1101
|
Bailey ML, O'Neil NJ, van Pel DM, Solomon DA, Waldman T, Hieter P. Glioblastoma cells containing mutations in the cohesin component STAG2 are sensitive to PARP inhibition. Mol Cancer Ther 2014; 13:724-32. [PMID: 24356817 PMCID: PMC4130349 DOI: 10.1158/1535-7163.mct-13-0749] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Recent data have identified STAG2, a core subunit of the multifunctional cohesin complex, as a highly recurrently mutated gene in several types of cancer. We sought to identify a therapeutic strategy to selectively target cancer cells harboring inactivating mutations of STAG2 using two independent pairs of isogenic glioblastoma cell lines containing either an endogenous mutant STAG2 allele or a wild-type STAG2 allele restored by homologous recombination. We find that mutations in STAG2 are associated with significantly increased sensitivity to inhibitors of the DNA repair enzyme PARP. STAG2-mutated, PARP-inhibited cells accumulated in G2 phase and had a higher percentage of micronuclei, fragmented nuclei, and chromatin bridges compared with wild-type STAG2 cells. We also observed more 53BP1 foci in STAG2-mutated glioblastoma cells, suggesting that these cells have defects in DNA repair. Furthermore, cells with mutations in STAG2 were more sensitive than cells with wild-type STAG2 when PARP inhibitors were used in combination with DNA-damaging agents. These data suggest that PARP is a potential target for tumors harboring inactivating mutations in STAG2, and strongly recommend that STAG2 status be determined and correlated with therapeutic response to PARP inhibitors, both prospectively and retrospectively, in clinical trials.
Collapse
Affiliation(s)
- Melanie L Bailey
- Corresponding Author: Philip Hieter, Michael Smith Laboratories, 2185 East Mall, Room 323, University of British Columbia, Vancouver, British Columbia, Canada V6T1Z4.
| | | | | | | | | | | |
Collapse
|
1102
|
Appels R, Nystrom-Persson J, Keeble-Gagnere G. Advances in genome studies in plants and animals. Funct Integr Genomics 2014; 14:1-9. [PMID: 24626952 PMCID: PMC3968518 DOI: 10.1007/s10142-014-0364-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 01/30/2023]
Abstract
The area of plant and animal genomics covers the entire suite of issues in biology because it aims to determine the structure and function of genetic material. Although specific issues define research advances at an organism level, it is evident that many of the fundamental features of genome structure and the translation of encoded information to function share common ground. The Plant and Animal Genome (PAG) conference held in San Diego (California), in January each year provides an overview across all organisms at the genome level, and often it is evident that investments in the human area provide leadership, applications, and discoveries for researchers studying other organisms. This mini-review utilizes the plenary lectures as a basis for summarizing the trends in the genome-level studies of organisms, and the lectures include presentations by Ewan Birney (EBI, UK), Eric Green (NIH, USA), John Butler (NIST, USA), Elaine Mardis (Washington, USA), Caroline Dean (John Innes Centre, UK), Trudy Mackay (NC State University, USA), Sue Wessler (UC Riverside, USA), and Patrick Wincker (Genoscope, France). The work reviewed is based on published papers. Where unpublished information is cited, permission to include the information in this manuscript was obtained from the presenters.
Collapse
Affiliation(s)
- R Appels
- Veterinary and Life Sciences, Murdoch University, 90 South Street, Murdoch, Perth, WA, 6150, Australia,
| | | | | |
Collapse
|
1103
|
Behrens A, van Deursen JM, Rudolph KL, Schumacher B. Impact of genomic damage and ageing on stem cell function. Nat Cell Biol 2014; 16:201-7. [PMID: 24576896 PMCID: PMC4214082 DOI: 10.1038/ncb2928] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Impairment of stem cell function contributes to the progressive deterioration of tissue maintenance and repair with ageing. Evidence is mounting that age-dependent accumulation of DNA damage in both stem cells and cells that comprise the stem cell microenvironment are partly responsible for stem cell dysfunction with ageing. Here, we review the impact of the various types of DNA damage that accumulate with ageing on stem cell functionality, as well as the development of cancer. We discuss DNA-damage-induced cell intrinsic and extrinsic alterations that influence these processes, and review recent advances in understanding systemic adjustments to DNA damage and how they affect stem cells.
Collapse
Affiliation(s)
- Axel Behrens
- Mammalian Genetics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK, and the School of Medicine, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | - K Lenhard Rudolph
- Leibniz Institute of Age Research, Fritz Lipmann Institute e.V., Jena, 07745, Germany, and the Research Group on Molecular Aging, Faculty of Medicine, Friedrich-Schiller-University, Jena, Germany
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931 Cologne, and the Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Institute for Genetics, and Systems Biology of Cologne, University of Cologne, Zülpicher Str. 47a, 50674 Cologne, Germany
| |
Collapse
|
1104
|
Rushworth SA, Murray MY, Zaitseva L, Bowles KM, MacEwan DJ. Identification of Bruton's tyrosine kinase as a therapeutic target in acute myeloid leukemia. Blood 2014; 123:1229-38. [PMID: 24307721 DOI: 10.1182/blood-2013-06-511154] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) is a cytoplasmic protein found in all hematopoietic cell lineages except for T cells. BTK mediates signaling downstream of a number of receptors. Pharmacologic targeting of BTK using ibrutinib (previously PCI-32765) has recently shown encouraging clinical activity in a range of lymphoid malignancies. This study reports for the first time that ibrutinib inhibits blast proliferation from human acute myeloid leukemia (AML) and that treatment with ibrutinib significantly augmented cytotoxic activities of standard AML chemotherapy cytarabine or daunorubicin. Here we describe that BTK is constitutively phosphorylated in the majority of AML samples tested, with BTK phosphorylation correlating highly with the cell's cytotoxic sensitivity toward ibrutinib. BTK-targeted RNAi knockdown reduced colony-forming capacity of primary AML blasts and proliferation of AML cell lines. We showed that ibrutinib binds at nanomolar range to BTK. Furthermore, we showed ibrutinib's antiproliferative effects in AML are mediated via an inhibitory effect on downstream nuclear factor-κB survival pathways. Moreover, ibrutinib inhibited AML cell adhesion to bone marrow stroma. Furthermore, these effects of ibrutinib in AML were seen at comparable concentrations efficacious in chronic lymphocytic leukemia. These results provide a biological rationale for clinical evaluation of BTK inhibition in AML patients.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Adult
- Agammaglobulinaemia Tyrosine Kinase
- Aged
- Aged, 80 and over
- Apoptosis/drug effects
- Cell Adhesion/drug effects
- Cell Proliferation/drug effects
- Enzyme Activation/drug effects
- Female
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Male
- Middle Aged
- NF-kappa B/metabolism
- Phosphorylation/physiology
- Piperidines
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Signal Transduction/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Stuart A Rushworth
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | | | | | | | | |
Collapse
|
1105
|
Kohlmann A, Bacher U, Schnittger S, Haferlach T. Perspective on how to approach molecular diagnostics in acute myeloid leukemia and myelodysplastic syndromes in the era of next-generation sequencing. Leuk Lymphoma 2014; 55:1725-34. [PMID: 24144312 DOI: 10.3109/10428194.2013.856427] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Molecular mutation information became essential for biological subclassification, risk stratification and therapeutic decisions in patients with acute myeloid leukemia (AML). In myelodysplastic syndromes (MDS), a broad spectrum of molecular biomarkers such as the spliceosome mutations has been identified in recent years. The currently established combination of various polymerase chain reaction (PCR) methods with capillary Sanger sequencing for mutation analysis in AML is time-consuming and labor-intensive. The constantly increasing spectrum of molecular mutations is a tremendous challenge for hematological laboratories. The introduction of high-throughput sequencing technology, which allows the massive parallel analysis of hundreds of thousands of alleles in the shortest time, provides new options for molecular mutation analyses and for follow-up diagnostics in myeloid neoplasms. In contrast to whole-genome or exome analyses, amplicon deep-sequencing focuses on distinct genomic loci and their mutation patterns and enables a comprehensive biomarker analysis in a multitude of patients per analysis. This review summarizes thus far established common molecular diagnostic strategies and intends to outline the perspective of distinct novel amplicon deep-sequencing panels for patients with AML and MDS. It is foreseeable that clearly defined algorithms for molecular investigations will revolutionize diagnosis in patients with AML and MDS in the near future.
