Cancer Letters xxx (2012) xxx–xxx
Contents lists available at SciVerse ScienceDirect
Cancer Letters journal homepage: www.elsevier.com/locate/canlet
Mini-review
Epigenetics in human gliomas Simone Kreth a, Niklas Thon b, Friedrich W. Kreth b,⇑ a b
Research Group of Molecular Medicine, Department of Anesthesiology, University of Munich (LMU), Germany Department of Neurosurgery, University of Munich (LMU), Germany
a r t i c l e
i n f o
Article history: Available online xxxx Keywords: Glioblastoma Epigenetic networks MicroRNAs Stereotactic biopsy
a b s t r a c t Aberrant epigenetic landscapes and their involvement in genesis and progression of tumors, as well as in treatment responses and prognosis, indicate one of the most emerging fields in cancer research. In gliomas, the most common human primary brain tumors, and in particular in glioblastoma, the most malignant and devastating brain tumor entity in adults, the elucidation of distinct patterns of aberrant DNA methylation, histone modification, and miRNA expression and their interrelationship has fundamentally changed our point of view on these highly heterogeneous tumors. In the current review article, we address the basic principles of epigenetic control in gliomas, their current and putative future role in prognostic and predictive models and possible interactions within the epigenetic network. We discuss diagnostic and therapeutic opportunities appearing at horizon of epigenetic research. Moreover, we present current and propose future clinical workflow models for molecular characterization of malignant gliomas. Ó 2012 Elsevier Ireland Ltd. All rights reserved.
1. Introduction Diffuse gliomas represent a broad diagnostic category that also includes glioblastoma (GBM), the most common and most malignant primary brain tumor in adults. Whereas GBMs are rapidly progressive, resistant to therapy, and typically follow fatal clinical courses with median survival times in the range of 15 months, their low grade counterparts (WHO grade II gliomas) can exhibit years of clinical and radiological stability without any applied therapy [1,2]. However, even these low grade variants clearly represent malignant tumor entities, which undergo more or less delayed malignant transformation and rapid clinical decline later on [3]. Traditional molecular approaches, which have mainly focused on structural changes in genes such as point mutations, gene deletions, and rearrangements, have added important insights to both the process of glioma-genesis itself and the diagnostic, prognostic and predictive network of these tumors. Recently, for example, mutations in the IDH1/IDH2 gene encoding isocitrate dehydrogenase 1 and 2 have been identified as one of the earliest molecular event in the pathway to grade II/III astrocytomas, oligodendrogliomas, oligo-astrocytomas, and secondary GBMs but not in the development of de novo GBMs. These findings suggest that histologically differently appearing tumor subclasses might share common precursor cells and that histologically indistinguishable
tumors (e.g. de novo GBM versus secondary GBM) could be separated into biologically distinct subclasses [4–7]. The additional focus on mechanisms of regulation of gene expression at the transcriptional/translational level indicates one of the most emerging fields in cancer research: Recent research has provided evidence that malignant transformation results from a complex interplay of both genetic alterations and epigenetic changes affecting various cellular processes, including cell proliferation and invasion, DNA repair, apoptosis, angiogenesis and cell cycle regulation, finally leading to the development of a malignant phenotype [8,9]. Among the most studied epigenetic changes occurring in gliomas, DNA methylation, and histone modifications can be mentioned. Recently, non-coding RNAs have been recognized to constitute another level of epigenetic control. Identification and characterization of epigenetic alterations in human gliomas have added important parts to the puzzle of glioma-genesis and will lead to the discovery of novel prognostic and predictive biomarkers. Moreover, the reversible nature of epigenetic modifications kindled strong efforts to develop new therapeutic approaches aiming at epigenetic landmarks of human gliomas [10]. Here, we will provide a brief survey about the basic mechanisms of epigenetic control found in human glioma, their clinical significance, and the resulting therapeutical perspectives. 2. DNA methylation
⇑ Corresponding author. Address: Department of Neurosurgery, Ludwig-Maximilians-University, Campus Großhadern, Marchioninistrasse 15, D-81377 München, Germany. Tel.: +49 (0) 89 7095 3555; fax: +49 (0) 89 7095 2592. E-mail address:
[email protected] (F.W. Kreth).
The most studied epigenetic modification in humans is cytosine methylation, which describes the covalent addition of a methyl group at the 50 -position of a cytosine nucleotide. In mammalian
0304-3835/$ - see front matter Ó 2012 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.canlet.2012.04.008
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
2
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx
cells, DNA methylation predominantly occurs on a cytosine residue followed by a guanine. These CpG dinucleotides tend to cluster to so-called CpG islands being located in the promoter regions of more than half of all human genes, or to CpG island shores, which are regions of lower CpG density that lie in close proximity of CpG islands [11,12]. DNA methylation is mediated by the DNMT (DNA methyltransferase) family of enzymes that catalyze the transfer of a methyl group from S-adenosyl methionine to DNA. So far, five members of the DNMT family have been identified: DNMT1, DNMT2, DNMT3a, DNMT3b, and DNMT3L. Whereas DNMT3a and DNMT3b target unmethylated CpGs and therefore are termed de novo methyltransferases, DNMT1 preferentially targets hemimethylated DNA to maintain the methylation pattern during DNA replication; DNMT3L is required for maternal genomic imprinting processes [13,14]. Generally, methylation of CpG islands and island shores is closely associated with transcriptional inactivation. Less frequently, when occurring at gene bodies, DNA methylation is associated with transcriptional activation. In human cancer, it is widely known that global alterations of DNA methylation profiles play an important role in tumor initiation and progression [15,16]. 2.1. DNA methylation in gliomas Human glioma exhibit tumor-typical changes of methylation patterns, which are characterized by a global loss of methylation combined with focal hypermethylation [17–20]. Hypomethylation mostly takes place at DNA-repetitive regions and may promote tumor growth by activation of oncogenes and by increasing genomic instability. Hypermethylation mostly occurs at the promoter CpG island of genes that are involved in processes leading to tumor formation and progression and has been shown for a wide variety of genes associated with tumor suppression [21,22], DNA repair [23], cell cycle regulation [24], apoptosis [25,26], invasion [27,28], and migration [29], and the list of examples is constantly increasing (Table 1). Interestingly, the methylation patterns between different glioma grades appear to differ, as differences in the methylation status of various genes has been detected between gliomas WHO grade II, III, and IV [30]. Within the framework of The Cancer Genome Atlas (TCGA) project, a methylation profiling study by Noushmehr et al. identified a GBM tumor phenotype that is characterized by a concerted hypermethylation of a large number of gene loci, which has been referred to as glioma CpG island methylator phenotype (G-CIMP) [6]. G-CIMP is associated with prolonged survival and with defined gene expression profiles (proneural expression patterns) [31]. Furthermore, a close association with IDH1 mutations was found, which may be explained by metabolic alterations resulting from these mutations: IDH proteins catalyze the oxydative decarboxylation of isocitrate to a-ketoglutarate (a-KG); IDH mutations lead to changes in enzymatic activities: a-KG production is reduced, and instead, the metabolite 2-hydroxyglutarate (2-HG) is produced, which competitively inhibits the activity of enzymes regulating DNA- and histone methylation (a-KG-dependent dioxygenases), including histone demethylases [32] and the TET family of 5mC hydroxylases [33–35]. Recently, TET proteins have been identified as a new class of enzymes that are capable to alter the methylation status of the DNA by converting 5-methylcytosine (5mC) to 5hydroxymethylcytosine (5hmC). The biological function of 5hmC has not conclusively been elucidated yet. Most recent data, however, support the theory that it is an intermediate in DNA demethylation processes. In human gliomas a remarkable reduction of 5hmC as compared to normal brain and an inverse relationship between 5hmC levels and cell proliferation have been reported [36,37]. These findings unveil an additional level of regulation and demonstrate the strong interrelation of genetic and epigenetic events in glioma-genesis [38,39].
