Miembros del Grupo
Coordinador
Investigadores
Benat
Ariceta Ganuza
María Erendira
Calleja Cervantes
Cristina
Calviño Sampedro
Miren
Lasaga
Juan Pablo
Romero Riojas
Asier
Ullate Agote
Amaia
Vilas Zornoza
Colaboradores
Almudena
Aguilera Díaz
Marina
Ainciburu Fernandez
Ane
Amundarain Iraola
Leire
Garate Iturriagagoitia
Naroa
Gimenez Camino
Nahia
Gomez Echarte
Óscar
González Moreno
Angel
Martin Mallo
Estibaliz
Miranda Elizalde
Luis Vitores
Valcárcel García
Ana Cristina
Viñado Solanas
Líneas de Investigación
- Desarrollo de estrategias terapéuticas basadas en células CART
- Estudio del transcriptoma, epigenoma y metabolismo de las células plasmáticas tumorales del Mieloma múltiple
- Mecanismos de regulación génica en leucemia aguda y síndromes mielodisplásicos
- Papel de las alteraciones epigenéticas en la patogénesis y tratamiento de tumores hematológicos
- Papel del nicho hematopoyético en los tumores hematológicos
Palabras Clave
- Epigenómica
- Histonas
- Leucemia
- Metabolismo
- Metilación
- Mieloma Múltiple
- Nicho hematopoyético
- Síndrome mielodisplásico
Publicaciones Científicas desde 2018
-
Autores: Simoes Pinto, Cátia Patricia; Chillón, M. C.; Martínez-Cuadrón, D.; et al.Revista: BLOOD ADVANCESISSN: 2473-9529 Vol.7 N° 1 2023 págs. 167 - 173ResumenClonal evolution in acute myeloid leukemia (AML) originates long before diagnosis and is a dynamic process that may affect survival. However, it remains uninvestigated during routine diagnostic workups. We hypothesized that the mutational status of bone marrow dysplastic cells and leukemic blasts, analyzed at the onset of AML using integrated multidimensional flow cytometry (MFC) immunophenotyping and fluorescence-activated cell sorting (FACS) with next-generation sequencing (NGS), could reconstruct leukemogenesis. Dysplastic cells were detected by MFC in 285 of 348 (82%) newly diagnosed patients with AML. Presence of dysplasia according to MFC and World Health Organization criteria had no prognostic value in older adults. NGS of dysplastic cells and blasts isolated at diagnosis identified 3 evolutionary patterns: stable (n= 12 of 21), branching (n= 4 of 21), and clonal evolution (n= 5 of 21). In patients achieving complete response (CR), integrated MFC and FACS with NGS showed persistent measurable residual disease (MRD) in phenotypically normal cell types, as well as the acquisition of genetic traits associated with treatment resistance. Furthermore, whole-exome sequencing of dysplastic and leukemic cells at diagnosis and of MRD uncovered different clonal involvement in dysplastic myelo-erythropoiesis, leukemic transformation, and chemoresistance. Altogether, we showed that it is possible to reconstruct leukemogenesis in 80% of patients with newly diagnosed AML, using techniques other than single-cell multiomics.
-
Autores: Amundarain, A.; Pastor, F.; Prosper Cardoso, Felipe (Autor de correspondencia); et al.Revista: CANCERSISSN: 2072-6694 Vol.14 N° 21 2022 págs. 5471ResumenSimple Summary Multiple Myeloma (MM) remains incurable due to high relapse rates and fast development of drug resistances. Monoclonal antibodies (mAb) have revolutionized MM treatment, opening the door to chemotherapy-free yet curative treatments. Nevertheless, antibody-based therapies face several difficulties which could be overcome by nucleic acid aptamers. Aptamers are short oligonucleotide ligands that bind their targets with great affinity and specificity, and can be easily conjugated to different cargoes for their cell-specific delivery. This review summarizes the aptamers that have been tested in MM so far with promising results, and proposes various strategies for the further development of aptamer-based strategies against MM. Multiple Myeloma (MM) remains an incurable disease due to high relapse rates and fast development of drug resistances. The introduction of monoclonal antibodies (mAb) has caused a paradigm shift in MM treatment, paving the way for targeted approaches with increased efficacy and reduced toxicities. Nevertheless, antibody-based therapies face several difficulties such as high immunogenicity, high production costs and limited conjugation capacity, which we believe could be overcome by the introduction of nucleic acid aptamers. Similar to antibodies, aptamers can bind to their targets with great affinity and specificity. However, their chemical nature reduces their immunogenicity and production costs, while it enables their conjugation to a wide variety of cargoes for their use as delivery agents. In this review, we summarize several aptamers that have been tested against MM specific targets with promising results, establishing the rationale for the further development of aptamer-based strategies against MM. In this direction, we believe that the study of novel plasma cell surface markers, the development of intracellular aptamers and further research on aptamers as building blocks for complex nanomedicines will lead to the generation of next-generation targeted approaches that will undoubtedly contribute to improve the management and life quality of MM patients.
-
Autores: Huerga Domínguez, Sofia; Villar Fernández, Sara; Prosper Cardoso, Felipe; et al.Revista: CANCERSISSN: 2072-6694 Vol.14 N° 19 2022 págs. 4756ResumenSimple Summary Acute myeloid leukemia is the most common type of acute leukemia in adults. It is associated with poor outcomes, especially in older patients. Treatments based exclusively on chemotherapy do not achieve high overall survival rates, or in any case, only in a small group of patients. The objective of this review is to discuss the treatment of acute myeloid leukemia from newly approved therapeutic drugs to newer strategies. In first line, the combination of new targeted therapies with standard chemotherapy achieves better outcomes in "fit" patients. For "unfit" patients the combination of different targeted therapies provides them with better overall survival rates, with limited toxicity. As for refractory and relapsed acute myeloid leukemia, development of immunotherapy or new targeted therapies brings new hope. Acute myeloid leukemia is a heterogeneous disease defined by a large spectrum of genetic aberrations that are potential therapeutic targets. New targeted therapies have changed the landscape for a disease with poor outcomes. They are more effective than standard chemotherapy with a good safety profile. For "fit patients" in first-line, the combination of gemtuzumab ozogamicin or midostaurin with intensive chemotherapy or Vyxeos is now considered the "standard of care" for selected patients. On the other hand, for "unfit patients", azacitidine-venetoclax has been consolidated as a frontline treatment, while other combinations with magrolimab or ivosidenib are in development. Nevertheless, global survival results, especially in relapsed or refractory patients, remain unfavorable. New immunotherapies or targeted therapies, such as Menin inhibitors or sabatolimab, represent an opportunity in this situation. Future directions will probably come from combinations of different targeted therapies ("triplets") and maintenance strategies guided by measurable residual disease.
