Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Eur J Haematol ; 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39129130

ABSTRACT

INTRODUCTION: Elderly acute myeloid leukemia (AML) patients with poor-risk cytogenetics have a poor outcome with intensive chemotherapy (IC). While Venetoclax (VEN) has changed the outcomes of elderly unfit patients treatment, it is unknown whether it could be effective in poor-risk cytogenetics 60-75 years old patients. MATERIALS AND METHODS: We included 60-75-year-old AML patients eligible to allogenic stem cell transplantation (allo-SCT) treated with VEN (combined with azacitidine or with Cladribin and Aracytine) at Institut Paoli Calmettes, between 2020 and 2023 and compared this cohort with patients treated by IC between 2010 and 2019. RESULTS: Twenty six patients were treated with VEN (17 in combination with azacitidine and 9 with Cladribin and Aracytine) and 90 were treated with IC. Thirteen patients (50%) had a TP53 mutation. The median time for leucocyte and platelet counts recovery was 26 days (range 0-103) and 26 days (range, 0-63). The median duration of the first hospitalization was 32 days (ranges, 7-79). The composite response rate was 69% (CR = 50%, CRi = 4%, MLFS = 15%). Allo-SCT could be performed in 42% of cases. Median overall survival (OS) was 7.9 months (20.9 months in the group of patients who transitioned to allo-SCT). We found no difference with the historical cohort of patients treated with IC except a trend toward less lower and upper tract gastro-intestinal (GI) tract infections in the VEN group (respectively 8% vs 26%, p = .06; and 0% vs. 13% p = .06). CONCLUSION: VEN-based treatment was found to be effective in high risk AML can be considered as an alternative to IC in patients aged 60-75 with adverse cytogenetics.

3.
Genes Chromosomes Cancer ; 59(1): 30-39, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31340059

ABSTRACT

INTRODUCTION: In BCR-ABL1-negative myeloproliferative neoplasms, myelofibrosis (MF) is either primary (PMF) or secondary (SMF) to polycythemia vera or essential thrombocythemia. MF is characterized by an increased risk of transformation to acute myeloid leukemia (AML) and a shortened life expectancy. METHODS: Because natural histories of PMF and SMF are different, we studied by targeted next generation sequencing the differences in the molecular landscape of 86 PMF and 59 SMF and compared their prognosis impact. RESULTS: PMF had more ASXL1 (47.7%) and SRSF2 (14%) gene mutations than SMF (respectively 27.1% and 3.4%, P = .04). Poorer survival was associated with RNA splicing mutations (especially SRSF2) and TP53 in PMF (P = .0003), and with ASXL1 and TP53 mutations in SMF (P < .0001). These mutations of poor prognosis were associated with biological features of scoring systems (DIPSS and MYSEC-PM score). Mutations in TP53/SRSF2 in PMF or TP53/ASXL1 in SMF were more frequent as the risk of these scores increased. This allowed for a better stratification of MF patients, especially within the DIPSS intermediate-1 risk group (DIPSS) or the MYSEC-PM high risk group. AML transformation occurred faster in SMF than in PMF and patients who transformed to AML were more SRSF2-mutated and less CALR-mutated at MF sampling. CONCLUSIONS: PMF and SMF have different but not specific molecular profiles and different prognosis depending on the molecular profile. This may be due to differences in disease history. Combining mutations and existing scores should improve prognosis assessment.

4.
Br J Haematol ; 188(6): 935-944, 2020 03.
Article in English | MEDLINE | ID: mdl-31710700

ABSTRACT

In myeloproliferative neoplasms (MPN), JAK2V617F allele burden measurement has an impact on prognosis that helps in patient monitoring. Less is known about its usefulness in CALR-mutated cases. Additional mutations found by next-generation sequencing have also shown an impact on prognosis that may drive therapeutic choices, especially in myelofibrosis, but few studies focused on CALR-mutated patients. We performed a molecular evaluation combining next-generation sequencing with a myeloid panel and CALR allele burden measurement at diagnosis and during follow-up in a cohort of 45 patients with CALR-mutated essential thrombocythaemia. The bone marrow histology was also blindly reviewed in order to apply the WHO2016 classification. The most frequently mutated gene was TET2 (11/21 mutations). CALR type 1-like patients appear to have a more complex molecular landscape. We found an association between disease progression and CALR allele burden increase during follow-up, independently of additional mutations and WHO2016-reviewed diagnosis. Patients with disease progression at the time of follow-up showed a significant increase in CALR allele burden (+16·7%, P = 0·005) whereas patients without disease progression had a stable allele burden (+3·7%, P = 0·194). This result argues for clinical interest in CALR allele burden monitoring.