Table 1 Major epigenetic alterations found in human gliomas. DNA methylation Genes GATA4, NDRG2 MGMT p14ARF TMS1/ASC, WWOX SOCS3, PCDH-gamma-A11 Sox2
Pathway Tumor suppression DNA repair Cell cycle regulation Apoptosis Invasion Migration
[21,22] [23] [24] [25,26] [27,28] [29]
Histone modifications Global aberrations Mutations Histone deacetylases Histone demethylases Histone methyltransferases
(HDAC2, HDAC9) (JMJD1A, JMJD1B) (SET7, SETD7, MLL, MLL3, MLL4)
Altered expression levels Histone deacetylases
(HDAC1, HDAC2, HDAC3)
Modifications of individual genes Genes Effects RRP22 Repressed expression P21 Repressed expression HOXA9 Enhanced expression
[5]
[53] [54] [55]
Micro-RNA deregulation Down-regulation of tumor suppressive miRNAs mi-RNA Target gene miR-34a c-Met, Notch miR-146a Notch miR-7 EGFR miR-128 Bmi-1 miR-195 E2F3, CCND3
[79] [80] [81] [82] [83]
Up-regulation of oncogenic miRNAs miR-21 RECK, TIMP3 miR-26a pTEN, RB1 miR-10b Cell-cycle inhibitor miR-30e IjBa miR-221/222 p27Kip1, PTPl, PUMA,
[85] [75,86] [76] [87] [88–90]
In the last decade, research on DNA methylation processes has led to the discovery of an important biomarker that has increasingly gained translational relevance: The O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme that removes alkyl adducts at the O6 position of guanine and thereby protects normal cells from carcinogens, but – unfortunately – also protects tumor cells from chemotherapy with alkylating agents such as temozolomide. MGMT expression can be epigenetically silenced by promoter methylation, a phenomenon that is found in 35–45% of malignant gliomas and in 80% of WHO grade II gliomas [40,41]. Promoter methylation status has been identified as a strong and independent predictive factor of treatment response for malignant glioma patients undergoing chemotherapy with alkylating agents. Generally, carriers of the methylated form of the MGMT promoter respond substantially better to therapy with temozolomide as compared to those with an unmethylated MGMT promoter [42,43]. Determination of the MGMT promoter methylation status has been established as an important clinical marker in neuro-oncology and is increasingly used for stratification in clinical trials. MGMT promoter methylation, however, is not unequivocally linked to a favorable treatment response. Not all patients with a methylated promoter showed treatment response after temozolomide, and a considerable number of unmethylated tumors experienced a surprisingly favorable outcome. In these patients, discordant findings of MGMT promoter methylation and mRNA MGMT expression have been demonstrated: High (low) mRNA MGMT expression was detected in approximately 25% of glioblastomas despite a methylated (unmethylated) MGMT promoter; those with low transcriptional
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx
activity exhibited a better treatment response, which was independent of MGMT promoter methylation. The underlying mechanisms of discordance still remain unclear. It has been hypothesized that low MGMT expression levels combined with an unmethylated promoter might result from transcript destabilization and/or transcription-repressing factors, such as miRNA regulation or histone modifications [44]. Existence of discordant findings explains that treatment decision with respect to chemotherapy cannot be based on the MGMT promoter methylation status alone. 3. Histone modifications Chromatin is the condensed form of DNA and histones within the nucleus of a cell. In eukaryotes, chromatin is composed of 147 base pairs of DNA that are tightly wrapped around an octamer of each two copies of the four core histones H2A, H2B, H3 and H4. The resulting nucleosomes are indicated as the fundamental repeating units of chromatin [45]. As each core histone possesses an amino-terminal ‘‘tail’’ which protrudes from the nucleosome, histones and particularly their tails can be subject to a remarkable number of post-translational modifications. Histone modifications comprise prominent marks like acetylation, methylation and phosphorylation, but also less studied modifications such as ubiquitylation, sumoylation, ADP ribosylation, deamination and proline isomerisation [46]. Each of these histone modifications is able to affect chromatin structure, thereby leading to alterations in DNA repair and replication as well as in gene transcription. Regarding the latter, histone modifications can broadly be categorized into active versus passive marks. In particular histone acetylation and methylation have been found to play a remarkable role in carcinogenesis [15,47]. The acetylation of lysine residues is regulated by the opposing action of histone acetyl transferases (HATs) and histone deacetylases (HDACs). Acetylation neutralizes the positive charge of lysine residue and thereby weakens the bond between DNA and histone tails. Thus, histone acetylation is linked to transcriptional activation, whereas deacetylation is generally associated with repression of transcription. Histone methylation mainly occurs on the side chains of lysines and arginines, which influences the activity of effector proteins of the transcriptional machinery. In contrast to acetylation of histones, which is generally associated with activation of gene expression, histone methylation may either activate (e.g. H3K4me2, H3K4me3) or repress (H3K9me2, H3K27me3) transcription depending on the respective methylation sites [48–50]. 3.1. Histone modifications in gliomas Recent data have mounted evidence that alterations at the histone level may also play a role in glioma-genesis. These alterations encompass a globally deregulated expression of genes involved in histone modifications as well as changes in the histone modification pattern of individual genes (Table 1). Global aberrations at the histone level result from mutations in regulatory genes, as detected in a large-scale genomic analysis of GBM samples, including HDACs (HDAC2 and HDAC9), histone demethylases (JMJD1A and JMJD1B), histone methylatransferases (SET7, SETD7, MLL, MLL3 and MLL4) [5]. Furthermore, altered expression levels of HDACs due to so far not defined reasons have been reported and have been linked to tumor recurrence and progression (HDAC1, HDAC2, and HDAC3) [51,52]. Histone modifications regulating individual genes have been reported in several studies. For example, a repressed expression of the tumor suppressor RRP22 and of the cell cycle regulator p21, and an enhanced expression of the pro-proliferative transcription factor HOXA9 have been linked to alterations in histone modification patterns [53–55]. However, the actual functional
3
roles of histone modifications in gliomas and their potential to serve as biomarkers and/or therapeutic targets, still remains to be fully elucidated. 4. MicroRNAs Non-coding RNAs have recently been found to play an important role in the epigenetic regulation of gene expression [56,57]. Among these, microRNAs (miRNA) are the best characterized modulators of gene function: MiRNAs are double-stranded RNA molecules of approximately 22 nucleotides (nt) length which – in several processing steps – derive from larger precursor transcripts that are ubiquitously encoded in the human genome. By binding to specific recognition sequences within the 30 -untranslated region (30 -UTR) of target mRNAs, miRNAs are potent regulators of gene expression by either repression of translation or mRNA degradation [58,59]. Target recognition appears to be mainly mediated through basepairing of an eight nucleotide short sequence in the 50 -region of the miRNA [60]. Whereas some miRNAs regulate specific individual targets, evidence from multiple studies suggests that a single key miRNA may regulate up to several hundreds of target genes, and many types of miRNAs regulate their targets cooperatively [61,62]. Using a combination of computational prediction methods and different sequencing techniques a significant number of these regulatory molecules have been identified [63,64]. The current release of ‘‘mirbase’’, the official registry for known miRNA-genes, contains more than 17,000 mature miRNA sequences in over 140 species [65]. Recent work suggests that large parts of the transcriptome are subject to regulatory control exerted by miRNAs [66]. It is therefore not surprising that dysregulation of miRNA expression has been associated with a wide range of pathologies and their prognosis. 4.1. MiRNAs in gliomas Aberrant expression of miRNAs has been detected in many types of human tumors, including gliomas [67,68]. Recent data have shown that human tumors are characterized by globally down-regulated miRNA expression profiles due to a general defect in the miRNA production process, which promotes the view that miRNAs may mainly serve as ‘‘guardians’’ of biologic processes [69]. Nevertheless, miRNA have been demonstrated not to act only as tumor suppressors, but also – dependent on the function of the targeted mRNA – as oncogenes [70,71]. Accordingly, altered miRNA expression levels exert major impact on oncogenic processes. The reasons for the widespread differential expression of miRNAs in malignant as compared to normal cells are not fully elucidated. However, epigenetic modifications within the transcriptional regulatory sequences of miRNAs, but also genetic alterations like mutations, genomic deletions or gene amplifications, which can affect miRNA maturation and/or interactions with mRNA targets, are thought to contribute to miRNA dysregulation [72–74]. In GBM, high-throughput analyses have identified various miRNAs that are differentially expressed when compared to non-neoplastic brain tissues [75–77]. Thus, miRNAs are thought to be important mediators of multiple biological characteristics of GBM, including cell proliferation, G1/S cell cycle progression, cell survival, cell migration and cell invasion [78]. Although the exact function of altered miRNAs in glioma and of the complex network they regulate have not been elucidated yet, a growing number of studies have assigned single miRNAs to distinct functions in the processes of glioma-genesis and progression (Table 1). For example, miRNAs that are downregulated in gliomas as compared to normal brain have been found to function as tumor suppressors by directly targeting the oncogenes c-Met, Notch [79,80], Bmi-1 [76], the epidermal growth factor receptor [81], receptor tyrosine
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
4
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx
kinases [82], and cell cycle components [83]. Contrarily, miRNAs with enhanced expression in glioma might be designated as oncogenes; i.e. miR-21 promoting invasion by targeting regulators of matrix metalloproteinases, miR-26a targeting PTEN, and miR-10b and miR-221 targeting cell cycle inhibitors, and miR30e targeting IjBa [84–90]. 5. The ‘‘epigenetic network’’ Epigenetic regulation pathways gain further complexity by interrelationships between the epigenetic players: (i) Expression of miRNA itself can be modified by changes in chromatin structures caused by covalent modifications on histones and/or DNA methylation. For instance, it has recently been shown that miR-127 and miR124a, two putative tumor suppressor miRNAs, are methylated in tumor cells [91,92]. (ii) Tumors can use miRNAs reciprocally to target the epigenetic machinery. For example, miR-29b was found to target DNMT3A and 3B in acute myeloid leukemia, and miR-449a controls HDAC1 in prostate cancer cells. In human glioma, miR185 has recently been described to be a regulator of DNMT1, and its overexpression results in a global DNA hypomethylation [93– 95]. Furthermore, miR-101 which targets the histone methyltransferase EZH2 has been found to be down-regulated in human GBM thereby promoting tumor growth [96,97]. (iii) Histone modification enzymes may be silenced by CpG hypermethylation. For instance, silencing of the NSD1 gene, which encodes a histone methyltransferase involved in chromatin regulation, results in diminished methylation of histone residues H4-K20 and H3-K36, which in turn leads to activation of the oncogene MEIS1. This epigenetic alteration has been found to be a common event in gliomas [98]. Further studies on a genome-wide scale are needed to fully elucidate epigenetic patterns in gliomas, which will elucidate the complex pattern of the epigenetic networks of these highly heterogeneous tumors [99]. 6. Epigenetics in glioma: perspectives Enormous recent advances in high throughput analyses have opened up not only DNA sequences of individual whole genomes of human GBM samples but also complete DNA methylomes, histonomes and transcriptomes [7,5]. Based on these new technical possibilities, current brain tumor research aims at the identification and characterization of new biomarkers that may allow to exactly determine the diagnosis and to estimate prognosis and treatment response of the individual patient [100–102]. It therefore can be expected that in human gliomas, determination of MGMT promoter methylation may only be the spearhead of new evolving biomarkers based on epigenetic landmarks. Since the approval of DNA demethylating agents and histone deacetylase inhibitors for the treatment of lymphoma patients, the potential of epigenetic proteins and modifications to serve as drug targets has widely been recognized [103,104]. The underlying idea is (i) to reverse tumor specific epigenetic alterations that may essentially drive tumor growth (for example by silencing tumor suppressors, activating oncogenes, or affecting DNA repair), and (ii) to induce changes in chromatin structure permitting better access of DNA damaging drugs [105,106]. In human gliomas, several approaches aiming at the global epigenetic machinery have been made: Treatment with histone deacetylase inhibitors (HDACi) such as aliphatic acids (valproic acid, butyrate), suberoylanilide hydrocamic acid (SAHA, vorinostat), alone or in combination with radiotherapy have been suggested to exert beneficial effects in preclinical models [107–109]; furthermore, pilot studies using SAHA or valproic acid in GBM have described low toxicity and moderate response rates [110–113]. However, these therapy approaches only
represent a first step in a new direction; future approaches should enable a specific targeting of defined epigenetic landmarks. Perspectives of clinical concepts based on miRNA regulatory networks are promising, because miRNAs may prove as both suitable biomarkers and therapeutic targets in oncology: Current studies indicate the presence of miRNAs in blood circulation, which are set free from tumor cells and thus, characteristical miRNA signatures may be linked to the diagnosis and prognosis for certain tumor types [114–116]. Concerning brain tumors, a specific miRNA signature in the peripheral blood and in cerebrospinal fluid of GBM patients was reported recently [117,118]. The rapidly expanding knowledge of the substantial role of miRNA in epigenetic regulation has led to the development of therapeutic concepts based on miRNA for a range of diseases [119–121]. Basically, two approaches exist: first, suppression of disease-specific miRNAs, and second, application of specific miRNAs in excess to the diseased tissue or organism to achieve therapeutic effects by selective gen silencing [122–124]. The evolving concepts are promising; however at present all techniques (i.e. anti-miR oligonucleotides, competitive miR-inhibitors) are still in early stages and not ready to be introduced in clinical practice [125]. One general key challenge is the targeted and effective delivery of the small nucleotide agents to cells and tissues, and in gliomas, this is further complicated by the blood–brain-barrier. Since one miRNA can influence the expression of various target genes, several important aspects must be reconsidered before miRNAs may be used therapeutically: First, manipulation of one single miRNA may affect the expression of many different proteins and may potentially cause devastating side effects. Second, miRNAs can affect different cell types and tissues with occasionally contrary effects and tissue specific delivery of miRNAs or antagonists is yet an unsolved problem. 7. Clinical workflow: present and future The consideration of molecular biomarkers has improved both the diagnostic precision within the framework of the current WHO classification and the discrimination of glioma subgroups exhibiting the same WHO grade but differences in prognosis and treatment response [2]. The detection of a 1p/19 co-deletetion, for example, supports the diagnosis of an oligidendroglioma/oligoastrocytoma and has been associated with a favorable outcome after chemotherapy and/or radiation therapy [126,127]. Screening for IDH1/2 mutations is helpful to distinguish WHO grade I pilocytic astrocytomas and ependymomas (harboring no IDH mutations) from diffuse astrocytomas and to differentiate diffuse tumor infiltration from reactive gliosis. IDH mutations are associated with a better prognosis in anaplastic astrocytomas and glioblastomas [128,129]. Given the fact that complete tumor resection is seldom possible in grade II–IV gliomas and incomplete resections usually did not gain prognostic impact, the development of minimal invasive molecular characterization strategies is of utmost interest [130]. Recently, novel molecular stereotactic biopsy procedures combining histopathologic diagnosis with size-adjusted molecular genetic analyses have been shown to achieve highly reproducible and valid results at the DNA and RNA level as well; remarkably, information about MGMT promoter methylation and MGMT mRNA expression, TP53 mutational status, and LOH on 1p/19q-status could be obtained from a single 1-mm3 stereotactic tissue sample collected from an exactly defined site of the tumor space [131,132]. The introduction of the massively parallel ‘‘next generation’’ sequencing (NGS) techniques into scientific practice marks the start of a revolution in genomic research [133]; whole-genome, transcriptome- and methylome-sequencing is currently at the horizon of clinical research and could be performed even in small-sized tumor tissue samples. NGS platforms provide a comprehensive and unbiased view of the genome and epigenome and the integration
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx
5
Fig. 1. Flow chart illustrating present and future clinical workflow for glioma treatment. Tumor samples are obtained by minimally invasive stereotactic biopsy procedure; samples are used for histologic analyses and for extraction of nucleic acids. Classical workflow (1): Tumor samples (smear and paraffine embedded) are used for histopathologic diagnosis; categorized treatment concepts are applied. Modern workflow (1 + 2) implementing molecular features of the tumor (MGMT promoter methylation, LOH 1p19q, IDH1/2 mutations, further optional markers); therapeutic proceedings are adapted to the results of the analyses. Future workflow (1 + 3): Tumor samples are used for histology and next generation sequencing analyses. Whole genome-, epigenome- and transcriptome analyses allow a complete molecular characterization of the tumor. Therapy comprises an individualized concept integrating all treatment modalities.
of NGS-based data will be essential to understand and identify the dense web of connected intracellular pathways that drive formation and progression of gliomas [134,135]. With falling costs of the NGS technology and improved bioinformatic handling of the data, the simultaneous genome wide measurement of multiple epigenetic modifications in conjunction with the transcriptome and genetic variations of the same biologic sample may become a clinical standard approach in the future. In this setting, removal of tumor samples by minimal-invasive methods, such the molecular stereotactic procedure, will be of particular value. The information gained from this approach will allow creating customized therapeutic regimens for each patient based on the unique genomic signature of the individual tumor. Also, therapeutic strategies for the treatment of gliomas have to be markedly improved, which includes the development of new epigenetic therapies and the gain of knowledge how to synchronize them with other treatment modalities. A modern present clinical workflow and the anticipated future developments are schematically illustrated in Fig. 1.
8. Conclusion Currently, we are at the beginning of a comprehensive understanding of the glioma epigenome. Before introducing therapeutic concepts in the treatment of brain tumors based on epigenetic aberrations, critical aspects have yet to be prospectively addressed in clinical and basic research, and technical tools have to be further developed. Nevertheless, we envision that the unique properties of epigenetic changes in gliomas offer a highly promising future diagnostic and therapeutic potential which will certainly lead to innovative clinical concepts.
Acknowledgement Grant support from the Institutional funding of the LMU Munich. We thank Dr. S. Schütz for suggestions on the manuscript.
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
6
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx
References [1] R. Stupp, M.E. Hegi, W.P. Mason, M.J. van den Bent, M.J. Taphoorn, R.C. Janzer, S.K. Ludwin, A. Allgeier, B. Fisher, K. Belanger, P. Hau, A.A. Brandes, J. Gijtenbeek, C. Marosi, C.J. Vecht, K. Mokhtari, P. Wesseling, S. Villa, E. Eisenhauer, T. Gorlia, M. Weller, D. Lacombe, J.G. Cairncross, R.O. Mirimanoff, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial, Lancet Oncol. 10 (2009) 459– 466. [2] M. Westphal, K. Lamszus, The neurobiology of gliomas: from cell biology to the development of therapeutic approaches, Nat. Rev. Neurosci. 12 (2011) 495–508. [3] F.W. Kreth, M. Faist, S. Grau, C.B. Ostertag, Interstitial 125I radiosurgery of supratentorial de novo WHO Grade 2 astrocytoma and oligoastrocytoma in adults: long-term results and prognostic factors, Cancer 106 (2006) 1372– 1381. [4] H. Ohgaki, P. Kleihues, Genetic alterations and signaling pathways in the evolution of gliomas, Cancer Sci. 100 (2009) 2235–2241. [5] D.W. Parsons, S. Jones, X. Zhang, J.C. Lin, R.J. Leary, P. Angenendt, P. Mankoo, H. Carter, I.M. Siu, G.L. Gallia, A. Olivi, R. McLendon, B.A. Rasheed, S. Keir, T. Nikolskaya, Y. Nikolsky, D.A. Busam, H. Tekleab, L.A. Diaz Jr., J. Hartigan, D.R. Smith, R.L. Strausberg, S.K. Marie, S.M. Shinjo, H. Yan, G.J. Riggins, D.D. Bigner, R. Karchin, N. Papadopoulos, G. Parmigiani, B. Vogelstein, V.E. Velculescu, K.W. Kinzler, An integrated genomic analysis of human glioblastoma multiforme, Science 321 (2008) 1807–1812. [6] H. Noushmehr, D.J. Weisenberger, K. Diefes, H.S. Phillips, K. Pujara, B.P. Berman, F. Pan, C.E. Pelloski, E.P. Sulman, K.P. Bhat, R.G. Verhaak, K.A. Hoadley, D.N. Hayes, C.M. Perou, H.K. Schmidt, L. Ding, R.K. Wilson, D. Van Den Berg, H. Shen, H. Bengtsson, P. Neuvial, L.M. Cope, J. Buckley, J.G. Herman, S.B. Baylin, P.W. Laird, K. Aldape, Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma, Cancer Cell 17 (2010) 510–522. [7] Cancer Genome Atlas Research Network, Comprehensive genomic characterization defines human glioblastoma genes and core pathways, Nature 455 (2008) 1061–1068. [8] M. Berdasco, M. Esteller, Aberrant epigenetic landscape in cancer: how cellular identity goes awry, Dev. Cell 19 (2010) 698–711. [9] A. Portela, M. Esteller, Epigenetic modifications and human disease, Nat. Biotechnol. 28 (2010) 1057–1068. [10] R. Martinez, G. Schackert, Epigenetic aberrations in malignant gliomas: an open door leading to better understanding and treatment, Epigenetics 2 (2007) 147–150. [11] M.M. Suzuki, A. Bird, DNA methylation landscapes: provocative insights from epigenomics, Nat. Rev. Genet. 9 (2008) 465–476. [12] R.A. Irizarry, C. Ladd-Acosta, B. Wen, Z. Wu, C. Montano, P. Onyango, H. Cui, K. Gabo, M. Rongione, M. Webster, H. Ji, J.B. Potash, S. Sabunciyan, A.P. Feinberg, The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores, Nat. Genet. 41 (2009) 178–186. [13] Z.X. Chen, A.D. Riggs, DNA methylation and demethylation in mammals, J. Biol. Chem. 286 (2011) 18347–18353. [14] A. Hellman, A. Chess, Gene body-specific methylation on the active X chromosome, Science 315 (2007) 1141–1143. [15] M. Esteller, Cancer epigenomics: DNA methylomes and histone-modification maps, Nat. Rev. Genet. 8 (2007) 286–298. [16] K.D. Robertson, DNA methylation and human disease, Nat. Rev. Genet. 6 (2005) 597–610. [17] M. Esteller, P.G. Corn, S.B. Baylin, J.G. Herman, A gene hypermethylation profile of human cancer, Cancer Res. 61 (2001) 3225–3229. [18] B. Cadieux, T.T. Ching, S.R. Vandenberg, J.F. Costello, Genome-wide hypomethylation in human glioblastomas associated with specific copy number alteration, methylenetetrahydrofolate reductase allele status, and increased proliferation, Cancer Res. 66 (2006) 8469–8476. [19] R. Martinez, J.I. Martin-Subero, V. Rohde, M. Kirsch, M. Alaminos, A.F. Fernandez, S. Ropero, G. Schackert, M. Esteller, A microarray-based DNA methylation study of glioblastoma multiforme, Epigenetics 4 (2009) 255– 264. [20] X. Wu, T.A. Rauch, X. Zhong, W.P. Bennett, F. Latif, D. Krex, G.P. Pfeifer, CpG island hypermethylation in human astrocytomas, Cancer Res. 70 (2010) 2718–2727. [21] S. Agnihotri, A. Wolf, D.M. Munoz, C.J. Smith, A. Gajadhar, A. Restrepo, I.D. Clarke, G.N. Fuller, S. Kesari, P.B. Dirks, C.J. McGlade, W.L. Stanford, K. Aldape, P.S. Mischel, C. Hawkins, A. Guha, A GATA4-regulated tumor suppressor network represses formation of malignant human astrocytomas, J. Exp. Med. 208 (2011) 689–702. [22] M. Tepel, P. Roerig, M. Wolter, D.H. Gutmann, A. Perry, G. Reifenberger, M.J. Riemenschneider, Frequent promoter hypermethylation and transcriptional downregulation of the NDRG2 gene at 14q11.2 in primary glioblastoma, Int. J. Cancer 123 (2008) 2080–2086. [23] M. Nakamura, T. Watanabe, Y. Yonekawa, P. Kleihues, H. Ohgaki, Promoter methylation of the DNA repair gene MGMT in astrocytomas is frequently associated with G:C ? A:T mutations of the TP53 tumor suppressor gene, Carcinogenesis 22 (2001) 1715–1719.
[24] M. Nakamura, T. Watanabe, U. Klangby, C. Asker, K. Wiman, Y. Yonekawa, P. Kleihues, H. Ohgaki, P14ARF deletion and methylation in genetic pathways to glioblastomas, Brain Pathol. 11 (2001) 159–168. [25] A.R. Stone, W. Bobo, D.J. Brat, N.S. Devi, E.G. Van Meir, P.M. Vertino, Aberrant methylation and down-regulation of TMS1/ASC in human glioblastoma, Am. J. Pathol. 165 (2004) 1151–1161. [26] K. Kosla, E. Pluciennik, A. Kurzyk, D. Jesionek-Kupnicka, R. Kordek, P. Potemski, A.K. Bednarek, Molecular analysis of WWOX expression correlation with proliferation and apoptosis in glioblastoma multiforme, J. Neurooncol. 101 (2011) 207–213. [27] A. Waha, S. Guntner, T.H. Huang, P.S. Yan, B. Arslan, T. Pietsch, O.D. Wiestler, A. Waha, Epigenetic silencing of the protocadherin family member PCDHgamma-A11 in astrocytomas, Neoplasia 7 (2005) 193–199. [28] C. Lindemann, O. Hackmann, S. Delic, N. Schmidt, G. Reifenberger, M.J. Riemenschneider, SOCS3 promoter methylation is mutually exclusive to EGFR amplification in gliomas and promotes glioma cell invasion through STAT3 and FAK activation, Acta Neuropathol. 122 (2011) 241–251. [29] M.M. Alonso, R. Diez-Valle, L. Manterola, A. Rubio, D. Liu, N. Cortes-Santiago, L. Urquiza, P. Jauregi, A.L. de Munain, N. Sampron, A. Aramburu, S. TejadaSolis, C. Vicente, M.D. Odero, E. Bandres, J. Garcia-Foncillas, M.A. Idoate, F.F. Lang, J. Fueyo, C. Gomez-Manzano, Genetic and epigenetic modifications of Sox2 contribute to the invasive phenotype of malignant gliomas, PLoS One 6 (2011) e26740. [30] K. Uhlmann, K. Rohde, C. Zeller, J. Szymas, S. Vogel, K. Marczinek, G. Thiel, P. Nurnberg, P.W. Laird, Distinct methylation profiles of glioma subtypes, Int. J. Cancer 106 (2003) 52–59. [31] R.G. Verhaak, K.A. Hoadley, E. Purdom, V. Wang, Y. Qi, M.D. Wilkerson, C.R. Miller, L. Ding, T. Golub, J.P. Mesirov, G. Alexe, M. Lawrence, M. O’Kelly, P. Tamayo, B.A. Weir, S. Gabriel, W. Winckler, S. Gupta, L. Jakkula, H.S. Feiler, J.G. Hodgson, C.D. James, J.N. Sarkaria, C. Brennan, A. Kahn, P.T. Spellman, R.K. Wilson, T.P. Speed, J.W. Gray, M. Meyerson, G. Getz, C.M. Perou, D.N. Hayes, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1, Cancer Cell 17 (2010) 98–110. [32] C. Lu, P.S. Ward, G.S. Kapoor, D. Rohle, S. Turcan, O. Abdel-Wahab, C.R. Edwards, R. Khanin, M.E. Figueroa, A. Melnick, K.E. Wellen, D.M. O’Rourke, S.L. Berger, T.A. Chan, R.L. Levine, I.K. Mellinghoff, C.B. Thompson, IDH mutation impairs histone demethylation and results in a block to cell differentiation, Nature 483 (2012) 474–478. [33] L. Dang, D.W. White, S. Gross, B.D. Bennett, M.A. Bittinger, E.M. Driggers, V.R. Fantin, H.G. Jang, S. Jin, M.C. Keenan, K.M. Marks, R.M. Prins, P.S. Ward, K.E. Yen, L.M. Liau, J.D. Rabinowitz, L.C. Cantley, C.B. Thompson, M.G. Vander Heiden, S.M. Su, Cancer-associated IDH1 mutations produce 2hydroxyglutarate, Nature 465 (2010) 966. [34] W. Xu, H. Yang, Y. Liu, Y. Yang, P. Wang, S.H. Kim, S. Ito, C. Yang, P. Wang, M.T. Xiao, L.X. Liu, W.Q. Jiang, J. Liu, J.Y. Zhang, B. Wang, S. Frye, Y. Zhang, Y.H. Xu, Q.Y. Lei, K.L. Guan, S.M. Zhao, Y. Xiong, Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases, Cancer Cell 19 (2011) 17–30. [35] J.U. Guo, Y. Su, C. Zhong, G.L. Ming, H. Song, Hydroxylation of 5methylcytosine by TET1 promotes active DNA demethylation in the adult brain, Cell 145 (2011) 423–434. [36] S.G. Jin, Y. Jiang, R. Qiu, T.A. Rauch, Y. Wang, G. Schackert, D. Krex, Q. Lu, G.P. Pfeifer, 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations, Cancer Res. 71 (2011) 7360– 7365. [37] T.F. Kraus, D. Globisch, M. Wagner, S. Eigenbrod, D. Widmann, M. Munzel, M. Muller, T. Pfaffeneder, B. Hackner, W. Feiden, U. Schuller, T. Carell, H.A. Kretzschmar, Low values of 5-hydroxymethylcytosine (5hmC), the ‘‘sixth base,’’ are associated with anaplasia in human brain tumors, Int. J. Cancer (2012), http://dx.doi.org/10.1002/ijc.27429 [Epub ahead of print]. [38] M.E. Figueroa, O. Abdel-Wahab, C. Lu, P.S. Ward, J. Patel, A. Shih, Y. Li, N. Bhagwat, A. Vasanthakumar, H.F. Fernandez, M.S. Tallman, Z. Sun, K. Wolniak, J.K. Peeters, W. Liu, S.E. Choe, V.R. Fantin, E. Paietta, B. Lowenberg, J.D. Licht, L.A. Godley, R. Delwel, P.J. Valk, C.B. Thompson, R.L. Levine, A. Melnick, Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation, Cancer Cell 18 (2010) 553–567. [39] K. Williams, J. Christensen, M.T. Pedersen, J.V. Johansen, P.A. Cloos, J. Rappsilber, K. Helin, TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity, Nature 473 (2011) 343–348. [40] M.E. Hegi, A.C. Diserens, T. Gorlia, M.F. Hamou, T.N. de, M. Weller, J.M. Kros, J.A. Hainfellner, W. Mason, L. Mariani, J.E. Bromberg, P. Hau, R.O. Mirimanoff, J.G. Cairncross, R.C. Janzer, R. Stupp, MGMT gene silencing and benefit from temozolomide in glioblastoma, N. Engl. J. Med. 352 (2005) 997–1003. [41] N. Thon, S. Eigenbrod, S. Kreth, J. Lutz, J.C. Tonn, H. Kretzschmar, A. Peraud, F.W. Kreth, IDH1 mutations in grade II astrocytomas are associated with unfavorable progression-free survival and prolonged postrecurrence survival, Cancer 118 (2012) 452–460. [42] W. Wick, C. Hartmann, C. Engel, M. Stoffels, J. Felsberg, F. Stockhammer, M.C. Sabel, S. Koeppen, R. Ketter, R. Meyermann, M. Rapp, C. Meisner, R.D. Kortmann, T. Pietsch, O.D. Wiestler, U. Ernemann, M. Bamberg, G. Reifenberger, D.A. von, M. Weller, NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide, J. Clin. Oncol. 27 (2009) 5874–5880.
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx [43] R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K. Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O. Mirimanoff, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma, N. Engl. J. Med. 352 (2005) 987–996. [44] S. Kreth, N. Thon, S. Eigenbrod, J. Lutz, C. Ledderose, R. Egensperger, J.C. Tonn, H.A. Kretzschmar, L.C. Hinske, F.W. Kreth, O-methylguanine-DNA methyltransferase (MGMT) mRNA expression predicts outcome in malignant glioma independent of MGMT promoter methylation, PLoS One 6 (2011) e17156. [45] R. Margueron, D. Reinberg, Chromatin structure and the inheritance of epigenetic information, Nat. Rev. Genet. 11 (2010) 285–296. [46] T. Kouzarides, Chromatin modifications and their function, Cell 128 (2007) 693–705. [47] M.F. Fraga, E. Ballestar, A. Villar-Garea, M. Boix-Chornet, J. Espada, G. Schotta, T. Bonaldi, C. Haydon, S. Ropero, K. Petrie, N.G. Iyer, A. Perez-Rosado, E. Calvo, J.A. Lopez, A. Cano, M.J. Calasanz, D. Colomer, M.A. Piris, N. Ahn, A. Imhof, C. Caldas, T. Jenuwein, M. Esteller, Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer, Nat. Genet. 37 (2005) 391–400. [48] R. Karlic, H.R. Chung, J. Lasserre, K. Vlahovicek, M. Vingron, Histone modification levels are predictive for gene expression, Proc. Natl. Acad. Sci. USA 107 (2010) 2926–2931. [49] A.J. Bannister, T. Kouzarides, Regulation of chromatin by histone modifications, Cell Res. 21 (2011) 381–395. [50] H. Santos-Rosa, R. Schneider, A.J. Bannister, J. Sherriff, B.E. Bernstein, N.C. Emre, S.L. Schreiber, J. Mellor, T. Kouzarides, Active genes are tri-methylated at K4 of histone H3, Nature 419 (2002) 407–411. [51] B. Campos, J.L. Bermejo, L. Han, J. Felsberg, R. Ahmadi, N. Grabe, G. Reifenberger, A. Unterberg, C. Herold-Mende, Expression of nuclear receptor corepressors and class I histone deacetylases in astrocytic gliomas, Cancer Sci. 102 (2011) 387–392. [52] A.K. Lucio-Eterovic, M.A. Cortez, E.T. Valera, F.J. Motta, R.G. Queiroz, H.R. Machado, C.G. Carlotti Jr., L. Neder, C.A. Scrideli, L.G. Tone, Differential expression of 12 histone deacetylase (HDAC) genes in astrocytomas and normal brain tissue: class II and IV are hypoexpressed in glioblastomas, BMC Cancer 8 (2008) 243. [53] N. Schmidt, S. Windmann, G. Reifenberger, M.J. Riemenschneider, DNA hypermethylation and histone modifications downregulate the candidate tumor suppressor gene RRP22 on 22q12 in human gliomas, Brain Pathol. 