Abstract
The current research in oncology is focused on genetics and molecular oncology in order to obtain better understanding of the etiology of tumor disease. Detailed knowledge of oncogenesis mechanisms could lead to invention of effective therapeutic tools against cancer. Under healthy conditions, cell cycle is regulated by oncogenes and tumor suppressor genes, which should be in strict balance. Genesis of tumor is a consequence of the accumulation of genetic alterations, which help the cell to escape normal cellular regulatory mechanisms and destruction by immune system. Glioblastoma (GBM) is a highly malignant primary brain tumor occurring mostly in population of adults. Patients suffering from GBM have very poor prognosis. Despite development in radiology methods promising earlier diagnosis and development of clinical and radiation oncology with newer treatment regimes, the effect of therapy remains limited and prognosis of patients has not improved as expected. Target of GBM research are genes involved in response to oxidative stress and DNA damage, genes regulating cell cycle, genes determining immune response, growth factors, and others. Genetic alterations are studied in connection to their possible relationship to susceptibility of brain tissue for tumor formation, to sensitivity of brain tissue for various environmental etiology factors, to effect of anticancer treatment or resistance of tumor tissue to therapy, to overall survival, and progression-free interval.
Keywords
- glioblastoma
- genes
- alteration
- mutations
- genetic pathways
1. Introduction
Glioblastoma (GBM) is a brain tumor of neuroectodermal origin. It is a second most common primary brain neoplasm and the most common from malignant brain tumors. This tumor arises from neural stem cells (NSCs), progenitor cells, dedifferentiated mature neural cells or neuroepithelial stem cells that transform into cancer stem cells (CSCs) or glioblastoma stem (GSC) or stem-like cells [1]. Stem cells have a high potential of self-renewal and differentiation. GBM is a tumor with highest degree of anaplasia within gliomas. It is classified as grade IV according to the WHO classification of brain tumors from 2007. This lesion has a rapid growth, is unbounded, infiltrates surrounding brain tissue, but rarely metastasizes. When metastases occur, it is usually within the central nervous system. Typical histopathological features of this tumor are cells of recognizably astrocyte origin, but displaying cellular pleomorphism with multinucleation, frequent mitoses, and areas of necrosis surrounded by palisading nuclei (increased tumor cell density) and endothelial proliferation as a manifestation of cellular hyperplasia with numerous clusters of blood vessels forming so-called glomeruloid formations. GBM cells spread along tracts in white matter infiltrate cerebrospinal fluid and vessels between meningeal layers. Invasion of GBM cells begins with the degradation of surrounding matrix proteins by proteases and proteinases. Movement of tumor cells through surrounding brain tissue requires receptor turnover, formation and degradation of focal adhesion molecules and rearrangement of cytoskeleton components. These changes are consequences of genetic alterations, such as overexpression, amplification, deletion or mutation in focal adhesion kinase and phosphatidylinositol 3-kinase (PI3K) pathways [2] and mainly caused by activation of growth factors and their receptors (integrins and protein deleted in colorectal cancer (DCC), hyaluron receptors CD44, RHAMM, BEHAB, ontogenetic protein SPARC, receptors for platelet-derived growth factor (PDGF), transforming growth factor (TGF)-α, epidermal growth factor (EGF), and basic fibroblast growth factor (bFGF)). Some components of extracellular matrix such as laminin or fibronectin are overexpressed in GBM and their silencing reduces invasiveness of GBM cells [3]. Besides microscopic characteristics, GBM is connected with a huge amount of genetic alterations causing higher proliferation, migration, and invasiveness of tumor cells. The intrinsic ability of GBM cells to invade normal brain tissue impedes complete surgical resection and predictably results in early local recurrence and mortality. Thoroughgoing explanation of GBM genetic pathways with their gene alterations is necessary for choosing of more suitable therapy and in predicting patient prognosis. This chapter offers an overview of the most common genetic alterations occurring in GBM.
2. Classification of GBM
According to the WHO classification of brain tumors based on histopathological origin, GBM is a primary brain tumor of neuroepithelial, glial origin. Ranking of GBM in clinical and prognostic significance is within the highest grade IV. GBM could also be classified according to mode of occurrence, as it could be a result of progression from less malignant glial tumor (so-called secondary type) or it could occur de novo (primary type). Another specific type is pediatric GBM. The most common is primary type. Primary, secondary, and pediatric GBMs have their own specific genetic and epigenetic alterations occurring in their gliomagenesis (see Figure 1). As diagnosis of GBM presents with heterogeneity of altered genetic pathways evidenced by The Cancer Genome Atlas Research Network’s study, classification based on gene expression profile distinguishes a classical, mesenchymal, proneural, and neural type of GBM. Classical type typically harbors no TP53 mutations, but very high rate of epidermal growth factor receptor (EGFR) mutations. This type has a slightly better survival. Mesenchymal type displays frequent mutations of neurofibromatosis gene 1 (NF1), TP53, and PTEN genes and aggressive anticancer therapy brings a significant increase in survival of these patients. Third, proneural subtype presents with highest mutation rate of TP53, PDGFRA, and isocitrate dehydrogenase (IDH) and usually affects younger adults. Last, neural type occurs more often in older population and contains several gene mutations at an average rate [4]. These classifications show high heterogeneity of genetic profile, aggressiveness, clinical characteristics, and expected prognosis of GBM patients with a strong need of definite uniform classification of subtypes leading to more specific treatment for each GBM subtype.
