1351
|
Abstract
Over the past decade, comprehensive sequencing efforts have revealed the genomic landscapes of common forms of human cancer. For most cancer types, this landscape consists of a small number of "mountains" (genes altered in a high percentage of tumors) and a much larger number of "hills" (genes altered infrequently). To date, these studies have revealed ~140 genes that, when altered by intragenic mutations, can promote or "drive" tumorigenesis. A typical tumor contains two to eight of these "driver gene" mutations; the remaining mutations are passengers that confer no selective growth advantage. Driver genes can be classified into 12 signaling pathways that regulate three core cellular processes: cell fate, cell survival, and genome maintenance. A better understanding of these pathways is one of the most pressing needs in basic cancer research. Even now, however, our knowledge of cancer genomes is sufficient to guide the development of more effective approaches for reducing cancer morbidity and mortality.
Collapse
Affiliation(s)
- Bert Vogelstein
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Nickolas Papadopoulos
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Victor E. Velculescu
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Shibin Zhou
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Luis A. Diaz
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Kenneth W. Kinzler
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| |
Collapse
|
1352
|
Abstract
Chemical modifications of histones and DNA, such as histone methylation, histone acetylation, and DNA methylation, play critical roles in epigenetic gene regulation. Many of the enzymes that add or remove such chemical modifications are known, or might be suspected, to be sensitive to changes in intracellular metabolism. This knowledge provides a conceptual foundation for understanding how mutations in the metabolic enzymes SDH, FH, and IDH can result in cancer and, more broadly, for how alterations in metabolism and nutrition might contribute to disease. Here, we review literature pertinent to hypothetical connections between metabolic and epigenetic states in eukaryotic cells.
Collapse
Affiliation(s)
- William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Steven L. McKnight
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
1353
|
Sahm F, Lass U, Herold-Mende C, von Deimling A, Hartmann C, Mueller W. Analysis of CIC-associated CpG island methylation in oligoastrocytoma. Neuropathol Appl Neurobiol 2013; 39:831-6. [PMID: 23521081 DOI: 10.1111/nan.12045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 03/15/2013] [Indexed: 11/29/2022]
Abstract
AIMS Combined deletion of the whole chromosomal arms 1p and 19q is a frequent event in oligodendroglial tumours. Recent identification of recurrent mutations in CIC on 19q and FUBP1 on 1p and their mutational patterns suggest a loss of function of the respective proteins. Surprisingly, oligoastrocytomas harbouring identical genetic characteristics regarding 1p/19q codeletion and frequent IDH1/2 mutations have been shown to carry CIC mutations in a significantly lower number of cases. The present study investigates whether epigenetic modification may result in silencing of CIC. METHODS As IDH1/2 mutation-mediated DNA hypermethylation is a prominent feature of these tumours, we analysed a set of CIC wild-type oligoastrocytomas and other diffuse gliomas with regard to 1p/19q status for presence of CIC-associated CpG island methylation by methylation-specific PCR. RESULTS Both methylation-specific PCR and subsequent bisulphite-sequencing of selected cases revealed an unmethylated status in all samples. CONCLUSION Despite the hypermethylator phenotype in IDH1/2 mutant tumours and recent detection of gene silencing particularly on retained alleles in oligodendroglial tumours, hypermethylation of CIC-associated CpG islands does not provide an alternative mechanism of functional CIC protein abrogation.
Collapse
Affiliation(s)
- F Sahm
- Department of Neuropathology, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology G380, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
1354
|
Ho AS, Turcan S, Chan TA. Epigenetic therapy: use of agents targeting deacetylation and methylation in cancer management. Onco Targets Ther 2013; 6:223-32. [PMID: 23569385 PMCID: PMC3615839 DOI: 10.2147/ott.s34680] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The emergence of epigenetic mechanisms as key regulators of gene expression has led to dramatic advances in understanding cancer biology. Driven by complex layers that include aberrant DNA methylation and histone modification, epigenetic aberrations have emerged as critical processes that disrupt cellular machinery and homeostasis. Recent discoveries have already translated into successful clinical trials and improved patient care, with several agents approved for hematologic disease and others undergoing study. As the field matures, substantial challenges persist that will require resolution. These include the need to decipher more fully the interplay between the epigenetic and genetic machinery, patient selection and improving treatment efficacy in solid tumors, and optimizing combination therapies to counteract chemoresistance and minimize adverse effects. Here, we review recent progress in epigenetic treatments and consider their implications for future cancer therapy.
Collapse
Affiliation(s)
- Allen S Ho
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
1355
|
Sze CI, Su WP, Chiang MF, Lu CY, Chen YA, Chang NS. Assessing current therapeutic approaches to decode potential resistance mechanisms in glioblastomas. Front Oncol 2013; 3:59. [PMID: 23516171 PMCID: PMC3601334 DOI: 10.3389/fonc.2013.00059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 03/04/2013] [Indexed: 12/12/2022] Open
Abstract
Unique astrocytic cell infiltrating growth and glial tumor growth in the confined skull make human glioblastoma (GBM) one of the most difficult cancers to treat in modern medicine. Prognosis for patients is very poor, as they die more or less within 12 months. Patients either die of the cancer itself, or secondary complications such as cerebral edema, herniations, or hemorrhages. GBMs rarely metastasize to other organs. However, GBM recurrence associated with resistance to therapeutic drugs is common. Patients die shortly after relapse. GBM is indeed an outstanding cancer model to search for potential mechanisms for drug resistance. Here, we reviewed the current cancer biology of gliomas and their pathophysiological events that contribute to the development of therapeutic resistance. We have addressed the potential roles of cancer stem cells, epigenetic modifications, and epithelial mesenchymal transition (EMT) in the development of resistance to inhibitor drugs in GBMs. The potential role of TIAF1 (TGF-β-induced antiapoptotic factor) overexpression and generation of intratumor amyloid fibrils for conferring drug resistance in GBMs is discussed.
Collapse
Affiliation(s)
- Chun-I Sze
- Department of Anatomy and Cell Biology, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
1356
|
A novel, diffusely infiltrative xenograft model of human anaplastic oligodendroglioma with mutations in FUBP1, CIC, and IDH1. PLoS One 2013; 8:e59773. [PMID: 23527265 PMCID: PMC3602110 DOI: 10.1371/journal.pone.0059773] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 02/18/2013] [Indexed: 12/28/2022] Open
Abstract
Oligodendroglioma poses a biological conundrum for malignant adult human gliomas: it is a tumor type that is universally incurable for patients, and yet, only a few of the human tumors have been established as cell populations in vitro or as intracranial xenografts in vivo. Their survival, thus, may emerge only within a specific environmental context. To determine the fate of human oligodendroglioma in an experimental model, we studied the development of an anaplastic tumor after intracranial implantation into enhanced green fluorescent protein (eGFP) positive NOD/SCID mice. Remarkably after nearly nine months, the tumor not only engrafted, but it also retained classic histological and genetic features of human oligodendroglioma, in particular cells with a clear cytoplasm, showing an infiltrative growth pattern, and harboring mutations of IDH1 (R132H) and of the tumor suppressor genes, FUBP1 and CIC. The xenografts were highly invasive, exhibiting a distinct migration and growth pattern around neurons, especially in the hippocampus, and following white matter tracts of the corpus callosum with tumor cells accumulating around established vasculature. Although tumors exhibited a high growth fraction in vivo, neither cells from the original patient tumor nor the xenograft exhibited significant growth in vitro over a six-month period. This glioma xenograft is the first to display a pure oligodendroglioma histology and expression of R132H. The unexpected property, that the cells fail to grow in vitro even after passage through the mouse, allows us to uniquely investigate the relationship of this oligodendroglioma with the in vivo microenvironment.
Collapse
|
1357
|
Breathing-in epigenetic change with vitamin C. EMBO Rep 2013; 14:337-46. [PMID: 23492828 PMCID: PMC3615655 DOI: 10.1038/embor.2013.29] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 02/19/2013] [Indexed: 01/01/2023] Open
Abstract
Vitamin C is an antioxidant that maintains the activity of iron and α-ketoglutarate-dependent dioxygenases. Despite these enzymes being implicated in a wide range of biological pathways, vitamin C is rarely included in common cell culture media. Recent studies show that reprogramming of pluripotent stem cells is enhanced when vitamin C is present, thereby illustrating previous limitations in reprogramming cultures. Here, we summarize understanding of dioxygenase function in reprogramming and epigenetic regulation. The available data suggest a link between dioxygenase function and stem cell differentiation, which is exposed to environmental influence and is relevant for human disease.