22 (2012) 17–25. [54] D. Mottet, S. Pirotte, V. Lamour, M. Hagedorn, S. Javerzat, A. Bikfalvi, A. Bellahcene, E. Verdin, V. Castronovo, HDAC4 represses p21(WAF1/Cip1) expression in human cancer cells through a Sp1-dependent, p53-independent mechanism, Oncogene 28 (2009) 243–256. [55] B.M. Costa, J.S. Smith, Y. Chen, J. Chen, H.S. Phillips, K.D. Aldape, G. Zardo, J. Nigro, C.D. James, J. Fridlyand, R.M. Reis, J.F. Costello, Reversing HOXA9 oncogene activation by PI3K inhibition: epigenetic mechanism and prognostic significance in human glioblastoma, Cancer Res. 70 (2010) 453– 462. [56] F.F. Costa, Non-coding RNAs, epigenetics and complexity, Gene 410 (2008) 9– 17. [57] M. Esteller, Non-coding RNAs in human disease, Nat. Rev. Genet. 12 (2011) 861–874. [58] N.C. Lau, L.P. Lim, E.G. Weinstein, D.P. Bartel, An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans, Science 294 (2001) 858–862. [59] J.T. Mendell, MicroRNAs: critical regulators of development, cellular physiology and malignancy, Cell Cycle 4 (2005) 1179–1184. [60] B.P. Lewis, C.B. Burge, D.P. Bartel, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets, Cell 120 (2005) 15–20. [61] K.K. Farh, A. Grimson, C. Jan, B.P. Lewis, W.K. Johnston, L.P. Lim, C.B. Burge, D.P. Bartel, The widespread impact of mammalian MicroRNAs on mRNA repression and evolution, Science 310 (2005) 1817–1821. [62] L.P. Lim, N.C. Lau, P. Garrett-Engele, A. Grimson, J.M. Schelter, J. Castle, D.P. Bartel, P.S. Linsley, J.M. Johnson, Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs, Nature 433 (2005) 769–773. [63] B. John, C. Sander, D.S. Marks, Prediction of human microRNA targets, Methods Mol. Biol. 342 (2006) 101–113. [64] P.H. Gunaratne, C. Coarfa, B. Soibam, A. Tandon, MiRNA data analysis: nextgen sequencing, Methods Mol. Biol. 822 (2012) 273–288. [65] A. Kozomara, S. Griffiths-Jones, MiRBase: integrating microRNA annotation and deep-sequencing data, Nucl. Acids Res. 39 (2011) D152–D157. [66] M. Selbach, B. Schwanhausser, N. Thierfelder, Z. Fang, R. Khanin, N. Rajewsky, Widespread changes in protein synthesis induced by microRNAs, Nature 455 (2008) 58–63. [67] H.S. Soifer, J.J. Rossi, P. Saetrom, MicroRNAs in disease and potential therapeutic applications, Mol. Ther. 15 (2007) 2070–2079. [68] C.M. Croce, Causes and consequences of microRNA dysregulation in cancer, Nat. Rev. Genet. 10 (2009) 704–714. [69] P. Carninci, T. Kasukawa, S. Katayama, J. Gough, M.C. Frith, N. Maeda, R. Oyama, T. Ravasi, B. Lenhard, C. Wells, R. Kodzius, K. Shimokawa, V.B. Bajic, S.E. Brenner, S. Batalov, A.R. Forrest, M. Zavolan, M.J. Davis, L.G. Wilming, V. Aidinis, J.E. Allen, A. Ambesi-Impiombato, R. Apweiler, R.N. Aturaliya, T.L.
[70] [71] [72] [73]
[74] [75]
[76]
[77]
[78] [79]
[80] [81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
7
Bailey, M. Bansal, L. Baxter, K.W. Beisel, T. Bersano, H. Bono, A.M. Chalk, K.P. Chiu, V. Choudhary, A. Christoffels, D.R. Clutterbuck, M.L. Crowe, E. Dalla, B.P. Dalrymple, B.B. de, G.G. Della, B.D. di, T. Down, P. Engstrom, M. Fagiolini, G. Faulkner, C.F. Fletcher, T. Fukushima, M. Furuno, S. Futaki, M. Gariboldi, P. Georgii-Hemming, T.R. Gingeras, T. Gojobori, R.E. Green, S. Gustincich, M. Harbers, Y. Hayashi, T.K. Hensch, N. Hirokawa, D. Hill, L. Huminiecki, M. Iacono, K. Ikeo, A. Iwama, T. Ishikawa, M. Jakt, A. Kanapin, M. Katoh, Y. Kawasawa, J. Kelso, H. Kitamura, H. Kitano, G. Kollias, S.P. Krishnan, A. Kruger, S.K. Kummerfeld, I.V. Kurochkin, L.F. Lareau, D. Lazarevic, L. Lipovich, J. Liu, S. Liuni, S. McWilliam, B.M. Madan, M. Madera, L. Marchionni, H. Matsuda, S. Matsuzawa, H. Miki, F. Mignone, S. Miyake, K. Morris, S. Mottagui-Tabar, N. Mulder, N. Nakano, H. Nakauchi, P. Ng, R. Nilsson, S. Nishiguchi, S. Nishikawa, F. Nori, O. Ohara, Y. Okazaki, V. Orlando, K.C. Pang, W.J. Pavan, G. Pavesi, G. Pesole, N. Petrovsky, S. Piazza, J. Reed, J.F. Reid, B.Z. Ring, M. Ringwald, B. Rost, Y. Ruan, S.L. Salzberg, A. Sandelin, C. Schneider, C. Schonbach, K. Sekiguchi, C.A. Semple, S. Seno, L. Sessa, Y. Sheng, Y. Shibata, H. Shimada, K. Shimada, D. Silva, B. Sinclair, S. Sperling, E. Stupka, K. Sugiura, R. Sultana, Y. Takenaka, K. Taki, K. Tammoja, S.L. Tan, S. Tang, M.S. Taylor, J. Tegner, S.A. Teichmann, H.R. Ueda, N.E. van, R. Verardo, C.L. Wei, K. Yagi, H. Yamanishi, E. Zabarovsky, S. Zhu, A. Zimmer, W. Hide, C. Bult, S.M. Grimmond, R.D. Teasdale, E.T. Liu, V. Brusic, J. Quackenbush, C. Wahlestedt, J.S. Mattick, D.A. Hume, C. Kai, D. Sasaki, Y. Tomaru, S. Fukuda, M. Kanamori-Katayama, M. Suzuki, J. Aoki, T. Arakawa, J. Iida, K. Imamura, M. Itoh, T. Kato, H. Kawaji, N. Kawagashira, T. Kawashima, M. Kojima, S. Kondo, H. Konno, K. Nakano, N. Ninomiya, T. Nishio, M. Okada, C. Plessy, K. Shibata, T. Shiraki, S. Suzuki, M. Tagami, K. Waki, A. Watahiki, Y. Okamura-Oho, H. Suzuki, J. Kawai, Y. Hayashizaki, The transcriptional landscape of the mammalian genome, Science 309 (2005) 1559–1563. Y.S. Lee, A. Dutta, MicroRNAs: small but potent oncogenes or tumor suppressors, Curr. Opin. Invest. Drugs 7 (2006) 560–564. B. Zhang, X. Pan, G.P. Cobb, T.A. Anderson, MicroRNAs as oncogenes and tumor suppressors, Dev. Biol. 302 (2007) 1–12. G.A. Calin, C.M. Croce, MicroRNAs and chromosomal abnormalities in cancer cells, Oncogene 25 (2006) 6202–6210. M.S. Kumar, J. Lu, K.L. Mercer, T.R. Golub, T. Jacks, Impaired microRNA processing enhances cellular transformation and tumorigenesis, Nat. Genet. 39 (2007) 673–677. A. Lujambio, M. Esteller, CpG island hypermethylation of tumor suppressor microRNAs in human cancer, Cell Cycle 6 (2007) 1455–1459. H. Kim, W. Huang, X. Jiang, B. Pennicooke, P.J. Park, M.D. Johnson, Integrative genome analysis reveals an oncomir/oncogene cluster regulating glioblastoma survivorship, Proc. Natl. Acad. Sci. USA 107 (2010) 2183–2188. J. Godlewski, M.O. Nowicki, A. Bronisz, S. Williams, A. Otsuki, G. Nuovo, A. Raychaudhury, H.B. Newton, E.A. Chiocca, S. Lawler, Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal, Cancer Res. 68 (2008) 9125–9130. A. Gaur, D.A. Jewell, Y. Liang, D. Ridzon, J.H. Moore, C. Chen, V.R. Ambros, M.A. Israel, Characterization of microRNA expression levels and their biological correlates in human cancer cell lines, Cancer Res. 67 (2007) 2456–2468. J.C. Pang, W.K. Kwok, Z. Chen, H.K. Ng, Oncogenic role of microRNAs in brain tumors, Acta Neuropathol. 