Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hematol ; 99(6): 1108-1118, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563187

ABSTRACT

We investigated using a custom NGS panel of 149 genes the mutational landscape of 64 consecutive adult patients with tyrosine kinase fusion-negative hypereosinophilia (HE)/hypereosinophilic syndrome (HES) harboring features suggestive of myeloid neoplasm. At least one mutation was reported in 50/64 (78%) patients (compared to 8/44 (18%) patients with idiopathic HE/HES/HEUS used as controls; p < .001). Thirty-five patients (54%) had at least one mutation involving the JAK-STAT pathway, including STAT5B (n = 18, among which the hotspot N642H, n = 13), JAK1 (indels in exon 13, n = 5; V658F/L, n = 2), and JAK2 (V617F, n = 6; indels in exon 13, n = 2). Other previously undescribed somatic mutations were also found in JAK2, JAK1, STAT5B, and STAT5A, including three patients who shared the same STAT5A V707fs mutation and features consistent with primary polycythemia. Nearly all JAK-STAT mutations were preceded by (or associated with) myelodysplasia-related gene mutations, especially in RNA-splicing genes or chromatin modifiers. In multivariate analysis, neurologic involvement (hazard ratio [HR] 4.95 [1.87-13.13]; p = .001), anemia (HR 5.50 [2.24-13.49]; p < .001), and the presence of a high-risk mutation (as per the molecular international prognosis scoring system: HR 6.87 [2.39-19.72]; p < .001) were independently associated with impaired overall survival. While corticosteroids were ineffective in all treated JAK-STAT-mutated patients, ruxolitinib showed positive hematological responses including in STAT5A-mutated patients. These findings emphasize the usefulness of NGS for the workup of tyrosine kinase fusion-negative HE/HES patients and support the use of JAK inhibitors in this setting. Updated classifications could consider patients with JAK-STAT mutations and eosinophilia as a new "gene mutated-entity" that could be differentiated from CEL, NOS, and idiopathic HES.


Subject(s)
Hypereosinophilic Syndrome , Mutation , STAT5 Transcription Factor , Humans , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/drug therapy , Male , Female , Middle Aged , Adult , Aged , STAT5 Transcription Factor/genetics , Janus Kinase 2/genetics , Signal Transduction , Janus Kinase 1/genetics , Aged, 80 and over , Pyrimidines/therapeutic use , Young Adult
2.
Neurogenetics ; 21(1): 67-72, 2020 01.
Article in English | MEDLINE | ID: mdl-31823155

ABSTRACT

Microdeletions encompassing 14q11.2 locus, involving SUPT16H and CHD8, were shown to cause developmental delay, intellectual disability, autism spectrum disorders and macrocephaly. Variations leading to CHD8 haploinsufficiency or loss of function were also shown to lead to a similar phenotype. Recently, a 14q11.2 microduplication syndrome, encompassing CHD8 and SUPT16H, has been described, highlighting the importance of a tight control of at least CHD8 gene-dosage for a normal development. There have been only a few reports of 14q11.2 microduplications. Patients showed variable neurodevelopmental issues of variable severity. Breakpoints of the microduplications were non-recurrent, making interpretation of the CNV and determination of their clinical relevance difficult. Here, we report on two patients with 14q11.2 microduplication encompassing CHD8 and SUPT16H, one of whom had normal intelligence. Review of previous reports describing patients with comparable microduplications allowed for a more precise delineation of the condition and widening of the phenotypic spectrum.


Subject(s)
Brain/pathology , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Gene Duplication , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Transcription Factors/genetics , Brain/diagnostic imaging , Child , Female , Humans , Male , Neurodevelopmental Disorders/diagnostic imaging , Phenotype
3.
Haematologica ; 104(6): 1150-1155, 2019 06.
Article in English | MEDLINE | ID: mdl-30573507

