1001
|
Abstract
Genetic analyses have shaped much of our understanding of cancer. However, it is becoming increasingly clear that cancer cells display features of normal tissue organization, where cancer stem cells (CSCs) can drive tumor growth. Although often considered as mutually exclusive models to describe tumor heterogeneity, we propose that the genetic and CSC models of cancer can be harmonized by considering the role of genetic diversity and nongenetic influences in contributing to tumor heterogeneity. We offer an approach to integrating CSCs and cancer genetic data that will guide the field in interpreting past observations and designing future studies.
Collapse
Affiliation(s)
- Antonija Kreso
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
1002
|
Caspase-9 is required for normal hematopoietic development and protection from alkylator-induced DNA damage in mice. Blood 2014; 124:3887-95. [PMID: 25349173 DOI: 10.1182/blood-2014-06-582551] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Apoptosis and the DNA damage responses have been implicated in hematopoietic development and differentiation, as well as in the pathogenesis of myelodysplastic syndromes (MDS) and leukemia. However, the importance of late-stage mediators of apoptosis in hematopoiesis and leukemogenesis has not been elucidated. Here, we examine the role of caspase-9 (Casp9), the initiator caspase of the intrinsic apoptotic cascade, in murine fetal and adult hematopoiesis. Casp9 deficiency resulted in decreased erythroid and B-cell progenitor abundance and impaired function of hematopoietic stem cells after transplantation. Mouse bone marrow chimeras lacking Casp9 or its cofactor Apaf1 developed low white blood cell counts, decreased B-cell numbers, anemia, and reduced survival. Defects in apoptosis have also been previously implicated in susceptibility to therapy-related leukemia, a disease caused by exposure to DNA-damaging chemotherapy. We found that the burden of DNA damage was increased in Casp9-deficient cells after exposure to the alkylator, N-ethyl-nitrosourea (ENU). Furthermore, exome sequencing revealed that oligoclonal hematopoiesis emerged in Casp9-deficient bone marrow chimeras after alkylator exposure. Taken together, these findings suggest that defects in apoptosis could be a key step in the pathogenesis of alkylator-associated secondary malignancies.
Collapse
|
1003
|
Peterson EA, Chavan SS, Bauer MA, Heuck CJ, Johann DJ. Revealing the inherent heterogeneity of human malignancies by variant consensus strategies coupled with cancer clonal analysis. BMC Bioinformatics 2014; 15 Suppl 11:S9. [PMID: 25350589 PMCID: PMC4251058 DOI: 10.1186/1471-2105-15-s11-s9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tumors are heterogeneous in composition. They are composed of cancer cells proper, along with stromal elements that collectively form a microenvironment, all of which are necessary to nurture the malignant process. In addition, many of the stromal cells are modified to support the unique needs of the malignant state. Tumors are composed of a variety of clones or subpopulations of cancer cells, which may differ in karyotype, growth rate, expression of cell surface markers, sensitivity to therapeutics, etc. New tools and methods to provide an improved understanding of tumor clonal architecture are needed to guide therapy.
Collapse
|
1004
|
Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, McMichael JF, Schmidt HK, Yellapantula V, Miller CA, Ozenberger BA, Welch JS, Link DC, Walter MJ, Mardis ER, Dipersio JF, Chen F, Wilson RK, Ley TJ, Ding L. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014; 20:1472-8. [PMID: 25326804 PMCID: PMC4313872 DOI: 10.1038/nm.3733] [Citation(s) in RCA: 1439] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 09/21/2014] [Indexed: 12/15/2022]
Abstract
Several genetic alterations characteristic of leukemia and lymphoma have been detected in the blood of individuals without apparent hematological malignancies. The Cancer Genome Atlas (TCGA) provides a unique resource for comprehensive discovery of mutations and genes in blood that may contribute to the clonal expansion of hematopoietic stem/progenitor cells. Here, we analyzed blood-derived sequence data from 2,728 individuals from TCGA and discovered 77 blood-specific mutations in cancer-associated genes, the majority being associated with advanced age. Remarkably, 83% of these mutations were from 19 leukemia and/or lymphoma-associated genes, and nine were recurrently mutated (DNMT3A, TET2, JAK2, ASXL1, TP53, GNAS, PPM1D, BCORL1 and SF3B1). We identified 14 additional mutations in a very small fraction of blood cells, possibly representing the earliest stages of clonal expansion in hematopoietic stem cells. Comparison of these findings to mutations in hematological malignancies identified several recurrently mutated genes that may be disease initiators. Our analyses show that the blood cells of more than 2% of individuals (5-6% of people older than 70 years) contain mutations that may represent premalignant events that cause clonal hematopoietic expansion.
Collapse
Affiliation(s)
- Mingchao Xie
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Charles Lu
- The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jiayin Wang
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael D McLellan
- The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kimberly J Johnson
- Brown School Master of Public Health Program, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael C Wendl
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, USA. [3] Department of Mathematics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Joshua F McMichael
- The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Heather K Schmidt
- The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Venkata Yellapantula
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Christopher A Miller
- The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bradley A Ozenberger
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - John S Welch
- 1] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel C Link
- 1] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Matthew J Walter
- 1] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Elaine R Mardis
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [3] Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, USA. [4] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - John F Dipersio
- 1] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Feng Chen
- 1] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Richard K Wilson
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [3] Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, USA. [4] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Timothy J Ley
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [3] Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, USA. [4] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Li Ding
- 1] The Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA. [2] Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA. [3] Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, USA. [4] Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
1005
|
Gojo I, Karp JE. New strategies in acute myelogenous leukemia: leukemogenesis and personalized medicine. Clin Cancer Res 2014; 20:6233-41. [PMID: 25324141 DOI: 10.1158/1078-0432.ccr-14-0900] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent advances in molecular technology have unraveled the complexity of leukemogenesis and provided the opportunity to design more personalized and pathophysiology-targeted therapeutic strategies. Despite the use of intensive chemotherapy, relapse remains the most common cause for therapeutic failure in acute myelogenous leukemia (AML). The interactions between leukemia stem cells (LSC) and marrow microenvironment appear to be critical in promoting therapeutic resistance through progressive acquisition of genetic and epigenetic changes within leukemia cells and immune evasion, resulting in leukemia cell survival. With advances in genomic-sequencing efforts, epigenetic and phenotypic characterization, personalized therapeutic strategies aimed at critical leukemia survival mechanisms may be feasible in the near future. Here, we review select novel approaches to therapy of AML such as targeting LSC, altering leukemia/marrow microenvironment interactions, inhibiting DNA repair or cell-cycle checkpoints, and augmenting immune-based antileukemia activity.
Collapse
Affiliation(s)
- Ivana Gojo
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Judith E Karp
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
1006
|
Somatic mutations, genome mosaicism, cancer and aging. Curr Opin Genet Dev 2014; 26:141-9. [PMID: 25282114 DOI: 10.1016/j.gde.2014.04.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/04/2014] [Accepted: 04/11/2014] [Indexed: 01/11/2023]
Abstract
Genomes are inherently unstable due to the need for DNA sequence variation in the germ line to fuel evolution through natural selection. In somatic tissues mutations accumulate during development and aging, generating genome mosaics. There is little information about the possible causal role of increased somatic mutation loads in late-life disease and aging, with the exception of cancer. Characterizing somatic mutations and their functional consequences in normal tissues remains a formidable challenge due to their low, individual abundance. Here, I will briefly review our current knowledge of somatic mutations in animals and humans in relation to aging, how they arise and lead to genome mosaicism, the technology to study somatic mutations and how they possibly could cause non-clonal disease.
Collapse
|
1007
|
Abstract
SIRT1 inhibition facilitates elimination of CML stem cells by Imatinib, in part via p53 activation. In this issue of Cell Stem Cell, Li et al. (2014) demonstrate a similar role for SIRT1 inhibition in eradicating FLT3-ITD AML stem cells, potentially through a positive feedback loop with c-MYC, highlighting SIRT1 as a potential target in combination cancer therapy.
