1251
|
He BL, Shi X, Man CH, Ma ACH, Ekker SC, Chow HCH, So CWE, Choi WWL, Zhang W, Zhang Y, Leung AYH. Functions of flt3 in zebrafish hematopoiesis and its relevance to human acute myeloid leukemia. Blood 2014; 123:2518-29. [PMID: 24591202 PMCID: PMC4017313 DOI: 10.1182/blood-2013-02-486688] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 02/18/2014] [Indexed: 12/22/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is expressed in human hematopoietic stem and progenitor cells (HSPCs) but its role during embryogenesis is unclear. In acute myeloid leukemia (AML), internal tandem duplication (ITD) of FLT3 at the juxtamembrane (JMD) and tyrosine kinase (TKD) domains (FLT3-ITD(+)) occurs in 30% of patients and is associated with inferior clinical prognosis. TKD mutations (FLT3-TKD(+)) occur in 5% of cases. We made use of zebrafish to examine the role of flt3 in developmental hematopoiesis and model human FLT3-ITD(+) and FLT3-TKD(+) AML. Zebrafish flt3 JMD and TKD were remarkably similar to their mammalian orthologs. Morpholino knockdown significantly reduced the expression of l-plastin (pan-leukocyte), csf1r, and mpeg1 (macrophage) as well as that of c-myb (definitive HSPCs), lck, and rag1 (T-lymphocyte). Expressing human FLT3-ITD in zebrafish embryos resulted in expansion and clustering of myeloid cells (pu.1(+), mpo(+), and cebpα(+)) which were ameliorated by AC220 and associated with stat5, erk1/2, and akt phosphorylation. Human FLT3-TKD (D835Y) induced significant, albeit modest, myeloid expansion resistant to AC220. This study provides novel insight into the role of flt3 during hematopoiesis and establishes a zebrafish model of FLT3-ITD(+) and FLT3-TKD(+) AML that may facilitate high-throughput screening of novel and personalized agents.
Collapse
Affiliation(s)
- Bai-Liang He
- Division of Haematology, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1252
|
Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis. Blood 2014; 123:e123-33. [PMID: 24740812 DOI: 10.1182/blood-2014-02-554634] [Citation(s) in RCA: 309] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Genomic studies have identified somatic alterations in the majority of myeloproliferative neoplasms (MPN) patients, including JAK2 mutations in the majority of MPN patients and CALR mutations in JAK2-negative MPN patients. However, the role of JAK-STAT pathway activation in different MPNs, and in patients without JAK2 mutations, has not been definitively delineated. We used expression profiling, single nucleotide polymorphism arrays, and mutational profiling to investigate a well-characterized cohort of MPN patients. MPN patients with homozygous JAK2V617F mutations were characterized by a distinctive transcriptional profile. Notably, a transcriptional signature consistent with activated JAK2 signaling is seen in all MPN patients regardless of clinical phenotype or mutational status. In addition, the activated JAK2 signature was present in patients with somatic CALR mutations. Conversely, we identified a gene expression signature of CALR mutations; this signature was significantly enriched in JAK2-mutant MPN patients consistent with a shared mechanism of transformation by JAK2 and CALR mutations. We also identified a transcriptional signature of TET2 mutations in MPN patent samples. Our data indicate that MPN patients, regardless of diagnosis or JAK2 mutational status, are characterized by a distinct gene expression signature with upregulation of JAK-STAT target genes, demonstrating the central importance of the JAK-STAT pathway in MPN pathogenesis.
Collapse
|
1253
|
Lymphohematopoietic cancers induced by chemicals and other agents and their implications for risk evaluation: An overview. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 761:40-64. [PMID: 24731989 DOI: 10.1016/j.mrrev.2014.04.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/13/2022]
Abstract
Lymphohematopoietic neoplasia are one of the most common types of cancer induced by therapeutic and environmental agents. Of the more than 100 human carcinogens identified by the International Agency for Research on Cancer, approximately 25% induce leukemias or lymphomas. The objective of this review is to provide an introduction into the origins and mechanisms underlying lymphohematopoietic cancers induced by xenobiotics in humans with an emphasis on acute myeloid leukemia, and discuss the implications of this information for risk assessment. Among the agents causing lymphohematopoietic cancers, a number of patterns were observed. Most physical and chemical leukemia-inducing agents such as the therapeutic alkylating agents, topoisomerase II inhibitors, and ionizing radiation induce mainly acute myeloid leukemia through DNA-damaging mechanisms that result in either gene or chromosomal mutations. In contrast, biological agents and a few immunosuppressive chemicals induce primarily lymphoid neoplasms through mechanisms that involve alterations in immune response. Among the environmental agents examined, benzene was clearly associated with acute myeloid leukemia in humans, with increasing but still limited evidence for an association with lymphoid neoplasms. Ethylene oxide and 1,3-butadiene were linked primarily to lymphoid cancers. Although the association between formaldehyde and leukemia remains controversial, several recent evaluations have indicated a potential link between formaldehyde and acute myeloid leukemia. The four environmental agents examined in detail were all genotoxic, inducing gene mutations, chromosomal alterations, and/or micronuclei in vivo. Although it is clear that rapid progress has been made in recent years in our understanding of leukemogenesis, many questions remain for future research regarding chemically induced leukemias and lymphomas, including the mechanisms by which the environmental agents reviewed here induce these diseases and the risks associated with exposures to such agents.
Collapse
|
1254
|
|
1255
|
Im AP, Sehgal AR, Carroll MP, Smith BD, Tefferi A, Johnson DE, Boyiadzis M. DNMT3A and IDH mutations in acute myeloid leukemia and other myeloid malignancies: associations with prognosis and potential treatment strategies. Leukemia 2014; 28:1774-83. [PMID: 24699305 DOI: 10.1038/leu.2014.124] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/05/2014] [Accepted: 03/25/2014] [Indexed: 02/08/2023]
Abstract
The development of effective treatment strategies for most forms of acute myeloid leukemia (AML) has languished for the past several decades. There are a number of reasons for this, but key among them is the considerable heterogeneity of this disease and the paucity of molecular markers that can be used to predict clinical outcomes and responsiveness to different therapies. The recent large-scale sequencing of AML genomes is now providing opportunities for patient stratification and personalized approaches to treatment that are based on individual mutational profiles. It is particularly notable that studies by The Cancer Genome Atlas and others have determined that 44% of patients with AML exhibit mutations in genes that regulate methylation of genomic DNA. In particular, frequent mutation has been observed in the genes encoding DNA methyltransferase 3A (DNMT3A), isocitrate dehydrogenase 1 (IDH1) and isocitrate dehydrogenase 2 (IDH2), as well as Tet oncogene family member 2. This review will summarize the incidence of these mutations, their impact on biochemical functions including epigenetic modification of genomic DNA and their potential usefulness as prognostic indicators. Importantly, the presence of DNMT3A, IDH1 or IDH2 mutations may confer sensitivity to novel therapeutic approaches, including the use of demethylating agents. Therefore, the clinical experience with decitabine and azacitidine in the treatment of patients harboring these mutations will be reviewed. Overall, we propose that understanding the role of these mutations in AML biology will lead to more rational therapeutic approaches targeting molecularly defined subtypes of the disease.
Collapse
Affiliation(s)
- A P Im
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - A R Sehgal
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - M P Carroll
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - B D Smith
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology at the Johns Hopkins University, Baltimore, MD, USA
| | - A Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - D E Johnson
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - M Boyiadzis
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
1256
|
Patnaik MM, Itzykson R, Lasho TL, Kosmider O, Finke CM, Hanson CA, Knudson RA, Ketterling RP, Tefferi A, Solary E. ASXL1 and SETBP1 mutations and their prognostic contribution in chronic myelomonocytic leukemia: a two-center study of 466 patients. Leukemia 2014; 28:2206-12. [PMID: 24695057 DOI: 10.1038/leu.2014.125] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 11/09/2022]
Abstract
In a cohort of 466 patients, we sought to clarify the prognostic relevance of ASXL1 and SETBP1 mutations, among others, in World Health Organization-defined chronic myelomonocytic leukemia (CMML) and its added value to the Mayo prognostic model. In univariate analysis, survival was adversely affected by ASXL1 (nonsense and frameshift) but not SETBP1 mutations. In multivariable analysis, ASXL1 mutations, absolute monocyte count >10 × 10(9)/l, hemoglobin <10 g/dl, platelets <100 × 10(9)/l and circulating immature myeloid cells were independently predictive of shortened survival: hazard ratio (95% confidence interval (CI)) values were 1.5 (1.1-2.0), 2.2 (1.6-3.1), 2.0 (1.6-2.6), 1.5 (1.2-1.9) and 2.0 (1.4-2.7), respectively. A regression coefficient-based prognostic model based on these five risk factors delineated high (≥3 risk factors; HR 6.2, 95% CI 3.7-10.4) intermediate-2 (2 risk factors; HR 3.4, 95% CI 2.0-5.6) intermediate-1 (one risk factor; HR 1.9, 95% CI 1.1-3.3) and low (no risk factors) risk categories with median survivals of 16, 31, 59 and 97 months, respectively. Neither ASXL1 nor SETBP1 mutations predicted leukemic transformation. The current study confirms the independent prognostic value of nonsense/frameshift ASXL1 mutations in CMML and signifies its added value to the Mayo prognostic model, as had been shown previously in the French consortium model.
Collapse
Affiliation(s)
- M M Patnaik
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - R Itzykson
- 1] Université Paris Descartes, Paris, France [2] Institut Gustave Roussy, Villejuif, France [3] Université Paris-Sud 11, Orsay, France
| | - T L Lasho
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - O Kosmider
- 1] Université Paris Descartes, Paris, France [2] Institut Cochin, Paris, France
| | - C M Finke
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - C A Hanson
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | - R A Knudson
- Division of Cytogenetics, Mayo Clinic, Rochester, MN, USA
| | - R P Ketterling
- Division of Cytogenetics, Mayo Clinic, Rochester, MN, USA
| | - A Tefferi
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - E Solary
- 1] Institut Gustave Roussy, Villejuif, France [2] Université Paris-Sud 11, Orsay, France [3] INSERM U1009, Villejuif, France
| |
Collapse
|
1257
|
Ostronoff F, Meshinchi S, Estey EH. Mutation profiling and prediction of outcome in acute myeloid leukemia. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Fabiana Ostronoff
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Pediatrics, Division of Hematology, University of Washington, Seattle, WA, USA
| | - Elihu H Estey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| |
Collapse
|
1258
|
Liu WJ, Tan XH, Luo XP, Guo BP, Wei ZJ, Ke Q, He S, Cen H. Prognostic significance of Tet methylcytosine dioxygenase 2 (TET2) gene mutations in adult patients with acute myeloid leukemia: a meta-analysis. Leuk Lymphoma 2014; 55:2691-8. [PMID: 24524305 DOI: 10.3109/10428194.2014.893308] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tet methylcytosine dioxygenase 2 (TET2) gene mutations have recently been recognized in acute myeloid leukemia (AML). We performed a meta-analysis to evaluate the controversial prognostic significance of TET2 mutations in AML. Eight studies, covering 2552 patients with AML, were included in this analysis. Pooled hazard ratios (HRs) indicated that TET2 mutations had a poor prognostic impact on the survival of patients with AML. The combined HR for overall survival (OS) was 1.53 and the summary HR for event-free survival (EFS) was 1.64. Additionally, TET2 mutations appeared to be an adverse prognostic indicator in both patients with cytogenetically normal (CN)-AML (HR for OS: 1.43 and HR for EFS: 1.76) and subgroups of patients with favorable-risk genotypes (HR for EFS: 2.35) and intermediate-I-risk genotypes (HR for EFS: 1.57). These findings indicate that TET2 mutations have an adverse impact on prognosis and may help to justify risk-adapted therapeutic strategies for patients with AML.
Collapse
Affiliation(s)
- Wen-Jian Liu
- Department of Chemotherapy, Tumor Hospital of Guangxi Medical University , Nanning , People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
1259
|
Clinical and biological impact of TET2 mutations and expression in younger adult AML patients treated within the EORTC/GIMEMA AML-12 clinical trial. Ann Hematol 2014; 93:1401-12. [PMID: 24994606 DOI: 10.1007/s00277-014-2055-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 01/24/2023]
Abstract
We assessed the prognostic impact of TET2 mutations and mRNA expression in a prospective cohort of 357 adult AML patients < 60 years of age enrolled in the European Organization For Research and Treatment of Cancer (EORTC)/Gruppo Italiano Malattie Ematologiche dell' Adulto (GIMEMA) AML-12 06991 clinical trial. In addition the co-occurrence with other genetic defects and the functional consequences of TET2 mutations were investigated. TET2 mutations occurred in 7.6 % of the patients and were an independent marker of poor prognosis (p = 0.024). TET2 and IDH1/2 mutations strongly associated with aberrations in the DNA methyltransferase DNMT3A. Functional studies confirmed previous work that neither nonsense truncations, nor missense TET2 mutations, induced 5-hydroxymethylcytosine formation. In addition, we now show that mutant TET2 forms did not act in a dominant negative manner when co-expressed with the wild-type protein. Finally, as loss-of-function TET2 mutations predicted poor outcome, we questioned whether low TET2 mRNA expression in cases of AML without TET2 mutations would affect overall survival. Notably, also AML patients with low TET2 mRNA expression levels showed inferior overall survival.
Collapse
|
1260
|
The epigenetic landscape of acute myeloid leukemia. Adv Hematol 2014; 2014:103175. [PMID: 24778653 PMCID: PMC3980839 DOI: 10.1155/2014/103175] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/27/2014] [Accepted: 02/03/2014] [Indexed: 11/29/2022] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous disease. Certain cytogenetic and molecular genetic mutations are recognized to have an impact on prognosis, leading to their inclusion in some prognostic stratification systems. Recently, the advent of high-throughput whole genome or exome sequencing has led to the identification of several novel recurrent mutations in AML, a number of which have been found to involve genes concerned with epigenetic regulation. These genes include in particular DNMT3A, TET2, and IDH1/2, involved with regulation of DNA methylation, and EZH2 and ASXL-1, which are implicated in regulation of histones. However, the precise mechanisms linking these genes to AML pathogenesis have yet to be fully elucidated as has their respective prognostic relevance. As massively parallel DNA sequencing becomes increasingly accessible for patients, there is a need for clarification of the clinical implications of these mutations. This review examines the literature surrounding the biology of these epigenetic modifying genes with regard to leukemogenesis and their clinical and prognostic relevance in AML when mutated.
Collapse
|
1261
|
Cagnetta A, Adamia S, Acharya C, Patrone F, Miglino M, Nencioni A, Gobbi M, Cea M. Role of genotype-based approach in the clinical management of adult acute myeloid leukemia with normal cytogenetics. Leuk Res 2014; 38:649-59. [PMID: 24726781 DOI: 10.1016/j.leukres.2014.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/03/2014] [Accepted: 03/09/2014] [Indexed: 02/02/2023]
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia affecting adults. Although it is a complex disease driven by numerous genetic and epigenetic abnormalities, nearly 50% of patients exhibit a normal karyotype (CN-AML) with an intermediate cytogenetic risk. However, a widespread genomic analysis has recently shown the recurrence of genomic aberrations in this category (mutations of FLT3, CEBPA, NPM1, RUNX1, TET2, IDH1/2, DNMT3A, ASXL1, MLL and WT1) thus revealing its marked genomic heterogeneity. In this perspective, a global gene expression analysis of AML patients provides an independent prognostic marker to categorize each patient into clinic-pathologic subgroups based on its molecular genetic defects. Consistently such classification, taking into account the uniqueness of each AML patient, furnishes an individualized treatment approach leading a step closer to personalized medicine. Overall the genome-wide analysis of AML patients, by providing novel insights into biology of this tumor, furnishes accurate prognostic markers as well as useful tools for selecting the most appropriate treatment option. Moreover it provides novel therapeutic targets useful to enhance efficacy of the current anti-AML therapeutics. Here we describe the prognostic relevance of such new genetic data and discuss how this approach can be used to improve survival and treatment of AML patients.
Collapse
Affiliation(s)
- Antonia Cagnetta
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy.
| | - Sophia Adamia
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chirag Acharya
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Franco Patrone
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Maurizio Miglino
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Alessio Nencioni
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Marco Gobbi
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Michele Cea
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy.
| |
Collapse
|
1262
|
|
1263
|
Abstract
Epigenetic alterations are associated with all aspects of cancer, from tumor initiation to cancer progression and metastasis. It is now well understood that both losses and gains of DNA methylation as well as altered chromatin organization contribute significantly to cancer-associated phenotypes. More recently, new sequencing technologies have allowed the identification of driver mutations in epigenetic regulators, providing a mechanistic link between the cancer epigenome and genetic alterations. Oncogenic activating mutations are now known to occur in a number of epigenetic modifiers (i.e. IDH1/2, EZH2, DNMT3A), pinpointing epigenetic pathways that are involved in tumorigenesis. Similarly, investigations into the role of inactivating mutations in chromatin modifiers (i.e. KDM6A, CREBBP/EP300, SMARCB1) implicate many of these genes as tumor suppressors. Intriguingly, a number of neoplasms are defined by a plethora of mutations in epigenetic regulators, including renal, bladder, and adenoid cystic carcinomas. Particularly striking is the discovery of frequent histone H3.3 mutations in pediatric glioma, a particularly aggressive neoplasm that has long remained poorly understood. Cancer epigenetics is a relatively new, promising frontier with much potential for improving cancer outcomes. Already, therapies such as 5-azacytidine and decitabine have proven that targeting epigenetic alterations in cancer can lead to tangible benefits. Understanding how genetic alterations give rise to the cancer epigenome will offer new possibilities for developing better prognostic and therapeutic strategies.
Collapse
|
1264
|
Chromatin modifiers and the promise of epigenetic therapy in acute leukemia. Leukemia 2014; 28:1396-406. [PMID: 24609046 DOI: 10.1038/leu.2014.94] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/04/2014] [Accepted: 02/14/2014] [Indexed: 12/31/2022]
Abstract
Hematopoiesis is a tightly regulated process involving the control of gene expression that directs the transition from hematopoietic stem and progenitor cells to terminally differentiated blood cells. In leukemia, the processes directing self-renewal, differentiation and progenitor cell expansion are disrupted, leading to the accumulation of immature, non-functioning malignant cells. Insights into these processes have come in stages, based on technological advances in genetic analyses, bioinformatics and biological sciences. The first cytogenetic studies of leukemic cells identified chromosomal translocations that generate oncogenic fusion proteins and most commonly affect regulators of transcription. This was followed by the discovery of recurrent somatic mutations in genes encoding regulators of the signal transduction pathways that control cell proliferation and survival. Recently, studies of global changes in methylation and gene expression have led to the understanding that the output of transcriptional regulators and the proliferative signaling pathways are ultimately influenced by chromatin structure. Candidate gene, whole-genome and whole-exome sequencing studies have identified recurrent somatic mutations in genes encoding epigenetic modifiers in both acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL). In contrast to the two-hit model of leukemogenesis, emerging evidence suggests that these epigenetic modifiers represent a class of mutations that are critical to the development of leukemia and affect the regulation of various other oncogenic pathways. In this review, we discuss the range of recurrent, somatic mutations in epigenetic modifiers found in leukemia and how these modifiers relate to the classical leukemogenic pathways that lead to impaired cell differentiation and aberrant self-renewal and proliferation.