-
Autores: Guedan, S.; Luu, M.; Ammar, D.; et al.Título: Time 2EVOLVE: predicting efficacy of engineered T-cells - how far is the bench from the bedside?Revista: JOURNAL FOR IMMUNOTHERAPY OF CANCERISSN: 2051-1426 Vol.10 N° 5 2022 págs. e003487ResumenImmunotherapy with gene engineered CAR and TCR transgenic T-cells is a transformative treatment in cancer medicine. There is a rich pipeline with target antigens and sophisticated technologies that will enable establishing this novel treatment not only in rare hematological malignancies, but also in common solid tumors. The T2EVOLVE consortium is a public private partnership directed at accelerating the preclinical development of and increasing access to engineered T-cell immunotherapies for cancer patients. A key ambition in T2EVOLVE is to assess the currently available preclinical models for evaluating safety and efficacy of engineered T cell therapy and developing new models and test parameters with higher predictive value for clinical safety and efficacy in order to improve and accelerate the selection of lead T-cell products for clinical translation. Here, we review existing and emerging preclinical models that permit assessing CAR and TCR signaling and antigen binding, the access and function of engineered T-cells to primary and metastatic tumor ligands, as well as the impact of endogenous factors such as the host immune system and microbiome. Collectively, this review article presents a perspective on an accelerated translational development path that is based on innovative standardized preclinical test systems for CAR and TCR transgenic T-cell products.
-
Autores: McLornan, D. P. (Autor de correspondencia); Gras, L.; Martin, I.; et al.Revista: BRITISH JOURNAL OF HAEMATOLOGYISSN: 0007-1048 Vol.198 N° 1 2022 págs. 209 - 213
-
Autores: Amundarain, A.; Valcárcel, L. V.; Ordóñez Ciriza, Raquel; et al.Revista: AMERICAN JOURNAL OF HEMATOLOGYISSN: 0361-8609 Vol.97 N° 3 2022 págs. E113 - E117
-
Autores: Vilarrasa-Blasi, R. (Autor de correspondencia); Verdaguer-Dot, N.; Belver, L.; et al.Título: Insights into the mechanisms underlying aberrant SOX11 oncogene expression in mantle cell lymphomaRevista: LEUKEMIAISSN: 0887-6924 Vol.36 N° 2 2022 págs. 583 - 587
-
Autores: Ghobrial, I. M. (Autor de correspondencia); Rodríguez Otero, Paula; Koh, Y.; et al.Revista: CLINICAL LYMPHOMA MYELOMA AND LEUKEMIAISSN: 2152-2650 Vol.22 N° 7 2022 págs. 545
-
Autores: Peng, J.; SERRANO SANZ, Guillermo; Traniello, I. M.; et al.Revista: COMMUNICATIONS BIOLOGYISSN: 2399-3642 Vol.5 N° 1 2022 págs. 351ResumenSingle-cell RNA-Sequencing has the potential to provide deep biological insights by revealing complex regulatory interactions across diverse cell phenotypes at single-cell resolution. However, current single-cell gene regulatory network inference methods produce a single regulatory network per input dataset, limiting their capability to uncover complex regulatory relationships across related cell phenotypes. We present SimiC, a single-cell gene regulatory inference framework that overcomes this limitation by jointly inferring distinct, but related, gene regulatory dynamics per phenotype. We show that SimiC uncovers key regulatory dynamics missed by previously proposed methods across a range of systems, both model and non-model alike. In particular, SimiC was able to uncover CAR T cell dynamics after tumor recognition and key regulatory patterns on a regenerating liver, and was able to implicate glial cells in the generation of distinct behavioral states in honeybees. SimiC hence establishes a new approach to quantitating regulatory architectures between distinct cellular phenotypes, with far-reaching implications for systems biology.
-
Autores: DePasquale, E. A. K.; Ssozi, D.; Ainciburu Fernandez, Marina; et al.Revista: FRONTIERS IN IMMUNOLOGYISSN: 1664-3224 Vol.13 2022 págs. 809414ResumenThe immune system represents a major barrier to cancer progression, driving the evolution of immunoregulatory interactions between malignant cells and T-cells in the tumor environment. Blastic plasmacytoid dendritic cell neoplasm (BPDCN), a rare acute leukemia with plasmacytoid dendritic cell (pDC) differentiation, provides a unique opportunity to study these interactions. pDCs are key producers of interferon alpha (IFNA) that play an important role in T-cell activation at the interface between the innate and adaptive immune system. To assess how uncontrolled proliferation of malignant BPDCN cells affects the tumor environment, we catalog immune cell heterogeneity in the bone marrow (BM) of five healthy controls and five BPDCN patients by analyzing 52,803 single-cell transcriptomes, including 18,779 T-cells. We test computational techniques for robust cell type classification and find that T-cells in BPDCN patients consistently upregulate interferon alpha (IFNA) response and downregulate tumor necrosis factor alpha (TNFA) pathways. Integrating transcriptional data with T-cell receptor sequencing via shared barcodes reveals significant T-cell exhaustion in BPDCN that is positively correlated with T-cell clonotype expansion. By highlighting new mechanisms of T-cell exhaustion and immune evasion in BPDCN, our results demonstrate the value of single-cell multiomics to understand immune cell interactions in the tumor environment.