Subject(s)
Calreticulin/genetics , Myeloproliferative Disorders/genetics , Thrombocytosis/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Disease Progression , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation , Prognosis , Young Adult
5.
Histopathology ; 74(4): 654-662, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30393995

ABSTRACT

AIMS: This study sought to clarify the molecular pathways underlying the putative evolution from lymphomatoid papulosis (LyP) to cutaneous anaplastic large-cell lymphoma (c-ALCL) and lymph node invasion (LNI). METHODS AND RESULTS: We analysed nine sequential tumours from the same patient presenting with parallel evolution of LyP (n = 3) and c-ALCL (n = 1) with LNI (n = 1), combined with systemic diffuse large B-cell lymphoma (DLBCL) (n = 4). Clonality analysis showed a common clonal T-cell origin in the five CD30+ lesions, and a common clonal B-cell origin in the four DLBCL relapses. Array-comparative genomic hybridisation and targeted next-generation sequencing analysis demonstrated relative genomic stability of LyP lesions as compared with clonally related anaplastic large-cell lymphoma (ALCL) tumours, which showed 4q and 22q13 deletions involving the PRDM8 and TIMP3 tumour suppressor genes, respectively. The three analysed CD30+ lesions showed mostly private (specific to each sample) genetic alterations, suggesting early divergence from a common precursor. In contrast, DLBCL tumours showed progressive accumulation of private alterations, indicating late divergence. CONCLUSIONS: Sequential cutaneous and nodal CD30+ tumours were clonally related. This suggests that LyP, c-ALCL and LNI represent a continuous spectrum of clonal evolution emerging from a common precursor of cutaneous CD30+ lymphoproliferations. Therefore, nodal ALCL tumours in the context of LyP should be considered as a form of transformation rather than composite lymphoma.


Subject(s)
Lymph Nodes/pathology , Lymphoma, Large-Cell, Anaplastic/pathology , Lymphomatoid Papulosis/pathology , Skin Neoplasms/pathology , Clonal Evolution , Disease Progression , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphomatoid Papulosis/genetics , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Skin Neoplasms/genetics
6.
Am J Hematol ; 93(3): 330-338, 2018 03.
Article in English | MEDLINE | ID: mdl-29148089

ABSTRACT

Acute myeloid leukemias secondary (sAML) to myeloproliferative neoplasms (MPN) have variable clinical courses and outcomes, but remain almost always fatal. Large cohorts of sAML to MPN are difficult to obtain and there is very little scientific literature or prospective trials for determining robust prognostic markers and efficient treatments. We analyzed event-free survival (EFS) and overall survival (OS) of 73 patients with MPN who progressed to sAML, based on their epidemiological characteristics, the preexisting MPN, the different treatments received, the different prognostic groups and the responses achieved according to the ELN, and their mutational status determined by next-generation DNA sequencing (NGS). For 24 patients, we were able to do a comparative NGS analysis at both MPN and sAML phase. After acute transformation EFS and OS were respectively of 2.9 months (range: 0-48.1) and 4.7 months (range: 0.1-58.8). No difference in EFS or OS regarding the previous MPN, the ELN2017 prognostic classification, the first-line therapy or the response was found. After univariate analysis, three genes, TP53, SRSF2 and TET2, impacted pejoratively sAML prognosis at sAML time. In multivariate analysis, TP53 (P = .0001), TET2 (P = .011) and SRSF2 (P = .018) remained independent prognostic factors. Time to sAML transformation was shorter in SRSF2-mutated patients (51.2 months, range: 14.7-98) than in SRSF2-unmutated patients (133.8 months, range: 12.6-411.2) (P < .001). Conventional clinical factors (age, karyotype, ELN2017 prognostic classification, treatments received, treatments response, Allo-SCT…) failed to predict the patients' outcome. Only the mutational status appeared relevant to predict patients' prognosis at sAML phase.