Collapse
|
1358
|
Ohno M, Narita Y, Miyakita Y, Matsushita Y, Yoshida A, Fukushima S, Ichimura K, Shibui S. Secondary glioblastomas with IDH1/2 mutations have longer glioma history from preceding lower-grade gliomas. Brain Tumor Pathol 2013; 30:224-32. [PMID: 23494632 DOI: 10.1007/s10014-013-0140-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/19/2013] [Indexed: 11/30/2022]
Abstract
Isocitrate dehydrogenase (IDH)1/2 mutations have been proposed as a genetic marker for secondary glioblastoma (sGBM). This study aimed to evaluate the impact of the IDH1/2 mutations on the clinical course and genetic alterations of sGBMs, which histopathologically progressed from lower-grade gliomas. We investigated 18 sGBMs, including 8 sGBMs with IDH1/2 mutations (sGBM-Mut) and 10 with wild-type IDH1/2 (sGBM-Wt). The median overall survival time of patients with sGBM-Mut was significantly longer than that of patients with sGBM-Wt (68.2 vs. 25.3 months). The median time from initial diagnosis to sGBM diagnosis was also significantly longer for sGBM-Mut than for sGBM-Wt (50.1 vs. 13.4 months). There was no difference in the median survival time from the sGBM diagnosis between sGBM-Mut and sGBM-Wt (6.75 vs. 6.8 months). All sGBM-Mut (7 of 7) and 6 of 9 sGBM-Wt had TP53 mutations, and the remaining one-thirds of sGBM-Wt had neither TP53 mutations nor 1p/19q codeletion. These observations suggest that IDH1/2 mutations have an impact on the glioma history of sGBM with different genetic pathway. The aggressive progression to sGBM-Wt suggest the need for more intense treatment to the IDH1/2 wild-type tumors.
Collapse
Affiliation(s)
- Makoto Ohno
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | | | | | | | | | | | | | | |
Collapse
|
1359
|
NNMT promotes epigenetic remodeling in cancer by creating a metabolic methylation sink. Nat Chem Biol 2013; 9:300-6. [PMID: 23455543 PMCID: PMC3631284 DOI: 10.1038/nchembio.1204] [Citation(s) in RCA: 306] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/06/2013] [Indexed: 12/11/2022]
Abstract
Nicotinamide N-methyltransferase (NNMT) is overexpressed in a variety of human cancers, where it contributes to tumorigenesis by a still poorly understood mechanism. Here, we show using metabolomics that NNMT impairs the methylation potential of cancer cells by consuming methyl units from S-adenosyl methionine to create the stable metabolic product 1-methylnicotinamide. As a result, NNMT-expressing cancer cells possess an altered epigenetic state that includes hypomethylated histones and other cancer-related proteins combined with heightened expression of pro-tumorigenic gene products. Our findings thus point to a direct mechanistic link between the deregulation of a metabolic enzyme and widespread changes in the methylation landscape of cancer cells.
Collapse
|
1360
|
Venneti S, Thompson CB. Metabolic modulation of epigenetics in gliomas. Brain Pathol 2013; 23:217-21. [PMID: 23432648 PMCID: PMC3615671 DOI: 10.1111/bpa.12022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 12/29/2012] [Indexed: 01/18/2023] Open
Abstract
Cancer metabolism and epigenetics are two relatively new areas of cancer research. Recent years have seen an explosion of studies implicating either altered tumor metabolism or epigenetic mechanisms in the pathogenesis or maintenance of brain tumors. A new paradigm is emerging in cancer biology that represents a convergence of these themes, the metabolic regulation of epigenetics. We discuss this interrelationship in the context of two metabolic enzymes that can influence the pathogenesis of gliomas by altering the epigenetic state. The first of these enzymes is isocitrate dehydrogenase 1 (IDH1), which is mutated in secondary glioblastomas and ~70% of grade II/III astrocytomas and oligodendrogliomas. Mutant IDH1 results in the production of a metabolite 2-hydroxyglutarate (2-HG) that can inhibit DNA and histone demethylating enzymes resulting in the glioma-CpG island phenotype (G-CIMP) and increased histone methylation marks. Pyruvate kinase M2 (PKM2), an enzyme that plays a critical role in the glycolytic pathway, is a second example of a metabolic enzyme that can affect histone modifications. In epidermal growth factor receptor (EGFR)-driven glioblastoma, PKM2 translocates to the nucleus and phosphorylates histone 3 at threonine 11 (H3-T11). This causes dissociation of HDAC3 from the CCND1 (Cyclin D1) and c-MYC promoters and subsequent histone acetylation, leading to transcription of Cyclin-D1 and c-MYC, and subsequent cell proliferation. Modification of the epigenetic state by alterations in metabolic enzymes is a novel phenomenon that contributes to the pathogenesis of gliomas and may help in the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Sriram Venneti
- Cancer Biology and Genetics ProgramMemorial Sloan‐Kettering Cancer CenterNew YorkNY
| | - Craig B. Thompson
- Cancer Biology and Genetics ProgramMemorial Sloan‐Kettering Cancer CenterNew YorkNY
| |
Collapse
|
1361
|
Kannan K, Inagaki A, Silber J, Gorovets D, Zhang J, Kastenhuber ER, Heguy A, Petrini JH, Chan TA, Huse JT. Whole-exome sequencing identifies ATRX mutation as a key molecular determinant in lower-grade glioma. Oncotarget 2013; 3:1194-203. [PMID: 23104868 PMCID: PMC3717947 DOI: 10.18632/oncotarget.689] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The molecular foundations of lower-grade gliomas (LGGs)—astrocytoma, oligodendroglioma, and oligoastrocytoma—remain less well characterized than those of their fully malignant counterpart, glioblastoma. Mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2) likely represent initiating pathogenic events. However, while IDH mutations appear to dramatically alter cellular epigenomic landscapes, definitive downstream transformative mechanisms have not been characterized. It remains likely, therefore, that additional genomic abnormalities collaborate with IDH mutation to drive oncogenesis in LGG. We performed whole exome sequencing in 4 LGGs, followed by focused resequencing in an additional 28, and found a high incidence of mutations in the ATRX gene (α thalassemia/mental retardation syndrome X-linked). ATRX forms a core component of a chromatin remodeling complex active in telomere biology. Mutations in ATRX have been identified in multiple tumor types and appear to cause alternative lengthening of telomeres (ALT), a presumed precursor to genomic instability. In our samples, ATRX mutation was entirely restricted to IDH-mutant tumors, closely correlated with TP53 mutation and astrocytic differentiation, and mutually exclusive with 1p/19q codeletion, the molecular hallmark of oligodendroglioma. Moreover, ATRX mutation was highly enriched in tumors of so-called early progenitor-like transcriptional subclass (~85%), which our prior work has linked to specific cells of origin in the forebrain subventricular zone. Finally, ATRX mutation correlated with ALT, providing a mechanistic link to genomic instability. In summary, our findings both identify ATRX mutation as a defining molecular determinant for a large subset of IDH-mutant gliomas and have direct implications on pathogenic mechanisms across the wide spectrum of LGGs.
Collapse
Affiliation(s)
- Kasthuri Kannan
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1362
|
Kurek KC, Pansuriya TC, van Ruler MAJH, van den Akker B, Luks VL, Verbeke SLJ, Kozakewich HP, Sciot R, Lev D, Lazar AJ, Fletcher CDM, Bovée JVMG. R132C IDH1 mutations are found in spindle cell hemangiomas and not in other vascular tumors or malformations. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1494-500. [PMID: 23485734 DOI: 10.1016/j.ajpath.2013.01.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/22/2012] [Accepted: 01/07/2013] [Indexed: 11/29/2022]
Abstract
Spindle cell hemangioma (SCH) is a rare, benign vascular tumor of the dermis and subcutis. The lesions can be multifocal and are overrepresented in Maffucci syndrome, in which patients also have multiple enchondromas. Somatic mosaic R132C IDH1 hotspot mutations were recently identified in Maffucci syndrome. We evaluated the presence of mutations in solitary and multiple SCHs in patients without multiple enchondromas and tested a range of other vascular lesions that enter into the differential diagnosis. The R132C IDH1 mutation was identified by hydrolysis probes assay and confirmed by Sanger sequencing in 18 of 28 (64%) SCHs; of the 10 negative cases, 2 harbored a mutation in IDH2 (R172T and R172M) by Sanger sequencing. None of 154 other vascular malformations and tumors harbored an IDH1 R132C mutation, and R132H IDH1 mutations were absent in all 182 cases. All 16 SCHs examined by immunohistochemistry were negative for expression of HIF-1α. In conclusion, 20 of 28 (71%) SCHs harbored mutations in exon 4 of IDH1 or IDH2. Given that mutations were absent in 154 other vascular lesions, the mutation seems to be highly specific for SCH. The mutation does not induce expression of HIF-1α in SCH, and therefore the exact mechanism by which mutations in IDH1 or IDH2 lead to vascular tumorigenesis remains to be established.
Collapse
Affiliation(s)
- Kyle C Kurek
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1363
|
Introduction. Brain Pathol 2013; 23:192. [PMID: 23432643 DOI: 10.1111/bpa.12030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
1364
|
Popov S, Jury A, Laxton R, Doey L, Kandasamy N, Al-Sarraj S, Jürgensmeier JM, Jones C. IDH1-associated primary glioblastoma in young adults displays differential patterns of tumour and vascular morphology. PLoS One 2013; 8:e56328. [PMID: 23451042 PMCID: PMC3579823 DOI: 10.1371/journal.pone.0056328] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/08/2013] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma is a highly aggressive tumour with marked heterogeneity at the morphological level in both the tumour cells and the associated highly prominent vasculature. As we begin to develop an increased biological insight into the underlying processes driving the disease, fewer attempts have thus far been made to understand these phenotypic differences. We sought to address this by carefully assessing the morphological characteristics of both the tumour cells and the associated vasculature, relating these observations to the IDH1/MGMT status, with a particular focus on the early onset population of young adults who develop primary glioblastoma. 276 primary glioblastoma specimens were classified into their predominant cell morphological type (fibrillary, gemistocytic, giant cell, small cell, oligodendroglial, sarcomatous), and assessed for specific tumour (cellularity, necrosis, palisades) and vascular features (glomeruloid structures, arcades, pericyte proliferation). IDH1 positive glioblastomas were associated with a younger age at diagnosis, better clinical outcome, prominent oligodendroglial and small cell tumour cell morphology, pallisading necrosis and glomeruloid vascular proliferation in the absence of arcade-like structures. These features widen the phenotype of IDH1 mutation-positive primary glioblastoma in young adults and provide correlative evidence for a functional role of mutant IDH1 in the differential nature of neo-angiogenesis in different subtypes of glioblastoma.