117 (2009) 599–611. Y. Li, F. Guessous, Y. Zhang, C. diPierro, B. Kefas, E. Johnson, L. Marcinkiewicz, J. Jiang, Y. Yang, T.D. Schmittgen, B. Lopes, D. Schiff, B. Purow, R. Abounader, MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes, Cancer Res. 69 (2009) 7569–7576. J. Mei, R. Bachoo, C.L. Zhang, MicroRNA-146a inhibits glioma development by targeting Notch1, Mol. Cell Biol. 31 (2011) 3584–3592. B. Kefas, J. Godlewski, L. Comeau, Y. Li, R. Abounader, M. Hawkinson, J. Lee, H. Fine, E.A. Chiocca, S. Lawler, B. Purow, MicroRNA-7 inhibits the epidermal growth factor receptor and the Akt pathway and is down-regulated in glioblastoma, Cancer Res. 68 (2008) 3566–3572. T. Papagiannakopoulos, D. Friedmann-Morvinski, P. Neveu, J.C. Dugas, R.M. Gill, E. Huillard, C. Liu, H. Zong, D.H. Rowitch, B.A. Barres, I.M. Verma, and K.S. Kosik, Pro-neural miR-128 is a glioma tumor suppressor that targets mitogenic kinases, Oncogene (2011). Q.Q. Zhang, H. Xu, M.B. Huang, L.M. Ma, Q.J. Huang, Q. Yao, H. Zhou, L.H. Qu, MicroRNA-195 plays a tumor-suppressor role in human glioblastoma cells by targeting signaling pathways involved in cellular proliferation and invasion, Neuro-Oncology 14 (2012) 278–287. G. Gabriely, M. Yi, R.S. Narayan, J.M. Niers, T. Wurdinger, J. Imitola, K.L. Ligon, S. Kesari, C. Esau, R.M. Stephens, B.A. Tannous, A.M. Krichevsky, Human glioma growth is controlled by microRNA-10b, Cancer Res. 71 (2011) 3563– 3572. G. Gabriely, T. Wurdinger, S. Kesari, C.C. Esau, J. Burchard, P.S. Linsley, A.M. Krichevsky, MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators, Mol. Cell Biol. 28 (2008) 5369–5380. J.T. Huse, C. Brennan, D. Hambardzumyan, B. Wee, J. Pena, S.H. Rouhanifard, C. Sohn-Lee, S.C. le, R. Agami, T. Tuschl, E.C. Holland, The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo, Genes Dev. 23 (2009) 1327–1337. L. Jiang, C. Lin, L. Song, J. Wu, B. Chen, Z. Ying, L. Fang, X. Yan, M. He, J. Li, M. Li, MicroRNA-30e promotes human glioma cell invasiveness in an orthotopic xenotransplantation model by disrupting the NF-kappaB/IkappaBalpha negative feedback loop, J. Clin. Invest. 122 (2012) 33–47. C. Quintavalle, M. Garofalo, C. Zanca, G. Romano, M. Iaboni, C.M. del Basso De, J.C. Martinez-Montero, M. Incoronato, G. Nuovo, C.M. Croce, G. Condorelli,
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008
8
[89]
[90] [91]
[92]
[93]
[94]
[95]
[96]
[97]
[98]
[99]
[100] [101]
[102]
[103]
[104] [105] [106]
[107]
[108]
[109]
[110]
[111]
S. Kreth et al. / Cancer Letters xxx (2012) xxx–xxx MiR-221/222 overexpession in human glioblastoma increases invasiveness by targeting the protein phosphate PTPmu, Oncogene 31 (2012) 858–868. C.Z. Zhang, J.X. Zhang, A.L. Zhang, Z.D. Shi, L. Han, Z.F. Jia, W.D. Yang, G.X. Wang, T. Jiang, Y.P. You, P.Y. Pu, J.Q. Cheng, C.S. Kang, MiR-221 and miR-222 target PUMA to induce cell survival in glioblastoma, Mol. Cancer 9 (2010) 229. J.K. Gillies, I.A. Lorimer, Regulation of p27Kip1 by miRNA 221/222 in glioblastoma, Cell Cycle 6 (2007) 2005–2009. Y. Saito, G. Liang, G. Egger, J.M. Friedman, J.C. Chuang, G.A. Coetzee, P.A. Jones, Specific activation of microRNA-127 with downregulation of the protooncogene BCL6 by chromatin-modifying drugs in human cancer cells, Cancer cell 9 (2006) 435–443. A. Lujambio, S. Ropero, E. Ballestar, M.F. Fraga, C. Cerrato, F. Setien, S. Casado, A. Suarez-Gauthier, M. Sanchez-Cespedes, A. Git, I. Spiteri, P.P. Das, C. Caldas, E. Miska, M. Esteller, Genetic unmasking of an epigenetically silenced microRNA in human cancer cells, Cancer Res. 67 (2007) 1424–1429. R. Garzon, S. Liu, M. Fabbri, Z. Liu, C.E. Heaphy, E. Callegari, S. Schwind, J. Pang, J. Yu, N. Muthusamy, V. Havelange, S. Volinia, W. Blum, L.J. Rush, D. Perrotti, M. Andreeff, C.D. Bloomfield, J.C. Byrd, K. Chan, L.C. Wu, C.M. Croce, G. Marcucci, MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1, Blood 113 (2009) 6411–6418. E.J. Noonan, R.F. Place, D. Pookot, S. Basak, J.M. Whitson, H. Hirata, C. Giardina, R. Dahiya, MiR-449a targets HDAC-1 and induces growth arrest in prostate cancer, Oncogene 28 (2009) 1714–1724. Z. Zhang, H. Tang, Z. Wang, B. Zhang, W. Liu, H. Lu, L. Xiao, X. Liu, R. Wang, X. Li, M. Wu, G. Li, MiR-185 targets the DNA methyltransferases 1 and regulates global DNA methylation in human glioma, Mol. Cancer 10 (2011) 124. S. Varambally, Q. Cao, R.S. Mani, S. Shankar, X. Wang, B. Ateeq, B. Laxman, X. Cao, X. Jing, K. Ramnarayanan, J.C. Brenner, J. Yu, J.H. Kim, B. Han, P. Tan, C. Kumar-Sinha, R.J. Lonigro, N. Palanisamy, C.A. Maher, A.M. Chinnaiyan, Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer, Science 322 (2008) 1695–1699. M. Smits, J. Nilsson, S.E. Mir, P.M. van der Stoop, E. Hulleman, J.M. Niers, P.C. de Witt Hamer, V.E. Marquez, J. Cloos, A.M. Krichevsky, D.P. Noske, B.A. Tannous, T. Wurdinger, MiR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis, Oncotarget 1 (2010) 710–720. M. Berdasco, S. Ropero, F. Setien, M.F. Fraga, P. Lapunzina, R. Losson, M. Alaminos, N.K. Cheung, N. Rahman, M. Esteller, Epigenetic inactivation of the Sotos overgrowth syndrome gene histone methyltransferase NSD1 in human neuroblastoma and glioma, Proc. Natl. Acad. Sci. USA 106 (2009) 21830– 21835. M. Wang, H. Xie, W. Stellpflug, V. Rajaram, M.F. Bonaldo, S. Goldman, T. Tomita, M.B. Soares, BTECH: a platform to integrate genomic, transcriptomic and epigenomic alterations in brain tumors, Neuroinformatics 9 (2011) 59– 67. M. Esteller, Cancer epigenetics for the 21st century: what’s next?, Genes Cancer 2 (2011) 604–606 B. Malzkorn, M. Wolter, M.J. Riemenschneider, G. Reifenberger, Unraveling the glioma epigenome: from molecular mechanisms to novel biomarkers and therapeutic targets, Brain Pathol. 21 (2011) 619–632. M.J. Riemenschneider, J.W. Jeuken, P. Wesseling, G. Reifenberger, Molecular diagnostics of gliomas: state of the art, Acta Neuropathol. 120 (2010) 567– 584. B.S. Mann, J.R. Johnson, M.H. Cohen, R. Justice, R. Pazdur, FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma, Oncologist 12 (2007) 1247–1252. T.K. Kelly, D.D. De Carvalho, P.A. Jones, Epigenetic modifications as therapeutic targets, Nat. Biotechnol. 28 (2010) 1069–1078. A.A. Lane, B.A. Chabner, Histone deacetylase inhibitors in cancer therapy, J. Clin. Oncol. 27 (2009) 5459–5468. P. Chinnaiyan, G. Vallabhaneni, E. Armstrong, S.M. Huang, P.M. Harari, Modulation of radiation response by histone deacetylase inhibition, Int. J. Radiat. Oncol. Biol. Phys. 62 (2005) 223–229. D. Yin, J.M. Ong, J. Hu, J.C. Desmond, N. Kawamata, B.M. Konda, K.L. Black, H.P. Koeffler, Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor: effects on gene expression and growth of glioma cells in vitro and in vivo, Clin. Cancer Res. 13 (2007) 1045–1052. I.Y. Eyupoglu, E. Hahnen, R. Buslei, F.A. Siebzehnrubl, N.E. Savaskan, M. Luders, C. Trankle, W. Wick, M. Weller, R. Fahlbusch, I. Blumcke, Suberoylanilide hydroxamic acid (SAHA) has potent anti-glioma properties in vitro, ex vivo and in vivo, J. Neurochem. 93 (2005) 992–999. M. Entin-Meer, X. Yang, S.R. VandenBerg, K.R. Lamborn, A. Nudelman, A. Rephaeli, D.A. Haas-Kogan, In vivo efficacy of a novel histone deacetylase inhibitor in combination with radiation for the treatment of gliomas, NeuroOncology 9 (2007) 82–88. J. Drappatz, E.Q. Lee, S. Hammond, S.A. Grimm, A.D. Norden, R. Beroukhim, M. Gerard, D. Schiff, A.S. Chi, T.T. Batchelor, L.M. Doherty, A.S. Ciampa, D.C. Lafrankie, S. Ruland, S.M. Snodgrass, J.J. Raizer, P.Y. Wen, Phase I study of panobinostat in combination with bevacizumab for recurrent high-grade glioma, J. Neurooncol. 107 (2012) 133–138. A. Masoudi, M. Elopre, E. Amini, M.E. Nagel, J.L. Ater, V. Gopalakrishnan, J.E. Wolff, Influence of valproic acid on outcome of high-grade gliomas in children, Anticancer Res. 28 (2008) 2437–2442.