3. Most frequent genetic alterations of GBM
As mentioned above, alterations of oncogenes and tumor suppressor genes could deflect cell from normal cell cycle. Accumulation of genetic alterations of these genes initiates oncogenesis. In oncogenes, lesion of one allele is sufficient to cause mutation. Activation of oncogene causes avoidance of apoptosis and cell further proliferates. Amplification and activating mutation are the most common genetic alteration of oncogenes. On the other hand, lesion of both alleles is required to evoke mutation in tumor suppressor genes. These genes and their protein products when activated inhibit cell proliferation. Frequently observed alterations are deletion and inhibitory mutation.
During years of genetic research, there have been established three critical genetic pathways whose alterations lead to the formation of GBM: inactivation of p53 and retinoblastoma (RB) pathways (see Figure 2), activation of the PI3K pathway, and deregulation of growth factor (receptor tyrosine kinase—RTK) signaling (see Figure 3). TP53 signaling pathway is altered in 87% of GBMs, mostly affecting p53, murine double minute-2 (MDM2), MDM4, and cyclin-dependent kinase (CDK)N2A genes. Approximately 78% of GBMs harbor RB signaling disruption with most frequently altered genes: RB1, CDK4, CDK6, CCND2, and cyclin-dependent kinase inhibitor 2 (CDKN2) family. Finally, RTK/RAS/PI3K activation was found in 88% of tumors, affecting usually NF1, PIK3R1, and PIK3CA genes.
GBM is classified as primary or secondary according to altered genes. Primary GBM is a tumor with de novo formation. On the other hand, GBM as a result of malignant transformation of lower-grade glial tumor is called secondary [1]. Also, clinical manifestation and age of diagnosis vary within these two types. Typical for primary GBM is very short anamnesis and age over 50. On the contrary, patients with secondary GBM usually have previous anamnesis of lower grade glioma and younger age depending on the age at the time of diagnosis of preexisting lower grade glioma. Both – primary and secondary GBM – have their own specific gene alterations occurring in their gliomagenesis (see Figure 1). Typical alterations for primary GBM are homozygous deletion or mutation of
Gene
Oncogene
Gene
On 9p21 are located genes
Protein
Genes
Gene phosphatase and tensin homology –
Signal transducer and activator of transcription 3 –
Oncogene
Paternally expressed gene 3 –
Vascular endothelial growth factor –
Proto-oncogene
Gene
The B-Raf proto-oncogene serine/threonine kinase –
H3 histone, family 3A –
Telomerase reverse transcriptase –
Tumor suppressor
4. Conclusion
GBM is the most aggressive and devastating primary brain tumor with very grim prognosis. The patient’s survival rarely exceeds a year and a half with all accessible therapy used. Only 3% of GBM patients have survival over 5 years. This fact makes this tumor a very severe diagnosis directly affecting life expectancy of the patient and quality of his/her life. Explanation of its genesis could bring us closer to invention of effective treatment and genetic profile of concrete GBM tissue would help to design the individualized therapy management for each patient. Understanding the genetic and epigenetic characterization could help to distinguish various GBM subgroups, indistinguishable by histological appearance, but classified according to molecular and genetic alterations. This could lead to establishment of GBM classification with clinical impact, subgroup-specific treatment, and better design of future trials. The aim was to achieve prolonged progression-free interval and overall survival with maintaining satisfactory quality of life. It is very important if concrete genetic alteration is connected to tumor formation or if it is prognostic factor. Other factors influencing prognosis are histological type and tumor grade, age of patient, Karnofsky performance score at the time of diagnosis, extend of surgical resection, tumor localization, and appropriate therapeutic management. Tumor localization and extend of its surgical resection influence progression-free interval as well as overall survival. Detailed and complete explanation and discovery of altered genes and whole genetic pathways of GBM is the basis for new distinctive GBM classification. Such a new tumor division could bring us closer to routine genetic examination of frequently altered genetic pathways from tumor sample and aiming of therapy directly against specific genetic and epigenetic targets. Such individualized therapy could decrease the number of adverse effects, but first of all, hopefully, would finally ameliorate survival and life quality of patients, suffering from this severe disease.
Acknowledgments
This work was supported by projects: Competence Center for Research and Development in the diagnosis and therapy of oncological diseases, ITMS 26220220153 and The application of PACS (Picture Archiving and Communication System) in the reasearch and development, ITMS 26210120004.
References
- 1.
Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, Delattre JY: Primary brain tumours in adults. Lancet. 2012; 379 :1984–1996. DOI: 10.1016/S0140-6736(11)61346-9 - 2.
Michael KE, Dumbauld DW, Burns KL, Hanks SK, Garcia AJ: Focal adhesion kinase modulates cell adhesion strengthening via integrin activation. Mol Biol Cell. 2009; 20 :2508–2519. DOI: 10.1091/mbc.E08-01-0076 - 3.
Sengupta SNS, Hindi ES, Wainwright DA, Han Y, Lesniak MS: Short hairpin RNA-mediated fibronectin knockdown delays tumor growth in a mouse glioma model. Neoplasia. 2010; 12 :837–847. - 4.
Cancer Genome Atlas Research Network: Comprehensive genomic characterization defines human GBM genes and core pathways. Nature. 2008; 455: 1061–1068. DOI: 10.1038/nature07385 - 5.
Iacob G, Dinca EB: Current data and strategy in GBM multiforme. J Med Life. 2009; 2 :386–393. - 6.