Collapse
Affiliation(s)
- Sergey Popov
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, United Kingdom
| | - Alexa Jury
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, United Kingdom
| | - Ross Laxton
- Department of Neuropathology, King’s College Hospital, London, United Kingdom
| | - Lawrence Doey
- Department of Neuropathology, King’s College Hospital, London, United Kingdom
| | - Naga Kandasamy
- Department of Neuroradiology, King’s College Hospital, London, United Kingdom
| | - Safa Al-Sarraj
- Department of Neuropathology, King’s College Hospital, London, United Kingdom
| | | | - Chris Jones
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, United Kingdom
- * E-mail:
| |
Collapse
|
1365
|
Marabita F, Almgren M, Lindholm ME, Ruhrmann S, Fagerström-Billai F, Jagodic M, Sundberg CJ, Ekström TJ, Teschendorff AE, Tegnér J, Gomez-Cabrero D. An evaluation of analysis pipelines for DNA methylation profiling using the Illumina HumanMethylation450 BeadChip platform. Epigenetics 2013; 8:333-46. [PMID: 23422812 PMCID: PMC3669124 DOI: 10.4161/epi.24008] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The proper identification of differentially methylated CpGs is central in most epigenetic studies. The Illumina HumanMethylation450 BeadChip is widely used to quantify DNA methylation; nevertheless, the design of an appropriate analysis pipeline faces severe challenges due to the convolution of biological and technical variability and the presence of a signal bias between Infinium I and II probe design types. Despite recent attempts to investigate how to analyze DNA methylation data with such an array design, it has not been possible to perform a comprehensive comparison between different bioinformatics pipelines due to the lack of appropriate data sets having both large sample size and sufficient number of technical replicates. Here we perform such a comparative analysis, targeting the problems of reducing the technical variability, eliminating the probe design bias and reducing the batch effect by exploiting two unpublished data sets, which included technical replicates and were profiled for DNA methylation either on peripheral blood, monocytes or muscle biopsies. We evaluated the performance of different analysis pipelines and demonstrated that: (1) it is critical to correct for the probe design type, since the amplitude of the measured methylation change depends on the underlying chemistry; (2) the effect of different normalization schemes is mixed, and the most effective method in our hands were quantile normalization and Beta Mixture Quantile dilation (BMIQ); (3) it is beneficial to correct for batch effects. In conclusion, our comparative analysis using a comprehensive data set suggests an efficient pipeline for proper identification of differentially methylated CpGs using the Illumina 450K arrays.
Collapse
Affiliation(s)
- Francesco Marabita
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1366
|
Leu S, von Felten S, Frank S, Vassella E, Vajtai I, Taylor E, Schulz M, Hutter G, Hench J, Schucht P, Boulay JL, Mariani L. IDH/MGMT-driven molecular classification of low-grade glioma is a strong predictor for long-term survival. Neuro Oncol 2013; 15:469-79. [PMID: 23408861 DOI: 10.1093/neuonc/nos317] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Low-grade gliomas (LGGs) are rare brain neoplasms, with survival spanning up to a few decades. Thus, accurate evaluations on how biomarkers impact survival among patients with LGG require long-term studies on samples prospectively collected over a long period. METHODS The 210 adult LGGs collected in our databank were screened for IDH1 and IDH2 mutations (IDHmut), MGMT gene promoter methylation (MGMTmet), 1p/19q loss of heterozygosity (1p19qloh), and nuclear TP53 immunopositivity (TP53pos). Multivariate survival analyses with multiple imputation of missing data were performed using either histopathology or molecular markers. Both models were compared using Akaike's information criterion (AIC). The molecular model was reduced by stepwise model selection to filter out the most critical predictors. A third model was generated to assess for various marker combinations. RESULTS Molecular parameters were better survival predictors than histology (ΔAIC = 12.5, P< .001). Forty-five percent of studied patients died. MGMTmet was positively associated with IDHmut (P< .001). In the molecular model with marker combinations, IDHmut/MGMTmet combined status had a favorable impact on overall survival, compared with IDHwt (hazard ratio [HR] = 0.33, P< .01), and even more so the triple combination, IDHmut/MGMTmet/1p19qloh (HR = 0.18, P< .001). Furthermore, IDHmut/MGMTmet/TP53pos triple combination was a significant risk factor for malignant transformation (HR = 2.75, P< .05). CONCLUSION By integrating networks of activated molecular glioma pathways, the model based on genotype better predicts prognosis than histology and, therefore, provides a more reliable tool for standardizing future treatment strategies.
Collapse
Affiliation(s)
- Severina Leu
- Department of Biomedicine, University Hospital of Basel, Spitalstrasse 21, CH-4031 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1367
|
CEBPA-CEBPG axis as a novel promising therapeutic target in acute myeloid leukemia. Acta Pharmacol Sin 2013; 34:185-6. [PMID: 23381107 DOI: 10.1038/aps.2012.191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
1368
|
van den Bent MJ, Hartmann C, Preusser M, Ströbel T, Dubbink HJ, Kros JM, von Deimling A, Boisselier B, Sanson M, Halling KC, Diefes KL, Aldape K, Giannini C. Interlaboratory comparison of IDH mutation detection. J Neurooncol 2013; 112:173-8. [PMID: 23358936 DOI: 10.1007/s11060-013-1056-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 01/14/2013] [Indexed: 11/28/2022]
Abstract
Isocitrate dehydrogenase (IDH) mutational testing is becoming increasingly important. For this, robust and reliable assays are needed. We tested the variation of results between six laboratories of testing for IDH mutations. Each laboratory received five unstained slides from 31 formalin-fixed paraffin-embedded (FFPE) glioma samples, and followed its own standard IDH diagnostic routine. All laboratories used immunohistochemistry (IHC) with an antibody against the most frequent IDH1 mutation (R132H) as a first step. Three laboratories then sequenced only IHC negative cases while the others sequenced all cases. Based on the overall analysis, 13 samples from 11 tumors had an R132H mutation and one tumor showed an R132G mutation. Results of IHC for IDH1 R132H mutations in all six laboratories were completely in agreement, and identified all R132H mutations. Upon sequencing the results of two laboratories deviated from those of the others. After a review of the entire diagnostic process, on repeat (blinded) testing one laboratory was completely in agreement with the overall result. A change in technique did only partially improve the results in the other laboratory. IHC for the IDH1 R132H mutation is very reliable and consistent across laboratories. IDH sequencing procedures yielded inconsistent results in 2 out of 6 laboratories. Quality assurance is pivotal before IDH testing is made part of clinical management of patients.
Collapse
Affiliation(s)
- Martin J van den Bent
- Department of Neurology/Neuro-Oncology, Erasmus MC - Cancer Institute, Groene Hilledijk 301, 3075EA, Rotterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1369
|
Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A 2013; 110:2916-21. [PMID: 23359684 DOI: 10.1073/pnas.1222577110] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Uterine serous carcinoma (USC) is a biologically aggressive subtype of endometrial cancer. We analyzed the mutational landscape of USC by whole-exome sequencing of 57 cancers, most of which were matched to normal DNA from the same patients. The distribution of the number of protein-altering somatic mutations revealed that 52 USC tumors had fewer than 100 (median 36), whereas 5 had more than 3,000 somatic mutations. The mutations in these latter tumors showed hallmarks of defects in DNA mismatch repair. Among the remainder, we found a significantly increased burden of mutation in 14 genes. In addition to well-known cancer genes (i.e., TP53, PIK3CA, PPP2R1A, KRAS, FBXW7), there were frequent mutations in CHD4/Mi2b, a member of the NuRD-chromatin-remodeling complex, and TAF1, an element of the core TFIID transcriptional machinery. Additionally, somatic copy-number variation was found to play an important role in USC, with 13 copy-number gains and 12 copy-number losses that occurred more often than expected by chance. In addition to loss of TP53, we found frequent deletion of a small segment of chromosome 19 containing MBD3, also a member of the NuRD-chromatin-modification complex, and frequent amplification of chromosome segments containing PIK3CA, ERBB2 (an upstream activator of PIK3CA), and CCNE1 (a target of FBXW7-mediated ubiquitination). These findings identify frequent mutation of DNA damage, chromatin remodeling, cell cycle, and cell proliferation pathways in USC and suggest potential targets for treatment of this lethal variant of endometrial cancer.
Collapse
|
1370
|
van den Bent MJ, Brandes AA, Taphoorn MJ, Kros JM, Kouwenhoven MC, Delattre JY, Bernsen HJ, Frenay M, Tijssen CC, Grisold W, Sipos L, Enting RH, French PJ, Dinjens WN, Vecht CJ, Allgeier A, Lacombe D, Gorlia T, Hoang-Xuan K. Adjuvant Procarbazine, Lomustine, and Vincristine Chemotherapy in Newly Diagnosed Anaplastic Oligodendroglioma: Long-Term Follow-Up of EORTC Brain Tumor Group Study 26951. J Clin Oncol 2013; 31:344-50. [PMID: 23071237 DOI: 10.1200/jco.2012.43.2229] [Citation(s) in RCA: 804] [Impact Index Per Article: 73.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Anaplastic oligodendroglioma are chemotherapy-sensitive tumors. We now present the long-term follow-up findings of a randomized phase III study on the addition of six cycles of procarbazine, lomustine, and vincristine (PCV) chemotherapy to radiotherapy (RT). Patients and Methods Adult patients with newly diagnosed anaplastic oligodendroglial tumors were randomly assigned to either 59.4 Gy of RT or the same RT followed by six cycles of adjuvant PCV. An exploratory analysis of the correlation between 1p/19q status and survival was part of the study. Retrospectively, the methylation status of the methyl-guanine methyl transferase gene promoter and the mutational status of the isocitrate dehydrogenase (IDH) gene were determined. The primary end points were overall survival (OS) and progression-free survival based on intent-to-treat analysis. Results A total of 368 patients were enrolled. With a median follow-up of 140 months, OS in the RT/PCV arm was significantly longer (42.3 v 30.6 months in the RT arm, hazard ratio [HR], 0.75; 95% CI, 0.60 to 0.95). In the 80 patients with a 1p/19q codeletion, OS was increased, with a trend toward more benefit from adjuvant PCV (OS not reached in the RT/PCV group v 112 months in the RT group; HR, 0.56; 95% CI, 0.31 to 1.03). IDH mutational status was also of prognostic significance. Conclusion The addition of six cycles of PCV after 59.4 Gy of RT increases both OS and PFS in anaplastic oligodendroglial tumors. 1p/19q-codeleted tumors derive more benefit from adjuvant PCV compared with non–1p/19q-deleted tumors.
Collapse
Affiliation(s)
- Martin J. van den Bent
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Alba A. Brandes
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Martin J.B. Taphoorn
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Johan M. Kros
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Mathilde C.M. Kouwenhoven
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Jean-Yves Delattre
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Hans J.J.A. Bernsen
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Marc Frenay
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Cees C. Tijssen
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Wolfgang Grisold
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - László Sipos
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Roelien H. Enting
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Pim J. French
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Winand N.M. Dinjens
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Charles J. Vecht
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Anouk Allgeier
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Denis Lacombe
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Thierry Gorlia
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| | - Khê Hoang-Xuan
- Martin J. van den Bent, Johan M. Kros, Mathilde C.M. Kouwenhoven, Roelien H. Enting, Pim J. French, and Winand N.M. Dinjens, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam; Martin J.B. Taphoorn and Charles J. Vecht, Medical Center Haaglanden, The Hague; Martin J.B. Taphoorn, Vrije Universiteit Medisch Centrum, Amsterdam; Hans J.J.A. Bernsen, Canisius Wilhelmina Ziekenhuis, Nijmegen; Cees C. Tijssen, St Elisabeth Hospital, Tilburg; Roelien H. Enting, University Medical Center Groningen, Groningen,
| |
Collapse
|
1371
|
Gozé C, Mansour L, Rigau V, Duffau H. Distinct IDH1/IDH2 mutation profiles in purely insular versus paralimbic WHO Grade II gliomas. J Neurosurg 2013; 118:866-72. [PMID: 23330999 DOI: 10.3171/2012.11.jns121100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The molecular profile of diffuse WHO Grade II gliomas involving the insular lobe, with a possible impact on outcome, is controversial. The authors undertook this study to investigate a possible difference of molecular patterns between purely insular Grade II gliomas and paralimbic Grade II gliomas that involve both the insular lobe and the frontal and/or temporal structures. METHODS From a consecutive series of 47 patients who underwent resection of a Grade II glioma invading the insula, 2 subgroups were identified. The first subgroup included 11 patients with a purely insular tumor. The second subgroup included 36 patients with a paralimbic Grade II glioma also involving the frontal and/or temporal lobe. The authors searched systematically for TP53 mutations, 1p19q codeletion, and IDH1/IDH2 mutations. RESULTS There was no significant difference between the 2 subgroups with respect to 1p19q codeletion or TP53 mutations rates. Conversely, IDH1/IDH2 mutations were found in all 11 (100%) of the insular Grade II gliomas but only 20 (55%) of 36 paralimbic Grade II gliomas (p = 0.008). Ten (28%) of the 36 patients in the paralimbic tumor group experienced a malignant transformation, and 6 of them died; whereas neither transformation nor death occurred in the insular tumor group (trend toward significance, p = 0.088). CONCLUSIONS These findings demonstrate for the first time distinct IDH1/IDH2 and consequently distinct "triplenegative" patterns in purely insular versus paralimbic Grade II gliomas. Such findings could explain discrepancies reported in the literature, because insular and paralimbic gliomas have not been separated in previous reports. These results may enable physicians to refine the management of Grade II gliomas involving the insula according to the presence or lack of invasion of the frontal and/or temporal areas.
Collapse
Affiliation(s)
- Catherine Gozé
- Hormone and Cell Biology Laboratory, Hôpital Arnaud de Villeneuve, Montpellier University Medical Center, Montpellier, France.
| | | | | | | |
Collapse
|
1372
|
Hatzimichael E, Georgiou G, Benetatos L, Briasoulis E. Gene mutations and molecularly targeted therapies in acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2013; 3:29-51. [PMID: 23358589 PMCID: PMC3555190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/07/2013] [Indexed: 06/01/2023]
Abstract
Acute myelogenous leukemia (AML) can progress quickly and without treatment can become fatal in a short period of time. However, over the last 30 years fine-tuning of therapeutics have increased the rates of remission and cure. Cytogenetics and mutational gene profiling, combined with the option of allogeneic hematopoietic stem cell transplantation offered in selected patients have further optimized AML treatment on a risk stratification basis in younger adults. However there is still an unmet medical need for effective therapies in AML since disease relapses in almost half of adult patients becoming refractory to salvage therapy. Improvements in the understanding of molecular biology of cancer and identification of recurrent mutations in AML provide opportunities to develop targeted therapies and improve the clinical outcome. In the spectrum of identified gene mutations, primarily targetable lesions are gain of function mutations of tyrosine kinases FLT3, JAK2 and cKIT for which specific, dual and multi-targeted small molecule inhibitors have been developed. A number of targeted compounds such as sorafenib, quizartinib, lestaurtinib, midostaurin, pacritinib, PLX3397 and CCT137690 are in clinical development. For loss-of-function gene mutations, which are mostly biomarkers of favorable prognosis, combined therapeutic approaches can maximize the therapeutic efficacy of conventional therapy. Apart from mutated gene products, proteins aberrantly overexpressed in AML appear to be clinically significant therapeutic targets. Such a molecule for which targeted inhibitors are currently in clinical development is PLK1. We review characteristic gene mutations, discuss their biological functions and clinical significance and present small molecule compounds in clinical development, which are expected to have a role in treating AML subtypes with characteristic molecular alterations.
Collapse
|
1373
|
Varley KE, Gertz J, Bowling KM, Parker SL, Reddy TE, Pauli-Behn F, Cross MK, Williams BA, Stamatoyannopoulos JA, Crawford GE, Absher DM, Wold BJ, Myers RM. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res 2013; 23:555-67. [PMID: 23325432 PMCID: PMC3589544 DOI: 10.1101/gr.147942.112] [Citation(s) in RCA: 520] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As studies of DNA methylation increase in scope, it has become evident that methylation has a complex relationship with gene expression, plays an important role in defining cell types, and is disrupted in many diseases. We describe large-scale single-base resolution DNA methylation profiling on a diverse collection of 82 human cell lines and tissues using reduced representation bisulfite sequencing (RRBS). Analysis integrating RNA-seq and ChIP-seq data illuminates the functional role of this dynamic mark. Loci that are hypermethylated across cancer types are enriched for sites bound by NANOG in embryonic stem cells, which supports and expands the model of a stem/progenitor cell signature in cancer. CpGs that are hypomethylated across cancer types are concentrated in megabase-scale domains that occur near the telomeres and centromeres of chromosomes, are depleted of genes, and are enriched for cancer-specific EZH2 binding and H3K27me3 (repressive chromatin). In noncancer samples, there are cell-type specific methylation signatures preserved in primary cell lines and tissues as well as methylation differences induced by cell culture. The relationship between methylation and expression is context-dependent, and we find that CpG-rich enhancers bound by EP300 in the bodies of expressed genes are unmethylated despite the dense gene-body methylation surrounding them. Non-CpG cytosine methylation occurs in human somatic tissue, is particularly prevalent in brain tissue, and is reproducible across many individuals. This study provides an atlas of DNA methylation across diverse and well-characterized samples and enables new discoveries about DNA methylation and its role in gene regulation and disease.
