decreasing telomerase activity. J Surg Res. 2006;131(1):143–149.
    1. Ohgaki H, Kleihues P. Genetic alterations and signaling pathways in the evolution of gliomas. Cancer science. 2009;100(12):2235–2241.
    1. Bigner SH, Matthews MR, Rasheed BK, Wiltshire RN, Friedman HS, Friedman AH, Stenzel TT, Dawes DM, McLendon RE, Bigner DD. Molecular genetic aspects of oligodendrogliomas including analysis by comparative genomic hybridization. The American journal of pathology. 1999;155(2):375–386.
    1. Cairncross G, Jenkins R. Gliomas with 1p/19q codeletion: a.k.a. oligodendroglioma. Cancer journal. 2008;14(6):352–357.
    1. Jenkins RB, Blair H, Ballman KV, Giannini C, Arusell RM, Law M, Flynn H, Passe S, Felten S, Brown PD, Shaw EG, Buckner JC. A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer research. 2006;66(20):9852–9861.
    1. Smith JS, Perry A, Borell TJ, Lee HK, O'Fallon J, Hosek SM, Kimmel D, Yates A, Burger PC, Scheithauer BW, Jenkins RB. Alterations of chromosome arms 1p and 19q as predictors of survival in oligodendrogliomas, astrocytomas, and mixed oligoastrocytomas. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2000;18(3):636–645.
    1. Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, Mandelker D, Leary RJ, Ptak J, Silliman N, Szabo S, Buckhaults P, Farrell C, Meeh P, Markowitz SD, Willis J, et al. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314(5797):268–274.
    1. Reifenberger J, Reifenberger G, Liu L, James CD, Wechsler W, Collins VP. Molecular genetic analysis of oligodendroglial tumors shows preferential allelic deletions on 19q and 1p. The American journal of pathology. 1994;145(5):1175–1190.

Source: PubMed

Подписаться