1501
|
Shibahara I, Sonoda Y, Kanamori M, Saito R, Kumabe T, Tominaga T. New insights into glioma classification based on isocitrate dehydrogenase 1 and 2 gene status. Brain Tumor Pathol 2011; 28:203-8. [PMID: 21735252 DOI: 10.1007/s10014-011-0050-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 06/14/2011] [Indexed: 11/24/2022]
Abstract
In glioma, mutations in the isocitrate dehydrogenase 1 and 2 (IDH1/2) genes have been receiving attention. IDH1/2 mutations are frequently found in grade II and III gliomas. These genetic alterations occur very early in gliomagenesis and strongly predict favorable outcome in patients with high-grade gliomas. Despite the evolution of studies on this topic, the underlying mechanism of the IDH1/2 mutations remains unknown. Here, we briefly review the current knowledge of IDH1/2 and discuss molecular diagnostics based on IDH1/2 gene status.
Collapse
Affiliation(s)
- Ichiyo Shibahara
- Department of Neurosurgery, Tohoku University School of Medicine, 1-1 Seiryo-machi Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | |
Collapse
|
1502
|
Estécio MRH, Issa JPJ. Dissecting DNA hypermethylation in cancer. FEBS Lett 2011; 585:2078-86. [PMID: 21146531 PMCID: PMC3378045 DOI: 10.1016/j.febslet.2010.12.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 12/02/2010] [Accepted: 12/03/2010] [Indexed: 12/31/2022]
Abstract
There is compelling evidence to support the importance of DNA methylation alterations in cancer development. Both losses and gains of DNA methylation are observed, thought to contribute pathophysiologically by inactivating tumor suppressor genes, inducing chromosomal instability and ectopically activating gene expression. Lesser known are the causes of aberrant DNA methylation. Recent studies have pointed out that intrinsic gene susceptibility to DNA methylation, environmental factors and gene function all have an intertwined participation in this process. Overall, these data support a deterministic rather than a stochastic mechanism for de novo DNA methylation in cancer. In this review article, we discuss the technologies available to study DNA methylation and the endogenous and exogenous factors that influence the onset of de novo methylation in cancer.
Collapse
Affiliation(s)
- Marcos R H Estécio
- Department of Leukemia, UT MD Anderson Cancer Center, Houston, TX 77030, United States.
| | | |
Collapse
|
1503
|
Falini B, Gionfriddo I, Cecchetti F, Ballanti S, Pettirossi V, Martelli MP. Acute myeloid leukemia with mutated nucleophosmin (NPM1): any hope for a targeted therapy? Blood Rev 2011; 25:247-54. [PMID: 21724308 DOI: 10.1016/j.blre.2011.06.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute myeloid leukemia (AML) carrying nucleophosmin (NPM1) mutations displays distinct molecular and clinical-pathological features that led to its inclusion as provisional entity in 2008 WHO classification of myeloid neoplasms. Since NPM1 mutations behave as a founder genetic lesion in AML, they could be an attractive target for therapeutic intervention. Here, we discuss the potential for developing targeted therapies for NPM1-mutated AML with focus on: (i) interfering with the abnormal traffic of the NPM1 leukemic mutant, i.e., its cytoplasmic dislocation; (ii) disrupting the nucleolar structure/function by interfering with residual wild-type nucleophosmin and other nucleolar components acting as hub proteins; and (iii) evaluating the activity of epigenetic drugs (e.g., 5-azacytidine) or agents acting on differentiation and apoptosis. As quantitative assessment of NPM1 mutated transcript copies now provides the means to measure minimal residual disease, we also discuss the potential for intervening in NPM1-mutated AML before overt hematological relapse occurs (so-called pre-emptive therapy).
Collapse
Affiliation(s)
- Brunangelo Falini
- Institute of Hematology, Strada Sant'Andrea delle Fratte, University of Perugia, 06122 Perugia, Italy.
| | | | | | | | | | | |
Collapse
|
1504
|
Nannini M, Biasco G, Maleddu A, Pantaleo MA. New molecular targets beyond KIT and PDGFRA in gastrointestinal stromal tumors: present and future. Expert Opin Ther Targets 2011; 15:803-15. [DOI: 10.1517/14728222.2011.566215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
1505
|
Abstract
The effects of genomic medicine on child health promise to be profound. Medical applications will eventually include characterizing patients' genomes to detect predictive mutations for pre-symptomatic counseling where treatment exists; to search for causes of diseases of unknown etiology, and to detect carriers for prenatal counseling; to define cancer and other disease-based genomes to design individualized therapy; and to understand our microbiomes to modify these in health and disease. Rapid advances in technology and bioinformatics have reduced the cost and the time and increased the accuracy necessary to sequence whole genomes or whole exomes. However, complete understanding of disease will also require correlation of genomic information with high-quality phenotypic data. In addition, several critical ethical, psycho-social, and public policy issues will require clarity in the coming years. Ultimately these advances will improve the effectiveness of health care for children and for society.
Collapse
|
1506
|
Fu Y, Huang R, Zheng Y, Zhang Z, Liang A. Glioma-derived mutations in isocitrate dehydrogenase 2 beneficial to traditional chemotherapy. Biochem Biophys Res Commun 2011; 410:218-23. [DOI: 10.1016/j.bbrc.2011.05.108] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 05/18/2011] [Indexed: 10/18/2022]
|
1507
|
|
1508
|
Deng X. SeqGene: a comprehensive software solution for mining exome- and transcriptome- sequencing data. BMC Bioinformatics 2011; 12:267. [PMID: 21714929 PMCID: PMC3148209 DOI: 10.1186/1471-2105-12-267] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 06/29/2011] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The popularity of massively parallel exome and transcriptome sequencing projects demands new data mining tools with a comprehensive set of features to support a wide range of analysis tasks. RESULTS SeqGene, a new data mining tool, supports mutation detection and annotation, dbSNP and 1000 Genome data integration, RNA-Seq expression quantification, mutation and coverage visualization, allele specific expression (ASE), differentially expressed genes (DEGs) identification, copy number variation (CNV) analysis, and gene expression quantitative trait loci (eQTLs) detection. We also developed novel methods for testing the association between SNP and expression and identifying genotype-controlled DEGs. We showed that the results generated from SeqGene compares favourably to other existing methods in our case studies. CONCLUSION SeqGene is designed as a general-purpose software package. It supports both paired-end reads and single reads generated on most sequencing platforms; it runs on all major types of computers; it supports arbitrary genome assemblies for arbitrary organisms; and it scales well to support both large and small scale sequencing projects. The software homepage is http://seqgene.sourceforge.net.
Collapse
Affiliation(s)
- Xutao Deng
- Bioinformatics Core Facility, Department of Molecular Medicine, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
1509
|
Analysis of cancer metabolism by imaging hyperpolarized nuclei: prospects for translation to clinical research. Neoplasia 2011; 13:81-97. [PMID: 21403835 DOI: 10.1593/neo.101102] [Citation(s) in RCA: 568] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/18/2010] [Accepted: 10/22/2010] [Indexed: 12/13/2022] Open
Abstract
A major challenge in cancer biology is to monitor and understand cancer metabolism in vivo with the goal of improved diagnosis and perhaps therapy. Because of the complexity of biochemical pathways, tracer methods are required for detecting specific enzyme-catalyzed reactions. Stable isotopes such as (13)C or (15)N with detection by nuclear magnetic resonance provide the necessary information about tissue biochemistry, but the crucial metabolites are present in low concentration and therefore are beyond the detection threshold of traditional magnetic resonance methods. A solution is to improve sensitivity by a factor of 10,000 or more by temporarily redistributing the populations of nuclear spins in a magnetic field, a process termed hyperpolarization. Although this effect is short-lived, hyperpolarized molecules can be generated in an aqueous solution and infused in vivo where metabolism generates products that can be imaged. This discovery lifts the primary constraint on magnetic resonance imaging for monitoring metabolism-poor sensitivity-while preserving the advantage of biochemical information. The purpose of this report was to briefly summarize the known abnormalities in cancer metabolism, the value and limitations of current imaging methods for metabolism, and the principles of hyperpolarization. Recent preclinical applications are described. Hyperpolarization technology is still in its infancy, and current polarizer equipment and methods are suboptimal. Nevertheless, there are no fundamental barriers to rapid translation of this exciting technology to clinical research and perhaps clinical care.
Collapse
|
1510
|
Abstract
CONTEXT Gliomas are the most common primary brain tumors of adults and include a variety of histologic types and morphologies. Histologic evaluation remains the gold standard for glioma diagnosis; however, diagnostic difficulty may arise from tumor heterogeneity, overlapping morphologic features, and tumor sampling. Recently, our knowledge about the genetics of these tumors has expanded, and new molecular markers have been developed. Some of these markers have shown diagnostic value, whereas others are useful prognosticators for patient survival and therapeutic response. OBJECTIVE To review the most clinically useful molecular markers and their detection techniques in gliomas. DATA SOURCES Review of the pertinent literature and personal experience with the molecular testing in gliomas. CONCLUSIONS This article provides an overview of the most common molecular markers in neurooncology, including 1p/19q codeletion in oligodendroglial tumors, mutations in the isocitrate dehydrogenase 1 and 2 genes in diffuse gliomas, hypermethylation of the O(6)-methylguanine-DNA methyltransferase gene promoter in glioblastomas and anaplastic gliomas, alterations in the epidermal growth factor receptor and phosphatase and tensin homolog genes in high-grade gliomas, as well as BRAF alterations in pilocytic astrocytomas. Molecular testing of gliomas is increasingly used in routine clinical practice and requires that neuropathologists be familiar with these genetic markers and the molecular diagnostic techniques for their detection.
Collapse
Affiliation(s)
- Marina N Nikiforova
- Department of Pathology, University of Pittsburgh, 200 Lothrop Ave, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
1511
|
Mutations in epigenetic modifiers in myeloid malignancies and the prospect of novel epigenetic-targeted therapy. Adv Hematol 2011; 2012:469592. [PMID: 21811504 PMCID: PMC3145345 DOI: 10.1155/2012/469592] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 06/02/2011] [Indexed: 11/17/2022] Open
Abstract
In the recent years, the discovery of a series of mutations in patients with myeloid malignancies has provided insight into the pathogenesis of myelodysplastic syndromes (MDSs), myeloproliferative neoplasms (MPNs), and acute myeloid leukemia (AML). Among these alterations have been mutations in genes, such as IDH1/2, TET2, DNMT3A, and EZH2, which appear to affect DNA and/or histone lysine methylation. Large clinical correlative studies are beginning to decipher the clinical importance, prevalence, and potential prognostic significance of these mutations. Additionally, burgeoning insight into the role of epigenetics in the pathogenesis of myeloid malignancies has prompted increased interest in development of novel therapies which target DNA and histone posttranslational modifications. DNA demethylating agents have been demonstrated to be clinically active in a subset of patients with MDS and AML and are used extensively. However, newer, more specific agents which alter DNA and histone modification are under preclinical study and development and are likely to expand our therapeutic options for these diseases in the near future. Here, we review the current understanding of the clinical importance of these newly discovered mutations in AML and MDS patients. We also discuss exciting developments in DNA methyltransferase inhibitor strategies and the prospect of novel histone lysine methyltransferase inhibitors.
Collapse
|
1512
|
Abstract
This mini-review describes the rapid changes in genome technologies that are leading to comprehensive views of genetic alterations in cancer, and presents high-level thoughts on ways to accelerate translation into clinical medicine. Issues that are more relevant to children, adolescents, and young adult patients with cancer are highlighted.
Collapse
Affiliation(s)
- Thomas J Hudson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| |
Collapse
|
1513
|
Juliusson G, Tadmor T, Polliack A. 'Hairy' cells: where are the roots of this leukemia? Leuk Lymphoma 2011; 52:2205-6. [PMID: 21689026 DOI: 10.3109/10428194.2011.593277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Gunnar Juliusson
- Department of Hematology, Skåne University Hospital, Lund University, Lund, Sweden.
| | | | | |
Collapse
|
1514
|
Pusch S, Sahm F, Meyer J, Mittelbronn M, Hartmann C, von Deimling A. Scientific correspondence. Neuropathol Appl Neurobiol 2011; 37:428-30. [DOI: 10.1111/j.1365-2990.2010.01127.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
1515
|
Acquisition of Genetic Aberrations by Activation-Induced Cytidine Deaminase (AID) during Inflammation-Associated Carcinogenesis. Cancers (Basel) 2011; 3:2750-66. [PMID: 24212831 PMCID: PMC3757441 DOI: 10.3390/cancers3022750] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 06/09/2011] [Accepted: 06/14/2011] [Indexed: 02/06/2023] Open
Abstract
Genetic abnormalities such as nucleotide alterations and chromosomal disorders that accumulate in various tumor-related genes have an important role in cancer development. The precise mechanism of the acquisition of genetic aberrations, however, remains unclear. Activation-induced cytidine deaminase (AID), a nucleotide editing enzyme, is essential for the diversification of antibody production. AID is expressed only in activated B lymphocytes under physiologic conditions and induces somatic hypermutation and class switch recombination in immunoglobulin genes. Inflammation leads to aberrant AID expression in various gastrointestinal organs and increased AID expression contributes to cancer development by inducing genetic alterations in epithelial cells. Studies of how AID induces genetic disorders are expected to elucidate the mechanism of inflammation-associated carcinogenesis.
Collapse
|
1516
|
Kim YH, Pierscianek D, Mittelbronn M, Vital A, Mariani L, Hasselblatt M, Ohgaki H. TET2 promoter methylation in low-grade diffuse gliomas lacking IDH1/2 mutations: Figure 1. J Clin Pathol 2011; 64:850-2. [DOI: 10.1136/jclinpath-2011-200133] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BackgroundMiscoding mutations of the TET2 gene, which encodes the α-ketoglutarate-dependent enzyme that catalyses the conversion of 5-methylcytosine to 5-hydroxymethylcytosine, thus producing DNA demethylation, have been detected in 10–25% of acute myeloid leukaemias lacking IDH1/2 mutations. Most low-grade diffuse gliomas carry IDH1/2 mutations (>85%), but molecular mechanisms of pathogenesis in those lacking IDH1/2 mutations remain to be elucidated.MethodsMiscoding mutations and promoter methylation of the TET2 gene were screened for in 29 low-grade diffuse gliomas lacking IDH1/2 mutations.ResultsSingle-strand conformational polymorphism followed by direct sequencing showed the absence of miscoding mutations in TET2. Methylation-specific PCR revealed methylation of the TET2 promoter in 5 of 35 cases (14%). In contrast, none of 38 low-grade diffuse gliomas with IDH1/2 mutations had TET2 promoter methylation (p=0.0216).ConclusionResults suggest that TET2 promoter methylation, but not TET2 mutation, may be an alternative mechanism of pathogenesis in a small fraction of low-grade diffuse gliomas lacking IDH1/2 mutations.
Collapse
|
1517
|
Noris P, Perrotta S, Seri M, Pecci A, Gnan C, Loffredo G, Pujol-Moix N, Zecca M, Scognamiglio F, De Rocco D, Punzo F, Melazzini F, Scianguetta S, Casale M, Marconi C, Pippucci T, Amendola G, Notarangelo LD, Klersy C, Civaschi E, Balduini CL, Savoia A. Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families. Blood 2011; 117:6673-80. [PMID: 21467542 DOI: 10.1182/blood-2011-02-336537] [Citation(s) in RCA: 200] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Until recently, thrombocytopenia 2 (THC2) was considered an exceedingly rare form of autosomal dominant thrombocytopenia and only 2 families were known. However, we recently identified mutations in the 5'-untranslated region of the ANKRD26 gene in 9 THC2 families. Here we report on 12 additional pedigrees with ANKRD26 mutations, 6 of which are new. Because THC2 affected 21 of the 210 families in our database, it has to be considered one of the less rare forms of inherited thrombocytopenia. Analysis of all 21 families with ANKRD26 mutations identified to date revealed that thrombocytopenia and bleeding tendency were usually mild. Nearly all patients had no platelet macrocytosis, and this characteristic distinguishes THC2 from most other forms of inherited thrombocytopenia. In the majority of cases, platelets were deficient in glycoprotein Ia and α-granules, whereas in vitro platelet aggregation was normal. Bone marrow examination and serum thrombopoietin levels suggested that thrombocytopenia was derived from dysmegakaryopoiesis. Unexplained high values of hemoglobin and leukocytes were observed in a few cases. An unexpected finding that warrants further investigation was a high incidence of acute leukemia. Given the scarcity of distinctive characteristics, the ANKRD26-related thrombocytopenia has to be taken into consideration in the differential diagnosis of isolated thrombocytopenias.
Collapse
Affiliation(s)
- Patrizia Noris
- Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo Foundation, University of Pavia, Piazzale Golgi, Pavia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1518
|
Bralten LBC, Kloosterhof NK, Balvers R, Sacchetti A, Lapre L, Lamfers M, Leenstra S, de Jonge H, Kros JM, Jansen EEW, Struys EA, Jakobs C, Salomons GS, Diks SH, Peppelenbosch M, Kremer A, Hoogenraad CC, Smitt PAES, French PJ. IDH1 R132H decreases proliferation of glioma cell lines in vitro and in vivo. Ann Neurol 2011; 69:455-63. [PMID: 21446021 DOI: 10.1002/ana.22390] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE A high percentage of grade II and III gliomas have mutations in the gene encoding isocitrate dehydrogenase (IDH1). This mutation is always a heterozygous point mutation that affects the amino acid arginine at position 132 and results in loss of its native enzymatic activity and gain of alternative enzymatic activity (producing D-2-hydroxyglutarate). The objective of this study was to investigate the cellular effects of R132H mutations in IDH1. METHODS Functional consequences of IDH1(R132H) mutations were examined among others using fluorescence-activated cell sorting, kinome and expression arrays, biochemical assays, and intracranial injections on 3 different (glioma) cell lines with stable overexpression of IDH1(R132H) . RESULTS IDH1(R132H) overexpression in established glioma cell lines in vitro resulted in a marked decrease in proliferation, decreased Akt phosphorylation, altered morphology, and a more contact-dependent cell migration. The reduced proliferation is related to accumulation of D-2-hydroxyglutarate that is produced by IDH1(R132H) . Mice injected with IDH1(R132H) U87 cells have prolonged survival compared to mice injected with IDH1(wt) or green fluorescent protein-expressing U87 cells. INTERPRETATION Our results demonstrate that IDH1(R132H) dominantly reduces aggressiveness of established glioma cell lines in vitro and in vivo. In addition, the IDH1(R132H) -IDH1(wt) heterodimer has higher enzymatic activity than the IDH1(R132H) -IDH1(R132H) homodimer. Our observations in model systems of glioma might lead to a better understanding of the biology of IDH1 mutant gliomas, which are typically low grade and often slow growing.
Collapse
Affiliation(s)
- Linda B C Bralten
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1519
|
Fabbri G, Rasi S, Rossi D, Trifonov V, Khiabanian H, Ma J, Grunn A, Fangazio M, Capello D, Monti S, Cresta S, Gargiulo E, Forconi F, Guarini A, Arcaini L, Paulli M, Laurenti L, Larocca LM, Marasca R, Gattei V, Oscier D, Bertoni F, Mullighan CG, Foá R, Pasqualucci L, Rabadan R, Dalla-Favera R, Gaidano G. Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation. ACTA ACUST UNITED AC 2011; 208:1389-401. [PMID: 21670202 PMCID: PMC3135373 DOI: 10.1084/jem.20110921] [Citation(s) in RCA: 491] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Next generation sequencing and copy number analysis provide insights into the complexity of the CLL coding genome, and reveal an association between NOTCH1 mutational activation and poor prognosis. The pathogenesis of chronic lymphocytic leukemia (CLL), the most common leukemia in adults, is still largely unknown. The full spectrum of genetic lesions that are present in the CLL genome, and therefore the number and identity of dysregulated cellular pathways, have not been identified. By combining next-generation sequencing and copy number analysis, we show here that the typical CLL coding genome contains <20 clonally represented gene alterations/case, including predominantly nonsilent mutations, and fewer copy number aberrations. These analyses led to the discovery of several genes not previously known to be altered in CLL. Although most of these genes were affected at low frequency in an expanded CLL screening cohort, mutational activation of NOTCH1, observed in 8.3% of CLL at diagnosis, was detected at significantly higher frequency during disease progression toward Richter transformation (31.0%), as well as in chemorefractory CLL (20.8%). Consistent with the association of NOTCH1 mutations with clinically aggressive forms of the disease, NOTCH1 activation at CLL diagnosis emerged as an independent predictor of poor survival. These results provide initial data on the complexity of the CLL coding genome and identify a dysregulated pathway of diagnostic and therapeutic relevance.
Collapse
Affiliation(s)
- Giulia Fabbri
- Institute for Cancer Genetics and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1520
|
Prevalence and prognostic value of IDH1 and IDH2 mutations in childhood AML: a study of the AML–BFM and DCOG study groups. Leukemia 2011; 25:1704-10. [DOI: 10.1038/leu.2011.142] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
1521
|
Ddx18 is essential for cell-cycle progression in zebrafish hematopoietic cells and is mutated in human AML. Blood 2011; 118:903-15. [PMID: 21653321 DOI: 10.1182/blood-2010-11-318022] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In a zebrafish mutagenesis screen to identify genes essential for myelopoiesis, we identified an insertional allele hi1727, which disrupts the gene encoding RNA helicase dead-box 18 (Ddx18). Homozygous Ddx18 mutant embryos exhibit a profound loss of myeloid and erythroid cells along with cardiovascular abnormalities and reduced size. These mutants also display prominent apoptosis and a G1 cell-cycle arrest. Loss of p53, but not Bcl-xl overexpression, rescues myeloid cells to normal levels, suggesting that the hematopoietic defect is because of p53-dependent G1 cell-cycle arrest. We then sequenced primary samples from 262 patients with myeloid malignancies because genes essential for myelopoiesis are often mutated in human leukemias. We identified 4 nonsynonymous sequence variants (NSVs) of DDX18 in acute myeloid leukemia (AML) patient samples. RNA encoding wild-type DDX18 and 3 NSVs rescued the hematopoietic defect, indicating normal DDX18 activity. RNA encoding one mutation, DDX18-E76del, was unable to rescue hematopoiesis, and resulted in reduced myeloid cell numbers in ddx18(hi1727/+) embryos, indicating this NSV likely functions as a dominant-negative allele. These studies demonstrate the use of the zebrafish as a robust in vivo system for assessing the function of genes mutated in AML, which will become increasingly important as more sequence variants are identified by next-generation resequencing technologies.
Collapse
|
1522
|
Andersson AK, Miller DW, Lynch JA, Lemoff AS, Cai Z, Pounds SB, Radtke I, Yan B, Schuetz JD, Rubnitz JE, Ribeiro RC, Raimondi SC, Zhang J, Mullighan CG, Shurtleff SA, Schulman BA, Downing JR. IDH1 and IDH2 mutations in pediatric acute leukemia. Leukemia 2011; 25:1570-7. [PMID: 21647154 DOI: 10.1038/leu.2011.133] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To investigate the frequency of isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) mutations in pediatric acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL), we sequenced these genes in diagnostic samples from 515 patients (227 AMLs and 288 ALLs). Somatic IDH1/IDH2 mutations were rare in ALL (N=1), but were more common in AML, occurring in 3.5% (IDH1 N=3 and IDH2 N=5), with the frequency higher in AMLs with a normal karyotype (9.8%). The identified IDH1 mutations occurred in codon 132 resulting in replacement of arginine with either cysteine (N=3) or histidine (N=1). By contrast, mutations in IDH2 did not affect the homologous residue but instead altered codon 140, resulting in replacement of arginine with either glutamine (N=4) or tryptophan (N=1). Structural modeling of IDH2 suggested that codon 140 mutations disrupt the enzyme's ability to bind its substrate isocitrate. Accordingly, recombinant IDH2 R140Q/W were unable to carry out the decarboxylation of isocitrate to α-ketoglutarate (α-KG), but instead gained the neomorphic activity to reduce α-KG to R(-)-2-hydroxyglutarete (2-HG). Analysis of primary leukemic blasts confirmed high levels of 2-HG in AMLs with IDH1/IDH2 mutations. Interestingly, 3/5 AMLs with IDH2 mutations had FLT3-activating mutations, raising the possibility that these mutations cooperate in leukemogenesis.
Collapse
Affiliation(s)
- A K Andersson
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1523
|
Abstract
PURPOSE OF REVIEW Since the discovery of the JAK2V617F mutation in 2005, an increasing number of somatic and germline genetic events responsible for myeloproliferative neoplasm (MPN) pathogenesis have been uncovered. The purpose of this review is to outline the most recent discoveries of the genetic alterations found in patients with MPNs. RECENT FINDINGS In addition to the JAK2V617F mutation, additional mutations in the JAK–STAT pathway have been discovered including a series of mutations in exon 12 of JAK2, the thrombopoietic receptor gene MPL, and in the gene encoding the JAK–STAT inhibitory adaptor protein LNK. Additionally, mutations in genes which appear to affect the epigenome of MPN patients have been discovered including mutations in TET2, IDH1/ 2, EZH2, and ASXL1. Lastly, some insights into the genetic events which contribute to transformation of a chronic MPN phenotype to acute myeloid leukemia have been elucidated, including deletion of the transcription factor Ikaros. SUMMARY The spectrum of genetic abnormalities found in the classic MPNs has increased over the last 6 years and somatic mutations in JAK2, MPL, LNK, TET2, EZH2, ASXL1, and IDH1/2 have all been described. Despite this, the initiating genetic events responsible for the development of MPNs is still not totally understood.
Collapse
|
1524
|
Metellus P, Colin C, Taieb D, Guedj E, Nanni-Metellus I, de Paula AM, Colavolpe C, Fuentes S, Dufour H, Barrie M, Chinot O, Ouafik L, Figarella-Branger D. IDH mutation status impact on in vivo hypoxia biomarkers expression: new insights from a clinical, nuclear imaging and immunohistochemical study in 33 glioma patients. J Neurooncol 2011; 105:591-600. [PMID: 21643985 DOI: 10.1007/s11060-011-0625-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 05/25/2011] [Indexed: 12/15/2022]
Abstract
Mutations in the gene encoding isocitrate dehydrogenase enzyme isoforms 1 (IDH1) and 2 (IDH2) have recently been identified in a large proportion of glial tumors of the CNS, but their mechanistic role in tumor development remains unclear. Here, we assessed the actual impact of IDH1 and IDH2 mutations in patients harboring WHO grade II and III gliomas. We sequenced IDH1 at codon 132 and IDH2 at codon 172 in 33 patients with WHO grade II and III gliomas who benefited from a preoperative (18)F-FDG positron emission tomography (PET). Immunohistochemical expression of Hypoxia Inducible Factor-1alpha (HIF-1α), Carbonic Anhydrase IX (CAIX), Glucose Transporter 1 (GLUT1) and Caspase 3 active form (CASP3) along with the R132HIDH1 mutation was assessed in all cases as well as 1p/19q deletion status and p53 expression. HIF-1α expression was found in 15% of IDH-mutated compared to 7.7% of IDH-nonmutated tumors (P = 0.954). Also, GLUT-1 positive staining was found in 5% of IDH-mutated and in 7.1% of IDH-nonmutated tumors (P = 0.794). Finally, CA-IX expression was found in 15% of IDH-mutated and in 7.7% of IDH-nonmutated tumors (P = 0.484). The combined expression of these three hypoxic markers was found in two WHO grade III tumors, one of which was IDH-mutated whereas the other was IDH-nonmutated (P = 0.794). In IDH-mutated tumors, the median SUVmax ratio was 2.24 versus 2.15 in IDH-nonmutated tumors (P = 0.775). Together, these data question the actual relationship between IDH mutation status and in vivo hypoxic biomarkers expression in WHO grade II and III gliomas.
Collapse
Affiliation(s)
- Philippe Metellus
- Hôpital de la Timone, Service de Neurochirurgie, Assistance Publique-Hôpitaux de Marseille, 13000 Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1525
|
Stieber D, Abdul Rahim SA, Niclou SP. Novel ways to target brain tumour metabolism. Expert Opin Ther Targets 2011; 15:1227-39. [PMID: 21635150 DOI: 10.1517/14728222.2011.588211] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Glioblastoma remains a highly aggressive primary brain cancer with very poor prognosis. The detection of mutations in the metabolic enzyme isocitrate dehydrogenase in gliomas, has broadened our view of tumourigenic mechanisms. Together with renewed awareness of tumour-specific energy metabolism, research is pointed towards novel ways for targeting brain cancer. AREAS COVERED This paper reviews recent knowledge on the possible tumourigenic mechanism of mutant isocitrate dehydrogenase, and provides a detailed overview of cancer-specific metabolic enzymes associated with glycolysis and intracellular pH regulation. It also discusses available drugs that may serve as a basis for novel drug development to target metabolic transformation in gliomas. EXPERT OPINION Despite the fact that energy metabolism is a very basic cellular process, tumour specific alterations in key metabolic processes represent promising targets for glioma treatment. Novel therapies against gliomas, including those that target metabolic transformation, need to consider the genetic background of the individual tumours, to allow the correlation of treatment response with the underlying biological status, both in preclinical and clinical studies.
Collapse
Affiliation(s)
- Daniel Stieber
- Centre de Recherche Public de la Santé (CRP-Santé), Oncology Department , NorLux Neuro-Oncology Laboratory, Luxembourg
| | | | | |
Collapse
|
1526
|
Abstract
Recent data on DNA sequencing of human tumours have established that cancer cells contain thousands of mutations. These data support the concept that cancer cells express a mutator phenotype. This Perspective considers the evidence supporting the mutator phenotype hypothesis, the origin and consequences of a mutator phenotype, the implications for personalized medicine and the feasibility of ablating tumours by error catastrophe.
Collapse
Affiliation(s)
- Lawrence A Loeb
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA.
| |
Collapse
|
1527
|
Mullighan C, Petersdorf E, Davies SM, DiPersio J. From trees to the forest: genes to genomics. Biol Blood Marrow Transplant 2011; 17:S52-7. [PMID: 21195310 DOI: 10.1016/j.bbmt.2010.10.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Crick, Watson, and colleagues revealed the genetic code in 1953, and since that time, remarkable progress has been made in understanding what makes each of us who we are. Identification of single genes important in disease, and the development of a mechanistic understanding of genetic elements that regulate gene function, have cast light on the pathophysiology of many heritable and acquired disorders. In 1990, the human genome project commenced, with the goal of sequencing the entire human genome, and a "first draft" was published with astonishing speed in 2001. The first draft, although an extraordinary achievement, reported essentially an imaginary haploid mix of alleles rather than a true diploid genome. In the years since 2001, technology has further improved, and efforts have been focused on filling in the gaps in the initial genome and starting the huge task of looking at normal variation in the human genome. This work is the beginning of understanding human genetics in the context of the structure of the genome as a complete entity, and as more than simply the sum of a series of genes. We present 3 studies in this review that apply genomic approaches to leukemia and to transplantation to improve and extend therapies.
Collapse
Affiliation(s)
- Charles Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | |
Collapse
|
1528
|
Shibata T, Kokubu A, Miyamoto M, Sasajima Y, Yamazaki N. Mutant IDH1 confers an in vivo growth in a melanoma cell line with BRAF mutation. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1395-402. [PMID: 21356389 DOI: 10.1016/j.ajpath.2010.12.011] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 11/14/2010] [Accepted: 12/07/2010] [Indexed: 11/19/2022]
Abstract
Melanoma is the most deadly tumor of the skin, and systemic therapies for the advanced stage are still limited. Recent genetic analyses have revealed the molecular diversity of melanoma and potential therapeutic targets. By screening a cohort of 142 primary nonepithelial tumors, we discovered that about 10% of melanoma cases (4/39) harbored an IDH1 or IDH2 mutation. These mutations were found to coexist with BRAF or KIT mutation, and all IDH1 mutations were detected in metastatic lesions. BRAF-mutated melanoma cells, additionally expressing the cancer-related IDH1 mutant, acquired increased colony-forming and in vivo growth activities and showed enhanced activation of the MAPK and STAT3 pathways. Genome-wide gene expression profiling demonstrated that mutant IDH1 affected the expression of a set of genes. Especially, it caused the induction of growth-related transcriptional regulators (Jun, N-myc, Atf3) and the reduction of Rassf1 and two dehydrogenase genes (Dhrs1 and Adh5), which may be involved in the carcinogenesis of IDH1-mutated tumors. Our analyses demonstrate that IDH1 mutation works with other oncogenic mutations and could contribute to the metastasis in melanoma.
Collapse
Affiliation(s)
- Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
1529
|
Somatic mutations in acute promyelocytic leukemia (APL) identified by exome sequencing. Leukemia 2011; 25:1519-22. [DOI: 10.1038/leu.2011.114] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1530
|
Tibes R, Mesa RA. Myeloproliferative neoplasms 5 years after discovery of JAK2V617F: what is the impact of JAK2 inhibitor therapy? Leuk Lymphoma 2011; 52:1178-87. [DOI: 10.3109/10428194.2011.566952] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
1531
|
Risk stratification of intermediate-risk acute myeloid leukemia: integrative analysis of a multitude of gene mutation and gene expression markers. Blood 2011; 118:1069-76. [PMID: 21596848 DOI: 10.1182/blood-2011-02-334748] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Numerous molecular markers have been recently discovered as potential prognostic factors in acute myeloid leukemia (AML). It has become of critical importance to thoroughly evaluate their interrelationships and relative prognostic importance. Gene expression profiling was conducted in a well-characterized cohort of 439 AML patients (age < 60 years) to determine expression levels of EVI1, WT1, BCL2, ABCB1, BAALC, FLT3, CD34, INDO, ERG and MN1. A variety of AML-specific mutations were evaluated, that is, FLT3, NPM1, N-RAS, K-RAS, IDH1, IDH2, and CEBPA(DM/SM) (double/single). Univariable survival analysis shows that (1) patients with FLT3(ITD) mutations have inferior overall survival (OS) and event-free survival (EFS), whereas CEBPA(DM) and NPM1 mutations indicate favorable OS and EFS in intermediate-risk AML, and (2) high transcript levels of BAALC, CD34, MN1, EVl1, and ERG predict inferior OS and EFS. In multivariable survival analysis, CD34, ERG, and CEBPA(DM) remain significant. Using survival tree and regression methodologies, we show that CEBPA(DM), CD34, and IDH2 mutations are capable of separating the intermediate group into 2 AML subgroups with highly distinctive survival characteristics (OS at 60 months: 51.9% vs 14.9%). The integrated statistical approach demonstrates that from the multitude of biomarkers a greatly condensed subset can be selected for improved stratification of intermediate-risk AML.
Collapse
|
1532
|
Abstract
We have investigated the prognostic significance of isocitrate dehydrogenase 2 (IDH2) mutations in 1473 younger adult acute myeloid leukemia patients treated in 2 United Kingdom Medical Research Council trials. An IDH2 mutation was present in 148 cases (10%), 80% at R140 and 20% at R172. Patient characteristics and outcome differed markedly between the 2 mutations. IDH2(R140) significantly correlated with nucleophosmin mutations (NPM1(MUT)), whereas IDH2(R172) cases generally lacked other molecular mutations. An IDH2(R140) mutation was an independent favorable prognostic factor for relapse (P = .004) and overall survival (P = .008), and there was no significant heterogeneity with regard to NPM1 or FLT3 internal tandem duplication (FLT3/ITD) genotype. Relapse in FLT3/ITD(WT)NPM1(MUT)IDH2(R140) patients was lower than in favorable-risk cytogenetics patients in the same cohort (20% and 38% at 5 years, respectively). The presence of an IDH2(R172) mutation was associated with a significantly worse outcome than IDH2(R140), and relapse in FLT3/ITD(WT)NPM1(WT)IDH2(R172) patients was comparable with adverse-risk cytogenetics patients (76% and 72%, respectively).
Collapse
|
1533
|
Godley LA, Cunningham J, Dolan ME, Huang RS, Gurbuxani S, McNerney ME, Larson RA, Leong H, Lussier Y, Onel K, Odenike O, Stock W, White KP, Le Beau MM. An integrated genomic approach to the assessment and treatment of acute myeloid leukemia. Semin Oncol 2011; 38:215-24. [PMID: 21421111 DOI: 10.1053/j.seminoncol.2011.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Traditionally, new scientific advances have been applied quickly to the leukemias based on the ease with which relatively pure samples of malignant cells can be obtained. Currently, our arsenal of approaches used to characterize an individual's acute myeloid leukemia (AML) combines hematopathologic evaluation, flow cytometry, cytogenetic analysis, and molecular studies focused on a few key genes. The advent of high-throughput methods capable of full-genome evaluation presents new options for a revolutionary change in the way we diagnose, characterize, and treat AML. Next-generation DNA sequencing techniques allow full sequencing of a cancer genome or transcriptome, with the hope that this will be affordable for routine clinical care within the decade. Microarray-based testing will define gene and miRNA expression, DNA methylation patterns, chromosomal imbalances, and predisposition to disease and chemosensitivity. The vision for the future entails an integrated and automated approach to these analyses, bringing the possibility of formulating an individualized treatment plan within days of a patient's initial presentation. With these expectations comes the hope that such an approach will lead to decreased toxicities and prolonged survival for patients.
Collapse
Affiliation(s)
- Lucy A Godley
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA. lgodley@medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1534
|
|
1535
|
Stoddart A, McNerney ME, Bartom E, Bergerson R, Young DJ, Qian Z, Wang J, Fernald AA, Davis EM, Larson RA, White KP, Le Beau MM. Genetic pathways leading to therapy-related myeloid neoplasms. Mediterr J Hematol Infect Dis 2011; 3:e2011019. [PMID: 21713073 PMCID: PMC3113274 DOI: 10.4084/mjhid.2011.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/21/2011] [Indexed: 01/03/2023] Open
Abstract
Therapy-related myeloid neoplasm (t-MN) is a distinctive clinical syndrome occurring after exposure to chemotherapy or radiotherapy. t-MN arises in most cases from a multipotential hematopoietic stem cell or, less commonly, in a lineage committed progenitor cell. The prognosis for patients with t-MN is poor, as current forms of therapy are largely ineffective. Cytogenetic analysis, molecular analysis and gene expression profiling analysis of t-MN has revealed that there are distinct subtypes of the disease; however, our understanding of the genetic basis of t-MN is incomplete. Elucidating the genetic pathways and molecular networks that are perturbed in t-MNs, may facilitate the identification of therapeutic targets that can be exploited for the development of urgently-needed targeted therapies.
Collapse
|
1536
|
Abstract
PURPOSE OF REVIEW This review describes recent advances in technologies for massive parallel sequencing of human genomes and discusses their application to the analysis of cancer genomes. RECENT FINDINGS Several different instruments are now available for next-generation sequencing (NGS). Although they use different sample preparation and sequencing technologies, they all rely on large computing capacity for assembling sequences and identifying somatic mutations against the background of genetic variations. Recent examples of NGS application to cancer genomes include the sequencing of 22 cases of glioblastoma multiforme that identified IDH1, the gene encoding isocitrate dehydrogenase 1, as target for cancer-driving mutations. Analysis of entire genomes of single samples of lung cancer and melanoma has brought unprecedented details on how tobacco carcinogens and UV exposure, respectively, may sculpt specific mutation landscapes. In breast cancer, comparative genome sequencing of primary and secondary lesions of a single patient has revealed clues on the phylogeny of tumor cells. SUMMARY NGS is opening a new era for understanding how environmental factors alter the human genome to generate cancerous cells, paving the way to a better understanding of the origins of human cancer.
Collapse
|
1537
|
Abstract
Common or sporadic systolic heart failure (heart failure) is the clinical syndrome of insufficient forward cardiac output resulting from myocardial disease. Most heart failure is the consequence of ischemic or idiopathic cardiomyopathy. There is a clear familial predisposition to heart failure, with a genetic component estimated to confer between 20% and 30% of overall risk. The multifactorial etiology of this syndrome has complicated identification of its genetic underpinnings. Until recently, almost all genetic studies of heart failure were designed and deployed according to the common disease-common variant hypothesis, in which individual risk alleles impart a small positive or negative effect and overall genetic risk is the cumulative impact of all functional genetic variations. Early studies used a candidate gene approach focused mainly on factors within adrenergic and renin-angiotensin pathways that affect heart failure progression and are targeted by standard pharmacotherapeutics. Many of these reported allelic associations with heart failure have not been replicated. However, the preponderance of data supports risk-modifier effects for the Arg389Gly polymorphism of β1-adrenergic receptors and the intron 16 in/del polymorphism of angiotensin-converting enzyme. Recent unbiased studies using genome-wide single nucleotide polymorphism microarrays have shown fewer positive results than when these platforms were applied to hypertension, myocardial infarction, or diabetes, possibly reflecting the complex etiology of heart failure. A new cardiovascular gene-centric subgenome single nucleotide polymorphism array identified a common heat failure risk allele at 1p36 in multiple independent cohorts, but the biological mechanism for this association is still uncertain. It is likely that common gene polymorphisms account for only a fraction of individual genetic heart failure risk, and future studies using deep resequencing are likely to identify rare gene variants with larger biological effects.
Collapse
Affiliation(s)
- Gerald W Dorn
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
1538
|
Abstract
High-throughput tools for nucleic acid characterization now provide the means to conduct comprehensive analyses of all somatic alterations in the cancer genomes. Both large-scale and focused efforts have identified new targets of translational potential. The deluge of information that emerges from these genome-scale investigations has stimulated a parallel development of new analytical frameworks and tools. The complexity of somatic genomic alterations in cancer genomes also requires the development of robust methods for the interrogation of the function of genes identified by these genomics efforts. Here we provide an overview of the current state of cancer genomics, appraise the current portals and tools for accessing and analyzing cancer genomic data, and discuss emerging approaches to exploring the functions of somatically altered genes in cancer.
Collapse
Affiliation(s)
- Lynda Chin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | | | |
Collapse
|
1539
|
Pietrak B, Zhao H, Qi H, Quinn C, Gao E, Boyer JG, Concha N, Brown K, Duraiswami C, Wooster R, Sweitzer S, Schwartz B. A tale of two subunits: how the neomorphic R132H IDH1 mutation enhances production of αHG. Biochemistry 2011; 50:4804-12. [PMID: 21524095 DOI: 10.1021/bi200499m] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heterozygously expressed single-point mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2, respectively) render these dimeric enzymes capable of producing the novel metabolite α-hydroxyglutarate (αHG). Accumulation of αHG is used as a biomarker for a number of cancer types, helping to identify tumors with similar IDH mutations. With IDH1, it has been shown that one role of the mutation is to increase the rate of conversion from αKG to αHG. To improve our understanding of the function of this mutation, we have detailed the kinetics of the normal (isocitrate to αKG) and neomorphic (αKG to αHG) reactions, as well as the coupled conversion of isocitrate to αHG. We find that the mutant IDH1 is very efficient in this coupled reaction, with the ability to form αHG from isocitrate and NADP(+). The wild type/wild type IDH1 is also able to catalyze this conversion, though it is much more sensitive to concentrations of isocitrate. This difference in behavior can be attributed to the competitive binding between isocitrate and αKG, which is made more favorable for αKG by the neomorphic mutation at arginine 132. Thus, each partial reaction in the heterodimer is functionally isolated from the other. To test whether there is a cooperative effect resulting from the two subunits being in a dimer, we selectively inactivated each subunit with a secondary mutation in the NADP/H binding site. We observed that the remaining, active subunit was unaffected in its associated activity, reinforcing the notion of each subunit being functionally independent. This was further demonstrated using a monomeric form of IDH from Azotobacter vinelandii, which can be shown to gain the same neomorphic reaction when a homologous mutation is introduced into that protein.
Collapse
Affiliation(s)
- Beth Pietrak
- Departments of Biological Reagents and Assay Development, Screening and Compound Profiling, Statistical Sciences, Computational and Structural Sciences, and Cancer Metabolism, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1540
|
Patnaik MM, Lasho TL, Finke CM, Knudson RA, Ketterling RP, Chen D, Hoyer JD, Hanson CA, Tefferi A. Isolated del(5q) in myeloid malignancies: clinicopathologic and molecular features in 143 consecutive patients. Am J Hematol 2011; 86:393-8. [PMID: 21523797 DOI: 10.1002/ajh.21984] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
World Health Organization (WHO) criteria were used to identify 143 consecutive patients (median age 73 years; 90 females) with myeloid neoplasms and isolated del(5q) seen between 1989 and 2009. We have previously reported on 88 (61%) of these patients who met criteria for WHO defined "myelodysplastic syndromes (MDS) with isolated del(5q)." The remaining 55 patients were classified as having "other" MDS variants (n = 29; 20%), acute myeloid leukemia (AML; n = 14; 10%), or myeloproliferative neoplasms (MPN; n = 12; 8%). DNA was available in 138 patients and mutation screening revealed 20 cases with JAK2, 6 with IDH, and 3 with MPL mutations; JAK2 and MPL mutations were seen mostly in MPN or "MDS with isolated del(5q)" whereas IDH mutations were frequent in other MDS variants. Overall median survival for the 143 patient cohort was 35 months and leukemic transformation (LT) was documented in 19 (~13%) cases. "MDS with isolated del(5q)" had the best prognosis with median survival of 66 months and LT rate of ~6%. Survival was poor among the other myeloid neoplasm subgroups regardless of specific morphologic diagnosis. Multivariable analysis identified higher leukocyte count and percentage of bone marrow and circulating blasts as independent predictors of shortened survival. The first two parameters and the presence of IDH mutations predicted inferior leukemia-free survival. The current study validates the prognostic relevance of considering "MDS with isolated del(5q)" as a separate WHO subcategory and identifies leukocytosis, higher blast count, and IDH mutations as being prognostically detrimental, in myeloid neoplasms associated with isolated del(5q).
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Cell Transformation, Neoplastic/genetics
- Chromosome Deletion
- Chromosomes, Human, Pair 15
- Female
- Follow-Up Studies
- Humans
- Isocitrate Dehydrogenase/genetics
- Janus Kinase 2/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/physiopathology
- Leukemia, Myeloid, Acute/therapy
- Leukocytosis
- Male
- Medical Records
- Middle Aged
- Mutation
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/physiopathology
- Myelodysplastic Syndromes/therapy
- Myeloproliferative Disorders/diagnosis
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/physiopathology
- Myeloproliferative Disorders/therapy
- Prognosis
- Receptors, Thrombopoietin/genetics
- Retrospective Studies
- Survival Analysis
- United States
Collapse
Affiliation(s)
- Mrinal M Patnaik
- Department of Medicine, Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1541
|
Abstract
BACKGROUND Advances in the understanding of complex trait genetics have always been enabled by advances in genomic technology. Next-generation sequencing (NGS) is set to revolutionize the way complex trait genetics research is carried out. RESULTS NGS has multiple applications in the field of human genetics, but is accompanied by substantial study design, analysis and interpretation challenges. This review discusses key aspects of study design considerations, data handling issues and required analytical developments. We also highlight early successes in mapping genetic traits using NGS. CONCLUSION NGS opens the entire spectrum of genomic alterations for the genetic analysis of complex traits and there are early publications illustrating its power. Continuing development in analytical tools will allow the promise of NGS to be realized.
Collapse
|
1542
|
Kim RY, Xu H, Myllykangas S, Ji H. Genetic-based biomarkers and next-generation sequencing: the future of personalized care in colorectal cancer. Per Med 2011; 8:331-345. [PMID: 23662107 PMCID: PMC3646399 DOI: 10.2217/pme.11.16] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The past 5 years have witnessed extraordinary advances in the field of DNA sequencing technology. What once took years to accomplish with Sanger sequencing can now be accomplished in a matter of days with next-generation sequencing (NGS) technology. This has allowed researchers to sequence individual genomes and match combinations of mutations with specific diseases. As cancer is inherently a disease of the genome, it is not surprising to see NGS technology already being applied to cancer research with promises of greater understanding of carcinogenesis. While the task of deciphering the cancer genomic code remains ongoing, we are already beginning to see the application of genetic-based testing in the area of colorectal cancer. In this article we will provide an overview of current colorectal cancer genetic-based biomarkers, namely mutations and other genetic alterations in cancer genome DNA, discuss recent advances in NGS technology and speculate on future directions for the application of NGS technology to colorectal cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Redecca Y Kim
- Author for correspondence: Department of General Surgery, Stanford University, CCSR 1115, 269 Campus Drive, Stanford, CA 94305, USA Tel.:+1 650 723 4000
| | - Hua Xu
- Stanford Genome Technology Center, Stanford University, Stanford, CA, USA
| | - Samuel Myllykangas
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, USA
| | - Hanlee Ji
- Stanford Genome Technology Center, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
1543
|
Natrajan R, Reis-Filho JS. Next-generation sequencing applied to molecular diagnostics. Expert Rev Mol Diagn 2011; 11:425-44. [PMID: 21545259 DOI: 10.1586/erm.11.18] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Next-generation sequencing technologies have begun to revolutionize the field of cancer genetics through rapid and accurate assessment of a patient's DNA makeup with minimal cost. These technologies have already led to the realization of the inter- and intra-tumor genetic heterogeneity and the identification of novel mutations and chimeric genes, however, several challenges lie ahead. Given the low number of recurrent somatic genetic aberrations in common types of cancer, the identification of 'driver' genetic aberrations has proven challenging. Furthermore, implementation of next-generation sequencing and/or some of its derivatives into routine practice as diagnostic tests will require in-depth understanding of the pitfalls of these technologies and a great degree of bioinformatic expertise. This article focuses on the contribution of next-generation sequencing technologies to diagnosis and cancer prognostication and prediction.
Collapse
Affiliation(s)
- Rachael Natrajan
- Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK.
| | | |
Collapse
|
1544
|
Lin J, Qian J, Yao DM, Li Y, Yang J, Chen Q, Chai HY, Xiao GF, Xu WR. Rapid and reliable detection of IDH1 R132 mutations in acute myeloid leukemia using high-resolution melting curve analysis. Clin Biochem 2011; 44:779-83. [PMID: 21539821 DOI: 10.1016/j.clinbiochem.2011.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/13/2011] [Accepted: 04/17/2011] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The mutations of isocitrate dehydrogenase 1 (IDH1) gene have been identified in a proportion of hematologic malignancies including acute myeloid leukemia (AML). The aim of the present study was to explore the reliability of the high-resolution melting analysis (HRMA) for the identification of IDH1 R132 mutations in AML. DESIGNS AND METHODS We evaluated the sensitivity of HRMA in the detection of IDH1 R132 mutation and screened IDH1 mutations in 110 AML patients using HRMA. The results of HRMA were validated by direct DNA sequencing. RESULTS The reproducible sensitivity of HRMA was 5% for the detection of IDH1 R132 mutation, higher than 10% of direct DNA sequencing. Heterozygous IDH1 mutations were identified in 4 (3.6%) AML cases, which were R132H in 3 cases and R132S in 1 case confirmed by DNA sequencing. CONCLUSION The HRMA is a rapid, accurate, reliable, high-throughput method to screen IDH1 gene mutations.
Collapse
Affiliation(s)
- Jiang Lin
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
1545
|
Welch JS, Westervelt P, Ding L, Larson DE, Klco JM, Kulkarni S, Wallis J, Chen K, Payton JE, Fulton RS, Veizer J, Schmidt H, Vickery TL, Heath S, Watson MA, Tomasson MH, Link DC, Graubert TA, DiPersio JF, Mardis ER, Ley TJ, Wilson RK. Use of whole-genome sequencing to diagnose a cryptic fusion oncogene. JAMA 2011; 305:1577-84. [PMID: 21505136 PMCID: PMC3156695 DOI: 10.1001/jama.2011.497] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CONTEXT Whole-genome sequencing is becoming increasingly available for research purposes, but it has not yet been routinely used for clinical diagnosis. OBJECTIVE To determine whether whole-genome sequencing can identify cryptic, actionable mutations in a clinically relevant time frame. DESIGN, SETTING, AND PATIENT We were referred a difficult diagnostic case of acute promyelocytic leukemia with no pathogenic X-RARA fusion identified by routine metaphase cytogenetics or interphase fluorescence in situ hybridization (FISH). The case patient was enrolled in an institutional review board-approved protocol, with consent specifically tailored to the implications of whole-genome sequencing. The protocol uses a "movable firewall" that maintains patient anonymity within the entire research team but allows the research team to communicate medically relevant information to the treating physician. MAIN OUTCOME MEASURES Clinical relevance of whole-genome sequencing and time to communicate validated results to the treating physician. RESULTS Massively parallel paired-end sequencing allowed identification of a cytogenetically cryptic event: a 77-kilobase segment from chromosome 15 was inserted en bloc into the second intron of the RARA gene on chromosome 17, resulting in a classic bcr3 PML-RARA fusion gene. Reverse transcription polymerase chain reaction sequencing subsequently validated the expression of the fusion transcript. Novel FISH probes identified 2 additional cases of t(15;17)-negative acute promyelocytic leukemia that had cytogenetically invisible insertions. Whole-genome sequencing and validation were completed in 7 weeks and changed the treatment plan for the patient. CONCLUSION Whole-genome sequencing can identify cytogenetically invisible oncogenes in a clinically relevant time frame.
Collapse
MESH Headings
- Adult
- Chromosome Breakpoints
- Chromosomes, Human, Pair 15/genetics
- Chromosomes, Human, Pair 17/genetics
- Gene Fusion
- Genome, Human
- Humans
- Introns
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/therapy
- Male
- Nuclear Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Promyelocytic Leukemia Protein
- Receptors, Retinoic Acid/genetics
- Retinoic Acid Receptor alpha
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
- Transcription Factors/genetics
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- John S. Welch
- Department of Medicine, Washington University, St. Louis, MO
| | | | - Li Ding
- The Genome Institute, Washington University, St. Louis, MO
| | | | - Jeffery M. Klco
- Department of Pathology and Immunology, Washington University, St. Louis, MO
| | - Shashikant Kulkarni
- Department of Pathology and Immunology, Washington University, St. Louis, MO
- Department of Genetics, Washington University, St. Louis, MO
- Department of Pediatrics, Washington University, St. Louis, MO
| | - John Wallis
- The Genome Institute, Washington University, St. Louis, MO
| | - Ken Chen
- The Genome Institute, Washington University, St. Louis, MO
| | | | | | - Joelle Veizer
- The Genome Institute, Washington University, St. Louis, MO
| | | | | | - Sharon Heath
- Department of Medicine, Washington University, St. Louis, MO
| | - Mark A. Watson
- The Genome Institute, Washington University, St. Louis, MO
- Department of Pathology and Immunology, Washington University, St. Louis, MO
| | | | - Daniel C. Link
- Department of Medicine, Washington University, St. Louis, MO
| | | | | | - Elaine R. Mardis
- The Genome Institute, Washington University, St. Louis, MO
- Department of Genetics, Washington University, St. Louis, MO
| | - Timothy J. Ley
- Department of Medicine, Washington University, St. Louis, MO
- The Genome Institute, Washington University, St. Louis, MO
- Department of Genetics, Washington University, St. Louis, MO
| | - Richard K. Wilson
- The Genome Institute, Washington University, St. Louis, MO
- Department of Genetics, Washington University, St. Louis, MO
| |
Collapse
|
1546
|
Link DC, Schuettpelz LG, Shen D, Wang J, Walter MJ, Kulkarni S, Payton JE, Ivanovich J, Goodfellow PJ, Le Beau M, Koboldt DC, Dooling DJ, Fulton RS, Bender RHF, Fulton LL, Delehaunty KD, Fronick CC, Appelbaum EL, Schmidt H, Abbott R, O'Laughlin M, Chen K, McLellan MD, Varghese N, Nagarajan R, Heath S, Graubert TA, Ding L, Ley TJ, Zambetti GP, Wilson RK, Mardis ER. Identification of a novel TP53 cancer susceptibility mutation through whole-genome sequencing of a patient with therapy-related AML. JAMA 2011; 305:1568-76. [PMID: 21505135 PMCID: PMC3170052 DOI: 10.1001/jama.2011.473] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CONTEXT The identification of patients with inherited cancer susceptibility syndromes facilitates early diagnosis, prevention, and treatment. However, in many cases of suspected cancer susceptibility, the family history is unclear and genetic testing of common cancer susceptibility genes is unrevealing. OBJECTIVE To apply whole-genome sequencing to a patient without any significant family history of cancer but with suspected increased cancer susceptibility because of multiple primary tumors to identify rare or novel germline variants in cancer susceptibility genes. DESIGN, SETTING, AND PARTICIPANT: Skin (normal) and bone marrow (leukemia) DNA were obtained from a patient with early-onset breast and ovarian cancer (negative for BRCA1 and BRCA2 mutations) and therapy-related acute myeloid leukemia (t-AML) and analyzed with the following: whole-genome sequencing using paired-end reads, single-nucleotide polymorphism (SNP) genotyping, RNA expression profiling, and spectral karyotyping. MAIN OUTCOME MEASURES Structural variants, copy number alterations, single-nucleotide variants, and small insertions and deletions (indels) were detected and validated using the described platforms. RESULTS; Whole-genome sequencing revealed a novel, heterozygous 3-kilobase deletion removing exons 7-9 of TP53 in the patient's normal skin DNA, which was homozygous in the leukemia DNA as a result of uniparental disomy. In addition, a total of 28 validated somatic single-nucleotide variations or indels in coding genes, 8 somatic structural variants, and 12 somatic copy number alterations were detected in the patient's leukemia genome. CONCLUSION Whole-genome sequencing can identify novel, cryptic variants in cancer susceptibility genes in addition to providing unbiased information on the spectrum of mutations in a cancer genome.
Collapse
MESH Headings
- Adult
- Age of Onset
- Breast Neoplasms/therapy
- Cystadenocarcinoma, Serous/therapy
- DNA, Neoplasm/genetics
- Female
- Genes, p53/genetics
- Genetic Predisposition to Disease
- Genome, Human/genetics
- Humans
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/genetics
- Ovarian Neoplasms/therapy
- Polymorphism, Single Nucleotide
- Sequence Analysis, DNA
- Sequence Deletion
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Daniel C. Link
- Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | | | - Dong Shen
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Jinling Wang
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN
| | - Matthew J. Walter
- Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Shashikant Kulkarni
- Department of Pediatrics, Washington University, St. Louis, MO
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
- Department of Pathology and Immunology, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Jacqueline E. Payton
- Department of Pathology and Immunology, Siteman Cancer Center, Washington University, St. Louis, MO
| | | | | | | | - Daniel C. Koboldt
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - David J. Dooling
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Robert S. Fulton
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - R. Hugh F. Bender
- Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Lucinda L. Fulton
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | | | - Catrina C. Fronick
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | | | - Heather Schmidt
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Rachel Abbott
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Michelle O'Laughlin
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Ken Chen
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Michael D. McLellan
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Nobish Varghese
- Department of Pathology and Immunology, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Rakesh Nagarajan
- Department of Pathology and Immunology, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Sharon Heath
- Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Timothy A. Graubert
- Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - Li Ding
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Timothy J. Ley
- Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Gerard P. Zambetti
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN
| | - Richard K. Wilson
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| | - Elaine R. Mardis
- Department of Genetics, The Genome Center, Washington University, St. Louis, MO
| |
Collapse
|
1547
|
Genome interrupted: sequencing of prostate cancer reveals the importance of chromosomal rearrangements. Genome Med 2011; 3:23. [PMID: 21542879 PMCID: PMC3129639 DOI: 10.1186/gm237] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A recent study involving whole genome sequencing of seven prostate cancers has provided the first comprehensive assessment of genomic changes that underlie this common malignancy. Point mutations were found to be infrequent but changes in chromosome structure were common. Rearrangements were linked to chromatin organization and associated with regions involved in transcription factor binding. Novel candidate prostate cancer genes were also identified, highlighting the importance of genome sequencing to identify oncogenic changes that are otherwise invisible to detection.
Collapse
|
1548
|
Sardina JL, López-Ruano G, Sánchez-Sánchez B, Llanillo M, Hernández-Hernández A. Reactive oxygen species: are they important for haematopoiesis? Crit Rev Oncol Hematol 2011; 81:257-74. [PMID: 21507675 DOI: 10.1016/j.critrevonc.2011.03.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 03/15/2011] [Accepted: 03/22/2011] [Indexed: 02/07/2023] Open
Abstract
The production of reactive oxygen species (ROS) has traditionally been related to deleterious effects for cells. However, it is now widely accepted that ROS can play an important role in regulating cellular signalling and gene expression. NADPH oxidase ROS production seems to be especially important in this regard. Some lines of evidence suggest that ROS may be important modulators of cell differentiation, including haematopoietic differentiation, in both physiologic and pathologic conditions. Here we shall review how ROS can regulate cell signalling and gene expression. We shall also focus on the importance of ROS for haematopoietic stem cell (HSC) biology and for haematopoietic differentiation. We shall review the involvement of ROS and NADPH oxidases in cancer, and in particular what is known about the relationship between ROS and haematological malignancies. Finally, we shall discuss the use of ROS as cancer therapeutic targets.
Collapse
Affiliation(s)
- José L Sardina
- Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca, Spain
| | | | | | | | | |
Collapse
|
1549
|
High-resolution characterization of a hepatocellular carcinoma genome. Nat Genet 2011; 43:464-9. [PMID: 21499249 DOI: 10.1038/ng.804] [Citation(s) in RCA: 239] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 03/14/2011] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma, one of the most common virus-associated cancers, is the third most frequent cause of cancer-related death worldwide. By massively parallel sequencing of a primary hepatitis C virus-positive hepatocellular carcinoma (36× coverage) and matched lymphocytes (>28× coverage) from the same individual, we identified more than 11,000 somatic substitutions of the tumor genome that showed predominance of T>C/A>G transition and a decrease of the T>C substitution on the transcribed strand, suggesting preferential DNA repair. Gene annotation enrichment analysis of 63 validated non-synonymous substitutions revealed enrichment of phosphoproteins. We further validated 22 chromosomal rearrangements, generating four fusion transcripts that had altered transcriptional regulation (BCORL1-ELF4) or promoter activity. Whole-exome sequencing at a higher sequence depth (>76× coverage) revealed a TSC1 nonsense substitution in a subpopulation of the tumor cells. This first high-resolution characterization of a virus-associated cancer genome identified previously uncharacterized mutation patterns, intra-chromosomal rearrangements and fusion genes, as well as genetic heterogeneity within the tumor.
Collapse
|
1550
|
Wendl MC, Wallis JW, Lin L, Kandoth C, Mardis ER, Wilson RK, Ding L. PathScan: a tool for discerning mutational significance in groups of putative cancer genes. ACTA ACUST UNITED AC 2011; 27:1595-602. [PMID: 21498403 DOI: 10.1093/bioinformatics/btr193] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
MOTIVATION The expansion of cancer genome sequencing continues to stimulate development of analytical tools for inferring relationships between somatic changes and tumor development. Pathway associations are especially consequential, but existing algorithms are demonstrably inadequate. METHODS Here, we propose the PathScan significance test for the scenario where pathway mutations collectively contribute to tumor development. Its design addresses two aspects that established methods neglect. First, we account for variations in gene length and the consequent differences in their mutation probabilities under the standard null hypothesis of random mutation. The associated spike in computational effort is mitigated by accurate convolution-based approximation. Second, we combine individual probabilities into a multiple-sample value using Fisher-Lancaster theory, thereby improving differentiation between a few highly mutated genes and many genes having only a few mutations apiece. We investigate accuracy, computational effort and power, reporting acceptable performance for each. RESULTS As an example calculation, we re-analyze KEGG-based lung adenocarcinoma pathway mutations from the Tumor Sequencing Project. Our test recapitulates the most significant pathways and finds that others for which the original test battery was inconclusive are not actually significant. It also identifies the focal adhesion pathway as being significantly mutated, a finding consistent with earlier studies. We also expand this analysis to other databases: Reactome, BioCarta, Pfam, PID and SMART, finding additional hits in ErbB and EPHA signaling pathways and regulation of telomerase. All have implications and plausible mechanistic roles in cancer. Finally, we discuss aspects of extending the method to integrate gene-specific background rates and other types of genetic anomalies. AVAILABILITY PathScan is implemented in Perl and is available from the Genome Institute at: http://genome.wustl.edu/software/pathscan.
Collapse
Affiliation(s)
- Michael C Wendl
- The Genome Institute, Washington University, St Louis, MO 63108, USA.
| | | | | | | | | | | | | |
Collapse
|