Collapse
|
1265
|
Järås M, Miller PG, Chu LP, Puram RV, Fink EC, Schneider RK, Al-Shahrour F, Peña P, Breyfogle LJ, Hartwell KA, McConkey ME, Cowley GS, Root DE, Kharas MG, Mullally A, Ebert BL. Csnk1a1 inhibition has p53-dependent therapeutic efficacy in acute myeloid leukemia. ACTA ACUST UNITED AC 2014; 211:605-12. [PMID: 24616378 PMCID: PMC3978274 DOI: 10.1084/jem.20131033] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Despite extensive insights into the underlying genetics and biology of acute myeloid leukemia (AML), overall survival remains poor and new therapies are needed. We found that casein kinase 1 α (Csnk1a1), a serine-threonine kinase, is essential for AML cell survival in vivo. Normal hematopoietic stem and progenitor cells (HSPCs) were relatively less affected by shRNA-mediated knockdown of Csnk1a1. To identify downstream mediators of Csnk1a1 critical for leukemia cells, we performed an in vivo pooled shRNA screen and gene expression profiling. We found that Csnk1a1 knockdown results in decreased Rps6 phosphorylation, increased p53 activity, and myeloid differentiation. Consistent with these observations, p53-null leukemias were insensitive to Csnk1a1 knockdown. We further evaluated whether D4476, a casein kinase 1 inhibitor, would exhibit selective antileukemic effects. Treatment of leukemia stem cells (LSCs) with D4476 showed highly selective killing of LSCs over normal HSPCs. In summary, these findings demonstrate that Csnk1a1 inhibition causes reduced Rps6 phosphorylation and activation of p53, resulting in selective elimination of leukemia cells, revealing Csnk1a1 as a potential therapeutic target for the treatment of AML.
Collapse
Affiliation(s)
- Marcus Järås
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1266
|
Schiller GJ. Evolving treatment strategies in patients with high-risk acute myeloid leukemia. Leuk Lymphoma 2014; 55:2438-48. [DOI: 10.3109/10428194.2014.881479] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Gary J. Schiller
- Hematological Malignancy, Stem Cell Transplant Program, David Geffen School of Medicine at UCLA,
Los Angeles, CA, USA
| |
Collapse
|
1267
|
|
1268
|
Loberiza FR, Cannon AC, Cannon AJ, Bierman PJ. Insights on practice variations in the management of lymphoma and leukemia. Leuk Lymphoma 2014; 55:2449-56. [DOI: 10.3109/10428194.2014.881480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Fausto R. Loberiza
- Section of Oncology/Hematology, Internal Medicine, University of Nebraska Medical Center,
Omaha, NE, USA
| | - Andrew C. Cannon
- Section of Oncology/Hematology, Internal Medicine, University of Nebraska Medical Center,
Omaha, NE, USA
| | - Anthony J. Cannon
- Section of Oncology/Hematology, Internal Medicine, University of Nebraska Medical Center,
Omaha, NE, USA
| | - Philip J. Bierman
- Section of Oncology/Hematology, Internal Medicine, University of Nebraska Medical Center,
Omaha, NE, USA
| |
Collapse
|
1269
|
Abdel-Wahab O, Klimek VM, Gaskell AA, Viale A, Cheng D, Kim E, Rampal R, Bluth M, Harding JJ, Callahan MK, Merghoub T, Berger MF, Solit DB, Rosen N, Levine RL, Chapman PB. Efficacy of intermittent combined RAF and MEK inhibition in a patient with concurrent BRAF- and NRAS-mutant malignancies. Cancer Discov 2014; 4:538-45. [PMID: 24589925 DOI: 10.1158/2159-8290.cd-13-1038] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vemurafenib, a RAF inhibitor, extends survival in patients with BRAF(V600)-mutant melanoma but activates extracellular signal-regulated kinase (ERK) signaling in RAS-mutant cells. In a patient with a BRAF(V600K)-mutant melanoma responding to vemurafenib, we observed accelerated progression of a previously unrecognized NRAS-mutant leukemia. We hypothesized that combining vemurafenib with a MAP-ERK kinase (MEK) inhibitor would inhibit ERK activation in the melanoma and prevent ERK activation by vemurafenib in the leukemia, and thus suppress both malignancies. We demonstrate that intermittent administration of vemurafenib led to a near-complete remission of the melanoma, and the addition of the MEK inhibitor cobimetinib (GDC-0973) caused suppression of vemurafenib-induced leukemic proliferation and ERK activation. Antimelanoma and antileukemia responses have been maintained for nearly 20 months, as documented by serial measurements of tumor-derived DNA in plasma in addition to conventional radiographic and clinical assessments of response. These data support testing of intermittent ERK pathway inhibition in the therapy for both RAS-mutant leukemia and BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Omar Abdel-Wahab
- 1Human Oncology and Pathogenesis Program, 2Leukemia Service, 3Gastrointestinal Oncology Service, 4Melanoma and Immunotherapeutics Service, Department of Medicine, 5Molecular Diagnostics Service, Department of Pathology, 6Department of Radiology, 7Center for Molecular Oncology, 8Ludwig Center for Cancer Immunotherapy, and 9Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center; and 10Weill Cornell Medical College, New York, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1270
|
Yamaguchi S, Iwanaga E, Tokunaga K, Nanri T, Shimomura T, Suzushima H, Mitsuya H, Asou N. IDH1 and IDH2 mutations confer an adverse effect in patients with acute myeloid leukemia lacking the NPM1 mutation. Eur J Haematol 2014; 92:471-7. [PMID: 24443894 DOI: 10.1111/ejh.12271] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2014] [Indexed: 01/07/2023]
Abstract
We examined the incidence and prognostic effect of IDH1 and IDH2 mutations in 233 Japanese adults with acute myeloid leukemia (AML). IDH1 R132 mutations were detected in 20 (8.6%) patients with AML. IDH2 mutations were found in 19 (8.2%, 17 R140 and two R172) patients. IDH1 and IDH2 mutations were mutually exclusive and were associated with normal karyotype AML, cytogenetic intermediate-risk group, and NPM1 mutations. Five-year overall survival (OS) rates were significantly lower (15.6%) in patients harboring the IDH mutations than in patients lacking the IDH mutation (32.0%) in the entire cohort of AML (P = 0.005). Among patients aged 59 yr or younger with IDH mutations, 5-yr OS in patients who underwent allogeneic stem cell transplantation (SCT) was significantly higher than that in those not receiving allogeneic SCT (50% vs. 10.6%, P = 0.020). Of 51 patients with NPM1 mutations, there was no significant difference in 5-yr OS rates between patients with and those without the IDH mutations. In contrast, among 175 patients lacking the NPM1 mutations, 5-yr OS rate in patients with IDH mutations was significantly lower than that in those without IDH mutations (0% vs. 34.7%, P = <0.001). These data suggest that IDH mutations have an unfavorable effect in AML, especially AML with the NPM1 wild type and younger AML patients with IDH mutations may benefit from allogeneic SCT.
Collapse
Affiliation(s)
- Shunichiro Yamaguchi
- Department of Hematology, Kumamoto University School of Medicine, Kumamoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
1271
|
|
1272
|
Greim H, Kaden DA, Larson RA, Palermo CM, Rice JM, Ross D, Snyder R. The bone marrow niche, stem cells, and leukemia: impact of drugs, chemicals, and the environment. Ann N Y Acad Sci 2014; 1310:7-31. [PMID: 24495159 PMCID: PMC4002179 DOI: 10.1111/nyas.12362] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hematopoietic stem cells (HSCs) are a unique population of somatic stem cells that can both self-renew for long-term reconstitution of HSCs and differentiate into hematopoietic progenitor cells (HPCs), which in turn give rise, in a hierarchical manner, to the entire myeloid and lymphoid lineages. The differentiation and maturation of these lineages occurs in the bone marrow (BM) niche, a microenvironment that regulates self-renewal, survival, differentiation, and proliferation, with interactions among signaling pathways in the HSCs and the niche required to establish and maintain homeostasis. The accumulation of genetic mutations and cytogenetic abnormalities within cells of the partially differentiated myeloid lineage, particularly as a result of exposure to benzene or cytotoxic anticancer drugs, can give rise to malignancies like acute myeloid leukemia and myelodysplastic syndrome. Better understanding of the mechanisms driving these malignancies and susceptibility factors, both within HPCs and cells within the BM niche, may lead to the development of strategies for prevention of occupational and cancer therapy-induced disease.
Collapse
|
1273
|
Liu LL, Zhang DH, Mao X, Zhang XH, Zhang B. Over-expression of FoxM1 is associated with adverse prognosis and FLT3-ITD in acute myeloid leukemia. Biochem Biophys Res Commun 2014; 446:280-5. [PMID: 24582753 DOI: 10.1016/j.bbrc.2014.02.094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 02/22/2014] [Indexed: 11/25/2022]
Abstract
Forkhead box M1 (FoxM1) drives cell cycle progression and the prevention of growth arrest and is over-expressed in many human malignancies. However, the characteristics of FoxM1 in acute myeloid leukemia (AML) are not clearly understood. We investigated the expression level of FoxM1 and analyzed the correlation of FoxM1 expression with AML patient characteristics and prognoses. Changes in FoxM1 expression were detected after MV4-11 cells, which have an internal tandem duplication (ITD) of the fms-like tyrosine kinase 3 gene (FLT3-ITD), and control THP1 cells (encoding wild-type FLT3) were treated with the FLT3 receptor tyrosine kinase inhibitor AC220 (quizartinib) or FLT3 ligand (FL). Finally, we determined the apoptosis rates after the addition of the FoxM1 inhibitor thiostrepton (TST) to AML cells with or without FLT3-ITD. The expression of FoxM1 in AML patients was correlated with the presence of FLT3-ITD, genetic groups, and possibly overall survival. Inhibition of FLT3-ITD by AC220 down-regulated FoxM1 expression in MV4-11 cells, and stimulation of FLT3 by FL up-regulated FoxM1 expression in MV4-11 and THP1 cells. TST induced the apoptosis of MV4-11 and THP1 cells in a dose-dependent manner. Thus, FoxM1 is a potential prognostic marker and a promising therapeutic target in AML.
Collapse
Affiliation(s)
- Long-long Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Hematology, Wuhan General Hospital of Guangzhou Military, Wuhan, Hubei, China
| | - Dong-hua Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin-hua Zhang
- Department of Hematology, Wuhan General Hospital of Guangzhou Military, Wuhan, Hubei, China
| | - Bing Zhang
- Department of Hematology, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu, China
| |
Collapse
|
1274
|
Incidence and prognostic significance of karyotypic subgroups in older patients with acute myeloid leukemia: the Swedish population-based experience. Blood Cancer J 2014; 4:e188. [PMID: 24583534 PMCID: PMC3944658 DOI: 10.1038/bcj.2014.10] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 01/24/2014] [Accepted: 01/29/2014] [Indexed: 12/21/2022] Open
Abstract
The Swedish population-based acute myeloid leukemia registry contains data from 3251 patients (excluding acute promyelocytic leukemia) diagnosed between 1997 and 2006. Informative cytogenetic data from 1893 patients were retrospectively added, including 1054 patients aged between 60 and 79 years. Clonal abnormalities were found in 57% of the informative karyotypes. Karyotypic patterns differed by age: t(8;21), inv(16) and t(11q23) were more common in younger patients, whereas loss of 5q, 7q and 17p, monosomal karyotype (MK) and complex karyotypes were more common in older patients. Loss of 5q, 7q and 17p often occurred together within MK. Patients with 5 chromosome abnormalities had worse overall survival than those with fewer abnormalities or normal karyotype in all age groups. Loss of 5q, 7q and/or 17p had, in contrast to MK, a further negative impact on survival. Multivariable Cox regression analyses on risk factors in patients <80 years with cytogenetic abnormalities and intensive treatment revealed that age and performance status had the most significant impact on survival (both P<0.001), followed by sex (P=0.0135) and a karyotype including -7/del(7q) (P=0.048).
Collapse
|
1275
|
Gritsman K, Yuzugullu H, Von T, Yan H, Clayton L, Fritsch C, Maira SM, Hollingworth G, Choi C, Khandan T, Paktinat M, Okabe RO, Roberts TM, Zhao JJ. Hematopoiesis and RAS-driven myeloid leukemia differentially require PI3K isoform p110α. J Clin Invest 2014; 124:1794-809. [PMID: 24569456 DOI: 10.1172/jci69927] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 12/17/2013] [Indexed: 01/19/2023] Open
Abstract
The genes encoding RAS family members are frequently mutated in juvenile myelomonocytic leukemia (JMML) and acute myeloid leukemia (AML). RAS proteins are difficult to target pharmacologically; therefore, targeting the downstream PI3K and RAF/MEK/ERK pathways represents a promising approach to treat RAS-addicted tumors. The p110α isoform of PI3K (encoded by Pik3ca) is an essential effector of oncogenic KRAS in murine lung tumors, but it is unknown whether p110α contributes to leukemia. To specifically examine the role of p110α in murine hematopoiesis and in leukemia, we conditionally deleted p110α in HSCs using the Cre-loxP system. Postnatal deletion of p110α resulted in mild anemia without affecting HSC self-renewal; however, deletion of p110α in mice with KRASG12D-associated JMML markedly delayed their death. Furthermore, the p110α-selective inhibitor BYL719 inhibited growth factor-independent KRASG12D BM colony formation and sensitized cells to a low dose of the MEK inhibitor MEK162. Furthermore, combined inhibition of p110α and MEK effectively reduced proliferation of RAS-mutated AML cell lines and disease in an AML murine xenograft model. Together, our data indicate that RAS-mutated myeloid leukemias are dependent on the PI3K isoform p110α, and combined pharmacologic inhibition of p110α and MEK could be an effective therapeutic strategy for JMML and AML.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Class I Phosphatidylinositol 3-Kinases
- Erythropoiesis/genetics
- Erythropoiesis/physiology
- Genes, ras
- Hematopoiesis/genetics
- Hematopoiesis/physiology
- Heterografts
- Humans
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelomonocytic, Juvenile/enzymology
- Leukemia, Myelomonocytic, Juvenile/genetics
- Leukemia, Myelomonocytic, Juvenile/pathology
- MAP Kinase Signaling System
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphatidylinositol 3-Kinases/deficiency
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Signal Transduction
Collapse
|
1276
|
Kohlmann A, Bacher U, Schnittger S, Haferlach T. Perspective on how to approach molecular diagnostics in acute myeloid leukemia and myelodysplastic syndromes in the era of next-generation sequencing. Leuk Lymphoma 2014; 55:1725-34. [PMID: 24144312 DOI: 10.3109/10428194.2013.856427] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Molecular mutation information became essential for biological subclassification, risk stratification and therapeutic decisions in patients with acute myeloid leukemia (AML). In myelodysplastic syndromes (MDS), a broad spectrum of molecular biomarkers such as the spliceosome mutations has been identified in recent years. The currently established combination of various polymerase chain reaction (PCR) methods with capillary Sanger sequencing for mutation analysis in AML is time-consuming and labor-intensive. The constantly increasing spectrum of molecular mutations is a tremendous challenge for hematological laboratories. The introduction of high-throughput sequencing technology, which allows the massive parallel analysis of hundreds of thousands of alleles in the shortest time, provides new options for molecular mutation analyses and for follow-up diagnostics in myeloid neoplasms. In contrast to whole-genome or exome analyses, amplicon deep-sequencing focuses on distinct genomic loci and their mutation patterns and enables a comprehensive biomarker analysis in a multitude of patients per analysis. This review summarizes thus far established common molecular diagnostic strategies and intends to outline the perspective of distinct novel amplicon deep-sequencing panels for patients with AML and MDS. It is foreseeable that clearly defined algorithms for molecular investigations will revolutionize diagnosis in patients with AML and MDS in the near future.
Collapse
|
1277
|
Shlush LI, Zandi S, Mitchell A, Chen WC, Brandwein JM, Gupta V, Kennedy JA, Schimmer AD, Schuh AC, Yee KW, McLeod JL, Doedens M, Medeiros JJF, Marke R, Kim HJ, Lee K, McPherson JD, Hudson TJ, Brown AMK, Yousif F, Trinh QM, Stein LD, Minden MD, Wang JCY, Dick JE. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 2014; 506:328-33. [PMID: 24522528 PMCID: PMC4991939 DOI: 10.1038/nature13038] [Citation(s) in RCA: 1110] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/20/2014] [Indexed: 12/17/2022]
Abstract
In acute myeloid leukemia (AML), the cell of origin, nature and biological consequences of initiating lesions and order of subsequent mutations remain poorly understood, as AML is typically diagnosed without observation of a pre-leukemic phase. Here, highly purified hematopoietic stem cells (HSC), progenitor and mature cell fractions from the blood of AML patients were found to contain recurrent DNMT3a mutations (DNMT3amut) at high allele frequency, but without coincident NPM1 mutations (NPM1c) present in AML blasts. DNMT3amut-bearing HSC exhibited multilineage repopulation advantage over non-mutated HSC in xenografts, establishing their identity as pre-leukemic-HSC (preL-HSC). preL-HSC were found in remission samples indicating that they survive chemotherapy. Thus DNMT3amut arises early in AML evolution, likely in HSC, leading to a clonally expanded pool of preL-HSC from which AML evolves. Our findings provide a paradigm for the detection and treatment of pre-leukemic clones before the acquisition of additional genetic lesions engenders greater therapeutic resistance.