Subject(s)
Genes, Neoplasm , Leukemia, Myeloid, Acute/genetics , Myeloproliferative Disorders/pathology , Precancerous Conditions/pathology , Adult , Aged , Aged, 80 and over , Allografts , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Female , Genes, p53 , Hematopoietic Stem Cell Transplantation , Humans , Karyotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myeloproliferative Disorders/genetics , Palliative Care , Precancerous Conditions/genetics , Prognosis , Progression-Free Survival , Proto-Oncogene Proteins/genetics , Sequence Analysis, DNA , Serine-Arginine Splicing Factors/genetics , Survival Analysis
9.
Am J Hematol ; 90(8): 737-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26017030

ABSTRACT

Acquired α-thalassemia myelodysplastic syndrome (MDS) (ATMDS) is an acquired syndrome characterized by a somatic point mutation or splicing defect in the ATRX gene in patients with myeloid disorders, primarily MDS. In a large MDS patient series, the incidence of ATMDS was below 0.5%. But no large series has yet assessed the incidence of ATMDS in microcytic MDS. In this study, we focused on patients with MDS and unexplained microcytosis, which was defined as absence of iron deficiency, inflammatory disease, or history of inherited hemoglobinopathy. Our data confirm the low frequency of ATRX mutations in MDS: 0% in an unselected clinical trial cohort of 80 low risk MDS, 0.2-0.8% in a multicenter registry of 2,980 MDS and 43% of MDS with unexplained microcytosis in this same registry. In addition, we reported four novel mutations of the ATRX gene in ATMDS. This study further determines the frequency of ATRX mutations and highlights the importance of microcytosis to detect ATRX mutations within MDS patients.


Subject(s)
DNA Helicases/genetics , Hematopoietic Stem Cells/pathology , Mutation Rate , Myelodysplastic Syndromes/genetics , Nuclear Proteins/genetics , alpha-Thalassemia/genetics , Aged , Aged, 80 and over , DNA Mutational Analysis , Gene Expression , Hematopoietic Stem Cells/metabolism , Humans , Middle Aged , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/pathology , Survival Analysis , X-linked Nuclear Protein , alpha-Thalassemia/metabolism , alpha-Thalassemia/mortality , alpha-Thalassemia/pathology
10.
Haematologica ; 99(1): 37-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23996481

ABSTRACT

Myelofibrosis is a myeloproliferative neoplasm that occurs de novo (primary myelofibrosis) or results from the progression of polycythemia vera or essential thrombocytemia (hereafter designated as secondary myelofibrosis or post-polycythemia vera/ essential thrombocythemia myelofibrosis). To progress in the understanding of myelofibrosis and to find molecular prognostic markers we studied 104 samples of primary and secondary myelofibrosis at chronic (n=68) and acute phases (n=12) from 80 patients, by using array-comparative genomic hybridization and sequencing of 23 genes (ASXL1, BMI1, CBL, DNMT3A, EZH2, IDH1/2, JAK2, K/NRAS, LNK, MPL, NF1, PPP1R16B, PTPN11, RCOR1, SF3B1, SOCS2, SRSF2, SUZ12, TET2, TP53, TRPS1). We found copy number aberrations in 54% of samples, often involving genes with a known or potential role in leukemogenesis. We show that cases carrying a del(20q), del(17) or del(12p) evolve in acute myeloid leukemia (P=0.03). We found that 88% of the cases were mutated, mainly in signaling pathway (JAK2 69%, NF1 6%) and epigenetic genes (ASXL1 26%, TET2 14%, EZH2 8%). Overall survival was poor in patients with more than one mutation (P=0.001) and in patients with JAK2/ASXL1 mutations (P=0.02). Our study highlights the heterogeneity of myelofibrosis, and points to several interesting copy number aberrations and genes with diagnostic and prognostic impact.


Subject(s)
Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Chromosome Deletion , Comparative Genomic Hybridization , DNA Copy Number Variations , Disease Progression , Female , Genetic Association Studies , Humans , Male , Middle Aged , Mutation , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/mortality , Prognosis , Sequence Analysis, DNA
11.
Am J Hematol ; 89(6): 604-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24595958

ABSTRACT

Initially classified in the myelodysplastic syndromes (MDSs), chronic myelomonocytic leukemia (CMML) is currently considered as a MDS/myeloproliferative neoplasm. Two classes-myelodysplastic and myeloproliferative-have been distinguished upon the level of the white blood cell count (threshold 13 G/L). We analyzed mutations in 19 genes reported in CMML to determine if and how these mutations impacted the respective prognosis of the two classes. We defined four major mutated pathways (DNA methylation, ASXL1, splicing, and signaling) and determined their prognostic impact. The number of mutated pathways impacted overall survival in the myelodysplastic class but not in the myeloproliferative class. The myeloproliferative class had a worse prognosis than the myelodysplastic class and was impacted by RUNX1 mutations only. Our results argue for a reclassification of CMML based on the myelodysplastic/myeloproliferative status.


