73 results on '"Preudhomme, Claude"'
Search Results
2. Cytogénétique hématologique
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
3. Cytologie
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Groupes sanguins érythrocytaires
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
5. Examens complémentaires pour l’exploration d’un syndrome hémorragique
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
6. Exploration des proliférations myéloïdes
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
7. Exploration des pathologies érythrocytaires
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
8. Autres contextes clinico-biologiques
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
9. Anticorps anti-érythrocytaires
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
10. Exploration des anomalies de l’hémoglobine
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
11. Exploration des anomalies du métabolisme du fer et hémolyse
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
12. Autres techniques moléculaires
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
13. Maladie résiduelle moléculaire
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
14. Exploration des facteurs de risque biologiques de maladie thromboembolique
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
15. Suivi biologique des traitements antithrombotiques
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
16. Cytométrie en flux
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
17. Tests globaux et facteurs de coagulation
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
18. Exploration des proliférations lymphoïdes
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
19. Annexes
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
20. Diagnostic d’exclusion de maladie thromboembolique veineuse
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
21. Autres anticorps anti-éléments figurés du sang
- Author
-
Béné, Marie Christine, primary, Martinez-Aguilar, Patricia, additional, Lasne, Dominique, additional, Pirenne, France, additional, Ugo, Valérie, additional, Fischer, Anne-Marie, additional, Ajzenberg, Nadine, additional, Preudhomme, Claude, additional, and Maynadié, Marc, additional
- Published
- 2018
- Full Text
- View/download PDF
22. Genomic imbalance analysis provides new insight into prognostic factors in adult and pediatric T-ALL.
- Author
-
Balducci E, Simonin M, Duployez N, Steimlé T, Dourthe ME, Villarese P, Ducassou S, Arnoux I, Cayuela JM, Balsat M, Courtois L, Andrieu G, Touzart A, Huguet F, Petit A, Ifrah N, Dombret H, Baruchel A, Macintyre E, Preudhomme C, Boissel N, and Asnafi V
- Subjects
- Adolescent, Adult, Female, Humans, Male, Middle Aged, Young Adult, Chromosome Aberrations, Prognosis, Polymorphism, Single Nucleotide, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma diagnosis
- Abstract
Abstract: Given the poor outcome of refractory and relapsing T-cell acute lymphoblastic leukemia (T-ALL), identifying prognostic markers is still challenging. Using single nucleotide polymorphism (SNP) array analysis, we provide a comprehensive analysis of genomic imbalances in a cohort of 317 newly diagnosed patients with T-ALL including 135 children and 182 adults with respect to clinical and biological features and outcomes. SNP array results identified at least 1 somatic genomic imbalance in virtually all patients with T-ALL (∼96%). Del(9)(p21) (∼70%) and UPD(9)p21)/CDKN2A/B (∼28%) were the most frequent genomic imbalances. Unexpectedly del(13)(q14)/RB1/DLEU1 (∼14%) was the second most frequent copy number variant followed by del(6)(q15)/CASP8AP2 (∼11%), del(1)(p33)/SIL-TAL1 (∼11%), del(12)(p13)ETV6/CDKN1B (∼9%), del(18)(p11)/PTPN2 (∼9%), del(1)(p36)/RPL22 (∼9%), and del(17)(q11)/NF1/SUZ12 (∼8%). SNP array also revealed distinct profiles of genomic imbalances according to age, immunophenotype, and oncogenetic subgroups. In particular, adult patients with T-ALL demonstrated a significantly higher incidence of del(1)(p36)/RPL22, and del(13)(q14)/RB1/DLEU1, and lower incidence of del(9)(p21) and UPD(9p21)/CDKN2A/B. We determined a threshold of 15 genomic imbalances to stratify patients into high- and low-risk groups of relapse. Survival analysis also revealed the poor outcome, despite the low number of affected cases, conferred by the presence of chromothripsis (n = 6, ∼2%), del(16)(p13)/CREBBP (n = 15, ∼5%) as well as the newly-identified recurrent gain at 6q27 involving MLLT4 (n = 10, ∼3%). Genomic complexity, del(16)(p13)/CREBBP and gain at 6q27 involving MLLT4, maintained their significance in multivariate analysis for survival outcome. Our study thus demonstrated that whole genome analysis of imbalances provides new insights to refine risk stratification in T-ALL. This trial was registered at www.ClinicalTrials.gov as #NCT00222027 and #NCT00327678, and as #FRALLE 2000T trial., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
23. Prognostic impact of DDX41 germline mutations in intensively treated acute myeloid leukemia patients: an ALFA-FILO study.
- Author
-
Duployez N, Largeaud L, Duchmann M, Kim R, Rieunier J, Lambert J, Bidet A, Larcher L, Lemoine J, Delhommeau F, Hirsch P, Fenwarth L, Kosmider O, Decroocq J, Bouvier A, Le Bris Y, Ochmann M, Santagostino A, Adès L, Fenaux P, Thomas X, Micol JB, Gardin C, Itzykson R, Soulier J, Clappier E, Recher C, Preudhomme C, Pigneux A, Dombret H, Delabesse E, and Sébert M
- Subjects
- DEAD-box RNA Helicases genetics, Female, Germ-Line Mutation, Humans, Male, Prognosis, Prospective Studies, Retrospective Studies, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute therapy
- Abstract
DDX41 germline mutations (DDX41MutGL) are the most common genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia (AML). Recent reports suggest that DDX41MutGL myeloid malignancies could be considered as a distinct entity, even if their specific presentation and outcome remain to be defined. We describe here the clinical and biological features of 191 patients with DDX41MutGL AML. Baseline characteristics and outcome of 86 of these patients, treated with intensive chemotherapy in 5 prospective Acute Leukemia French Association/French Innovative Leukemia Organization trials, were compared with those of 1604 patients with DDX41 wild-type (DDX41WT) AML, representing a prevalence of 5%. Patients with DDX41MutGL AML were mostly male (75%), in their seventh decade, and with low leukocyte count (median, 2 × 109/L), low bone marrow blast infiltration (median, 33%), normal cytogenetics (75%), and few additional somatic mutations (median, 2). A second somatic DDX41 mutation (DDX41MutSom) was found in 82% of patients, and clonal architecture inference suggested that it could be the main driver for AML progression. DDX41MutGL patients displayed higher complete remission rates (94% vs 69%; P < .0001) and longer restricted mean overall survival censored at hematopoietic stem cell transplantation (HSCT) than 2017 European LeukemiaNet intermediate/adverse (Int/Adv) DDX41WT patients (5-year difference in restricted mean survival times, 13.6 months; P < .001). Relapse rates censored at HSCT were lower at 1 year in DDX41MutGL patients (15% vs 44%) but later increased to be similar to Int/Adv DDX41WT patients at 3 years (82% vs 75%). HSCT in first complete remission was associated with prolonged relapse-free survival (hazard ratio, 0.43; 95% confidence interval, 0.21-0.88; P = .02) but not with longer overall survival (hazard ratio, 0.77; 95% confidence interval, 0.35-1.68; P = .5)., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
24. 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party.
- Author
-
Heuser M, Freeman SD, Ossenkoppele GJ, Buccisano F, Hourigan CS, Ngai LL, Tettero JM, Bachas C, Baer C, Béné MC, Bücklein V, Czyz A, Denys B, Dillon R, Feuring-Buske M, Guzman ML, Haferlach T, Han L, Herzig JK, Jorgensen JL, Kern W, Konopleva MY, Lacombe F, Libura M, Majchrzak A, Maurillo L, Ofran Y, Philippe J, Plesa A, Preudhomme C, Ravandi F, Roumier C, Subklewe M, Thol F, van de Loosdrecht AA, van der Reijden BA, Venditti A, Wierzbowska A, Valk PJM, Wood BL, Walter RB, Thiede C, Döhner K, Roboz GJ, and Cloos J
- Subjects
- Europe, Flow Cytometry methods, High-Throughput Nucleotide Sequencing methods, Humans, Leukemia, Myeloid, Acute genetics, Neoplasm, Residual genetics, Prognosis, Leukemia, Myeloid, Acute diagnosis, Neoplasm, Residual diagnosis
- Abstract
Measurable residual disease (MRD) is an important biomarker in acute myeloid leukemia (AML) that is used for prognostic, predictive, monitoring, and efficacy-response assessments. The European LeukemiaNet (ELN) MRD Working Party evaluated standardization and harmonization of MRD in an ongoing manner and has updated the 2018 ELN MRD recommendations based on significant developments in the field. New and revised recommendations were established during in-person and online meetings, and a 2-stage Delphi poll was conducted to optimize consensus. All recommendations are graded by levels of evidence and agreement. Major changes include technical specifications for next-generation sequencing-based MRD testing and integrative assessments of MRD irrespective of technology. Other topics include use of MRD as a prognostic and surrogate end point for drug testing; selection of the technique, material, and appropriate time points for MRD assessment; and clinical implications of MRD assessment. In addition to technical recommendations for flow- and molecular-MRD analysis, we provide MRD thresholds and define MRD response, and detail how MRD results should be reported and combined if several techniques are used. MRD assessment in AML is complex and clinically relevant, and standardized approaches to application, interpretation, technical conduct, and reporting are of critical importance., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
25. Genetic identification of patients with AML older than 60 years achieving long-term survival with intensive chemotherapy.
- Author
-
Itzykson R, Fournier E, Berthon C, Röllig C, Braun T, Marceau-Renaut A, Pautas C, Nibourel O, Lemasle E, Micol JB, Adès L, Lebon D, Malfuson JV, Gastaud L, Goursaud L, Raffoux E, Wattebled KJ, Rousselot P, Thomas X, Chantepie S, Cluzeau T, Serve H, Boissel N, Terré C, Celli-Lebras K, Preudhomme C, Thiede C, Dombret H, Gardin C, and Duployez N
- Subjects
- Aged, Aged, 80 and over, Cytogenetics, Disease-Free Survival, Female, Follow-Up Studies, Humans, Male, Middle Aged, Survival Rate, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Mutation, Neoplasm Proteins genetics
- Abstract
To design a simple and reproducible classifier predicting the overall survival (OS) of patients with acute myeloid leukemia (AML) ≥60 years of age treated with 7 + 3, we sequenced 37 genes in 471 patients from the ALFA1200 (Acute Leukemia French Association) study (median age, 68 years). Mutation patterns and OS differed between the 84 patients with poor-risk cytogenetics and the 387 patients with good (n = 13), intermediate (n = 339), or unmeasured (n = 35) cytogenetic risk. TP53 (hazards ratio [HR], 2.49; P = .0003) and KRAS (HR, 3.60; P = .001) mutations independently worsened the OS of patients with poor-risk cytogenetics. In those without poor-risk cytogenetics, NPM1 (HR, 0.57; P = .0004), FLT3 internal tandem duplications with low (HR, 1.85; P = .0005) or high (HR, 3.51; P < 10-4) allelic ratio, DNMT3A (HR, 1.86; P < 10-4), NRAS (HR, 1.54; P = .019), and ASXL1 (HR, 1.89; P = .0003) mutations independently predicted OS. Combining cytogenetic risk and mutations in these 7 genes, 39.1% of patients could be assigned to a "go-go" tier with a 2-year OS of 66.1%, 7.6% to the "no-go" group (2-year OS 2.8%), and 3.3% of to the "slow-go" group (2-year OS of 39.1%; P < 10-5). Across 3 independent validation cohorts, 31.2% to 37.7% and 11.2% to 13.5% of patients were assigned to the go-go and the no-go tiers, respectively, with significant differences in OS between tiers in all 3 trial cohorts (HDF [Hauts-de-France], n = 141, P = .003; and SAL [Study Alliance Leukemia], n = 46; AMLSG [AML Study Group], n = 223, both P < 10-5). The ALFA decision tool is a simple, robust, and discriminant prognostic model for AML patients ≥60 years of age treated with intensive chemotherapy. This model can instruct the design of trials comparing the 7 + 3 standard of care with less intensive regimens., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
26. Prognostic significance of concurrent gene mutations in intensively treated patients with IDH-mutated AML: an ALFA study.
- Author
-
Duchmann M, Micol JB, Duployez N, Raffoux E, Thomas X, Marolleau JP, Braun T, Adès L, Chantepie S, Lemasle E, Berthon C, Malfuson JV, Pautas C, Lambert J, Boissel N, Celli-Lebras K, Caillot D, Turlure P, Vey N, Pigneux A, Recher C, Terré C, Gardin C, Itzykson R, Preudhomme C, Dombret H, and de Botton S
- Subjects
- Abnormal Karyotype, Aged, Chromosome Aberrations, Clinical Trials as Topic statistics & numerical data, DNA Methyltransferase 3A genetics, Disease-Free Survival, Female, France epidemiology, Humans, In Situ Hybridization, Fluorescence, Isocitrate Dehydrogenase deficiency, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Neoplasm Proteins deficiency, Nucleophosmin genetics, Proportional Hazards Models, Prospective Studies, Retrospective Studies, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute genetics, Neoplasm Proteins genetics, Point Mutation
- Abstract
In patients with isocitrate dehydrogenase (IDH)-mutated acute myeloid leukemia (AML) treated by intensive chemotherapy (IC), prognostic significance of co-occurring genetic alterations and allogeneic hematopoietic stem cell transplantation (HSCT) are of particular interest with the advent of IDH1/2 mutant inhibitors. We retrospectively analyzed 319 patients with newly diagnosed AML (127 with IDH1, 135 with IDH2R140, and 57 with IDH2R172 mutations) treated with IC in 3 Acute Leukemia French Association prospective trials. In each IDH subgroup, we analyzed the prognostic impact of clinical and genetic covariates, and the role of HSCT. In patients with IDH1 mutations, the presence of NPM1 mutations was the only variable predicting improved overall survival (OS) in multivariate analysis (P < .0001). In IDH2R140-mutated AML, normal karyotype (P = .008) and NPM1 mutations (P = .01) predicted better OS. NPM1 mutations were associated with better disease-free survival (DFS; P = .0009), whereas the presence of DNMT3A mutations was associated with shorter DFS (P = .0006). In IDH2R172-mutated AML, platelet count was the only variable retained in the multivariate model for OS (P = .002). Among nonfavorable European LeukemiaNet 2010-eligible patients, 71 (36%) underwent HSCT in first complete remission (CR1) and had longer OS (P = .03) and DFS (P = .02) than nontransplanted patients. Future clinical trials testing frontline IDH inhibitors combined with IC may consider stratification on NPM1 mutational status, the primary prognostic factor in IDH1- or IDH2R140-mutated AML. HSCT improve OS of nonfavorable IDH1/2-mutated AML and should be fully integrated into the treatment strategy., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
27. Germline PAX5 mutation predisposes to familial B-cell precursor acute lymphoblastic leukemia.
- Author
-
Duployez N, Jamrog LA, Fregona V, Hamelle C, Fenwarth L, Lejeune S, Helevaut N, Geffroy S, Caillault A, Marceau-Renaut A, Poulain S, Roche-Lestienne C, Largeaud L, Prade N, Dufrechou S, Hébrard S, Berthon C, Nelken B, Fernandes J, Villenet C, Figeac M, Gerby B, Delabesse E, Preudhomme C, and Broccardo C
- Subjects
- Adult, Child, Female, Genetic Predisposition to Disease, Humans, Male, Pedigree, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma congenital, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Germ-Line Mutation, PAX5 Transcription Factor genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2021
- Full Text
- View/download PDF
28. A personalized approach to guide allogeneic stem cell transplantation in younger adults with acute myeloid leukemia.
- Author
-
Fenwarth L, Thomas X, de Botton S, Duployez N, Bourhis JH, Lesieur A, Fortin G, Meslin PA, Yakoub-Agha I, Sujobert P, Dumas PY, Récher C, Lebon D, Berthon C, Michallet M, Pigneux A, Nguyen S, Chantepie S, Vey N, Raffoux E, Celli-Lebras K, Gardin C, Lambert J, Malfuson JV, Caillot D, Maury S, Ducourneau B, Turlure P, Lemasle E, Pautas C, Chevret S, Terré C, Boissel N, Socié G, Dombret H, Preudhomme C, and Itzykson R
- Subjects
- Adolescent, Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Clinical Decision-Making, Clinical Trials, Phase II as Topic statistics & numerical data, Combined Modality Therapy, Datasets as Topic, Female, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Models, Theoretical, Multicenter Studies as Topic statistics & numerical data, Neoplasm, Residual, Nuclear Proteins genetics, Nucleophosmin, Prognosis, Randomized Controlled Trials as Topic statistics & numerical data, Remission Induction, Risk Assessment, Transplantation, Homologous, Young Adult, Algorithms, Hematopoietic Stem Cell Transplantation standards, Leukemia, Myeloid, Acute therapy, Precision Medicine
- Abstract
A multistage model instructed by a large dataset (knowledge bank [KB] algorithm) has recently been developed to improve outcome predictions and tailor therapeutic decisions, including hematopoietic stem cell transplantation (HSCT) in acute myeloid leukemia (AML). We assessed the performance of the KB in guiding HSCT decisions in first complete remission (CR1) in 656 AML patients younger than 60 years from the ALFA-0702 trial (NCT00932412). KB predictions of overall survival (OS) were superior to those of European LeukemiaNet (ELN) 2017 risk stratification (C-index, 68.9 vs 63.0). Among patients reaching CR1, HSCT in CR1, as a time-dependent covariate, was detrimental in those with favorable ELN 2017 risk and those with negative NPM1 minimal residual disease (MRD; interaction tests, P = .01 and P = .02, respectively). Using KB simulations of survival at 5 years in a scenario without HSCT in CR1 (KB score), we identified, in a similar time-dependent analysis, a significant interaction between KB score and HSCT, with HSCT in CR1 being detrimental only in patients with a good prognosis based on KB simulations (KB score ≥40; interaction test, P = .01). We could finally integrate ELN 2017, NPM1 MRD, and KB scores to sort 545 CR1 patients into 278 (51.0%) HSCT candidates and 267 (49.0%) chemotherapy-only candidates. In both time-dependent and 6-month landmark analyses, HSCT significantly improved OS in HSCT candidates, whereas it significantly shortened OS in chemotherapy-only candidates. Integrating KB predictions with ELN 2017 and MRD may thus represent a promising approach to optimize HSCT timing in younger AML patients., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
29. Mutational profile and benefit of gemtuzumab ozogamicin in acute myeloid leukemia.
- Author
-
Fournier E, Duployez N, Ducourneau B, Raffoux E, Turlure P, Caillot D, Thomas X, Marceau-Renaut A, Chantepie S, Malfuson JV, Lemasle E, Cheok M, Celli-Lebras K, Guerin E, Terré C, Lambert J, Pautas C, Dombret H, Castaigne S, Preudhomme C, and Boissel N
- Subjects
- Aged, Antineoplastic Agents, Immunological adverse effects, Gemtuzumab adverse effects, Gene Expression Regulation, Leukemic, Humans, Leukemia, Myeloid, Acute genetics, Middle Aged, Proportional Hazards Models, Sialic Acid Binding Ig-like Lectin 3 genetics, Antineoplastic Agents, Immunological therapeutic use, Gemtuzumab therapeutic use, Leukemia, Myeloid, Acute drug therapy, Mutation
- Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous disease both in terms of genetic background and response to chemotherapy. Although molecular aberrations are routinely used to stratify AML patients into prognostic subgroups when receiving standard chemotherapy, the predictive value of the genetic background and co-occurring mutations remains to be assessed when using newly approved antileukemic drugs. In the present study, we retrospectively addressed the question of the predictive value of molecular events on the benefit of the addition of gemtuzumab ozogamicin (GO) to standard front-line chemotherapy. Using the more recent European LeukemiaNet (ELN) 2017 risk classification, we confirmed that the benefit of GO was restricted to the favorable (hazard ratio [HR], 0.54, 95% confidence interval [CI], 0.30-0.98) and intermediate (HR, 0.57; 95% CI, 0.33-1.00) risk categories, whereas it did not influence the outcome of patients within the adverse risk subgroup (HR, 0.93; 95% CI, 0.61-1.43). Interestingly, the benefit of GO was significant for patients with activating signaling mutations (HR, 0.43; 95% CI, 0.28-0.65), which correlated with higher CD33 expression levels. These results suggest that molecular aberrations could be critical for future differentially tailored treatments based on integrated genetic profiles that are able to predict the benefit of GO on outcome., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
30. Inherited transmission of the CSF3R T618I mutational hotspot in familial chronic neutrophilic leukemia.
- Author
-
Duployez N, Willekens C, Plo I, Marceau-Renaut A, de Botton S, Fenwarth L, Boyer T, Huet G, Nibourel O, Rose C, Nelken B, Quesnel B, and Preudhomme C
- Subjects
- Adolescent, Adult, Child, Female, Humans, Male, Pedigree, Signal Transduction, Genetic Predisposition to Disease, Leukemia, Neutrophilic, Chronic genetics, Leukemia, Neutrophilic, Chronic pathology, Mutation, Receptors, Colony-Stimulating Factor genetics
- Published
- 2019
- Full Text
- View/download PDF
31. Clonal interference of signaling mutations worsens prognosis in core-binding factor acute myeloid leukemia.
- Author
-
Itzykson R, Duployez N, Fasan A, Decool G, Marceau-Renaut A, Meggendorfer M, Jourdan E, Petit A, Lapillonne H, Micol JB, Cornillet-Lefebvre P, Ifrah N, Leverger G, Dombret H, Boissel N, Haferlach T, and Preudhomme C
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor, Child, Child, Preschool, Chromosome Aberrations, Female, Gene Expression Regulation, Leukemic, High-Throughput Nucleotide Sequencing, Humans, Infant, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Prognosis, Retrospective Studies, Survival Analysis, Young Adult, Clonal Evolution genetics, Core Binding Factors metabolism, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute metabolism, Mutation, Signal Transduction
- Abstract
Mutations in receptor tyrosine kinase/RAS signaling pathway genes are frequent in core-binding factor (CBF) acute myeloid leukemias (AMLs), but their prognostic relevance is debated. A subset of CBF AML patients harbors several signaling gene mutations. Genotyping of colonies and of relapse samples indicates that these arise in independent clones, thus defining a process of clonal interference (or parallel evolution). Clonal interference is pervasive in cancers, but the mechanisms underlying this process remain unclear, and its prognostic impact remains unknown. We analyzed a cohort of 445 adult and pediatric patients with CBF AML treated with intensive chemotherapy and with deep sequencing of 6 signaling genes ( KIT , NRAS , KRAS , FLT3 , JAK2 , CBL ). A total of 152 (34%), 167 (38%), and 126 (28%) patients harbored no, a single, and multiple signaling clones (clonal interference), respectively. Clonal interference of signaling mutations was associated with older age ( P = .004) and inv(16) subtype ( P = .025) but not with white blood cell count or mutations in chromatin or cohesin genes. The median allele frequency of signaling mutations was 31% in patients with a single clone or clonal interference ( P = .14). The repertoire of KIT , FLT3 , and NRAS / KRAS variants differed between groups. Clonal interference did not affect complete remission rate or minimal residual disease after 1-2 courses, but it did convey inferior event-free survival ( P < 10
-4 ), whereas the presence of a single signaling clone did not ( P = .44). This inferior outcome was independent of clinical parameters and of the presence of specific signaling clones. Our results suggest that specific clonal architectures can herald distinct prognoses in AML., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
32. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party.
- Author
-
Schuurhuis GJ, Heuser M, Freeman S, Béné MC, Buccisano F, Cloos J, Grimwade D, Haferlach T, Hills RK, Hourigan CS, Jorgensen JL, Kern W, Lacombe F, Maurillo L, Preudhomme C, van der Reijden BA, Thiede C, Venditti A, Vyas P, Wood BL, Walter RB, Döhner K, Roboz GJ, and Ossenkoppele GJ
- Subjects
- Clinical Trials as Topic, Consensus Development Conferences as Topic, Europe, Guidelines as Topic, Humans, Leukemia, Myeloid, Acute pathology, Neoplasm, Residual, Prognosis, United States, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute therapy
- Abstract
Measurable residual disease (MRD; previously termed minimal residual disease) is an independent, postdiagnosis, prognostic indicator in acute myeloid leukemia (AML) that is important for risk stratification and treatment planning, in conjunction with other well-established clinical, cytogenetic, and molecular data assessed at diagnosis. MRD can be evaluated using a variety of multiparameter flow cytometry and molecular protocols, but, to date, these approaches have not been qualitatively or quantitatively standardized, making their use in clinical practice challenging. The objective of this work was to identify key clinical and scientific issues in the measurement and application of MRD in AML, to achieve consensus on these issues, and to provide guidelines for the current and future use of MRD in clinical practice. The work was accomplished over 2 years, during 4 meetings by a specially designated MRD Working Party of the European LeukemiaNet. The group included 24 faculty with expertise in AML hematopathology, molecular diagnostics, clinical trials, and clinical medicine, from 19 institutions in Europe and the United States., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
33. Oncogenetic mutations combined with MRD improve outcome prediction in pediatric T-cell acute lymphoblastic leukemia.
- Author
-
Petit A, Trinquand A, Chevret S, Ballerini P, Cayuela JM, Grardel N, Touzart A, Brethon B, Lapillonne H, Schmitt C, Thouvenin S, Michel G, Preudhomme C, Soulier J, Landman-Parker J, Leverger G, Macintyre E, Baruchel A, and Asnafi V
- Subjects
- Adolescent, Child, Child, Preschool, Disease-Free Survival, Genes, Neoplasm, Humans, Infant, Infant, Newborn, Leukocyte Count, Neoplasm, Residual blood, Prognosis, Recurrence, Treatment Outcome, Mutation genetics, Neoplasm, Residual genetics, Oncogenes genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Risk stratification in childhood T-cell acute lymphoblastic leukemia (T-ALL) is mainly based on minimal residual disease (MRD) quantification. Whether oncogenetic mutation profiles can improve the discrimination of MRD-defined risk categories was unknown. Two hundred and twenty FRALLE2000T-treated patients were tested retrospectively for NOTCH1/FBXW7/RAS and PTEN alterations. Patients with NOTCH1/FBXW7 ( N/F ) mutations and RAS/PTEN ( R/P ) germ line (GL) were classified as oncogenetic low risk (gLoR; n = 111), whereas those with N/F GL and R/P GL mutations or N/F and R/P mutations were classified as high risk (gHiR; n = 109). Day 35 MRD status was available for 191 patients. Five-year cumulative incidence of relapse (CIR) and disease-free survival were 36% and 60% for gHiR patients and 11% and 89% for gLoR patients, respectively. Importantly, among the 60% of patients with MRD <10
-4 , 5-year CIR was 29% for gHiR patients and 4% for gLoR patients. Based on multivariable Cox models and stepwise selection, the 3 most discriminating variables were the oncogenetic classifier, MRD, and white blood cell (WBC) count. Patients harboring a WBC count ≥200 × 109 /L, gHiR classifier, and MRD ≥10-4 demonstrated a 5-year CIR of 46%, whereas the 58 patients (30%) with a WBC count <200 × 109 /L, gLoR classifier, and MRD <10-4 had a very low risk of relapse, with a 5-year CIR of only 2%. In childhood T-ALL, the N/F/R/P mutation profile is an independent predictor of relapse. When combined with MRD and a WBC count ≥200 × 109 /L, it identifies a significant subgroup of patients with a low risk of relapse., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
34. LXR agonist treatment of blastic plasmacytoid dendritic cell neoplasm restores cholesterol efflux and triggers apoptosis.
- Author
-
Ceroi A, Masson D, Roggy A, Roumier C, Chagué C, Gauthier T, Philippe L, Lamarthée B, Angelot-Delettre F, Bonnefoy F, Perruche S, Biichle S, Preudhomme C, Macintyre E, Lagrost L, Garnache-Ottou F, and Saas P
- Subjects
- ATP Binding Cassette Transporter 1 metabolism, ATP Binding Cassette Transporter, Subfamily G, Member 1 metabolism, Animals, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Dendritic Cells pathology, Female, Humans, Interleukin-3 metabolism, Liver X Receptors metabolism, Male, Mice, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Proto-Oncogene Proteins c-akt metabolism, STAT5 Transcription Factor metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Cholesterol metabolism, Dendritic Cells metabolism, Liver X Receptors agonists, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Blastic plasmacytoid dendritic cell (PDC) neoplasm (BPDCN) is an aggressive hematological malignancy with a poor prognosis that derives from PDCs. No consensus for optimal treatment modalities is available today and the full characterization of this leukemia is still emerging. We identified here a BPDCN-specific transcriptomic profile when compared with those of acute myeloid leukemia and T-acute lymphoblastic leukemia, as well as the transcriptomic signature of primary PDCs. This BPDCN gene signature identified a dysregulation of genes involved in cholesterol homeostasis, some of them being liver X receptor (LXR) target genes. LXR agonist treatment of primary BPDCN cells and BPDCN cell lines restored LXR target gene expression and increased cholesterol efflux via the upregulation of adenosine triphosphate-binding cassette (ABC) transporters, ABCA1 and ABCG1. LXR agonist treatment was responsible for limiting BPDCN cell proliferation and inducing intrinsic apoptotic cell death. LXR activation in BPDCN cells was shown to interfere with 3 signaling pathways associated with leukemic cell survival, namely: NF-κB activation, as well as Akt and STAT5 phosphorylation in response to the BPDCN growth/survival factor interleukin-3. These effects were increased by the stimulation of cholesterol efflux through a lipid acceptor, the apolipoprotein A1. In vivo experiments using a mouse model of BPDCN cell xenograft revealed a decrease of leukemic cell infiltration and BPDCN-induced cytopenia associated with increased survival after LXR agonist treatment. This demonstrates that cholesterol homeostasis is modified in BPDCN and can be normalized by treatment with LXR agonists which can be proposed as a new therapeutic approach., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
35. Comprehensive mutational profiling of core binding factor acute myeloid leukemia.
- Author
-
Duployez N, Marceau-Renaut A, Boissel N, Petit A, Bucci M, Geffroy S, Lapillonne H, Renneville A, Ragu C, Figeac M, Celli-Lebras K, Lacombe C, Micol JB, Abdel-Wahab O, Cornillet P, Ifrah N, Dombret H, Leverger G, Jourdan E, and Preudhomme C
- Subjects
- Adolescent, Adult, Alleles, Cell Cycle Proteins genetics, Child, Child, Preschool, Chromatin genetics, Chromatin ultrastructure, Chromosomal Proteins, Non-Histone genetics, Core Binding Factor Alpha 2 Subunit genetics, DNA Mutational Analysis, Female, Genetic Association Studies, High-Throughput Nucleotide Sequencing, Humans, Infant, Male, Middle Aged, Oncogene Proteins, Fusion genetics, Prognosis, RUNX1 Translocation Partner 1 Protein, Young Adult, Cohesins, Chromosome Inversion, Chromosomes, Human, Pair 16 genetics, Chromosomes, Human, Pair 21 genetics, Chromosomes, Human, Pair 8 genetics, Core Binding Factors genetics, DNA, Neoplasm genetics, Leukemia, Myeloid, Acute genetics, Mutation, Translocation, Genetic
- Abstract
Acute myeloid leukemia (AML) with t(8;21) or inv(16) have been recognized as unique entities within AML and are usually reported together as core binding factor AML (CBF-AML). However, there is considerable clinical and biological heterogeneity within this group of diseases, and relapse incidence reaches up to 40%. Moreover, translocations involving CBFs are not sufficient to induce AML on its own and the full spectrum of mutations coexisting with CBF translocations has not been elucidated. To address these issues, we performed extensive mutational analysis by high-throughput sequencing in 215 patients with CBF-AML enrolled in the Phase 3 Trial of Systematic Versus Response-adapted Timed-Sequential Induction in Patients With Core Binding Factor Acute Myeloid Leukemia and Treating Patients with Childhood Acute Myeloid Leukemia with Interleukin-2 trials (age, 1-60 years). Mutations in genes activating tyrosine kinase signaling (including KIT, N/KRAS, and FLT3) were frequent in both subtypes of CBF-AML. In contrast, mutations in genes that regulate chromatin conformation or encode members of the cohesin complex were observed with high frequencies in t(8;21) AML (42% and 18%, respectively), whereas they were nearly absent in inv(16) AML. High KIT mutant allele ratios defined a group of t(8;21) AML patients with poor prognosis, whereas high N/KRAS mutant allele ratios were associated with the lack of KIT or FLT3 mutations and a favorable outcome. In addition, mutations in epigenetic modifying or cohesin genes were associated with a poor prognosis in patients with tyrosine kinase pathway mutations, suggesting synergic cooperation between these events. These data suggest that diverse cooperating mutations may influence CBF-AML pathophysiology as well as clinical behavior and point to potential unique pathogenesis of t(8;21) vs inv(16) AML., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
36. The level of blast CD33 expression positively impacts the effect of gemtuzumab ozogamicin in patients with acute myeloid leukemia.
- Author
-
Olombel G, Guerin E, Guy J, Perrot JY, Dumezy F, de Labarthe A, Bastie JN, Legrand O, Raffoux E, Plesa A, Wagner-Ballon O, Cornet E, Salaun V, Preudhomme C, Thomas X, Pautas C, Chantepie S, Turlure P, Castaigne S, Dombret H, and Feuillard J
- Subjects
- Aged, Female, Gemtuzumab, Humans, Leukemia, Myeloid, Acute epidemiology, Male, Middle Aged, Survival Analysis, Treatment Outcome, Aminoglycosides therapeutic use, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Leukemia, Myeloid, Acute drug therapy, Sialic Acid Binding Ig-like Lectin 3 analysis
- Published
- 2016
- Full Text
- View/download PDF
37. Effect of lenalidomide treatment on clonal architecture of myelodysplastic syndromes without 5q deletion.
- Author
-
Chesnais V, Renneville A, Toma A, Lambert J, Passet M, Dumont F, Chevret S, Lejeune J, Raimbault A, Stamatoullas A, Rose C, Beyne-Rauzy O, Delaunay J, Solary E, Fenaux P, Dreyfus F, Preudhomme C, Kosmider O, and Fontenay M
- Subjects
- Aged, Anemia, Macrocytic drug therapy, Anemia, Macrocytic genetics, Anemia, Macrocytic pathology, Cell Proliferation drug effects, Cell Proliferation genetics, Chromosome Deletion, Chromosomes, Human, Pair 5 genetics, Clonal Evolution genetics, Clone Cells drug effects, Clone Cells metabolism, Clone Cells pathology, DNA Mutational Analysis, Erythropoietin administration & dosage, Female, Humans, Lenalidomide, Male, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology, Recombinant Proteins administration & dosage, Thalidomide administration & dosage, Thalidomide pharmacology, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Clonal Evolution drug effects, Myelodysplastic Syndromes drug therapy, Thalidomide analogs & derivatives
- Abstract
Non-del(5q) transfusion-dependent low/intermediate-1 myelodysplastic syndrome (MDS) patients achieve an erythroid response with lenalidomide in 25% of cases. Addition of an erythropoiesis-stimulating agent could improve response rate. The impact of recurrent somatic mutations identified in the diseased clone in response to lenalidomide and the drug's effects on clonal evolution remain unknown. We investigated recurrent mutations by next-generation sequencing in 94 non-del(5q) MDS patients randomized in the GFM-Len-Epo-08 clinical trial to lenalidomide or lenalidomide plus epoetin β. Clonal evolution was analyzed after 4 cycles of treatment in 42 cases and reanalyzed at later time points in 18 cases. The fate of clonal architecture of single CD34(+)CD38(-) hematopoietic stem cells was also determined in 5 cases. Mutation frequency was >10%: SF3B1 (74.5%), TET2 (45.7%), DNMT3A (20.2%), and ASXL1 (19.1%). Analysis of variant allele frequencies indicated a decrease of major mutations in 15 of 20 responders compared with 10 of 22 nonresponders after 4 cycles. The decrease in the variant allele frequency of major mutations was more significant in responders than in nonresponders (P < .001). Genotyping of single CD34(+)CD38(-) cell-derived colonies showed that the decrease in the size of dominant subclones could be associated with the rise of founding clones or of hematopoietic stem cells devoid of recurrent mutations. These effects remained transient, and disease escape was associated with the re-emergence of the dominant subclones. In conclusion, we show that, although the drug initially modulates the distribution of subclones, loss of treatment efficacy coincides with the re-expansion of the dominant subclone. This trial was registered at www.clinicaltrials.gov as #NCT01718379., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
38. Disease evolution and outcomes in familial AML with germline CEBPA mutations.
- Author
-
Tawana K, Wang J, Renneville A, Bödör C, Hills R, Loveday C, Savic A, Van Delft FW, Treleaven J, Georgiades P, Uglow E, Asou N, Uike N, Debeljak M, Jazbec J, Ancliff P, Gale R, Thomas X, Mialou V, Döhner K, Bullinger L, Mueller B, Pabst T, Stelljes M, Schlegelberger B, Wozniak E, Iqbal S, Okosun J, Araf S, Frank AK, Lauridsen FB, Porse B, Nerlov C, Owen C, Dokal I, Gribben J, Smith M, Preudhomme C, Chelala C, Cavenagh J, and Fitzgibbon J
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Disease Progression, Female, Genetic Predisposition to Disease, High-Throughput Nucleotide Sequencing, Humans, Infant, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Neoplasm Recurrence, Local mortality, Pedigree, Young Adult, CCAAT-Enhancer-Binding Proteins genetics, Germ-Line Mutation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology
- Abstract
In-depth molecular investigation of familial leukemia has been limited by the rarity of recognized cases. This study examines the genetic events initiating leukemia and details the clinical progression of disease across multiple families harboring germ-line CEBPA mutations. Clinical data were collected from 10 CEBPA-mutated families, representing 24 members with acute myeloid leukemia (AML). Whole-exome (WES) and deep sequencing were performed to genetically profile tumors and define patterns of clonal evolution. Germline CEBPA mutations clustered within the N-terminal and were highly penetrant, with AML presenting at a median age of 24.5 years (range, 1.75-46 years). In all diagnostic tumors tested (n = 18), double CEBPA mutations (CEBPAdm) were detected, with acquired (somatic) mutations preferentially targeting the C-terminal. Somatic CEBPA mutations were unstable throughout the disease course, with different mutations identified at recurrence. Deep sequencing of diagnostic and relapse paired samples confirmed that relapse-associated CEBPA mutations were absent at diagnosis, suggesting recurrence was triggered by novel, independent clones. Integrated WES and deep sequencing subsequently revealed an entirely new complement of mutations at relapse, verifying the presentation of a de novo leukemic episode. The cumulative incidence of relapse in familial AML was 56% at 10 years (n = 11), and 3 patients experienced ≥3 disease episodes over a period of 17 to 20 years. Durable responses to secondary therapies were observed, with prolonged median survival after relapse (8 years) and long-term overall survival (10-year overall survival, 67%). Our data reveal that familial CEBPA-mutated AML exhibits a unique model of disease progression, associated with favorable long-term outcomes., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
39. Frequent ASXL2 mutations in acute myeloid leukemia patients with t(8;21)/RUNX1-RUNX1T1 chromosomal translocations.
- Author
-
Micol JB, Duployez N, Boissel N, Petit A, Geffroy S, Nibourel O, Lacombe C, Lapillonne H, Etancelin P, Figeac M, Renneville A, Castaigne S, Leverger G, Ifrah N, Dombret H, Preudhomme C, Abdel-Wahab O, and Jourdan E
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Chromosomes, Human, Pair 21 genetics, Chromosomes, Human, Pair 8 genetics, Female, Gene Frequency, Humans, Leukemia, Myeloid, Acute classification, Leukemia, Myeloid, Acute drug therapy, Male, Middle Aged, Neoplasm, Residual genetics, RUNX1 Translocation Partner 1 Protein, Translocation, Genetic, Young Adult, Core Binding Factor Alpha 2 Subunit genetics, Leukemia, Myeloid, Acute genetics, Mutation, Oncogene Proteins, Fusion genetics, Repressor Proteins genetics
- Abstract
Acute myeloid leukemia (AML) with t(8;21) (q22;q22) is considered to have favorable risk; however, nearly half of t(8;21) patients are not cured, and recent studies have highlighted remarkable genetic heterogeneity in this subset of AML. Here we identify somatic mutations in additional sex combs-like 2 (ASXL2) in 22.7% (25/110) of patients with t(8;21), but not in patients with inv(16)/t(16;16) (0/60) or RUNX1-mutated AML (0/26). ASXL2 mutations were similarly frequent in adults and children t(8;21) and were mutually exclusive with ASXL1 mutations. Although overall survival was similar between ASXL1 and ASXL2 mutant t(8;21) AML patients and their wild-type counterparts, patients with ASXL1 or ASXL2 mutations had a cumulative incidence of relapse of 54.6% and 36.0%, respectively, compared with 25% in ASXL1/2 wild-type counterparts (P = .226). These results identify a high-frequency mutation in t(8;21) AML and identify the need for future studies to investigate the clinical and biological relevance of ASXL2 mutations in this unique subset of AML., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
40. SET-NUP214 is a recurrent γδ lineage-specific fusion transcript associated with corticosteroid/chemotherapy resistance in adult T-ALL.
- Author
-
Ben Abdelali R, Roggy A, Leguay T, Cieslak A, Renneville A, Touzart A, Banos A, Randriamalala E, Caillot D, Lioure B, Devidas A, Mossafa H, Preudhomme C, Ifrah N, Dombret H, Macintyre E, and Asnafi V
- Subjects
- Adrenal Cortex Hormones pharmacology, Adult, Antineoplastic Agents pharmacology, DNA-Binding Proteins, Female, Gene Rearrangement, delta-Chain T-Cell Antigen Receptor, Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor, Humans, Male, Middle Aged, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma immunology, Receptors, Antigen, T-Cell, gamma-delta genetics, Young Adult, Drug Resistance, Neoplasm genetics, Histone Chaperones genetics, Nuclear Pore Complex Proteins genetics, Oncogene Fusion, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Transcription Factors genetics
- Abstract
The SET-NUP214 (TAF1/CAN) fusion gene is a rare genetic event in T-cell acute lymphoblastic leukemia (T-ALL). Eleven (6%) of 196 T-ALL patients enrolled in the French Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) 2003 and 2005 trials harbored a SET-NUP214 transcript. SET-NUP214-positive patients were predominantly (10 [91%] of 11) T-cell receptor (TCR)-negative and strikingly associated with TCRγδ lineage T-ALLs, as defined by expression of TCRγδ, TCRδ and/or TCRγ rearrangements but no complete TCRβ variable diversity joining rearrangement in surface CD3/TCR-negative cases. When compared with SET-NUP214-negative patients, SET-NUP214-positive patients showed a significantly higher rate of corticosteroid resistance (91% vs 44%; P = .003) and chemotherapy resistance (100% vs 44%; P = .0001). All SET-NUP214-positive patients but one achieved complete remission, and 9 were allografted. Despite the poor early-treatment sensitivity, the outcome of SET-NUP214-positive patients was similar to that of SET-NUP214-negative patients.
- Published
- 2014
- Full Text
- View/download PDF
41. BCOR and BCORL1 mutations in myelodysplastic syndromes and related disorders.
- Author
-
Damm F, Chesnais V, Nagata Y, Yoshida K, Scourzic L, Okuno Y, Itzykson R, Sanada M, Shiraishi Y, Gelsi-Boyer V, Renneville A, Miyano S, Mori H, Shih LY, Park S, Dreyfus F, Guerci-Bresler A, Solary E, Rose C, Cheze S, Prébet T, Vey N, Legentil M, Duffourd Y, de Botton S, Preudhomme C, Birnbaum D, Bernard OA, Ogawa S, Fontenay M, and Kosmider O
- Subjects
- Acute Disease, Adult, Aged, Aged, 80 and over, Cohort Studies, Core Binding Factor Alpha 2 Subunit genetics, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A, DNA Mutational Analysis, Exome genetics, Female, Gene Expression Regulation, Neoplastic, Gene Frequency, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid genetics, Leukemia, Myelomonocytic, Chronic genetics, Male, Middle Aged, Multivariate Analysis, Reverse Transcriptase Polymerase Chain Reaction, Mutation, Myelodysplastic Syndromes genetics, Proto-Oncogene Proteins genetics, Repressor Proteins genetics
- Abstract
Patients with low-risk myelodysplastic syndromes (MDS) that rapidly progress to acute myeloid leukemia (AML) remain a challenge in disease management. Using whole-exome sequencing of an MDS patient, we identified a somatic mutation in the BCOR gene also mutated in AML. Sequencing of BCOR and related BCORL1 genes in a cohort of 354 MDS patients identified 4.2% and 0.8% of mutations respectively. BCOR mutations were associated with RUNX1 (P = .002) and DNMT3A mutations (P = .015). BCOR is also mutated in chronic myelomonocytic leukemia patients (7.4%) and BCORL1 in AML patients with myelodysplasia-related changes (9.1%). Using deep sequencing, we show that BCOR mutations arise after mutations affecting genes involved in splicing machinery or epigenetic regulation. In univariate analysis, BCOR mutations were associated with poor prognosis in MDS (overall survival [OS]: P = .013; cumulative incidence of AML transformation: P = .005). Multivariate analysis including age, International Prognostic Scoring System, transfusion dependency, and mutational status confirmed a significant inferior OS to patients with a BCOR mutation (hazard ratio, 3.3; 95% confidence interval, 1.4-8.1; P = .008). These data suggest that BCOR mutations define the clinical course rather than disease initiation. Despite infrequent mutations, BCOR analyses should be considered in risk stratification.
- Published
- 2013
- Full Text
- View/download PDF
42. MYD88 L265P mutation in Waldenstrom macroglobulinemia.
- Author
-
Poulain S, Roumier C, Decambron A, Renneville A, Herbaux C, Bertrand E, Tricot S, Daudignon A, Galiègue-Zouitina S, Soenen V, Theisen O, Grardel N, Nibourel O, Roche-Lestienne C, Quesnel B, Duthilleul P, Preudhomme C, and Leleu X
- Subjects
- 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, Myeloid Differentiation Factor 88 genetics, Point Mutation, Waldenstrom Macroglobulinemia genetics
- 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.
- Published
- 2013
- Full Text
- View/download PDF
43. Clonal architecture of chronic myelomonocytic leukemias.
- Author
-
Itzykson R, Kosmider O, Renneville A, Morabito M, Preudhomme C, Berthon C, Adès L, Fenaux P, Platzbecker U, Gagey O, Rameau P, Meurice G, Oréar C, Delhommeau F, Bernard OA, Fontenay M, Vainchenker W, Droin N, and Solary E
- Subjects
- Aged, Aged, 80 and over, Case-Control Studies, Cell Differentiation genetics, Clonal Evolution immunology, Cohort Studies, Female, Hematopoiesis genetics, Hematopoiesis immunology, Humans, Leukemia, Myelomonocytic, Chronic immunology, Loss of Heterozygosity, Male, Middle Aged, Mutation physiology, Mutation Rate, Myeloid Cells metabolism, Myeloid Cells physiology, Clonal Evolution genetics, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic pathology
- Abstract
Genomic studies in chronic myeloid malignancies, including myeloproliferative neoplasms (MPN), myelodysplastic syndromes (MDS), and MPN/MDS, have identified common mutations in genes encoding signaling, epigenetic, transcription, and splicing factors. In the present study, we interrogated the clonal architecture by mutation-specific discrimination analysis of single-cell-derived colonies in 28 patients with chronic myelomonocytic leukemias (CMML), the most frequent MPN/MDS. This analysis reveals a linear acquisition of the studied mutations with limited branching through loss of heterozygosity. Serial analysis of untreated and treated samples demonstrates a dynamic architecture on which most current therapeutic approaches have limited effects. The main disease characteristics are early clonal dominance, arising at the CD34(+)/CD38(-) stage of hematopoiesis, and granulomonocytic differentiation skewing of multipotent and common myeloid progenitors. Comparison of clonal expansions of TET2 mutations in MDS, MPN, and CMML, together with functional invalidation of TET2 in sorted progenitors, suggests a causative link between early clonal dominance and skewed granulomonocytic differentiation. Altogether, early clonal dominance may distinguish CMML from other chronic myeloid neoplasms with similar gene mutations.
- Published
- 2013
- Full Text
- View/download PDF
44. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia.
- Author
-
Jourdan E, Boissel N, Chevret S, Delabesse E, Renneville A, Cornillet P, Blanchet O, Cayuela JM, Recher C, Raffoux E, Delaunay J, Pigneux A, Bulabois CE, Berthon C, Pautas C, Vey N, Lioure B, Thomas X, Luquet I, Terré C, Guardiola P, Béné MC, Preudhomme C, Ifrah N, and Dombret H
- Subjects
- Adolescent, Adult, Antimetabolites, Antineoplastic administration & dosage, Cytarabine administration & dosage, DNA Mutational Analysis, Drug Resistance, Neoplasm genetics, Female, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Male, Middle Aged, Neoplasm, Residual, Prognosis, Prospective Studies, Treatment Outcome, Young Adult, Core Binding Factors genetics, Genes, Neoplasm genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mutation physiology
- Abstract
Not all patients with core binding factor acute myeloid leukemia (CBF-AML) display a good outcome. Modern risk factors include KIT and/or FLT3 gene mutations and minimal residual disease (MRD) levels, but their respective values have never been prospectively assessed. A total of 198 CBF-AML patients were randomized between a reinforced and a standard induction course, followed by 3 high-dose cytarabine consolidation courses. MRD levels were monitored prospectively. Gene mutations were screened at diagnosis. Despite a more rapid MRD decrease after reinforced induction, induction arm did not influence relapse-free survival (RFS) (64% in both arms; P = .91). Higher WBC, KIT, and/or FLT3-ITD/TKD gene mutations, and a less than 3-log MRD reduction after first consolidation, were associated with a higher specific hazard of relapse, but MRD remained the sole prognostic factor in multivariate analysis. At 36 months, cumulative incidence of relapse and RFS were 22% vs 54% (P < .001) and 73% vs 44% (P < .001) in patients who achieved 3-log MRD reduction vs the others. These results suggest that MRD, rather than gene mutations, should be used for future treatment stratifications in CBF-AML patients. This trial was registered at EudraCT as #2006-005163-26 and at www.clinicaltrials.gov as #NCT 00428558.
- Published
- 2013
- Full Text
- View/download PDF
45. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study.
- Author
-
Castaigne S, Pautas C, Terré C, Raffoux E, Bordessoule D, Bastie JN, Legrand O, Thomas X, Turlure P, Reman O, de Revel T, Gastaud L, de Gunzburg N, Contentin N, Henry E, Marolleau JP, Aljijakli A, Rousselot P, Fenaux P, Preudhomme C, Chevret S, and Dombret H
- Subjects
- Aged, Aminoglycosides adverse effects, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents adverse effects, Disease-Free Survival, Female, Gemtuzumab, Humans, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Survival Rate, Aminoglycosides therapeutic use, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
Background: The results of the addition of gemtuzumab ozogamicin, an anti-CD33 antibody conjugate, to the standard treatment for patients with acute myeloid leukaemia in phase 3 trials were contradictory. We investigated whether the addition of low fractionated-dose gemtuzumab ozogamicin to standard front-line chemotherapy would improve the outcome of patients with this leukaemia without causing excessive toxicity., Methods: In a phase 3, open-label study, undertaken in 26 haematology centres in France, patients aged 50-70 years with previously untreated de novo acute myeloid leukaemia were randomly assigned with a computer-generated sequence in a 1:1 ratio with block sizes of four to standard treatment (control group) with or without five doses of intravenous gemtuzumab ozogamicin (3 mg/m(2) on days 1, 4, and 7 during induction and day 1 of each of the two consolidation chemotherapy courses). The primary endpoint was event-free survival (EFS). Secondary endpoints were relapse-free (RFS), overall survival (OS), and safety. Analysis was by intention to treat. This study is registered with EudraCT, number 2007-002933-36., Findings: 280 patients were randomly assigned to the control (n=140) and gemtuzumab ozogamicin groups (n=140), and 139 patients were analysed in each group. Complete response with or without incomplete platelet recovery to induction was 104 (75%) in the control group and 113 (81%) in the gemtuzumab ozogamicin group (odds ratio 1·46, 95% CI 0·20-2·59; p=0·25). At 2 years, EFS was estimated as 17·1% (10·8-27·1) in the control group versus 40·8% (32·8-50·8) in the gemtuzumab ozogamicin group (hazard ratio 0·58, 0·43-0·78; p=0·0003), OS 41·9% (33·1-53·1) versus 53·2% (44·6-63·5), respectively (0·69, 0·49-0·98; p=0·0368), and RFS 22·7% (14·5-35·7) versus 50·3% (41·0-61·6), respectively (0·52, 0·36-0·75; p=0·0003). Haematological toxicity, particularly persistent thrombocytopenia, was more common in the gemtuzumab ozogamicin group than in the control group (22 [16%] vs 4 [3%]; p<0·0001), without an increase in the risk of death from toxicity., Interpretation: The use of fractionated lower doses of gemtuzumab ozogamicin allows the safe delivery of higher cumulative doses and substantially improves outcomes in patients with acute myeloid leukaemia. The findings warrant reassessment of gemtuzumab ozogamicin as front-line therapy for acute myeloid leukaemia., Funding: Wyeth (Pfizer)., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
46. Mutations affecting mRNA splicing define distinct clinical phenotypes and correlate with patient outcome in myelodysplastic syndromes.
- Author
-
Damm F, Kosmider O, Gelsi-Boyer V, Renneville A, Carbuccia N, Hidalgo-Curtis C, Della Valle V, Couronné L, Scourzic L, Chesnais V, Guerci-Bresler A, Slama B, Beyne-Rauzy O, Schmidt-Tanguy A, Stamatoullas-Bastard A, Dreyfus F, Prébet T, de Botton S, Vey N, Morgan MA, Cross NC, Preudhomme C, Birnbaum D, Bernard OA, and Fontenay M
- Subjects
- Adult, Aged, Aged, 80 and over, Cell Transformation, Neoplastic genetics, Female, Genetic Association Studies, Humans, Leukemia, Myeloid, Acute genetics, Male, Middle Aged, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes mortality, Nuclear Proteins genetics, Phosphoproteins genetics, Prognosis, RNA Splicing Factors, Ribonucleoprotein, U2 Small Nuclear genetics, Ribonucleoproteins genetics, Serine-Arginine Splicing Factors, Splicing Factor U2AF, Survival Analysis, Mutation, Myelodysplastic Syndromes genetics, Phenotype, RNA Splicing genetics
- Abstract
A cohort of MDS patients was examined for mutations affecting 4 splice genes (SF3B1, SRSF2, ZRSR2, and U2AF35) and evaluated in the context of clinical and molecular markers. Splice gene mutations were detected in 95 of 221 patients. These mutations were mutually exclusive and less likely to occur in patients with complex cytogenetics or TP53 mutations. SF3B1(mut) patients presented with lower hemoglobin levels, increased WBC and platelet counts, and were more likely to have DNMT3A mutations. SRSF2(mut) patients clustered in RAEB-1 and RAEB-2 subtypes and exhibited pronounced thrombocytopenias. ZRSR2(mut) patients clustered in International Prognostic Scoring System intermediate-1 and intermediate-2 risk groups, had higher percentages of bone marrow blasts, and more often displayed isolated neutropenias. SRSF2 and ZRSR2 mutations were more common in TET2(mut) patients. U2AF35(mut) patients had an increased prevalence of chromosome 20 deletions and ASXL1 mutations. Multivariate analysis revealed an inferior overall survival and a higher AML transformation rate for the genotype ZRSR2(mut)/TET2(wt) (overall survival: hazard ratio = 3.3; 95% CI, 1.4-7.7; P = .006; AML transformation: hazard ratio = 3.6; 95% CI, 2-4.2; P = .026). Our results demonstrate that splice gene mutations are among the most frequent molecular aberrations in myelodysplastic syndrome, define distinct clinical phenotypes, and show preferential associations with mutations targeting transcriptional regulation.
- Published
- 2012
- Full Text
- View/download PDF
47. Changes in the dynamics of the excess mortality rate in chronic phase-chronic myeloid leukemia over 1990-2007: a population study.
- Author
-
Corm S, Roche L, Micol JB, Coiteux V, Bossard N, Nicolini FE, Iwaz J, Preudhomme C, Roche-Lestienne C, Facon T, and Remontet L
- Subjects
- 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, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Chronic-Phase drug therapy, Leukemia, Myeloid, Chronic-Phase mortality, Piperazines therapeutic use, Pyrimidines therapeutic use
- 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.
- Published
- 2011
- Full Text
- View/download PDF
48. Molecular predictors of response to decitabine in advanced chronic myelomonocytic leukemia: a phase 2 trial.
- Author
-
Braun T, Itzykson R, Renneville A, de Renzis B, Dreyfus F, Laribi K, Bouabdallah K, Vey N, Toma A, Recher C, Royer B, Joly B, Vekhoff A, Lafon I, Sanhes L, Meurice G, Oréar C, Preudhomme C, Gardin C, Ades L, Fontenay M, Fenaux P, Droin N, and Solary E
- Subjects
- Aged, Aged, 80 and over, Azacitidine therapeutic use, Decitabine, Female, Gene Expression Regulation, Leukemic, Humans, Leukemia, Myelomonocytic, Chronic diagnosis, Male, Middle Aged, Mutation, Prognosis, Survival Analysis, Antimetabolites, Antineoplastic therapeutic use, Azacitidine analogs & derivatives, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic genetics
- Abstract
Hydroxyurea is the standard therapy of chronic myelomonocytic leukemia (CMML) presenting with advanced myeloproliferative and/or myelodysplastic features. Response to hypomethylating agents has been reported in heterogeneous series of CMML. We conducted a phase 2 trial of decitabine (DAC) in 39 patients with advanced CMML defined according to a previous trial. Median number of DAC cycles was 10 (range, 1-24). Overall response rate was 38% with 4 complete responses (10%), 8 marrow responses (21%), and 3 stable diseases with hematologic improvement (8%). Eighteen patients (46%) demonstrated stable disease without hematologic improvement, and 6 (15%) progressed to acute leukemia. With a median follow-up of 23 months, overall survival was 48% at 2 years. Mutations in ASXL1, TET2, AML1, NRAS, KRAS, CBL, FLT3, and janus kinase 2 (JAK2) genes, and hypermethylation of the promoter of the tumor suppressor gene TIF1γ, did not predict response or survival on DAC therapy. Lower CJUN and CMYB gene expression levels independently predicted improved overall survival. This trial confirmed DAC efficacy in approximately 40% of CMML patients with advanced myeloproliferative or myelodysplastic features and suggested that CJUN and CMYB expression could be potential biomarkers in this setting. This trial is registered at EudraCT (eudract.ema.europa.eu) as #2008-000470-21 and www.clinicaltrials.gov as #NCT01098084.
- Published
- 2011
- Full Text
- View/download PDF
49. Comparison of high-dose cytarabine and timed-sequential chemotherapy as consolidation for younger adults with AML in first remission: the ALFA-9802 study.
- Author
-
Thomas X, Elhamri M, Raffoux E, Renneville A, Pautas C, de Botton S, de Revel T, Reman O, Terré C, Gardin C, Chelghoum Y, Boissel N, Quesnel B, Hicheri Y, Bourhis JH, Fenaux P, Preudhomme C, Michallet M, Castaigne S, and Dombret H
- Subjects
- Adolescent, Adult, Age Factors, Cytogenetic Analysis, Disease-Free Survival, Drug Therapy methods, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Middle Aged, Nucleophosmin, Prospective Studies, Remission Induction, Young Adult, Antimetabolites, Antineoplastic administration & dosage, Antimetabolites, Antineoplastic therapeutic use, Cytarabine administration & dosage, Cytarabine therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
To assess the value of administering timed-sequential chemotherapy (TSC; 2 therapeutic sequences separated by a 4-day interval-free chemotherapy) or high-dose cytarabine (HDAraC) cycles in consolidation therapy for acute myeloid leukemia (AML), 459 patients 15 to 50 years of age were enrolled in the prospective randomized Acute Leukemia French Association-9802 trial. Complete remission was achieved in 89%. A total of 237 patients were then randomized to either TSC consolidation (120 patients) or HDAraC consolidation cycles (117 patients). Overall, there was no significant difference between the 2 consolidation arms (5-year event-free survival [EFS]: 41% for HDAraC vs 35% for TSC), or cumulative incidence of relapse, or treatment-related mortality. Cytogenetically normal AML NPM1(+) or CEBPA(+) and FLT3-ITD(-) had the same outcome as those with favorable cytogenetics. When considering favorable and unfavorable risk groups, the trend was in favor of HDAraC. However, the difference became significant when considering intermediate cytogenetics (5-year EFS: 49% vs 29%; P = .02), especially cytogenetically normal AML (5-year EFS: 48% vs 31%; P = .04), which was related to lower relapse rate and less toxicity. This study demonstrates that TSC did not produce any benefit when used as consolidation therapy in younger adults compared with HDAraC. This trial was registered at www.clinicaltrials.gov as #NCT00880243.
- Published
- 2011
- Full Text
- View/download PDF
50. Myelodysplasia and leukemia of Fanconi anemia are associated with a specific pattern of genomic abnormalities that includes cryptic RUNX1/AML1 lesions.
- Author
-
Quentin S, Cuccuini W, Ceccaldi R, Nibourel O, Pondarre C, Pagès MP, Vasquez N, Dubois d'Enghien C, Larghero J, Peffault de Latour R, Rocha V, Dalle JH, Schneider P, Michallet M, Michel G, Baruchel A, Sigaux F, Gluckman E, Leblanc T, Stoppa-Lyonnet D, Preudhomme C, Socié G, and Soulier J
- Subjects
- Adolescent, Adult, Bone Marrow physiology, Child, Child, Preschool, Fanconi Anemia complications, Female, Gene Dosage genetics, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Genes, Tumor Suppressor, Homozygote, Humans, Leukemia, Myeloid, Acute etiology, Male, Middle Aged, Myelodysplastic Syndromes etiology, Nucleophosmin, Polymorphism, Single Nucleotide, Young Adult, Core Binding Factor Alpha 2 Subunit genetics, Fanconi Anemia genetics, Genomic Instability genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics
- Abstract
Fanconi anemia (FA) is a genetic condition associated with bone marrow (BM) failure, myelodysplasia (MDS), and acute myeloid leukemia (AML). We studied 57 FA patients with hypoplastic or aplastic anemia (n = 20), MDS (n = 18), AML (n = 11), or no BM abnormality (n = 8). BM samples were analyzed by karyotype, high-density DNA arrays with respect to paired fibroblasts, and by selected oncogene sequencing. A specific pattern of chromosomal abnormalities was found in MDS/AML, which included 1q+ (44.8%), 3q+ (41.4%), -7/7q (17.2%), and 11q- (13.8%). Moreover, cryptic RUNX1/AML1 lesions (translocations, deletions, or mutations) were observed for the first time in FA (20.7%). Rare mutations of NRAS, FLT3-ITD, MLL-PTD, ERG amplification, and ZFP36L2-PRDM16 translocation, but no TP53, TET2, CBL, NPM1, and CEBPα mutations were found. Frequent homozygosity regions were related not to somatic copy-neutral loss of heterozygosity but to consanguinity, suggesting that homologous recombination is not a common progression mechanism in FA. Importantly, the RUNX1 and other chromosomal/genomic lesions were found at the MDS/AML stages, except for 1q+, which was found at all stages. These data have implications for staging and therapeutic managing in FA patients, and also to analyze the mechanisms of clonal evolution and oncogenesis in a background of genomic instability and BM failure.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.