Collapse
|
1106
|
Affiliation(s)
- Nicola E Potter
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM2 5NG, UK
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
1107
|
DNMT3A Arg882 mutation drives chronic myelomonocytic leukemia through disturbing gene expression/DNA methylation in hematopoietic cells. Proc Natl Acad Sci U S A 2014; 111:2620-5. [PMID: 24497509 DOI: 10.1073/pnas.1400150111] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The gene encoding DNA methyltransferase 3A (DNMT3A) is mutated in ∼20% of acute myeloid leukemia cases, with Arg882 (R882) as the hotspot. Here, we addressed the transformation ability of the DNMT3A-Arg882His (R882H) mutant by using a retroviral transduction and bone marrow transplantation (BMT) approach and found that the mutant gene can induce aberrant proliferation of hematopoietic stem/progenitor cells. At 12 mo post-BMT, all mice developed chronic myelomonocytic leukemia with thrombocytosis. RNA microarray analysis revealed abnormal expressions of some hematopoiesis-related genes, and the DNA methylation assay identified corresponding changes in methylation patterns in gene body regions. Moreover, DNMT3A-R882H increased the CDK1 protein level and enhanced cell-cycle activity, thereby contributing to leukemogenesis.
Collapse
|
1108
|
Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci U S A 2014; 111:2548-53. [PMID: 24550281 DOI: 10.1073/pnas.1324297111] [Citation(s) in RCA: 574] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer is widely characterized by the sequential acquisition of genetic lesions in a single lineage of cells. Our previous studies have shown that, in acute myeloid leukemia (AML), mutation acquisition occurs in functionally normal hematopoietic stem cells (HSCs). These preleukemic HSCs harbor some, but not all, of the mutations found in the leukemic cells. We report here the identification of patterns of mutation acquisition in human AML. Our findings support a model in which mutations in "landscaping" genes, involved in global chromatin changes such as DNA methylation, histone modification, and chromatin looping, occur early in the evolution of AML, whereas mutations in "proliferative" genes occur late. Additionally, we analyze the persistence of preleukemic mutations in patients in remission and find CD34+ progenitor cells and various mature cells that harbor preleukemic mutations. These findings indicate that preleukemic HSCs can survive induction chemotherapy, identifying these cells as a reservoir for the reevolution of relapsed disease. Finally, through the study of several cases of relapsed AML, we demonstrate various evolutionary patterns for the generation of relapsed disease and show that some of these patterns are consistent with involvement of preleukemic HSCs. These findings provide key insights into the monitoring of minimal residual disease and the identification of therapeutic targets in human AML.
Collapse
|
1109
|
Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia 2014; 28:1586-95. [PMID: 24487413 DOI: 10.1038/leu.2014.55] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 12/12/2022]
Abstract
To clarify the cooperative roles of recurrently identified mutations and to establish a more precise risk classification system in acute myeloid leukemia (AML), we comprehensively analyzed mutations in 51 genes, as well as cytogenetics and 11 chimeric transcripts, in 197 adult patients with de novo AML who were registered in the Japan Adult Leukemia Study Group AML201 study. We identified a total of 505 mutations in 44 genes, while only five genes, FLT3, NPM1, CEBPA, DNMT3A and KIT, were mutated in more than 10% of the patients. Although several cooperative and exclusive mutation patterns were observed, the accumulated mutation number was higher in cytogenetically normal AML and lower in AML with RUNX1-RUNX1T1 and CBFB-MYH11, indicating a strong potential of these translocations for the initiation of AML. Furthermore, we evaluated the prognostic impacts of each sole mutation and the combinations of mutations and/or cytogenetics, and demonstrated that AML patients could be clearly stratified into five risk groups for overall survival by including the mutation status of DNMT3A, MLL-PTD and TP53 genes in the risk classification system of the European LeukemiaNet. These results indicate that the prognosis of AML could be stratified by the major mutation status in combination with cytogenetics.
Collapse
|
1110
|
Nguyen L, Makarem M, Carles A, Moksa M, Kannan N, Pandoh P, Eirew P, Osako T, Kardel M, Cheung A, Kennedy W, Tse K, Zeng T, Zhao Y, Humphries R, Aparicio S, Eaves C, Hirst M. Clonal Analysis via Barcoding Reveals Diverse Growth and Differentiation of Transplanted Mouse and Human Mammary Stem Cells. Cell Stem Cell 2014; 14:253-63. [DOI: 10.1016/j.stem.2013.12.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/08/2013] [Accepted: 12/16/2013] [Indexed: 10/25/2022]
|
1111
|
Burrell RA, Swanton C. The evolution of the unstable cancer genome. Curr Opin Genet Dev 2014; 24:61-7. [PMID: 24657538 DOI: 10.1016/j.gde.2013.11.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/17/2013] [Indexed: 12/16/2022]
Abstract
Cancer next-generation sequencing and genomics studies published over the last five years have provided unprecedented insights into the forces shaping cancer genome evolution. In particular, these studies have revealed a high level of heterogeneity not only between different tumours, but also within individual tumours; the 'cancer genome' may evolve along several independent trajectories within a single tumour. There is an increasing appreciation of the importance of intratumour genetic heterogeneity in determining disease progression and clinical outcome in cancer medicine, and thus an increasing awareness of the need to understand the processes that both generate genetic diversity and shape genome evolution in human tumours.
Collapse
Affiliation(s)
- Rebecca A Burrell
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK; UCL Cancer Institute, Paul O'Gorman Building University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
1112
|
Janin M, Mylonas E, Saada V, Micol JB, Renneville A, Quivoron C, Koscielny S, Scourzic L, Forget S, Pautas C, Caillot D, Preudhomme C, Dombret H, Berthon C, Barouki R, Rabier D, Auger N, Griscelli F, Chachaty E, Leclercq E, Courtier MH, Bennaceur-Griscelli A, Solary E, Bernard OA, Penard-Lacronique V, Ottolenghi C, de Botton S. Serum 2-Hydroxyglutarate Production in IDH1- and IDH2-Mutated De Novo Acute Myeloid Leukemia: A Study by the Acute Leukemia French Association Group. J Clin Oncol 2014; 32:297-305. [DOI: 10.1200/jco.2013.50.2047] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Purpose Mutated isocitrate dehydrogenases (IDHs) 1 and 2 produce high levels of 2-hydroxyglutarate (2-HG). We investigated whether, in acute myeloid leukemia (AML), serum 2-HG would predict the presence of IDH1/2 mutations at diagnosis and provide a marker of minimal residual disease (MRD). Patients and Methods Serum samples from 82 patients at diagnosis of de novo AML (IDH1/2 mutated, n = 53) and 68 patients without AML were analyzed for total 2-HG and its ratio of D to L stereoisomers by mass spectrometry. We measured 2-HG levels and molecular markers of MRD (WT1 and NPM1) in serial samples of 36 patients with IDH1/2 mutations after induction therapy. Results In patients with AML with IDH1/2 mutations, 2-HG serum levels were significantly higher than in patients with IDH1/2 wild type (P < .001). Area under the receiver operating characteristic curve was 99%. The optimum diagnostic cutoff between IDH1/2 mutated and normal was 2 μmol/L (sensitivity, 100%; specificity, 79%). Quantification of the D/L stereoisomers increased specificity (100%; 95% CI, 83% to 100%) compared with total 2-HG (P = .031). In patients with IDH2 R172 mutations, 2-HG levels were higher relative to those with other IDH1/2 mutations (P < .05). During follow-up, serum 2-HG levels showed strong positive correlation with WT1 and NPM1 (P < .001). After induction therapy, total 2-HG serum levels < 2 μmol/L were associated with better overall (P = .008) and disease-free survival (P = .005). Conclusion Serum 2-HG is a predictor of the presence of IDH1/2 mutations and outcome in these patients. Discrimination between D/L stereoisomers improved specificity.
Collapse
Affiliation(s)
- Maxime Janin
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Elena Mylonas
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Véronique Saada
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Jean-Baptiste Micol
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Aline Renneville
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Cyril Quivoron
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Serge Koscielny
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Laurianne Scourzic
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Sébastien Forget
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Cécile Pautas
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Denis Caillot
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Claude Preudhomme
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Hervé Dombret
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Céline Berthon
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Robert Barouki
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Daniel Rabier
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Nathalie Auger
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Frank Griscelli
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Elisabeth Chachaty
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Edwige Leclercq
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Marie-Hélène Courtier
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Annelise Bennaceur-Griscelli
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Eric Solary
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Olivier Adrien Bernard
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Virginie Penard-Lacronique
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Chris Ottolenghi
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Stéphane de Botton
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| |
Collapse
|
1113
|
White BS, DiPersio JF. Genomic tools in acute myeloid leukemia: From the bench to the bedside. Cancer 2014; 120:1134-44. [PMID: 24474533 DOI: 10.1002/cncr.28552] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 11/14/2013] [Indexed: 12/28/2022]
Abstract
Since its use in the initial characterization of an acute myeloid leukemia (AML) genome, next-generation sequencing (NGS) has continued to molecularly refine the disease. Here, the authors review the spectrum of NGS applications that have subsequently delineated the prognostic significance and biologic consequences of these mutations. Furthermore, the role of this technology in providing a high-resolution glimpse of AML clonal heterogeneity, which may inform future choice of targeted therapy, is discussed. Although obstacles remain in applying these techniques clinically, they have already had an impact on patient care.