Collapse
|
1374
|
Chemical approaches to study metabolic networks. Pflugers Arch 2013; 465:427-40. [PMID: 23296751 DOI: 10.1007/s00424-012-1201-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/26/2012] [Accepted: 12/04/2012] [Indexed: 12/14/2022]
Abstract
One of the more provocative realizations that have come out of the genome sequencing projects is that organisms possess a large number of uncharacterized or poorly characterized enzymes. This finding belies the commonly held notion that our knowledge of cell metabolism is nearly complete, underscoring the vast landscape of unannotated metabolic and signaling networks that operate under normal physiological conditions, let alone in disease states where metabolic networks may be rewired, dysregulated, or altered to drive disease progression. Consequently, the functional annotation of enzymatic pathways represents a grand challenge for researchers in the post-genomic era. This review will highlight the chemical technologies that have been successfully used to characterize metabolism, and put forth some of the challenges we face as we expand our map of metabolic pathways.
Collapse
|
1375
|
Shinawi T, Hill VK, Krex D, Schackert G, Gentle D, Morris MR, Wei W, Cruickshank G, Maher ER, Latif F. DNA methylation profiles of long- and short-term glioblastoma survivors. Epigenetics 2013; 8:149-56. [PMID: 23291739 DOI: 10.4161/epi.23398] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant type of primary brain tumor in adults and prognosis of most GBM patients is poor. However, a small percentage of patients show a long term survival of 36 mo or longer after diagnosis. Epigenetic profiles can provide molecular markers for patient prognosis: recently, a G-CIMP positive phenotype associated with IDH1 mutations has been described for GBMs with good prognosis. In the present analysis we performed genome-wide DNA methylation profiling of short-term survivors (STS; overall survival < 1 y) and long-term survivors (LTS; overall survival > 3 y) by utilizing the HumanMethylation450K BeadChips to assess quantitative methylation at > 480,000 CpG sites. Cluster analysis has shown that a subset of LTS showed a G-CIMP positive phenotype that was tightly associated with IDH1 mutation status and was confirmed by analysis of the G-CIMP signature genes. Using high stringency criteria for differential hypermethylation between non-cancer brain and tumor samples, we identified 2,638 hypermethylated CpG loci (890 genes) in STS GBMs, 3,101 hypermethylated CpG loci (1,062 genes) in LTS (wild type IDH1) and 11,293 hypermethylated CpG loci in LTS (mutated for IDH1), reflecting the CIMP positive phenotype. The location of differentially hypermethylated CpG loci with respect to CpG content, neighborhood context and functional genomic distribution was similar in our sample set, with the majority of CpG loci residing in CpG islands and in gene promoters. Our preliminary study also identified a set of CpG loci differentially hypermethylated between STS and LTS cases, including members of the homeobox gene family (HOXD8, HOXD13 and HOXC4), the transcription factors NR2F2 and TFAP2A, and Dickkopf 2, a negative regulator of the wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Thoraia Shinawi
- Centre for Rare Diseases and Personalized Medicine, Department of Medical & Molecular Genetics, School of Clinical and Experimental Medicine, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1376
|
Abstract
Heterozygous hotspot mutations in isocitrate dehydrogenases (IDH) IDH1 or IDH2 are frequently observed in specific types of cartilaginous tumors, gliomas, and leukemias. Mutant IDH enzyme loses its normal activity to convert isocitrate into α-ketoglutarate (αKG) and instead acquires the ability to reduce αKG to D-2-hydroxyglutarate. Through direct competition with αKG, accumulation of the oncometabolite D-2-hydroxyglutarate in IDH mutated tumors results in inhibition of αKG-dependent dioxygenases involved in DNA and histone demethylation. Apart from epigenetic alterations, perturbations in the tricarboxylic acid cycle (depletion of intermediates) and activation of the intricately linked hypoxia signaling pathway are apparent in IDH mutated cells. As molecular details are being unraveled, the emerging concept is that IDH mutations result in tumor formation by epigenetic alterations that affect gene expression and result in inhibition of cellular differentiation. Activation of hypoxic stress signaling reprograms cellular energy metabolism and promotes anabolic processes and angiogenesis, thus, providing an advantage to promote neoplastic growth.
Collapse
|
1377
|
Ntziachristos P, Mullenders J, Trimarchi T, Aifantis I. Mechanisms of epigenetic regulation of leukemia onset and progression. Adv Immunol 2013; 117:1-38. [PMID: 23611284 DOI: 10.1016/b978-0-12-410524-9.00001-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the past decade, it has become clear that both genetics and epigenetics play pivotal roles in cancer onset and progression. The importance of epigenetic regulation in proper maintenance of cellular state is highlighted by the frequent mutation of chromatin modulating factors across cancer subtypes. Identification of these mutations has created an interest in designing drugs that target enzymes involved in DNA methylation and posttranslational modification of histones. In this review, we discuss recurrent genetic alterations to epigenetic modulators in both myeloid and lymphoid leukemias. Furthermore, we review how these perturbations contribute to leukemogenesis and impact disease outcome and treatment efficacy. Finally, we discuss how the recent advances in our understanding of chromatin biology may impact treatment of leukemia.
Collapse
|
1378
|
Wang E. Understanding genomic alterations in cancer genomes using an integrative network approach. Cancer Lett 2012; 340:261-9. [PMID: 23266571 DOI: 10.1016/j.canlet.2012.11.050] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/28/2012] [Accepted: 11/28/2012] [Indexed: 12/21/2022]
Abstract
In recent years, cancer genome sequencing and other high-throughput studies of cancer genomes have generated many notable discoveries. In this review, novel genomic alteration mechanisms, such as chromothripsis (chromosomal crisis) and kataegis (mutation storms), and their implications for cancer are discussed. Genomic alterations spur cancer genome evolution. Thus, the relationship between cancer clonal evolution and cancer stems cells is commented. The key question in cancer biology concerns how these genomic alterations support cancer development and metastasis in the context of biological functioning. Thus far, efforts such as pathway analysis have improved the understanding of the functional contributions of genetic mutations and DNA copy number variations to cancer development, progression and metastasis. However, the known pathways correspond to a small fraction, plausibly 5-10%, of somatic mutations and genes with an altered copy number. To develop a comprehensive understanding of the function of these genomic alterations in cancer, an integrative network framework is proposed and discussed. Finally, the challenges and the directions of studying cancer omic data using an integrative network approach are commented.
Collapse
Affiliation(s)
- Edwin Wang
- Lab of Bioinformatics and Systems Biology, National Research Council Canada, Montreal, Canada; McGill University Center for Bioinformatics, Montreal, Canada.
| |
Collapse
|
1379
|
Ward PS, Lu C, Cross JR, Abdel-Wahab O, Levine RL, Schwartz GK, Thompson CB. The potential for isocitrate dehydrogenase mutations to produce 2-hydroxyglutarate depends on allele specificity and subcellular compartmentalization. J Biol Chem 2012; 288:3804-15. [PMID: 23264629 DOI: 10.1074/jbc.m112.435495] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Monoallelic point mutations in cytosolic isocitrate dehydrogenase 1 (IDH1) and its mitochondrial homolog IDH2 can lead to elevated levels of 2-hydroxyglutarate (2HG) in multiple cancers. Here we report that cellular 2HG production from cytosolic IDH1 mutation is dependent on the activity of a retained wild-type IDH1 allele. In contrast, expression of mitochondrial IDH2 mutations led to robust 2HG production in a manner independent of wild-type mitochondrial IDH function. Among the recurrent IDH2 mutations at Arg-172 and Arg-140, IDH2 Arg-172 mutations consistently led to greater 2HG accumulation than IDH2 Arg-140 mutations, and the degree of 2HG accumulation correlated with the ability of these mutations to block cellular differentiation. Cytosolic IDH1 Arg-132 mutations, although structurally analogous to mutations at mitochondrial IDH2 Arg-172, were only able to elevate intracellular 2HG to comparable levels when an equivalent level of wild-type IDH1 was co-expressed. Consistent with 2HG production from cytosolic IDH1 being limited by substrate production from wild-type IDH1, we observed 2HG levels to increase in cancer cells harboring an endogenous monoallelic IDH1 mutation when mitochondrial IDH flux was diverted to the cytosol. Finally, expression of an IDH1 construct engineered to localize to the mitochondria rather than the cytosol resulted in greater 2HG accumulation. These data demonstrate that allelic and subcellular compartment differences can regulate the potential for IDH mutations to produce 2HG in cells. The consequences of 2HG elevation are dose-dependent, and the non-equivalent 2HG accumulation resulting from IDH1 and IDH2 mutations may underlie their differential prognosis and prevalence in various cancers.
Collapse
Affiliation(s)
- Patrick S Ward
- Cancer Biology and Genetics Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | | | | | |
Collapse
|
1380
|
Flanagan S, Lee M, Li CCY, Suter CM, Buckland ME. Promoter Methylation Analysis of IDH Genes in Human Gliomas. Front Oncol 2012; 2:193. [PMID: 23267435 PMCID: PMC3525876 DOI: 10.3389/fonc.2012.00193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/29/2012] [Indexed: 01/22/2023] Open
Abstract
Mutations in isocitrate dehydrogenase (IDH)-1 or -2 are found in the majority of WHO grade II and III astrocytomas and oligodendrogliomas, and secondary glioblastomas. Almost all described mutations are heterozygous missense mutations affecting a conserved arginine residue in the substrate binding site of IDH1 (R132) or IDH2 (R172). But the exact mechanism of IDH mutations in neoplasia is not understood. It has been proposed that IDH mutations impart a “toxic gain-of-function” to the mutant protein, however a dominant-negative effect of mutant IDH has also been described, implying that IDH may function as a tumor suppressor gene. As most, if not all, tumor suppressor genes are inactivated by epigenetic silencing, in a wide variety of tumors, we asked if IDH1 or IDH2 carry the epigenetic signature of a tumor suppressor by assessing cytosine methylation at their promoters. Methylation was quantified in 68 human brain tumors, including both IDH-mutant and IDH wildtype, by bisulfite pyrosequencing. In all tumors examined, CpG methylation levels were less than 8%. Our data demonstrate that inactivation of IDH function through promoter hypermethylation is not common in human gliomas and other brain tumors. These findings do not support a tumor suppressor role for IDH genes in human gliomas.