Collapse
Affiliation(s)
- Bernd B Zeisig
- Leukaemia and Stem Cell Biology Group, Department of Haematological Medicine, Division of Cancer Studies, King's College London, London WC2R 2LS, UK
| | - Chi Wai Eric So
- Leukaemia and Stem Cell Biology Group, Department of Haematological Medicine, Division of Cancer Studies, King's College London, London WC2R 2LS, UK.
| |
Collapse
|
1008
|
Rich A, Sun J, Aldayel AS, Yin CC, Medeiros LJ, Konoplev S. Myelomastocytic leukemia with aberrant CD25 expression: case report and review of the literature. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2014; 14:e173-7. [PMID: 25022599 DOI: 10.1016/j.clml.2014.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 04/15/2014] [Accepted: 06/04/2014] [Indexed: 02/03/2023]
MESH Headings
- Adenocarcinoma/surgery
- Antigens, Neoplasm/analysis
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Bone Marrow/pathology
- Brain/pathology
- Cerebral Hemorrhage/etiology
- Cytarabine/administration & dosage
- Fatal Outcome
- Humans
- Hydroxamic Acids/administration & dosage
- Idarubicin/administration & dosage
- Interleukin-2 Receptor alpha Subunit/analysis
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/classification
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemic Infiltration
- Male
- Mast Cells/pathology
- Middle Aged
- Neoplasms, Second Primary/blood
- Neoplasms, Second Primary/diagnosis
- Neoplasms, Second Primary/drug therapy
- Neoplasms, Second Primary/metabolism
- Neoplastic Stem Cells/pathology
- Opportunistic Infections/etiology
- Prostatectomy
- Prostatic Neoplasms/surgery
- Vorinostat
Collapse
Affiliation(s)
- Amy Rich
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jinlan Sun
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Abdulrahman S Aldayel
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - C Cameron Yin
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - L Jeffrey Medeiros
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sergej Konoplev
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
1009
|
Abstract
Acute myeloid leukemia (AML) arises within the bone marrow from a malignant hematopoietic progenitor cell. Though AML is still often fatal, cure rates overall continue to improve incrementally yet steadily, primarily for two reasons: first, insights into the pathogenesis of AML over the last several decades have led to the development of a relatively sophisticated classification scheme that allows more nuanced risk stratification to guide treatment choices; second, improvements in stem cell transplantation have allowed many more patients to take advantage of this highly effective therapeutic technique. Improvements in overall survival for patients with AML are expected to continue rising because of the anticipated introduction of targeted therapies into this treatment platform.
Collapse
|
1010
|
Mazzarella L, Riva L, Luzi L, Ronchini C, Pelicci PG. The Genomic and Epigenomic Landscapes of AML. Semin Hematol 2014; 51:259-72. [DOI: 10.1053/j.seminhematol.2014.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
1011
|
Abstract
Subclonal cancer populations change spatially and temporally during the disease course. Studies are revealing branched evolutionary cancer growth with low-frequency driver events present in subpopulations of cells, providing escape mechanisms for targeted therapeutic approaches. Despite such complexity, evidence is emerging for parallel evolution of subclones, mediated through distinct somatic events converging on the same gene, signal transduction pathway, or protein complex in different subclones within the same tumor. Tumors may follow gradualist paths (microevolution) as well as major shifts in evolutionary trajectories (macroevolution). Although macroevolution has been subject to considerable controversy in post-Darwinian evolutionary theory, we review evidence that such nongradual, saltatory leaps, driven through chromosomal rearrangements or genome doubling, may be particularly relevant to tumor evolution. Adapting cancer care to the challenges imposed by tumor micro- and macroevolution and developing deeper insight into parallel evolutionary events may prove central to improving outcome and reducing drug development costs.
Collapse
Affiliation(s)
- Marco Gerlinger
- Cancer Research UK London Research Institute, London, United Kingdom WC2A 3LY;
| | | | | | | | | | | |
Collapse
|
1012
|
Valent P, Sperr WR, Sotlar K, Reiter A, Akin C, Gotlib J, Horny HP, Arock M. The serum tryptase test: an emerging robust biomarker in clinical hematology. Expert Rev Hematol 2014; 7:683-90. [PMID: 25169217 PMCID: PMC4603354 DOI: 10.1586/17474086.2014.955008] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During the past few years, a number of molecular markers have been developed in clinical hematology, most of them related to specific gene defects. However, there is also an unmet need to develop novel serologic parameters to improve diagnostics and prognostication in daily practice. Among these, the serum tryptase appears to be a most reliable biomarker of myeloid neoplasms. Elevated tryptase levels are found in subgroups of patients with mastocytosis, myelodysplastic syndrome, myeloproliferative neoplasm, acute myeloid leukemia, chronic myeloid leukemia and chronic eosinophilic leukemia. In these patients, the tryptase level is of diagnostic and/or prognostic significance. In mastocytosis, an elevated tryptase level is a minor criterion of systemic disease and in BCR-ABL1(+) chronic myeloid leukemia, elevated tryptase at diagnosis correlates with treatment responses and overall survival. In patients with elevated tryptase, the enzyme also serves as follow-up parameter and can be employed to measure treatment-responses. In the current article, we review and update the perspectives of tryptase and provide recommendations for use of this conventional biomarker in daily practice.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Karl Sotlar
- Institute of Pathology, Ludwig-Maximilian University, Munich, Germany
| | - Andreas Reiter
- III. Medizinische Klinik, Universitäts-Medizin Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Cem Akin
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason Gotlib
- Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Hans-Peter Horny
- Institute of Pathology, Ludwig-Maximilian University, Munich, Germany
| | - Michel Arock
- Molecular Oncology and Pharmacology Unit, LBPA CNRS UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| |
Collapse
|
1013
|
Abstract
The BCR-ABL1 oncoprotein is the cause of chronic myeloid leukemia and occurs as a consequence of the translocation t(9;22), a well-defined genetic event that results in the formation of the Philadelphia chromosome. While this genomic aberration is recognized to be the main culprit of the chronic phase of chronic myeloid leukemia, the natural clonal evolution of this myeloproliferative neoplasm involves the accumulation of secondary alterations through genomic instability. Thus, efforts to dissect the frequency and nature of the genomic events at diagnosis and at later stages are producing valuable insights into understanding the mechanisms of blastic transformation and development of resistance in chronic myeloid leukemia. The identification of alternative BCR-ABL1-dependent and BCR-ABL1-independent targets that sustain the survival of leukemic blasts and/or leukemia-initiating cells will facilitate the development of novel viable therapeutic options for patients who become resistant or intolerant to the currently available therapeutic options based on tyrosine kinase inhibitors.
Collapse
|
1014
|
Kim TM, Jung SH, Baek IP, Lee SH, Choi YJ, Lee JY, Chung YJ, Lee SH. Regional biases in mutation screening due to intratumoural heterogeneity of prostate cancer. J Pathol 2014; 233:425-35. [PMID: 24870262 DOI: 10.1002/path.4380] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/13/2014] [Accepted: 05/19/2014] [Indexed: 12/19/2022]
Abstract
Intratumoural heterogeneity (ITH) leads to regional biases of the mutational landscape in a single tumour and may influence the single biopsy-based clinical diagnosis and treatment decision. To evaluate the extent of ITH in unifocal prostate cancers (PCAs), we analysed multiple regional biopsies from three PCAs, using whole-exome sequencing, DNA copy number and gene expression profiling analyses. A substantial level of ITH was identified, in that 0-61% and 18-71% of somatic variants were common or private, respectively, within a given cancer. The enhanced mutation detection rate in the combined sequencing dataset across intratumoural biopsies was demonstrated with respect to the total number of mutations identified in a given tumour. Allele frequencies of the mutations were positively correlated with the levels of intratumoural recurrence (private < shared < common), but some common mutations showed low allele frequency, suggesting that not all were clonally fixed. Regional biases in the presentation of a well-known TMPRSS2-ERG fusion was noted in one PCA and the somatic mutation- and copy number-based phylogenetic relationships between intratumoural biopsies were largely concordant. Genes showing intratumoural expression variability were commonly enriched in the molecular function of eicosanoid metabolism and PCA-relevant clinical markers. Taken together, our analyses identified a substantial level of genetic ITH in unifocal PCAs at the mutation, copy number and expression levels, which should be taken into account for the identification of biomarkers in the clinical setting.
Collapse
Affiliation(s)
- Tae-Min Kim
- Cancer Evolution Research Centre, Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
1015
|
Beerman I, Rossi DJ. Epigenetic regulation of hematopoietic stem cell aging. Exp Cell Res 2014; 329:192-9. [PMID: 25261778 DOI: 10.1016/j.yexcr.2014.09.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 09/11/2014] [Indexed: 12/27/2022]
Abstract
Aging is invariably associated with alterations of the hematopoietic stem cell (HSC) compartment, including loss of functional capacity, altered clonal composition, and changes in lineage contribution. Although accumulation of DNA damage occurs during HSC aging, it is unlikely such consistent aging phenotypes could be solely attributed to changes in DNA integrity. Another mechanism by which heritable traits could contribute to the changes in the functional potential of aged HSCs is through alterations in the epigenetic landscape of adult stem cells. Indeed, recent studies on hematopoietic stem cells have suggested that altered epigenetic profiles are associated with HSC aging and play a key role in modulating the functional potential of HSCs at different stages during ontogeny. Even small changes of the epigenetic landscape can lead to robustly altered expression patterns, either directly by loss of regulatory control or through indirect, additive effects, ultimately leading to transcriptional changes of the stem cells. Potential drivers of such changes in the epigenetic landscape of aged HSCs include proliferative history, DNA damage, and deregulation of key epigenetic enzymes and complexes. This review will focus largely on the two most characterized epigenetic marks - DNA methylation and histone modifications - but will also discuss the potential role of non-coding RNAs in regulating HSC function during aging.
Collapse
Affiliation(s)
- Isabel Beerman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children׳s Hospital, MA 02116, USA.