Hede SM, Nazarenko I, Nistér M, Lindström MS: Novel perspectives on p53 function in neural stem cells and brain tumors. J Oncol. 2011;2011:852970. DOI: 10.1155/2011/852970 - 7.
Zhao Z, Xu Y: p53 and stem cells: new developments and new concerns. Trends Cell Biol. 2010; 20 :170–175. DOI: 10.1016/j.tcb.2009.12.004 - 8.
Krakstad C, Chekenya M: Survival signaling and apoptosis resistance in GBMs: opportunities for targeted therapeutics. Mol Cancer. 2010; 9 :135–149. DOI: 10.1186/1476-4598-9-135 - 9.
Wade M, Wang YV, Wahl GM: The p53 orchestra: Mdm2 and Mdmx set the tone. Trends Cell Biol. 2010; 20 :299–309. DOI: 10.1016/j.tcb.2010.01.009 - 10.
Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, Chin L, DePinho RA, Cavenee WK: Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 2007; 21 :2683–2710. DOI: 10.1101/gad.1596707 - 11.
Thomasova D, Mulay SR, Bruns H, Anders HJ: p53-independent roles of MDM2 in NF-κB signaling: implications for cancer therapy, wound healing, and autoimmune diseases. Neoplasia. 2012; 14 :1097–1101. - 12.
Wang AL, Liu ZX, Li G, Zhang LW: Expression and significance of P53 protein and MDM-2 protein in human gliomas. Chin Med J (Engl). 2011; 124 :2530–2533. - 13.
Wang CL, Wang JY, Liu ZY, Ma XM, Wang XW, Jin H, Zhang XP, Fu D, Hou LJ, Lu YC: Ubiquitin-specific protease 2a stabilizes MDM4 and facilitates the p53-mediated intrinsic apoptotic pathway in GBM. Carcinogenesis. 2014; 35 :1500–1509. DOI: 10.1093/carcin/bgu015 - 14.
Goldhoff P, Clarke J, Smirnov I, Berger MS, Prados MD, James CD, Perry A, Phillips JJ: Clinical stratification of GBM based on alterations in retinoblastoma tumor suppressor protein (RB1) and association with the proneural subtype. J Neuropathol Exp Neurol. 2012; 71 :83–89. DOI: 10.1097/NEN.0b013e31823fe8f1 - 15.
Chow LM, Endersby R, Zhu X, Rankin S, Qu C, Zhang J, Broniscer A, Ellison DW, Baker SJ: Cooperativity within and among Pten, p53, and Rb pathways induces high-grade astrocytoma in adult brain. Cancer Cell. 2011; 19 :305–316. DOI: 10.1016/j.ccr.2011.01.039 - 16.
Wiedemeyer WR, Dunn IF, Quayle SN, Zhang J, Chheda MG, Dunn GP, Zhuang L, Rosenbluh J, Chen S, Xiao Y, Shapiro GI, Hahn WC, Chin L: Pattern of retinoblastoma pathway inactivation dictates response to CDK4/6 inhibition in GBM. Proc Natl Acad Sci USA. 2010; 107 :11501–11506. DOI: 10.1073/pnas.1001613107 - 17.
Kim CS, Jung S, Jung TY, Jang WY, Sun HS, Ryu HH: Characterization of invading glioma cells using molecular analysis of leading-edge tissue. J Korean Neurosurg Soc. 2011; 50 :157–165. DOI: 10.3340/jkns.2011.50.3.157 - 18.
Walker C, Baborie A, Crooks D, Wilkins S, Jenkinson MD: Biology, genetics and imaging of glial cell tumours. Br J Radiol. 2011; 84 :90–106. DOI: 10.1259/bjr/23430927 - 19.
Yue WY, Sai K, Wu QL, Xia YF, Yu SH, Chen ZP: Long-term molecular changes in WHO grade II astrocytomas following radiotherapy. Chin J Cancer. 2012; 31 :159–165. DOI: 10.5732/cjc.011.10149 - 20.
Liu Y, Shete S, Hosking FJ, Robertson LB, Bondy ML, Houlston RS: New insights into susceptibility to glioma. Arch Neurol. 2010; 67 :275–278. DOI: 10.1001/archneurol.2010.4 - 21.
Weller M, Felsberg J, Hartmann C, Berger H, Steinbach JP, Schramm J, Westphal M, Schackert G, Simon M, Tonn JC, Heese O, Krex D, Nikkhah G, Pietsch T, Wiestler O, Reifenberger G, von Deimling A, Loeffler M: Molecular predictors of progression-free and overall. Survival in patients with newly diagnosed GBM: a prospective translational study of the German glioma network. J Clin Oncol. 2009; 27 :5743–5750. DOI: 10.1200/JCO.2009.23.0805 - 22.
Shete S, Hosking FJ, Robertson LB, Dobbins SE, Sanson M, Malmer B, Simon M, Marie Y, Boisselier B, Delattre JY, Hoang-Xuan K, El Hallani S, Idbaih A, Zelenika D, Andersson U, Henriksson R, Bergenheim AT, Feychting M, Lönn S, Ahlbom A, Schramm J, Linnebank M, Hemminki K, Kumar R, Hepworth SJ, Price A, Armstrong G, Liu Y, Gu X, Yu R, Lau C, Schoemaker M, Muir K, Swerdlow A, Lathrop M, Bondy M, Houlston RS: Genome-wide association study identifies five susceptibility loci for glioma. Nat Genet. 2009; 41 :899–904. DOI: 10.1038/ng.407 - 23.
