12 results on '"Bailén, R."'
Search Results
2. Sinusoidal Obstruction Syndrome/Veno-Occlusive Disease after Unmanipulated Haploidentical Hematopoietic Stem Cell Transplantation with Post-Transplantation Cyclophosphamide: A Study on Behalf of the Spanish Hematopoietic Stem Cell Transplantation and Cellular Therapy Group (GETH)
- Author
-
Gómez-Centurión, I., Gallardo Morillo, A.I., Pérez Martínez, A., Cabrero Calvo, M., Chinea, A., González, L., Pedraza, A., Jiménez Lorenzo, M.J., Robles, M. Calbacho, Bailén, R., Cascón, M. J. Pascual, Cabero, A., Piñana Sánchez, J.L., Luna, A., Perera Alvarez, M., Rovira, M., Torrent Catarineu, A., Sánchez-Pina, J., and Kwon, M.
- Abstract
•This study describes characteristics of patients with SOS/VOD after haplo-HSCT with PT-Cy.•Among the 797 haplo-HSCT recipients, 46 patients (5.77%) were diagnosed from SOS/VOD.•Most of the patients developed very severe SOS/VOD according to revised EBMT severity criteria.•Despite a promising SOS/VOD CR rate (65%), 100-day mortality remained high (43%).
- Published
- 2024
- Full Text
- View/download PDF
3. Successful allogeneic CD34 + hematopoietic stem cell boost for prolonged cytopenias following CAR T-cell therapy in B-cell acute lymphoblastic leukemia. On behalf of the Spanish Group for Hematopoietic Transplantation and Cellular Therapy (GETH-TC).
- Author
-
Molinos-Quintana Á, Martínez-Cibrian N, Alonso-Saladrigues A, Galán-Gómez V, Bailén R, Buendía-López S, Fuentes-Socorro C, Kwon M, González-Vincent M, Pérez de Soto C, González-Martínez B, Rives S, Pérez-Hurtado JM, Ortiz-Maldonado V, and Pérez-Simón JA
- Abstract
Competing Interests: Competing interests The authors declare no competing interests. Ethics The study involving humans was approved by the local institutional ethics committee: CEI de los Hospitales Universitarios Virgen Macarena y Virgen del Rocı́o (Code: 0600-N-22). The study was conducted in accordance with the local legislation and institutional requirements. Written Informed consent was obtained from all subjects or participants’ legal guardians protected by the GETH-TC.
- Published
- 2024
- Full Text
- View/download PDF
4. Post-transplant cyclophosphamide compared to sirolimus/tacrolimus in reduced intensity conditioning transplants for patients with lymphoid malignancies.
- Author
-
Fox ML, García-Cadenas I, Navarro V, Martínez AP, Kara M, Bazán IS, Ferra Coll C, Bailén R, Bento L, Parody R, Esquirol A, Ortí G, Mussetti A, Salamero O, Martino R, González AP, Barba P, Kwon M, Solano C, Bosch F, and Valcárcel D
- Subjects
- Humans, Female, Male, Adult, Middle Aged, Aged, Young Adult, Adolescent, Cyclophosphamide therapeutic use, Tacrolimus therapeutic use, Tacrolimus administration & dosage, Sirolimus therapeutic use, Sirolimus pharmacology, Sirolimus administration & dosage, Transplantation Conditioning methods, Hematopoietic Stem Cell Transplantation methods, Graft vs Host Disease prevention & control, Graft vs Host Disease etiology
- Abstract
Despite novel cellular and immunomodulatory therapies, allogeneic hematopoietic stem cell transplantation (HSCT) remains a treatment option for lymphoid malignancies. Post-transplant cyclophosphamide (PTCY) is increasingly employed for graft vs. host disease (GVHD) prophylaxis. This study aims to evaluate the safety and efficacy of PTCY in reduce intensity (RIC) HSCT for patients with lymphoid neoplasms compared to sirolimus with tacrolimus (SIR/TAC). The primary endpoint was to compare grade III-IV acute GVHD, severe chronic GVHD, and relapse-free survival (GRFS) between the two GVHD prophylaxis strategies. This study, conducted from January 2012 to December 2020, included 171 consecutive patients (82 in the PTCY and 89 in the SIR/TAC group). Results revealed a significantly decreased incidence of moderate and severe forms of chronic GVHD in PTCY cohort (5.8% [95% CI, 1.8 to 13.1]) versus the SIR/TAC cohort (39.6% [95% CI, 29.3 to 49.7] (p < 0.001)). Other outcomes, including GRFS (PTCY [45.9% (95% CI, 35.8-58.7)] and SIR/TAC groups [36.8% (95% CI, 28-48.4)], (p = 0.72)), non-relapse mortality (NRM), relapse and overall survival (OS) were similar in both groups. Interestingly, the failure to achieve GRFS was mainly attributed to GVHD in the SIR/TAC group, while disease relapse was the primary reason in the PTCY cohort., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