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children׳s Hospital, MA 02116, USA
| |
Collapse
|
1016
|
Zhou J, Chng WJ. Identification and targeting leukemia stem cells: The path to the cure for acute myeloid leukemia. World J Stem Cells 2014; 6:473-484. [PMID: 25258669 PMCID: PMC4172676 DOI: 10.4252/wjsc.v6.i4.473] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/22/2014] [Accepted: 08/30/2014] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence support the notion that acute myeloid leukemia (AML) is organized in a hierarchical system, originating from a special proportion of leukemia stem cells (LSC). Similar to their normal counterpart, hematopoietic stem cells (HSC), LSC possess self-renewal capacity and are responsible for the continued growth and proliferation of the bulk of leukemia cells in the blood and bone marrow. It is believed that LSC are also the root cause for the treatment failure and relapse of AML because LSC are often resistant to chemotherapy. In the past decade, we have made significant advancement in identification and understanding the molecular biology of LSC, but it remains a daunting task to specifically targeting LSC, while sparing normal HSC. In this review, we will first provide a historical overview of the discovery of LSC, followed by a summary of identification and separation of LSC by either cell surface markers or functional assays. Next, the review will focus on the current, various strategies for eradicating LSC. Finally, we will highlight future directions and challenges ahead of our ultimate goal for the cure of AML by targeting LSC.
Collapse
Affiliation(s)
- Jianbiao Zhou
- Jianbiao Zhou, Wee-Joo Chng, Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore
| | - Wee-Joo Chng
- Jianbiao Zhou, Wee-Joo Chng, Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore
| |
Collapse
|
1017
|
Abstract
PURPOSE OF REVIEW A plethora of studies over the past two decades have identified many genes that are recurrently mutated in acute myeloid leukemia (AML). Although great advances have been made in understanding the role of these mutated genes in AML disease pathogenesis, to date relatively few have been demonstrated to have direct clinical relevance. RECENT FINDINGS Genomic techniques have allowed for the identification of many mutated genes that appear to drive disease pathogenesis and prognosis in AML. Integrated analyses examining the co-occurrence of these genes in well annotated AML patient cohorts has helped to significantly refine prognostic models, allowing for a more nuanced selection of patients for optimal postremission therapies. Furthermore, there are emerging data that gene mutations may be useful to select patients for optimal doses and/or modalities of upfront AML therapy. Finally, mutated genes themselves hold promise as therapeutic targets, as supported by strong preclinical studies. SUMMARY Recent advances in our knowledge of the molecular genetics of AML have significantly improved our tools for clinical decision-making and promise to identify new therapies for patients.
Collapse
|
1018
|
Annesley CE, Brown P. The Biology and Targeting of FLT3 in Pediatric Leukemia. Front Oncol 2014; 4:263. [PMID: 25295230 PMCID: PMC4172015 DOI: 10.3389/fonc.2014.00263] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/08/2014] [Indexed: 12/22/2022] Open
Abstract
Despite remarkable improvement in treatment outcomes in pediatric leukemia over the past several decades, the prognosis for high-risk groups of acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), as well as for relapsed leukemia, remains poor. Intensification of chemotherapy regimens for those at highest risk has improved success rates, but at the cost of significantly increased morbidity and long-term adverse effects. With the success of imatinib in Philadelphia-chromosome-positive leukemia and all-trans retinoic acid in acute promyelocytic leukemia, the quest to find additional molecularly targeted therapies has generated much excitement over recent years. Another such possible target in pediatric acute leukemia is FMS-like tyrosine kinase 3 (FLT3). FLT3 aberrations are among the most frequently identified transforming events in AML, and have significant clinical implications in both high-risk pediatric AML and in certain high-risk groups of pediatric ALL. Therefore, the successful targeting of FLT3 has tremendous potential to improve outcomes in these subsets of patients. This article will give an overview of the molecular function and signaling of the FLT3 receptor, as well as its pathogenic role in leukemia. We review the discovery of targeting FLT3, discuss currently available FLT3 inhibitors in pediatric leukemia and results of clinical trials to date, and finally, consider the future promise and challenges of FLT3 inhibitor therapy.
Collapse
Affiliation(s)
- Colleen E. Annesley
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick Brown
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
1019
|
Wouters R, Cucchi D, Kaspers GJL, Schuurhuis GJ, Cloos J. Relevance of leukemic stem cells in acute myeloid leukemia: heterogeneity and influence on disease monitoring, prognosis and treatment design. Expert Rev Hematol 2014; 7:791-805. [PMID: 25242511 DOI: 10.1586/17474086.2014.959921] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Acute myeloid leukemia is a bone marrow disease characterized by a block in differentiation of the myeloid lineage with a concomitant uncontrolled high proliferation rate. Development of acute myeloid leukemia from stem cells with specific founder mutations, leads to an oligoclonal disease that progresses into a very heterogeneous leukemia at diagnosis. Measurement of leukemic stem cell load and characterization of these cells are essential for prediction of relapse and target identification, respectively. Prediction of relapse by monitoring the disease during minimal residual disease detection is challenged by clonal shifts during therapy. To overcome this, characterization of the potential relapse-initiating cells is required using both flow cytometry and molecular analysis since leukemic stem cells can be targeted both on extracellular features and on stem-cell specific signal transduction pathways.
Collapse
Affiliation(s)
- Rolf Wouters
- Departments of Pediatric Oncology/Hematology and Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
1020
|
Varetti G, Pellman D, Gordon DJ. Aurea mediocritas: the importance of a balanced genome. Cold Spring Harb Perspect Biol 2014; 6:a015842. [PMID: 25237130 DOI: 10.1101/cshperspect.a015842] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Aneuploidy, defined as an abnormal number of chromosomes, is a hallmark of cancer. Paradoxically, aneuploidy generally has a negative impact on cell growth and fitness in nontransformed cells. In this work, we review recent progress in identifying how aneuploidy leads to genomic and chromosomal instability, how cells can adapt to the deleterious effects of aneuploidy, and how aneuploidy contributes to tumorigenesis in different genetic contexts. Finally, we also discuss how aneuploidy might be a target for anticancer therapies.
Collapse
Affiliation(s)
- Gianluca Varetti
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - David Pellman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115 Howard Hughes Medical Institute, Chevy Chase, Maryland 20815-6789
| | - David J Gordon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| |
Collapse
|
1021
|
DeLaBarre B, Hurov J, Cianchetta G, Murray S, Dang L. Action at a distance: allostery and the development of drugs to target cancer cell metabolism. CHEMISTRY & BIOLOGY 2014; 21:1143-61. [PMID: 25237859 DOI: 10.1016/j.chembiol.2014.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/03/2014] [Accepted: 08/12/2014] [Indexed: 01/14/2023]
Abstract
Cancer cells must carefully regulate their metabolism to maintain growth and division under varying nutrient and oxygen levels. Compelling data support the investigation of numerous enzymes as therapeutic targets to exploit metabolic vulnerabilities common to several cancer types. We discuss the rationale for developing such drugs and review three targets with central roles in metabolic pathways crucial for cancer cell growth: pyruvate kinase muscle isozyme splice variant 2 (PKM2) in glycolysis, glutaminase in glutaminolysis, and mutations in isocitrate dehydrogenase 1 and 2 isozymes (IDH1/2) in the tricarboxylic acid cycle. These targets exemplify the drugging approach to cancer metabolism, with allosteric modulation being the common theme. The first glutaminase and mutant IDH1/2 inhibitors have entered clinical testing, and early data are promising. Cancer metabolism provides a wealth of novel targets, and targeting allosteric sites promises to yield selective drugs with the potential to transform clinical outcomes across many cancer types.
Collapse
Affiliation(s)
- Byron DeLaBarre
- Agios Pharmaceuticals, Inc., 38 Sidney Street, Cambridge, MA 02139, USA
| | - Jonathan Hurov
- Agios Pharmaceuticals, Inc., 38 Sidney Street, Cambridge, MA 02139, USA
| | | | - Stuart Murray
- Agios Pharmaceuticals, Inc., 38 Sidney Street, Cambridge, MA 02139, USA
| | - Lenny Dang
- Agios Pharmaceuticals, Inc., 38 Sidney Street, Cambridge, MA 02139, USA.
| |
Collapse
|
1022
|
Eiring AM, Deininger MW. Individualizing kinase-targeted cancer therapy: the paradigm of chronic myeloid leukemia. Genome Biol 2014; 15:461. [PMID: 25316524 PMCID: PMC4318205 DOI: 10.1186/s13059-014-0461-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The success of tyrosine kinase inhibitors in treating chronic myeloid leukemia highlights the potential of targeting oncogenic kinases with small molecules. By using drug activity profiles and individual patient genotypes, one can guide personalized therapy selection for patients with resistance.