ABSTRACT

Clonal chromosome abnormalities in Philadelphia-negative cells could concern chronic myeloid leukemia patients treated by tyrosine kinase inhibitors. The European LeukemiaNet distinguishes -7/del(7q) abnormalities as a "warning". However, the impact of clonal chromosome abnormalities, and specifically those of -7/del(7q), in Philadelphia-negative cells on clinical outcomes is unclear and based on case-reports showing morphological dysplasia and increased risk of acute myeloid leukemia, suggesting the coexistence of chronic myeloid leukemia and high-risk myelodysplastic syndrome. The aim of this study was to determine whether the impact of -7/del(7q) clonal chromosome abnormalities in Philadelphia-negative cells on the clinical outcome is different from that of other types of abnormalities, and we argue for an underlying associated high-risk myelodysplastic syndrome. Among 102 chronic myeloid leukemia patients with clonal chromosome abnormalities in Philadelphia-negative cells with more than a median of 6 years of follow up, patients with -7/del(7q) more frequently had signs of dysplasia, a lower cumulative incidence of deep molecular response and often needed further treatment lines, with the consequent impact on event-free and progression-free survival. Morphological features of dysplasia are associated with myelodysplastic syndrome/acute myeloid leukemia mutations and compromise the optimal response to tyrosine kinase inhibitors, irrespectively of the type of clonal chromosome abnormalities in Philadelphia-negative cells. However, mutation patterns determined by next-generation sequencing could not clearly explain the underlying high-risk disease. We hereby confirm the pejorative prognostic value of -7/del(7q) clonal chromosome abnormalities in Philadelphia-negative cells and suggest that myelodysplastic features constitute a warning signal that response to tyrosine kinase inhibitors may be less than optimal.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 7 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Metaphase/genetics , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Alleles , Chromosome Deletion , Disease Progression , Female , Genetic Association Studies , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Male , Middle Aged , Molecular Targeted Therapy , Mutation , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Prognosis , Survival Analysis
4.
Cancer ; 124(14): 2956-2963, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29723417

ABSTRACT

BACKGROUND: The ultimate goal of chronic myeloid leukemia management in the tyrosine kinase inhibitor (TKI) era for patients who obtain deep molecular responses is maintaining a durable off-treatment response after treatment discontinuation; this situation is called treatment-free remission (TFR). Knowledge accumulated during the last 10 years justifies moving TFR strategies from research to clinical practice. METHODS: Twenty experts from the French Chronic Myeloid Leukemia Study Group (France Intergroupe des Leucémies Myéloïdes Chroniques), including 17 hematologists, 2 molecular biologists, and 1 cytogeneticist, critically reviewed published data with the goal of developing evidence-based recommendations for TKI discontinuation in clinical practice. RESULTS: Clinically relevant questions were addressed, including the selection of candidate patients (with known prognostic factors for outcomes taken into account), detailed monitoring procedures during the treatment-free phase, a definition of relapse requiring therapy resumption, and monitoring after treatment reintroduction. CONCLUSIONS: This work presents consensus statements with the aim of guiding physicians and biologists by means of pragmatic recommendations for safe TKI discontinuation in daily practice. Cancer 2018;124:2956-63. © 2018 American Cancer Society.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Hematology/standards , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Medical Oncology/standards , Neoplasm Recurrence, Local/diagnosis , Protein Kinase Inhibitors/therapeutic use , Adult , Age Factors , Consensus , France , Fusion Proteins, bcr-abl/blood , Fusion Proteins, bcr-abl/isolation & purification , Fusion Proteins, bcr-abl/metabolism , Hematology/methods , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Medical Oncology/methods , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/prevention & control , Patient Education as Topic , Patient Selection , Prognosis , Remission Induction/methods , Treatment Outcome , Watchful Waiting/standards , Young Adult
7.
Am J Med Genet A ; 173(6): 1690-1693, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28398607

ABSTRACT

Wilm's tumor, aniridia, genitourinary anomalies, and mental retardation (WAGR) syndrome, a rare genetic disorder, is caused by the loss of 11p13 region including PAX6 and WT1. We report novel findings in a 28-month-old boy with aniridia, Wilm's tumor, congenital hypothyroidism, and sublingual thyroid ectopia. He was found to have a mosaic 5.28 Mb interstitial deletion of chromosome 11p13 deleting PAX6 and WT1. In order to clarify the mechanism underlying his thyroid dysgenesis, sequence analysis of candidate thyroid developmental genes was performed. We identified a FOXE1: c.532_537delGCCGCC p.(Ala178_Ala179del) variant that predisposes to thyroid ectopia. Taken together, this is the first report of mosaic 11p13 deletion in association with thyroid dysgenesis. We also propose a model of complex interactions of different genetic variants for this particular phenotype in the present patient.


Subject(s)
Congenital Hypothyroidism/genetics , Forkhead Transcription Factors/genetics , Thyroid Dysgenesis/genetics , WAGR Syndrome/genetics , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 11 , Congenital Hypothyroidism/physiopathology , Humans , In Situ Hybridization, Fluorescence , Male , Mosaicism , PAX6 Transcription Factor/genetics , Phenotype , Thyroid Dysgenesis/physiopathology , WAGR Syndrome/physiopathology , WT1 Proteins/genetics
9.
Eur J Haematol ; 97(4): 399-402, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26935241

ABSTRACT

We report a case of myeloproliferative neoplasm (MPN) with an atypical t(9;22;15)(p24;q11;q21) translocation, leading to a BCR-JAK2 fusion, associated with a trisomy of chromosome 8 in clonal evolution at karyotype. Patient's evolution was marked by an aggressive clinical course with rapid progression to blast phase within the first year after diagnosis. Examination of matched chronic phase and blast crisis samples by SNP-array karyotyping identified secondary acquired cryptic genetic events at the time of lymphoblastic transformation, including biallelic IKZF1 alteration and EBF1 and CDKN2A/B codeletions. This case is the first report describing acquisition of secondary genetic events leading to acute lymphoblastic progression in a rare MPN with BCR-JAK2 fusion.


Subject(s)
Cell Transformation, Neoplastic/genetics , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Oncogene Proteins, Fusion , Proto-Oncogene Proteins c-bcr/genetics , Translocation, Genetic , Chromosome Banding , Disease Progression , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Sequence Analysis, DNA
10.
Blood ; 121(22): 4504-11, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23532735

ABSTRACT

Mutation of the MYD88 gene has recently been identified in activated B-cell-like diffuse cell lymphoma and enhanced Janus kinase/signal transducer and activator of transcription (JAK-STAT) and nuclear factor κB (NF-κB) signaling pathways. A whole exome-sequencing study of Waldenstrom macroglobulinemia (WM) suggested a high frequency of MYD88 L265P mutation in WM. The genetic background is not fully deciphered in WM, although the role of NF-κB and JAK-STAT has been demonstrated. We analyzed MYD88 mutation in exon 5 and characterized the clinical significance of this genetic alteration in 67 WM patients. Clinical features; immunophenotypic markers; and conventional cytogenetic, fluorescence in situ hybridization, and single nucleotide polymorphism array data were analyzed. MYD88 L265P mutation was acquired in 79% of patients. Overall, we have identified alteration of the MYD88 locus in 91% of WM patients, including 12% with gain on chromosome 3 at the 3p22 locus that included the MYD88 gene. Patients with absence of MYD88 mutation were WM characterized with a female predominance, a splenomegaly, gain of chromosome 3, and CD27 expression. Importantly, inhibition of MYD88 signaling induced cytotoxicity and inhibited cell growth of cell lines issued from patients with WM. In conclusion, these results confirm a high frequency of MYD88 L265P mutation in WM. The discovery of MYD88 L265P mutation may contribute to a better understanding of the physiopathogeny of WM.


Subject(s)
Myeloid Differentiation Factor 88/genetics , Point Mutation , Waldenstrom Macroglobulinemia/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation , Female , Flow Cytometry , Humans , Male , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Polymorphism, Single Nucleotide , Signal Transduction/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Waldenstrom Macroglobulinemia/metabolism , Waldenstrom Macroglobulinemia/therapy
11.
Haematologica ; 100(8): 1086-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25682606

ABSTRACT

The CD3(-)CD4(+) lymphoid variant of hypereosinophilic syndrome is characterized by hypereosinophilia and clonal circulating CD3(-)CD4(+) T cells. Peripheral T-cell lymphoma has been described during this disease course, and we observed in our cohort of 23 patients 2 cases of angio-immunoblastic T-cell lymphoma. We focus here on histopathological (n=12 patients) and immunophenotypic (n=15) characteristics of CD3(-)CD4(+) lymphoid variant of hypereosinophilic syndrome. Atypical CD4(+) T cells lymphoid infiltrates were found in 10 of 12 CD3(-)CD4(+) L-HES patients, in lymph nodes (n=4 of 4 patients), in skin (n=9 of 9) and other extra-nodal tissues (gut, lacrymal gland, synovium). Lymph nodes displayed infiltrates limited to the interfollicular areas or even an effacement of nodal architecture, associated with proliferation of arborizing high endothelial venules and increased follicular dendritic cell meshwork. Analysis of 2 fresh skin samples confirmed the presence of CD3(-)CD4(+) T cells. Clonal T cells were detected in at least one tissue in 8 patients, including lymph nodes (n=4 of 4): the same clonal T cells were detected in blood and in at least one biopsy, with a maximum delay of 23 years between samples. In the majority of cases, circulating CD3(-)CD4(+) T cells were CD2(hi) (n=9 of 14), CD5(hi) (n=12 of 14), and CD7(-)(n=4 of 14) or CD7(low) (n=10 of 14). Angio-immunoblastic T-cell lymphoma can also present with CD3(-)CD4(+) T cells; despite other common histopathological and immunophenotypic features, CD10 expression and follicular helper T-cell markers were not detected in lymphoid variant of hypereosinophilic syndrome patients, except in both patients who developed angio-immunoblastic T-cell lymphoma, and only at T-cell lymphoma diagnosis. Taken together, persistence of tissular clonal T cells and histopathological features define CD3(-)CD4(+) lymphoid variant of hypereosinophilic syndrome as a peripheral indolent clonal T-cell lymphoproliferative disorder, which should not be confused with angio-immunoblastic T-cell lymphoma.


Subject(s)
CD3 Complex/metabolism , CD4 Antigens/metabolism , Clonal Evolution , Hypereosinophilic Syndrome/metabolism , Hypereosinophilic Syndrome/pathology , Immunophenotyping , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Adolescent , Adult , Aged , Bone Marrow/metabolism , Bone Marrow/pathology , Diagnosis, Differential , Female , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Humans , Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/therapy , Immunohistochemistry , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Male , Middle Aged , Skin/metabolism , Skin/pathology , Young Adult
12.
Ann Hematol ; 94(2): 187-93, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25193356

ABSTRACT

The dicentric chromosome (9;20) (dic(9;20)) is described in 2 % of childhood B-acute lymphoblastic leukaemia. Fluorescence in situ hybridization (FISH) is the most reliable method to identify dic(9;20) when compared with conventional cytogenetics. To define the prognostic importance of dic(9;20), we evaluated treatment response and patient survival. This was a retrospective study in three French university centres. Patients' clinical and laboratory characteristics and treatment response are described. Nine children with dic(9;20) have been identified since 1995. All patients had at least one poor prognostic feature either among the clinical features, the initial laboratory results or in the initial treatment response: central nervous system involvement (2/9), high median leucocyte count (≥50 G/L) (8/9) and poor response to prednisone (2/9). All patients were in complete cytological remission after induction therapy but only three had a good molecular response with minimal residual disease (MRD) <10(-3). Five out of nine patients relapsed and two died, 4 and 12 months after diagnosis, respectively. The event-free survival rate in this population was 44 % (95 % confidence interval (CI) = 0.09-0.79) and overall survival 78 % (95 % CI = 0.51-1.05). In this population, dic(9;20) is associated with a relatively poor prognosis. Patients showing dic(9;20), whether this cytogenetic abnormality is associated with other poor prognostic factors or not, should be identified at the outset in order to be offered a more intensive treatment protocol.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 20/genetics , Chromosomes, Human, Pair 9/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Chromosome Banding , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Karyotype , Male , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prognosis , Retrospective Studies , Treatment Outcome
13.
Genes Chromosomes Cancer ; 53(1): 106-16, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24249260

ABSTRACT

Mantle cell lymphoma (MCL) is usually an aggressive disease. However, a few patients do have an "indolent" evolution (iMCL) defined by a long survival time without intensive therapy. Many studies highlight the prognostic role of additional genetic abnormalities, but these abnormalities are not routinely tested for and do not yet influence the treatment decision. We aimed to evaluate the prognostic impact of these additional abnormalities detected by conventional cytogenetic testing, as well as their relationships with the clinical characteristics and their value in identifying iMCL. All consecutive MCL cases diagnosed between 1995 and 2011 at four institutions were retrospectively selected on the basis of an informative karyotype with a t(11;14) translocation at the time of diagnosis. A total of 125 patients were included and followed for an actual median time of 35 months. The median overall survival (OS) and survival without treatment (TFS) were 73.7 and 1.3 months, respectively. In multivariable Cox models, a high mantle cell lymphoma international prognostic index score, a complex karyotype, and blastoid morphology were independently associated with a shortened OS. Spleen enlargement, nodal presentation, extra-hematological involvement, and complex karyotypes were associated with shorter TFS. A score based on these factors allowed for the identification of "indolent" patients (median TFS 107 months) from other patients (median TFS: 1 month). In conclusion, in this multicentric cohort of MCL patients, a complex karyotype was associated with a shorter survival time and allowed for the identification of iMCL at the time of diagnosis.


Subject(s)
Chromosomes, Human/genetics , Karyotype , Lymphoma, Mantle-Cell/diagnosis , Adult , Aged , Aged, 80 and over , Chromosome Aberrations , Female , Humans , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate
15.
N Engl J Med ; 363(26): 2511-21, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21175313

ABSTRACT

BACKGROUND: Imatinib (400 mg daily) is considered the best initial therapy for patients with newly diagnosed chronic myeloid leukemia (CML) in the chronic phase. However, only a minority of patients treated with imatinib have a complete molecular remission. METHODS: We randomly assigned 636 patients with untreated chronic-phase CML to receive imatinib alone at a dose of 400 mg daily, imatinib (400 mg daily) plus cytarabine (20 mg per square meter of body-surface area per day on days 15 through 28 of each 28-day cycle) or pegylated interferon (peginterferon) alfa-2a (90 µg weekly), or imatinib alone at a dose of 600 mg daily. Molecular and cytogenetic responses, time to treatment failure, overall and event-free survival, and adverse events were assessed. An analysis of molecular response at 12 months was planned. A superior molecular response was defined as a decrease in the ratio of transcripts of the tyrosine kinase gene BCR-ABL to transcripts of ABL of 0.01% or less, corresponding to a reduction of 4 log(10) units or more from the baseline level, as assessed by means of a real-time quantitative polymerase-chain-reaction assay. RESULTS: At 12 months, the rates of cytogenetic response were similar among the four groups. The rate of a superior molecular response was significantly higher among patients receiving imatinib and peginterferon alfa-2a (30%) than among patients receiving 400 mg of imatinib alone (14%) (P=0.001). The rate was significantly higher among patients treated for more than 12 months than among those treated for 12 months or less. Gastrointestinal events were more frequent among patients receiving cytarabine, whereas rash and depression were more frequent among patients receiving peginterferon alfa-2a. CONCLUSIONS: As compared with other treatments, the addition of peginterferon alfa-2a to imatinib therapy resulted in significantly higher rates of molecular response in patients with chronic-phase CML. (Funded by the French Ministry of Health and others; ClinicalTrials.gov number, NCT00219739.).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon-alpha/administration & dosage , Leukemia, Myeloid, Chronic-Phase/drug therapy , Piperazines/administration & dosage , Polyethylene Glycols/administration & dosage , Pyrimidines/administration & dosage , Adult , Anemia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides , Cytarabine/administration & dosage , Cytarabine/adverse effects , Female , Fusion Proteins, bcr-abl/analysis , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Interferon alpha-2 , Interferon-alpha/adverse effects , Leukemia, Myeloid, Chronic-Phase/mortality , Leukemia, Myeloid, Chronic-Phase/pathology , Male , Middle Aged , Neutropenia/chemically induced , Piperazines/adverse effects , Polyethylene Glycols/adverse effects , Proto-Oncogene Proteins c-abl/analysis , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/adverse effects , RNA, Neoplasm/analysis , Recombinant Proteins , Remission Induction , Stem Cells/drug effects , Survival Analysis , Thrombocytopenia/chemically induced , Transcription, Genetic , Treatment Outcome
16.
Blood ; 118(16): 4331-7, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21849485

ABSTRACT

Imatinib has transformed the prognosis and the management of chronic myeloid leukemia (CML) and has probably changed the patterns of mortality rates. We explored this change at each disease severity level (Sokal score) through a flexible statistical modeling of the effect of the year of diagnosis on the excess mortality rate. The study included 691 chronic-phase patients from Nord-Pas-de-Calais French CML registry diagnosed from 1990 to 2007. Imatinib was given to 93% of the patients diagnosed after 2000. Comparing the 1990-1994, 1995-1999, and 2000-2007 periods of diagnosis, the 5-year relative survival improved from 64% to 66% and 88%. The year of diagnosis was associated with a significant reduction of the excess mortality, but only in patients with intermediate to high Sokal scores. In high-risk patients diagnosed in the early 1990s, a peak of excess mortality was observed during the second year of follow-up. That peak decreased progressively over the years of diagnosis until disappearing in patients diagnosed after 2000. This study showed different effects according to Sokal scores of the use of imatinib on mortality in patients with chronic-phase CML and showed that since 2000 the pattern of mortality of high-risk patients became similar to that of intermediate-risk ones.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/mortality , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Benzamides , Cohort Studies , Female , Humans , Imatinib Mesylate , Leukemia, Myeloid, Chronic-Phase/diagnosis , Male , Middle Aged , Multivariate Analysis , Severity of Illness Index , Survival Analysis , Young Adult
17.
Haematologica ; 98(10): 1510-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23716543

ABSTRACT

The BCR-ABL T315I mutation confers resistance to currently licensed tyrosine kinase inhibitors in chronic myelogenous leukemia. However, the impact of this mutation on survival in early stages of disease, in chronic phase, has never been detailed. Using matched pair analysis, a cohort of 64 patients with chronic phase chronic myelogenous leukemia harboring a T315I mutation and resistant to imatinib mesylate was compared to a similar cohort of 53 chronic phase patients resistant to imatinib, but with no detectable T315I mutation, in the pre-ponatinib era. These patients were matched according to age at diagnosis, interval between disease diagnosis and start of imatinib treatment, and duration of imatinib therapy. Kaplan-Meier survival analyses demonstrated the significant negative impact of the presence of the T315I mutation on overall survival (since imatinib-resistance: 48.4 months for T315I(+) patients versus not reached for T315I(-) ones; P=0.006) and failure-free survival (since imatinib-resistance: 34.7 months for T315I(+) patients versus not reached for T315I(-) patients; P=0.003). In addition, Cox proportional hazard models adjusted on overall survival demonstrated the negative influence of the T315I mutation (P=0.02, HR=2.54). These results confirm early assumptions concerning the poor prognosis of chronic phase chronic myelogenous leukemia patients with the T315I mutation who are not eligible for allogeneic transplantation, and demonstrate the need for more therapeutic options.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Genes, abl , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Adolescent , Adult , Aged , Cohort Studies , Drug Resistance, Neoplasm/genetics , Female , Humans , Leukemia, Myeloid, Chronic-Phase/mortality , Male , Middle Aged , Prospective Studies , Protein Kinase Inhibitors/pharmacology , Retrospective Studies , Survival Rate , Young Adult
18.
Am J Hematol ; 88(4): 306-11, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23460398

ABSTRACT

Germline heterozygous alterations of the tumor-suppressor gene neurofibromatosis-1 (NF1) lead to neurofibromatosis type 1, a genetic disorder characterized by a higher risk to develop juvenile myelomonocytic leukemia and/or acute myeloid leukemia (AML). More recently, somatic 17q11 deletions encompassing NF1 have been described in many adult myeloid malignancies. In this context, we aimed to define NF1 involvement in AML. We screened a total of 488 previously untreated de novo AML patients for the NF1 deletion using either array comparative genomic hybridization (aCGH) or real-time quantitative PCR/fluorescence in situ hybridization approaches. We also applied massively parallel sequencing for in depth mutation analysis of NF1 in 20 patients including five NF1-deleted patients. We defined a small ∼0.3 Mb minimal deleted region involving NF1 by aCGH and an overall frequency of NF1 deletion of 3.5% (17/485). NF1 deletion is significantly associated with unfavorable cytogenetics and with monosomal karyotype notably. We discovered six NF1 variants of unknown significance in 7/20 patients of which only one out of four disappeared in corresponding complete remission sample. In addition, only one out of five NF1-deleted patients has an acquired coding mutation in the remaining allele. In conclusion, direct NF1 inactivation is infrequent in de novo AML and may be a secondary event probably involved in leukemic progression.


Subject(s)
Gene Deletion , Leukemia, Myeloid, Acute/genetics , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Adolescent , Adult , Aged , Alleles , Comparative Genomic Hybridization , Female , Gene Frequency , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Middle Aged , Mutation Rate , Neurofibromin 1/deficiency , Real-Time Polymerase Chain Reaction
19.
Am J Hematol ; 88(11): 948-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23861223

ABSTRACT

SNP array (SNPa) was developed to detect copy number alteration (CNA) and loss of heterozygosity (LOH) without copy number changes, CN-LOH. We aimed to identify novel genomic aberrations using SNPa in 31 WM with paired samples. Methylation status and mutation were analyzed on target genes. A total of 61 genetic aberrations were observed, 58 CNA (33 gains, 25 losses) in 58% of patients and CN-LOH in 6% of patients. The CNA were widely distributed throughout the genome, including 12 recurrent regions and identified new cryptic clonal chromosomal lesions that were mapped. Gene set expression analysis demonstrated a relationship between either deletion 6q or gain of chromosome 4 and alteration of gene expression profiling. We then studied methylation status and sought for mutations in altered regions on target genes. We observed methylation of DLEU7 on chromosome 13 in all patients (n = 12) with WM, and mutations of CD79B/CD79A genes (17q region), a key component of the BCR pathway, in 15% of cases. Most importantly, higher frequency of ≥3 CNA was observed in symptomatic WM. In conclusion, this study expands the view of the genomic complexity of WM, especially in symptomatic WM, including a potentially new mechanism of gene dysfunction, acquired uniparental disomy/CN-LOH. Finally, we have identified new potential target genes in WM, such as DLEU7 and CD79A/B.


Subject(s)
Chromosome Aberrations , DNA Copy Number Variations , DNA Methylation , Gene Expression Regulation , Loss of Heterozygosity , Mutation , Waldenstrom Macroglobulinemia/genetics , Adult , Aged , Aged, 80 and over , CD79 Antigens/genetics , CD79 Antigens/metabolism , Chromosome Deletion , Chromosome Duplication , Cohort Studies , Female , France , Genome-Wide Association Study , Humans , Male , Middle Aged , Neoplasm Proteins , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Waldenstrom Macroglobulinemia/metabolism
20.
Curr Res Transl Med ; 71(4): 103424, 2023.
Article in English | MEDLINE | ID: mdl-38011761

ABSTRACT

Myeloproliferative neoplasms, mastocytosis, myeloid/lymphoid neoplasms with hypereosinophilia and tyrosine kinase gene fusions, and myelodysplastic/myeloproliferative neoplasms are clonal hematopoietic cancers that, with the exception of certain entities, have an indolent course. In addition to their increasingly important role in the diagnosis of these entities, as shown by the recent classification of hematolymphoid tumors in the 5th edition of the World Health Organization and the International Consensus Classification of myeloid neoplasms and acute leukemias, identification of the profile of acquired genetic abnormalities is essential for adapting patient management and early detection of patients at high risk of progression. Alongside molecular abnormalities, cytogenetic abnormalities play an important role in the diagnosis, prognosis and follow-up of these diseases. Here, we review the recent literature on the impact of chromosomal abnormalities in these different entities and provide updated cytogenetic recommendations and guidelines for their management.


Subject(s)
Hematology , Mastocytosis , Myeloproliferative Disorders , Humans , Chromosome Aberrations , Cytogenetic Analysis , Mastocytosis/diagnosis , Mastocytosis/genetics , Mastocytosis/therapy , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL