7 results on '"Díez-Campelo M"'
Search Results
2. Chronic graft-versus-host disease could ameliorate the impact of adverse somatic mutations in patients with myelodysplastic syndromes and hematopoietic stem cell transplantation
- Author
-
Caballero JC, Sánchez Barba M, Hernández Sánchez JM, Such E, Janusz K, Sanz G, Cabrero M, Chillón C, Cervera J, Hurtado AM, Jerez A, Calderón Cabrera C, Valcárcel D, Lumbreras E, Abáigar M, López Cadenas F, Hernández Rivas JM, Del Cañizo MC, and Díez Campelo M
- Subjects
hemic and lymphatic diseases ,Allogeneic hematopoietic stem cell transplantation, Chronic graft-versus-host disease, Myelodysplastic syndromes, Somatic mutations, TP53 - Abstract
Somatic mutations in patients with myelodysplastic syndromes (MDS) undergoing allogeneic hematopoietic stem cell transplantation (HSTC) are associated with adverse outcome, but the role of chronic graft-versus-host disease (cGVHD) in this subset of patients remains unknown. We analyzed bone marrow samples from 115 patients with MDS collected prior to HSCT using next-generation sequencing. Seventy-one patients (61%) had at least one mutated gene. We found that patients with a higher number of mutated genes (more than 2) had a worse outcome (2years overall survival [OS] 54.8% vs. 31.1%, p = 0.035). The only two significant variables in the multivariate analysis for OS were TET2 mutations (p = 0.046) and the development of cGVHD, considered as a time-dependent variable (p 0.001), correlated with a worse and a better outcome, respectively. TP53 mutations also demonstrated impact on the cumulative incidence of relapse (CIR) (1year CIR 47.1% vs. 9.8%, p = 0.006) and were related with complex karyotype (p = 0.003). cGVHD improved the outcome even among patients with more than 2 mutated genes (1-year OS 88.9% at 1year vs. 31.3%, p = 0.02) and patients with TP53 mutations (1-year CIR 20% vs. 42.9%, p = 0.553). These results confirm that cGVHD could ameliorate the adverse impact of somatic mutations in patients with MDS with HSCT.
- Published
- 2019
3. Erythroleukemia shares biological features and outcome with myelodysplastic syndromes with excess blasts: a rationale for its inclusion into future classifications of myelodysplastic syndromes
- Author
-
Calvo X, Arenillas L, Luño E, Senent L, Arnan M, Ramos F, Ardanaz MT, Pedro C, Tormo M, Montoro J, Díez-Campelo M, Arrizabalaga B, Xicoy B, Bonanad S, Jerez A, Nomdedeu B, Ferrer A, Sanz GF, and Florensa L
- Subjects
hemic and lymphatic diseases - Abstract
Erythroleukemia was considered an acute myeloid leukemia in the 2008 World Health Organization (WHO) classification and is defined by the presence of >= 50% bone marrow erythroblasts, having = 20% bone marrow myeloblasts from nonerythroid cells. Erythroleukemia shares clinicopathologic features with myelodysplastic syndromes, especially with erythroid-predominant myelodysplastic syndromes (>= 50% bone marrow erythroblasts). The upcoming WHO revision proposes to eliminate the nonerythroid blast cell count rule and to move erythroleukemia patients into the appropriate myelodysplastic syndrome category on the basis of the absolute blast cell count. We conducted a retrospective study of patients with de novo erythroleukemia and compared their clinico-biological features and outcome with those of de novo myelodysplastic syndromes, focusing on erythroid-predominant myelodysplastic syndromes. Median overall survival of 405 erythroid-predominant myelodysplastic syndromes without excess blasts was significantly longer than that observed in 57 erythroid-predominant refractory anemias with excess blasts-1 and in 59 erythroleukemias, but no significant difference was observed between erythroid-predominant refractory anemias with excess blasts-1 and erythroleukemias. In this subset of patients with >= 50% bone marrow erythroblasts and excess blasts, the presence of a high-risk karyotype defined by the International Prognostic Scoring System or by the Revised International Prognostic Scoring System was the main prognostic factor. In the same way, the survival of 459 refractory anemias with excess blasts-2, independently of having >= 20% bone marrow blasts from nonerythroid cells or not, was almost identical to the observed in 59 erythroleukemias. Interestingly, 11 low-blast count erythroleukemias with 5 to < 10% bone marrow blasts from total nucleated cells showed similar survival than the rest of erythroleukemias. Our data suggest that de novo erythroleukemia is in the spectrum of myelodysplastic syndromes with excess blasts and support its inclusion into future classifications of myelodysplastic syndromes.
- Published
- 2016
4. Azacitidine frontline therapy for unfit acute myeloid leukemia patients: Clinical use and outcome prediction
- Author
-
Ramos F., Maurillo L., Itzykson R., Bargay J., Stauder R., Venditti A., Seegers V., Gaidano G., Gardin C., Musto P., Greil R., Sánchez-Guijo F., Fenaux P., Thépot S., Récher C., Raffoux E., Quesnel B., Delaunay J., Cluzeau T., Koka A.M., Stamatoullas A., Chaury M.-P., Gyan E., Cheze S., Banos A., Morel P., Plantier I., Taksin A.-L., Shanti A., Sanhes L., De Botton S., Marolleau J.P., Pautas C., Wattel E., Isnard F., Guerci A., Vey N., Dreyfus F., Ifrah N., Ades L., Martínez-Robles V., Debén G., Garrido A., Casaño J., Salamero O., Bergua J.M., Colado E., García R., Pedro C., Redondo S., Tormo M., Bonanad S., Díez-Campelo M., Pérez-Encinas M., de la Fuente A., Xicoy B., Falantes J.F., Font P., González-López T.J., Martín-Núñez G., Montesinos P., Pleyer L., Burgstaller S., Schreder M., Tinchon C., Pfeilstoecker M., Steinkirchner S., Melchardt T., Mitrovic M., Girschikofsky M., Lang A., Krippl P., Sliwa T., Egle A., Linkesch W., Voskova D., Angermann H., Spagnoli A., Lunghi M., Pietrantuono G., and Villani O.
- Subjects
blood toxicity ,cancer patient ,leukocyte count ,very elderly ,retrospective study ,blast cell ,ear disease ,cytogenetics ,middle aged ,neurotoxicity ,Leukemia ,antineoplastic antimetabolite ,Aged, 80 and over ,adult ,nephrotoxicity ,European ALMA score ,clinical trial ,acute granulocytic leukemia ,aged ,Leukemia, Myeloid, Acute ,female ,Italy ,priority journal ,validation study ,drug withdrawal ,Azacitidine ,France ,survival rate ,Antimetabolites, Antineoplastic ,overall survival ,cardiotoxicity ,Article ,eye toxicity ,multiple cycle treatment ,remission ,male ,follow up ,Humans ,controlled study ,drug fatality ,human ,gastrointestinal toxicity ,survival time ,lung toxicity ,human cell ,practice guideline ,Australia ,scoring system ,treatment response ,hearing impairment ,major clinical study ,skin toxicity ,mortality ,infection ,multicenter study ,Spain ,treatment outcome ,population research ,musculoskeletal disease ,Follow-Up Studies - Abstract
Hypomethylating agents are able to prolong the overall survival of some patients diagnosed with acute myeloid leukemia. The aim of this study was to evaluate the clinical use of azacitidine as front-line therapy in unfit acute myeloid leukemia (AML) patients and to develop a clinical prediction model to identify which patients may benefit more from the drug. One hundred and ten untreated unfit AML patients received front-line azacitidine therapy in Spain, and response and survival were evaluated in them following European LeukemiaNet (ELN) guidelines. A clinical prediction rule was obtained from this population that was validated and refined in 261 patients treated in France, Austria and Italy. ELN response was achieved in 21.0% of the 371 patients (CI95% 17.0-25.5) and did not depend on bone marrow blast cell percentage. Median overall survival was 9.6 months (CI95% 8.5-10.8) and 40.6% of the patients were alive at 1 year (CI95% 35.5-45.7). European ALMA score (E-ALMA), based on performance status, white blood cell counts at azacitidine onset and cytogenetics, discriminated three risk groups with different survival and response rates. Azacitidine seems a reasonable therapeutic option for most unfit AML patients, i.e. those displaying a favorable or intermediate E-ALMA score. © 2014 Elsevier Ltd.
- Published
- 2015
5. Panobinostat as part of induction and maintenance for elderly patients with newly diagnosed acute myeloid leukemia: phase Ib/II panobidara study
- Author
-
Ocio EM, Herrera P, Olave MT, Castro N, Pérez-Simón JA, Brunet S, Oriol A, Mateo M, Sanz MÁ, López J, Montesinos P, Chillón MC, Prieto-Conde MI, Díez-Campelo M, González M, Vidriales MB, Mateos MV, San Miguel JF, and PETHEMA Group
- Subjects
II TRIAL ,OUTCOMES ,CELL TRANSPLANTATION ,POOR-PROGNOSIS ,MYELODYSPLASTIC SYNDROMES ,MINIMAL RESIDUAL DISEASE ,VORINOSTAT ,OLDER-ADULTS ,OPEN-LABEL ,POSTREMISSION THERAPY - Abstract
This phase Ib/II trial combined the pan-deacetylase inhibitor panobinostat with chemotherapy followed by panobinostat maintenance in elderly patients with newly diagnosed acute myeloid leukemia. Patients with prior history of myelodysplastic syndrome were excluded and 38 evaluable patients were included in the study (median age: 71 years; range: 65-83). Study patients received an induction with idarubicin (8 mg/m(2) iv days 1-3) plus cytarabine (100 mg/m2 iv days 1-7) plus panobinostat po at escalating doses (days 8, 10, 12, 15, 17 and 19) that could be repeated in non-responding patients. Patients achieving complete remission received a consolidation cycle with the same schema, followed by panobinostat maintenance (40 mg po 3 days/week) every other week until progression. Thirty-one patients were treated at the maximum tolerated dose of panobinostat in the combination (10 mg) with good tolerability. Complete remission rate was 64% with a time to relapse of 17.0 months (12.8-21.1). Median overall survival for the whole series was 17 months (5.5-28.4). Moreover, in 4 of 5 patients with persistent minimal residual disease before maintenance, panobinostat monotherapy reduced its levels, with complete negativization in two of them. Maintenance phase was well tolerated. The most frequent adverse events were thrombocytopenia (25% grades 3/4), and gastrointestinal toxicity, asthenia and anorexia (mainly grades 1/2). Five patients required dose reduction during this phase, but only one discontinued therapy due to toxicity. These results suggest that panobinostat is one of the first novel agents with activity in elderly acute myeloid leukemia patients, and suggest further investigation is warranted, particularly in the context of maintenance therapy.
- Published
- 2015
6. Results of allogeneic stem cell transplantation in the Spanish MDS registry: Prognostic factors for low risk patients
- Author
-
Díez Campelo M, Sánchez-Barba M, de Soria VG, Martino R, Sanz G, Insunza A, Bernal T, Duarte R, Amigo ML, Xicoy B, Tormo M, Iniesta F, Bailén A, Benlloch L, Córdoba I, López-Villar O, Del Cañizo MC, and Spanish Registry of MDS
- Subjects
MDS ,Allogeneic transplant ,Low risk MDS - Abstract
Although new agents have been approved for the treatment of MDS, the only curative approach is allogeneic hematopoietic stem cell transplantation (HSCT) and thus, in particular circumstances this procedure has been proposed as a treatment option for low risk patients. We have retrospectively analyzed the results of HSCT in 291 patients from the Spanish MDS registry with special attention to low risk MDS (LR-MDS) in order to define the variables that could impact their clinical evolution after transplantation. At 2 years OS was 51% and EFS was 50% (95% CI 0.7-4.5 years for OS and 95% CI 0.1-3.9 years for EFS). Among 43 LR-MDS, transplant-related mortality was 28%. At 3 years, OS was 67% (95% CI 264.7-8927.2 days for OS) and EFS was 64% (95% CI 0-9697.2 days for EFS). In the multivariate analysis only cytogenetics retained statistical significant effect on both OS (p=.047) and EFS (p=.046). Conditioning regimen could improve outcome among this subset of patients (OS 86% and RFS 100% for patients receiving RIC regimen). The present study confirms that specific disease characteristic as well as transplant characteristics have a significant impact on transplant outcome. Regarding low risk patients a non-myeloablative conditioning would be preferable especially in cases without high-risk cytogenetics. Copyright (C) 2014 Elsevier Ltd. All rights reserved.
- Published
- 2014
7. Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML
- Author
-
Lionel Adès, Larisa Girshova, Vadim A. Doronin, María Díez-Campelo, David Valcárcel, Suman Kambhampati, Nora-Athina Viniou, Dariusz Woszczyk, Raquel De Paz Arias, Argiris Symeonidis, Achilles Anagnostopoulos, Eduardo Ciliao Munhoz, Uwe Platzbecker, Valeria Santini, Robert J. Fram, Ying Yuan, Sharon Friedlander, Douglas V. Faller, Mikkael A. Sekeres, Institut Català de la Salut, [Adès L] INSERM U944, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Louis and University of Paris, Paris, France. [Girshova L] Federal Almazov North-West Medical Research Centre, Saint-Petersburg, Russia. [Doronin VA] City Clinical Hospital 40, Moscow, Russia. [Díez-Campelo M] Institute for Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain. [Valcárcel D] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Barcelona, Spain. [Kambhampati S] Sarah Cannon at Research Medical Center, Kansas City, MO, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
acciones y usos químicos::acciones farmacológicas::mecanismos moleculares de acción farmacológica::antimetabolitos::antimetabolitos antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,Antimetabolites, Antineoplastic ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Chemical Actions and Uses::Pharmacologic Actions::Molecular Mechanisms of Pharmacological Action::Antimetabolites::Antimetabolites, Antineoplastic [CHEMICALS AND DRUGS] ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Leukemia, Myelomonocytic, Chronic ,neoplasias::neoplasias por tipo histológico::leucemia::leucemia mieloide::leucemia mielomonocítica crónica [ENFERMEDADES] ,Cyclopentanes ,Hematology ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Otros calificadores::Otros calificadores::/efectos adversos [Otros calificadores] ,Quimioteràpia combinada ,Medicaments antineoplàstics - Efectes secundaris ,Neoplasms::Neoplasms by Histologic Type::Leukemia::Leukemia, Myeloid::Leukemia, Myelomonocytic, Chronic [DISEASES] ,Pyrimidines ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Leucèmia mieloide aguda - Tractament ,Other subheadings::Other subheadings::/adverse effects [Other subheadings] ,Azacitidine ,Humans ,Drug Therapy, Combination - Abstract
Pevonedistat; Chronic myelomonocytic leukemia Pevonedistat; Leucemia mielomonocítica crónica Pevonedistat; Leucèmia mielomonocítica crònica PANTHER is a global, randomized phase 3 trial of pevonedistat+azacitidine (n = 227) vs azacitidine monotherapy (n = 227) in patients with newly diagnosed higher-risk myelodysplastic syndromes (MDS; n = 324), higher-risk chronic myelomonocytic leukemia (n = 27), or acute myeloid leukemia (AML) with 20% to 30% blasts (n = 103). The primary end point was event-free survival (EFS). In the intent-to-treat population, the median EFS was 17.7 months with pevonedistat+azacitidine vs 15.7 months with azacitidine (hazard ratio [HR], 0.968; 95% confidence interval [CI], 0.757-1.238; P = .557) and in the higher-risk MDS cohort, median EFS was 19.2 vs 15.6 months (HR, 0.887; 95% CI, 0.659-1.193; P = .431). Median overall survival (OS) in the higher-risk MDS cohort was 21.6 vs 17.5 months (HR, 0.785; P = .092), and in patients with AML with 20% to 30% blasts was 14.5 vs 14.7 months (HR, 1.107; P = .664). In a post hoc analysis, median OS in the higher-risk MDS cohort for patients receiving >3 cycles was 23.8 vs 20.6 months (P = .021) and for >6 cycles was 27.1 vs 22.5 months (P = .008). No new safety signals were identified, and the azacitidine dose intensity was maintained. Common hematologic grade ≥3 treatment emergent adverse events were anemia (33% vs 34%), neutropenia (31% vs 33%), and thrombocytopenia (30% vs 30%). These results underscore the importance of large, randomized controlled trials in these heterogeneous myeloid diseases and the value of continuing therapy for >3 cycles. The trial was registered on clinicaltrials.gov as #NCT03268954. This study was sponsored by Takeda Development Centers Inc (TDCA; Lexington, MA).
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.