5. Anti-CD19 CAR-T Cell Therapy in Elderly Patients: Multicentric Real-World Experience from GETH-TC/GELTAMO.
- Author
-
Bailén R, Iacoboni G, Delgado J, López-Corral L, Hernani-Morales R, Ortiz-Maldonado V, Guerreiro M, Caballero AC, Guerra-Domínguez ML, Sánchez-Pina JM, Peña M, Torrent A, Pérez-Martínez A, Bastos-Oreiro M, Reguera-Ortega JL, Martín A, Hernandez-Boluda JC, Martínez-Cibrián N, Sanz J, Briones J, Henriquez HL, Calbacho M, Mussetti A, Sancho JM, Barba P, and Kwon M
- Subjects
- Humans, Aged, Male, Female, Retrospective Studies, Middle Aged, Antigens, CD19 therapeutic use, Antigens, CD19 immunology, Aged, 80 and over, Lymphoma, Large B-Cell, Diffuse therapy, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse mortality, Treatment Outcome, Adult, Biological Products therapeutic use, Receptors, Chimeric Antigen therapeutic use, Receptors, Antigen, T-Cell, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods
- Abstract
Chimeric antigen receptor (CAR)-T cell therapy is approved for the treatment of relapsed/refractory (R/R) large B cell lymphoma (LBCL). However, elderly patients might not be candidates for this therapy due to its toxicity, and criteria for candidate selection are lacking. Our aim was to analyze efficacy and toxicity results of CAR-T cell therapy in the population of patients 70 years and older as compared to those obtained in younger patients in the real-world setting. A multicentric retrospective study was performed including patients with R/R aggressive LBCL who received commercial CAR-T cell therapy with either tisagenlecleucel or axicabtagene ciloleucel within the Spanish Group of Hematopoietic Transplant and Cell Therapy/Spanish Group of Lymphomas and Autologous Transplant (GETH-TC/GELTAMO) centers between 2019 and 2023. As of August 2023, 442 adult patients with aggressive LBCL underwent apheresis for CAR-T cell therapy as third or subsequent line and follow-up data was collected. Of 412 infused patients, 71 (17%) were 70 years or older. Baseline characteristics, product selection, and characteristics at apheresis (including disease status, Ann Arbor stage, revised international prognosis index (R-IPI), bulky disease, lactate dehydrogenase [LDH] and ECOG [Eastern Cooperative Group performance status]) were comparable between groups. Median time from both approval to infusion and apheresis to infusion did not differ. No differences were found between groups in overall and complete response rates at 1 and 3 months. With a median follow-up of 12.2 months (range 1-44), 12-month progression-free survival (PFS) and overall survival (OS) were comparable between groups (35.2% in <70 years vs. 35.9% in ≥70 years (P = .938) and 51.1% and 52.1% (P = .885), respectively). Age ≥70 years did not affect PFS (hazard ratio (HR) 0.98, P = .941) and OS (HR 0.97, P = .890) in the univariate and multivariate analysis. Cytokine release syndrome (CRS) was observed in 82% of patients <70 years old and 84.5% in ≥ 70 years old (P = .408). Grade ≥3 CRS was more frequent in the older group (5% vs. 15%, P = .002). In the multivariate analysis, age ≥70 years was associated with an increased risk of grade ≥3 CRS (OR 3.7, P = .013). No differences were observed in terms of overall neurotoxicity (35% vs. 42%, P = .281) or grade ≥3 (12% vs. 17%, P = .33). The proportion of patients with infections, admission to the intensive care unit within the first month, and non-relapse mortality were similar between both groups. CAR-T cell therapy in patients older than 70 years showed similar efficacy to that observed in younger patients in the real-world setting. However, age ≥70 years was an independent risk factor for grades 3-4 CRS. The need for additional strategies to reduce toxicity in this population should be addressed in future studies., (Copyright © 2024 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Identification of predictive models including polymorphisms in cytokines genes and clinical variables associated with post-transplant complications after identical HLA-allogeneic stem cell transplantation.