Collapse
Affiliation(s)
- Anna M Eiring
- />Huntsman Cancer Institute, The University of Utah, Circle of Hope, Salt Lake City, UT 84112-5550 USA
| | - Michael W Deininger
- />Huntsman Cancer Institute, The University of Utah, Circle of Hope, Salt Lake City, UT 84112-5550 USA
- />Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT 84132 USA
| |
Collapse
|
1023
|
Thompson AM, Paguirigan AL, Kreutz JE, Radich JP, Chiu DT. Microfluidics for single-cell genetic analysis. LAB ON A CHIP 2014; 14:3135-42. [PMID: 24789374 PMCID: PMC4117719 DOI: 10.1039/c4lc00175c] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The ability to correlate single-cell genetic information to cellular phenotypes will provide the kind of detailed insight into human physiology and disease pathways that is not possible to infer from bulk cell analysis. Microfluidic technologies are attractive for single-cell manipulation due to precise handling and low risk of contamination. Additionally, microfluidic single-cell techniques can allow for high-throughput and detailed genetic analyses that increase accuracy and decrease reagent cost compared to bulk techniques. Incorporating these microfluidic platforms into research and clinical laboratory workflows can fill an unmet need in biology, delivering the highly accurate, highly informative data necessary to develop new therapies and monitor patient outcomes. In this perspective, we describe the current and potential future uses of microfluidics at all stages of single-cell genetic analysis, including cell enrichment and capture, single-cell compartmentalization and manipulation, and detection and analyses.
Collapse
Affiliation(s)
- A M Thompson
- Department of Chemistry, University of Washington, Seattle, WA, USA.
| | | | | | | | | |
Collapse
|
1024
|
Schleiermacher G, Javanmardi N, Bernard V, Leroy Q, Cappo J, Rio Frio T, Pierron G, Lapouble E, Combaret V, Speleman F, de Wilde B, Djos A, Øra I, Hedborg F, Träger C, Holmqvist BM, Abrahamsson J, Peuchmaur M, Michon J, Janoueix-Lerosey I, Kogner P, Delattre O, Martinsson T. Emergence of New ALK Mutations at Relapse of Neuroblastoma. J Clin Oncol 2014; 32:2727-34. [DOI: 10.1200/jco.2013.54.0674] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose In neuroblastoma, the ALK receptor tyrosine kinase is activated by point mutations. We investigated the potential role of ALK mutations in neuroblastoma clonal evolution. Methods We analyzed ALK mutations in 54 paired diagnosis–relapse neuroblastoma samples using Sanger sequencing. When an ALK mutation was observed in one paired sample, a minor mutated component in the other sample was searched for by more than 100,000× deep sequencing of the relevant hotspot, with a sensitivity of 0.17%. Results All nine ALK-mutated cases at diagnosis demonstrated the same mutation at relapse, in one case in only one of several relapse nodules. In five additional cases, the mutation seemed to be relapse specific, four of which were investigated by deep sequencing. In two cases, no mutation evidence was observed at diagnosis. In one case, the mutation was present at a subclonal level (0.798%) at diagnosis, whereas in another case, two different mutations resulting in identical amino acid changes were detected, one only at diagnosis and the other only at relapse. Further evidence of clonal evolution of ALK-mutated cells was provided by establishment of a fully ALK-mutated cell line from a primary sample with an ALK-mutated cell population at subclonal level (6.6%). Conclusion In neuroblastoma, subclonal ALK mutations can be present at diagnosis with subsequent clonal expansion at relapse. Given the potential of ALK-targeted therapy, the significant spatiotemporal variation of ALK mutations is of utmost importance, highlighting the potential of deep sequencing for detection of subclonal mutations with a sensitivity 100-fold that of Sanger sequencing and the importance of serial samplings for therapeutic decisions.
Collapse
Affiliation(s)
- Gudrun Schleiermacher
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Niloufar Javanmardi
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Virginie Bernard
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Quentin Leroy
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Julie Cappo
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Thomas Rio Frio
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Gaelle Pierron
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Eve Lapouble
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Valérie Combaret
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Frank Speleman
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Bram de Wilde
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Anna Djos
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Ingrid Øra
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Fredrik Hedborg
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Catarina Träger
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Britt-Marie Holmqvist
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Jonas Abrahamsson
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Michel Peuchmaur
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Jean Michon
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Isabelle Janoueix-Lerosey
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Per Kogner
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Olivier Delattre
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| | - Tommy Martinsson
- Gudrun Schleiermacher, Julie Cappo, Isabelle Janoueix-Lerosey, and Olivier Delattre, L'Institut National de la Santé et de la Recherche Médicale U830, Laboratoire de Génétique et Biologie des Cancers; Gudrun Schleiermacher, Virginie Bernard, Quentin Leroy, Thomas Rio Frio, Gaelle Pierron, Eve Lapouble, and Jean Michon, Institut Curie; Michel Peuchmaur, Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Robert Debré, Service de Pathologie; Michel Peuchmaur, Université Diderot Paris 7, Sorbonne
| |
Collapse
|
1025
|
Ossenkoppele GJ, Schuurhuis GJ. MRD in AML: It is time to change the definition of remission. Best Pract Res Clin Haematol 2014; 27:265-71. [DOI: 10.1016/j.beha.2014.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
1026
|
Matynia AP, Szankasi P, Shen W, Kelley TW. Molecular genetic biomarkers in myeloid malignancies. Arch Pathol Lab Med 2014; 139:594-601. [PMID: 25152312 DOI: 10.5858/arpa.2014-0096-ra] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Recent studies using massively parallel sequencing technologies, so-called next-generation sequencing, have uncovered numerous recurrent, single-gene variants or mutations across the spectrum of myeloid malignancies. OBJECTIVES To review the recent advances in the understanding of the molecular basis of myeloid neoplasms, including their significance for diagnostic and prognostic purposes and the possible implications for the development of novel therapeutic strategies. DATA SOURCES Literature review. CONCLUSIONS The recurrent mutations found in myeloid malignancies fall into distinct functional categories. These include (1) cell signaling factors, (2) transcription factors, (3) regulators of the cell cycle, (4) regulators of DNA methylation, (5) regulators of histone modification, (6) RNA-splicing factors, and (7) components of the cohesin complex. As the clinical significance of these mutations and mutation combinations is established, testing for their presence is likely to become a routine part of the diagnostic workup. This review will attempt to establish a framework for understanding these mutations in the context of myeloproliferative neoplasms, myelodysplastic syndromes, and acute myeloid leukemia.
Collapse
Affiliation(s)
- Anna P Matynia
- From the Department of Pathology, University of Utah, Salt Lake City (Drs Matynia and Kelley); and Research and Development, ARUP Laboratories, Salt Lake City, Utah (Drs Szankasi and Shen)
| | | | | | | |
Collapse
|
1027
|
Falandry C, Bonnefoy M, Freyer G, Gilson E. Biology of Cancer and Aging: A Complex Association With Cellular Senescence. J Clin Oncol 2014; 32:2604-10. [DOI: 10.1200/jco.2014.55.1432] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Over the last 50 years, major improvements have been made in our understanding of the driving forces, both parallel and opposing, that lead to aging and cancer. Many theories on aging first proposed in the 1950s, including those associated with telomere biology, senescence, and adult stem-cell regulation, have since gained support from cumulative experimental evidence. These views suggest that the accumulation of mutations might be a common driver of both aging and cancer. Moreover, some tumor suppressor pathways lead to aging in line with the theory of antagonist pleiotropy. According to the evolutionary-selected disposable soma theory, aging should affect primarily somatic cells. At the cellular level, both intrinsic and extrinsic pathways regulate aging and senescence. However, increasing lines of evidence support the hypothesis that these driving forces might be regulated by evolutionary-conserved pathways that modulate energy balance. According to the hyperfunction theory, aging is a quasi-program favoring both age-related diseases and cancer that could be inhibited by the regulation of longevity pathways. This review summarizes these hypotheses, as well as the experimental data that have accumulated over the last 60 years linking aging and cancer.
Collapse
Affiliation(s)
- Claire Falandry
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| | - Marc Bonnefoy
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| | - Gilles Freyer
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| | - Eric Gilson
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| |
Collapse
|
1028
|
Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome. Blood 2014; 124:2698-704. [PMID: 25139356 DOI: 10.1182/blood-2014-05-574889] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The distinction between acquired aplastic anemia (AA) and hypocellular myelodysplastic syndrome (hMDS) is often difficult, especially nonsevere AA. We postulated that somatic mutations are present in a subset of AA, and predict malignant transformation. From our database, we identified 150 AA patients with no morphological evidence of MDS, who had stored bone marrow (BM) and constitutional DNA. We excluded Fanconi anemia, mutations of telomere maintenance, and a family history of BM failure (BMF) or cancer. The initial cohort of 57 patients was screened for 835 known genes associated with BMF and myeloid cancer; a second cohort of 93 patients was screened for mutations in ASXL1, DNMT3A, BCOR, TET2, and MPL. Somatic mutations were detected in 19% of AA, and included ASXL1 (n = 12), DNMT3A (n = 8) and BCOR (n = 6). Patients with somatic mutations had a longer disease duration (37 vs 8 months, P < .04), and shorter telomere lengths (median length, 0.9 vs 1.1, P < .001), compared with patients without mutations. Somatic mutations in AA patients with a disease duration of >6 months were associated with a 40% risk of transformation to MDS (P < .0002). Nearly one-fifth of AA patients harbor mutations in genes typically seen in myeloid malignancies that predicted for later transformation to MDS.