Collapse
Affiliation(s)
- Liran I Shlush
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2]
| | - Sasan Zandi
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2]
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada
| | - Weihsu Claire Chen
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada
| | - Joseph M Brandwein
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada
| | - Vikas Gupta
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada
| | - James A Kennedy
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada
| | - Aaron D Schimmer
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada [4] Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Andre C Schuh
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada
| | - Karen W Yee
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada
| | - Jessica L McLeod
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada
| | - Monica Doedens
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada
| | - Jessie J F Medeiros
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada
| | - Rene Marke
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Radboud University, Nijmegen Medical Centre, Nijmegen 6500 HB, The Netherlands
| | - Hyeoung Joon Kim
- Chonnam National University Hwasun Hospital, Genome Research Center for Hematopoietic Diseases, Gwangju 519-809, South Korea
| | - Kwon Lee
- Chonnam National University Hwasun Hospital, Genome Research Center for Hematopoietic Diseases, Gwangju 519-809, South Korea
| | - John D McPherson
- 1] Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada [2] Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Thomas J Hudson
- 1] Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada [2] Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada [3] Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | - Andrew M K Brown
- Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | | | - Quang M Trinh
- Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Lincoln D Stein
- 1] Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada [2] Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Mark D Minden
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada [4] Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Jean C Y Wang
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Medicine, University of Toronto, Toronto, Ontario M5S 2J7, Canada [3] Division of Medical Oncology and Hematology, UHN, Toronto, Ontario M5G 2M9, Canada
| | - John E Dick
- 1] Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario M5G 2M9, Canada [2] Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
1278
|
Puissant A, Fenouille N, Alexe G, Pikman Y, Bassil CF, Mehta S, Du J, Kazi JU, Luciano F, Rönnstrand L, Kung AL, Aster JC, Galinsky I, Stone RM, DeAngelo DJ, Hemann MT, Stegmaier K. SYK is a critical regulator of FLT3 in acute myeloid leukemia. Cancer Cell 2014; 25:226-42. [PMID: 24525236 PMCID: PMC4106711 DOI: 10.1016/j.ccr.2014.01.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 11/14/2013] [Accepted: 01/22/2014] [Indexed: 10/25/2022]
Abstract
Cooperative dependencies between mutant oncoproteins and wild-type proteins are critical in cancer pathogenesis and therapy resistance. Although spleen tyrosine kinase (SYK) has been implicated in hematologic malignancies, it is rarely mutated. We used kinase activity profiling to identify collaborators of SYK in acute myeloid leukemia (AML) and determined that FMS-like tyrosine kinase 3 (FLT3) is transactivated by SYK via direct binding. Highly activated SYK is predominantly found in FLT3-ITD positive AML and cooperates with FLT3-ITD to activate MYC transcriptional programs. FLT3-ITD AML cells are more vulnerable to SYK suppression than FLT3 wild-type counterparts. In a FLT3-ITD in vivo model, SYK is indispensable for myeloproliferative disease (MPD) development, and SYK overexpression promotes overt transformation to AML and resistance to FLT3-ITD-targeted therapy.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Apoptosis
- Blotting, Western
- Cell Proliferation
- Cell Transformation, Neoplastic
- Cells, Cultured
- Drug Resistance, Neoplasm
- Fluorouracil/pharmacology
- Humans
- Immunoenzyme Techniques
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred BALB C
- Mutation/genetics
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Syk Kinase
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Alexandre Puissant
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Nina Fenouille
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA; The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Bioinformatics Graduate Program, Boston University, Boston, MA 02215, USA
| | - Yana Pikman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Christopher F Bassil
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Swapnil Mehta
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Jinyan Du
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Julhash U Kazi
- Experimental Clinical Chemistry, Department of Laboratory Medicine, Lund University, Medicon Village, 221 00 Lund, Sweden
| | - Frédéric Luciano
- C3M/ INSERM U1065 Team Cell Death, Differentiation, Inflammation and Cancer, 06204 Nice, France
| | - Lars Rönnstrand
- Experimental Clinical Chemistry, Department of Laboratory Medicine, Lund University, Medicon Village, 221 00 Lund, Sweden
| | - Andrew L Kung
- Pediatric Department, Columbia University Medical Center, New York, NY 10032, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Ilene Galinsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Michael T Hemann
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA; The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
1279
|
El Ghannam D, Taalab MM, Ghazy HF, Eneen AF. DNMT3A R882 mutations in patients with cytogenetically normal acute myeloid leukemia and myelodysplastic syndrome. Blood Cells Mol Dis 2014; 53:61-6. [PMID: 24512939 DOI: 10.1016/j.bcmd.2014.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 01/07/2014] [Accepted: 01/07/2014] [Indexed: 01/22/2023]
Abstract
Several molecular markers have been described that help to classify patients with acute myeloid leukemia (AML), a heterogeneous hematopoietic tissue neoplasm, into risk groups. We determined the frequency of DNMT3A mutations, their associations with clinical and molecular characteristics and outcome, in primary, cytogenetically-normal AML (CN-AML) and CN-myelodysplastic syndrome (MDS). A total of 63 CN-AML and 16 CN-MDS patients were analyzed for mutations in DNMT3A, codon R822 by direct sequencing and mutation of NPM1 and FLT3/ITD. DNMT3A mutations were found in 17/63 (27%) of CN-AML and in 1/16 (6.3%) of CN-MDS patients. Patients with DNMT3A mutations were older (p=0.047), had higher white blood cell (WBC) counts (p=0.046), more often belonged to FAB groups M4 and M5 (p=0.017), and were more associated with NPM1 mutations (p=0.017), than those with wild-type DNMT3A. DNMT3A-mutated patients had shorter overall disease survival (p<0.001) and disease-free survival (p=0.014) when the entire patient cohort was considered, which remained significant in multivariate analysis. We conclude that DNMT3A R882 mutations are recurrent molecular aberrations in CN-AML, less frequent in CN-MDS, and that testing for R882 mutations may provide a useful tool for refining risk classification of CN-AML.
Collapse
Affiliation(s)
- Doaa El Ghannam
- Departments of Clinical Pathology, Faculty of Medicine, Mansoura University, Egypt.
| | - Mona M Taalab
- Clinical Hematology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hayam F Ghazy
- Medical Oncology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Asmaa F Eneen
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
1280
|
Mannelli F, Cutini I, Gianfaldoni G, Bencini S, Scappini B, Pancani F, Ponziani V, Bonetti MI, Biagiotti C, Longo G, Bosi A. CXCR4 expression accounts for clinical phenotype and outcome in acute myeloid leukemia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2014; 86:340-9. [DOI: 10.1002/cyto.b.21156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/27/2013] [Accepted: 01/03/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Francesco Mannelli
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Ilaria Cutini
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Giacomo Gianfaldoni
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Sara Bencini
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Barbara Scappini
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Fabiana Pancani
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Vanessa Ponziani
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Maria Ida Bonetti
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Caterina Biagiotti
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Giovanni Longo
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| | - Alberto Bosi
- UF di Ematologia, Dipartimento di Medicina Sperimentale e Clinica; Università degli Studi, Firenze, and Istituto Toscano Tumori; Florence Italy
| |
Collapse
|
1281
|
DiNardo CD, Patel KP, Garcia-Manero G, Luthra R, Pierce S, Borthakur G, Jabbour E, Kadia T, Pemmaraju N, Konopleva M, Faderl S, Cortes J, Kantarjian HM, Ravandi F. Lack of association of IDH1, IDH2 and DNMT3A mutations with outcome in older patients with acute myeloid leukemia treated with hypomethylating agents. Leuk Lymphoma 2014; 55:1925-9. [PMID: 24138309 DOI: 10.3109/10428194.2013.855309] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Courtney D DiNardo
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center , Houston, TX , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1282
|
Tabarroki A, Tiu RV. Molecular genetics of myelofibrosis and its associated disease phenotypes. Transl Med UniSa 2014; 8:53-64. [PMID: 24778998 PMCID: PMC4000463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/04/2014] [Indexed: 11/06/2022] Open
Abstract
In 2005, the discovery of Janus kinase 2 (JAK2) V617F mutation in approximately half of patients with myelofibrosis (MF) marked an important milestone in our understanding of the pathophysiology of MF. This has broadened our understanding of the disease pathogenesis and became the foundation for the development and subsequent clinical use of JAK inhibitors for MF. However, it is clear that other pathogenetic modifiers contribute to the disease diversity and phenotypic variability of MF. Novel genome scanning technologies were useful in the identification of recurrent molecular mutations in other genes including MPL, TET2, IDH1/2, DNMT3A, SH3B2 (LNK) and CBL in MF pointing out that other pathways might be important in addition to the JAK/STAT pathway. The biologic role and clinical implications of these molecular mutations in MF is currently under investigation. The main challenge is to understand the mechanisms whereby molecular mutations whether alone or in combination with other genetic and non-genetic events contribute to the pathogenesis of MF and eventually can explain the phenotypic variability among the MF patients. In the present review we will provide an overview of the molecular pathogenesis of MF describing past and recent discoveries in molecular markers and their possible relevance in disease phenotype.
Collapse
Affiliation(s)
- Ali Tabarroki
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland OH, USA
| | - Ramon V. Tiu
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland OH, USA,Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland OH, USA,
| |
Collapse
|
1283
|
Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia 2014; 28:1586-95. [PMID: 24487413 DOI: 10.1038/leu.2014.55] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 12/12/2022]
Abstract
To clarify the cooperative roles of recurrently identified mutations and to establish a more precise risk classification system in acute myeloid leukemia (AML), we comprehensively analyzed mutations in 51 genes, as well as cytogenetics and 11 chimeric transcripts, in 197 adult patients with de novo AML who were registered in the Japan Adult Leukemia Study Group AML201 study. We identified a total of 505 mutations in 44 genes, while only five genes, FLT3, NPM1, CEBPA, DNMT3A and KIT, were mutated in more than 10% of the patients. Although several cooperative and exclusive mutation patterns were observed, the accumulated mutation number was higher in cytogenetically normal AML and lower in AML with RUNX1-RUNX1T1 and CBFB-MYH11, indicating a strong potential of these translocations for the initiation of AML. Furthermore, we evaluated the prognostic impacts of each sole mutation and the combinations of mutations and/or cytogenetics, and demonstrated that AML patients could be clearly stratified into five risk groups for overall survival by including the mutation status of DNMT3A, MLL-PTD and TP53 genes in the risk classification system of the European LeukemiaNet. These results indicate that the prognosis of AML could be stratified by the major mutation status in combination with cytogenetics.
Collapse
|
1284
|
Janin M, Mylonas E, Saada V, Micol JB, Renneville A, Quivoron C, Koscielny S, Scourzic L, Forget S, Pautas C, Caillot D, Preudhomme C, Dombret H, Berthon C, Barouki R, Rabier D, Auger N, Griscelli F, Chachaty E, Leclercq E, Courtier MH, Bennaceur-Griscelli A, Solary E, Bernard OA, Penard-Lacronique V, Ottolenghi C, de Botton S. Serum 2-Hydroxyglutarate Production in IDH1- and IDH2-Mutated De Novo Acute Myeloid Leukemia: A Study by the Acute Leukemia French Association Group. J Clin Oncol 2014; 32:297-305. [DOI: 10.1200/jco.2013.50.2047] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Purpose Mutated isocitrate dehydrogenases (IDHs) 1 and 2 produce high levels of 2-hydroxyglutarate (2-HG). We investigated whether, in acute myeloid leukemia (AML), serum 2-HG would predict the presence of IDH1/2 mutations at diagnosis and provide a marker of minimal residual disease (MRD). Patients and Methods Serum samples from 82 patients at diagnosis of de novo AML (IDH1/2 mutated, n = 53) and 68 patients without AML were analyzed for total 2-HG and its ratio of D to L stereoisomers by mass spectrometry. We measured 2-HG levels and molecular markers of MRD (WT1 and NPM1) in serial samples of 36 patients with IDH1/2 mutations after induction therapy. Results In patients with AML with IDH1/2 mutations, 2-HG serum levels were significantly higher than in patients with IDH1/2 wild type (P < .001). Area under the receiver operating characteristic curve was 99%. The optimum diagnostic cutoff between IDH1/2 mutated and normal was 2 μmol/L (sensitivity, 100%; specificity, 79%). Quantification of the D/L stereoisomers increased specificity (100%; 95% CI, 83% to 100%) compared with total 2-HG (P = .031). In patients with IDH2 R172 mutations, 2-HG levels were higher relative to those with other IDH1/2 mutations (P < .05). During follow-up, serum 2-HG levels showed strong positive correlation with WT1 and NPM1 (P < .001). After induction therapy, total 2-HG serum levels < 2 μmol/L were associated with better overall (P = .008) and disease-free survival (P = .005). Conclusion Serum 2-HG is a predictor of the presence of IDH1/2 mutations and outcome in these patients. Discrimination between D/L stereoisomers improved specificity.
Collapse
Affiliation(s)
- Maxime Janin
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Elena Mylonas
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Véronique Saada
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Jean-Baptiste Micol
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Aline Renneville
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Cyril Quivoron
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Serge Koscielny
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Laurianne Scourzic
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Sébastien Forget
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Cécile Pautas
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Denis Caillot
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Claude Preudhomme
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Hervé Dombret
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Céline Berthon
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Robert Barouki
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Daniel Rabier
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Nathalie Auger
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Frank Griscelli
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Elisabeth Chachaty
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Edwige Leclercq
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Marie-Hélène Courtier
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Annelise Bennaceur-Griscelli
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Eric Solary
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Olivier Adrien Bernard
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Virginie Penard-Lacronique
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Chris Ottolenghi
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| | - Stéphane de Botton
- Maxime Janin, Robert Barouki, Daniel Rabier, and Chris Ottolenghi, Biochimie Métabolique, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Necker-Enfants Malades; Maxime Janin, Robert Barouki, and Chris Ottolenghi, Institut National de la Santé et de la Recherche Médicale (INSERM) U747, Université Paris Descartes; Hervé Dombret, Hôpital Saint-Louis, Paris; Elena Mylonas, Cyril Quivoron, Laurianne Scourzic, Olivier Adrien Bernard, Virginie Penard-Lacronique, and Stéphane de Botton, INSERM U985,
| |
Collapse
|
1285
|
Liersch R, Müller-Tidow C, Berdel WE, Krug U. Prognostic factors for acute myeloid leukaemia in adults - biological significance and clinical use. Br J Haematol 2014; 165:17-38. [DOI: 10.1111/bjh.12750] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Ruediger Liersch
- Department of Haematology and Oncology; Internal Medicine III; Clemenshospital Muenster; Muenster Germany
| | - Carsten Müller-Tidow
- Department of Medicine A - Haematology and Oncology; University Hospital of Muenster; Muenster Germany
| | - Wolfgang E. Berdel
- Department of Medicine A - Haematology and Oncology; University Hospital of Muenster; Muenster Germany
| | - Utz Krug
- Department of Medicine A - Haematology and Oncology; University Hospital of Muenster; Muenster Germany
| |
Collapse
|
1286
|
Dawson MA, Gudgin EJ, Horton SJ, Giotopoulos G, Meduri E, Robson S, Cannizzaro E, Osaki H, Wiese M, Putwain S, Fong CY, Grove C, Craig J, Dittmann A, Lugo D, Jeffrey P, Drewes G, Lee K, Bullinger L, Prinjha RK, Kouzarides T, Vassiliou GS, Huntly BJP. Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 2014; 28:311-20. [PMID: 24220271 PMCID: PMC3918873 DOI: 10.1038/leu.2013.338] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 11/25/2022]
Abstract
Recent evidence suggests that inhibition of bromodomain and extra-terminal (BET) epigenetic readers may have clinical utility against acute myeloid leukemia (AML). Here we validate this hypothesis, demonstrating the efficacy of the BET inhibitor I-BET151 across a variety of AML subtypes driven by disparate mutations. We demonstrate that a common 'core' transcriptional program, which is HOX gene independent, is downregulated in AML and underlies sensitivity to I-BET treatment. This program is enriched for genes that contain 'super-enhancers', recently described regulatory elements postulated to control key oncogenic driver genes. Moreover, our program can independently classify AML patients into distinct cytogenetic and molecular subgroups, suggesting that it contains biomarkers of sensitivity and response. We focus AML with mutations of the Nucleophosmin gene (NPM1) and show evidence to suggest that wild-type NPM1 has an inhibitory influence on BRD4 that is relieved upon NPM1c mutation and cytosplasmic dislocation. This leads to the upregulation of the core transcriptional program facilitating leukemia development. This program is abrogated by I-BET therapy and by nuclear restoration of NPM1. Finally, we demonstrate the efficacy of I-BET151 in a unique murine model and in primary patient samples of NPM1c AML. Taken together, our data support the use of BET inhibitors in clinical trials in AML.
Collapse
MESH Headings
- Animals
- Benzodiazepines/administration & dosage
- Benzodiazepines/pharmacology
- Cell Cycle Proteins
- Cell Line, Tumor
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Mice
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nucleophosmin
- Transcription Factors/metabolism
- Transcription, Genetic
- Transcriptional Activation
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- M A Dawson
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge UK
| | - E J Gudgin
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - S J Horton
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - G Giotopoulos
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - E Meduri
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - S Robson
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge UK
| | - E Cannizzaro
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge UK
| | - H Osaki
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - M Wiese
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge UK
| | - S Putwain
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - C Y Fong
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge UK
| | - C Grove
- Haematological Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - J Craig
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - A Dittmann
- Discovery Research, Cellzome AG, Heidelberg, Germany
| | - D Lugo
- Epinova DPU, Immuno-Inflammation Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - P Jeffrey
- Epinova DPU, Immuno-Inflammation Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - G Drewes
- Discovery Research, Cellzome AG, Heidelberg, Germany
| | - K Lee
- Epinova DPU, Immuno-Inflammation Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - L Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - R K Prinjha
- Epinova DPU, Immuno-Inflammation Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - T Kouzarides
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge UK
| | - G S Vassiliou
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Haematological Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - B J P Huntly
- Department of Haematology, Cambridge Institute for Medical Research and Addenbrookes Hospital, University of Cambridge, Cambridge, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| |
Collapse
|
1287
|
Prognostic significance of flow cytometric residual disease, dysregulated neutrophils/monocytes, and hematogones in adult acute myeloid leukemia in first remission. Int J Hematol 2014; 99:296-304. [PMID: 24481944 DOI: 10.1007/s12185-014-1525-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
Abstract
Fifty-one consecutive non-M3 acute myeloid leukemia (AML) patients who had achieved morphologic complete remission (mCR) after induction chemotherapy were enrolled in the present study. Three characteristics of bone marrow (BM) composition analyzed by flow cytometry were combined to determine the prognostic impact. A standardized panel of reagents was used to detect residual disease of aberrant myeloid progenitor cells (RD), identify neutrophils/monocytes with dysregulated immunophenotype (dysregulated neutro/mono) and quantify the appearance of CD34(+) B-progenitor-related cluster (hematogones) simultaneously in post-induction BM of adult AML patients. Patients who had detectable RD ≥0.2 % exhibited significantly lower median leukemia-free survival (LFS) than those who did not (13.5 vs. 48.0 months; P = 0.042). Dysregulated neutro/mono abnormalities assessed by this flow cytometric scoring system (FCSS ≥2) predicted shorter LFS (8.0 vs. 39.0 months; P = 0.008). While B-progenitor-related cluster size ≥5 % predicted improved outcome, with longer LFS (not reached vs. 13.5 months; P = 0.023) and better overall survival (not reached vs. 24.0 months; P = 0.027). The proposed RD/dysregulated neutro/mono/hematogones score showed a new risk groups with different LFS in the overall patients (P = 0.0006) as well as in the subgroup of intermediate cytogenetic risk (P = 0.001). The RD/dysregulated neutro/mono/hematogones score assessed by flow cytometry for adult AML in mCR may offer a rapid and practical risk assessment providing better refinement in risk-adapted management after induction chemotherapy.
Collapse
|
1288
|
Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood 2014; 123:2220-8. [PMID: 24478400 DOI: 10.1182/blood-2013-11-537167] [Citation(s) in RCA: 457] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a group of clonal disorders characterized by aberrant hematopoietic proliferation and an increased tendency toward leukemic transformation. We used targeted next-generation sequencing (NGS) of 104 genes to detect somatic mutations in a cohort of 197 MPN patients and followed clonal evolution and the impact on clinical outcome. Mutations in calreticulin (CALR) were detected using a sensitive allele-specific polymerase chain reaction. We observed somatic mutations in 90% of patients, and 37% carried somatic mutations other than JAK2 V617F and CALR. The presence of 2 or more somatic mutations significantly reduced overall survival and increased the risk of transformation into acute myeloid leukemia. In particular, somatic mutations with loss of heterozygosity in TP53 were strongly associated with leukemic transformation. We used NGS to follow and quantitate somatic mutations in serial samples from MPN patients. Surprisingly, the number of mutations between early and late patient samples did not significantly change, and during a total follow-up of 133 patient years, only 2 new mutations appeared, suggesting that the mutation rate in MPN is rather low. Our data show that comprehensive mutational screening at diagnosis and during follow-up has considerable potential to identify patients at high risk of disease progression.
Collapse
|
1289
|
White BS, DiPersio JF. Genomic tools in acute myeloid leukemia: From the bench to the bedside. Cancer 2014; 120:1134-44. [PMID: 24474533 DOI: 10.1002/cncr.28552] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 11/14/2013] [Indexed: 12/28/2022]
Abstract
Since its use in the initial characterization of an acute myeloid leukemia (AML) genome, next-generation sequencing (NGS) has continued to molecularly refine the disease. Here, the authors review the spectrum of NGS applications that have subsequently delineated the prognostic significance and biologic consequences of these mutations. Furthermore, the role of this technology in providing a high-resolution glimpse of AML clonal heterogeneity, which may inform future choice of targeted therapy, is discussed. Although obstacles remain in applying these techniques clinically, they have already had an impact on patient care.