- Author
-
Muñiz P, Martínez-García M, Bailén R, Chicano M, Oarbeascoa G, Triviño JC, de la Iglesia-San Sebastian I, Fernández de Córdoba S, Anguita J, Kwon M, Díez-Martín JL, Olmos PM, Martínez-Laperche C, and Buño I
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Young Adult, Adolescent, Hematologic Neoplasms therapy, Hematologic Neoplasms genetics, Hematologic Neoplasms mortality, HLA Antigens genetics, HLA Antigens immunology, Polymorphism, Genetic, Aged, Hematopoietic Stem Cell Transplantation adverse effects, Cytokines genetics, Graft vs Host Disease genetics, Graft vs Host Disease etiology, Transplantation, Homologous adverse effects
- Abstract
Backgrounds: Although allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for hematological malignancies, it can be associated with relevant post-transplant complications. Several reports have shown that polymorphisms in immune system genes are correlated with the development of post-transplant complications. Within this context, this work focuses on identifying novel polymorphisms in cytokine genes and developing predictive models to anticipate the risk of developing graft-versus-host disease (GVHD), transplantation-related mortality (TRM), relapse and overall survival (OS)., Methods: Our group developed a 132-cytokine gene panel which was tested in 90 patients who underwent an HLA-identical sibling-donor allo-HSCT. Bayesian logistic regression (BLR) models were used to select the most relevant variables. Based on the cut-off points selected for each model, patients were classified as being at high or low-risk for each of the post-transplant complications (aGVHD II-IV, aGVHD III-IV, cGVHD, mod-sev cGVHD, TRM, relapse and OS)., Results: A total of 737 polymorphisms were selected from the custom panel genes. Of these, 41 polymorphisms were included in the predictive models in 30 cytokine genes were selected (17 interleukins and 13 chemokines). Of these polymorphisms, 5 (12.2%) were located in coding regions, and 36 (87.8%) in non-coding regions. All models had a statistical significance of p<0.0001., Conclusion: Overall, genomic polymorphisms in cytokine genes make it possible to anticipate the development all complications studied following allo-HSCT and, consequently, to optimize the clinical management of patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Muñiz, Martínez-García, Bailén, Chicano, Oarbeascoa, Triviño, de la Iglesia-San Sebastian, Fernández de Córdoba, Anguita, Kwon, Díez-Martín, Olmos, Martínez-Laperche and Buño.)
- Published
- 2024
- Full Text
- View/download PDF
7. Efficacy and safety of bendamustine-containing bridging therapy in R/R LBCL patients receiving CD19 CAR T-cells.
- Author
-
Iacoboni G, Sánchez-Salinas MA, Rejeski K, Martín-López AÁ, Kwon M, Navarro V, Jalowiec KA, Hernani R, Reguera-Ortega JL, Gallur L, Blumenberg V, Herrero-García M, Roddie C, Benzaquén A, Delgado-Serrano J, Bailén R, Carpio C, Amat P, López-Corral L, Martín-Martín L, Bastos M, Subklewe M, O'Reilly M, and Barba P
- Abstract
Bridging therapy (BT) after leukapheresis is required in most relapsed/refractory (R/R) large B-cell lymphoma (LBCL) patients receiving chimeric antigen receptor (CAR) T cells. Bendamustine-containing regimens are a potential BT option. We aimed to assess if this agent had a negative impact on CAR-T outcomes when it was administered as BT. We included R/R LBCL patients from six centers who received systemic BT after leukapheresis from February 2019 to September 2022; patients who only received steroids or had pre-apheresis bendamustine exposure were excluded. Patients were divided into two BT groups, with and without bendamustine. Separate safety and efficacy analyses were carried out for axi-cel and tisa-cel. Of 243 patients who received BT, bendamustine (benda) was included in 62 (26%). There was a higher rate of BT progressors in the non-benda group (62% vs. 45%, p = 0.02). Concerning CAR-T efficacy, complete responses were comparable for benda versus non-benda BT cohorts with axi-cel (70% vs. 53%, p = 0.12) and tisa-cel (44% vs. 36%, p = 0.70). Also, 12-month progression-free and overall survival were not significantly different between BT groups with axi-cel (56% vs. 43% and 71% vs. 63%) and tisa-cel (25% vs. 26% and 52% vs. 48%); there were no differences when BT response was considered. CAR T-cell expansion for each construct was similar between BT groups. Regarding safety, CRS G ≥3 (6% vs. 6%, p = 0.79), ICANS G ≥3 (15% vs. 17%, p = 0.68), severe infections, and neutropenia post-infusion were comparable among BT regimens. BT with bendamustine-containing regimens is safe for patients requiring disease control during CAR T-cell manufacturing., Competing Interests: Gloria Iacoboni: Consultancy and Honoraria: Novartis, Roche, Kite/Gilead, Bristol‐Myers Squibb, Abbvie, Janssen, Sandoz, Miltenyi, AstraZeneca. Mario A. Sánchez‐Salinas Honoraria for presentations: Kite. Support for attending meetings: Takeda. Kai Rejeski: Kite/Gilead: Research Funding, Consultancy, Honoraria and travel support; Novartis: Honoraria; BMS/Celgene: Consultancy, Honoraria; Pierre‐Fabre: travel support. Mi Kwon Consulting and lectures: Gilead, Jazz, Pfizer. Katarzyna A. Jalowiec Honoraria: Kite/Gilead. Rafael Hernani: Research: Gilead. Travel support: Gilead. Honoraria: Gilead, Janssen, MSD, Celgene, Novartis. Viktoria Blumenberg: BMS/Celgene: Research Funding; Kite/Gilead: Consultancy, Honoraria, Research Funding; Janssen: Research Funding, Honoraria; Novartis: Research Funding, Honoraria; Roche: Consultancy, Research Funding; Takeda: Research Funding. Claire Roddie: Honoraria from Kite/Gilead, Novartis, BMS, Amgen. Javier Delgado‐Serrano: Honoraria from Kite‐Gilead, Novartis, Bristol Myers Squibb, Janssen. Rebeca Bailén: Speaker and travel: Kite. Cecilia Carpio: Regeneron: Consultancy/Advisory, BMS: Consultancy/Advisory, Takeda: Consultancy/Advisory/Honoraria, Novartis: Honoraria. Marion Subklewe: receives industry research support from Amgen, Bristol‐Myers Squibb/Celgene, Gilead, Janssen, Miltenyi Biotec, Morphosys, Novartis, Roche, Seattle Genetics, and Takeda, and serves as a consultant/advisor to AvenCell, CDR‐Life, Ichnos Sciences, Incyte Biosciences, Janssen, Molecular Partners, and Takeda. She serves on the speakers' bureau at Amgen, AstraZeneca, BMS/Celgene, Gilead, GSK, Janssen, Novartis, Pfizer, Roche, and Takeda. Maeve O'Reilly: Honoraria from Kite, Novartis, Janssen. Advisory boards Kite and Autolus. Travel grant Kite and Novartis. Pere Barba: Allogene: Honoraria; Amgen: Honoraria; BMS: Honoraria; Kite/Gilead: Honoraria; Janssen: Honoraria; Jazz Pharmaceuticals: Honoraria; Miltenyi: Honoraria; Novartis: Honoraria; Nektar: Honoraria., (© 2024 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2024
- Full Text
- View/download PDF
8. Dynamics of virological and immunological markers of HIV persistence after allogeneic haematopoietic stem-cell transplantation in the IciStem cohort: a prospective observational cohort study.