Collapse
Affiliation(s)
- Simon Flanagan
- Discipline of Pathology, University of Sydney Sydney, NSW, Australia ; Department of Neuropathology, Royal Prince Alfred Hospital Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
1381
|
Goodenberger ML, Jenkins RB. Genetics of adult glioma. Cancer Genet 2012; 205:613-21. [PMID: 23238284 DOI: 10.1016/j.cancergen.2012.10.009] [Citation(s) in RCA: 567] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 10/22/2012] [Accepted: 10/26/2012] [Indexed: 12/13/2022]
Abstract
Gliomas make up approximately 30% of all brain and central nervous system tumors and 80% of all malignant brain tumors. Despite the frequency of gliomas, the etiology of these tumors remains largely unknown. Diffuse gliomas, including astrocytomas and oligodendrogliomas, belong to a single pathologic class but have very different histologies and molecular etiologies. Recent genomic studies have identified separate molecular subtypes within the glioma classification that appear to correlate with biological etiology, prognosis, and response to therapy. The discovery of these subtypes suggests that molecular genetic tests are and will be useful, beyond classical histology, for the clinical classification of gliomas. While a familial susceptibility to glioma has been identified, only a small percentage of gliomas are thought to be due to single-gene hereditary cancer syndromes. Through the use of linkage studies and genome-wide association studies, multiple germline variants have been identified that are beginning to define the genetic susceptibility to glioma.
Collapse
Affiliation(s)
- McKinsey L Goodenberger
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | |
Collapse
|
1382
|
Histone demethylase Jumonji D3 (JMJD3) as a tumor suppressor by regulating p53 protein nuclear stabilization. PLoS One 2012; 7:e51407. [PMID: 23236496 PMCID: PMC3517524 DOI: 10.1371/journal.pone.0051407] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 11/01/2012] [Indexed: 01/20/2023] Open
Abstract
Histone methylation regulates normal stem cell fate decisions through a coordinated interplay between histone methyltransferases and demethylases at lineage specific genes. Malignant transformation is associated with aberrant accumulation of repressive histone modifications, such as polycomb mediated histone 3 lysine 27 (H3K27me3) resulting in a histone methylation mediated block to differentiation. The relevance, however, of histone demethylases in cancer remains less clear. We report that JMJD3, a H3K27me3 demethylase, is induced during differentiation of glioblastoma stem cells (GSCs), where it promotes a differentiation-like phenotype via chromatin dependent (INK4A/ARF locus activation) and chromatin independent (nuclear p53 protein stabilization) mechanisms. Our findings indicate that deregulation of JMJD3 may contribute to gliomagenesis via inhibition of the p53 pathway resulting in a block to terminal differentiation.
Collapse
|
1383
|
Leibetseder A, Ackerl M, Flechl B, Wöhrer A, Widhalm G, Dieckmann K, Kreinecker SS, Pichler J, Hainfellner J, Preusser M, Marosi C. Outcome and molecular characteristics of adolescent and young adult patients with newly diagnosed primary glioblastoma: a study of the Society of Austrian Neurooncology (SANO). Neuro Oncol 2012; 15:112-21. [PMID: 23223340 DOI: 10.1093/neuonc/nos283] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Young age is a favorable prognostic factor for patients with glioblastoma multiforme (GBM). We reviewed the outcomes and molecular tumor characteristics of adolescent and young adult patients with GBM treated in 2 Austrian centers. PATIENTS AND METHODS Data on patients with histologically proven primary GBM diagnosed from 18 through 40 years of age were retrospectively analyzed. All patients were treated with standard first-line therapy. The primary end points were overall survival (OS) and time to progression (TTP). IDH1-R132H mutation status was analyzed using immunohistochemistry, and MGMT promoter methylation was assessed using methylation-specific polymerase chain reaction. RESULTS We included 70 patients (36 men and 34 women) with a median age of 33 years. IDH1-R132H mutations were detected in 22 (39.3%) of 56 cases and MGMT promoter methylation in 33 (61.1%) of 54 cases with available tissue samples. In patients with wild-type IDH, median TTP was 8.2 months and median OS was 24 months, compared with 18 months and 44 months, respectively, observed in patients with mutated IDH. Neither IDH1 nor MGMT status showed a statistically significant association with TTP or OS. Of note, the social and economical situation of the young patients with GBM was alarming, because only 17% succeeded in staying employed after receiving the diagnosis. CONCLUSIONS We found a high frequency of IDH1 mutations and MGMT promoter methylation among young adult patients with primary GBM that may contribute to the generally favorable outcome associated with young age. The social and economic coverage of patients with glioma remains an unsolved socio-ethical problem.
Collapse
Affiliation(s)
- Annette Leibetseder
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1384
|
Abstract
Glioblastoma is the most frequent and malignant brain tumor. The vast majority of glioblastomas (~90%) develop rapidly de novo in elderly patients, without clinical or histologic evidence of a less malignant precursor lesion (primary glioblastomas). Secondary glioblastomas progress from low-grade diffuse astrocytoma or anaplastic astrocytoma. They manifest in younger patients, have a lesser degree of necrosis, are preferentially located in the frontal lobe, and carry a significantly better prognosis. Histologically, primary and secondary glioblastomas are largely indistinguishable, but they differ in their genetic and epigenetic profiles. Decisive genetic signposts of secondary glioblastoma are IDH1 mutations, which are absent in primary glioblastomas and which are associated with a hypermethylation phenotype. IDH1 mutations are the earliest detectable genetic alteration in precursor low-grade diffuse astrocytomas and in oligodendrogliomas, indicating that these tumors are derived from neural precursor cells that differ from those of primary glioblastomas. In this review, we summarize epidemiologic, clinical, histopathologic, genetic, and expression features of primary and secondary glioblastomas and the biologic consequences of IDH1 mutations. We conclude that this genetic alteration is a definitive diagnostic molecular marker of secondary glioblastomas and more reliable and objective than clinical criteria. Despite a similar histologic appearance, primary and secondary glioblastomas are distinct tumor entities that originate from different precursor cells and may require different therapeutic approaches.
Collapse
Affiliation(s)
- Hiroko Ohgaki
- Molecular Pathology Section, International Agency for Research on Cancer, Lyon, France.
| | | |
Collapse
|
1385
|
Jin G, Reitman ZJ, Duncan CG, Spasojevic I, Gooden DM, Rasheed BA, Yang R, Lopez GY, He Y, McLendon RE, Bigner DD, Yan H. Disruption of wild-type IDH1 suppresses D-2-hydroxyglutarate production in IDH1-mutated gliomas. Cancer Res 2012. [PMID: 23204232 DOI: 10.1158/0008-5472.can-12-2852] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Point mutations at Arg132 of the cytoplasmic NADP(+)-dependent isocitrate dehydrogenase 1 (IDH1) occur frequently in gliomas and result in a gain of function to produce the "oncometabolite" D-2-hydroxyglutarate (D-2HG). The mutated IDH1 allele is usually associated with a wild-type IDH1 allele (heterozygous) in cancer. Here, we identify 2 gliomas that underwent loss of the wild-type IDH1 allele but retained the mutant IDH1 allele following tumor progression from World Health Organization (WHO) grade III anaplastic astrocytomas to WHO grade IV glioblastomas. Intratumoral D-2HG was 14-fold lower in the glioblastomas lacking wild-type IDH1 than in glioblastomas with heterozygous IDH1 mutations. To characterize the contribution of wild-type IDH1 to cancer cell D-2HG production, we established an IDH1-mutated astrocytoma (IMA) cell line from a WHO grade III anaplastic astrocytoma. Disruption of the wild-type IDH1 allele in IMA cells by gene targeting resulted in an 87-fold decrease in cellular D-2HG levels, showing that both wild-type and mutant IDH1 alleles are required for D-2HG production in glioma cells. Expression of wild-type IDH1 was also critical for mutant IDH1-associated D-2HG production in the colorectal cancer cell line HCT116. These insights may aid in the development of therapeutic strategies to target IDH1-mutated cancers.