Collapse
|
1029
|
Ablain J, de Thé H. Retinoic acid signaling in cancer: The parable of acute promyelocytic leukemia. Int J Cancer 2014; 135:2262-72. [PMID: 25130873 DOI: 10.1002/ijc.29081] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/04/2014] [Accepted: 05/09/2014] [Indexed: 12/22/2022]
Abstract
Inevitably fatal some 40 years, acute promyelocytic leukemia (APL) can now be cured in more than 95% of cases. This clinical success story is tightly linked to tremendous progress in our understanding of retinoic acid (RA) signaling. The discovery of retinoic acid receptor alpha (RARA) was followed by the cloning of the chromosomal translocations driving APL, all of which involve RARA. Since then, new findings on the biology of nuclear receptors have progressively enlightened the basis for the clinical efficacy of RA in APL. Reciprocally, the disease offered a range of angles to approach the cellular and molecular mechanisms of RA action. This virtuous circle contributed to make APL one of the best-understood cancers from both clinical and biological standpoints. Yet, some important questions remain unanswered including how lessons learnt from RA-triggered APL cure can help design new therapies for other malignancies.
Collapse
Affiliation(s)
- Julien Ablain
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris Cedex 10, France; INSERM U 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris Cedex 10, France; CNRS UMR 7212, Hôpital St. Louis, Paris Cedex 10, France
| | | |
Collapse
|
1030
|
Abstract
The use of genomic profiling in acute myeloid leukemia (AML) has led to an improved understanding of disease pathogenesis. Genomic profiling has given rise to fundamental observations about the biology of AML and has served to better define clinical outcomes for patients based on somatic mutational status. As additional mutations are identified with a known or postulated role in AML pathogenesis, the challenge ahead will be learning how to integrate these findings into clinical practice in such a way that they have a meaningful impact on patient care and, ultimately, on patient outcomes. Potential goals include using genomic information for refined risk stratification and clinical decision making, and to identify genetic lesions that guide the use of molecularly targeted therapies. The development of next-generation sequencing technologies has made genomic profiling a viable option for use in clinical practice because it can provide robust, high-coverage sequencing data for multiple genes in 1 assay, within a clinically reasonable time frame. The present article discusses recent candidate gene sequencing studies, the development of prognostic models based on these studies, and the current and potential future uses of next-generation sequencing technologies in making treatment decisions for patients with AML.
Collapse
Affiliation(s)
- Matias Sanchez
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ross L Levine
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY.
| | - Raajit Rampal
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
1031
|
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease, characterized by frequent resistance to available chemotherapeutic agents. The basic therapy for patients with AML has changed little over the past 30 years. Improvements in outcome in recent decades in younger adult cohorts have generally been ascribed to better supportive care (ie, transfusion and antimicrobial therapy); older adults with AML continue to fare poorly. The explosion of new knowledge regarding the AML genome has yet to be translated into therapeutic benefit, but analysis of specific molecular features in AML samples has enabled the field to more accurately parse out prognosis and assign appropriate therapies (eg, chemotherapy vs allogeneic stem cell transplantation) for groups of patients. Cytogenetic analysis, whether by metaphase or interphase analysis, has been the main tool used to divide patients into varying prognostic subsets, but it has been modified in recent years to include assessment of mutations in a small number of genes. In the past several years, new technologies have provided strategies to interrogate individual cancer genomes in a broad and in-depth fashion. The present article discusses the potential of these new technologies, particularly gene panel and whole-exome or whole-genome sequencing, to improve diagnosis, prognosis, and therapeutic outcome in AML.
Collapse
|
1032
|
Frequent cases of RAS-mutated Down syndrome acute lymphoblastic leukaemia lack JAK2 mutations. Nat Commun 2014; 5:4654. [PMID: 25105841 DOI: 10.1038/ncomms5654] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/09/2014] [Indexed: 12/12/2022] Open
Abstract
Children with Down syndrome (DS) and acute lymphoblastic leukaemia (ALL) have poorer survival and more relapses than non-DS children with ALL, highlighting an urgent need for deeper mechanistic understanding of DS-ALL. Here, using full-exome or cancer genes-targeted sequencing of 42 ALL samples from 39 DS patients, we uncover driver mutations in RAS, (KRAS and NRAS) recurring to a similar extent (15/42) as JAK2 (12/42) mutations or P2RY8-CRLF2 fusions (14/42). RAS mutations are almost completely mutually exclusive with JAK2 mutations (P=0.016), driving a combined total of two-thirds of analysed cases. Clonal architecture analysis reveals that both RAS and JAK2 drove sub-clonal expansions primarily initiated by CRLF2 rearrangements, and/or mutations in chromatin remodellers and lymphocyte differentiation factors. Remarkably, in 2/3 relapsed cases, there is a switch from a primary JAK2- or PTPN11-mutated sub-clone to a RAS-mutated sub-clone in relapse. These results provide important new insights informing the patient stratification strategies for targeted therapeutic approaches for DS-ALL.
Collapse
|
1033
|
Abstract
Cytogenetic data suggest that acute myeloid leukemia (AML) develops through a process of branching evolution, especially during relapse and progression. Recent genomic data from AML cases using digital sequencing, temporal comparisons, xenograft cloning, and single-cell analysis indicate that most, if not all, AML cases emerge through branching evolution. According to a review of the current literature, the balanced translocations (t[15;17], t[8;21], and inv[16]) and nucleotide variants in DNMT3A and TET2 most commonly occur in the founding clone at diagnosis. These mutations are rarely gained or lost at relapse, and the latter 2 mutations are observed in elderly subjects with mosaic hematopoiesis antedating overt leukemia. In contrast, +8, +13, +22, -X, -Y, and nucleotide variants in FLT3, NRAS/KRAS, WT1, and KIT frequently occur in subclones and are observed either to emerge or to be lost at relapse. Because drugs that target mutations within a subclone are unlikely to eliminate all leukemic cells, it will be essential to understand not only which mutations a patient has but also how they organize within the leukemic subclonal architecture.
Collapse
Affiliation(s)
- John S Welch
- Washington University School of Medicine, Division of Oncology, Stem Cell Biology Section, St. Louis, MO.
| |
Collapse
|
1034
|
Hematopoietic stem cell aging and chronic lymphocytic leukemia pathogenesis. Int J Hematol 2014; 100:335-40. [PMID: 25100497 DOI: 10.1007/s12185-014-1651-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/22/2014] [Accepted: 07/22/2014] [Indexed: 01/15/2023]
Abstract
Human malignancies develop through the multistep acquisition of critical somatic mutations during the clinical course. Regarding hematological malignancies, recent novel findings have indicated that hematopoietic stem cells (HSCs), which have the potential to self-renew and differentiate into multilineage hematopoietic cells, are an important cellular target for the accumulation of critical somatic mutations and play a central role in myeloid malignancy development. In contrast to myeloid malignancies, mature lymphoid malignancies, such as chronic lymphocytic leukemia (CLL), are considered to directly originate from differentiated mature lymphocytes; however, we previously reported that the propensity to generate clonal B cells had already been acquired at the HSC stage in CLL patients. Similarly, HSC involvement has been reported in the pathogenesis of mature T cell lymphomas. These studies indicate that, in mature lymphoid, if not all, malignancies, HSCs should be considered as the critical cellular target in the oncogenic process. The prevalence of these hematological malignancies dramatically increases with age, and the effect of aging HSCs should thus be taken into account when investigating the stepwise malignant transformation process of these age-associated malignancies.
Collapse
|
1035
|
Abstract
Leukemia is a disease that develops as a result of changes in the genomes of hematopoietic cells, a fact first appreciated by microscopic examination of the bone marrow cell chromosomes of affected patients. These studies revealed that specific subtypes of leukemia diagnoses correlated with specific chromosomal abnormalities, such as the t(15;17) of acute promyelocytic leukemia and the t(9;22) of chronic myeloid leukemia. Over time, our genomic characterization of hematologic malignancies has moved beyond the resolution of the microscope to that of individual nucleotides in the analysis of whole-genome sequencing (WGS) data using state-of-the-art massively parallel sequencing (MPS) instruments and algorithmic analyses of the resulting data. In addition to studying the genomic sequence alterations that occur in patients' genomes, these same instruments can decode the methylation landscape of the leukemia genome and the resulting RNA expression landscape of the leukemia transcriptome. Broad correlative analyses can then integrate these 3 data types to better inform researchers and clinicians about the biology of individual acute myeloid leukemia (AML) cases, facilitating improvements in care and prognosis.
Collapse
Affiliation(s)
- Elaine R Mardis
- The Genome Institute, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
1036
|
Meyer SC, Levine RL. Translational implications of somatic genomics in acute myeloid leukaemia. Lancet Oncol 2014; 15:e382-94. [DOI: 10.1016/s1470-2045(14)70008-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
1037
|
Miller CA, White BS, Dees ND, Griffith M, Welch JS, Griffith OL, Vij R, Tomasson MH, Graubert TA, Walter MJ, Ellis MJ, Schierding W, DiPersio JF, Ley TJ, Mardis ER, Wilson RK, Ding L. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput Biol 2014; 10:e1003665. [PMID: 25102416 PMCID: PMC4125065 DOI: 10.1371/journal.pcbi.1003665] [Citation(s) in RCA: 334] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 04/28/2014] [Indexed: 01/06/2023] Open
Abstract
The sensitivity of massively-parallel sequencing has confirmed that most cancers are oligoclonal, with subpopulations of neoplastic cells harboring distinct mutations. A fine resolution view of this clonal architecture provides insight into tumor heterogeneity, evolution, and treatment response, all of which may have clinical implications. Single tumor analysis already contributes to understanding these phenomena. However, cryptic subclones are frequently revealed by additional patient samples (e.g., collected at relapse or following treatment), indicating that accurately characterizing a tumor requires analyzing multiple samples from the same patient. To address this need, we present SciClone, a computational method that identifies the number and genetic composition of subclones by analyzing the variant allele frequencies of somatic mutations. We use it to detect subclones in acute myeloid leukemia and breast cancer samples that, though present at disease onset, are not evident from a single primary tumor sample. By doing so, we can track tumor evolution and identify the spatial origins of cells resisting therapy.
Collapse
Affiliation(s)
- Christopher A. Miller
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
| | - Brian S. White
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nathan D. Dees
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
| | - Malachi Griffith
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
| | - John S. Welch
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Obi L. Griffith
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ravi Vij
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael H. Tomasson
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Timothy A. Graubert
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Matthew J. Walter
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Matthew J. Ellis
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | | | - John F. DiPersio
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Timothy J. Ley
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elaine R. Mardis
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Richard K. Wilson
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Li Ding
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
1038
|
Tubio JMC, Li Y, Ju YS, Martincorena I, Cooke SL, Tojo M, Gundem G, Pipinikas CP, Zamora J, Raine K, Menzies A, Roman-Garcia P, Fullam A, Gerstung M, Shlien A, Tarpey PS, Papaemmanuil E, Knappskog S, Van Loo P, Ramakrishna M, Davies HR, Marshall J, Wedge DC, Teague JW, Butler AP, Nik-Zainal S, Alexandrov L, Behjati S, Yates LR, Bolli N, Mudie L, Hardy C, Martin S, McLaren S, O'Meara S, Anderson E, Maddison M, Gamble S, Foster C, Warren AY, Whitaker H, Brewer D, Eeles R, Cooper C, Neal D, Lynch AG, Visakorpi T, Isaacs WB, Veer LV, Caldas C, Desmedt C, Sotiriou C, Aparicio S, Foekens JA, Eyfjörd JE, Lakhani SR, Thomas G, Myklebost O, Span PN, Børresen-Dale AL, Richardson AL, Van de Vijver M, Vincent-Salomon A, Van den Eynden GG, Flanagan AM, Futreal PA, Janes SM, Bova GS, Stratton MR, McDermott U, Campbell PJ, ICGC Breast Cancer Group, ICGC Bone Cancer Group, ICGC Prostate Cancer Group. Mobile DNA in cancer. Extensive transduction of nonrepetitive DNA mediated by L1 retrotransposition in cancer genomes. Science 2014; 345:1251343. [PMID: 25082706 PMCID: PMC4380235 DOI: 10.1126/science.1251343] [Citation(s) in RCA: 301] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Long interspersed nuclear element-1 (L1) retrotransposons are mobile repetitive elements that are abundant in the human genome. L1 elements propagate through RNA intermediates. In the germ line, neighboring, nonrepetitive sequences are occasionally mobilized by the L1 machinery, a process called 3' transduction. Because 3' transductions are potentially mutagenic, we explored the extent to which they occur somatically during tumorigenesis. Studying cancer genomes from 244 patients, we found that tumors from 53% of the patients had somatic retrotranspositions, of which 24% were 3' transductions. Fingerprinting of donor L1s revealed that a handful of source L1 elements in a tumor can spawn from tens to hundreds of 3' transductions, which can themselves seed further retrotranspositions. The activity of individual L1 elements fluctuated during tumor evolution and correlated with L1 promoter hypomethylation. The 3' transductions disseminated genes, exons, and regulatory elements to new locations, most often to heterochromatic regions of the genome.
Collapse
Affiliation(s)
| | - Yilong Li
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Young Seok Ju
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | | | | | - Marta Tojo
- Department of Physiology, School of Medicine–Center for Resesarch in Molecular Medicine and Chronic Diseases, Instituto de Investigaciones Sanitarias, University of Santiago de Compostela, Spain
| | - Gunes Gundem
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | | | - Jorge Zamora
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Keiran Raine
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Andrew Menzies
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | | | - Anthony Fullam
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Moritz Gerstung
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Adam Shlien
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | | | | | - Stian Knappskog
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Peter Van Loo
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Human Genome Laboratory, Department of Human Genetics, VIB and KU Leuven, Leuven, Belgium
| | | | - Helen R. Davies
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - John Marshall
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - David C. Wedge
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Jon W. Teague
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Adam P. Butler
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Serena Nik-Zainal
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, UK
| | | | - Sam Behjati
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Lucy R. Yates
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Niccolo Bolli
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Laura Mudie
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Claire Hardy
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Sancha Martin
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Stuart McLaren
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Sarah O'Meara
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | | | - Mark Maddison
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Stephen Gamble
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | | | - Anne Y. Warren
- Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, UK
| | - Hayley Whitaker
- Cancer Research UK (CRUK) Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Daniel Brewer
- Institute of Cancer Research, Sutton, London, UK
- University of East Anglia, Norwich, UK
| | | | - Colin Cooper
- Institute of Cancer Research, Sutton, London, UK
- University of East Anglia, Norwich, UK
| | - David Neal
- Cancer Research UK (CRUK) Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Andy G. Lynch
- Cancer Research UK (CRUK) Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Tapio Visakorpi
- Institute of Biosciences and Medical Technology–BioMediTech, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | | | - Carlos Caldas
- Cancer Research UK (CRUK) Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Christine Desmedt
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sam Aparicio
- British Columbia Cancer Agency, Vancouver, Canada
| | - John A. Foekens
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Sunil R. Lakhani
- School of Medicine, University of Queensland, Brisbane, Australia
- Pathology Queensland, Royal Brisbane and Women’s Hospital, Brisbane, Australia
- UQ Centre for Clinical Research, University of Queensland, Brisbane, Australia
| | - Gilles Thomas
- Université Lyon 1, Institut National du Cancer (INCa)–Synergie, Lyon, France
| | - Ola Myklebost
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Paul N. Span
- Department of Radiation Oncology and Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | - Marc Van de Vijver
- Department of Pathology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Anne Vincent-Salomon
- Institut Bergonié, 229 cours de l’Argone, 33076 Bordeaux, France
- Institut Curie, Department of Tumor Biology, 26 rue d’Ulm, 75248 Paris cédex 05, France
| | - Gert G. Van den Eynden
- Translational Cancer Research Unit and Department of Pathology, GZA Hospitals, Antwerp, Belgium
| | - Adrienne M. Flanagan
- Royal National Orthopaedic Hospital, Middlesex, UK
- UCL Cancer Institute, University College London, London, UK
| | - P. Andrew Futreal
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- MD Anderson Cancer Center, Houston, TX, USA
| | - Sam M. Janes
- Lungs for Living Research Centre, Rayne Institute, University College London (UCL), London, UK
| | - G. Steven Bova
- Institute of Biosciences and Medical Technology–BioMediTech, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | - Ultan McDermott
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Peter J. Campbell
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | | | - ICGC Prostate Cancer Group
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Institute of Cancer Research, Sutton, London, UK
- Cancer Research UK (CRUK) Cambridge Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
1039
|
Abstract
Acute myeloid leukaemia (AML) is an uncontrolled clonal proliferation of abnormal myeloid progenitor cells in the bone marrow and blood. Advances in cancer genomics have revealed the spectrum of somatic mutations that give rise to human AML and drawn our attention to its molecular evolution and clonal architecture. It is now evident that most AML genomes harbour small numbers of mutations, which are acquired in a stepwise manner. This characteristic, combined with our ability to identify mutations in individual leukaemic cells and our detailed understanding of normal human and murine haematopoiesis, makes AML an excellent model for understanding the principles of cancer evolution. Furthermore, a better understanding of how AML evolves can help us devise strategies to improve the therapy and prognosis of AML patients. Here, we draw from recent advances in genomics, clinical studies and experimental models to describe the current knowledge of the clonal evolution of AML and its implications for the biology and treatment of leukaemias and other cancers.