Collapse
Affiliation(s)
- Brian S White
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri; The Genome Institute, Washington University, St. Louis, Missouri
| | | |
Collapse
|
1290
|
Damaj G, Joris M, Chandesris O, Hanssens K, Soucie E, Canioni D, Kolb B, Durieu I, Gyan E, Livideanu C, Chèze S, Diouf M, Garidi R, Georgin-Lavialle S, Asnafi V, Lhermitte L, Lavigne C, Launay D, Arock M, Lortholary O, Dubreuil P, Hermine O. ASXL1 but not TET2 mutations adversely impact overall survival of patients suffering systemic mastocytosis with associated clonal hematologic non-mast-cell diseases. PLoS One 2014; 9:e85362. [PMID: 24465546 PMCID: PMC3897447 DOI: 10.1371/journal.pone.0085362] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 11/25/2013] [Indexed: 01/08/2023] Open
Abstract
Systemic mastocytosis with associated hematologic clonal non-mast cell disease (SM-AHNMD) is a rare and heterogeneous subtype of SM and few studies on this specific entity have been reported. Sixty two patients with Systemic mastocytosis with associated hematologic clonal non-mast cell disease (SM-AHNMD) were presented. Myeloid AHNMD was the most frequent (82%) cases. This subset of patients were older, had more cutaneous lesions, splenomegaly, liver enlargement, ascites; lower bone mineral density and hemoglobin levels and higher tryptase level than lymphoid AHNMD. Defects in KIT, TET2, ASXL1 and CBL were positive in 87%, 27%, 14%, and 11% of cases respectively. The overall survival of patients with SM-AHNMD was 85.2 months. Within the myeloid group, SM-MPN fared better than SM-MDS or SM-AML (p = 0.044,). In univariate analysis, the presence of C-findings, the AHNMD subtypes (SM-MDS/CMML/AML versus SM-MPN/hypereosinophilia) (p = 0.044), Neutropenia (p = 0.015), high monocyte level (p = 0.015) and the presence of ASXL1 mutation had detrimental effects on OS (p = 0.007). In multivariate analysis and penalized Cox model, only the presence of ASXL1 mutation remained an independent prognostic factor that negatively affected OS (p = 0.035). SM-AHNMD is heterogeneous with variable prognosis according to the type of the AHNMD. ASXL1 is mutated in a subset of myeloid AHNMD and adversely impact on OS.
Collapse
Affiliation(s)
- Gandhi Damaj
- Service d'Hématologie, Centre Hospitalier Universitaire, Hôpital Sud; Amiens, France
- Centre de Référence des Mastocytoses, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
- CNRS UMR 8147 and Institut Imagine, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
- * E-mail:
| | - Magalie Joris
- Service d'Hématologie, Centre Hospitalier Universitaire, Hôpital Sud; Amiens, France
| | - Olivia Chandesris
- Centre de Référence des Mastocytoses, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
- Service d'Hématologie Adulte, Université Paris Descartes, Paris Sorbonne Cité, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
| | - Katia Hanssens
- Inserm, U1068, CRCM, (Signaling, Hematopoiesis and Mechanism of Oncogenesis); Institut Paoli-Calmettes,Marseille; Aix-Marseille Univ; CNRS, UMR7258, Marseille, France
| | - Erinn Soucie
- Inserm, U1068, CRCM, (Signaling, Hematopoiesis and Mechanism of Oncogenesis); Institut Paoli-Calmettes,Marseille; Aix-Marseille Univ; CNRS, UMR7258, Marseille, France
| | - Danielle Canioni
- Service d'Anatomo-pathologie, Université Paris Descartes, Paris Sorbonne Cité, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
| | - Brigitte Kolb
- Service d'Hématologie, Centre Hospitalier Universitaire, Reims, France
| | - Isabelle Durieu
- Service de médecine interne, Groupe Hospitalier Sud. Hospices Civils, Lyon, France
| | - Emanuel Gyan
- Service d'Hématologie et thérapie cellulaire, CIC INSERMU202, Centre Hospitalier Universitaire, Tours, France
| | - Cristina Livideanu
- Département de Dermatologie, Centre Hospitalier Universitaire, Toulouse, France
| | - Stephane Chèze
- Service d'Hématologie, Centre Hospitalier Universitaire, Caen, France
| | - Momar Diouf
- Département de bio-statistiques et de Recherche clinique, Centre Hospitalier Universitaire, Amiens, France
| | - Reda Garidi
- Service d'Hématologie, Centre Hospitalier, St Quentin, France
| | - Sophie Georgin-Lavialle
- Service de Médecine Interne, Hôpital Tenon, Assistance Publique-Hôpitaux, Université Pierre et Marie Curie, Paris, France
| | - Vahid Asnafi
- Laboratoire d'hématologie Biologique et UMR CNRS 8147, Université Paris Descartes, Paris Sorbonne Cité, Faculté de Médecine et Assistance Publique-Hôpitaux de Paris (AP-HP) Necker-Enfants Malades, Paris, France
| | - Ludovic Lhermitte
- Laboratoire d'hématologie Biologique et UMR CNRS 8147, Université Paris Descartes, Paris Sorbonne Cité, Faculté de Médecine et Assistance Publique-Hôpitaux de Paris (AP-HP) Necker-Enfants Malades, Paris, France
| | - Christian Lavigne
- Service d'Hématologie, Centre Hospitalier Universitaire, Angers, France
| | - David Launay
- Service de Médecine Interne, CHRU, Lille, France
| | - Michel Arock
- CNRS UMR 8113, Laboratoire de Biologie et Pharmacologie Appliquée, Ecole Normale Supérieure, Cachan, France
- Laboratoire Central d'Hématologie, Groupe Hospitalier Pitié-Salpetrière, Paris, France
| | - Olivier Lortholary
- Service de Médecine Interne et de Maladie Infectieuses, Université Paris Descartes, Paris Sorbonne Cité, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
| | - Patrice Dubreuil
- Inserm, U1068, CRCM, (Signaling, Hematopoiesis and Mechanism of Oncogenesis); Institut Paoli-Calmettes,Marseille; Aix-Marseille Univ; CNRS, UMR7258, Marseille, France
| | - Olivier Hermine
- Centre de Référence des Mastocytoses, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
- CNRS UMR 8147 and Institut Imagine, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
- Service d'Hématologie Adulte, Université Paris Descartes, Paris Sorbonne Cité, Faculté de Médecine et AP-HP Necker-Enfants Malades, Paris, France
| |
Collapse
|
1291
|
Kats LM, Reschke M, Taulli R, Pozdnyakova O, Burgess K, Bhargava P, Straley K, Karnik R, Meissner A, Small D, Su SM, Yen K, Zhang J, Pandolfi PP. Proto-oncogenic role of mutant IDH2 in leukemia initiation and maintenance. Cell Stem Cell 2014; 14:329-41. [PMID: 24440599 DOI: 10.1016/j.stem.2013.12.016] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 11/13/2013] [Accepted: 12/24/2013] [Indexed: 01/28/2023]
Abstract
Mutations in the metabolic enzymes isocitrate dehydrogenase-1 (IDH1) and IDH2 that produce the oncometabolite D-2-hydroxyglutarate (2-HG) occur frequently in human acute myeloid leukemia (AML). 2-HG modulates numerous biological pathways implicated in malignant transformation, but the contribution of mutant IDH proteins to maintenance and progression of AML in vivo is currently unknown. To answer this crucial question we have generated transgenic mice that express IDH2(R140Q) in an on/off- and tissue-specific manner using a tetracycline-inducible system. We found that IDH2(R140Q) can cooperate with overexpression of HoxA9 and Meis1a and with mutations in FMS-like tyrosine kinase 3 (FLT3) to drive acute leukemia in vivo. Critically, we show that genetic deinduction of mutant IDH2 in leukemic cells in vivo has profound effects on their growth and/or maintenance. Our data demonstrate the proto-oncogenic role of mutant IDH2 and support its relevance as a therapeutic target for the treatment of human AML.
Collapse
Affiliation(s)
- Lev M Kats
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Markus Reschke
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Riccardo Taulli
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Olga Pozdnyakova
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Kerri Burgess
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Parul Bhargava
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Rahul Karnik
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alexander Meissner
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Donald Small
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | - Jiangwen Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
1292
|
Fu L, Huang W, Jing Y, Jiang M, Zhao Y, Shi J, Huang S, Xue X, Zhang Q, Tang J, Dou L, Wang L, Nervi C, Li Y, Yu L. AML1-ETO triggers epigenetic activation of early growth response gene l, inducing apoptosis in t(8;21) acute myeloid leukemia. FEBS J 2014; 281:1123-31. [PMID: 24314118 DOI: 10.1111/febs.12673] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 11/06/2013] [Accepted: 12/02/2013] [Indexed: 11/30/2022]
Abstract
The t(8;21)(q22;q22) translocation is the most common chromosomal translocation in acute myeloid leukemia (AML), and it gives rise to acute myeloid gene 1 (AML1)-myeloid transforming gene 8 (ETO)-positive AML, which has a relatively favorable prognosis. However, the molecular mechanism related to a favorable prognosis in AML1-ETO-positive AML is still not fully understood. Our results show that the AML1-ETO fusion protein triggered activation of early growth response gene l (EGR1) by binding at AML1-binding sites on the EGR1 promoter and, subsequently, recruiting acetyltransferase P300, which is known to acetylate histones. However, AML1-ETO could not recruit DNA methyltransferases and histone deacetylases; therefore, EGR1 expression was affected by histone acetylation but not by DNA methylation. Both transcription and translation of EGR1 were higher in AML1-ETO-positive AML cell lines than in AML1-ETO-negative AML cell lines, owing to acetylation. Furthermore, when AML1-ETO-positive AML cell lines were treated with C646 (P300 inhibitor) and trichostatin A (histone deacetylase inhibitor), EGR1 expression was significantly decreased and increased, respectively. In addition, treatment with 5-azacytidine (methyltransferase inhibitor) did not cause any significant change in EGR1 expression. Overexpression of EGR1 inhibited cell proliferation and promoted apoptosis, and EGR1 knockout promoted cell proliferation. Thus, EGR1 could be a novel prognostic factor for a favorable outcome in AML1-ETO-positive AML. The results of our study may explain the molecular mechanisms underlying the favorable prognosis in AML1-ETO-positive AML.
Collapse
Affiliation(s)
- Lin Fu
- Department of Hematology, Chinese PLA General Hospital, Beijing, China; Nankai University School of Medicine, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1293
|
Affiliation(s)
- Joanne Mason
- West Midlands Regional Genetics Laboratory, Birmingham Women’s NHS Foundation Trust, Edgbaston, Birmingham, B15 2TG, UK
| | - Michael Griffiths
- West Midlands Regional Genetics Laboratory, Birmingham Women’s NHS Foundation Trust, Edgbaston, Birmingham, B15 2TG, UK
| |
Collapse
|
1294
|
Jeong M, Sun D, Luo M, Huang Y, Challen GA, Rodriguez B, Zhang X, Chavez L, Wang H, Hannah R, Kim SB, Yang L, Ko M, Chen R, Göttgens B, Lee JS, Gunaratne P, Godley LA, Darlington GJ, Rao A, Li W, Goodell MA. Large conserved domains of low DNA methylation maintained by Dnmt3a. Nat Genet 2014; 46:17-23. [PMID: 24270360 PMCID: PMC3920905 DOI: 10.1038/ng.2836] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/01/2013] [Indexed: 02/07/2023]
Abstract
Gains and losses in DNA methylation are prominent features of mammalian cell types. To gain insight into the mechanisms that promote shifts in DNA methylation and contribute to changes in cell fate, including malignant transformation, we performed genome-wide mapping of 5-methylcytosine and 5-hydroxymethylcytosine in purified mouse hematopoietic stem cells. We discovered extended regions of low methylation (canyons) that span conserved domains frequently containing transcription factors and are distinct from CpG islands and shores. About half of the genes in these methylation canyons are coated with repressive histone marks, whereas the remainder are covered by activating histone marks and are highly expressed in hematopoietic stem cells (HSCs). Canyon borders are demarked by 5-hydroxymethylcytosine and become eroded in the absence of DNA methyltransferase 3a (Dnmt3a). Genes dysregulated in human leukemias are enriched for canyon-associated genes. The new epigenetic landscape we describe may provide a mechanism for the regulation of hematopoiesis and may contribute to leukemia development.
Collapse
Affiliation(s)
- Mira Jeong
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Deqiang Sun
- Division of Biostatistics, Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Min Luo
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yun Huang
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Grant A. Challen
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin Rodriguez
- Division of Biostatistics, Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Xiaotian Zhang
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Lukas Chavez
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Hui Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Rebecca Hannah
- Department of Hematology, Cambridge Institute for Medical Research and Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge University, Hills Road, Cambridge, UK
| | - Sang-Bae Kim
- Department of Systems Biology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Liubin Yang
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Myunggon Ko
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Rui Chen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Berthold Göttgens
- Department of Hematology, Cambridge Institute for Medical Research and Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge University, Hills Road, Cambridge, UK
| | - Ju-Seog Lee
- Department of Systems Biology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Preethi Gunaratne
- Department of Pathology, Baylor College of Medicine, and Department of Biology & Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Lucy A. Godley
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Wei Li
- Division of Biostatistics, Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Margaret A. Goodell
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
1295
|
Ahmad F, Mohota R, Sanap S, Mandava S, Das BR. Molecular evaluation of DNMT3A and IDH1/2 gene mutation: frequency, distribution pattern and associations with additional molecular markers in normal karyotype Indian acute myeloid leukemia patients. Asian Pac J Cancer Prev 2014; 15:1247-1253. [PMID: 24606448 DOI: 10.7314/apjcp.2014.15.3.1247] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
Mutations in the DNMT3A and IDH genes represent the most common genetic alteration after FLT3/NPM1 in acute myeloid leukemia (AML). We here analyzed the frequency and distribution pattern of DNMT3A and IDH mutations and their associations with other molecular markers in normal karyotype AML patients. Forty- five patients were screened for mutations in DNMT3A (R882), IDH1 (R132) and IDH2 (R140 and R172) genes by direct sequencing. Of the 45 patients screened, DNMT3A and IDH mutations were observed in 6 (13.3%) and 7 (15.4%), respectively. Patients with isolated DNMT3A mutations were seen in 4 cases (9%), isolated IDH mutations in 5 (11.1%), while interestingly, two cases showed both DNMT3A and IDH mutations (4.3%). Nucleotide sequencing of DNMT3A revealed missense mutations (R882H and R882C), while that of IDH revealed R172K, R140Q, R132H and R132S. Both DNMT3A and IDH mutations were observed only in adults, with a higher frequency in males. DNMT3A and IDH mutations were significantly associated with NPM1, while trends towards higher coexistence with FLT3 mutations were observed. This is the first study to evaluate DNMT3A/ IDH mutations in Indian patients. Significant associations among the various molecular markers was observed, that highlights cooperation between them and possible roles in improved risk stratification.
Collapse
Affiliation(s)
- Firoz Ahmad
- Research and Development, SRL Ltd, Mumbai, India E-mail :
| | | | | | | | | |
Collapse
|
1296
|
Ok CY, Hasserjian RP, Fox PS, Stingo F, Zuo Z, Young KH, Patel K, Medeiros LJ, Garcia-Manero G, Wang SA. Application of the international prognostic scoring system-revised in therapy-related myelodysplastic syndromes and oligoblastic acute myeloid leukemia. Leukemia 2014; 28:185-9. [PMID: 23787392 DOI: 10.1038/leu.2013.191] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- C Y Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R P Hasserjian
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - P S Fox
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - F Stingo
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Z Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L J Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
1297
|
|
1298
|
A new assay to identify recurrent mutations in acute myeloid leukemia using next-generation sequencing. REV ROMANA MED LAB 2014. [DOI: 10.2478/rrlm-2014-0003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
1299
|
Wertheim GBW, Smith C, Figueroa ME, Kalos M, Bagg A, Carroll M, Master SR. Microsphere-based multiplex analysis of DNA methylation in acute myeloid leukemia. J Mol Diagn 2013; 16:207-15. [PMID: 24373919 DOI: 10.1016/j.jmoldx.2013.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 01/19/2023] Open
Abstract
Aberrant regulation of DNA methylation is characteristic of cancer cells and clearly influences phenotypes of various malignancies. Despite clear correlations between DNA methylation and patient outcome, tests that directly measure multiple-locus DNA methylation are typically expensive and technically challenging. Previous studies have demonstrated that the prognosis of patients with acute myeloid leukemia can be predicted by the DNA methylation pattern of 18 loci. We have developed a novel strategy, termed microsphere HpaII tiny fragment enrichment by ligation-mediated PCR (MELP), to simultaneously analyze the DNA methylation pattern at these loci using methylation-specific DNA digestion, fluorescently labeled microspheres, and branched DNA hybridization. The method uses techniques that are inexpensive and easily performed in a molecular laboratory. MELP accurately reflects the methylation levels at each locus analyzed and segregates patients with acute myeloid leukemia into prognostic subgroups. Our results demonstrate the usefulness of MELP as a platform for simultaneous evaluation of DNA methylation of multiple loci.
Collapse
Affiliation(s)
- Gerald B W Wertheim
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Catherine Smith
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Maria E Figueroa
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Michael Kalos
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin Carroll
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen R Master
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
1300
|
Elias HK, Schinke C, Bhattacharyya S, Will B, Verma A, Steidl U. Stem cell origin of myelodysplastic syndromes. Oncogene 2013; 33:5139-50. [PMID: 24336326 DOI: 10.1038/onc.2013.520] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/17/2013] [Accepted: 10/17/2013] [Indexed: 01/18/2023]
Abstract
Myelodysplastic syndromes (MDS) are common hematologic disorders that are characterized by decreased blood counts due to ineffective hematopoiesis. MDS is considered a 'preleukemic' disorder linked to a significantly elevated risk of developing an overt acute leukemia. Cytopenias can be observed in all three myeloid lineages suggesting the involvement of multipotent, immature hematopoietic cells in the pathophysiology of this disease. Recent studies using murine models of MDS as well as primary patient-derived bone marrow samples have provided direct evidence that the most immature, self-renewing hematopoietic stem cells (HSC), as well as lineage-committed progenitor cells, are critically altered in patients with MDS. Besides significant changes in the number and distribution of stem as well as immature progenitor cells, genetic and epigenetic aberrations have been identified, which confer functional changes to these aberrant stem cells, impairing their ability to proliferate and differentiate. Most importantly, aberrant stem cells can persist and further expand after treatment, even upon transient achievement of clinical complete remission, pointing to a critical role of these cells in disease relapse. Ongoing preclinical and clinical studies are particularly focusing on the precise molecular and functional characterization of aberrant MDS stem cells in response to therapy, with the goal to develop stem cell-targeted strategies for therapy and disease monitoring that will allow for achievement of longer-lasting remissions in MDS.
Collapse
Affiliation(s)
- H K Elias
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - C Schinke
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - S Bhattacharyya
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - B Will
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - A Verma
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| | - U Steidl
- 1] Albert Einstein College of Medicine, Albert Einstein Cancer Center, New York, NY, USA [2] Departments of Cell Biology and Developmental and Molecular Biology, New York, NY, USA [3] Division of Hematologic Malignancies, Department of Medicine (Oncology), New York, NY, USA [4] Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Chanin Institute for Cancer Research, New York, NY, USA
| |
Collapse
|