Subject(s)
Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Disease-Free Survival , Humans , Leukemia, Myelomonocytic, Chronic/classification , Prognosis , Survival Analysis
12.
Haematologica ; 98(4): 576-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23065512

ABSTRACT

Chronic myelomonocytic leukemia is similar to but a separate entity from both myeloproliferative neoplasms and myelodysplastic syndromes, and shows either myeloproliferative or myelodysplastic features. We ask whether this distinction may have a molecular basis. We established the gene expression profiles of 39 samples of chronic myelomonocytic leukemia (including 12 CD34-positive) and 32 CD34-positive samples of myelodysplastic syndromes by using Affymetrix microarrays, and studied the status of 18 genes by Sanger sequencing and array-comparative genomic hybridization in 53 samples. Analysis of 12 mRNAS from chronic myelomonocytic leukemia established a gene expression signature of 122 probe sets differentially expressed between proliferative and dysplastic cases of chronic myelomonocytic leukemia. As compared to proliferative cases, dysplastic cases over-expressed genes involved in red blood cell biology. When applied to 32 myelodysplastic syndromes, this gene expression signature was able to discriminate refractory anemias with ring sideroblasts from refractory anemias with excess of blasts. By comparing mRNAS from these two forms of myelodysplastic syndromes we derived a second gene expression signature. This signature separated the myelodysplastic and myeloproliferative forms of chronic myelomonocytic leukemias. These results were validated using two independent gene expression data sets. We found that myelodysplastic chronic myelomonocytic leukemias are characterized by mutations in transcription/epigenetic regulators (ASXL1, RUNX1, TET2) and splicing genes (SRSF2) and the absence of mutations in signaling genes. Myelodysplastic chronic myelomonocytic leukemias and refractory anemias with ring sideroblasts share a common expression program suggesting they are part of a continuum, which is not totally explained by their similar but not, however, identical mutation spectrum.


Subject(s)
Anemia, Refractory/genetics , Anemia, Sideroblastic/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Myelodysplastic Syndromes/genetics , Adult , Aged , Aged, 80 and over , Anemia, Refractory/diagnosis , Anemia, Refractory/metabolism , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/metabolism , Antigens, CD34/metabolism , Cluster Analysis , Comparative Genomic Hybridization , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , Diagnosis, Differential , Dioxygenases , Female , Gene Expression Profiling , Humans , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/metabolism , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/metabolism , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Ribonucleoproteins/genetics , Sequence Analysis, DNA , Serine-Arginine Splicing Factors
13.
Genes Chromosomes Cancer ; 51(8): 743-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22489043

ABSTRACT

Since the discovery of the JAK2V617F tyrosine kinase-activating mutation several genes have been found mutated in nonchronic myeloid leukemia (CML) myeloproliferative neoplasms (MPNs), which mainly comprise three subtypes of "classic" MPNs; polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF). We searched for mutations in ASXL1, CBL, DNMT3A, IDH1, IDH2, JAK2, MPL, NF1, SF3B1, SUZ12, and TET2 genes in 149 non-CML MPNs, including 127 "classic" MPNs cases. JAK2 was mutated in 100% PV, 66% ET and 68% MF. We found a high incidence of ASXL1 mutation in MF patients (20%) and a low incidence in PV (7%) and ET (4%) patients. Mutations in the other genes were rare (CBL, DNMT3A, IDH2, MPL, SF3B1, SUZ12, NF1) or absent (IDH1).


Subject(s)
DNA-Binding Proteins/genetics , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Carrier Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA Mutational Analysis , Dioxygenases , Female , Humans , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Proteins , Neurofibromin 1/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins c-cbl/genetics , RNA Splicing Factors , Receptors, Thrombopoietin/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Transcription Factors
15.
BMC Cancer ; 12: 304, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22823977