Simon M, Hosking FJ, Marie Y, Gousias K, Boisselier B, Carpentier C, Schramm J, Mokhtari K, Hoang-Xuan K, Idbaih A, Delattre JY, Lathrop M, Robertson LB, Houlston RS, Sanson M: Genetic risk profiles identify different molecular etiologies for glioma. Clin Cancer Res. 2010; 16 :5252–5259. DOI: 10.1158/1078-0432.CCR-10-1502 - 24.
Zerrouqi A, Pyrzynska B, Febbraio M, Brat DJ, Van Meir EG: P14ARF inhibits human GBM-induced angiogenesis by upregulating the expression of TIMP3. J Clin Invest. 2012; 122 :1283–1295. DOI: 10.1172/JCI38596 - 25.
Solomon DA, Kim JS, Jenkins S, Ressom H, Huang M, Coppa N, Mabanta L, Bigner D, Yan H, Jean W, Waldman T: Identification of p18 INK4c as a tumor suppressor gene in GBM multiforme. Cancer Res. 2008; 68 :2564–2569. DOI: 10.1158/0008-5472.CAN-07-6388 - 26.
Koyama-Nasu R, Nasu-Nishimura Y, Todo T, Ino Y, Saito N, Aburatani H, Funato K, Echizen K, Sugano H, Haruta R, Matsui M, Takahashi R, Manabe E, Oda T, Akiyama T: The critical role of cyclin D2 in cell cycle progression and tumorigenicity of GBM stem cells. Oncogene. 2013; 32 :3840–3845. DOI: 10.1038/onc.2012.399 - 27.
Courtney KD, Corcoran RB, Engelman JA: The PI3K pathway as drug target in human cancer. J Clin Oncol. 2010; 28 :1075–1083. DOI: 10.1200/JCO.2009.25.3641 - 28.
Wen PY, Lee EQ, Reardon DA, Ligon KL, Alfred Yung WK: Current clinical development of PI3K pathway inhibitors in GBM. Neuro Oncol. 2012; 14 :819–829. DOI: 10.1093/neuonc/nos117. - 29.
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, Busam DA, Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW: An integrated genomic analysis of human GBM multiforme. Science. 2008; 321 :1807–1812. DOI: 10.1126/science.1164382 - 30.
Varghese RT, Liang Y, Guan T, Franck CT, Kelly DF, Sheng Z: Survival kinase genes present prognostic significance in glioblastoma. Oncotarget. 2016;7:20140-20151. DOI: 10.18632/oncotarget.7917 - 31.
Sun MHP, Hofmann BT, Hart JR, Vogt PK: Cancer derived mutations in the regulatory subunit p85alpha of phosphoinositide 3-kinase function through the catalytic subunit p110alpha. Proc Natl Acad Sci USA. 2010; 107 :15,547–15,552. DOI: 10.1073/pnas.1009652107 - 32.
Weber GL, Parat MO, Binder ZA, Gallia GL, Riggins GJ: Abrogation of PIK3CA or PIK3R1 reduces proliferation, migration, and invasion in GBM multiforme cells. Oncotarget. 201; 2 :833–849. - 33.
Lee JJ, Kim BC, Park MJ, Lee YS, Kim YN, Lee BL, Lee JS: PTEN status switches cell fate between premature senescence and apoptosis in glioma exposed to ionizing radiation. Cell Death Differ. 2011; 18 :666–677. DOI: 10.1038/cdd.2010.139 - 34.
Sherry MM, Reeves A, Wu JK, Cochran BH: STAT3 is required for proliferation and maintenance of multipotency in GBM stem cells. Stem Cells. 2009; 27 :2383–2392. DOI: 10.1002/stem.185 - 35.
Turner KM, Sun Y, Ji P, Granberg KJ, Bernard B, Hu L, Cogdell DE, Zhou X, Yli-Harja O, Nykter M, Shmulevich I, Yung WK, Fuller GN, Zhang W: Genomically amplified Akt3 activates DNA repair pathway and promotes glioma progression. Proc Natl Acad Sci USA. 2015; 112 :3421–3426. DOI: 10.1073/pnas.1414573112 - 36.
Omuro AM, Faivre S, Raymond E: Lessons learned in the development of targeted therapy for malignant gliomas. Mol Cancer Ther. 2007; 6 :1909–1919. - 37.
Easton RM, Cho H, Roovers K, Shineman DW, Mizrahi M, Forman MS, Lee VM, Szabolcs M, de Jong R, Oltersdorf T, Ludwig T, Efstratiadis A, Birnbaum MJ: Role for Akt3/protein kinase Bgamma in attainment of normal brain size. Mol Cell Biol. 2005; 25 :1869–1878. - 38.
Mure H, Matsuzaki K, Kitazato KT, Mizobuchi Y, Kuwayama K, Kageji T, Nagahiro S: Akt2 and Akt3 play a pivotal role in malignant gliomas. Neuro Oncol. 2010; 12 :221–232. DOI: 10.1093/neuonc/nop026 - 39.
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, Winckler W, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT, Wilson RK, Speed TP, Gray JW, Meyerson M, Getz G, Perou CM, Hayes DN; Cancer Genome Atlas Research Network: Integrated genomic analysis identifies clinically relevant subtypes of GBM characterized by abnormalities in PDGFRA, IDH1, EGFR and NF1. Cancer Cell. 2010; 17 :98–110. DOI: 10.1016/j.ccr.2009.12.020 - 40.
Faria MH, Gonçalves BP, do Patrocínio RM, de Moraes-Filho MO, Rabenhorst SH: Expression of Ki-67, topoisomerase II alpha and c-MYC in astrocytic tumors: correlation with the histopathological grade and proliferative status. Neuropathology. 2006; 26 :519–527. - 41.
Cenci T, Martini M, Montano N, D'Alessandris QG, Falchetti ML, Annibali D, Savino M, Bianchi F, Pierconti F, Nasi S, Pallini R, Larocca LM: Prognostic relevance of c-Myc and BMI1 expression in patients with glioblastoma. Am J Clin Pathol. 2012; 138 :390–396. DOI: 10.1309/AJCPRXHNJQLO09QA - 42.
De Salvo M, Maresca G, D’agnano I, Marchese R, Stigliano A, Gagliassi R, Brunetti E, Raza GH, De Paula U, Bucci B: Temozolomide induced c-Myc-mediated apoptosis via Akt signaling in MGMT expressing glioblastoma cells. Int J Radiat Biol. 2011; 87 :518–533. DOI: 10.3109/09553002.2011.556173 - 43.
Shakhova O, Leung C, Marino S: Bmi1 in development and tumorigenesis of the central nervous system. J Mol Med. 2005; 83 :596–600. - 44.
Guo WJ, Datta S, Band V, Dimri GP: Mel-18, a polycomb group protein, regulates cell proliferation and senescence via transcriptional repression of Bmi-1 and c-Myc oncoproteins. Mol Biol Cell. 2007; 18 :536–546. - 45.
Pusch S, Sahm F, Meyer J, Mittelbronn M, Hartmann C, von Deimling A: Glioma IDH1 mutation patterns off the beaten track. Neuropathol Appl Neurobiol. 2011; 37 :428–430. DOI: 10.1111/j.1365-2990.2010.01127.x - 46.
Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, Yu W, Li Z, Gong L, Peng Y, Ding J, Lei Q, Guan KL, Xiong Y: Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha. Science. 2009; 324 :261–265. DOI: 10.1126/science.1170944 - 47.
Toedt G, Barbus S, Wolter M, Felsberg J, Tews B, Blond F, Sabel MC, Hofmann S, Becker N, Hartmann C, Ohgaki H, von Deimling A, Wiestler OD, Hahn M, Lichter P, Reifenberger G, Radlwimmer B: Molecular signatures classify astrocytic gliomas by IDH1 mutation status. Int J Cancer. 2011; 128 :1095–1103. DOI: 10.1002/ijc.25448 - 48.
Kim W, Liau LM: IDH mutations in human glioma. Neurosurg Clin N Am. 2012; 23 :471–480. DOI: 10.1016/j.nec.2012.04.009 - 49.
Yan H, Parsons DW, Jin G, McLendon R, Rasheed A, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, Friedman H, Friedman A, Reardon D, Herndon J, Kinzler KW, Velculescu VE, Vogelstein B, Bigner DD: IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009; 360 :765–773. DOI: 10.1056/NEJMoa0808710 - 50.
Ichimura K, Pearson DM, Kocialkowski S, Bäcklund LM, Chan R, Jones DT, Collins VP: IDH1 mutations are present in the majority of common adult gliomas but rare in primary GBMs. Neuro Oncol. 2009; 11 :341–347. DOI: 10.1215/15228517-2009-025 - 51.
Reitman ZJ, Yan H: Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J Natl Cancer Inst. 2010; 102 :932–941. DOI: 10.1093/jnci/djq187 - 52.
SongTao Q, Lei Y, Si G, YanQing D, HuiXia H, XueLin Z, LanXiao W, Fei Y: IDH mutations predict longer survival and response to temozolomide in secondary GBM. Cancer Sci. 2012; 103 :269–273. DOI: 10.1111/j.1349-7006.2011.02134.x - 53.
Nobusawa S, Watanabe T, Kleihues P, Ohgaki H: IDH1 mutations as molecular signature and predictive factor of secondary GBMs. Clin Cancer Res. 2009; 15 :6002–6007. DOI: 10.1158/1078-0432.CCR-09-0715 - 54.
Beiko J, Suki D, Hess KR, Fox BD, Cheung V, Cabral M, Shonka N, Gilbert MR, Sawaya R, Prabhu SS, Weinberg J, Lang FF, Aldape KD, Sulman EP, Rao G, McCutcheon IE, Cahill DP: IDH1 mutant malignant astrocytomas are more amenable to surgical resection and have a survival benefit associated with maximal surgical resection. Neuro-Oncology. 2014; 16 :81–91. DOI: 10.1093/neuonc/not159 - 55.
Reitman ZJ, Yan H: Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J Natl Cancer Inst. 2010; 102 :932–941. DOI: 10.1093/jnci/djq187 - 56.
Takahashi Y, Nakamura H, Makino K, Hide T, Muta D, Kamada H, Kuratsu J: Prognostic value of isocitrate dehydrogenase 1, O6-methylguanine-DNA methyltransferase promoter methylation, and 1p19q co-deletion in Japanese malignant glioma patients. World J Surg Oncol. 2013; 11 :284. DOI: 10.1186/1477-7819-11-284 - 57.
Rohle D, Popovici-Muller J, Palaskas N, Turcan S, Grommes C, Campos C, Tsoi J, Clark O, Oldrini B, Komisopoulou E, Kunii K, Pedraza A, Schalm S, Silverman L, Miller A, Wang F, Yang H, Chen Y, Kernytsky A, Rosenblum MK, Liu W, Biller SA, Su SM, Brennan CW, Chan TA, Graeber TG, Yen KE, Mellinghoff IK: An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science. 2013; 340 :626–630. DOI: 10.1126/science.1236062 - 58.
Rajasekharan S, Kennedy TE: The netrin protein family. Genome Biol. 2009; 10 :239. DOI: 10.1186/GBM-2009-10-9-239 - 59.
Jarjour AA, Durko M, Luk TL, Marçal N, Shekarabi M, Kennedy TE: Autocrine netrin function inhibits glioma cell motility and promotes focal adhesion formation. PLoS One. 2011; 6 :e25408. DOI:10.1371/journal.pone.0025408 - 60.
Jiang X, Yu Y, Yang HW, Agar NY, Frado L, Johnson MD: The imprinted gene PEG3 inhibits Wnt signaling and regulates glioma growth. J Biol Chem. 2010; 285 :8472–8480. DOI: 10.1074/jbc.M109.069450 - 61.
Otsuka S, Maegawa S, Takamura A, Kamitani H, Watanabe T, Oshimura M, Nanba E: Aberrant promoter methylation and expression of the imprinted PEG3 gene in glioma. Proc Jpn Acad Ser B. 2009; 85 :157–165. DOI: 10.2183/pjab.85.157 - 62.
Kilburn L, Okcu F, Wang T, Cao Y, Renfro-Spelman A, Aldape KD, Gilbert MR, Bondy M: Glutathione S-transferase polymorphism are associated with survival in anaplastic glioma patients. Cancer. 2010; 116 :2242–2249. DOI: 10.1002/cncr.25006 - 63.
Lo H, Ali-Osman F: Genetic polymorphism and function of glutathione S-transferases in tumor drug resistance. Curr Opin Pharmacol. 2007; 7 :367–374. DOI: 10.1016/j.coph.2007.06.009 - 64.
Schwartzbaum JA, Ahlbom A, Lönn S, Warholm M, Rannug A, Auvinen A, Christensen HC, Henriksson R, Johansen C, Lindholm C, Malmer B, Salminen T, Schoemaker MJ, Swerdlow AJ, Feychting M: An international casecontrol study of glutathione transferase and functionally related polymorphisms and risk of primary adult brain tumors. Cancer Epidemiol Biomarkers Prev. 2007; 16 :559–565. DOI: 10.1158/1055-9965.EPI-06-0918 - 65.
Sjöström S, Anderson U, Liu Y, Brännström T, Broholm H, Johansen C, Collatz-Laier H, Henriksson R, Bondy M, Melin B: Genetic variation in EGF and EGFR and GBM outcome. Neuro-Oncol. 2010; 12 :815–821. DOI:10.1093/neuonc/noq018 - 66.
Chen X, Yang G, Zhang D, Zhang W, Zou H, Zhao H, Zhang X, Zhao S: Association between the epidermal growth factor +61 G/A polymorphism and glioma risk: a meta-analysis. PLoS One. 2014; 9 :e95139. DOI: 10.1371/journal.pone.0095139 - 67.
Pinto GR, Yoshioka FK, Clara CA, Santos MJ, Almeida JR, Burbano RR, Rey JA, Casartelli C: Association study of an epidermal growth factor gene functional polymorphism with the risk and prognosis of gliomas in Brazil. Int J Biol Markers. 2009; 24 :277–281. - 68.
Hu X, Miao W, Zou Y, Zhang W, Zhang Y, Liu H: Expression of p53, epidermal growth factor receptor, Ki-67 and O6-methylguanine-DNA methyltransferase in human gliomas. Oncol Lett. 2013; 6 :130–134. - 69.
Ozawa T, Brennan CW, Wang L, Squatrito M, Sasayama T, Nakada M, Huse JT, Pedraza A, Utsuki S, Yasui Y, Tandon A, Fomchenko EI, Oka H, Levine RL, Fujii K, Ladanyi M, Holland EC: PDGFRA gene rearrangements are frequent genetic events in PDGFRA-amplified GBMs. Genes Dev. 2010; 24 :2205–2218. DOI: 10.1101/gad.1972310 - 70.
Kim Y, Kim E, Wu Q, Guryanova O, Hitomi M, Lathia JD, Serwanski D, Sloan AE, Weil RJ, Lee J, Nishiyama A, Bao S, Hjelmeland AB, Rich JN: Platelet-derived growth factor receptors differentially inform intertumoral and intratumoral heterogeneity. Genes Dev. 2012; 26 :1247–1262. DOI: 10.1101/gad.193565.112 - 71.
Westermark B: Platelet-derived growth factor in GBM-driver or biomarker? Ups J Med Sci. 2014; 119 :298–305. DOI: 10.3109/03009734.2014.970304 - 72.
Lönn S, Rothman N, Shapiro WR, Fine HA, Selker RG, Black PM, Loeffler JS, Hutchinson AA, Inskip PD: Genetic variation in insulin-like growth factors and brain tumor risk. Neuro-Oncol. 2008; 10 :553–559. DOI: 10.1215/15228517-2008-026 - 73.
Pardo OE, Wellbrock C, Khanzada UK, Aubert M, Arozarena I, Davidson S, Bowen F, Parker PJ, Filonenko VV, Gout IT, Sebire N, Marais R, Downward J, Seckl MJ: FGF-2 protects small cell lung cancer cells from apoptosis through a complex involving PKCepsilon, B-Raf and S6K2. The EMBO Journal. 2006; 25 :3078–3088. - 74.
Haley EM, Kim Y: The role of basic fibroblast growth factor in GBM multiforme and GBM stem cells and in their in vitro culture. Cancer Lett. 2014; 346 :1–5. DOI: 10.1016/j.canlet.2013.12.003 - 75.
Desjardins A, Friedman HS: Bevacizumab therapy for GBM: a passionate discussion. CNS Oncol. 2014; 3 :1–3. DOI: 10.2217/cns.13.53 - 76.
Wang K, Wang YY, Wang JF, Ma J, Jiang T, Dai JP: Radiologic features and expression of vascular endothelial growth factor stratify survival outcomes in patients with GBM. AJNR Am J Neuroradiol. 2016;37:629-635. DOI:10.3174/ajnr.A4567 - 77.
Santibanez JF, Quintanilla M, Bernabeu C: TGF-beta/ TGF-beta receptor system and its role in physiological and pathological conditions. Clin Sci (Lond). 2011; 121 :233–251. DOI: 10.1042/CS20110086 - 78.
Weiss A, Attisano L: The TGFbeta superfamily signaling pathway. Wiley Interdisciplinary Reviews. Developmental Biol. 2013; 2 :47–63. DOI: 10.1002/wdev.86 - 79.
Kaminska B, Kocyk M, Kijewska M: TGF beta signaling and its role in glioma pathogenesis. Adv Exp Med Biol. 2013; 986 :171–187. DOI: 10.1007/978-94-007-4719-7_9 - 80.
Frei K, Gramatzki D, Tritschler I, Schroeder JJ, Espinoza L, Rushing EJ, Weller M: Transforming growth factor-β pathway activity in GBM. Oncotarget. 2015; 6 :5963–5977. - 81.
Organ SL, Tsao MS: An overview of the c-MET signaling pathway. Ther Adv Med Oncol. 2011; 3 :7–19. DOI: 10.1177/1758834011422556 - 82.
Kwak Y, Kim SI, Park CK, Paek SH, Lee ST, Park SH: C-MET overexpression and amplification in gliomas. Int J Clin Exp Pathol. 2015; 8 :14932–14938. - 83.
Kong DS, Song SY, Kim DH, Joo KM, Yoo JS, Koh JS, Dong SM, Suh YL, Lee JI, Park K, Kim JH, Nam DH: Prognostic significance of c-Met expression in GBMs. Cancer. 2009; 115 :140–148. DOI: 10.1002/cncr.23972 - 84.
Basilico C, Arnesano A, Galluzzo M, Comoglio PM, Michieli P: A high affinity hepatocyte growth factor-binding site in the immunoglobulin-like region of Met. J Biol Chem. 2008; 283 :21,267–21,277. DOI: 10.1074/jbc.M800727200 - 85.
Xie Q, Bradley R, Kang L, Koeman J, Ascierto ML, Worschech A, De Giorgi V, Wang E, Kefene L, Su Y, Essenburg C, Kaufman DW, DeKoning T, Enter MA, O'Rourke TJ, Marincola FM, Vande Woude GF: Hepatocyte growth factor (HGF) autocrine activation predicts sensitivity to MET inhibition in GBM. Proc Natl Acad Sci USA. 2012; 109 :570–575. DOI: 10.1073/pnas.1119059109 - 86.
Garcia-Navarrete R, Garcia E, Arrieta O, Sotelo J: Hepatocyte growth factor in cerebrospinal fluid is associated with mortality and recurrence of GBM, and could be of prognostic value. J Neurooncol. 2010; 97 :347–351. DOI: 10.1007/s11060-009-0037-8 - 87.
Watson LA, Goldberg H, Bérubé NG: Emerging roles of ATRX in cancer. Epigenomics. 2015; 7 :1365–13678. DOI: 10.2217/epi.15.82 - 88.
Liu XY, Gerges N, Korshunov A, Sabha N, Khuong-Quang DA, Fontebasso AM, Fleming A, Hadjadj D, Schwartzentruber J, Majewski J, Dong Z, Siegel P, Albrecht S, Croul S, Jones DT, Kool M, Tonjes M, Reifenberger G, Faury D, Zadeh G, Pfister S, Jabado N: Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations. Acta Neuropathol. 2012; 124 :615–625. DOI: 10.1007/s00401-012-1031-3 - 89.
Appin CL, Brat DJ: Molecular pathways in gliomagenesis and their relevance to neuropathologic diagnosis. Adv Anat Pathol. 2015; 22 :50–58. DOI: 10.1097/PAP.0000000000000048 - 90.
Kleinschmidt-DeMasters BK, Aisner DL, Birks DK, Foreman NK: Epithelioid GBMs show a high percentage of BRAF V600E mutation. Am J Surg Pathol. 2013; 37 :685–698. DOI: 10.1097/PAS.0b013e31827f9c5e - 91.
Schindler G, Capper D, Meyer J, Janzarik W, Omran H, Herold-Mende C, Schmieder K, Wesseling P, Mawrin C, Hasselblatt M, Louis DN, Korshunov A, Pfister S, Hartmann C, Paulus W, Reifenberger G, von Deimling A: Analysis of BRAF V600E mutation in 1320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuropathol. 2011; 121 :397–405. DOI: 10.1007/s00401-011-0802-6 - 92.
Gutmann DH: BRAF-V600E mutation in pediatric and adult GBM. Neuro-Oncology. 2014; 16 :318–319. DOI: 10.1093/neuonc/not146 - 93.
Takahashi Y, Akahane T, Sawada T, Ikeda H, Tempaku A, Yamauchi S, Nishihara H, Tanaka S, Nitta K, Ide W, Hashimoto I, Kamada H: Adult classical GBM with a BRAFV600E mutation. World J Surg Oncol. 2015; 13 :100. DOI: 10.1186/s12957-015-0521-x - 94.
Lewis PW, Elsaesser SJ, Noh KM, Stadler SC, Allis CD: Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci USA. 2010; 107 :14,075–14,080. DOI: 10.1073/pnas.1008850107 - 95.
Schwartzentruber J, Korshunov A, Liu XY, Jones DTW, Pfaff E, Jacob K, Sturm D, Fontebasso AM, Quang DA, Tonjes M, Hovestadt V, Albrecht S, Kool M, Nantel A, Konermann C, Lindroth A, Jäger N, Rausch T, Ryzhova M, Korbel JO, Hielscher T, Hauser P, Garami M, Klekner A, Bognar L, Ebinger M, Schuhmann MU, Scheurlen W, Pekrun A, Frühwald MC, Roggendorf W, Kramm C, Dürken M, Atkinson J, Lepage P, Montpetit A, Zakrzewska M, Zakrzewski K, Liberski PP, Dong Z, Siegel P, Kulozik AE, Zapatka M, Guha A, Malkin D, Felsberg J, Reifenberger G, von Deimling A, Ichimura K, Collins VP, Witt H, Milde T, Witt O, Zhang C, Castelo-Branco P, Lichter P, Faury D, Tabori U, Plass C, Majewski J, Pfister SM, Jabado N: Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric GBM. Nature. 2012; 482 :226–231. DOI: 10.1038/nature10833 - 96.
Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DT, Konermann C, Pfaff E, Tönjes M, Sill M, Bender S, Kool M, Zapatka M, Becker N, Zucknick M, Hielscher T, Liu XY, Fontebasso AM, Ryzhova M, Albrecht S, Jacob K, Wolter M, Ebinger M, Schuhmann MU, van Meter T, Frühwald MC, Hauch H, Pekrun A, Radlwimmer B, Niehues T, von Komorowski G, Dürken M, Kulozik AE, Madden J, Donson A, Foreman NK, Drissi R, Fouladi M, Scheurlen W, von Deimling A,Monoranu C, Roggendorf W, Herold-Mende C, Unterberg A, Kramm CM, Felsberg J, Hartmann C, Wiestler B, Wick W, Milde T, Witt O, Lindroth AM, Schwartzentruber J, Faury D, Fleming A, Zakrzewska M, Liberski PP, Zakrzewski K, Hauser P, Garami M, Klekner A, Bognar L, Morrissy S, Cavalli F, Taylor MD,van Sluis P, Koster J, Versteeg R, Volckmann R, Mikkelsen T, Aldape K, Reifenberger G, Collins VP, Majewski J, Korshunov A, Lichter P, Plass C, Jabado N, Pfister SM: Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of GBM. Cancer Cell. 2012; 22 :425–437. DOI: 10.1016/j.ccr.2012.08.024 - 97.
Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA: Highly recurrent TERT promoter mutations in human melanoma. Science. 2013; 339 :957–959. DOI: 10.1126/science.1229259 - 98.
Simon M, Hosen I, Gousias K, Rachakonda S, Heidenreich B, Gessi M, Schramm J, Hemminki K, Waha A, Kumar R: TERT promoter mutations: a novel independent prognostic factor in primary GBMs. Neuro Oncol. 2015; 17 :45–52. DOI: 10.1093/neuonc/nou158 - 99.
Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I, Janakiraman M, Schultz N, Hanrahan AJ, Pao W, Ladanyi M, Sander C, Heguy A, Holland EC, Paty PB, Mischel PS, Liau L, Cloughesy TF, Mellinghoff IK, Solit DB, Chan TA: Somatic mutations of the Parkinson's disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet. 2010; 42 :77–82. DOI: 10.1038/ng.491 - 100.
Lin DC, Xu L, Chen Y, Yan H, Hazawa M, Doan N, Said JW, Ding LW, Liu LZ, Yang H, Yu S, Kahn M, Yin D, Koeffler HP: Genomic and Functional Analysis of the E3 Ligase PARK2 in Glioma. Cancer Res. 2015; 75 :1815–1827. DOI: 10.1158/0008-5472.CAN-14-1433 - 101.
Von Deimling A, Korshunov A, Hartmann C: The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and IDH1 mutations. Brain Pathol. 2011; 21 :74–87. DOI: 10.1111/j.1750-3639.2010.00454.x - 102.
Glas M, Happold C, Rieger J, Wiewrodt D, Bähr O, Steinbach JP, Wick W, Kortmann RD, Reifenberger G, Weller M, Herrlinger U: Long-term survival of GBM patients treated with radiotherapy and lomustine plus temozolomide. J Clin Oncol. 2009; 27 :1257–1261. DOI: 10.1200/JCO.2008.19.2195