Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Am J Hematol ; 99(6): 1108-1118, 2024 06.
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.
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
4.
Eur J Med Genet ; 65(11): 104603, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36049610

ABSTRACT

TRIT1 encodes a tRNA isopentenyl transferase that allows a strong interaction between the mini helix and the codon. Recent reports support the TRIT1 bi-allelic alterations as the cause of an autosomal recessive disorder, named combined oxydative phophorylation deficiency 35, with microcephaly, developmental disability, and epilepsy. The phenotype is due to decreased mitochondrial function, with deficit of i6A37 in cytosolic and mitochondrial tRNA. Only 10 patients have been reported. We report on two new patients with four novel variants, and confirm the published clinical TRIT1 deficient phenotype stressing the possibility of both very severe, with generalized pharmaco-resistant seizures, and mild phenotypes.


Subject(s)
Alkyl and Aryl Transferases , Microcephaly , Humans , Alkyl and Aryl Transferases/genetics , Alleles , Codon , Microcephaly/genetics , Mitochondria/genetics , Phenotype , RNA, Transfer
10.
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
13.
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
14.
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
15.
Oncotarget ; 8(34): 57451-57459, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28924457

ABSTRACT

Approximately 30% of the patients who fulfil the criteria of Waldenström's macroglobulinemia (WM) are diagnosed while asymptomatic (indolent), and will not require immediate therapy. Conversely, patients with a disease-related event will be considered for therapy. The physiopathology of these 2 groups remains unclear, and the mechanisms of progression from indolent to symptomatic WM have yet to be fully understood. Seventeen patients diagnosed with WM were included in this study, 8 asymptomatic WM (A-WM) and 9 symptomatic WM (S-WM). A differential analysis was performed on a first series of 11 patients and identified 48 genes whose expression separated samples from A- to S-WM. This gene signature was then confirmed on a second independent validation set of 6 WM. Within this expression profile, BACH2, a B-cell transcription factor known to be a tumor suppressor gene, was found to be over-expressed in A-MW relatively to S-MW. We specifically over-expressed BACH2 in a WM-related cell line and observed a significant reduction of the clonogenic activity. To the best of our knowledge, we report for the first time a specific gene expression signature that differentiates A-WM and S-WM. Within this expression profile, BACH2 was identified as a candidate gene that may help to understand better the behavior of tumor cells in indolent WM.

16.
Clin Cancer Res ; 23(20): 6325-6335, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28754818

ABSTRACT

Purpose:TP53 is a tumor-suppressor gene that functions as a regulator influencing cellular responses to DNA damage, and TP53 alterations are associated with pejorative outcome in most B-lymphoid disorders. Little is known regarding TP53 alteration in Waldenstrom's macroglobulinemia (WM).Experimental Design: Here, we have explored the incidence of TP53 alteration using Sanger sequencing and ultradeep-targeted sequencing in 125 WM and 10 immunoglobulin M (IgM) monoclonal gammopathy of undetermined significance (MGUS), along with the clinical features and the associated genomic landscape using single-nucleotide polymorphism array and mutational landscape in an integrative study.Results: Overall, we have identified alteration of TP53 locus including mutation, deletion, and copy-neutral LOH in 11.2% of WM. TP53 mutation was acquired in 7.3% of patients with WM at diagnosis, being absent in IgM MGUS, and was highly correlated to deletion 17p. No correlation with CXCR4 mutations was observed. Patients with TP53 alteration had a greater number of genomic abnormalities. Importantly, WM with TP53 alteration had a significantly shorter overall survival, particularly in symptomatic WM, and independently of the international prognostic scoring system for Waldenstrom macroglobulinemia (IPSSWM) score. Specific treatment for WM with TP53 may have to be studied. Nutlin-3a-targeted p53 signaling induced cytotoxicity preclinically, along with new compounds such as ibrutinib, PrimaMet, or CP31398 that bypass p53 pathway in WM, paving the path for future treatment-tailored options.Conclusions: Our results highlight the clinical significance of detection of TP53 alteration in WM to determine the prognosis of WM and guide the treatment choice. Clin Cancer Res; 23(20); 6325-35. ©2017 AACR.


Subject(s)
Mutation , Tumor Suppressor Protein p53/genetics , Waldenstrom Macroglobulinemia/diagnosis , Waldenstrom Macroglobulinemia/genetics , Adult , Aged , Aged, 80 and over , Apoptosis , Cell Survival/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17 , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Prognosis , Protein Binding , Protein Interaction Domains and Motifs/genetics , Survival Analysis , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Waldenstrom Macroglobulinemia/mortality
17.
Sci Transl Med ; 9(389)2017 05 10.
Article in English | MEDLINE | ID: mdl-28490664

ABSTRACT

Multiple myeloma (MM) is a frequently incurable hematological cancer in which overactivity of MYC plays a central role, notably through up-regulation of ribosome biogenesis and translation. To better understand the oncogenic program driven by MYC and investigate its potential as a therapeutic target, we screened a chemically diverse small-molecule library for anti-MM activity. The most potent hits identified were rocaglate scaffold inhibitors of translation initiation. Expression profiling of MM cells revealed reversion of the oncogenic MYC-driven transcriptional program by CMLD010509, the most promising rocaglate. Proteome-wide reversion correlated with selective depletion of short-lived proteins that are key to MM growth and survival, most notably MYC, MDM2, CCND1, MAF, and MCL-1. The efficacy of CMLD010509 in mouse models of MM confirmed the therapeutic relevance of these findings in vivo and supports the feasibility of targeting the oncogenic MYC-driven translation program in MM with rocaglates.


Subject(s)
Multiple Myeloma/genetics , Multiple Myeloma/therapy , Animals , Cell Line, Tumor , Cyclin D1/genetics , Humans , Mice , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays
18.
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
19.
Oncotarget ; 7(38): 62377-62385, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27566555

ABSTRACT

CD81 is a cell surface protein which belongs to the tetraspanin family. While in multiple myeloma its expression on plasma cells is associated with worse prognosis, this has not yet been explored in acute myeloid leukemia (AML). We measured membrane expression of CD81 on AML cells at diagnosis, evaluated its association with AML characteristics and its influence on patient outcome after intensive chemotherapy in a cohort of 134 patients. CD81 was detected in 92/134 (69%) patients. Patients with AML expressing CD81 had elevated leukocyte count (P=0.02) and were more likely classified as intermediate or adverse-risk by cytogenetics (P<0.001). CD81 expression had a negative impact on survival (event-free survival, overall survival and relapse-free survival) in univariate (P<0.001) and in multivariate analyses (P=0.003, 0.002 and <0.001, respectively). CD81 has a negative impact on OS in patients with NPM1 mutation (P=0.01) and in patients ELN-favorable (P=0.002). In conclusion, this cell surface marker may be a new prognostic marker for diagnostic risk classification and a potential therapeutic target for drug development in AML.


Subject(s)
Biomarkers, Tumor/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Tetraspanin 28/metabolism , Adult , Age Factors , Aged , Cell Membrane/metabolism , Disease-Free Survival , Female , Flow Cytometry , Follow-Up Studies , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/ultrastructure , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/genetics , Leukocyte Count , Male , Middle Aged , Mutation , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Young Adult
20.
Ann Biol Clin (Paris) ; 74(5): 511-515, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27477825

ABSTRACT

Cytogenetic evaluation is one the most important criteria for diagnosis and response to treatment in chronic myeloid leukemia, and recent baseline prognostic factors including particular additional clonal cytogenetic abnormalities have been established. The French cytogenetic group in hematology GFCH proposes here an updating of recommendations for cytogenetic assessment of CML in the era of tyrosine kinase inhibitors.


Subject(s)
Cytogenetic Analysis/standards , Hematology/standards , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Chromosome Aberrations , Cytogenetic Analysis/methods , Cytogenetic Analysis/trends , France , Hematology/organization & administration , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Monitoring, Physiologic/methods , Monitoring, Physiologic/standards , Societies, Medical , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL