CANCER
CB16/12/00400
•
Nombre agencia financiadora Ministerio de Economía y Competitividad
Acrónimo agencia financiadora MINECO
Programa Programa Estatal de I+D+I Orientada a los Retos de la Sociedad
Subprograma Salud, cambio demográfico y bienestar
Convocatoria Ayudas a la incorporación de nuevas áreas temáticas y nuevos grupos al Consorcio CIBER (AE de Salud 2016)
Año convocatoria 2016
Unidad de gestión Instituto de Salud Carlos III (ISCIII)
Centro beneficiario FUNDACIÓN DE INVESTIGACIÓN DEL CÁNCER DE LA UNIVERSIDAD DE SALAMANCA (FICUS)
Centro realización CENTRO DE INVESTIGACION DEL CANCER -IBMCC
Identificador persistente http://dx.doi.org/10.13039/501100003329
Publicaciones
Resultados totales (Incluyendo duplicados): 8
Encontrada(s) 1 página(s)
Encontrada(s) 1 página(s)
Clinical, histopathologic and genetic features of rhabdoid meningiomas
Academica-e. Repositorio Institucional de la Universidad Pública de Navarra
- Garrido Ruiz, Patricia Alejandra
- González-Tablas, María
- Pasco Peña, Alejandro
- Zelaya Huerta, María Victoria
- Ortiz, Javier
- Otero, Álvaro
- Corchete, Luis Antonio
- Ludeña, María Dolores
- Caballero Martínez, María Cristina
- Córdoba Iturriagagoitia, Alicia
- Fernández, Inmaculada Catalina
- González-Carreró Fojón, Joaquín
- Hernández Laín, Aurelio
- Orfao, Alberto
- Tabernero, María Dolores
Rhabdoid meningiomas (RM) shows heterogeneous histological findings, and a wide variety of chromosomal copy number alterations (CNA) are associated with an unpredictable course of the disease. In this study, we analyzed a series of 305 RM samples from patients previously reported in the literature and 33 samples from 23 patients studied in our laboratory. Monosomy 22-involving the minimal but most common recurrent region loss of the 22q11.23 chromosomal region was the most observed chromosomal alteration, followed by losses of chromosomes 14, 1, 6, and 19, polysomies of chromosomes 17, 1q, and 20, and gains of 13q14.2, 10p13, and 21q21.2 chromosomal regions. Based on their CNA profile, RM could be classified into two genetic subgroups with distinct clinicopathologic features characterized by the presence of (1) chromosomal losses only and (2) combined losses and gains of several chromosomes. The latter displays a higher frequency of WHO grade 3 tumors and poorer clinical outcomes., This research was funded by Consejería de Sanidad JCYL, Gerencia Regional de Salud,
Spain grant numbers GRS 2132/A/20 and GRS 2315/A/21 and Instituto de Salud Carlos III, Ministerio
de Economía y Competitividad, Madrid, Spain grant number CIBERONC CB16/12/00400.
Spain grant numbers GRS 2132/A/20 and GRS 2315/A/21 and Instituto de Salud Carlos III, Ministerio
de Economía y Competitividad, Madrid, Spain grant number CIBERONC CB16/12/00400.
Proyecto: MINECO//CB16-12-00400
Unique clinico-biological, genetic and prognostic features of adult early T-cell precursor acute lymphoblastic leukemia
Dipòsit Digital de Documents de la UAB
- Genescà, Eulàlia|||0000-0002-5657-4842
- Morgades, Mireia|||0000-0003-0295-2534
- Montesinos, Pau|||0000-0002-3275-5593
- Barba, Pere|||0000-0001-7076-7969
- Gil, Cristina
- Guàrdia, Ramón
- Moreno, Maria José
- Martínez-Carballeira, Daniel|||0000-0003-4840-7822
- Garcia Cadenas, Irene|||0000-0002-2994-9055
- Vives Polo, Susana|||0000-0003-2217-5285
- Ribera Salas, Jordi|||0000-0003-4796-6470
- González-Campos, José
- González Gil, Celia|||0000-0002-7582-1482
- Zamora, Lurdes|||0000-0003-1713-7110
- Ramirez, Jose Luis
- Díaz-Beyá, Marina|||0000-0001-9624-2597
- Mercadal, Santiago|||0000-0003-4741-7885
- Artola, Maria Teresa
- Cladera, Antonia
- Tormo, Mar|||0000-0001-9622-1649
- Bermúdez, Arancha
- Vall-Llovera, Ferran|||0000-0002-0718-5955
- Martínez, Pilar N.
- Amigo, María-Luz|||0000-0002-3356-9290
- Monsalvo, Silvia
- Novo, Andrés
- Cervera, Marta
- Garcia-Guiñón, Antoni
- Junca, Jordi|||0000-0003-0142-772X
- Ciudad Pizarro, Juana
- Orfao, Alberto|||0000-0002-0007-7230
- Ribera, Jose-Maria|||0000-0003-1042-6024
Comparison of next-generation sequencing (NGS) and next-generation flow (NGF) for minimal residual disease (MRD) assessment in multiple myeloma
Dipòsit Digital de Documents de la UAB
- Medina, Alejandro|||0000-0001-5609-2969
- Puig, Noemí
- Flores-Montero, Juan|||0000-0002-1119-4387
- Jimenez, Cristina
- Sarasquete, M. Eugenia|||0000-0001-7335-3657
- García-Álvarez, María|||0000-0001-8864-1168
- Prieto-Conde, Isabel|||0000-0002-2510-8355
- Chillon, Carmen
- Alcoceba, Miguel|||0000-0002-3819-4846
- Gutierrez, Norma C.
- Oriol, Albert|||0000-0001-6804-2221
- Rosinol, Laura
- Bladé Creixenti, Juan|||0000-0002-4563-3405
- Gironella, Mercedes|||0000-0002-8106-8909
- Hernandez, Miguel T.
- Gonzalez-Calle, Verónica
- Cedena, María Teresa|||0000-0001-5851-3720
- Paiva, Bruno|||0000-0003-1977-3815
- San-Miguel, J|||0000-0002-9183-4857
- Lahuerta, J. J.|||0000-0002-3393-9570
- Mateos, M. V.|||0000-0003-2390-1218
- Martínez-López, Joaquín|||0000-0001-7908-0063
- Orfao, Alberto|||0000-0002-0007-7230
- Gonzalez, Marcos
- García-Sanz, Ramón|||0000-0003-4120-2787
Detecting persistent minimal residual disease (MRD) allows the identification of patients with an increased risk of relapse and death. In this study, we have evaluated MRD 3 months after transplantation in 106 myeloma patients using a commercial next-generation sequencing (NGS) strategy (LymphoTrack®), and compared the results with next-generation flow (NGF, EuroFlow). The use of different marrow pulls and the need of concentrating samples for NGS biased the applicability for MRD evaluation and favored NGF. Despite that, correlation between NGS and NGF was high (R = 0.905). The 3-year progression-free survival (PFS) rates by NGS and NGF were longer for undetectable vs. positive patients (NGS: 88.7% vs. 56.6%; NGF: 91.4% vs. 50%; p < 0.001 for both comparisons), which resulted in a 3-year overall survival (OS) advantage (NGS: 96.2% vs. 77.3%; NGF: 96.6% vs. 74.9%, p < 0.01 for both comparisons). In the Cox regression model, NGS and NGF negativity had similar results but favoring the latter in PFS (HR: 0.20, 95% CI: 0.09-0.45, p < 0.001) and OS (HR: 0.21, 95% CI: 0.06-0.75, p = 0.02). All these results reinforce the role of MRD detection by different strategies in patient prognosis and highlight the use of MRD as an endpoint for multiple myeloma treatment.
Mass spectrometry vs immunofixation for treatment monitoring in multiple myeloma
Dipòsit Digital de Documents de la UAB
- Puig, Noemí
- Contreras, María Teresa|||0000-0002-2264-7791
- Agulló, Cristina
- Martínez-López, Joaquín|||0000-0001-7908-0063
- Oriol, Albert|||0000-0001-6804-2221
- Blanchard, María Jesús
- Ríos-Tamayo, Rafael|||0000-0001-8193-1402
- Martín, Jesús
- Iñigo Rodríguez, María Belén
- Sureda, Anna|||0000-0002-1238-6970
- Hernández, Miguel Teodoro|||0000-0002-6576-7881
- de la Rubia, Javier|||0000-0002-8354-768X
- González-Calle, Verónica|||0000-0002-5493-6707
- Krsnik, Isabel|||0000-0002-2912-5072
- Cabañas Perianes, Valentín|||0000-0002-6248-2549
- Palomera, Luis|||0000-0003-0359-7191
- Moraleda, José María|||0000-0001-9080-1466
- Bargay, Joan|||0000-0002-6739-3916
- Cedena, María Teresa|||0000-0001-5851-3720
- Paiva, Bruno|||0000-0003-1977-3815
- Rosiñol, Laura|||0000-0002-2534-9239
- Bladé Creixenti, Juan|||0000-0002-4563-3405
- San-Miguel, J|||0000-0002-9183-4857
- Lahuerta, J. J|||0000-0002-3393-9570
- Mateos, M. V|||0000-0003-2390-1218
Monitoring of the monoclonal protein (M-protein) by electrophoresis and/or immunofixation (IFE) has long been used to assess treatment response in multiple myeloma (MM). However, with the use of highly effective therapies, the M-protein becomes frequently undetectable, and more sensitive methods had to be explored. We applied IFE and mass spectrometry (EXENT&FLC-MS) in serum samples from newly diagnosed MM patients enrolled in the PETHEMA/GEM2012MENOS65 obtained at baseline (n 5 223), and after induction (n 5 183), autologous stem cell transplantation (n 5 173), and consolidation (n 5 173). At baseline, the isotypes identified with both methods fully matched in 82.1% of samples; in the rest but 2 cases, EXENT&FLC-MS provided additional information to IFE with regards to the M-protein(s). Overall, the results of EXENT&FLC-MS and IFE were concordant in.80% of cases, being most discordances due to EXENT&FLC-MS but IFE cases. After consolidation, IFE was not able to discriminate 2 cohorts with different median progression-free survival (PFS), but EXENT&FLC-MS did so; furthermore, among IFE patients, EXENT&FLC-MS identified 2 groups with significantly different median PFS (P 5.0008). In conclusion, compared with IFE, EXENT&FLC-MS is more sensitive to detect the M-protein of patients with MM, both at baseline and during treatment, and provides a more accurate prediction of patients' outcome. This trial was registered at www.clinicaltrials.gov as #NCT01916252.
Next generation flow for minimally-invasive blood characterization of MGUS and multiple myeloma at diagnosis based on circulating tumor plasma cells (CTPC)
Zaguán. Repositorio Digital de la Universidad de Zaragoza
- Sanoja-Flores, L.
- Flores-Montero, J.
- Garces, J.J.
- Paiva, B.
- Puig, N.
- Garcia-Mateo, A.
- Garcia-Sanchez, O.
- Corral-Mateos, A.
- Burgos, L.
- Blanco, E.
- Hernandez-Martin, J.
- Pontes, R.
- Diez-Campelo, M.
- Millacoy, P.
- Rodriguez-Otero, P.
- Prosper, F.
- Merino, J.
- Vidriales, M.B.
- Garcia-Sanz, R.
- Romero, A.
- Palomera, L.
- Rios-Tamayo, R.
- Perez-Andres, M.
- Blanco, J.F.
- Gonzalez, M.
- van Dongen, J.J.M.
- Durie, B.
- Mateos, M.V.
- San-Miguel, J.
- Orfao, A.
Here, we investigated for the first time the frequency and number of circulating tumor plasma cells (CTPC) in peripheral blood (PB) of newly diagnosed patients with localized and systemic plasma cell neoplasms (PCN) using next-generation flow cytometry (NGF) and correlated our findings with the distinct diagnostic and prognostic categories of the disease. Overall, 508 samples from 264 newly diagnosed PCN patients, were studied. CTPC were detected in PB of all active multiple myeloma (MM; 100%), and smoldering MM (SMM) patients (100%), and in more than half (59%) monoclonal gammopathy of undetermined significance (MGUS) cases (p < 0.0001); in contrast, CTPC were present in a small fraction of solitary plasmacytoma patients (18%). Higher numbers of CTPC in PB were associated with higher levels of BM infiltration and more adverse prognostic features, together with shorter time to progression from MGUS to MM (p < 0.0001) and a shorter survival in MM patients with active disease requiring treatment (p <= 0.03). In summary, the presence of CTPC in PB as assessed by NGF at diagnosis, emerges as a hallmark of disseminated PCN, higher numbers of PB CTPC being strongly associated with a malignant disease behavior and a poorer outcome of both MGUS and MM.
Measurable Residual Disease by Next-Generation Flow Cytometry in Multiple Myeloma
Zaguán. Repositorio Digital de la Universidad de Zaragoza
- Paiva, B.
- Puig, N.
- Cedena, M.T.
- Rosiñol, L.
- Cordón, L.
- Vidriales, M.B.
- Burgos, L.
- Flores-Montero, J.
- Sanoja-Flores, L.
- Lopez-Anglada, L.
- Maldonado, R.
- de la Cruz, J.
- Gutierrez, N.C.
- Calasanz, M.J.
- Martin-Ramos, M.L.
- Garcia-Sanz, R.
- Martinez-Lopez, J.
- Oriol, A.
- Blanchard, M.J.
- Rios, R.
- Martin, J.
- Martinez-Martinez, R.
- Sureda, A.
- Hernandez, M.T.
- de la Rubia, J.
- Krsnik, I.
- Moraleda, J.M.
- Palomera, L.
- Bargay, J.
- Van Dongen, J.J.M.
- Orfao, A.
- Mateos, M.V.
- Blade, J.
- San-Miguel, J.F.
- Lahuerta, J.J.
PURPOSE: Assessing measurable residual disease (MRD) has become standard with many tumors, but the clinical meaning of MRD in multiple myeloma (MM) remains uncertain, particularly when assessed by next-generation flow (NGF) cytometry. Thus, we aimed to determine the applicability and sensitivity of the flow MRD-negative criterion defined by the International Myeloma Working Group (IMWG).
PATIENTS AND METHODS: In the PETHEMA/GEM2012MENOS65 trial, 458 patients with newly diagnosed MM had longitudinal assessment of MRD after six induction cycles with bortezomib, lenalidomide, and dexamethasone (VRD), autologous transplantation, and two consolidation courses with VRD. MRD was assessed in 1, 100 bone marrow samples from 397 patients; the 61 patients without MRD data discontinued treatment during induction and were considered MRD positive for intent-to-treat analysis. The median limit of detection achieved by NGF was 2.9 × 10-6. Patients received maintenance (lenalidomide ± ixazomib) according to the companion PETHEMA/GEM2014MAIN trial.
RESULTS: Overall, 205 (45%) of 458 patients had undetectable MRD after consolidation, and only 14 of them (7%) have experienced progression thus far; seven of these 14 displayed extraosseous plasmacytomas at diagnosis and/or relapse. Using time-dependent analysis, patients with undetectable MRD had an 82% reduction in the risk of progression or death (hazard ratio, 0.18; 95% CI, 0.11 to 0.30; P < .001) and an 88% reduction in the risk of death (hazard ratio, 0.12; 95% CI, 0.05 to 0.29; P < .001). Timing of undetectable MRD (after induction v intensification) had no impact on patient survival. Attaining undetectable MRD overcame poor prognostic features at diagnosis, including high-risk cytogenetics. By contrast, patients with Revised International Staging System III status and positive MRD had dismal progression-free and overall survivals (median, 14 and 17 months, respectively). Maintenance increased the rate of undetectable MRD by 17%.
CONCLUSION: The IMWG flow MRD-negative response criterion is highly applicable and sensitive to evaluate treatment efficacy in MM.
PATIENTS AND METHODS: In the PETHEMA/GEM2012MENOS65 trial, 458 patients with newly diagnosed MM had longitudinal assessment of MRD after six induction cycles with bortezomib, lenalidomide, and dexamethasone (VRD), autologous transplantation, and two consolidation courses with VRD. MRD was assessed in 1, 100 bone marrow samples from 397 patients; the 61 patients without MRD data discontinued treatment during induction and were considered MRD positive for intent-to-treat analysis. The median limit of detection achieved by NGF was 2.9 × 10-6. Patients received maintenance (lenalidomide ± ixazomib) according to the companion PETHEMA/GEM2014MAIN trial.
RESULTS: Overall, 205 (45%) of 458 patients had undetectable MRD after consolidation, and only 14 of them (7%) have experienced progression thus far; seven of these 14 displayed extraosseous plasmacytomas at diagnosis and/or relapse. Using time-dependent analysis, patients with undetectable MRD had an 82% reduction in the risk of progression or death (hazard ratio, 0.18; 95% CI, 0.11 to 0.30; P < .001) and an 88% reduction in the risk of death (hazard ratio, 0.12; 95% CI, 0.05 to 0.29; P < .001). Timing of undetectable MRD (after induction v intensification) had no impact on patient survival. Attaining undetectable MRD overcame poor prognostic features at diagnosis, including high-risk cytogenetics. By contrast, patients with Revised International Staging System III status and positive MRD had dismal progression-free and overall survivals (median, 14 and 17 months, respectively). Maintenance increased the rate of undetectable MRD by 17%.
CONCLUSION: The IMWG flow MRD-negative response criterion is highly applicable and sensitive to evaluate treatment efficacy in MM.
Comprehensive and systematic characterization of multi-functionalized cisplatin nano-conjugate: from the chemistry and proteomic biocompatibility to the animal model
Zaguán. Repositorio Digital de la Universidad de Zaragoza
- Hernández, Ángela-Patricia
- Micaelo, Ania
- Piñol, Rafael
- García-Vaquero, Marina L.
- Aramayona, José J.
- Criado, Julio J.
- Rodriguez, Emilio
- Sánchez-Gallego, José Ignacio
- Landeira-Viñuela, Alicia
- Juanes-Velasco, Pablo
- Díez, Paula
- Góngora, Rafael
- Jara-Acevedo, Ricardo
- Orfao, Alberto
- Miana-Mena, Javier
- Muñoz, María Jesús
- Villanueva, Sergio
- Millán, Ángel
- Fuentes, Manuel
Background
Nowadays, nanoparticles (NPs) have evolved as multifunctional systems combining different custom anchorages which opens a wide range of applications in biomedical research. Thus, their pharmacological involvements require more comprehensive analysis and novel nanodrugs should be characterized by both chemically and biological point of view. Within the wide variety of biocompatible nanosystems, iron oxide nanoparticles (IONPs) present mostly of the required features which make them suitable for multifunctional NPs with many biopharmaceutical applications.
Results
Cisplatin-IONPs and different functionalization stages have been broadly evaluated. The potential application of these nanodrugs in onco-therapies has been assessed by studying in vitro biocompatibility (interactions with environment) by proteomics characterization the determination of protein corona in different proximal fluids (human plasma, rabbit plasma and fetal bovine serum),. Moreover, protein labeling and LC–MS/MS analysis provided more than 4000 proteins de novo synthetized as consequence of the nanodrugs presence defending cell signaling in different tumor cell types (data available via ProteomeXchanges with identified PXD026615). Further in vivo studies have provided a more integrative view of the biopharmaceutical perspectives of IONPs.
Conclusions
Pharmacological proteomic profile different behavior between species and different affinity of protein coating layers (soft and hard corona). Also, intracellular signaling exposed differences between tumor cell lines studied. First approaches in animal model reveal the potential of theses NPs as drug delivery vehicles and confirm cisplatin compounds as strengthened antitumoral agents.
Nowadays, nanoparticles (NPs) have evolved as multifunctional systems combining different custom anchorages which opens a wide range of applications in biomedical research. Thus, their pharmacological involvements require more comprehensive analysis and novel nanodrugs should be characterized by both chemically and biological point of view. Within the wide variety of biocompatible nanosystems, iron oxide nanoparticles (IONPs) present mostly of the required features which make them suitable for multifunctional NPs with many biopharmaceutical applications.
Results
Cisplatin-IONPs and different functionalization stages have been broadly evaluated. The potential application of these nanodrugs in onco-therapies has been assessed by studying in vitro biocompatibility (interactions with environment) by proteomics characterization the determination of protein corona in different proximal fluids (human plasma, rabbit plasma and fetal bovine serum),. Moreover, protein labeling and LC–MS/MS analysis provided more than 4000 proteins de novo synthetized as consequence of the nanodrugs presence defending cell signaling in different tumor cell types (data available via ProteomeXchanges with identified PXD026615). Further in vivo studies have provided a more integrative view of the biopharmaceutical perspectives of IONPs.
Conclusions
Pharmacological proteomic profile different behavior between species and different affinity of protein coating layers (soft and hard corona). Also, intracellular signaling exposed differences between tumor cell lines studied. First approaches in animal model reveal the potential of theses NPs as drug delivery vehicles and confirm cisplatin compounds as strengthened antitumoral agents.
Enhancement of tumor cell immunogenicity and antitumor properties derived from platinum-conjugated iron nanoparticles
Zaguán. Repositorio Digital de la Universidad de Zaragoza
- Hernández, Ángela-Patricia
- Iglesias-Anciones, Laura
- Vaquero-González, José Javier
- Piñol, Rafael
- Criado, Julio J.
- Rodriguez, Emilio
- Juanes-Velasco, Pablo
- García-Vaquero, Marina L.
- Arias-Hidalgo, Carlota
- Orfao, Alberto
- Millán, Ángel
- Fuentes, Manuel
From chemistry design to clinical application, several approaches have been developed to overcome platinum drawbacks in antitumoral therapies. An in-depth understanding of intracellular signaling may hold the key to the relationship of both conventional drugs and nanoparticles. Within these strategies, first, nanotechnology has become an essential tool in oncotherapy, improving biopharmaceutical properties and providing new immunomodulatory profiles to conventional drugs mediated by activation of endoplasmic reticulum (ER) stress. Secondly, functional proteomics techniques based on microarrays have proven to be a successful method for high throughput screening of proteins and profiling of biomolecule mechanisms of action. Here, we conducted a systematic characterization of the antitumor profile of a platinum compound conjugated with iron oxide nanoparticles (IONPs). As a result of the nano-conjugation, cytotoxic and proteomics profiles revealed a significant improvement in the antitumor properties of the starting material, providing selectivity in certain tumor cell lines tested. Moreover, cell death patterns associated with immunogenic cell death (ICD) response have also been identified when ER signaling pathways have been triggered. The evaluation in several tumor cell lines and the analysis by functional proteomics techniques have shown novel perspectives on the design of new cisplatin-derived conjugates, the high value of IONPs as drug delivery systems and ICD as a rewarding approach for targeted oncotherapy and onco-immunotherapies.