[112] E. Galanis, K.A. Jaeckle, M.J. Maurer, J.M. Reid, M.M. Ames, J.S. Hardwick, J.F. Reilly, A. Loboda, M. Nebozhyn, V.R. Fantin, V.M. Richon, B. Scheithauer, C. Giannini, P.J. Flynn, D.F. Moore Jr., J. Zwiebel, J.C. Buckner, Phase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study, J. Clin. Oncol. 27 (2009) 2052–2058. [113] M. Weller, T. Gorlia, J.G. Cairncross, M.J. van den Bent, W. Mason, K. Belanger, A.A. Brandes, U. Bogdahn, D.R. Macdonald, P. Forsyth, A.O. Rossetti, D. Lacombe, R.O. Mirimanoff, C.J. Vecht, R. Stupp, Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma, Neurology 77 (2011) 1156–1164. [114] K. Wang, S. Zhang, B. Marzolf, P. Troisch, A. Brightman, Z. Hu, L.E. Hood, D.J. Galas, Circulating microRNAs, potential biomarkers for drug-induced liver injury, Proc. Natl. Acad. Sci. USA 106 (2009) 4402–4407. [115] M.P. Hunter, N. Ismail, X. Zhang, B.D. Aguda, E.J. Lee, L. Yu, T. Xiao, J. Schafer, M.L. Lee, T.D. Schmittgen, S.P. Nana-Sinkam, D. Jarjoura, C.B. Marsh, Detection of microRNA expression in human peripheral blood microvesicles, PLoS One 3 (2008) e3694. [116] M.V. Iorio, C.M. Croce, MicroRNAs in cancer: small molecules with a huge impact, J. Clin. Oncol. 27 (2009) 5848–5856. [117] P. Roth, J. Wischhusen, C. Happold, P.A. Chandran, S. Hofer, G. Eisele, M. Weller, A. Keller, A specific miRNA signature in the peripheral blood of glioblastoma patients, J. Neurochem. 118 (2011) 449–457. [118] A. Baraniskin, J. Kuhnhenn, U. Schlegel, A. Maghnouj, H. Zollner, W. Schmiegel, S. Hahn, R. Schroers, Identification of microRNAs in the cerebrospinal fluid as biomarker for the diagnosis of glioma, NeuroOncology 14 (2012) 29–33. [119] A. Petri, M. Lindow, S. Kauppinen, MicroRNA silencing in primates: towards development of novel therapeutics, Cancer Res. 69 (2009) 393–395. [120] R.E. Lanford, E.S. Hildebrandt-Eriksen, A. Petri, R. Persson, M. Lindow, M.E. Munk, S. Kauppinen, H. Orum, Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection, Science 327 (2010) 198– 201. [121] R.E. van, W.S. Marshall, E.N. Olson, Toward microRNA-based therapeutics for heart disease: the sense in antisense, Circ. Res. 103 (2008) 919–928. [122] J. Elmen, M. Lindow, S. Schutz, M. Lawrence, A. Petri, S. Obad, M. Lindholm, M. Hedtjarn, H.F. Hansen, U. Berger, S. Gullans, P. Kearney, P. Sarnow, E.M. Straarup, S. Kauppinen, LNA-mediated microRNA silencing in non-human primates, Nature 452 (2008) 896–899. [123] J. Weiler, J. Hunziker, J. Hall, Anti-miRNA oligonucleotides (AMOs): ammunition to target miRNAs implicated in human disease?, Gene Ther 13 (2006) 496–502. [124] M.S. Ebert, J.R. Neilson, P.A. Sharp, MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells, Nat. Methods 4 (2007) 721–726. [125] D. Castanotto, J.J. Rossi, The promises and pitfalls of RNA-interference-based therapeutics, Nature 457 (2009) 426–433. [126] G. Cairncross, B. Berkey, E. Shaw, R. Jenkins, B. Scheithauer, D. Brachman, J. Buckner, K. Fink, L. Souhami, N. Laperierre, M. Mehta, W. Curran, Phase III trial of chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402, J. Clin. Oncol. 24 (2006) 2707–2714. [127] M.J. van den Bent, A.F. Carpentier, A.A. Brandes, M. Sanson, M.J. Taphoorn, H.J. Bernsen, M. Frenay, C.C. Tijssen, W. Grisold, L. Sipos, H. Haaxma-Reiche, J.M. Kros, M.C. van Kouwenhoven, C.J. Vecht, A. Allgeier, D. Lacombe, T. Gorlia, Adjuvant procarbazine, lomustine, and vincristine improves progression-free survival but not overall survival in newly diagnosed anaplastic oligodendrogliomas and oligoastrocytomas: a randomized European Organisation for Research and Treatment of Cancer phase III trial, J. Clin. Oncol. 24 (2006) 2715–2722. [128] H. Ohgaki, P. Kleihues, Genetic profile of astrocytic and oligodendroglial gliomas, Brain Tumor Pathol. 28 (2011) 177–183. [129] Y.H. Kim, S. Nobusawa, M. Mittelbronn, W. Paulus, B. Brokinkel, K. Keyvani, U. Sure, K. Wrede, Y. Nakazato, Y. Tanaka, A. Vital, L. Mariani, R. Stawski, T. Watanabe, G.U. De, P. Kleihues, H. Ohgaki, Molecular classification of lowgrade diffuse gliomas, Am. J. Pathol. 177 (2010) 2708–2714. [130] W. Stummer, U. Pichlmeier, T. Meinel, O.D. Wiestler, F. Zanella, H.J. Reulen, Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial, Lancet Oncol. 7 (2006) 392–401. [131] N. Thon, S. Eigenbrod, E.M. Grasbon-Frodl, M. Ruiter, J.H. Mehrkens, S. Kreth, J.C. Tonn, H.A. Kretzschmar, F.W. Kreth, Novel molecular stereotactic biopsy procedures reveal intratumoral homogeneity of loss of heterozygosity of 1p/ 19q and TP53 mutations in World Health Organization grade II gliomas, J. Neuropathol. Exp. Neurol. 68 (2009) 1219–1228. [132] N. Thon, S. Eigenbrod, E.M. Grasbon-Frodl, J. Lutz, S. Kreth, G. Popperl, C. Belka, H.A. Kretzschmar, J.C. Tonn, F.W. Kreth, Predominant influence of MGMT methylation in non-resectable glioblastoma after radiotherapy plus temozolomide, J. Neurol. Neurosurg. Psychiatry 82 (2011) 441–446. [133] E.R. Mardis, A decade’s perspective on DNA sequencing technology, Nature 470 (2011) 198–203. [134] E. Meaburn, R. Schulz, Next generation sequencing in epigenetics: insights and challenges, Semin. Cell Dev. Biol. (2011. [135] G. Riddick, H.A. Fine, Integration and analysis of genome-scale data from gliomas, Nat. Rev. Neurol. 7 (2011) 439–450.
Please cite this article in press as: S. Kreth et al., Epigenetics in human gliomas, Cancer Lett. (2012), http://dx.doi.org/10.1016/j.canlet.2012.04.008