Collapse
Affiliation(s)
- Carolyn S Grove
- Haematological Cancer Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - George S Vassiliou
- Haematological Cancer Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK.
| |
Collapse
|
1040
|
Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 2014; 512:198-202. [PMID: 25079315 DOI: 10.1038/nature13619] [Citation(s) in RCA: 491] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 06/27/2014] [Indexed: 02/07/2023]
Abstract
Haematopoietic stem cells (HSCs) self-renew for life, thereby making them one of the few blood cells that truly age. Paradoxically, although HSCs numerically expand with age, their functional activity declines over time, resulting in degraded blood production and impaired engraftment following transplantation. While many drivers of HSC ageing have been proposed, the reason why HSC function degrades with age remains unknown. Here we show that cycling old HSCs in mice have heightened levels of replication stress associated with cell cycle defects and chromosome gaps or breaks, which are due to decreased expression of mini-chromosome maintenance (MCM) helicase components and altered dynamics of DNA replication forks. Nonetheless, old HSCs survive replication unless confronted with a strong replication challenge, such as transplantation. Moreover, once old HSCs re-establish quiescence, residual replication stress on ribosomal DNA (rDNA) genes leads to the formation of nucleolar-associated γH2AX signals, which persist owing to ineffective H2AX dephosphorylation by mislocalized PP4c phosphatase rather than ongoing DNA damage. Persistent nucleolar γH2AX also acts as a histone modification marking the transcriptional silencing of rDNA genes and decreased ribosome biogenesis in quiescent old HSCs. Our results identify replication stress as a potent driver of functional decline in old HSCs, and highlight the MCM DNA helicase as a potential molecular target for rejuvenation therapies.
Collapse
|
1041
|
Abstract
Patient-specific ex vivo drug sensitivity and resistance screening can identify rational drug candidates for the testing of personalized targeted therapy. An iterative approach of genomic and drug susceptibility characterization at sequential time points during clinical trials of targeted therapy in acute myeloid leukemia may be useful both for characterizing mechanisms of resistance and clonal evolution and also for identification of novel therapeutic targets and drug combinations.
Collapse
Affiliation(s)
- Christopher S Hourigan
- 1Myeloid Malignancies Section, Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, and 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | | |
Collapse
|
1042
|
Tumoral reprogramming: Plasticity takes a walk on the wild side. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:436-47. [PMID: 25038581 DOI: 10.1016/j.bbagrm.2014.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 07/08/2014] [Accepted: 07/10/2014] [Indexed: 12/12/2022]
Abstract
Cellular plasticity is the capacity that cells have to change their fate and adopt a new identity. Plasticity is essential for normal development and for tissue regeneration and, in an experimental setting, for the induction of pluripotency. All these processes involve a reprogramming of the cellular identity, mediated by signals from the environment and/or by internal changes at the transcriptional and epigenetic levels. Tumorigenesis is a process in which normal cells acquire a new malignant identity and give rise to a clonal aberrant population. This is only possible if the initiating cell has the necessary plasticity to undergo such changes, and if the oncogenic event(s) initiating cancer has the essential reprogramming capacity so as to be able to lead a change in cellular identity. The molecular mechanisms underlying tumoral reprogramming are the pathological counterparts of the normal processes regulating developmental plasticity or experimentally-induced reprogramming. In this review we will first revise the main features of non-pathological examples of reprogramming, and then we will describe the parallelisms with tumoral reprogramming, and we will also delineate how the precise knowledge of the reprogramming mechanisms offers the potential for the development of new therapeutical interventions. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.
Collapse
|
1043
|
Meier B, Cooke SL, Weiss J, Bailly AP, Alexandrov LB, Marshall J, Raine K, Maddison M, Anderson E, Stratton MR, Gartner A, Campbell PJ. C. elegans whole-genome sequencing reveals mutational signatures related to carcinogens and DNA repair deficiency. Genome Res 2014; 24:1624-36. [PMID: 25030888 PMCID: PMC4199376 DOI: 10.1101/gr.175547.114] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mutation is associated with developmental and hereditary disorders, aging, and cancer. While we understand some mutational processes operative in human disease, most remain mysterious. We used Caenorhabditis elegans whole-genome sequencing to model mutational signatures, analyzing 183 worm populations across 17 DNA repair-deficient backgrounds propagated for 20 generations or exposed to carcinogens. The baseline mutation rate in C. elegans was approximately one per genome per generation, not overtly altered across several DNA repair deficiencies over 20 generations. Telomere erosion led to complex chromosomal rearrangements initiated by breakage–fusion–bridge cycles and completed by simultaneously acquired, localized clusters of breakpoints. Aflatoxin B1 induced substitutions of guanines in a GpC context, as observed in aflatoxin-induced liver cancers. Mutational burden increased with impaired nucleotide excision repair. Cisplatin and mechlorethamine, DNA crosslinking agents, caused dose- and genotype-dependent signatures among indels, substitutions, and rearrangements. Strikingly, both agents induced clustered rearrangements resembling “chromoanasynthesis,” a replication-based mutational signature seen in constitutional genomic disorders, suggesting that interstrand crosslinks may play a pathogenic role in such events. Cisplatin mutagenicity was most pronounced in xpf-1 mutants, suggesting that this gene critically protects cells against platinum chemotherapy. Thus, experimental model systems combined with genome sequencing can recapture and mechanistically explain mutational signatures associated with human disease.
Collapse
Affiliation(s)
- Bettina Meier
- Centre for Gene Regulation and Expression, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Susanna L Cooke
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Joerg Weiss
- Centre for Gene Regulation and Expression, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Aymeric P Bailly
- Centre for Gene Regulation and Expression, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom; CRBM/CNRS UMR5237, University of Montpellier, Montpellier 34293, France
| | - Ludmil B Alexandrov
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - John Marshall
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Keiran Raine
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Mark Maddison
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Elizabeth Anderson
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Michael R Stratton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Anton Gartner
- Centre for Gene Regulation and Expression, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom;
| | - Peter J Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom; Department of Haematology, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
1044
|
Hughes AEO, Magrini V, Demeter R, Miller CA, Fulton R, Fulton LL, Eades WC, Elliott K, Heath S, Westervelt P, Ding L, Conrad DF, White BS, Shao J, Link DC, DiPersio JF, Mardis ER, Wilson RK, Ley TJ, Walter MJ, Graubert TA. Clonal architecture of secondary acute myeloid leukemia defined by single-cell sequencing. PLoS Genet 2014; 10:e1004462. [PMID: 25010716 PMCID: PMC4091781 DOI: 10.1371/journal.pgen.1004462] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 05/13/2014] [Indexed: 12/30/2022] Open
Abstract
Next-generation sequencing has been used to infer the clonality of heterogeneous tumor samples. These analyses yield specific predictions—the population frequency of individual clones, their genetic composition, and their evolutionary relationships—which we set out to test by sequencing individual cells from three subjects diagnosed with secondary acute myeloid leukemia, each of whom had been previously characterized by whole genome sequencing of unfractionated tumor samples. Single-cell mutation profiling strongly supported the clonal architecture implied by the analysis of bulk material. In addition, it resolved the clonal assignment of single nucleotide variants that had been initially ambiguous and identified areas of previously unappreciated complexity. Accordingly, we find that many of the key assumptions underlying the analysis of tumor clonality by deep sequencing of unfractionated material are valid. Furthermore, we illustrate a single-cell sequencing strategy for interrogating the clonal relationships among known variants that is cost-effective, scalable, and adaptable to the analysis of both hematopoietic and solid tumors, or any heterogeneous population of cells. Human cancers are genetically diverse populations of cells that evolve over the course of their natural history or in response to the selective pressure of therapy. In theory, it is possible to infer how this variation is structured into related populations of cells based on the frequency of individual mutations in bulk samples, but the accuracy of these models has not been evaluated across a large number of variants in individual cells. Here, we report a strategy for analyzing hundreds of variants within a single cell, and we apply this method to assess models of tumor clonality derived from bulk samples in three cases of leukemia. The data largely support the predicted population structure, though they suggest specific refinements. This type of approach not only illustrates the biological complexity of human cancer, but it also has the potential to inform patient management. That is, precise knowledge of which variants are present in which populations of cells may allow physicians to more effectively target combinations of mutations and predict how patients will respond to therapy.
Collapse
Affiliation(s)
- Andrew E. O. Hughes
- Center for Genome Sciences and Systems Biology, Washington University, St. Louis, Missouri, United States of America
| | - Vincent Magrini
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
| | - Ryan Demeter
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
| | - Christopher A. Miller
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
| | - Robert Fulton
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
| | - Lucinda L. Fulton
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
| | - William C. Eades
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
| | - Kevin Elliott
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
| | - Sharon Heath
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
| | - Peter Westervelt
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - Li Ding
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
| | - Donald F. Conrad
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, United States of America
| | - Brian S. White
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
| | - Jin Shao
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - Daniel C. Link
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - John F. DiPersio
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - Elaine R. Mardis
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
| | - Richard K. Wilson
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - Timothy J. Ley
- The Genome Institute, Washington University, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - Matthew J. Walter
- Department of Genetics, Washington University, St. Louis, Missouri, United States of America
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
| | - Timothy A. Graubert
- Department of Internal Medicine, Division of Oncology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
1045
|
Zare H, Wang J, Hu A, Weber K, Smith J, Nickerson D, Song C, Witten D, Blau CA, Noble WS. Inferring clonal composition from multiple sections of a breast cancer. PLoS Comput Biol 2014; 10:e1003703. [PMID: 25010360 PMCID: PMC4091710 DOI: 10.1371/journal.pcbi.1003703] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 05/20/2014] [Indexed: 12/13/2022] Open
Abstract
Cancers arise from successive rounds of mutation and selection, generating clonal populations that vary in size, mutational content and drug responsiveness. Ascertaining the clonal composition of a tumor is therefore important both for prognosis and therapy. Mutation counts and frequencies resulting from next-generation sequencing (NGS) potentially reflect a tumor's clonal composition; however, deconvolving NGS data to infer a tumor's clonal structure presents a major challenge. We propose a generative model for NGS data derived from multiple subsections of a single tumor, and we describe an expectation-maximization procedure for estimating the clonal genotypes and relative frequencies using this model. We demonstrate, via simulation, the validity of the approach, and then use our algorithm to assess the clonal composition of a primary breast cancer and associated metastatic lymph node. After dividing the tumor into subsections, we perform exome sequencing for each subsection to assess mutational content, followed by deep sequencing to precisely count normal and variant alleles within each subsection. By quantifying the frequencies of 17 somatic variants, we demonstrate that our algorithm predicts clonal relationships that are both phylogenetically and spatially plausible. Applying this method to larger numbers of tumors should cast light on the clonal evolution of cancers in space and time. Cancers arise from a series of mutations that occur over time. As a result, as a tumor grows each cell inherits a distinctive genotype, defined by the set of all somatic mutations that distinguish the tumor cell from normal cells. Acertaining these genotype patterns, and identifying which ones are associated with the growth of the cancer and its ability to metastasize, can potentially give clinicians insights into how to treat the cancer. In this work, we describe a method for inferring the predominant genotypes within a single tumor. The method requires that a tumor be sectioned and that each section be subjected to a high-throughput sequencing procedure. The resulting mutations and their associated frequencies within each tumor section are then used as input to a probabilistic model that infers the underlying genotypes and their relative frequencies within the tumor. We use simulated data to demonstrate the validity of the approach, and then we apply our algorithm to data from a primary breast cancer and associated metastatic lymph node. We demonstrate that our algorithm predicts genotypes that are consistent with an evolutionary model and with the physical topology of the tumor itself. Applying this method to larger numbers of tumors should cast light on the evolution of cancers in space and time.
Collapse
Affiliation(s)
- Habil Zare
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Junfeng Wang
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Alex Hu
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Kris Weber
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Josh Smith
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Debbie Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - ChaoZhong Song
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Daniela Witten
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DW); (CAB); (WSN)
| | - C. Anthony Blau
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DW); (CAB); (WSN)
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
- Department of Computer Science and Engineering, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DW); (CAB); (WSN)
| |
Collapse
|
1046
|
Klevebring D, Rosin G, Ma R, Lindberg J, Czene K, Kere J, Fredriksson I, Bergh J, Hartman J. Sequencing of breast cancer stem cell populations indicates a dynamic conversion between differentiation states in vivo. Breast Cancer Res 2014; 16:R72. [PMID: 24998755 PMCID: PMC4227057 DOI: 10.1186/bcr3687] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/25/2014] [Indexed: 12/18/2022] Open
Abstract
Introduction The cancer stem cell model implies a hierarchical organization within breast tumors maintained by cancer stem-like cells (CSCs). Accordingly, CSCs are a subpopulation of cancer cells with capacity for self-renewal, differentiation and tumor initiation. These cells can be isolated through the phenotypic markers CD44+/CD24-, expression of ALDH1 and an ability to form nonadherent, multicellular spheres in vitro. However, controversies to describe the stem cell model exist; it is unclear whether the tumorigenicity of CSCs in vivo is solely a proxy for a certain genotype. Moreover, in vivo evidence is lacking to fully define the reversibility of CSC differentiation. Methods In order to answer these questions, we undertook exome sequencing of CSCs from 12 breast cancer patients, along with paired primary tumor samples. As suggested by stem classical cell biology, we assumed that the number of mutations in the CSC subpopulation should be lower and distinct compared to the differentiated tumor cells with higher proliferation. Results Our analysis revealed that the majority of somatic mutations are shared between CSCs and bulk primary tumor, with similar frequencies in the two. Conclusions The data presented here exclude the possibility that CSCs are only a phenotypic consequence of certain somatic mutations, that is a distinct and non-reversible population of cells. In addition, our results imply that CSCs must be a population of cells that can dynamically switch from differentiated tumor cells, and vice versa. This finding increases our understanding of CSC function in tumor heterogeneity and the importance of identifying drugs to counter de-differentiation rather than targeting CSCs.
Collapse
|
1047
|
Gerber JM. Isolated clonal cytogenetic abnormalities after high-dose therapy: do they matter? Biol Blood Marrow Transplant 2014; 20:1077-8. [PMID: 24953018 DOI: 10.1016/j.bbmt.2014.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 11/16/2022]
Affiliation(s)
- Jonathan M Gerber
- Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina.
| |
Collapse
|
1048
|
Reikvam H, Brenner AK, Nepstad I, Sulen A, Bruserud Ø. Heat shock protein 70 – the next chaperone to target in the treatment of human acute myelogenous leukemia? Expert Opin Ther Targets 2014; 18:929-44. [DOI: 10.1517/14728222.2014.924925] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
1049
|
Thanasopoulou A, Tzankov A, Schwaller J. Potent co-operation between the NUP98-NSD1 fusion and the FLT3-ITD mutation in acute myeloid leukemia induction. Haematologica 2014; 99:1465-71. [PMID: 24951466 DOI: 10.3324/haematol.2013.100917] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The NUP98-NSD1 fusion, product of the t(5;11)(q35;p15.5) chromosomal translocation, is one of the most prevalent genetic alterations in cytogenetically normal pediatric acute myeloid leukemias and is associated with poor prognosis. Co-existence of an FLT3-ITD activating mutation has been found in more than 70% of NUP98-NSD1-positive patients. To address functional synergism, we determined the transforming potential of retrovirally expressed NUP98-NSD1 and FLT3-ITD in the mouse. Expression of NUP98-NSD1 provided mouse strain-dependent, aberrant self-renewal potential to bone marrow progenitor cells. Co-expression of FLT3-ITD increased proliferation and maintained self-renewal in vitro. Transplantation of immortalized progenitors co-expressing NUP98-NSD1 and FLT3-ITD into mice resulted in acute myeloid leukemia after a short latency. In contrast, neither NUP98-NSD1 nor FLT3-ITD single transduced cells were able to initiate leukemia. Interestingly, as reported for patients carrying NUP98-NSD1, an increased Flt3-ITD to wild-type Flt3 mRNA expression ratio with increased FLT3-signaling was associated with rapidly induced disease. In contrast, there was no difference in the expression levels of the NUP98-NSD1 fusion or its proposed targets HoxA5, HoxA7, HoxA9 or HoxA10 between animals with different latencies to develop disease. Finally, leukemic cells co-expressing NUP98-NSD1 and FLT3-ITD were very sensitive to a small molecule FLT3 inhibitor, which underlines the significance of aberrant FLT3 signaling for NUP98-NSD1-positive leukemias and suggests new therapeutic approaches that could potentially improve patient outcome.
Collapse
Affiliation(s)
| | | | - Juerg Schwaller
- Department of Biomedicine, University Children's Hospital of Basel (UKBB), Switzerland
| |
Collapse
|
1050
|
Chiaretti S, Gianfelici V, Ceglie G, Foà R. Genomic characterization of acute leukemias. Med Princ Pract 2014; 23:487-506. [PMID: 24968698 PMCID: PMC5586934 DOI: 10.1159/000362793] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 04/10/2014] [Indexed: 01/09/2023] Open
Abstract
Over the past two decades, hematologic malignancies have been extensively evaluated due to the introduction of powerful technologies, such as conventional karyotyping, FISH analysis, gene and microRNA expression profiling, array comparative genomic hybridization and SNP arrays, and next-generation sequencing (including whole-exome sequencing and RNA-seq). These analyses have allowed for the refinement of the mechanisms underlying the leukemic transformation in several oncohematologic disorders and, more importantly, they have permitted the definition of novel prognostic algorithms aimed at stratifying patients at the onset of disease and, consequently, treating them in the most appropriate manner. Furthermore, the identification of specific molecular markers is opening the door to targeted and personalized medicine. The most important findings on novel acquisitions in the context of acute lymphoblastic leukemia of both B and T lineage and de novo acute myeloid leukemia are described in this review.
Collapse
Affiliation(s)
- Sabina Chiaretti
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | | | | | | |
Collapse
|