Collapse
Affiliation(s)
- Genglin Jin
- The Preston Robert Tisch Brain Tumor Center, The Pediatric Brain Tumor Foundation Institute, and The Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1386
|
Teschendorff AE, Marabita F, Lechner M, Bartlett T, Tegner J, Gomez-Cabrero D, Beck S. A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. ACTA ACUST UNITED AC 2012; 29:189-96. [PMID: 23175756 PMCID: PMC3546795 DOI: 10.1093/bioinformatics/bts680] [Citation(s) in RCA: 1118] [Impact Index Per Article: 93.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Motivation: The Illumina Infinium 450 k DNA Methylation Beadchip is a prime candidate technology for Epigenome-Wide Association Studies (EWAS). However, a difficulty associated with these beadarrays is that probes come in two different designs, characterized by widely different DNA methylation distributions and dynamic range, which may bias downstream analyses. A key statistical issue is therefore how best to adjust for the two different probe designs. Results: Here we propose a novel model-based intra-array normalization strategy for 450 k data, called BMIQ (Beta MIxture Quantile dilation), to adjust the beta-values of type2 design probes into a statistical distribution characteristic of type1 probes. The strategy involves application of a three-state beta-mixture model to assign probes to methylation states, subsequent transformation of probabilities into quantiles and finally a methylation-dependent dilation transformation to preserve the monotonicity and continuity of the data. We validate our method on cell-line data, fresh frozen and paraffin-embedded tumour tissue samples and demonstrate that BMIQ compares favourably with two competing methods. Specifically, we show that BMIQ improves the robustness of the normalization procedure, reduces the technical variation and bias of type2 probe values and successfully eliminates the type1 enrichment bias caused by the lower dynamic range of type2 probes. BMIQ will be useful as a preprocessing step for any study using the Illumina Infinium 450 k platform. Availability: BMIQ is freely available from http://code.google.com/p/bmiq/. Contact:a.teschendorff@ucl.ac.uk Supplementary information:Supplementary data are available at Bioinformatics online
Collapse
Affiliation(s)
- Andrew E Teschendorff
- Statistical Genomics Group, UCL Cancer Institute, University College London, London WC1E 6BT, UK.
| | | | | | | | | | | | | |
Collapse
|
1387
|
Zhou S, Liu R, Yuan K, Yi T, Zhao X, Huang C, Wei Y. Proteomics analysis of tumor microenvironment: Implications of metabolic and oxidative stresses in tumorigenesis. MASS SPECTROMETRY REVIEWS 2012; 32:267-311. [PMID: 23165949 DOI: 10.1002/mas.21362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 08/22/2012] [Accepted: 08/22/2012] [Indexed: 02/05/2023]
Abstract
Tumorigenesis is always concomitant with microenvironmental alterations. The tumor microenvironment is a heterogeneous and complex milieu, which exerts a variety of stresses on tumor cells for proliferation, survival, or death. Recently, accumulated evidence revealed that metabolic and oxidative stresses both play significant roles in tumor development and progression that converge on a common autophagic pathway. Tumor cells display increased metabolic autonomy, and the hallmark is the exploitation of aerobic glycolysis (termed Warburg effect), which increased glucose consumption and decreased oxidative phosphorylation to support growth and proliferation. This characteristic renders cancer cells more aggressive; they devour tremendous amounts of nutrients from microenvironment to result in an ever-growing appetite for new tumor vessel formation and the release of more "waste," including key determinants of cell fate like lactate and reactive oxygen species (ROS). The intracellular ROS level of cancer cells can also be modulated by a variety of stimuli in the tumor microenvironment, such as pro-growth and pro-inflammatory factors. The intracellular redox state serves as a double-edged sword in tumor development and progression: ROS overproduction results in cytotoxic effects and might lead to apoptotic cell death, whereas certain level of ROS can act as a second-messenger for regulation of such cellular processes as cell survival, proliferation, and metastasis. The molecular mechanisms for cancer cell responses to metabolic and oxidative stresses are complex and are likely to involve multiple molecules or signaling pathways. In addition, the expression and modification of these proteins after metabolic or oxidative stress challenge are diverse in different cancer cells and endow them with different functions. Therefore, MS-based high-throughput platforms, such as proteomics, are indispensable in the global analysis of cancer cell responses to metabolic and oxidative stress. Herein, we highlight recent advances in the understanding of the metabolic and oxidative stresses associated with tumor progression with proteomics-based systems biology approaches.
Collapse
Affiliation(s)
- Shengtao Zhou
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | | | | | | | | | | | | |
Collapse
|
1388
|
Di Stefano AL, Enciso-Mora V, Marie Y, Desestret V, Labussière M, Boisselier B, Mokhtari K, Idbaih A, Hoang-Xuan K, Delattre JY, Houlston RS, Sanson M. Association between glioma susceptibility loci and tumour pathology defines specific molecular etiologies. Neuro Oncol 2012; 15:542-7. [PMID: 23161787 DOI: 10.1093/neuonc/nos284] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Genome-wide association studies have identified single-nucleotide polymorphisms (SNPs) at 7 loci influencing glioma risk: rs2736100 (TERT), rs11979158 and rs2252586 (EGFR), rs4295627 (CCDC26), rs4977756 (CDKN2A/CDKN2B), rs498872 (PHLDB1), and rs6010620 (RTEL1). MATERIALS AND METHODS We studied the relationship among these 7 glioma-risk SNPs and characteristics of tumors from 1374 patients, including grade, IDH (ie IDH1 or IDH2) mutation, EGFR amplification, CDKN2A-p16-INK4a homozygous deletion, 9p and 10q loss, and 1p-19q codeletion. RESULTS rs2736100 (TERT) and rs6010620 (RTEL1) risk alleles were associated with high-grade disease, EGFR amplification, CDKN2A-p16-INK4a homozygous deletion, and 9p and 10q deletion; rs4295627 (CCDC26) and rs498872 (PHLDB1) were associated with low-grade disease, IDH mutation, and 1p-19q codeletion. In contrast, rs4977756 (CDKN2A/B), rs11979158 (EGFR), and to a lesser extent, rs2252586 (EGFR) risk alleles were independent of tumor grade and genetic profile. Adjusting for tumor grade showed a significant association between rs2736100 and IDH status (P = .01), 10q loss (P = .02); rs4295627 and 1p-19q codeletion (P = .04), rs498872 and IDH (P = .02), 9p loss (P = .04), and 10q loss (P = .02). Case-control analyses stratified into 4 molecular classes (defined by 1p-19q status, IDH mutation, and EGFR amplification) showed an association of rs4295627 and rs498872 with IDH-mutated gliomas (P < 10(-3)) and rs2736100 and rs6010620 with IDH wild-type gliomas (P < 10(-3) and P = .03). CONCLUSION The frequency of EGFR and CDKN2A/B risk alleles were largely independent of tumor genetic profile, whereas TERT, RTEL1, CCDC26, and PHLDB1 variants were associated with different genetic profiles that annotate distinct molecular pathways. Our findings provide further insight into the biological basis of glioma etiology.
Collapse
Affiliation(s)
- Anna Luisa Di Stefano
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière (CRICM), UMR-S975, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1389
|
Yun J, Johnson JL, Hanigan CL, Locasale JW. Interactions between epigenetics and metabolism in cancers. Front Oncol 2012; 2:163. [PMID: 23162793 PMCID: PMC3498627 DOI: 10.3389/fonc.2012.00163] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 10/24/2012] [Indexed: 12/31/2022] Open
Abstract
Cancer progression is accompanied by widespread transcriptional changes and metabolic alterations. While it is widely accepted that the origin of cancer can be traced to the mutations that accumulate over time, relatively recent evidence favors a similarly fundamental role for alterations in the epigenome during tumorigenesis. Changes in epigenetics that arise from post-translational modifications of histones and DNA are exploited by cancer cells to upregulate and/or downregulate the expression levels of oncogenes and tumor suppressors, respectively. Although the mechanisms behind these modifications, in particular how they lead to gene silencing and activation, are still being understood, most of the enzymatic machinery of epigenetics require metabolites as substrates or cofactors. As a result, their activities can be influenced by the metabolic state of the cell. The purpose of this review is to give an overview of cancer epigenetics and metabolism and provide examples of where they converge.
Collapse
Affiliation(s)
- Jihye Yun
- Department of Systems Biology, Harvard Medical School Boston, MA, USA
| | | | | | | |
Collapse
|
1390
|
|
1391
|
Benjamin DI, Cravatt BF, Nomura DK. Global profiling strategies for mapping dysregulated metabolic pathways in cancer. Cell Metab 2012; 16:565-77. [PMID: 23063552 PMCID: PMC3539740 DOI: 10.1016/j.cmet.2012.09.013] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/16/2012] [Accepted: 07/31/2012] [Indexed: 12/27/2022]
Abstract
Cancer cells possess fundamentally altered metabolism that provides a foundation to support tumorigenicity and malignancy. Our understanding of the biochemical underpinnings of cancer has benefited from the integrated utilization of large-scale profiling platforms (e.g., genomics, proteomics, and metabolomics), which, together, can provide a global assessment of how enzymes and their parent metabolic networks become altered in cancer to fuel tumor growth. This review presents several examples of how these integrated platforms have yielded fundamental insights into dysregulated metabolism in cancer. We will also discuss questions and challenges that must be addressed to more completely describe, and eventually control, the diverse metabolic pathways that support tumorigenesis.
Collapse
Affiliation(s)
- Daniel I Benjamin
- Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 127 Morgan Hall, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
1392
|
G-cimp status prediction of glioblastoma samples using mRNA expression data. PLoS One 2012; 7:e47839. [PMID: 23139755 PMCID: PMC3490960 DOI: 10.1371/journal.pone.0047839] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/21/2012] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is a tumor with high mortality and no known cure. The dramatic molecular and clinical heterogeneity seen in this tumor has led to attempts to define genetically similar subgroups of GBM with the hope of developing tumor specific therapies targeted to the unique biology within each of these subgroups. Recently, a subset of relatively favorable prognosis GBMs has been identified. These glioma CpG island methylator phenotype, or G-CIMP tumors, have distinct genomic copy number aberrations, DNA methylation patterns, and (mRNA) expression profiles compared to other GBMs. While the standard method for identifying G-CIMP tumors is based on genome-wide DNA methylation data, such data is often not available compared to the more widely available gene expression data. In this study, we have developed and evaluated a method to predict the G-CIMP status of GBM samples based solely on gene expression data.