ABSTRACT

Myeloid malignant diseases comprise chronic (including myelodysplastic syndromes, myeloproliferative neoplasms and chronic myelomonocytic leukemia) and acute (acute myeloid leukemia) stages. They are clonal diseases arising in hematopoietic stem or progenitor cells. Mutations responsible for these diseases occur in several genes whose encoded proteins belong principally to five classes: signaling pathways proteins (e.g. CBL, FLT3, JAK2, RAS), transcription factors (e.g. CEBPA, ETV6, RUNX1), epigenetic regulators (e.g. ASXL1, DNMT3A, EZH2, IDH1, IDH2, SUZ12, TET2, UTX), tumor suppressors (e.g. TP53), and components of the spliceosome (e.g. SF3B1, SRSF2). Large-scale sequencing efforts will soon lead to the establishment of a comprehensive repertoire of these mutations, allowing for a better definition and classification of myeloid malignancies, the identification of new prognostic markers and therapeutic targets, and the development of novel therapies. Given the importance of epigenetic deregulation in myeloid diseases, the use of drugs targeting epigenetic regulators appears as a most promising therapeutic approach.


Subject(s)
Leukemia, Myeloid/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Cell Transformation, Neoplastic/genetics , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/therapy , Models, Biological , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/therapy , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/therapy
16.
Am J Hematol ; 87(7): 659-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22535592

ABSTRACT

To determine whether the distinct and heterogeneous WHO category called "AML with myelodysplasia-related changes" (MRC-AML), presents specific molecular alterations we searched for mutations in genes known to be mutated in malignant myeloid diseases. In 48 MRC-AML patients analyzed, we found 17 mutations in ASXL1 (35%), eight in RUNX1 (17%), seven in TET2 (15%), 12 in IDH (n = 2) or IDH2 (n = 10) (25%), four in DNMT3A (8%), four in NPM1 (8%), and one in FLT3 (2%). Mutations were more frequent in the intermediate cytogenetic (IC) subgroup of 36 patients than in the unfavorable karyotype subgroup, with an average ratio mutations/patients of 1.36 [0-3] vs. 0.33 [0-2] (P < 0.001). Then, we compared these 36 patients with IC MRC-AML with a control panel of 37 no-MRC-AML patients, who had both IC and no dysplasia. IC MRC-AMLs were associated with higher incidence of ASXL1 mutations (47% vs. 0%, P < 0.001) and lower incidence of DNMT3A (6% vs. 38%, P = 0.001), NPM1 (11% vs. 62%, P < 0.001) and FLT3 (3% vs. 49%, P < 0.001) mutations. No difference was found in the incidence of IDH1/2 or TET2 mutations according to the presence of dysplasia. Complete remission rate after intensive treatment was lower in the MRC-AML group than in the no-MRC-AML group (48% vs. 78%, P = 0.023) and in wild type NPM1 patients (50% vs. 84%, P = 0.009). Our study showed that MRC-AML as defined in the WHO 2008 classification presents a specific mutation pattern characterized by a high frequency of ASXL1 mutations and a low rate of NPM1, FLT3, and DNMT3A mutations.


Subject(s)
Bone Marrow/pathology , DNA, Intergenic/chemistry , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation Rate , Repressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Female , France , Genetic Association Studies , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Remission Induction , Repressor Proteins/metabolism , World Health Organization , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
17.
Blood Adv ; 5(5): 1442-1451, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33666653

ABSTRACT

We aimed to study the prognostic impact of the mutational landscape in primary and secondary myelofibrosis. The study included 479 patients with myelofibrosis recruited from 24 French Intergroup of Myeloproliferative Neoplasms (FIM) centers. The molecular landscape was studied by high-throughput sequencing of 77 genes. A Bayesian network allowed the identification of genomic groups whose prognostic impact was studied in a multistate model considering transitions from the 3 conditions: myelofibrosis, acute leukemia, and death. Results were validated using an independent, previously published cohort (n = 276). Four genomic groups were identified: patients with TP53 mutation; patients with ≥1 mutation in EZH2, CBL, U2AF1, SRSF2, IDH1, IDH2, NRAS, or KRAS (high-risk group); patients with ASXL1-only mutation (ie, no associated mutation in TP53 or high-risk genes); and other patients. A multistate model found that both TP53 and high-risk groups were associated with leukemic transformation (hazard ratios [HRs] [95% confidence interval], 8.68 [3.32-22.73] and 3.24 [1.58-6.64], respectively) and death from myelofibrosis (HRs, 3.03 [1.66-5.56] and 1.77 [1.18-2.67], respectively). ASXL1-only mutations had no prognostic value that was confirmed in the validation cohort. However, ASXL1 mutations conferred a worse prognosis when associated with a mutation in TP53 or high-risk genes. This study provides a new definition of adverse mutations in myelofibrosis with the addition of TP53, CBL, NRAS, KRAS, and U2AF1 to previously described genes. Furthermore, our results argue that ASXL1 mutations alone cannot be considered detrimental.