- Author
-
Salgado M, Gálvez C, Nijhuis M, Kwon M, Cardozo-Ojeda EF, Badiola J, Gorman MJ, Huyveneers LEP, Urrea V, Bandera A, Jensen BO, Vandekerckhove L, Jurado M, Raj K, Schulze Zur Wiesch J, Bailén R, Eberhard JM, Nabergoj M, Hütter G, Saldaña-Moreno R, Oldford S, Barrett L, Ramirez MLM, Garba S, Gupta RK, Revollo B, Ferra-Coll C, Kuball J, Alter G, Sáez-Cirión A, Diez-Martin JL, Duke ER, Schiffer JT, Wensing A, and Martinez-Picado J
- Subjects
- Humans, Male, Prospective Studies, Female, Adult, Middle Aged, HIV-1 immunology, Transplantation, Homologous, Biomarkers blood, Viral Load, HIV Antibodies blood, Hematopoietic Stem Cell Transplantation adverse effects, HIV Infections immunology, HIV Infections virology
- Abstract
Background: Allogeneic haematopoietic stem-cell transplantation (allo-HSCT) markedly reduces HIV reservoirs, but the mechanisms by which this occurs are only partly understood. In this study, we aimed to describe the dynamics of virological and immunological markers of HIV persistence after allo-HSCT., Methods: In this prospective observational cohort study, we analysed the viral reservoir and serological dynamics in IciStem cohort participants with HIV who had undergone allo-HSCT and were receiving antiretroviral therapy, ten of whom had received cells from donors with the CCR5Δ32 mutation. Participants from Belgium, Canada, Germany, Italy, the Netherlands, Spain, Switzerland, and the UK were included in the cohort both prospectively and retrospectively between June 1, 2014 and April 30, 2019. In the first 6 months after allo-HSCT, participants had monthly assessments, with annual assessments thereafter, with the protocol tailored to accommodate for the individual health status of each participant. HIV reservoirs were measured in blood and tissues and HIV-specific antibodies were measured in plasma. We used the Wilcoxon signed-rank test to compare data collected before and after allo-HSCT in participants for whom longitudinal data were available. When the paired test was not possible, we used the Mann-Whitney U test. We developed a mathematical model to study the factors influencing HIV reservoir reduction in people with HIV after allo-HSCT., Findings: We included 30 people with HIV with haematological malignancies who received a transplant between Sept 1, 2009 and April 30, 2019 and were enrolled within the IciStem cohort and included in this analysis. HIV reservoirs in peripheral blood were reduced immediately after full donor chimerism was achieved, generally accompanied by undetectable HIV-DNA in bone marrow, ileum, lymph nodes, and cerebrospinal fluid, regardless of donor CCR5 genotype. HIV-specific antibody levels and functionality values declined more slowly than direct HIV reservoir values, decaying significantly only months after full donor chimerism. Mathematical modelling suggests that allogeneic immunity mediated by donor cells is the main viral reservoir depletion mechanism after massive reservoir reduction during conditioning chemotherapy before allo-HSCT (half-life of latently infected replication-competent cells decreased from 44 months to 1·5 months)., Interpretation: Our work provides, for the first time, data on the effects of allo-HSCT in the context of HIV infection. Additionally, we raise the question of which marker can serve as the last reporter of the residual viraemia, postulating that the absence of T-cell immune responses might be a more reliable marker than antibody decline after allo-HSCT., Funding: amfAR (American Foundation for AIDS Research; ARCHE Program), National Institutes of Health, National Institute of Allergy and Infectious Diseases, and Dutch Aidsfonds., Competing Interests: Declaration of interests AB reports grants from Gilead Sciences and participating on the advisory board of ViiV Healthcare. AW reports funding for this manuscript from the American Foundation for AIDS Research (amfAR) and Aidsfunds; grants from Gilead and NOW; consulting fees from ViiV Healthcare/GSK, MSD, and Gilead Sciences; participating on the board of the Dutch Federation of Medical Microbiology, the board of the European Society for Translational Antiviral Research, chair on the IAS-USA mutations work group, the Committee of ZonMW (Dutch research organization) Research, and the Committee of the Dutch Federation for Long Covid; and received funding from Ark. AS-C reports funding for this manuscript from amfAR; grants from ANRS, the National Institutes of Health (NIH), Institute Pasteur, and MSDAVENIR; honoraria from MSD, ViiV Healthcare, and Gilead Sciences; and is chair of the Scientific and Medical Committee of Sidaction. B-EOJ reports consulting fees from Gilead Sciences, ViiV Healthcare, and Merck Sharp & Dohme; honoraria from Gilead Sciences and ViiV Healthcare; travel expenses for attending meetings from Gilead; and is scientific secretary for the German AIDS Society. BR reports honoraria from Gilead Sciences, Janssen, and ViiV Healthcare; payment for advice from ViiV Healthcare; and travel expenses for attending meetings and travel from ViiV Healthcare and Gilead Sciences. GH reports travel expenses for attending the meeting and travel for the HIV Persistence Workshop 2022. JB reports receiving honoraria from AbbVie, Pfizer, and Gilead Sciences; and travel expenses for attending meetings from AbbVie, Pfizer, and Gilead Sciences. JK reports grants from Novartis and Miltenyi Biotech; royalties from GADETA and Miltenyi Biotech; a patent with GADETA; and holds stock interest in GADETA. JM-P reports funding for this manuscript from amfAR. JSZW reports funding for this manuscript from The German Center for Infection Research, EU H2020 Research and Innovation Programme, HW & J Hector Foundation, the German Research Foundation, The Hamburg Investment and Development Bank, and amfAR; and honoraria from Nobite, GSK, and Gilead Sciences. JTS reports funding for this manuscript from the NIH and National Institute of Allergy and Infectious Diseases. LB report grants from Abbvie and Gilead Sciences; consulting fees from Abbvie and Gilead Sciences; and honoraria from AbbVie and Gilead Sciences. LV reports receiving grants from ViiV Healthcare and Gilead Sciences; and consulting fees from ViiV Healthcare and Gilead Sciences. MJG and GA declare being an employee of Ragon Institute of Mass General, MIT, and Harvard during the study; and an employee of Moderna afterwards. MNi reports receiving consulting fees from Gilead Sciences; and honoraria for lectures from ViiV Healthcare. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
9. Correction: Need for ICU and outcome of critically ill patients with COVID-19 and haematological malignancies: results from the EPICOVIDEHA survey.
- Author
-
Lahmer T, Salmanton-García J, Marchesi F, El-Ashwah S, Nucci M, Besson C, Itri F, Jaksic O, Čolović N, Weinbergerová B, Seval GC, Adžić-Vukičević T, Szotkowski T, Sili U, Dargenio M, van Praet J, van Doesum J, Schönlein M, Ráčil Z, Žák P, Poulsen CB, Magliano G, Jiménez M, Bonuomo V, Piukovics K, Dragonetti G, Demirkan F, Blennow O, Valković T, Gomes Da Silva M, Maertens J, Glenthøj A, Fernández N, Bergantim R, Verga L, Petzer V, Omrani AS, Méndez GA, Machado M, Ledoux MP, Bailén R, Duarte RF, Del Principe MI, Farina F, Martín-Pérez S, Dávila-Valls J, Marchetti M, Bilgin YM, Fracchiolla NS, Cattaneo C, Espigado I, Cordoba R, Collins GP, Labrador J, Falces-Romero I, Prezioso L, Meers S, Passamonti F, Buquicchio C, López-García A, Kulasekararaj A, Ormazabal-Vélez I, Cuccaro A, Garcia-Vidal C, Busca A, Navrátil M, de Jonge N, Biernat MM, Guidetti A, Abu-Zeinah G, Samarkos M, Anastasopoulou A, de Ramón C, González-López TJ, Hoenigl M, Finizio O, Pinczés LI, Ali N, Vena A, Tascini C, Stojanoski Z, Merelli M, Emarah Z, Kohn M, Barać A, Mladenović M, Mišković B, Ilhan O, Çolak GM, Čerňan M, Gräfe SK, Ammatuna E, Hanakova M, Víšek B, Cabirta A, Nordlander A, Nunes Rodrigues R, Hersby DS, Zambrotta GPM, Wolf D, Núñez-Martín-Buitrago L, Arellano E, Aiello TF, García-Sanz R, Prattes J, Egger M, Limongelli A, Bavastro M, Cvetanoski M, Dibos M, Rasch S, Rahimli L, Cornely OA, and Pagano L
- Published
- 2024
- Full Text
- View/download PDF
10. Need for ICU and outcome of critically ill patients with COVID-19 and haematological malignancies: results from the EPICOVIDEHA survey.