Collapse
|
1393
|
Mullen AR, DeBerardinis RJ. Genetically-defined metabolic reprogramming in cancer. Trends Endocrinol Metab 2012; 23:552-9. [PMID: 22858391 PMCID: PMC3466334 DOI: 10.1016/j.tem.2012.06.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 06/22/2012] [Accepted: 06/27/2012] [Indexed: 12/21/2022]
Abstract
Oncogenes and tumor suppressors regulate cell metabolism. Evidence demonstrates that tumorigenic mutations in these genes tend to orchestrate metabolic activity into a platform that promotes cell survival, growth, and proliferation. Recent work has shown that some metabolic enzymes are also mutated in cancer, and that these mutations may influence malignancy directly. Thus, these enzymes seem to function as oncogenes and tumor suppressors, and would appear to be compelling targets for therapeutic intervention. Here, we review several enzymes mutated in cancer - phosphoglycerate dehydrogenase, isocitrate dehydrogenases 1 and 2, succinate dehydrogenase, and fumarate hydratase - and discuss exciting new work that has begun to pull back the curtain on how mutations in these enzymes influence tumorigenesis.
Collapse
Affiliation(s)
- Andrew R Mullen
- Children's Research Institute, Department of Pediatrics and McDermott Center for Human Growth and Development, University of Texas - Southwestern Medical Center, Dallas, TX 75390-8502, USA
| | | |
Collapse
|
1394
|
Seidel C, Florean C, Schnekenburger M, Dicato M, Diederich M. Chromatin-modifying agents in anti-cancer therapy. Biochimie 2012; 94:2264-79. [DOI: 10.1016/j.biochi.2012.05.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 05/14/2012] [Indexed: 01/12/2023]
|
1395
|
Theeler BJ, Yung WKA, Fuller GN, De Groot JF. Moving toward molecular classification of diffuse gliomas in adults. Neurology 2012; 79:1917-26. [PMID: 23109653 PMCID: PMC3525311 DOI: 10.1212/wnl.0b013e318271f7cb] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 06/26/2012] [Indexed: 12/20/2022] Open
Abstract
Diffuse gliomas are a heterogenous group of neoplasms traditionally classified as grades II to IV based on histologic features, and with prognosis determined mainly by histologic grade and pretreatment clinical factors. Our understanding of the molecular basis of glioma initiation, tumor progression, and treatment failure is rapidly evolving. A molecular profile of diffuse gliomas is emerging. Studies evaluating gene expression and DNA methylation profile have found multiple glioma subtypes and an association between subtype and survival. The recent discovery of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) mutations in glioma has provided reproducible prognostic biomarkers and novel therapeutic targets. Glioblastomas that exhibit CpG island hypermethylator phenotype, proneural gene expression, or IDH1 mutation identify a subset of patients with markedly improved prognosis. Accumulated evidence supports the stratification of both low-grade and anaplastic diffuse gliomas into prognostic groups using 1p/19q codeletion and IDH mutation status. A classification scheme incorporating clinical, pathologic, and molecular information may facilitate improved prognostication for patients treated in the clinic, the development of more effective clinical trials, and rational testing of targeted therapeutics.
Collapse
Affiliation(s)
- Brett J Theeler
- Department of Pathology, Division of Pathology and Laboratory Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | |
Collapse
|
1396
|
Abstract
In this issue of Cancer Cell, Sturm et al. report that global DNA methylation patterns in glioblastoma multiforme divide adult and pediatric tumors into subgroups that have characteristic DNA mutations, mRNA profiles, and most importantly, different clinical behaviors. These findings suggest novel opportunities for therapeutics for this dreaded disease.
Collapse
Affiliation(s)
- Eric H Raabe
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | |
Collapse
|
1397
|
Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DTW, 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 glioblastoma. Cancer Cell 2012; 22:425-37. [PMID: 23079654 DOI: 10.1016/j.ccr.2012.08.024] [Citation(s) in RCA: 1347] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 06/03/2012] [Accepted: 08/24/2012] [Indexed: 12/30/2022]
Abstract
Glioblastoma (GBM) is a brain tumor that carries a dismal prognosis and displays considerable heterogeneity. We have recently identified recurrent H3F3A mutations affecting two critical amino acids (K27 and G34) of histone H3.3 in one-third of pediatric GBM. Here, we show that each H3F3A mutation defines an epigenetic subgroup of GBM with a distinct global methylation pattern, and that they are mutually exclusive with IDH1 mutations, which characterize a third mutation-defined subgroup. Three further epigenetic subgroups were enriched for hallmark genetic events of adult GBM and/or established transcriptomic signatures. We also demonstrate that the two H3F3A mutations give rise to GBMs in separate anatomic compartments, with differential regulation of transcription factors OLIG1, OLIG2, and FOXG1, possibly reflecting different cellular origins.
Collapse
Affiliation(s)
- Dominik Sturm
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1398
|
Abstract
The understanding that oncogenes can have profound effects on cellular metabolism and the discovery of mutations and alterations in several metabolism-related enzymes--isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), succinate dehydrogenase (SDH), fumarate hydratase (FH), and pyruvate kinase M2 (PKM2)--has renewed interest in cancer metabolism and renewed hope of taking therapeutic advantage of cancer metabolism. Otto Warburg observed that aerobic glycolysis was a characteristic of cancer cells. More than 50 years later, we understand that aerobic glycolysis and uptake of glutamine and glycine allow cancer cells to produce energy (ATP) and the nucleotides, amino acids, and lipids required for proliferation. Expression of the MYC oncogene drives the increase in cellular biomass facilitating proliferation. PKM2 expression in cancer cells stimulates aerobic glycolysis. Among intermediary metabolism enzymes, mutations in SDH occur in gastointestinal stromal tumors and result in a pseudohypoxic metabolic milieu. FH mutations lead to a characteristic renal cell carcinoma. Isocitrate dehydrogenase (IDH1/2) mutations have been found in leukemias, gliomas, prostate cancer, colon cancer, thyroid cancer, and sarcomas. These recently recognized oncogenic metabolic lesions may be selective targets for new anticancer therapeutics.
Collapse
Affiliation(s)
- Beverly A Teicher
- Molecular Pharmacology Branch, Developmental Therapeutics Program, National Cancer Institute, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
1399
|
Yang H, Ye D, Guan KL, Xiong Y. IDH1 and IDH2 mutations in tumorigenesis: mechanistic insights and clinical perspectives. Clin Cancer Res 2012; 18:5562-71. [PMID: 23071358 PMCID: PMC3897211 DOI: 10.1158/1078-0432.ccr-12-1773] [Citation(s) in RCA: 317] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genes encoding for isocitrate dehydrogenases 1 and 2, IDH1 and IDH2, are frequently mutated in multiple types of human cancer. Mutations targeting IDH1 and IDH2 result in simultaneous loss of their normal catalytic activity, the production of α-ketoglutarate (α-KG), and gain of a new function, the production of 2-hydroxyglutarate (2-HG). 2-HG is structurally similar to α-KG, and acts as an α-KG antagonist to competitively inhibit multiple α-KG-dependent dioxygenases, including both lysine histone demethylases and the ten-eleven translocation family of DNA hydroxylases. Abnormal histone and DNA methylation are emerging as a common feature of tumors with IDH1 and IDH2 mutations and may cause altered stem cell differentiation and eventual tumorigenesis. Therapeutically, unique features of IDH1 and IDH2 mutations make them good biomarkers and potential drug targets.
Collapse
Affiliation(s)
- Hui Yang
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences and School of Life Sciences, Fudan University, Shanghai, P R China
| | | | | | | |
Collapse
|
1400
|
Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell 2012; 48:612-26. [PMID: 23063526 DOI: 10.1016/j.molcel.2012.08.033] [Citation(s) in RCA: 610] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 06/15/2012] [Accepted: 08/27/2012] [Indexed: 12/13/2022]
Abstract
Widespread changes in gene expression drive tumorigenesis, yet our knowledge of how aberrant epigenomic and transcriptome profiles arise in cancer cells is poorly understood. Here, we demonstrate that metabolic transformation plays an important role. Butyrate is the primary energy source of normal colonocytes and is metabolized to acetyl-CoA, which was shown to be important not only for energetics but also for HAT activity. Due to the Warburg effect, cancerous colonocytes rely on glucose as their primary energy source, so butyrate accumulated and functioned as an HDAC inhibitor. Although both mechanisms increased histone acetylation, different target genes were upregulated. Consequently, butyrate stimulated the proliferation of normal colonocytes and cancerous colonocytes when the Warburg effect was prevented from occurring, whereas it inhibited the proliferation of cancerous colonocytes undergoing the Warburg effect. These findings link a common metabolite to epigenetic mechanisms that are differentially utilized by normal and cancerous cells because of their inherent metabolic differences.
Collapse
Affiliation(s)
- Dallas R Donohoe
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|