Subject(s)
Primary Myelofibrosis , Bayes Theorem , Genomics , Humans , Mutation , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Prognosis , Repressor Proteins/genetics
18.
Br J Haematol ; 151(4): 365-75, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20880116

ABSTRACT

Chronic myelomonocytic leukaemia (CMML) is a haematological disease currently classified in the category of myelodysplastic syndromes/myeloproliferative neoplasm (MDS/MPN) because of its dual clinical and biological presentation. The molecular biology of CMML is poorly characterized. We studied a series of 53 CMML samples including 31 cases of myeloproliferative form (MP-CMML) and 22 cases of myelodysplastic forms (MD-CMML) using array-comparative genomic hybridisation (aCGH) and sequencing of 13 candidate genes including ASXL1, CBL, FLT3, IDH1, IDH2, JAK2, KRAS, NPM1, NRAS, PTPN11, RUNX1, TET2 and WT1. Mutations in ASXL1 and in the genes associated with proliferation (CBL, FLT3, PTPN11, NRAS) were mainly found in MP-CMML cases. Mutations of ASXL1 correlated with an evolution toward an acutely transformed state: all CMMLs that progressed to acute phase were mutated and none of the unmutated patients had evolved to acute leukaemia. The overall survival of ASXL1 mutated patients was lower than that of unmutated patients.


Subject(s)
Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Repressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Comparative Genomic Hybridization , DNA Mutational Analysis/methods , DNA, Neoplasm/genetics , Disease Progression , Female , Follow-Up Studies , Genes, Neoplasm , Genetic Association Studies , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Nucleophosmin , Prognosis , Survival Analysis
19.
BMC Cancer ; 10: 401, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20678218

ABSTRACT

BACKGROUND: Gene mutation is an important mechanism of myeloid leukemogenesis. However, the number and combination of gene mutated in myeloid malignancies is still a matter of investigation. METHODS: We searched for mutations in the ASXL1, CBL, FLT3, IDH1, IDH2, JAK2, KRAS, NPM1, NRAS, RUNX1, TET2 and WT1 genes in 65 myelodysplastic syndromes (MDSs) and 64 acute myeloid leukemias (AMLs) without balanced translocation or complex karyotype. RESULTS: Mutations in ASXL1 and CBL were frequent in refractory anemia with excess of blasts. Mutations in TET2 occurred with similar frequency in MDSs and AMLs and associated equally with either ASXL1 or NPM1 mutations. Mutations of RUNX1 were mutually exclusive with TET2 and combined with ASXL1 but not with NPM1. Mutations in FLT3 (mutation and internal tandem duplication), IDH1, IDH2, NPM1 and WT1 occurred primarily in AMLs. CONCLUSION: Only 14% MDSs but half AMLs had at least two mutations in the genes studied. Based on the observed combinations and exclusions we classified the 12 genes into four classes and propose a highly speculative model that at least a mutation in one of each class is necessary for developing AML with simple or normal karyotype.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Myelodysplastic Syndromes/genetics , Neoplasm Proteins/genetics , Aged , Aged, 80 and over , Core Binding Factor Alpha 2 Subunit/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Female , Genes, ras/physiology , Humans , Isocitrate Dehydrogenase/genetics , Janus Kinase 2/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Nuclear Proteins/genetics , Nucleophosmin , Polymerase Chain Reaction , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins p21(ras) , Repressor Proteins/genetics , WT1 Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics , ras Proteins/genetics
20.
Am J Hematol ; 85(9): 717-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20687102

ABSTRACT

New genes involved in leukemogenesis, such as ASXL1 and TET2, have been identified recently using genomic analyses of DNA from patient samples. We have studied by array-comparative genomic hybridization (aCGH) a series of 167 samples including myelodysplastic syndromes, chronic myelomonocytic leukemias, and acute myeloid leukemias. We found a deletion of the RAD21 and STAG2 genes, which encode two components of the cohesin complex. We propose that these alterations may compromise the cohesin complex and its regulation of the transcription of genes.


Subject(s)
Antigens, Nuclear/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Deletion , Leukemia, Myeloid/genetics , Myelodysplastic Syndromes/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Female , Humans , Male , Nucleic Acid Hybridization , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL