36 results on '"Dombret, H."'
Search Results
2. An early thymic precursor phenotype predicts outcome exclusively in HOXA-overexpressing adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study
- Author
-
Bond, J., primary, Marchand, T., additional, Touzart, A., additional, Cieslak, A., additional, Trinquand, A., additional, Sutton, L., additional, Radford-Weiss, I., additional, Lhermitte, L., additional, Spicuglia, S., additional, Dombret, H., additional, Macintyre, E., additional, Ifrah, N., additional, Hamel, J.-F., additional, and Asnafi, V., additional
- Published
- 2016
- Full Text
- View/download PDF
3. A phase I study of danusertib (PHA-739358) in adult patients with accelerated or blastic phase chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia resistant or intolerant to imatinib and/or other second generation c-ABL therapy
- Author
-
Borthakur, G., primary, Dombret, H., additional, Schafhausen, P., additional, Brummendorf, T. H., additional, Boissel, N., additional, Jabbour, E., additional, Mariani, M., additional, Capolongo, L., additional, Carpinelli, P., additional, Davite, C., additional, Kantarjian, H., additional, and Cortes, J. E., additional
- Published
- 2015
- Full Text
- View/download PDF
4. Tailoring iron chelation by iron intake and serum ferritin: the prospective EPIC study of deferasirox in 1744 patients with transfusion-dependent anemias
- Author
-
Cappellini, M. D., Porter, J., El Beshlawy, A., C. K., Li, Seymour, J. F., Elalfy, M., Gattermann, N., Giraudier, S., Lee, J. W., Chan, L. L., Lin, K. H., Rose, C., Taher, A., Thein, S. L., Viprakasit, V., Habr, D., Domokos, G., Roubert, B., Kattamis, A., Agaoglu, L., Alimena, G., Alonso, D., Ame, S., Angelucci, E., Businco, A., Arrizabalaga, B., Athanasiou Metaxa, M., Augustson, B., Aydinok, Y., Baba, A., Baccarani, M., Beck, J., Beris, P., Beyne Rauzy, O., Birgens, H., Bordessoule, D., Borgna, Caterina, Bosly, A., Bouabdallah, K., Bowden, D., Bowen, D., Bron, D., Capra, M., Cartron, G., Cazzola, M., Chalkias, C., Chancharunee, S., Chapman, C., Charoenkwan, P., Chasapopoulou, E., Cheze, S., Chuansumrit, A., Cianciulli, P., Dauriac, C., Delforge, M., Dölken, G., Dombret, H., Duyster, J., Economopoulos, T., Ehninger, G., Enggaard, L., Fillet, G., Filosa, A., Forni, G., Galanello, R., Gastl, G., Goldfarb, A., Grigg, A., Gumruk, F., S. Y., Ha, Haase, D., Heinrich, B., Hertzberg, M., Ho, J., Hsu, H. C., Huang, S., Hunault Berger, M., Inusa, B., Jaulmes, D., Jensen, J., Kilinc, Y., Kim, K. H., Kinsey, S., Kjeldsen, L., Koren, A., Lai, M. E., Lai, Y., Lee, K. H., Lee, S. H., Legros, L., Li, C., Li, Q., Linkesch, W., Lübbert, M., Lutz, D., Mohamed Thalha, A. J., Mufti, G., Muus, P., Nobile, F., Papadopoulos, N., Perrotta, S., Petrini, M., Pfeilstöcker, M., Piga, A., Poole, J., Porter, J. B., Pungolino, E., Quarta, G., Ravoet, C., Jolimont Lobbes, H. H., Remacha, A. F., Roy, L., Saglio, G., Sanz, G., Schmugge, M., Schots, H., Secchi, G., Shah, F., Shah, H., Shen, Z., Slama, B., Sutcharitchan, P., Tamary, H., Tesch, H. J., Troncy, J., Villegas, A., Wainwright, L., Wassmann, B., Wettervald, M., Will, A., Wörmann, B., Wright, J., Yeh, S. P., Yoon, S. S., Zoumbos, N. S., and Zweegman, S.
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pediatrics ,Blood transfusion ,Iron Overload ,Adolescent ,Anemia ,Thalassemia ,medicine.medical_treatment ,Iron chelation therapy ,Transfusion medicine ,Transfusion-dependent anemias ,Hemosiderosis ,Iron Chelating Agents ,Gastroenterology ,Benzoates ,Young Adult ,IRON CHELATION ,Internal medicine ,medicine ,Humans ,Blood Transfusion ,Tissue Distribution ,SERUM FERRITIN ,Chelation therapy ,Prospective Studies ,Child ,Aged ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Deferasirox ,Hematology ,Middle Aged ,Triazoles ,medicine.disease ,DEFERASOROX ,Deferoxamine ,Child, Preschool ,Ferritins ,Female ,Original Article ,business ,TRANSFUSION-DEPENDENT ANEMIAS ,Iron, Dietary ,medicine.drug - Abstract
Background Following a clinical evaluation of deferasirox (Exjade) it was concluded that, in addition to baseline body iron burden, ongoing transfusional iron intake should be considered when selecting doses. The 1-year EPIC study, the largest ever investigation conducted for an iron chelator, is the first to evaluate whether fixed starting doses of deferasirox, based on transfusional iron intake, with dose titration guided by serum ferritin trends and safety markers, provides clinically acceptable chelation in patients (agedor=2 years) with transfusional hemosiderosis from various types of anemia.The recommended initial dose was 20 mg/kg/day for patients receiving 2-4 packed red blood cell units/month and 10 or 30 mg/kg/day was recommended for patients receiving less or more frequent transfusions, respectively. Dose adjustments were based on 3-month serum ferritin trends and continuous assessment of safety markers. The primary efficacy end-point was change in serum ferritin after 52 weeks compared with baseline.The 1744 patients enrolled had the following conditions; thalassemia (n=1115), myelodysplastic syndromes (n=341), aplastic anemia (n=116), sickle cell disease (n=80), rare anemias (n=43) and other transfused anemias (n=49). Overall, there was a significant reduction in serum ferritin from baseline (-264 ng/mL; P0.0001), reflecting dosage adjustments and ongoing iron intake. The most common (5%) adverse events were gastrointestinal disturbances (28%) and skin rash (10%). Conclusions Analysis of this large, prospectively collected data set confirms the response to chelation therapy across various anemias, supporting initial deferasirox doses based on transfusional iron intake, with subsequent dose titration guided by trends in serum ferritin and safety markers (clinicaltrials.gov identifier: NCT00171821).
- Published
- 2009
5. Dasatinib in high-risk core binding factor acute myeloid leukemia in first complete remission: a French Acute Myeloid Leukemia Intergroup trial
- Author
-
Boissel, N., primary, Renneville, A., additional, Leguay, T., additional, Lefebvre, P. C., additional, Recher, C., additional, Lecerf, T., additional, Delabesse, E., additional, Berthon, C., additional, Blanchet, O., additional, Prebet, T., additional, Pautas, C., additional, Chevallier, P., additional, Lepretre, S., additional, Girault, S., additional, Bonmati, C., additional, Guieze, R., additional, Himberlin, C., additional, Randriamalala, E., additional, Preudhomme, C., additional, Jourdan, E., additional, Dombret, H., additional, and Ifrah, N., additional
- Published
- 2015
- Full Text
- View/download PDF
6. Impact of additional genetic alterations on the outcome of patients with NPM1-mutated cytogenetically normal acute myeloid leukemia
- Author
-
Peterlin, P., primary, Renneville, A., additional, Ben Abdelali, R., additional, Nibourel, O., additional, Thomas, X., additional, Pautas, C., additional, de Botton, S., additional, Raffoux, E., additional, Cayuela, J.-M., additional, Boissel, N., additional, Terre, C., additional, Celli-Lebras, K., additional, Castaigne, S., additional, Preudhomme, C., additional, Gardin, C., additional, and Dombret, H., additional
- Published
- 2014
- Full Text
- View/download PDF
7. Vincristine, dexamethasone and epratuzumab for older relapsed/refractory CD22+ B-acute lymphoblastic leukemia patients: a phase II study
- Author
-
Chevallier, P., primary, Huguet, F., additional, Raffoux, E., additional, Etienne, A., additional, Leguay, T., additional, Isnard, F., additional, Robillard, N., additional, Guillaume, T., additional, Delaunay, J., additional, Charbonnier, A., additional, Pigneux, A., additional, Peterlin, P., additional, Bene, M. C., additional, Wegener, W. A., additional, Goldenberg, D. M., additional, and Dombret, H., additional
- Published
- 2014
- Full Text
- View/download PDF
8. Early onset hypercholesterolemia induced by the 2nd-generation tyrosine kinase inhibitor nilotinib in patients with chronic phase-chronic myeloid leukemia
- Author
-
Rea, D., primary, Mirault, T., additional, Cluzeau, T., additional, Gautier, J.-F., additional, Guilhot, F., additional, Dombret, H., additional, and Messas, E., additional
- Published
- 2014
- Full Text
- View/download PDF
9. Successful tyrosine kinase inhibitor therapy in a refractory B-cell precursor acute lymphoblastic leukemia with EBF1-PDGFRB fusion
- Author
-
Lengline, E., primary, Beldjord, K., additional, Dombret, H., additional, Soulier, J., additional, Boissel, N., additional, and Clappier, E., additional
- Published
- 2013
- Full Text
- View/download PDF
10. Intensive induction is effective in selected octogenarian acute myeloid leukemia patients: prognostic significance of karyotype and selected molecular markers used in the European LeukemiaNet classification
- Author
-
Wetzler, M., primary, Mrozek, K., additional, Kohlschmidt, J., additional, Dombret, H., additional, Dohner, H., additional, Pilorge, S., additional, Krug, U., additional, Carroll, A. J., additional, Larson, R. A., additional, Marcucci, G., additional, Hiddemann, W., additional, Buchner, T., additional, and Bloomfield, C. D., additional
- Published
- 2013
- Full Text
- View/download PDF
11. The prognosis of CALM-AF10-positive adult T-cell acute lymphoblastic leukemias depends on the stage of maturation arrest
- Author
-
Ben Abdelali, R., primary, Asnafi, V., additional, Petit, A., additional, Micol, J.-B., additional, Callens, C., additional, Villarese, P., additional, Delabesse, E., additional, Reman, O., additional, Lepretre, S., additional, Cahn, J.-Y., additional, Guillerm, G., additional, Berthon, C., additional, Gardin, C., additional, Corront, B., additional, Leguay, T., additional, Bene, M.-C., additional, Ifrah, N., additional, Leverger, G., additional, Dombret, H., additional, and Macintyre, E., additional
- Published
- 2013
- Full Text
- View/download PDF
12. Impact of central nervous system involvement in adult patients with Philadelphia-negative acute lymphoblastic leukemia: a GRAALL-2005 study.
- Author
-
Orvain C, Chantepie S, Thomas X, Escofrre-Barbe M, Huguet F, Desbrosses Y, Guillerm G, Uzunov M, Leguay T, Barbieux S, Vey N, Chevallier P, Malfuson JV, Lepretre S, Baumann M, Aykut M, Chaib A, Joris M, Zerazhi H, Stussi G, Chapiro J, Berthon C, Bonmati C, Jourdan E, Carp D, Marcais AR, Gallego-Hernanz MP, Vaida I, Bilger K, Villate A, Pasquier F, Chalandon Y, Maury S, Lheritier V, Ifrah N, Dombret H, Boissel N, and Hunault-Berger M
- Subjects
- Young Adult, Humans, Prospective Studies, Cyclophosphamide, Central Nervous System, Treatment Outcome, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Hematopoietic Stem Cell Transplantation
- Abstract
Whereas the prognosis of adult patients with Philadelphia-negative acute lymphoblastic leukemia (ALL) has greatly improved since the advent of pediatric-inspired regimens, the impact of initial central nervous system (CNS) involvement has not been formerly re-evaluated. We report here the outcome of patients with initial CNS involvement included in the pediatric-inspired prospective randomized GRAALL-2005 study. Between 2006 and 2014, 784 adult patients (aged 18-59 years) with newly diagnosed Philadelphia-negative ALL were included, of whom 55 (7%) had CNS involvement. In CNSpositive patients, overall survival was shorter (median 1.9 years vs. not reached, HR=1.8 [1.3-2.6], P<0.001). While there was no statistical difference in cumulative incidence of relapse between CNS+ and CNS- patients (HR=1.5 [0.9-2.5], P=0.11), non-relapse mortality was significantly higher in those with initial CNS disease (HR=2.1 [1.2-3.5], P=0.01). This increase in toxicity was mostly observed in patients randomized to the high-dose cyclophosphamide arm and in those who received allogeneic stem cell transplantation. Exploratory landmark analyses did not show any association between either cranial irradiation or allogeneic stem cell transplantation and outcome. Despite improved outcome in young adult ALL patients with pediatric-inspired protocols, CNS involvement is associated with a worse outcome mainly due to excess toxicity, without improved outcome with allogeneic SCT.
- Published
- 2023
- Full Text
- View/download PDF
13. Survival outcomes with oral azacitidine maintenance in patients with acute myeloid leukemia in remission by receipt of initial chemotherapy: subgroup analyses from the phase III QUAZAR AML-001 trial.
- Author
-
Wei AH, Roboz GJ, Dombret H, Dohner H, Schuh AC, Montesinos P, Selleslag D, Bondarenko SN, Prebet T, Lai Y, Skikne B, Beach CL, and Ravandi F
- Subjects
- Humans, Antimetabolites, Antineoplastic therapeutic use, Remission Induction, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality
- Published
- 2023
- Full Text
- View/download PDF
14. Allogeneic transplantation in acute myelogenous leukemia: a comprehensive single institution's experience.
- Author
-
Socie G, Galimard JE, Raffoux E, Itzykson R, Debureaux PE, Michonneau D, Lengliné E, Robin M, De Fontbrune FS, Sébert M, Xhaard A, Kim R, Couprie A, Dhedin N, Dragani M, Lemaire P, Larcher L, Clappier E, Boissel N, Soulier J, Dombret H, Fenaux P, De Latour RP, and Adès L
- Subjects
- Adult, Humans, Aged, Adolescent, Young Adult, Middle Aged, Transplantation, Homologous, Remission Induction, Proportional Hazards Models, Retrospective Studies, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute therapy
- Abstract
Debates on the role and timing of allogeneic hemtopoietic stem cell transplantation (HSCT) in acute myelogenous leukemia (AML) have persisted for decades. Time to transplant introduces an immortal time and current treatment algorithm mainly relies on the European LeukemiaNet disease risk classification. Previous studies are also limited to age groups, remission status and other ill-defined parameters. We studied all patients at diagnosis irrespective of age and comorbidities to estimate the cumulative incidence and potential benefit or disadvantage of HSCT in a single center. As a time-dependent covariate, HSCT improved overall survival in intermediate- and poor-risk patients (hazard ratio =0.51; P=0.004). In goodrisk patients only eight were transplanted in first complete remission. Overall, the 4-year cumulative incidence of HSCT was only 21.9% but was higher (52.1%) for patients in the first age quartile (16-57 years old) and 26.4% in older patients (57-70 years old) (P<0.001). It was negligible in patients older than 70 years reflecting our own transplant policy but also barriers to transplantation (comorbidities and remission status). However, HSCT patients need to survive, be considered eligible both by the referring and the HSCT physicians and have a suitable donor to get transplantation. We, thus, comprehensively analyzed the complete decision-making and outcome of all our AML patients from diagnosis to last followup to decipher how patient allocation and therapy inform the value of HSCT. The role of HSCT in AML is shifting with broad access to different donors including haploidentical ones. Thus, it may (or may not) lead to increased numbers of allogeneic HSCT in AML in adults.
- Published
- 2023
- Full Text
- View/download PDF
15. High tumor burden before blinatumomab has a negative impact on the outcome of adult patients with B-cell precursor acute lymphoblastic leukemia. A real-world study by the GRAALL.
- Author
-
Cabannes-Hamy A, Brissot E, Leguay T, Huguet F, Chevallier P, Hunault M, Escoffre-Barbe M, Cluzeau T, Balsat M, Nguyen S, Pasquier F, Alexis M, Lheritier V, Pastoret C, Delabesse E, Clappier E, Dombret H, and Boissel N
- Subjects
- Adult, Humans, Neoplasm, Residual drug therapy, Recurrence, Retrospective Studies, Tumor Burden, Antibodies, Bispecific therapeutic use, Antineoplastic Agents therapeutic use, Burkitt Lymphoma drug therapy, Lymphoma, B-Cell drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Blinatumomab is a bispecific T-cell engager approved for B-cell precursor acute lymphoblastic leukemia (B-ALL) with persistent minimal residual disease (MRD) or in relapse. The prognostic impact of tumor load has been suggested before other immunotherapies but remains poorly explored before blinatumomab. We retrospectively analyzed the outcome of 73 patients who received blinatumomab either in first complete remission (CR) with MRD (n=35) or at relapse (n=38). Among MRD patients, 91% had MRD >0.01% before blinatumomab, and 89% achieved complete MRD response after blinatumomab. High pre-blinatumomab MRD levels were associated with shorter relapse-free survival (P=0.049) and overall survival (OS) (P=0.011). At 3 years, OS was 33%, 58% and 86% for pre-blinatumomab MRD >1%, between MRD 0.1- 1% and <0.1% respectively. Among relapsed patients, 23 received blinatumomab with overt relapse and 15 were in complete response (CR) after bridging chemotherapy. At 3 years, overall CR rate was 68% and complete MRD response rate was 84%. Patients who directly received blinatumomab had shorter relapse-free survival (P=0.033) and OS (P=0.003) than patients bridged to blinatumomab. Three-year OS was 66% in the latter group compared to 16% in the former group. Our observations suggest that pre-blinatumomab tumor burden should help to design more tailored strategies including tumor load reduction in relapsed patients.
- Published
- 2022
- Full Text
- View/download PDF
16. Oral azacitidine preserves favorable level of fatigue and health-related quality of life for patients with acute myeloid leukemia in remission: results from the phase 3, placebo-controlled QUAZAR AML-001 trial.
- Author
-
Roboz GJ, Döhner H, Pocock C, Dombret H, Ravandi F, Jang JH, Selleslag D, Mayer J, Martens UM, Liesveld J, Bernal T, Wang MC, Yu P, Shi L, Guo S, La Torre I, Skikne B, Dong Q, Braverman J, Nehme SA, Beach CL, and Wei AH
- Subjects
- Azacitidine therapeutic use, Fatigue drug therapy, Fatigue etiology, Humans, Remission Induction, Treatment Outcome, Leukemia, Myeloid, Acute drug therapy, Quality of Life
- Published
- 2021
- Full Text
- View/download PDF
17. Minimal residual disease monitoring in acute myeloid leukemia with non-A/B/D-NPM1 mutations by digital polymerase chain reaction: feasibility and clinical use.
- Author
-
Lesieur A, Thomas X, Nibourel O, Boissel N, Fenwarth L, De Botton S, Fournier E, Celli-Lebras K, Raffoux E, Recher C, Lambert J, Berthon C, Pigneux A, Itzykson R, Turlure P, Pautas C, Vargaftig J, Preudhomme C, Dombret H, and Duployez N
- Subjects
- Feasibility Studies, Humans, Mutation, Neoplasm, Residual, Nucleophosmin, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics
- Abstract
Not available.
- Published
- 2021
- Full Text
- View/download PDF
18. Low level CpG island promoter methylation predicts a poor outcome in adult T-cell acute lymphoblastic leukemia.
- Author
-
Touzart A, Boissel N, Belhocine M, Smith C, Graux C, Latiri M, Lhermitte L, Mathieu EL, Huguet F, Lamant L, Ferrier P, Ifrah N, Macintyre E, Dombret H, Asnafi V, and Spicuglia S
- Subjects
- Adult, CpG Islands, DNA Methylation, Humans, Promoter Regions, Genetic, T-Lymphocytes, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Cancer cells undergo massive alterations in their DNA methylation patterns which result in aberrant gene expression and malignant phenotypes. Abnormal DNA methylation is a prognostic marker in several malignancies, but its potential prognostic significance in adult T-cell acute lymphoblastic leukemia (T-ALL) is poorly defined. Here, we performed methylated DNA immunoprecipitation to obtain a comprehensive genome-wide analysis of promoter methylation in adult T-ALL (n=24) compared to normal thymi (n=3). We identified a CpG hypermethylator phenotype that distinguishes two T-ALL subgroups and further validated it in an independent series of 17 T-lymphoblastic lymphoma. Next, we identified a methylation classifier based on nine promoters which accurately predict the methylation phenotype. This classifier was applied to an independent series of 168 primary adult T-ALL treated accordingly to the GRAALL03/05 trial using methylation-specific multiplex ligation-dependent probe amplification. Importantly hypomethylation correlated with specific oncogenic subtypes of T-ALL and identified patients associated with a poor clinical outcome. This methylation-specific multiplex ligation-dependent probe amplification based methylation profiling could be useful for therapeutic stratification of adult T-ALL in routine practice. The GRAALL-2003 and -2005 studies were registered at http://www.clinicaltrials.gov as #NCT00222027 and #NCT00327678, respectively., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
19. DNMT3A mutation is associated with increased age and adverse outcome in adult T-cell acute lymphoblastic leukemia.
- Author
-
Bond J, Touzart A, Leprêtre S, Graux C, Bargetzi M, Lhermitte L, Hypolite G, Leguay T, Hicheri Y, Guillerm G, Bilger K, Lhéritier V, Hunault M, Huguet F, Chalandon Y, Ifrah N, Macintyre E, Dombret H, Asnafi V, and Boissel N
- Subjects
- Adult, Alleles, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, DNA Methyltransferase 3A, Drug Resistance, Neoplasm genetics, Female, Genotype, Hematopoiesis genetics, High-Throughput Nucleotide Sequencing, Humans, Immunophenotyping, Male, Middle Aged, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Sequence Analysis, DNA, Treatment Outcome, Aging genetics, DNA (Cytosine-5-)-Methyltransferases genetics, Mutation, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma mortality
- Abstract
The prognostic implications of DNMT3A genotype in T-cell acute lymphoblastic leukemia are incompletely understood. We performed comprehensive genetic and clinico-biological analyses of T-cell acute lymphoblastic leukemia patients with DNMT3A mutations treated during the GRAALL-2003 and -2005 studies. Eighteen of 198 cases (9.1%) had DNMT3A alterations. Two patients also had DNMT3A mutations in non-leukemic cell DNA, providing the first potential evidence of age-related clonal hematopoiesis in T-cell acute lymphoblastic leukemia. DNMT3A mutation was associated with older age (median 43.9 years vs 29.4 years, P <0.001), immature T-cell receptor genotype (53.3% vs 24.4%, P =0.016) and lower remission rates (72.2% mutated vs 94.4% non-mutated, P =0.006). DNMT3A alterations were significantly associated with worse clinical outcome, with higher cumulative incidence of relapse (HR 2.33, 95% CI: 1.05-5.16, P =0.037) and markedly poorer event-free survival (HR 3.22, 95% CI: 1.81-5.72, P <0.001) and overall survival (HR 2.91, 95% CI: 1.56-5.43, P =0.001). Adjusting for age as a covariate, or restricting the analysis to patients over 40 years, who account for almost 90% of DNMT3A -mutated cases, did not modify these observations. In multivariate analysis using the risk factors that were used to stratify treatment during the GRAALL studies, DNMT3A mutation was significantly associated with shorter event-free survival (HR 2.33, 95% CI: 1.06 - 4.04, P =0.02). Altogether, these results identify DNMT3A genotype as a predictor of aggressive T-cell acute lymphoblastic leukemia biology. The GRAALL-2003 and -2005 studies were registered at http://www.ClinicalTrials.gov as #NCT00222027 and #NCT00327678 , respectively., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
20. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: final efficacy and safety updates from the open-label, phase III ALFA-0701 trial.
- Author
-
Lambert J, Pautas C, Terré C, Raffoux E, Turlure P, Caillot D, Legrand O, Thomas X, Gardin C, Gogat-Marchant K, Rubin SD, Benner RJ, Bousset P, Preudhomme C, Chevret S, Dombret H, and Castaigne S
- Subjects
- Adult, Aged, Cytarabine administration & dosage, Daunorubicin administration & dosage, Disease-Free Survival, Female, Gemtuzumab administration & dosage, Humans, Male, Middle Aged, Survival Rate, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology
- Abstract
The randomized, phase III ALFA-0701 trial showed that a reduced and fractionated dose of gemtuzumab ozogamicin added to standard front-line chemotherapy significantly improves event-free survival (EFS) in adults with de novo acute myeloid leukemia (AML). Here we report an independent review of EFS, final overall survival (OS), and additional safety results from ALFA-0701. Patients (n=271) aged 50-70 years with de novo AML were randomized to receive conventional front-line induction chemotherapy (3+7daunorubicin+cytarabine) with/without gemtuzumab ozogamicin 3 mg/m
2 on days 1, 4, and 7 during induction. Patients in remission following induction therapy received 2 courses of consolidation therapy (daunorubicin+cytarabine) with/without gemtuzumab ozogamicin (3 mg/m2 /day on day 1) according to their initial randomization. The primary end point was investigator-assessed EFS. Secondary end points included OS and safety. A blinded independent review confirmed the investigator-assessed EFS results [August 1, 2011; hazard ratio (HR) 0.66; 95% Confidence Interval (CI): 0.49-0.89; 2-sided P =0.006], corresponding to a 34% reduction in risk of events in the gemtuzumab ozogamicin versus control arm. Final OS at April 30, 2013 favored gemtuzumab ozogamicin but was not significant. No differences in early death rate were observed between arms. The main toxicity associated with gemtuzumab ozogamicin was prolonged thrombocytopenia. Veno-occlusive disease (including after transplant) was observed in 6 patients in the gemtuzumab ozogamicin arm and 2 in the control arm. In conclusion, gemtuzumab ozogamicin added to standard intensive chemotherapy has a favorable benefit/risk ratio. These results expand front-line treatment options for adult patients with previously untreated AML. (Trial registered at clinicaltrials.gov; identifier: 00927498 )., (Copyright© 2019 Ferrata Storti Foundation.)- Published
- 2019
- Full Text
- View/download PDF
21. Arsenic trioxide is required in the treatment of newly diagnosed acute promyelocytic leukemia. Analysis of a randomized trial (APL 2006) by the French Belgian Swiss APL group.
- Author
-
Adès L, Thomas X, Bresler AG, Raffoux E, Spertini O, Vey N, Marchand T, Récher C, Pigneux A, Girault S, Deconinck E, Gardin C, Tournilhac O, Lambert JF, Chevallier P, de Botton S, Lejeune J, Dombret H, Chevret S, and Fenaux P
- Subjects
- Adult, Anthracyclines administration & dosage, Arsenic Trioxide administration & dosage, Belgium, Disease-Free Survival, Female, France, Humans, Leukemia, Promyelocytic, Acute diagnosis, Male, Middle Aged, Switzerland, Treatment Outcome, Tretinoin administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Promyelocytic, Acute drug therapy
- Abstract
In standard-risk acute promyelocytic leukemia, recent results have shown that all- trans retinoic acid plus arsenic trioxide combinations are at least as effective as classical all- trans retinoic acid plus anthracycline-based chemotherapy while being less myelosuppressive. However, the role of frontline arsenic trioxide is less clear in higher-risk acute promyelocytic leukemia, and access to arsenic remains limited for front-line treatment of standard-risk acute promyelocytic leukemia in many countries. In this randomized trial, we compared arsenic, all- trans retinoic acid and the "classical" cytarabine for consolidation treatment (after all- trans retinoic acid and chemotherapy induction treatment) in standard-risk acute promyelocytic leukemia, and evaluated the addition of arsenic during consolidation in higher-risk disease. Patients with newly diagnosed acute promyelocytic leukemia with a white blood cell count <10x10
9 /L, after an induction treatment consisting of all- trans retinoic acid plus idarubicin and cytarabine, received consolidation chemotherapy with idarubicin and cytarabine, arsenic or all- trans retinoic acid. Patients with a white blood cell count >10x109 /L received consolidation chemotherapy with or without arsenic. Overall, 795 patients with acute promyelocytic leukemia were enrolled in this trial. Among those with standard-risk acute promyelocytic leukemia (n=581), the 5-year event-free survival rates from randomization were 88.7%, 95.7% and 85.4% in the cytarabine, arsenic and all- trans retinoic acid consolidation groups, respectively ( P =0.0067), and the 5-year cumulative incidences of relapse were was 5.5%, 0% and 8.2%. ( P =0.001). Among those with higher-risk acute promyelocytic leukemia (n=214), the 5-year event-free survival rates were 85.5% and 92.1% ( P =0.38) in the chemotherapy and chemotherapy plus arsenic groups, respectively, and the corresponding 5-year cumulative incidences of relapse were 4.6% and 3.5% ( P =0.99). Given the prolonged myelosuppression that occurred in the chemotherapy plus arsenic arm, a protocol amendment excluded cytarabine during consolidation cycles in the chemotherapy plus arsenic group, resulting in no increase in relapse. Our results therefore advocate systematic introduction of arsenic in the first-line treatment of acute promyelocytic leukemia, but probably not concomitantly with intensive chemotherapy, a situation in which we found myelosuppression to be significant. ( ClinicalTrials.gov Identifier: NCT00378365 )., (Copyright© 2018 Ferrata Storti Foundation.)- Published
- 2018
- Full Text
- View/download PDF
22. Clinical relevance of IDH1/2 mutant allele burden during follow-up in acute myeloid leukemia. A study by the French ALFA group.
- Author
-
Ferret Y, Boissel N, Helevaut N, Madic J, Nibourel O, Marceau-Renaut A, Bucci M, Geffroy S, Celli-Lebras K, Castaigne S, Thomas X, Terré C, Dombret H, Preudhomme C, and Renneville A
- Subjects
- Adolescent, Adult, Aged, Combined Modality Therapy, Female, Follow-Up Studies, Humans, Incidence, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Neoplasm, Residual epidemiology, Neoplasm, Residual genetics, Nucleophosmin, Prognosis, Survival Rate, Young Adult, Biomarkers, Tumor genetics, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute therapy, Mutation, Neoplasm, Residual diagnosis
- Abstract
Assessment of minimal residual disease has emerged as a powerful prognostic factor in acute myeloid leukemia. In this study, we investigated the potential of IDH1/2 mutations as targets for minimal residual disease assessment in acute myeloid leukemia, since these mutations collectively occur in 15-20% of cases of acute myeloid leukemia and now represent druggable targets. We employed droplet digital polymerase chain reaction assays to quantify IDH1R132 , IDH2R140 , and IDH2R172 mutations on genomic DNA in 322 samples from 103 adult patients with primary IDH1/2 mutant acute myeloid leukemia and enrolled on Acute Leukemia French Association (ALFA) - 0701 or -0702 clinical trials. The median IDH1/2 mutant allele fraction in bone marrow samples was 42.3% (range, 8.2 - 49.9%) at diagnosis of acute myeloid leukemia, and below the detection limit of 0.2% (range, <0.2 - 39.3%) in complete remission after induction therapy. In univariate analysis, the presence of a normal karyotype, a NPM1 mutation, and an IDH1/2 mutant allele fraction <0.2% in bone marrow after induction therapy were statistically significant predictors of longer disease-free survival. In multivariate analysis, these three variables remained significantly predictive of disease-free survival. In 7/103 (7%) patients, IDH1/2 mutations persisted at high levels in complete remission, consistent with the presence of an IDH1/2 mutation in pre-leukemic hematopoietic stem cells. Five out of these seven patients subsequently relapsed or progressed toward myelodysplastic syndrome, suggesting that patients carrying the IDH1/2 mutation in a pre-leukemic clone may be at high risk of hematologic evolution., (Copyright © 2018 Ferrata Storti Foundation.)
- Published
- 2018
- Full Text
- View/download PDF
23. Hyper-CVAD + epratuzumab as a salvage regimen for younger patients with relapsed/refractory CD22-positive precursor B-cell acute lymphocytic leukemia.
- Author
-
Chevallier P, Chantepie S, Huguet F, Raffoux E, Thomas X, Leguay T, Marchand T, Isnard F, Charbonnier A, Maury S, Gallego-Hernanz MP, Robillard N, Guillaume T, Peterlin P, Garnier A, Rialland F, Le Houerou C, Goldenberg DM, Wegener WA, Béné MC, and Dombret H
- Subjects
- Adult, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Biomarkers, Cyclophosphamide adverse effects, Cyclophosphamide therapeutic use, Dexamethasone adverse effects, Dexamethasone therapeutic use, Doxorubicin adverse effects, Doxorubicin therapeutic use, Drug Resistance, Neoplasm, Female, Humans, Male, Middle Aged, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Recurrence, Retreatment, Treatment Outcome, Vincristine adverse effects, Vincristine therapeutic use, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Sialic Acid Binding Ig-like Lectin 2 metabolism
- Published
- 2017
- Full Text
- View/download PDF
24. Lenalidomide combined with intensive chemotherapy in acute myeloid leukemia and higher-risk myelodysplastic syndrome with 5q deletion. Results of a phase II study by the Groupe Francophone Des Myélodysplasies .
- Author
-
Ades L, Prebet T, Stamatoullas A, Recher C, Guieze R, Raffoux E, Bouabdallah K, Hunault M, Wattel E, Stalnikiewicz L, Toma A, Dombret H, Vey N, Sebert M, Gardin C, Chaffaut C, Chevret S, and Fenaux P
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Female, Humans, Lenalidomide, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes mortality, Prognosis, Thalidomide administration & dosage, Thalidomide analogs & derivatives, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chromosome Deletion, Chromosomes, Human, Pair 5, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics
- Abstract
Patients with acute myeloblastic leukemia or higher risk myelodysplastic syndromes with 5q deletion (generally within a complex karyotype) respond poorly to intensive chemotherapy and have very poor survival. In this population, we evaluated escalating doses of lenalidomide combined with intensive chemotherapy in a phase II study. Treatment consisted of daunorubicin (45 mg/m
2 /day, days 1-3 in cohort 1, escalated to 60 mg/m2 /day, days 1-3 in cohorts 2 and 3) combined with cytosine arabinoside (200 mg/m2 /day, days 1-7) and lenalidomide (10 mg/day, days 1-21 in cohorts 1 and 2, escalated to 25 mg/day, days 1-21 in cohort 3). Eighty-two patients with 5q deletion were enrolled, including 62 with acute myeloblastic leukemia, 62/79 (78%) of whom had a complex karyotype (median 7 cytogenetic abnormalities, all but 2 of them monosomal) and three had unknown karyotypes. Thirty-eight patients (46%) achieved complete remission and the overall response rate was 58.5%. Among the 62 patients with a complex karyotype, 27 achieved complete remission (44%) and 21 had cytogenetic responses. A lower response rate was observed in patients with acute myeloblastic leukemia but other pretreatment factors, including cytogenetic complexity and treatment cohort, did not significantly influence response. Fifteen patients underwent allogeneic stem cell transplantation, including 11 patients in first remission. The 1-year cumulative incidence of relapse was 64.6% and the median overall survival was 8.2 months. By comparison with conventional intensive chemotherapy, the treatment protocol we used appeared to produce higher hematologic and cytogenetic complete remission rates in patients with very poor cytogenetics, but response duration was short in this very poor risk population, highlighting the need for better post-induction strategies. Clinical trial registry number: NCT00885508 ., (Copyright© Ferrata Storti Foundation.)- Published
- 2017
- Full Text
- View/download PDF
25. Prospective long-term minimal residual disease monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: results of the French CBF-2006 trial.
- Author
-
Willekens C, Blanchet O, Renneville A, Cornillet-Lefebvre P, Pautas C, Guieze R, Ifrah N, Dombret H, Jourdan E, Preudhomme C, and Boissel N
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Adolescent, Adult, Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Blood Cells metabolism, Blood Cells pathology, Bone Marrow metabolism, Bone Marrow pathology, Chromosomes, Human, Pair 22, Chromosomes, Human, Pair 8, Consolidation Chemotherapy methods, Core Binding Factor Alpha 2 Subunit metabolism, Cytoskeletal Proteins metabolism, Female, France, Gene Expression, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Monitoring, Physiologic, Neoplasm, Residual, Prospective Studies, Proto-Oncogene Proteins metabolism, RUNX1 Translocation Partner 1 Protein, Real-Time Polymerase Chain Reaction, Recurrence, Remission Induction, Survival Analysis, Transcription Factors metabolism, Translocation, Genetic, Adaptor Proteins, Signal Transducing genetics, Biomarkers, Tumor genetics, Core Binding Factor Alpha 2 Subunit genetics, Cytoskeletal Proteins genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Proto-Oncogene Proteins genetics, Transcription Factors genetics
- Abstract
In t(8;21)(q22;q22) acute myeloid leukemia, the prognostic value of early minimal residual disease assessed with real-time quantitative polymerase chain reaction is the most important prognostic factor, but how long-term minimal residual disease monitoring may contribute to drive individual patient decisions remains poorly investigated. In the multicenter CBF-2006 study, a prospective monitoring of peripheral blood and bone marrow samples was performed every 3 months and every year, respectively, for 2 years following intensive chemotherapy in 94 patients in first complete remission. A complete molecular remission was defined as a (RUNX1-RUNX1T1/ABL1)×100 ≤ 0.001%. After the completion of consolidation therapy, a bone marrow complete molecular remission was observed in 30% of the patients, but was not predictive of subsequent relapse. Indeed, 8 patients (9%) presented a positive bone marrow minimal residual disease for up to 2 years of follow-up while still remaining in complete remission. Conversely, a peripheral blood complete molecular remission was statistically associated with a lower risk of relapse whatever the time-point considered after the completion of consolidation therapy. During the 2-year follow-up, the persistence of peripheral blood complete molecular remission was associated with a lower risk of relapse (4-year cumulative incidence, 8.2%), while molecular relapse confirmed on a subsequent peripheral blood sample predicted hematological relapse (4-year cumulative incidence, 86.9%) within a median time interval of 3.9 months. In t(8;21)(q22;q22) acute myeloid leukemia, minimal residual disease monitoring on peripheral blood every 3 months allows for the prediction of hematological relapse, and to identify patients who could potentially benefit from intervention therapy. (ClinicalTrials.gov ID #NCT00428558)., (Copyright© Ferrata Storti Foundation.)
- Published
- 2016
- Full Text
- View/download PDF
26. Impact of additional genetic alterations on the outcome of patients with NPM1-mutated cytogenetically normal acute myeloid leukemia.
- Author
-
Peterlin P, Renneville A, Ben Abdelali R, Nibourel O, Thomas X, Pautas C, de Botton S, Raffoux E, Cayuela JM, Boissel N, Terré C, Celli-Lebras K, Castaigne S, Preudhomme C, Gardin C, and Dombret H
- Subjects
- Biomarkers, Tumor genetics, Humans, Leukemia, Myeloid, Acute therapy, Nucleophosmin, Patient Outcome Assessment, Prognosis, Recurrence, Treatment Outcome, Karyotype, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Mutation, Nuclear Proteins genetics
- Published
- 2015
- Full Text
- View/download PDF
27. Vincristine, dexamethasone and epratuzumab for older relapsed/refractory CD22+ B-acute lymphoblastic leukemia patients: a phase II study.
- Author
-
Chevallier P, Huguet F, Raffoux E, Etienne A, Leguay T, Isnard F, Robillard N, Guillaume T, Delaunay J, Charbonnier A, Pigneux A, Peterlin P, Bené MC, Wegener WA, Goldenberg DM, and Dombret H
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Dexamethasone administration & dosage, Humans, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Recurrence, Sialic Acid Binding Ig-like Lectin 2 metabolism, Treatment Outcome, Vincristine administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Published
- 2015
- Full Text
- View/download PDF
28. Intensive induction is effective in selected octogenarian acute myeloid leukemia patients: prognostic significance of karyotype and selected molecular markers used in the European LeukemiaNet classification.
- Author
-
Wetzler M, Mrózek K, Kohlschmidt J, Dombret H, Döhner H, Pilorge S, Krug U, Carroll AJ, Larson RA, Marcucci G, Hiddemann W, Büchner T, and Bloomfield CD
- Subjects
- Abnormal Karyotype, Aged, Disease-Free Survival, Female, Humans, Nucleophosmin, Survival Rate, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Leukemia, Myeloid, Acute classification, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Mutation, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
We investigated whether octogenarian patients with acute myeloid leukemia enrolled onto Cooperative Group clinical trials and treated with intensive induction therapy could be cured, and whether karyotype and selected molecular markers had any prognostic significance in these patients. Among 138 patients with cytogenetic information, normal karyotype was the most common (47.1%) followed by complex karyotype (14.5%) and sole +8 (9.4%). Among these patients, the relapse-free survival rate at 1 year was 37% and 13% at 3 years, and the respective overall survival rates were 24% and 8%. Whereas the 90 patients who survived beyond 30 days had the same relapse-free survival rates, their 1-year and 3-year overall survival rates were 36% and 11%, respectively. Of the 66 patients surviving beyond 30 days who could be classified into European LeukemiaNet genetic groups, those in the intermediate-I group had better overall survival than patients in the adverse group (P=0.01). Among patients with cytogenetically normal acute myeloid leukemia who were tested for the European LeukemiaNet-associated molecular alterations, FLT3-internal tandem duplication and NPM1 mutations, it was found that FLT3-internal tandem duplication (detected in 29% of patients) did not associate with overall survival (P=0.31), whereas NPM1 mutations (30%) were associated with a significantly longer overall survival (P=0.002). We conclude that intensive induction is effective and indicated in selected octogenarians with acute myeloid leukemia, that their overall survival varies among the European LeukemiaNet genetic groups and that NPM1 mutations may be of prognostic significance among octogenarian patients with cytogenetically normal acute myeloid leukemia.
- Published
- 2014
- Full Text
- View/download PDF
29. Impact of post-remission therapy in patients aged 65-70 years with de novo acute myeloid leukemia: a comparison of two concomitant randomized ALFA trials with overlapping age inclusion criteria.
- Author
-
Itzykson R, Gardin C, Pautas C, Thomas X, Turlure P, Raffoux E, Terré C, Fenaux P, Castaigne S, Dombret H, and Boissel N
- Subjects
- Aged, Antineoplastic Agents therapeutic use, Female, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute mortality, Male, Prognosis, Remission Induction, Survival Analysis, Treatment Outcome, Leukemia, Myeloid, Acute therapy
- Abstract
Background: There is no standard post-remission therapy in older patients with acute myeloid leukemia., Design and Methods: From 1999 to 2006, the Acute Leukemia French Association group ran two concurrent randomized trials with overlapping inclusion criteria for patients aged 65 to 70 with acute myeloid leukemia, with different post-remission strategies: two intensive courses in the 9801 trial, one intensive course or six outpatient courses in the 9803 trial. We analyzed the outcome of these patients per protocol and per post-remission therapy., Results: Two hundred and eleven patients aged 65 to 70 years with de novo acute myeloid leukemia were enrolled in trial 9801 (n=76) or 9803 (n=135). The patients in the two trials had comparable white blood cell counts (P=0.3), cytogenetics (P=0.49), and complete remission rates (70% and 57%, respectively; P=0.17). Overall survival was identical in both trials (32% and 34% at 2 years, respectively; P=0.71). Overall survival after complete remission was identical in the 103 of 130 patients who received the planned post-remission courses (n=44 with two intensive courses, n=28 with one intensive course, n=31 with six outpatient courses; 41%, 55%, and 58% at 2 years, respectively; P=0.34). Even in patients with favorable or normal karyotype (n=97), overall survival from complete remission was not improved by more intensive post-remission therapy., Conclusions: In patients aged 65 to 70 years with de novo acute myeloid leukemia in complete remission after standard intensive induction chemotherapy, there is no apparent benefit from intensive post-remission therapy. (ClinicalTrials.gov Identifiers: NCT00931138 and NCT00363025).
- Published
- 2011
- Full Text
- View/download PDF
30. Liposomal cytarabine is effective and tolerable in the treatment of central nervous system relapse of acute lymphoblastic leukemia and very aggressive lymphoma.
- Author
-
Gökbuget N, Hartog CM, Bassan R, Derigs HG, Dombret H, Greil R, Hernández-Rivas JM, Huguet F, Intermesoli T, Jourdan E, Junghanss C, Leimer L, Moreno MJ, Reichle A, Ribera J, Schmid M, Serve H, Stelljes M, Stuhlmann R, and Hoelzer D
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Burkitt Lymphoma complications, Central Nervous System Neoplasms etiology, Female, Humans, International Agencies, Liposomes, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Recurrence, Local diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Prospective Studies, Risk Factors, Survival Rate, Treatment Outcome, Young Adult, Antimetabolites, Antineoplastic therapeutic use, Burkitt Lymphoma drug therapy, Central Nervous System Neoplasms drug therapy, Cytarabine therapeutic use, Neoplasm Recurrence, Local drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Background: Treatment of central nervous system relapse in adult acute lymphoblastic leukemia is a challenge and outcome is poor. Liposomal cytarabine has a prolonged half-life and, given intrathecally, has produced high response rates in patients with central nervous system relapse of non-Hodgkin's lymphoma. The aim of this study was to evaluate the efficacy and tolerability of liposomal cytarabine in central nervous system relapse of acute lymphoblastic leukemia or Burkitt's lymphoma/leukemia., Design and Methods: Liposomal cytarabine (50 mg) was given intrathecally together with systemic or intrathecal dexamethasone once every 2 weeks in a phase II European trial. The primary end-point, cytological response in the cerebrospinal fluid after one or two cycles, was evaluated at the time of next treatment., Results: Nineteen heavily pretreated patients (median age, 53 years; range 24-76 years) were evaluable: 14 with acute lymphoblastic leukemia and 5 with Burkitt's lymphoma/leukemia). Complete cytological remission as best response after two cycles of liposomal cytarabine was confirmed in 74% of the patients: 86% of those with acute lymphoblastic leukemia and 40% of those with Burkitt's lymphoma/leukemia). Nine of the 14 patients who achieved complete remission relapsed after a median of 7 months. The median overall survival was 11 months. Adverse events were observed in 89% of the patients (57% of cycles). Grade III-IV events with potential correlation to liposomal cytarabine occurred in 32% of the patients. The most frequent adverse event was headache. One patient developed severe neurological complications with loss of vision and a conus syndrome., Conclusions: Overall, liposomal cytarabine showed excellent antileukemic activity. Toxicity was acceptable but appeared to increase with the number of cycles. Future evaluation in prophylaxis is of interest.
- Published
- 2011
- Full Text
- View/download PDF
31. A randomized study of pegylated liposomal doxorubicin versus continuous-infusion doxorubicin in elderly patients with acute lymphoblastic leukemia: the GRAALL-SA1 study.
- Author
-
Hunault-Berger M, Leguay T, Thomas X, Legrand O, Huguet F, Bonmati C, Escoffre-Barbe M, Legros L, Turlure P, Chevallier P, Larosa F, Garban F, Reman O, Rousselot P, Dhédin N, Delannoy A, Lafage-Pochitaloff M, Béné MC, Ifrah N, and Dombret H
- Subjects
- Adult, Aged, Aged, 80 and over, Cyclophosphamide administration & dosage, Dexamethasone administration & dosage, Doxorubicin administration & dosage, Doxorubicin analogs & derivatives, Female, Humans, Liposomes, Male, Middle Aged, Neoplasm Recurrence, Local diagnosis, Polyethylene Glycols administration & dosage, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Survival Rate, Treatment Outcome, Vincristine administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasm Recurrence, Local drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Background: The prognosis of acute lymphoblastic leukemia in the elderly is poor. The GRAALL-SA1 phase II, randomized trial compared the efficacy and toxicity of pegylated liposomal doxorubicin versus continuous-infusion doxorubicin in patients 55 years or older with Philadelphia chromosome-negative acute lymphoblastic leukemia., Design and Methods: Sixty patients received either continuous-infusion doxorubicin (12 mg/m(2)/day) and continuous-infusion vincristine (0.4 mg/day) on days 1-4 or pegylated liposomal doxorubicin (40 mg/m(2)) and standard vincristine (2 mg) on day 1, accompanied by dexamethasone, followed at day 28 by a second cycle, reinforced by cyclophosphamide. End-points were safety, outcome and prognostic factors., Results: Myelosuppression was reduced in the pegylated liposomal doxorubicin arm with shorter severe neutropenia (P=0.05), shorter severe thrombocytopenia (P=0.03), and fewer red blood cell transfusions (P=0.04). Grade 3/4 infections and Gram-negative bacteremia were reduced in the pegylated liposomal doxorubicin arm (P=0.04 and P=0.02, respectively). There was a trend towards fewer cardiac events among the patients who received pegylated liposomal doxorubicin (1/29 versus 6/31). The complete remission rate was 82% and, with a median follow-up of 4 years, median event-free survival and overall survival were 9 and 10 months, respectively. Despite the better tolerance of pegylated liposomal doxorubicin, no differences in survival were observed between the two arms, due to trends towards more induction refractoriness (17 versus 3%, P=0.10) and a higher cumulative incidence of relapse (52% versus 32% at 2 years, P=0.20) in the pegylated liposomal doxorubicin arm., Conclusions: With the drug schedules used in this study, pegylated liposomal doxorubicin did not improve the outcome of elderly patients with acute lymphoblastic leukemia despite reduced toxicities.
- Published
- 2011
- Full Text
- View/download PDF
32. The European LeukemiaNet: achievements and perspectives.
- Author
-
Hehlmann R, Grimwade D, Simonsson B, Apperley J, Baccarani M, Barbui T, Barosi G, Bassan R, Béné MC, Berger U, Büchner T, Burnett A, Cross NC, de Witte TJ, Döhner H, Dombret H, Einsele H, Engelich G, Foà R, Fonatsch C, Gökbuget N, Gluckman E, Gratwohl A, Guilhot F, Haferlach C, Haferlach T, Hallek M, Hasford J, Hochhaus A, Hoelzer D, Kiladjian JJ, Labar B, Ljungman P, Mansmann U, Niederwieser D, Ossenkoppele G, Ribera JM, Rieder H, Serve H, Schrotz-King P, Sanz MA, and Saussele S
- Subjects
- Europe, Humans, International Cooperation, Societies, Medical organization & administration, Biomedical Research organization & administration, Leukemia, Medical Oncology organization & administration
- Abstract
The only way to cure leukemia is by cooperative research. To optimize research, the European LeukemiaNet integrates 105 national leukemia trial groups and networks, 105 interdisciplinary partner groups and about 1,000 leukemia specialists from 175 institutions. They care for tens of thousands of leukemia patients in 33 countries across Europe. Their ultimate goal is to cure leukemia. Since its inception in 2002, the European LeukemiaNet has steadily expanded and has unified leukemia research across Europe. The European LeukemiaNet grew from two major roots: 1) the German Competence Network on Acute and Chronic Leukemias; and 2) the collaboration of European Investigators on Chronic Myeloid Leukemia. The European LeukemiaNet has improved leukemia research and management across Europe. Its concept has led to funding by the European Commission as a network of excellence. Other sources (European Science Foundation; European LeukemiaNet-Foundation) will take over when the support of the European Commission ends.
- Published
- 2011
- Full Text
- View/download PDF
33. Adverse prognostic significance of CD20 expression in adults with Philadelphia chromosome-negative B-cell precursor acute lymphoblastic leukemia.
- Author
-
Maury S, Huguet F, Leguay T, Lacombe F, Maynadié M, Girard S, de Labarthe A, Kuhlein E, Raffoux E, Thomas X, Chevallier P, Buzyn A, Delannoy A, Chalandon Y, Vernant JP, Rousselot P, Macintyre E, Ifrah N, Dombret H, and Béné MC
- Subjects
- Acute Disease, Adolescent, Adult, Clinical Trials as Topic, Humans, Leukocyte Count, Middle Aged, Philadelphia Chromosome, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Recurrence, Young Adult, Antigens, CD20 genetics, Gene Expression, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
The prognostic significance of CD20 expression in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) has been mostly studied in children and yielded conflicting results. In 143 adults with Philadelphia chromosome-negative BCP-ALL treated in the multicentric GRAALL 2003 trial, CD20 positivity over 20% was observed in 32% of patients. While not influencing complete remission achievement, CD20 expression was associated with a higher cumulative incidence of relapse (CIR) at 42 months (P=0.04), independently of the ALL high-risk subset (P=0.025). Notably, the negative impact of CD20 expression on CIR was only observed in patients with a white blood cell count (WBC) over 30x10(9)/L (P=0.006), while not in those with a lower WBC. In the former subgroup, this impact translated into lower event-free survival (15% vs. 59% at 42 months, P=0.003). CD20 expression thus appears to be associated with a worse outcome, which reinforces the interest of evaluating rituximab combined to chemotherapy in CD20-positive adult BCP-ALL. ClinicalTrials.gov ID, NCT00222027.
- Published
- 2010
- Full Text
- View/download PDF
34. Risk factors and decision criteria for intensive chemotherapy in older patients with acute myeloid leukemia.
- Author
-
Malfuson JV, Etienne A, Turlure P, de Revel T, Thomas X, Contentin N, Terré C, Rigaudeau S, Bordessoule D, Vey N, Gardin C, and Dombret H
- Subjects
- Age Factors, Aged, Aged, 80 and over, Clinical Trials as Topic, Cytogenetic Analysis, Daunorubicin therapeutic use, Decision Making, Female, Humans, Idarubicin therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukocyte Count, Male, Patient Selection, Risk Factors, Survival Analysis, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute diagnosis, Practice Guidelines as Topic
- Abstract
Background: There is a need for standardization of treatment decisions in older patients with acute myeloid leukemia. The aim of the present study was to analyze the decisional value of poor risk factors in 416 elderly patients treated in the ALFA-9803 trial in order to derive a decisional index., Design and Methods: Standard multivariate analysis was used to identify risk factors for overall survival. Risk factors were then considered as good decision tools if associated with a frequency >10% and a false positive rate <10% in predicting overall survival as poor as observed after low-dose cytarabine therapy (25% survival or less at 12 months)., Results: Among six independent risk factors (age, performance status, white blood cell count, hematopoietic cell transplantation comorbidity index, infection at baseline, and cytogenetics), cytogenetics was the only potent, independent decision tool. High hematopoietic cell transplantation comorbidity index scores or infections were found too rarely to guide further decisions. The three other factors (age, performance status, and white cell count) needed to be combined to provide a good specificity. The proposed decisional index, therefore, included high-risk cytogenetics and/or the presence of at least two of the following criteria: age > or =75 years, performance status > or =2, and white cell count > or =50 x 10(9)/L. This simple two-class decisional index, which was validated in an independent patient set, enabled us to discriminate 100 patients (24%) who had an estimated overall survival of only 19% at 12 months, with a good 9% false positive rate., Conclusions: We propose waiting for cytogenetic information before making treatment decisions in elderly patients with acute myeloid leukemia. Those patients with unfavorable cytogenetics, as well as patients with at least two of the following features, age > or =75 years, performance status > or =2, and white cell count > or =50 x 10(9)/L, should not be considered for standard intensive chemotherapy (ClinicalTrials.gov identifier: NCT00363025).
- Published
- 2008
- Full Text
- View/download PDF
35. Long-term follow-up of autologous stem cell transplantation after intensive chemotherapy in patients with myelodysplastic syndrome or secondary acute myeloid leukemia.
- Author
-
Ducastelle S, Adès L, Gardin C, Dombret H, Prébet T, Deconinck E, Rio B, Thomas X, Debotton S, Guerci A, Gratecos N, Stamatoullas A, Fegueux N, Dreyfus F, Fenaux P, and Wattel E
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Agents therapeutic use, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes mortality, Survival Rate, Time, Transplantation, Autologous, Transplantation, Homologous, Leukemia, Myeloid, Acute surgery, Myelodysplastic Syndromes surgery, Stem Cell Transplantation
- Abstract
We report on the outcomes of 53 patients with myelodysplastic syndromes (MDS) or acute myeloid leukemia secondary to MDS, autografted in first complete remission. Five (9.4%) died from the procedure whereas hematological reconstitution occurred in all the remaining patients. Forty patients (75%) relapsed, with 87.5% of the relapses occurring within 2 years of the autologous transplant. With a median follow-up of 6.2 years, the median actuarial disease-free survival and overall survival were 8 and 17 months after autograft, respectively. Karyotype was the only prognostic factor for disease-free and overall survival. The eight survivors (15%), including two patients with unfavorable or intermediate karyotype, remained in first complete remission 50+ to 119+ months after transplantation and are probably cured.
- Published
- 2006
36. Valproic acid and all-trans retinoic acid for the treatment of elderly patients with acute myeloid leukemia.
- Author
-
Raffoux E, Chaibi P, Dombret H, and Degos L
- Subjects
- Aged, Aged, 80 and over, Bone Marrow, Humans, Pilot Projects, Time Factors, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute drug therapy, Tretinoin administration & dosage, Valproic Acid administration & dosage
- Abstract
Valproic acid (VPA) has been demonstrated to be able to inhibit histone deacetylase activity and to synergize with all-trans retinoic acid (ATRA) in inducing the differentiation of acute myeloid leukemia (AML) cells. A pilot study of the VPA/ATRA combination was performed in 11 elderly patients with de novo AML (median age, 82 years). Complete marrow response was observed in 3 patients, including 1 complete remission. Two additional patients had hematologic improvement.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.