- Author
-
Lahmer T, Salmanton-García J, Marchesi F, El-Ashwah S, Nucci M, Besson C, Itri F, Jaksic O, Čolović N, Weinbergerová B, Seval GC, Adžić-Vukičević T, Szotkowski T, Sili U, Dargenio M, van Praet J, van Doesum J, Schönlein M, Ráčil Z, Žák P, Poulsen CB, Magliano G, Jiménez M, Bonuomo V, Piukovics K, Dragonetti G, Demirkan F, Blennow O, Valković T, Gomes Da Silva M, Maertens J, Glenthøj A, Fernández N, Bergantim R, Verga L, Petzer V, Omrani AS, Méndez GA, Machado M, Ledoux MP, Bailén R, Duarte RF, Del Principe MI, Farina F, Martín-Pérez S, Dávila-Valls J, Marchetti M, Bilgin YM, Fracchiolla NS, Cattaneo C, Espigado I, Cordoba R, Collins GP, Labrador J, Falces-Romero I, Prezioso L, Meers S, Passamonti F, Buquicchio C, López-García A, Kulasekararaj A, Ormazabal-Vélez I, Cuccaro A, Garcia-Vidal C, Busca A, Navrátil M, de Jonge N, Biernat MM, Guidetti A, Abu-Zeinah G, Samarkos M, Anastasopoulou A, de Ramón C, González-López TJ, Hoenigl M, Finizio O, Pinczés LI, Ali N, Vena A, Tascini C, Stojanoski Z, Merelli M, Emarah Z, Kohn M, Barać A, Mladenović M, Mišković B, Ilhan O, Çolak GM, Čerňan M, Gräfe SK, Ammatuna E, Hanakova M, Víšek B, Cabirta A, Nordlander A, Nunes Rodrigues R, Hersby DS, Zambrotta GPM, Wolf D, Núñez-Martín-Buitrago L, Arellano E, Aiello TF, García-Sanz R, Prattes J, Egger M, Limongelli A, Bavastro M, Cvetanoski M, Dibos M, Rasch S, Rahimli L, Cornely OA, and Pagano L
- Subjects
- Humans, Male, Middle Aged, Female, Aged, Surveys and Questionnaires, Adult, COVID-19 epidemiology, Hematologic Neoplasms complications, Hematologic Neoplasms epidemiology, Critical Illness, Intensive Care Units statistics & numerical data, SARS-CoV-2
- Published
- 2024
- Full Text
- View/download PDF
11. Digital PCR Improves Sensitivity and Quantification in Monitoring CAR-T Cells in B Cell Lymphoma Patients.
- Author
-
de la Iglesia-San Sebastián I, Carbonell D, Bastos-Oreiro M, Pérez-Corral A, Bailén R, Chicano M, Muñiz P, Monsalvo S, Escudero-Fernández A, Oarbeascoa G, Fernández-Caldas P, Gómez-Centurión I, Pion M, Gayoso J, Anguita J, Kwon M, Díez-Martín JL, Buño I, and Martínez-Laperche C
- Subjects
- Humans, Immunotherapy, Adoptive adverse effects, T-Lymphocytes, Polymerase Chain Reaction, Receptors, Chimeric Antigen genetics, Lymphoma, B-Cell etiology
- Abstract
Chimeric antigen receptor T cells (CAR-T) has emerged as a promising therapy, over 60% of patients fail to sustain a long-term response. The underlying factors that leads to the effectiveness of this therapy are not completely understood, CAR-T cell persistence and monitoring seems to be pivotal for ensuring a successful response. Various monitoring methods such as multiparametric flow cytometry (MFC) or quantitative PCR (qPCR) have been applied. Our objective is to develop digital PCR (dPCR) assays for detection and quantification of CAR-T cells, comparing them with MFC and qPCR. Samples taken at different follow-up times from 45 patients treated with CAR-T therapy were analyzed to assess the correlation between the different methodologies. dPCR presented a high correlation with MFC and qPCR (r = 0.97 and r = 0.87, respectively), while offering a higher sensitivity (0.01%) compared to MFC (0.1%) and qPCR (1%). dPCR emerged as an alternative and highly sensitivity method for monitoring CAR-T cell dynamics. This technique is well-suited for implementation in clinical practice as a complementary technique to MFC., (Copyright © 2024 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Recent Bendamustine Treatment Before Apheresis Has a Negative Impact on Outcomes in Patients With Large B-Cell Lymphoma Receiving Chimeric Antigen Receptor T-Cell Therapy.
- Author
-
Iacoboni G, Navarro V, Martín-López AÁ, Rejeski K, Kwon M, Jalowiec KA, Amat P, Reguera-Ortega JL, Gallur L, Blumenberg V, Gutiérrez-Herrero S, Roddie C, Benzaquén A, Delgado-Serrano J, Sánchez-Salinas MA, Bailén R, Carpio C, López-Corral L, Hernani R, Bastos M, O'Reilly M, Martín-Martín L, Subklewe M, and Barba P
- Subjects
- Humans, Bendamustine Hydrochloride adverse effects, Immunotherapy, Adoptive adverse effects, Antigens, CD19, Cell- and Tissue-Based Therapy, Receptors, Chimeric Antigen, Blood Component Removal, Lymphoma, Large B-Cell, Diffuse
- Abstract
Purpose: Approximately 30%-40% of patients with relapsed/refractory (R/R) large B-cell lymphoma (LBCL) infused with CD19-targeted chimeric antigen receptor (CAR) T cells achieve durable responses. Consensus guidelines suggest avoiding bendamustine before apheresis, but specific data in this setting are lacking. We report distinct outcomes after CAR T-cell therapy according to previous bendamustine exposure., Methods: The study included CAR T-cell recipients from seven European sites. Safety, efficacy, and CAR T-cell expansion kinetics were analyzed according to preapheresis bendamustine exposure. Additional studies on the impact of the washout period and bendamustine dose were performed. Inverse probability treatment weighting (IPTW) and propensity score matching (PSM) analyses were carried out for all efficacy comparisons between bendamustine-exposed and bendamustine-naïve patients., Results: The study included 439 patients with R/R LBCL infused with CD19-targeted commercial CAR T cells, of whom 80 had received bendamustine before apheresis. Exposed patients had significantly lower CD3
+ cells and platelets at apheresis. These patients had a lower overall response rate (ORR, 53% v 72%; P < .01), a shorter progression-free survival (PFS, 3.1 v 6.2 months; P = .04), and overall survival (OS, 10.3 v 23.5 months; P = .01) in comparison with the bendamustine-naïve group. Following adjustment methods for baseline variables, these differences were mitigated. Focusing on the impact of bendamustine washout before apheresis, those with recent (<9 months) exposure (N = 42) displayed a lower ORR (40% v 72%; P < .01), shorter PFS (1.3 v 6.2 months; P < .01), and OS (4.6 v 23.5 months; P < .01) in comparison with bendamustine-naïve patients. These differences remained significant after IPTW and PSM analysis. Conversely, the cumulative dose of bendamustine before apheresis did not affect CAR-T efficacy outcomes., Conclusion: Recent bendamustine exposure before apheresis was associated with negative treatment outcomes after CD19-targeted CAR T-cell therapy and should be therefore avoided in CAR T-cell candidates.- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.