Collapse
Affiliation(s)
- Brian S White
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri; The Genome Institute, Washington University, St. Louis, Missouri
| | | |
Collapse
|
1114
|
Whole-genome sequencing identifies genomic heterogeneity at a nucleotide and chromosomal level in bladder cancer. Proc Natl Acad Sci U S A 2014; 111:E672-81. [PMID: 24469795 DOI: 10.1073/pnas.1313580111] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Using complete genome analysis, we sequenced five bladder tumors accrued from patients with muscle-invasive transitional cell carcinoma of the urinary bladder (TCC-UB) and identified a spectrum of genomic aberrations. In three tumors, complex genotype changes were noted. All three had tumor protein p53 mutations and a relatively large number of single-nucleotide variants (SNVs; average of 11.2 per megabase), structural variants (SVs; average of 46), or both. This group was best characterized by chromothripsis and the presence of subclonal populations of neoplastic cells or intratumoral mutational heterogeneity. Here, we provide evidence that the process of chromothripsis in TCC-UB is mediated by nonhomologous end-joining using kilobase, rather than megabase, fragments of DNA, which we refer to as "stitchers," to repair this process. We postulate that a potential unifying theme among tumors with the more complex genotype group is a defective replication-licensing complex. A second group (two bladder tumors) had no chromothripsis, and a simpler genotype, WT tumor protein p53, had relatively few SNVs (average of 5.9 per megabase) and only a single SV. There was no evidence of a subclonal population of neoplastic cells. In this group, we used a preclinical model of bladder carcinoma cell lines to study a unique SV (translocation and amplification) of the gene glutamate receptor ionotropic N-methyl D-aspertate as a potential new therapeutic target in bladder cancer.
Collapse
|
1115
|
Abstract
Genomes are inherently unstable because of the need for DNA sequence variation as a substrate for evolution through natural selection. However, most multicellular organisms have postmitotic tissues, with limited opportunity for selective removal of cells harboring persistent damage and deleterious mutations, which can therefore contribute to functional decline, disease, and death. Key in this process is the role of genome maintenance, the network of protein products that repair DNA damage and signal DNA damage response pathways. Genome maintenance is beneficial early in life by swiftly eliminating DNA damage or damaged cells, facilitating rapid cell proliferation. However, at later ages accumulation of unrepaired damage and mutations, as well as ongoing cell depletion, promotes cancer, atrophy, and other deleterious effects associated with aging. As such, genome maintenance and its phenotypic sequelae provide yet another example of antagonistic pleiotropy in aging and longevity.
Collapse
Affiliation(s)
- Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
1116
|
Abstract
The emerging paradigm of Precision Oncology 3.0 uses panomics and sophisticated methods of statistical reverse engineering to hypothesize the putative networks that drive a given patient's tumour, and to attack these drivers with combinations of targeted therapies. Here, we review a paradigm termed Rapid Learning Precision Oncology wherein every treatment event is considered as a probe that simultaneously treats the patient and provides an opportunity to validate and refine the models on which the treatment decisions are based. Implementation of Rapid Learning Precision Oncology requires overcoming a host of challenges that include developing analytical tools, capturing the information from each patient encounter and rapidly extrapolating it to other patients, coordinating many patient encounters to efficiently search for effective treatments, and overcoming economic, social and structural impediments, such as obtaining access to, and reimbursement for, investigational drugs.
Collapse
|
1117
|
Abstract
For a decade, the technologies behind DNA sequencing have improved rapidly in cost reduction and speed. Sequencing in large populations of cancer patients is leading to dramatic advances in our understanding of the cancer genome. The wide variety of cancer types sequenced and analyzed using these technologies has revealed many novel fundamental genetic mechanisms driving cancer initiation, progression, and maintenance. We have deepened our understanding of the signaling pathways, demonstrating disruption in epigenetic regulation and destabilization of the splicing machinery. The molecular mechanisms of resistance to targeted therapies are being elucidated for the first time. The translation of genome-scale variation into clinically actionable information is still in its infancy; nevertheless, insights from sequencing studies have led to the discovery of a variety of novel diagnostic biomarkers and therapeutic targets. Here, we review recent advances in cancer genomics and discuss what the new findings have taught us about cancer biology and, more importantly, how these new findings guide more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Linghua Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030
| | - David A. Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
1118
|
Ablain J, Rice K, Soilihi H, de Reynies A, Minucci S, de Thé H. Activation of a promyelocytic leukemia-tumor protein 53 axis underlies acute promyelocytic leukemia cure. Nat Med 2014; 20:167-74. [PMID: 24412926 DOI: 10.1038/nm.3441] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/04/2013] [Indexed: 12/18/2022]
Abstract
Acute promyelocytic leukemia (APL) is driven by the promyelocytic leukemia (PML)-retinoic acid receptor-α (PML-RARA) fusion protein, which interferes with nuclear receptor signaling and PML nuclear body (NB) assembly. APL is the only malignancy definitively cured by targeted therapies: retinoic acid (RA) and/or arsenic trioxide, which both trigger PML-RARA degradation through nonoverlapping pathways. Yet, the cellular and molecular determinants of treatment efficacy remain disputed. We demonstrate that a functional Pml-transformation-related protein 53 (Trp53) axis is required to eradicate leukemia-initiating cells in a mouse model of APL. Upon RA-induced PML-RARA degradation, normal Pml elicits NB reformation and induces a Trp53 response exhibiting features of senescence but not apoptosis, ultimately abrogating APL-initiating activity. Apart from triggering PML-RARA degradation, arsenic trioxide also targets normal PML to enhance NB reformation, which may explain its clinical potency, alone or with RA. This Pml-Trp53 checkpoint initiated by therapy-triggered NB restoration is specific for PML-RARA-driven APL, but not the RA-resistant promyelocytic leukemia zinc finger (PLZF)-RARA variant. Yet, as NB biogenesis is druggable, it could be therapeutically exploited in non-APL malignancies.
Collapse
Affiliation(s)
- Julien Ablain
- 1] Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France. [2] INSERM UMR 944, Equipe Labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France. [3] CNRS UMR 7212, Hôpital St. Louis, Paris, France. [4]
| | - Kim Rice
- 1] Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France. [2] INSERM UMR 944, Equipe Labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France. [3] CNRS UMR 7212, Hôpital St. Louis, Paris, France
| | - Hassane Soilihi
- 1] Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France. [2] INSERM UMR 944, Equipe Labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France. [3] CNRS UMR 7212, Hôpital St. Louis, Paris, France
| | - Aurélien de Reynies
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale contre le Cancer, Paris, France
| | - Saverio Minucci
- 1] Department of Experimental Oncology, European Institute of Oncology, Milan, Italy. [2] Department of Biosciences, University of Milan, Milan, Italy
| | - Hugues de Thé
- 1] Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France. [2] INSERM UMR 944, Equipe Labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France. [3] CNRS UMR 7212, Hôpital St. Louis, Paris, France. [4] Assistance Publique Hôpitaux de Paris, Service de Biochimie, Hôpital St. Louis, Paris, France
| |
Collapse
|
1119
|
Genetics of follicular lymphoma transformation. Cell Rep 2014; 6:130-40. [PMID: 24388756 DOI: 10.1016/j.celrep.2013.12.027] [Citation(s) in RCA: 443] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 12/30/2022] Open
Abstract
Follicular lymphoma (FL) is an indolent disease, but 30%-40% of cases undergo histologic transformation to an aggressive malignancy, typically represented by diffuse large B cell lymphoma (DLBCL). The pathogenesis of this process remains largely unknown. Using whole-exome sequencing and copy-number analysis, we show here that the dominant clone of FL and transformed FL (tFL) arise by divergent evolution from a common mutated precursor through the acquisition of distinct genetic events. Mutations in epigenetic modifiers and antiapoptotic genes are introduced early in the common precursor, whereas tFL is specifically associated with alterations deregulating cell-cycle progression and DNA damage responses (CDKN2A/B, MYC, and TP53) as well as aberrant somatic hypermutation. The genomic profile of tFL shares similarities with that of germinal center B cell-type de novo DLBCL but also displays unique combinations of altered genes with diagnostic and therapeutic implications.
Collapse
|
1120
|
Abstract
The inherited bone marrow failure (BMF) syndromes are a rare and diverse group of genetic disorders that ultimately result in the loss of blood production. The molecular defects underlying many of these conditions have been elucidated, and great progress has been made toward understanding the normal function of these gene products. This review will focus on perhaps the most well-known and genetically heterogeneous BMF syndrome: Fanconi anemia. More specifically, this account will review the current state of our knowledge on why the bone marrow fails in this illness and what this might tell us about the maintenance of bone marrow function and hematopoiesis.
Collapse
Affiliation(s)
- Juan I Garaycoechea
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | | |
Collapse
|
1121
|
Marsman J, O'Neill AC, Kao BRY, Rhodes JM, Meier M, Antony J, Mönnich M, Horsfield JA. Cohesin and CTCF differentially regulate spatiotemporal runx1 expression during zebrafish development. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1839:50-61. [PMID: 24321385 DOI: 10.1016/j.bbagrm.2013.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/19/2013] [Accepted: 11/25/2013] [Indexed: 11/23/2022]
Abstract
Runx1 is a transcription factor essential for definitive hematopoiesis. In all vertebrates, the Runx1 gene is transcribed from two promoters: a proximal promoter (P2), and a distal promoter (P1). We previously found that runx1 expression in a specific hematopoietic cell population in zebrafish embryos depends on cohesin. Here we show that zebrafish runx1 is directly bound by cohesin and CCCTC binding factor (CTCF) at the P1 and P2 promoters, and within the intron between P1 and P2. Cohesin initiates expression of runx1 in the posterior lateral mesoderm and influences promoter use, while CTCF represses its expression in the newly emerging cells of the tail bud. The intronic binding sites for cohesin and CTCF coincide with histone modifications that confer enhancer-like properties, and two of the cohesin/CTCF sites behaved as insulators in an in vivo assay. The identified cohesin and CTCF binding sites are likely to be cis-regulatory elements (CREs) for runx1 since they also recruit RNA polymerase II (RNAPII). CTCF depletion excluded RNAPII from two intronic CREs but not the promoters of runx1. We propose that cohesin and CTCF have distinct functions in the regulation of runx1 during zebrafish embryogenesis, and that these regulatory functions are likely to involve runx1 intronic CREs. Cohesin (but not CTCF) depletion enhanced RUNX1 expression in a human leukemia cell line, suggesting conservation of RUNX1 regulation through evolution.
Collapse
Affiliation(s)
- Judith Marsman
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Adam C O'Neill
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Betty Rui-Yun Kao
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Jenny M Rhodes
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Michael Meier
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Jisha Antony
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Maren Mönnich
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Julia A Horsfield
- Department of Pathology, Dunedin School of Medicine, The University of Otago, P.O. Box 913, Dunedin, New Zealand.
| |
Collapse
|
1122
|
Bolli N, Avet-Loiseau H, Wedge DC, Van Loo P, Alexandrov LB, Martincorena I, Dawson KJ, Iorio F, Nik-Zainal S, Bignell GR, Hinton JW, Li Y, Tubio JM, McLaren S, O' Meara S, Butler AP, Teague JW, Mudie L, Anderson E, Rashid N, Tai YT, Shammas MA, Sperling AS, Fulciniti M, Richardson PG, Parmigiani G, Magrangeas F, Minvielle S, Moreau P, Attal M, Facon T, Futreal PA, Anderson KC, Campbell PJ, Munshi NC. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun 2014; 5:2997. [PMID: 24429703 PMCID: PMC3905727 DOI: 10.1038/ncomms3997] [Citation(s) in RCA: 704] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 11/25/2013] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma is an incurable plasma cell malignancy with a complex and incompletely understood molecular pathogenesis. Here we use whole-exome sequencing, copy-number profiling and cytogenetics to analyse 84 myeloma samples. Most cases have a complex subclonal structure and show clusters of subclonal variants, including subclonal driver mutations. Serial sampling reveals diverse patterns of clonal evolution, including linear evolution, differential clonal response and branching evolution. Diverse processes contribute to the mutational repertoire, including kataegis and somatic hypermutation, and their relative contribution changes over time. We find heterogeneity of mutational spectrum across samples, with few recurrent genes. We identify new candidate genes, including truncations of SP140, LTB, ROBO1 and clustered missense mutations in EGR1. The myeloma genome is heterogeneous across the cohort, and exhibits diversity in clonal admixture and in dynamics of evolution, which may impact prognostic stratification, therapeutic approaches and assessment of disease response to treatment.
Collapse
Affiliation(s)
- Niccolo Bolli
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Department of Haematology, University of Cambridge, CIMR, Cambridge CB2 0XY, UK
| | - Hervé Avet-Loiseau
- Unité de Génomique du Myélome, CHU Rangueil, Toulouse 31059, France
- CRCT, INSERM U1037, Toulouse 31400, France
| | - David C. Wedge
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Peter Van Loo
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Department of Human Genetics, VIB and University of Leuven, Leuven 3000, Belgium
| | | | - Inigo Martincorena
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Kevin J. Dawson
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Francesco Iorio
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- European Molecular Biology Laboratory—European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Serena Nik-Zainal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Department of Medical Genetics, Addenbrooke’s Hospital NHS Trust, Cambridge CB2 0QQ, UK
| | - Graham R. Bignell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Jonathan W. Hinton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Yilong Li
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Jose M.C. Tubio
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Stuart McLaren
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Sarah O' Meara
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Adam P. Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Jon W. Teague
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Laura Mudie
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Elizabeth Anderson
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Naim Rashid
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yu-Tzu Tai
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Masood A. Shammas
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Boston Veterans Administration Healthcare System, West Roxbury, Massachusetts 02132, USA
| | - Adam S. Sperling
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mariateresa Fulciniti
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Paul G. Richardson
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Giovanni Parmigiani
- Dana–Farber Cancer Institute and Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Florence Magrangeas
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm-6299 CNRS-University of Nantes, IRS-UN, Nantes 4407, France
- UMGC, University Hospital, Nantes 44093, France
| | - Stephane Minvielle
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm-6299 CNRS-University of Nantes, IRS-UN, Nantes 4407, France
- UMGC, University Hospital, Nantes 44093, France
| | - Philippe Moreau
- Department of Hematology, University Hospital, Nantes 44093, France
| | - Michel Attal
- Department of Hematology, University Hospital and CRCT, INSERM U1037, Toulouse 31400, France
| | - Thierry Facon
- Department of Hematology, University Hospital, Lille 59045, France
| | - P Andrew Futreal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Present address: MD Anderson Cancer Center, Houston, Texas, USA
| | - Kenneth C. Anderson
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Peter J. Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Department of Haematology, University of Cambridge, CIMR, Cambridge CB2 0XY, UK
| | - Nikhil C. Munshi
- Lebow Institute of Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana–Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Boston Veterans Administration Healthcare System, West Roxbury, Massachusetts 02132, USA
| |
Collapse
|
1123
|
Pellagatti A, Fernandez-Mercado M, Di Genua C, Larrayoz MJ, Killick S, Dolatshad H, Burns A, Calasanz MJ, Schuh A, Boultwood J. Whole-exome sequencing in del(5q) myelodysplastic syndromes in transformation to acute myeloid leukemia. Leukemia 2013; 28:1148-51. [PMID: 24365791 DOI: 10.1038/leu.2013.381] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- A Pellagatti
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - M Fernandez-Mercado
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - C Di Genua
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - M J Larrayoz
- Department of Genetics, University of Navarra, Pamplona, Spain
| | - S Killick
- Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK
| | - H Dolatshad
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - A Burns
- NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - M J Calasanz
- Department of Genetics, University of Navarra, Pamplona, Spain
| | - A Schuh
- NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - J Boultwood
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
1124
|
Elias HK, Schinke C, Bhattacharyya S, Will B, Verma A, Steidl U. Stem cell origin of myelodysplastic syndromes. Oncogene 2013; 33:5139-50. [PMID: 24336326 DOI: 10.1038/onc.2013.520] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/17/2013] [Accepted: 10/17/2013] [Indexed: 01/18/2023]
Abstract
Myelodysplastic syndromes (MDS) are common hematologic disorders that are characterized by decreased blood counts due to ineffective hematopoiesis. MDS is considered a 'preleukemic' disorder linked to a significantly elevated risk of developing an overt acute leukemia. Cytopenias can be observed in all three myeloid lineages suggesting the involvement of multipotent, immature hematopoietic cells in the pathophysiology of this disease. Recent studies using murine models of MDS as well as primary patient-derived bone marrow samples have provided direct evidence that the most immature, self-renewing hematopoietic stem cells (HSC), as well as lineage-committed progenitor cells, are critically altered in patients with MDS. Besides significant changes in the number and distribution of stem as well as immature progenitor cells, genetic and epigenetic aberrations have been identified, which confer functional changes to these aberrant stem cells, impairing their ability to proliferate and differentiate. Most importantly, aberrant stem cells can persist and further expand after treatment, even upon transient achievement of clinical complete remission, pointing to a critical role of these cells in disease relapse. Ongoing preclinical and clinical studies are particularly focusing on the precise molecular and functional characterization of aberrant MDS stem cells in response to therapy, with the goal to develop stem cell-targeted strategies for therapy and disease monitoring that will allow for achievement of longer-lasting remissions in MDS.
Collapse
Affiliation(s)
- H K Elias
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - C Schinke
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - S Bhattacharyya
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - B Will
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - A Verma
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - U Steidl
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| |
Collapse
|
1125
|
Mutations in the cohesin complex in acute myeloid leukemia: clinical and prognostic implications. Blood 2013; 123:914-20. [PMID: 24335498 DOI: 10.1182/blood-2013-07-518746] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mutations in the cohesin complex are novel, genetic lesions in acute myeloid leukemia (AML) that are not well characterized. In this study, we analyzed the frequency, clinical, and prognostic implications of mutations in STAG1, STAG2, SMC1A, SMC3, and RAD21, all members of the cohesin complex, in a cohort of 389 uniformly treated AML patients by next generation sequencing. We identified a total of 23 patients (5.9%) with somatic mutations in 1 of the cohesin genes. All gene mutations were mutually exclusive, and STAG1 (1.8%), STAG2 (1.3%), and SMC3 (1.3%) were most frequently mutated. Patients with any cohesin complex mutation had lower BAALC expression levels. We found a strong association between mutations affecting the cohesin complex and NPM1. Mutated allele frequencies were similar between NPM1 and cohesin gene mutations. Overall survival (OS), relapse-free survival (RFS), and complete remission rates (CR) were not influenced by the presence of cohesin mutations (OS: hazard ratio [HR] 0.98; 95% confidence interval [CI], 0.56-1.72 [P = .94]; RFS: HR 0.7; 95% CI, 0.36-1.38 [P = .3]; CR: mutated 83% vs wild-type 76% [P = .45]). The cohesin complex presents a novel pathway affected by recurrent mutations in AML. This study is registered at www.clinicaltrials.gov as #NCT00209833.
Collapse
|
1126
|
Abstract
Two lingering problems regarding transplantation in older adults have been how to select patients appropriately and whether to use older sibling donors. Allogeneic hematopoietic cell transplantation (HCT) of older patients may result in long-term survival due to GVL, but the data remain observational and mostly restricted to those 50 to 69 years of age. Patients with excellent performance status and low comorbidity have the best long-term survival after HCT. Novel measures of health status such as self-report or performance-based functional measures allow “staging the age” and may inform candidacy for less robust patients. Older matched sibling donors should be preferred over matched unrelated donors (MUDs) because outcomes are equivalent to superior for matched sibling donors compared with MUD. However, MUDs also achieve acceptable outcomes and long-term disease control. An alternative donor can be considered based on institutional protocols and expertise. Very limited information is available in patients or related donors 70 years of age and older. Future efforts to more completely characterize patient health status before transplantation will allow better application of HCT in older adults.
Collapse
|
1127
|
Berglund EC, Lindqvist CM, Hayat S, Övernäs E, Henriksson N, Nordlund J, Wahlberg P, Forestier E, Lönnerholm G, Syvänen AC. Accurate detection of subclonal single nucleotide variants in whole genome amplified and pooled cancer samples using HaloPlex target enrichment. BMC Genomics 2013; 14:856. [PMID: 24314227 PMCID: PMC4046713 DOI: 10.1186/1471-2164-14-856] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 11/25/2013] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Target enrichment and resequencing is a widely used approach for identification of cancer genes and genetic variants associated with diseases. Although cost effective compared to whole genome sequencing, analysis of many samples constitutes a significant cost, which could be reduced by pooling samples before capture. Another limitation to the number of cancer samples that can be analyzed is often the amount of available tumor DNA. We evaluated the performance of whole genome amplified DNA and the power to detect subclonal somatic single nucleotide variants in non-indexed pools of cancer samples using the HaloPlex technology for target enrichment and next generation sequencing. RESULTS We captured a set of 1528 putative somatic single nucleotide variants and germline SNPs, which were identified by whole genome sequencing, with the HaloPlex technology and sequenced to a depth of 792-1752. We found that the allele fractions of the analyzed variants are well preserved during whole genome amplification and that capture specificity or variant calling is not affected. We detected a large majority of the known single nucleotide variants present uniquely in one sample with allele fractions as low as 0.1 in non-indexed pools of up to ten samples. We also identified and experimentally validated six novel variants in the samples included in the pools. CONCLUSION Our work demonstrates that whole genome amplified DNA can be used for target enrichment equally well as genomic DNA and that accurate variant detection is possible in non-indexed pools of cancer samples. These findings show that analysis of a large number of samples is feasible at low cost, even when only small amounts of DNA is available, and thereby significantly increases the chances of indentifying recurrent mutations in cancer samples.
Collapse
Affiliation(s)
- Eva C Berglund
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1128
|
Solomon DA, Kim JS, Bondaruk J, Shariat SF, Wang ZF, Elkahloun AG, Ozawa T, Gerard J, Zhuang D, Zhang S, Navai N, Siefker-Radtker A, Phillips JJ, Robinson BD, Rubin MA, Volkmer B, Hautmann R, Küfer R, Hogendoorn PCW, Netto G, Theodorescu D, James CD, Czerniak B, Miettinen M, Waldman T. Frequent truncating mutations of STAG2 in bladder cancer. Nat Genet 2013; 45:1428-30. [PMID: 24121789 PMCID: PMC3875130 DOI: 10.1038/ng.2800] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 09/18/2013] [Indexed: 01/22/2023]
Abstract
Here we report the discovery of truncating mutations of the gene encoding the cohesin subunit STAG2, which regulates sister chromatid cohesion and segregation, in 36% of papillary non-invasive urothelial carcinomas and 16% of invasive urothelial carcinomas of the bladder. Our studies suggest that STAG2 has a role in controlling chromosome number but not the proliferation of bladder cancer cells. These findings identify STAG2 as one of the most commonly mutated genes in bladder cancer.
Collapse
Affiliation(s)
- David A. Solomon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC, 20057, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jung-Sik Kim
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC, 20057, USA
| | - Jolanta Bondaruk
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shahrokh F. Shariat
- Department of Urology, Weill Cornell College of Medicine, New York, NY, 10065, USA
- Division of Medical Oncology, Weill Cornell College of Medicine, New York, NY, 10065, USA
| | - Zeng-Feng Wang
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Abdel G. Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Tomoko Ozawa
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Julia Gerard
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC, 20057, USA
| | - DaZhong Zhuang
- Department of Urology, Weill Cornell College of Medicine, New York, NY, 10065, USA
| | - Shizhen Zhang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Neema Navai
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Arleen Siefker-Radtker
- Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joanna J. Phillips
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Brian D. Robinson
- Department of Urology, Weill Cornell College of Medicine, New York, NY, 10065, USA
- Department of Pathology, Weill Cornell College of Medicine, New York, NY, 10065, USA
| | - Mark A. Rubin
- Department of Urology, Weill Cornell College of Medicine, New York, NY, 10065, USA
- Department of Pathology, Weill Cornell College of Medicine, New York, NY, 10065, USA
| | - Björn Volkmer
- Department of Urology, Hospital Kassel, Kassel, Germany
| | | | - Rainer Küfer
- Department of Urology, Hospital Am Eichert, Göppingen, Germany
| | | | - George Netto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Dan Theodorescu
- Department of Surgery, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - C. David James
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Bogdan Czerniak
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Todd Waldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC, 20057, USA
| |
Collapse
|
1129
|
|
1130
|
Transcript level of nucleostemin in newly diagnosed acute myeloid leukemia patients. Leuk Res 2013; 37:1636-41. [DOI: 10.1016/j.leukres.2013.09.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 09/13/2013] [Accepted: 09/20/2013] [Indexed: 12/30/2022]
|
1131
|
Abstract
A longstanding endeavor to define the genetic lesions that drive myeloid malignances has stimulated a period of remarkable discovery. Enabled by technological advances that have sharply decreased the cost of DNA sequencing, the full compendium of common, recurrent somatic mutations in the coding genome of myeloid malignancies is nearly complete. As the focus of genetic discovery shifts to the noncoding genome, renewed attention is being applied to the clinical and biological implications of recent genomic advances. Although the potential for this newfound knowledge to influence the care of patients has not yet been realized, broad genetic surveys of patient samples are now being used to improve the accuracy of disease diagnosis, define a molecular taxonomy of myeloid malignancies, refine prognostic and predictive models, and identify novel therapeutic strategies. Here, we will review recent advances in the genetics of myeloid malignancies and discuss their potential impact on clinical practice.
Collapse
Affiliation(s)
- R Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; and
| | | |
Collapse
|
1132
|
Mutational landscape and significance across 12 major cancer types. Nature 2013; 502:333-339. [PMID: 24132290 PMCID: PMC3927368 DOI: 10.1038/nature12634] [Citation(s) in RCA: 3333] [Impact Index Per Article: 277.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/13/2013] [Indexed: 02/06/2023]
Abstract
The Cancer Genome Atlas (TCGA) has used the latest sequencing and analysis methods to identify somatic variants across thousands of tumours. Here we present data and analytical results for point mutations and small insertions/deletions from 3,281 tumours across 12 tumour types as part of the TCGA Pan-Cancer effort. We illustrate the distributions of mutation frequencies, types and contexts across tumour types, and establish their links to tissues of origin, environmental/carcinogen influences, and DNA repair defects. Using the integrated data sets, we identified 127 significantly mutated genes from well-known (for example, mitogen-activated protein kinase, phosphatidylinositol-3-OH kinase, Wnt/β-catenin and receptor tyrosine kinase signalling pathways, and cell cycle control) and emerging (for example, histone, histone modification, splicing, metabolism and proteolysis) cellular processes in cancer. The average number of mutations in these significantly mutated genes varies across tumour types; most tumours have two to six, indicating that the number of driver mutations required during oncogenesis is relatively small. Mutations in transcriptional factors/regulators show tissue specificity, whereas histone modifiers are often mutated across several cancer types. Clinical association analysis identifies genes having a significant effect on survival, and investigations of mutations with respect to clonal/subclonal architecture delineate their temporal orders during tumorigenesis. Taken together, these results lay the groundwork for developing new diagnostics and individualizing cancer treatment. As part of The Cancer Genome Atlas Pan-Cancer effort, data analysis for point mutations and small indels from 3,281 tumours and 12 tumour types is presented; among the findings are 127 significantly mutated genes from cellular processes with both established and emerging links in cancer, and an indication that the number of driver mutations required for oncogenesis is relatively small. As part of The Cancer Genome Atlas Pan-Cancer project, these authors present data analysis for point mutations and small indels from more than 3,000 tumours representing 12 tumour types. Among the findings are 127 significantly mutated genes from cellular processes with both established and emerging links to cancer, and an indication that the number of driver mutations required for oncogenesis is relatively small. Additional analyses also identify genes with significant impact on survival and a likely temporal order of mutational events during tumorigenesis.
Collapse
|
1133
|
The genome of polymorphonuclear neutrophils maintains normal coding sequences. PLoS One 2013; 8:e78685. [PMID: 24250807 PMCID: PMC3826757 DOI: 10.1371/journal.pone.0078685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/13/2013] [Indexed: 11/19/2022] Open
Abstract
Genetic studies often use genomic DNA from whole blood cells, of which the majority are the polymorphonuclear myeloid cells. Those cells undergo dramatic change of nuclear morphology following cellular differentiation. It remains elusive if the nuclear morphological change accompanies sequence alternations from the intact genome. If such event exists, it will cause a serious problem in using such type of genomic DNA for genetic study as the sequences will not represent the intact genome in the host individuals. Using exome sequencing, we compared the coding regions between neutrophil, which is the major type of polymorphonuclear cells, and CD4+ T cell, which has an intact genome, from the same individual. The results show that exon sequences between the two cell types are essentially the same. The minor differences represented by the missed exons and base changes between the two cell types were validated to be mainly caused by experimental errors. Our study concludes that genomic DNA from whole blood cells can be safely used for genetic studies.
Collapse
|
1134
|
Chanock SJ. Introduction: sifting through the characterization of hematologic malignancies. Semin Hematol 2013; 50:284-5. [PMID: 24246695 DOI: 10.1053/j.seminhematol.2013.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Stephen J Chanock
- Laboratory of Translational Genomics Division of Cancer Epidemiology and Genetics National Cancer Institute Bethesda, MD
| |
Collapse
|
1135
|
Reikvam H, Øyan AM, Kalland KH, Hovland R, Hatfield KJ, Bruserud Ø. Differences in proliferative capacity of primary human acute myelogenous leukaemia cells are associated with altered gene expression profiles and can be used for subclassification of patients. Cell Prolif 2013; 46:554-62. [PMID: 24073609 DOI: 10.1111/cpr.12057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/12/2013] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Proliferative capacity of acute myelogenous leukaemia (AML) blasts is important for leukaemogenesis, and we have investigated whether proliferative capacity of primary human AML cells could be used for subclassification of patients. MATERIALS AND METHODS In vitro proliferative capacity of AML cells derived from two independent groups was investigated. Cells were cultured under highly standardized conditions and proliferation assayed by (3) H-thymidine incorporation after seven days culture. Patients were subclassified by clustering models, and gene expression profile was examined by microarray analyses. RESULTS Based on proliferative capacity of the AML cells, three different patient clusters were identified: (i) autocrine proliferation that was increased by exogenous cytokines; (ii) detectable proliferation only in presence of exogenous cytokines; and (iii) low or undetectable proliferation even in presence of exogenous cytokines. Patients with highest proliferative capacity cells had no favourable prognostic impact by NPM-1 mutation. Analysis of gene expression profiles showed that the most proliferative cells generally had altered expression of genes involved in regulation of transcription/RNA functions, whereas patients with high proliferative capacity and internal tandem duplications (ITDs) in the FLT3 cytokine receptor gene had altered expression of several molecules involved in cytoplasmic signal transduction. CONCLUSIONS In vitro proliferative capacity of primary human AML cells was considerably variable between patients and could be used to identify biologically distinct patient subsets.
Collapse
Affiliation(s)
- H Reikvam
- Division for Hematology, Institute of Clinical Science, University of Bergen, Bergen, Norway; Division for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway; Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | | | | | | | | | | |
Collapse
|
1136
|
Carretero M, Ruiz-Torres M, Rodríguez-Corsino M, Barthelemy I, Losada A. Pds5B is required for cohesion establishment and Aurora B accumulation at centromeres. EMBO J 2013; 32:2938-49. [PMID: 24141881 PMCID: PMC3831313 DOI: 10.1038/emboj.2013.230] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 10/02/2013] [Indexed: 12/23/2022] Open
Abstract
Cohesin mediates sister chromatid cohesion and contributes to the organization of interphase chromatin through DNA looping. In vertebrate somatic cells, cohesin consists of Smc1, Smc3, Rad21, and either SA1 or SA2. Three additional factors Pds5, Wapl, and Sororin bind to cohesin and modulate its dynamic association with chromatin. There are two Pds5 proteins in vertebrates, Pds5A and Pds5B, but their functional specificity remains unclear. Here, we demonstrate that Pds5 proteins are essential for cohesion establishment by allowing Smc3 acetylation by the cohesin acetyl transferases (CoATs) Esco1/2 and binding of Sororin. While both proteins contribute to telomere and arm cohesion, Pds5B is specifically required for centromeric cohesion. Furthermore, reduced accumulation of Aurora B at the inner centromere region in cells lacking Pds5B impairs its error correction function, promoting chromosome mis-segregation and aneuploidy. Our work supports a model in which the composition and function of cohesin complexes differs between different chromosomal regions.
Collapse
Affiliation(s)
- María Carretero
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Miguel Ruiz-Torres
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Miriam Rodríguez-Corsino
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Isabel Barthelemy
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Losada
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
1137
|
Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, Schnittger S, Sanada M, Kon A, Alpermann T, Yoshida K, Roller A, Nadarajah N, Shiraishi Y, Shiozawa Y, Chiba K, Tanaka H, Koeffler HP, Klein HU, Dugas M, Aburatani H, Kohlmann A, Miyano S, Haferlach C, Kern W, Ogawa S. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 2013; 28:241-7. [PMID: 24220272 PMCID: PMC3918868 DOI: 10.1038/leu.2013.336] [Citation(s) in RCA: 1223] [Impact Index Per Article: 101.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 10/21/2013] [Accepted: 10/30/2013] [Indexed: 02/07/2023]
Abstract
High-throughput DNA sequencing significantly contributed to diagnosis and prognostication in patients with myelodysplastic syndromes (MDS). We determined the biological and prognostic significance of genetic aberrations in MDS. In total, 944 patients with various MDS subtypes were screened for known/putative mutations/deletions in 104 genes using targeted deep sequencing and array-based genomic hybridization. In total, 845/944 patients (89.5%) harbored at least one mutation (median, 3 per patient; range, 0-12). Forty-seven genes were significantly mutated with TET2, SF3B1, ASXL1, SRSF2, DNMT3A, and RUNX1 mutated in >10% of cases. Many mutations were associated with higher risk groups and/or blast elevation. Survival was investigated in 875 patients. By univariate analysis, 25/48 genes (resulting from 47 genes tested significantly plus PRPF8) affected survival (P<0.05). The status of 14 genes combined with conventional factors revealed a novel prognostic model ('Model-1') separating patients into four risk groups ('low', 'intermediate', 'high', 'very high risk') with 3-year survival of 95.2, 69.3, 32.8, and 5.3% (P<0.001). Subsequently, a 'gene-only model' ('Model-2') was constructed based on 14 genes also yielding four significant risk groups (P<0.001). Both models were reproducible in the validation cohort (n=175 patients; P<0.001 each). Thus, large-scale genetic and molecular profiling of multiple target genes is invaluable for subclassification and prognostication in MDS patients.
Collapse
Affiliation(s)
- T Haferlach
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - Y Nagata
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - V Grossmann
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - Y Okuno
- Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - U Bacher
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - G Nagae
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - S Schnittger
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - M Sanada
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - A Kon
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - T Alpermann
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - K Yoshida
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - A Roller
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - N Nadarajah
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - Y Shiraishi
- Laboratory of DNA Information Analysis, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Y Shiozawa
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - K Chiba
- Laboratory of DNA Information Analysis, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - H Tanaka
- Laboratory of Sequence Data Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - H P Koeffler
- 1] Department of Hematology/Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA [2] Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - H-U Klein
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - M Dugas
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - H Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - A Kohlmann
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - S Miyano
- 1] Laboratory of Sequence Data Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan [2] Laboratory of DNA Information Analysis, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - C Haferlach
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - W Kern
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - S Ogawa
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
1138
|
The causes and consequences of genetic heterogeneity in cancer evolution. Nature 2013; 501:338-45. [PMID: 24048066 DOI: 10.1038/nature12625] [Citation(s) in RCA: 1639] [Impact Index Per Article: 136.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/13/2013] [Indexed: 02/06/2023]
Abstract
Recent studies have revealed extensive genetic diversity both between and within tumours. This heterogeneity affects key cancer pathways, driving phenotypic variation, and poses a significant challenge to personalized cancer medicine. A major cause of genetic heterogeneity in cancer is genomic instability. This instability leads to an increased mutation rate and can shape the evolution of the cancer genome through a plethora of mechanisms. By understanding these mechanisms we can gain insight into the common pathways of tumour evolution that could support the development of future therapeutic strategies.
Collapse
|
1139
|
Spencer DH, Tyagi M, Vallania F, Bredemeyer AJ, Pfeifer JD, Mitra RD, Duncavage EJ. Performance of common analysis methods for detecting low-frequency single nucleotide variants in targeted next-generation sequence data. J Mol Diagn 2013; 16:75-88. [PMID: 24211364 DOI: 10.1016/j.jmoldx.2013.09.003] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/16/2013] [Accepted: 09/04/2013] [Indexed: 12/31/2022] Open
Abstract
Next-generation sequencing (NGS) is becoming a common approach for clinical testing of oncology specimens for mutations in cancer genes. Unlike inherited variants, cancer mutations may occur at low frequencies because of contamination from normal cells or tumor heterogeneity and can therefore be challenging to detect using common NGS analysis tools, which are often designed for constitutional genomic studies. We generated high-coverage (>1000×) NGS data from synthetic DNA mixtures with variant allele fractions (VAFs) of 25% to 2.5% to assess the performance of four variant callers, SAMtools, Genome Analysis Toolkit, VarScan2, and SPLINTER, in detecting low-frequency variants. SAMtools had the lowest sensitivity and detected only 49% of variants with VAFs of approximately 25%; whereas the Genome Analysis Toolkit, VarScan2, and SPLINTER detected at least 94% of variants with VAFs of approximately 10%. VarScan2 and SPLINTER achieved sensitivities of 97% and 89%, respectively, for variants with observed VAFs of 1% to 8%, with >98% sensitivity and >99% positive predictive value in coding regions. Coverage analysis demonstrated that >500× coverage was required for optimal performance. The specificity of SPLINTER improved with higher coverage, whereas VarScan2 yielded more false positive results at high coverage levels, although this effect was abrogated by removing low-quality reads before variant identification. Finally, we demonstrate the utility of high-sensitivity variant callers with data from 15 clinical lung cancers.
Collapse
Affiliation(s)
- David H Spencer
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Manoj Tyagi
- Department of Genetics, Washington University, St. Louis, Missouri
| | - Francesco Vallania
- Genomics and Pathology Services, Washington University School of Medicine, St. Louis, Missouri
| | | | - John D Pfeifer
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Rob D Mitra
- Genomics and Pathology Services, Washington University School of Medicine, St. Louis, Missouri
| | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri.
| |
Collapse
|
1140
|
Yoshida K, Toki T, Okuno Y, Kanezaki R, Shiraishi Y, Sato-Otsubo A, Sanada M, Park MJ, Terui K, Suzuki H, Kon A, Nagata Y, Sato Y, Wang R, Shiba N, Chiba K, Tanaka H, Hama A, Muramatsu H, Hasegawa D, Nakamura K, Kanegane H, Tsukamoto K, Adachi S, Kawakami K, Kato K, Nishimura R, Izraeli S, Hayashi Y, Miyano S, Kojima S, Ito E, Ogawa S. The landscape of somatic mutations in Down syndrome-related myeloid disorders. Nat Genet 2013; 45:1293-9. [PMID: 24056718 DOI: 10.1038/ng.2759] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/19/2013] [Indexed: 12/11/2022]
Abstract
Transient abnormal myelopoiesis (TAM) is a myeloid proliferation resembling acute megakaryoblastic leukemia (AMKL), mostly affecting perinatal infants with Down syndrome. Although self-limiting in a majority of cases, TAM may evolve as non-self-limiting AMKL after spontaneous remission (DS-AMKL). Pathogenesis of these Down syndrome-related myeloid disorders is poorly understood, except for GATA1 mutations found in most cases. Here we report genomic profiling of 41 TAM, 49 DS-AMKL and 19 non-DS-AMKL samples, including whole-genome and/or whole-exome sequencing of 15 TAM and 14 DS-AMKL samples. TAM appears to be caused by a single GATA1 mutation and constitutive trisomy 21. Subsequent AMKL evolves from a pre-existing TAM clone through the acquisition of additional mutations, with major mutational targets including multiple cohesin components (53%), CTCF (20%), and EZH2, KANSL1 and other epigenetic regulators (45%), as well as common signaling pathways, such as the JAK family kinases, MPL, SH2B3 (LNK) and multiple RAS pathway genes (47%).
Collapse
Affiliation(s)
- Kenichi Yoshida
- 1] Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. [2] Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan. [3]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1141
|
Luthra R, Patel KP, Reddy NG, Haghshenas V, Routbort MJ, Harmon MA, Barkoh BA, Kanagal-Shamanna R, Ravandi F, Cortes JE, Kantarjian HM, Medeiros LJ, Singh RR. Next-generation sequencing-based multigene mutational screening for acute myeloid leukemia using MiSeq: applicability for diagnostics and disease monitoring. Haematologica 2013; 99:465-73. [PMID: 24142997 DOI: 10.3324/haematol.2013.093765] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Routine molecular testing in acute myeloid leukemia involves screening several genes of therapeutic and prognostic significance for mutations. A comprehensive analysis using single-gene assays requires large amounts of DNA, is cumbersome and timely consolidation of results for clinical reporting is challenging. High throughput, next-generation sequencing platforms widely used in research have not been tested vigorously for clinical application. Here we describe the clinical application of MiSeq, a next-generation sequencing platform to screen mutational hotspots in 54 cancer-related genes including genes relevant in acute myeloid leukemia (NRAS, KRAS, FLT3, NPM1, DNMT3A, IDH1/2, JAK2, KIT and EZH2). We sequenced 63 samples from patients with acute myeloid leukemia/myelodysplastic syndrome using MiSeq and compared the results with those obtained using another next-generation sequencing platform, Ion-Torrent Personal Genome Machine and other conventional testing platforms. MiSeq detected a total of 100 single nucleotide variants and 23 NPM1 insertions that were confirmed by Ion Torrent or conventional platforms, indicating complete concordance. FLT3-internal tandem duplications (n=10) were not detected; however, re-analysis of the MiSeq output by Pindel, an indel detection algorithm, did detect them. Dilution studies of cancer cell-line DNA showed that the quantitative accuracy of mutation detection was up to an allelic frequency of 1.5% with a high level of inter- and intra-run assay reproducibility, suggesting potential utility for monitoring response to therapy, clonal heterogeneity and evolution. Examples demonstrating the advantages of MiSeq over conventional platforms for disease monitoring are provided. Easy work-flow, high throughput multiplexing capability, 4-day turnaround time and simultaneous assessment of routinely tested and emerging markers make MiSeq highly applicable for clinical molecular testing in acute myeloid leukemia.
Collapse
|
1142
|
Abstract
Myelodysplasia is a diagnostic feature of myelodysplastic syndromes (MDSs) but is also found in other myeloid neoplasms. Its molecular basis has been recently elucidated by means of massive parallel sequencing studies. About 90% of MDS patients carry ≥1 oncogenic mutations, and two thirds of them are found in individuals with a normal karyotype. Driver mutant genes include those of RNA splicing (SF3B1, SRSF2, U2AF1, and ZRSR2), DNA methylation (TET2, DNMT3A, and IDH1/2), chromatin modification (ASXL1 and EZH2), transcription regulation (RUNX1), DNA repair (TP53), signal transduction (CBL, NRAS, and KRAS), and cohesin complex (STAG2). Only 4 to 6 genes are consistently mutated in ≥10% MDS patients, whereas a long tail of ∼50 genes are mutated less frequently. At presentation, most patients typically have 2 or 3 driver oncogenic mutations and hundreds of background mutations. MDS driver genes are also frequently mutated in other myeloid neoplasms. Reliable genotype/phenotype relationships include the association of the SF3B1 mutation with refractory anemia with ring sideroblasts, TET2/SRSF2 comutation with chronic myelomonocytic leukemia, and activating CSF3R mutation with chronic neutrophilic leukemia. Although both founding and subclonal driver mutations have been shown to have prognostic significance, prospective clinical trials that include the molecular characterization of the patient's genome are now needed.
Collapse
|
1143
|
Jäger N, Schlesner M, Jones DTW, Raffel S, Mallm JP, Junge KM, Weichenhan D, Bauer T, Ishaque N, Kool M, Northcott PA, Korshunov A, Drews RM, Koster J, Versteeg R, Richter J, Hummel M, Mack SC, Taylor MD, Witt H, Swartman B, Schulte-Bockholt D, Sultan M, Yaspo ML, Lehrach H, Hutter B, Brors B, Wolf S, Plass C, Siebert R, Trumpp A, Rippe K, Lehmann I, Lichter P, Pfister SM, Eils R. Hypermutation of the inactive X chromosome is a frequent event in cancer. Cell 2013; 155:567-81. [PMID: 24139898 PMCID: PMC3898475 DOI: 10.1016/j.cell.2013.09.042] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/02/2013] [Accepted: 09/20/2013] [Indexed: 11/26/2022]
Abstract
Mutation is a fundamental process in tumorigenesis. However, the degree to which the rate of somatic mutation varies across the human genome and the mechanistic basis underlying this variation remain to be fully elucidated. Here, we performed a cross-cancer comparison of 402 whole genomes comprising a diverse set of childhood and adult tumors, including both solid and hematopoietic malignancies. Surprisingly, we found that the inactive X chromosome of many female cancer genomes accumulates on average twice and up to four times as many somatic mutations per megabase, as compared to the individual autosomes. Whole-genome sequencing of clonally expanded hematopoietic stem/progenitor cells (HSPCs) from healthy individuals and a premalignant myelodysplastic syndrome (MDS) sample revealed no X chromosome hypermutation. Our data suggest that hypermutation of the inactive X chromosome is an early and frequent feature of tumorigenesis resulting from DNA replication stress in aberrantly proliferating cells. X chromosome has up to 4× more mutations than the autosomes in female cancer genomes Hypermutations only affect the inactive X chromosome X hypermutation involves somatic point mutations and indels, but not germline mutations No X hypermutation is found in clonal expansions of normal or premalignant cells
Collapse
Affiliation(s)
- Natalie Jäger
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1144
|
Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, Leiserson MDM, Miller CA, Welch JS, Walter MJ, Wendl MC, Ley TJ, Wilson RK, Raphael BJ, Ding L. Mutational landscape and significance across 12 major cancer types. Nature 2013. [DOI: 78495111110.1038/nature12634' target='_blank'>'"<>78495111110.1038/nature12634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [78495111110.1038/nature12634','', '10.1016/j.cell.2012.06.023')">Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
78495111110.1038/nature12634" />
|
1145
|
Abstract
Specific combinations of mutations, including FLT3 and IDH1/IDH2/TET2, frequently co-occur in acute myeloid leukemia (AML) and are associated with poor prognosis. These mutation combinations can be modeled in mice to provide a more genetically accurate model of AML. Within these models, stem cells may be different depending on how experiments are conducted and based on context. No one mutation can turn on a gene; rather the perfect storm of the right genes in the right cell is necessary to produce AML. Furthermore, this understanding is therapeutically relevant. Rapid and accurate targeted DNA sequencing will identify mutations of prognostic and therapeutic significance and will guide treatment choices in the future.
Collapse
Affiliation(s)
- Ross L Levine
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
1146
|
Whole-genome and whole-exome sequencing of bladder cancer identifies frequent alterations in genes involved in sister chromatid cohesion and segregation. Nat Genet 2013; 45:1459-63. [PMID: 24121792 DOI: 10.1038/ng.2798] [Citation(s) in RCA: 369] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 09/16/2013] [Indexed: 12/15/2022]
Abstract
Bladder cancer is one of the most common cancers worldwide, with transitional cell carcinoma (TCC) being the predominant form. Here we report a genomic analysis of TCC by both whole-genome and whole-exome sequencing of 99 individuals with TCC. Beyond confirming recurrent mutations in genes previously identified as being mutated in TCC, we identified additional altered genes and pathways that were implicated in TCC. Notably, we discovered frequent alterations in STAG2 and ESPL1, two genes involved in the sister chromatid cohesion and segregation (SCCS) process. Furthermore, we also detected a recurrent fusion involving FGFR3 and TACC3, another component of SCCS, by transcriptome sequencing of 42 DNA-sequenced tumors. Overall, 32 of the 99 tumors (32%) harbored genetic alterations in the SCCS process. Our analysis provides evidence that genetic alterations affecting the SCCS process may be involved in bladder tumorigenesis and identifies a new therapeutic possibility for bladder cancer.
Collapse
|
1147
|
Balbás-Martínez C, Sagrera A, Carrillo-de-Santa-Pau E, Earl J, Márquez M, Vazquez M, Lapi E, Castro-Giner F, Beltran S, Bayés M, Carrato A, Cigudosa JC, Domínguez O, Gut M, Herranz J, Juanpere N, Kogevinas M, Langa X, López-Knowles E, Lorente JA, Lloreta J, Pisano DG, Richart L, Rico D, Salgado RN, Tardón A, Chanock S, Heath S, Valencia A, Losada A, Gut I, Malats N, Real FX. Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy. Nat Genet 2013; 45:1464-9. [PMID: 24121791 PMCID: PMC3840052 DOI: 10.1038/ng.2799] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 09/16/2013] [Indexed: 12/11/2022]
Abstract
Urothelial bladder cancer (UBC) is heterogeneous at the clinical, pathological, and genetic levels. Tumor invasiveness (T) and grade (G) are the main factors associated with outcome and determine patient management (1). A discovery exome sequencing screen (n=17), followed by a prevalence screen (n=60), identified new genes mutated in this tumor coding for proteins involved in chromatin modification (MLL2, ASXL2, BPTF), cell division (STAG2, SMC1A, SMC1B), and DNA repair (ATM, ERCC2, FANCA). STAG2, a subunit of cohesin, was significantly and commonly mutated/lost in UBC, mainly in tumors of low stage/grade, and its loss was associated with improved outcome. Loss of expression was often observed in chromosomally-stable tumors and STAG2 knockdown in bladder cancer cells did not increase aneuploidy. STAG2 reintroduction in non-expressing cells led to reduced colony formation. Our findings indicate that STAG2 is a novel UBC tumor suppressor acting through mechanisms that are different from its role to prevent aneuploidy.
Collapse
Affiliation(s)
- Cristina Balbás-Martínez
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1148
|
|
1149
|
Reikvam H, Nepstad I, Tamburini J. Predicting effects of kinase inhibitor in therapy for myeloid malignancies – the challenges in capturing disease heterogeneity. Expert Opin Investig Drugs 2013; 22:1365-70. [DOI: 10.1517/13543784.2013.841137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
1150
|
Gervasini C, Russo S, Cereda A, Parenti I, Masciadri M, Azzollini J, Melis D, Aravena T, Doray B, Ferrarini A, Garavelli L, Selicorni A, Larizza L. Cornelia de Lange individuals with new and recurrent SMC1A mutations enhance delineation of mutation repertoire and phenotypic spectrum. Am J Med Genet A 2013; 161A:2909-19. [PMID: 24124034 DOI: 10.1002/ajmg.a.36252] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/29/2013] [Indexed: 01/11/2023]
Abstract
We report on the clinical and molecular characterization of eight patients, one male and seven females, with clinical diagnosis of Cornelia de Lange syndrome (CdLS), who were found to carry distinct mutations of the SMC1A gene. Five of the eight mutations are novel, with two involving amino acid residues previously described as altered in a different way. The other three have been reported each in a single case. Comparison of pairs of individuals with the same mutation indicates only partial overlap of their clinical phenotypes. The following novel missense mutations, all affecting highly conserved amino acid residues, were found: p.R398G in the N-terminal coiled-coil domain, p.V651M in the C-terminal coiled-coil/hinge junction, p.R693G in the C-terminal coiled-coil, and p.N1166T and p.L1189F in the C-terminal ABC cassette. The latter is localized in the H-loop, and represents the first mutation involving a functional motif of SMC1A protein. The effect of the mutations on SMC1A protein function has been predicted using four bioinformatic tools. All mutations except p.V651M were scored as pathogenic by three or four of the tools. p.V651M was found in the only male individual of our cohort, who presented with the most severe phenotype. This raises the issue of gender effect when addressing mutation-phenotype correlation for genes such as SMC1A, which incompletely escapes X-inactivation. Our clinical and molecular findings expand the total number of characterized SMC1A-mutated patients (from 44 to 52) and the restricted repertoire of SMC1A mutations (from 29 to 34), contributing to the molecular and clinical signature of SMC1A-based CdLS.
Collapse
Affiliation(s)
- Cristina Gervasini
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|