-
Autores: Dorr, D.; Obermayer, B.; Weiner, J. M.; et al.Revista: SCIENCE IMMUNOLOGYISSN: 2470-9468 Vol.7 N° 75 2022 págs. eabj0140ResumenPulmonary alveolar proteinosis (PAP) is a syndrome characterized by accumulation of surfactant lipoproteins within the lung alveoli. Alveolar macrophages (AMs) are crucial for surfactant clearance, and their differentiation depends on colony-stimulating factor 2 (CSF2), which regulates the establishment of an AM-characteristic gene regulatory network. Here, we report that the transcription factor CCAAT/enhancer binding protein ss (C/EBP ss) is essential for the development of the AM identity, as demonstrated by transcriptome and chromatin accessibility analysis. Furthermore, C/EBP ss-deficient AMs showed severe defects in proliferation, phagocytosis, and lipid metabolism, collectively resulting in a PAP-like syndrome. Mechanistically, the long C/EBP ss protein variants LAP* and LAP together with CSF2 signaling induced the expression of Pparg isoform 2 but not Pparg isoform 1, a molecular regulatory mechanism that was also observed in other CSF2-primed macrophages. These results uncover C/EBP ss as a key regulator of AM cell fate and shed light on the molecular networks controlling lipid metabolism in macrophages.
-
Autores: Viñado, A. C.; Calvo Arnedo, Isabel; Cenzano Armendariz, Itziar; et al.Revista: LEUKEMIAISSN: 0887-6924 Vol.36 N° 8 2022 págs. 1969 - 1979ResumenEradicating leukemia requires a deep understanding of the interaction between leukemic cells and their protective microenvironment. The CXCL12/CXCR4 axis has been postulated as a critical pathway dictating leukemia stem cell (LSC) chemoresistance in AML due to its role in controlling cellular egress from the marrow. Nevertheless, the cellular source of CXCL12 in the acute myeloid leukemia (AML) microenvironment and the mechanism by which CXCL12 exerts its protective role in vivo remain unresolved. Here, we show that CXCL12 produced by Prx1+ mesenchymal cells but not by mature osteolineage cells provide the necessary cues for the maintenance of LSCs in the marrow of an MLL::AF9-induced AML model. Prx1+ cells promote survival of LSCs by modulating energy metabolism and the REDOX balance in LSCs. Deletion of Cxcl12 leads to the accumulation of reactive oxygen species and DNA damage in LSCs, impairing their ability to perpetuate leukemia in transplantation experiments, a defect that can be attenuated by antioxidant therapy. Importantly, our data suggest that this phenomenon appears to be conserved in human patients. Hence, we have identified Prx1+ mesenchymal cells as an integral part of the complex niche-AML metabolic intertwining, pointing towards CXCL12/CXCR4 as a target to eradicate parenchymal LSCs in AML.
-
Autores: Ainciburu Fernandez, Marina; Berastegui Zufiaurre, Nerea; Romero Riojas, Juan Pablo; et al.Revista: NATURE COMMUNICATIONSISSN: 2041-1723 Vol.13 N° 1 2022 págs. 7619ResumenMyelodysplastic syndromes (MDS) are hematopoietic stem cell (HSC) malignancies characterized by ineffective hematopoiesis, with increased incidence in older individuals. Here we analyze the transcriptome of human HSCs purified from young and older healthy adults, as well as MDS patients, identifying transcriptional alterations following different patterns of expression. While aging-associated lesions seem to predispose HSCs to myeloid transformation, disease-specific alterations may trigger MDS development. Among MDS-specific lesions, we detect the upregulation of the transcription factor DNA Damage Inducible Transcript 3 (DDIT3). Overexpression of DDIT3 in human healthy HSCs induces an MDS-like transcriptional state, and dyserythropoiesis, an effect associated with a failure in the activation of transcriptional programs required for normal erythroid differentiation. Moreover, DDIT3 knockdown in CD34+ cells from MDS patients with anemia is able to restore erythropoiesis. These results identify DDIT3 as a driver of dyserythropoiesis, and a potential therapeutic target to restore the inefficient erythroid differentiation characterizing MDS patients. © 2022, The Author(s).
-
Autores: Moreira-Silva, F.; Outeiro-Pinho, G.; Lobo, J.; et al.Revista: BIOMEDICINE AND PHARMACOTHERAPYISSN: 0753-3322 Vol.150 2022 págs. 113031ResumenCastration-resistant prostate cancer (CRPC) is an incurable form of prostate cancer (PCa), with DNMT1 and G9a being reported as overexpressed, rendering them highly attractive targets for precision medicine. CM-272 is a dual inhibitor of both methyltransferases' activity. Herein, we assessed the response of different PCa cell lines to CM-272, in both 2D and 3D models, and explored the molecular mechanisms underlying CM-272 inhibitory effects.CRPC tissues displayed significantly higher DNMT1, G9a and H3K9me2 expression than localized PCa. In vitro, CM-272 caused a significant decrease in PCa cell viability and proliferation alongside with increased apoptotic levels. We disclose that, under the evaluated dose, CM-272 led to G9a activity inhibition, while not significantly affecting DNMT1 activity. Upon G9a knockdown, DU145 and PC3 showed decreased cell viability. Remarkably, DU145 cells treated with CM-272 or with G9a knockdown displayed no differences in viability, suggesting a SET dependent mechanism. Contrarily, PC3 cell viability impact was higher in G9a knockdown, compared with CM 272 treatment, suggesting an additional G9a function. Moreover, DU145 cells overexpressing catalytically functional G9a disclosed higher resistance to CM-272 treatment, reinforcing that the drug mechanism of action is dependent on G9a catalytic function.Importantly, we successfully assembled spheroids from several prostate cell lines. Our results showed that CM 272 retained its anti-tumoral effects in 3D PCa models, leading to a clear reduction in cancer cell survival. We concluded that inhibition of G9a methyltransferase activity by CM-272 has anti-tumor effect in PCa cells, holding therapeutic potential against CRPC.
-
Autores: Gimeno, M.; San José Enériz, Edurne; Rubio Díaz-Cordoves, Ángel; et al.Revista: CANCERSISSN: 2072-6694 Vol.14 N° 13 2022 págs. 3251ResumenSimple Summary This work shows that the predictions of lethal dependencies (LEDs) between genes can be dramatically improved by incorporating the "HUb effect in Genetic Essentiality" (HUGE) of gene alterations. In three genome-wide loss-of-function screens-Project Score, CERES score and DEMETER score-LEDs are identified with 75 times larger statistical power than using state-of-the-art methods. In AML, we identified LEDs not recalled by previous pipelines, including FLT3-mutant genotypes sensitive to FLT3 inhibitors. Interestingly, in-vitro validations confirm lethal de-pendencies of either NRAS or PTPN11 depending on the NRAS mutational status. Recent functional genomic screens-such as CRISPR-Cas9 or RNAi screening-have fostered a new wave of targeted treatments based on the concept of synthetic lethality. These approaches identified LEthal Dependencies (LEDs) by estimating the effect of genetic events on cell viability. The multiple-hypothesis problem is related to a large number of gene knockouts limiting the statistical power of these studies. Here, we show that predictions of LEDs from functional screens can be dramatically improved by incorporating the "HUb effect in Genetic Essentiality" (HUGE) of gene alterations. We analyze three recent genome-wide loss-of-function screens-Project Score, CERES score and DEMETER score-identifying LEDs with 75 times larger statistical power than using state-of-the-art methods. Using acute myeloid leukemia, breast cancer, lung adenocarcinoma and colon adenocarcinoma as disease models, we validate that our predictions are enriched in a recent harmonized knowledge base of clinical interpretations of somatic genomic variants in cancer (AUROC > 0.87). Our approach is effective even in tumors with large genetic heterogeneity such as acute myeloid leukemia, where we identified LEDs not recalled by previous pipelines, including FLT3-mutant genotypes sensitive to FLT3 inhibitors. Interestingly, in-vitro validations confirm lethal dependencies of either NRAS or PTPN11 depending on the NRAS mutational status. HUGE will hopefully help discover novel genetic dependencies amenable for precision-targeted therapies in cancer. All the graphs showing lethal dependencies for the 19 tumor types analyzed can be visualized in an interactive tool.
-
Autores: Armes, H.; Bewicke-Copley, F.; Rio-Machin, A.; et al.Revista: BRITISH JOURNAL OF HAEMATOLOGYISSN: 0007-1048 Vol.199 N° 5 2022 págs. 754 - 764ResumenDespite the inclusion of inherited myeloid malignancies as a separate entity in the World Health Organization Classification, many established predisposing loci continue to lack functional characterization. While germline mutations in the DNA repair factor ERCC excision repair 6 like 2 (ERCC6L2) give rise to bone marrow failure and acute myeloid leukaemia, their consequences on normal haematopoiesis remain unclear. To functionally characterise the dual impact of germline ERCC6L2 loss on human primary haematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs), we challenged ERCC6L2-silenced and patient-derived cells ex vivo. Here, we show for the first time that ERCC6L2-deficiency in HSPCs significantly impedes their clonogenic potential and leads to delayed erythroid differentiation. This observation was confirmed by CIBERSORTx RNA-sequencing deconvolution performed on ERCC6L2-silenced erythroid-committed cells, which demonstrated higher proportions of polychromatic erythroblasts and reduced orthochromatic erythroblasts versus controls. In parallel, we demonstrate that the consequences of ERCC6L2-deficiency are not limited to HSPCs, as we observe a striking phenotype in patient-derived and ERCC6L2-silenced MSCs, which exhibit enhanced osteogenesis and suppressed adipogenesis. Altogether, our study introduces a valuable surrogate model to study the impact of inherited myeloid mutations and highlights the importance of accounting for the influence of germline mutations in HSPCs and their microenvironment.
-
Autores: Garreta, E.; Prado, P.; Stanifer, M. L.; et al.Revista: CELL METABOLISMISSN: 1550-4131 Vol.34 N° 6 2022 págs. 857 - 873.e9ResumenIt is not well understood why diabetic individuals are more prone to develop severe COVID-19. To this, we here established a human kidney organoid model promoting early hallmarks of diabetic kidney disease development. Upon SARS-CoV-2 infection, diabetic-like kidney organoids exhibited higher viral loads compared with their control counterparts. Genetic deletion of the angiotensin-converting enzyme 2 (ACE2) in kidney organoids under control or diabetic-like conditions prevented viral detection. Moreover, cells isolated from kidney biopsies from diabetic patients exhibited altered mitochondrial respiration and enhanced glycolysis, resulting in higher SARS-CoV-2 infections compared with non-diabetic cells. Conversely, the exposure of patient cells to dichloroacetate (DCA), an inhibitor of aerobic glycolysis, resulted in reduced SARS-CoV-2 infections. Our results provide insights into the identification of diabetic-induced metabolic programming in the kidney as a critical event increasing SARS-CoV-2 infection susceptibility, opening the door to the identification of new interventions in COVID-19 pathogenesis targeting energy metabolism.
-
Autores: Colyn, L.; Álvarez-Sola, G.; Latasa Sada, María Ujué; et al.Revista: JOURNAL OF EXPERIMENTAL AND CLINICAL CANCER RESEARCHISSN: 1756-9966 Vol.41 N° 1 2022 págs. 183ResumenBackground Cholangiocarcinoma (CCA) is still a deadly tumour. Histological and molecular aspects of thioacetamide (TAA)-induced intrahepatic CCA (iCCA) in rats mimic those of human iCCA. Carcinogenic changes and therapeutic vulnerabilities in CCA may be captured by molecular investigations in bile, where we performed bile proteomic and metabolomic analyses that help discovery yet unknown pathways relevant to human iCCA. Methods Cholangiocarcinogenesis was induced in rats (TAA) and mice (Jnk(Delta hepa) + CCl4 + DEN model). We performed proteomic and metabolomic analyses in bile from control and CCA-bearing rats. Differential expression was validated in rat and human CCAs. Mechanisms were addressed in human CCA cells, including Huh28-KRAS(G12D) cells. Cell signaling, growth, gene regulation and [U-C-13]-D-glucose-serine fluxomics analyses were performed. In vivo studies were performed in the clinically-relevant iCCA mouse model. Results Pathways related to inflammation, oxidative stress and glucose metabolism were identified by proteomic analysis. Oxidative stress and high amounts of the oncogenesis-supporting amino acids serine and glycine were discovered by metabolomic studies. Most relevant hits were confirmed in rat and human CCAs (TCGA). Activation of interleukin-6 (IL6) and epidermal growth factor receptor (EGFR) pathways, and key genes in cancer-related glucose metabolic reprogramming, were validated in TAA-CCAs. In TAA-CCAs, G9a, an epigenetic pro-tumorigenic writer, was also increased. We show that EGFR signaling and mutant KRAS(G12D) can both activate IL6 production in CCA cells. Furthermore, phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in serine-glycine pathway, was upregulated in human iCCA correlating with G9a expression. In a G9a activity-dependent manner, KRAS(G12D) promoted PHGDH expression, glucose flow towards serine synthesis, and increased CCA cell viability. KRAS(G12D) CAA cells were more sensitive to PHGDH and G9a inhibition than controls. In mouse iCCA, G9a pharmacological targeting reduced PHGDH expression. Conclusions In CCA, we identified new pro-tumorigenic mechanisms: Activation of EGFR signaling or KRAS mutation drives IL6 expression in tumour cells; Glucose metabolism reprogramming in iCCA includes activation of the serine-glycine pathway; Mutant KRAS drives PHGDH expression in a G9a-dependent manner; PHGDH and G9a emerge as therapeutic targets in iCCA.
-
Autores: Poza-Viejo, L.; Paya-Milans, M.; San Martín Úriz, Patxi; et al.Título: Conserved and distinct roles of H3K27me3 demethylases regulating flowering time in Brassica rapaRevista: PLANT CELL AND ENVIRONMENTISSN: 0140-7791 Vol.45 N° 5 2022 págs. 1428 - 1441ResumenEpigenetic regulation is necessary for optimal organism development and preservation of gene expression profiles in the cell. In plants, the trimethylation of histone H3 lysine 27 (H3K27me3) is a silencing epigenetic mark relevant for developmental transitions like flowering. The floral transition is a key agronomic trait; however, the epigenetic mechanisms of flowering time regulation in crops remain poorly understood. Here we study the Jumonji H3K27me3 demethylases BraA.REF6 and BraA.ELF6 in Brassica rapa. Phenotypic characterization of novel mutant lines and genome-wide H3K27me3 chromatin immunoprecipitation and transcriptomic analyses indicated that BraA.REF6 plays a greater role than BraA.ELF6 in fine-tuning H3K27me3 levels. In addition, we found that braA.elf6 mutants were early flowering due to high H3K27me3 levels at B. rapa homologs of the floral repressor FLC. Unlike mutations in Arabidopsis thaliana, braA.ref6 mutants were late flowering without altering the expression of B. rapa FLC genes. Remarkably, we found that BraA.REF6 regulated a number of gibberellic acid (GA) biosynthetic genes, including a homolog of GA1, and that GA-treatment complemented the late flowering mutant phenotype. This study increases our understanding of the epigenetic regulation of flowering time in B. rapa, highlighting conserved and distinct regulatory mechanisms between model and crop species.
-
Autores: Rodríguez Márquez, Paula; Calleja Cervantes, María Erendira; SERRANO SANZ, Guillermo; et al.Título: CAR density influences antitumoral efficacy of BCMA CAR T cells and correlates with clinical outcomeRevista: SCIENCE ADVANCESISSN: 2375-2548 Vol.8 N° 39 2022 págs. eabo0514ResumenIdentification of new markers associated with long-term efficacy in patients treated with CAR T cells is a current medical need, particularly in diseases such as multiple myeloma. In this study, we address the impact of CAR density on the functionality of BCMA CAR T cells. Functional and transcriptional studies demonstrate that CAR T cells with high expression of the CAR construct show an increased tonic signaling with up-regulation of exhaustion markers and increased in vitro cytotoxicity but a decrease in in vivo BM infiltration. Characterization of gene regulatory networks using scRNA-seq identified regulons associated to activation and exhaustion up-regulated in CARHigh T cells, providing mechanistic insights behind differential functionality of these cells. Last, we demonstrate that patients treated with CAR T cell products enriched in CARHigh T cells show a significantly worse clinical response in several hematological malignancies. In summary, our work demonstrates that CAR density plays an important role in CAR T activity with notable impact on clinical response.
-
Autores: Ye, J. ; Calvo Arnedo, Isabel; Cenzano Armendariz, Itziar; et al.Revista: ISCIENCEISSN: 2589-0042 Vol.25 N° 5 2022 págs. 104225ResumenUnderstanding the regulation of normal and malignant human hematopoiesis requires comprehensive cell atlas of the hematopoietic stem cell (HSC) regulatory microenvironment. Here, we develop a tailored bioinformatic pipeline to integrate public and proprietary single-cell RNA sequencing (scRNA-seq) datasets. As a result, we robustly identify for the first time 14 intermediate cell states and 11 stages of differentiation in the endothelial and mesenchymal BM compartments, respectively. Our data provide the most comprehensive description to date of the murine HSC-regulatory microenvironment and suggest a higher level of specialization of the cellular circuits than previously anticipated. Furthermore, this deep characterization allows inferring conserved features in human, suggesting that the layers of microenvironmental regulation of hematopoiesis may also be shared between species. Our resource and methodology is a stepping-stone toward a comprehensive cell atlas of the BM microenvironment.
-
Autores: Martínez Turrillas, Rebeca; Martin Mallo, Angel; Rodríguez Díaz, Saray; et al.Revista: MOLECULAR THERAPY. METHODS & CLINICAL DEVELOPMENTISSN: 2329-0501 Vol.25 2022 págs. 137 - 146ResumenGenome-editing strategies, especially CRISPR-Cas9 systems, have substantially increased the efficiency of innovative therapeutic approaches for monogenic diseases such as primary hyperoxalurias (PHs). We have previously demonstrated that inhibition of glycolate oxidase using CRISPR-Cas9 systems represents a promising therapeutic option for PH type I (PH1). Here, we extended our work evaluating the efficacy of liver-specific inhibition of lactate dehydrogenase (LDH), a key enzyme responsible for converting glyoxylate to oxalate; this strategy would not be limited to PH1, being applicable to other PH subtypes. In this work, we demonstrate a liver-specific inhibition of LDH that resulted in a drastic reduction of LDH levels in the liver of PH1 and PH3 mice after a single-dose delivery of AAV8 vectors expressing the CRISPR-Cas9 system, resulting in reduced urine oxalate levels and kidney damage without signs of toxicity. Deep sequencing analysis revealed that this approach was safe and specific, with no off-targets detected in the liver of treated animals and no on-target/off-tissue events. Altogether, our data provide evidence that in vivo genome editing using CRISPR-Cas9 systems would represent a valuable tool for improved therapeutic approaches for PH.
-
Autores: Ainciburu Fernandez, Marina; Morgan, D. M.; De Pasquale, E. A. K.; et al.Revista: BIOINFORMATICSISSN: 1367-4803 Vol.38 N° 14 2022 págs. 3645 - 3647ResumenDiversity of the T-cell receptor (TCR) repertoire is central to adaptive immunity. The TCR is composed of alpha and beta chains, encoded by the TRA and TRB genes, of which the variable regions determine antigen specificity. To generate novel biological insights into the complex functioning of immune cells, combined capture of variable regions and single-cell transcriptomes provides a compelling approach. Recent developments enable the enrichment of TRA and TRB variable regions from widely used technologies for 3'-based single-cell RNA-sequencing (scRNA-seq). However, a comprehensive computational pipeline to process TCR-enriched data from 3' scRNA-seq is not available. Here, we present an analysis pipeline to process TCR variable regions enriched from 3' scRNA-seq cDNA. The tool reports TRA and TRB nucleotide and amino acid sequences linked to cell barcodes, enabling the reconstruction of T-cell clonotypes with associated transcriptomes. We demonstrate the software using peripheral blood mononuclear cells from a healthy donor and detect TCR sequences in a high proportion of single T cells. Detection of TCR sequences is low in non-T-cell populations, demonstrating specificity. Finally, we show that TCR clones are larger in CD8 Memory T cells than in other T-cell types, indicating an association between T-cell clonotypes and differentiation states.
-
Autores: Ayala, R.; Carreno-Tarragona, G.; Barragan, E.; et al.Revista: CANCERSISSN: 2072-6694 Vol.14 N° 23 2022 págs. 5799ResumenSimple Summary The prognostic impact of FLT3-ITD allele ratio (AR) is a matter of controversy. We analyzed 2901 AML patients with long-term follow-up treated with PETHEMA protocols in the pre-FLT3 inhibitors era, with 579 of them harboring the FLT3-ITD mutation. We found that FLT3-ITD AR > 0.5 was associated with lower complete remission and rate and overall survival, while AR > 0.8 was associated with lower RFS. An AR of 0.44 was the best cutoff for OS and 0.8 for RFS. Overall, allo- and auto-hematopoietic stem cell transplant (HSCT) in first CR offered similar OS in patients with AR < 0.44, while allo-HSCT improved OS for those with higher AR. However, allo-HSCT resulted in better OS and RFS as compared to auto-HSCT in NPM1/FLT3-ITD-mutated AML regardless of pre-established AR cutoff (<= 0.5 vs. >0.5), supporting the use of other risk stratification tools, such as NPM1 MRD monitoring, in this setting. FLT3-ITD results in a poor prognosis in terms of overall survival (OS) and relapse-free survival (RFS) in acute myeloid leukemia (AML). However, the prognostic usefulness of the allelic ratio (AR) to select post-remission therapy remains controversial. Our study focuses on the prognostic impact of FLT3-ITD and its ratio in a series of 2901 adult patients treated intensively in the pre-FLT3 inhibitor era and reported in the PETHEMA registry. A total of 579 of these patients (20%) harbored FLT3-ITD mutations. In multivariate analyses, patients with an FLT3-ITD allele ratio (AR) of >0.5 showed a lower complete remission (CR rate) and OS (HR 1.47, p = 0.009), while AR > 0.8 was associated with poorer RFS (HR 2.1; p < 0.001). Among NPM1/FLT3-ITD-mutated patients, median OS gradually decreased according to FLT3-ITD status and ratio (34.3 months FLT3-ITD-negative, 25.3 months up to 0.25, 14.5 months up to 0.5, and 10 months >= 0.5, p < 0.001). Post-remission allogeneic transplant (allo-HSCT) resulted in better OS and RFS as compared to auto-HSCT in NPM1/FLT3-ITD-mutated AML regardless of pre-established AR cutoff (<= 0.5 vs. >0.5). Using the maximally selected log-rank statistics, we established an optimal cutoff of FLT3-ITD AR of 0.44 for OS, and 0.8 for RFS. We analyzed the OS and RFS according to FLT3-ITD status in all patients, and we found that the group of FLT3-ITD-positive patients with AR < 0.44 had similar 5-year OS after allo-HSCT or auto-HSCT (52% and 41%, respectively, p = 0.86), but worse RFS after auto-HSCT (p = 0.01). Among patients with FLT3-ITD AR > 0.44, allo-HSCT was superior to auto-HSCT in terms of OS and RFS. This study provides more evidence for a better characterization of patients with AML harboring FLT3-ITD mutations.
-
Autores: Saumell, S.; Fernández-Serrano, M.; Mesa, A.; et al.Revista: LEUKEMIA AND LYMPHOMAISSN: 1042-8194 Vol.63 N° 5 2022 págs. 1227 - 1235ResumenMicromegakaryocytes (microMKs) are considered a myelodysplastic feature of myeloid neoplasms in adults, with an adverse prognosis connotation. However, this notion in MDS has not been well proved. In our cohort of 287 MDS, patients with microMKs showed lower overall survival (OS) (HR, 2.12; 95% CI, 1.47-3.06; p = 0.000036) and higher risk of acute myeloid leukemia (AML) evolution (HR, 4.8; 95% CI, 2.9-11.01; p = 0.00021). Results were validated with an independent cohort. In multivariate analysis, the presence of microMKs maintained its independent association with OS (HR, 1.54, 95% CI, 1.13-2.1, p = 0.0059) and AML transformation (HR, 2.28, 95% CI, 1.2-4.4, p = 0.014). Moreover, by adding 1 point to the IPSS-R score in patients with microMKs, we improved the IPSS-R accuracy. Interestingly, adding that 1-point, 29% of intermediate IPSS-R risk group patients were upgraded to the high-risk group. In summary, we confirmed that the presence of microMKs implies worse outcomes in MDS and suggested a modification improving IPSS-R.
-
Autores: Palacios, M.; Giménez, Y.; Peral, J.; et al.Revista: HUMAN GENE THERAPYISSN: 1043-0342 Vol.33 N° 23-24 2022 págs. A154 - A154
-
Autores: Díez, B.; Calviño Sampedro, Cristina; Fernández-García, M.; et al.Revista: HUMAN GENE THERAPYISSN: 1043-0342 Vol.33 N° 23-24 2022 págs. A126 - A127
-
Autores: Redondo, S.; García-Cadenas, I.; Cuesta, M.; et al.Revista: BONE MARROW TRANSPLANTATIONISSN: 0268-3369 Vol.57 N° SUPPL 1 2022 págs. 276 - 276
-
Autores: Broijl, A.; Van De Donk, N. W. C. J.; Bosch, F.; et al.Revista: JOURNAL OF CLINICAL ONCOLOGYISSN: 0732-183X Vol.40 N° 16 2022
-
Autores: Zapata-Linares, N.; Toillon, I.; Wanherdrick, K.; et al.Revista: OSTEOARTHRITIS AND CARTILAGEISSN: 1063-4584 Vol.30 N° Supl. 1 2022 págs. S303
-
Autores: Pérez-Martínez, A.; Belendez-Bieler, C.; Molina-Angulo, B.; et al.Revista: BONE MARROW TRANSPLANTATIONISSN: 0268-3369 Vol.57 N° SUPPL 1 2022 págs. 372 - 372
-
Autores: Carrasco Leon, Arantxa; Amundarain, A.; Gomez Echarte, Nahia; et al.Revista: CANCERSISSN: 2072-6694 Vol.13 N° 8 2021 págs. 1976ResumenSimple Summary Multiple myeloma (MM), the second most common hematological neoplasm, is still considered an incurable disease. Long non-coding RNAs (lncRNAs), genes that do not encode proteins, participate in numerous biological processes, but their deregulation, like that of coding genes, can contribute to carcinogenesis. Increasing evidence points to the relevant role of lncRNAs in the development of human tumors, such that they emerge as attractive biomarkers and therapeutic targets for cancer treatment, including MM. Here we review the oncogenic or tumor-suppressor functions of lncRNAs in MM and provide an overview of novel therapeutic approaches based on lncRNAs that will help to improve the management of these patients. MM is a hematological neoplasm that is still considered an incurable disease. Besides established genetic alterations, recent studies have shown that MM pathogenesis is also characterized by epigenetic aberrations, such as the gain of de novo active chromatin marks in promoter and enhancer regions and extensive DNA hypomethylation of intergenic regions, highlighting the relevance of these non-coding genomic regions. A recent study described how long non-coding RNAs (lncRNAs) correspond to 82% of the MM transcriptome and an increasing number of studies have demonstrated the importance of deregulation of lncRNAs in MM. In this review we focus on the deregulated lncRNAs in MM, including their biological or functional mechanisms, their role as biomarkers to improve the prognosis and monitoring of MM patients, and their participation in drug resistance. Furthermore, we also discuss the evidence supporting the role of lncRNAs as therapeutic targets through different novel RNA-based strategies.
-
Autores: Palacios-Berraquero, M. L.; Alfonso Piérola, Ana (Autor de correspondencia)Revista: JOURNAL OF CLINICAL MEDICINEISSN: 2077-0383 Vol.10 N° 10 2021 págs. 2107ResumenMyelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis, dysplasia and peripheral cytopenias. Nowadays, MDS therapy is selected based on risk. The goals of therapy are different in low-risk and high-risk patients. In low-risk MDS, the goal is to decrease transfusion needs and to increase the quality of life. Currently, available drugs for newly diagnosed low-risk MDS include growth factor support, lenalidomide and immunosuppressive therapy. Additionally, luspatercept has recently been added to treat patients with MDS with ring sideroblasts, who are not candidates or have lost the response to erythropoiesis-stimulating agents. Treatment of high-risk patients is aimed to improve survival. To date, the only currently approved treatments are hypomethylating agents and allogeneic stem cell transplantation. However, the future for MDS patients is promising. In recent years, we are witnessing the emergence of multiple treatment combinations based on hypomethylating agents (pevonedistat, magrolimab, eprenetapopt, venetoclax) that have proven to be effective in MDS, even those with high-risk factors. Furthermore, the approval in the US of an oral hypomethylating agent opens the door to exclusively oral combinations for these patients and their consequent impact on the quality of life of these patients. Relapsed and refractory patients remain an unmet clinical need. We need more drugs and clinical trials for this profile of patients who have a dismal prognosis.
-
Autores: Maes, K. (Autor de correspondencia); Mondino, A.; Lasarte Sagastibelza, Juan José; et al.Revista: FRONTIERS IN IMMUNOLOGYISSN: 1664-3224 Vol.12 2021 págs. 652160ResumenCancer cells are under the surveillance of the host immune system. Nevertheless, a number of immunosuppressive mechanisms allow tumors to escape protective responses and impose immune tolerance. Epigenetic alterations are central to cancer cell biology and cancer immune evasion. Accordingly, epigenetic modulating agents (EMAs) are being exploited as anti-neoplastic and immunomodulatory agents to restore immunological fitness. By simultaneously acting on cancer cells, e.g. by changing expression of tumor antigens, immune checkpoints, chemokines or innate defense pathways, and on immune cells, e.g. by remodeling the tumor stroma or enhancing effector cell functionality, EMAs can indeed overcome peripheral tolerance to transformed cells. Therefore, combinations of EMAs with chemo- or immunotherapy have become interesting strategies to fight cancer. Here we review several examples of epigenetic changes critical for immune cell functions and tumor-immune evasion and of the use of EMAs in promoting anti-tumor immunity. Finally, we provide our perspective on how EMAs could represent a game changer for combinatorial therapies and the clinical management of cancer.
-
Autores: Cuenca, I. ; Alameda Serrano, Daniel; Sánchez, B.; et al.Revista: LEUKEMIAISSN: 0887-6924 Vol.35 N° 1 2021 págs. 245 - 249
-
Autores: Català-Moll, F.; Ferreté-Bonastre, A. G.; Li, T. L.; et al.Revista: NUCLEIC ACIDS RESEARCHISSN: 0305-1048 Vol.49 N° 9 2021 págs. 5057 - 5073ResumenActivation-induced deaminase (AID) initiates antibody diversification in germinal center B cells by deaminating cytosines, leading to somatic hypermutation and class-switch recombination. Loss-of-function mutations in AID lead to hyper-IgM syndrome type 2 (HIGM2), a rare human primary antibody deficiency. AID-mediated deamination has been proposed as leading to active demethylation of 5-methycytosines in the DNA, although evidence both supports and casts doubt on such a role. In this study, using whole-genome bisulfite sequencing of HIGM2 B cells, we investigated direct AID involvement in active DNA demethylation. HIGM2 naive and memory B cells both display widespread DNA methylation alterations, of which similar to 25% are attributable to active DNA demethylation. For genes that undergo active demethylation that is impaired in HIGM2 individuals, our analysis indicates that AID is not directly involved. We demonstrate that the widespread alterations in the DNA methylation and expression profiles of HIGM2 naive B cells result from premature overstimulation of the B-cell receptor prior to the germinal center reaction. Our data support a role for AID in B cell central tolerance in preventing the expansion of autoreactive cell clones, affecting the correct establishment of DNA methylation patterns.
-
Autores: Haertle, L.; Barrio, S.; Munawar, U.; et al.Revista: BLOODISSN: 0006-4971 Vol.138 N° 18 2021 págs. 1721 - 1726ResumenCereblon is the direct binding target of the immunomodulatory drugs (IMiDs) that are commonly used to treat multiple myeloma (MM), the second most frequent hematologic malignancy. Patients respond well to initial treatment with IMiDs, but virtually all patients develop drug resistance over time, and the underlying mechanisms are poorly understood. We identified an as yet undescribed DNA hypermethylation in an active intronic CRBN enhancer. Differential hypermethylation in this region was found to be increased in healthy plasma cells, but was more pronounced in IMiD-refractory MM. Methylation significantly correlated with decreased CRBN expression levels. DNA methyltransferase inhibitor (DNTMi) in vitro experiments induced CRBN enhancer demethylation, and sensitizing effects on lenalidomide treatment were observed in 2 MM cell lines. Thus, we provide first evidence that aberrant CRBN DNA methylation is a novel mechanism of IMiD resistance in MM and may predict IMiD response prior to treatment.
-
Autores: Vilarrasa-Blasi, R.; Soler-Vila, P.; Verdaguer-Dot, N.; et al.Revista: NATURE COMMUNICATIONSISSN: 2041-1723 Vol.12 N° 1 2021ResumenTo investigate the three-dimensional (3D) genome architecture across normal B cell differentiation and in neoplastic cells from different subtypes of chronic lymphocytic leukemia and mantle cell lymphoma patients, here we integrate in situ Hi-C and nine additional omics layers. Beyond conventional active (A) and inactive (B) compartments, we uncover a highly-dynamic intermediate compartment enriched in poised and polycomb-repressed chromatin. During B cell development, 28% of the compartments change, mostly involving a widespread chromatin activation from naive to germinal center B cells and a reversal to the naive state upon further maturation into memory B cells. B cell neoplasms are characterized by both entity and subtype-specific alterations in 3D genome organization, including large chromatin blocks spanning key disease-specific genes. This study indicates that 3D genome interactions are extensively modulated during normal B cell differentiation and that the genome of B cell neoplasias acquires a tumor-specific 3D genome architecture.
-
Autores: Sargas, C.; Ayala, R.; Chillón, M. C.; et al.Revista: HAEMATOLOGICA-THE HEMATOLOGY JOURNALISSN: 0390-6078 Vol.106 N° 12 2021 págs. 3079 - 3089ResumenNext-Generation Sequencing has recently been introduced to efficiently and simultaneously detect genetic variations in acute myeloid leukemia. However, its implementation in the clinical routine raises new challenges focused on the diversity of assays and variant reporting criteria. To overcome this challenge, the PETHEMA group established a nationwide network of reference laboratories aimed to deliver molecular results in the clinics. We report the technical cross-validation results for next-generation sequencing panel genes during the standardization process and the clinical validation in 823 samples of 751 patients with newly diagnosed or refractory/relapse acute myeloid leukemia. Two cross-validation rounds were performed in seven nationwide reference laboratories in order to reach a consensus regarding quality metrics criteria and variant reporting. In the pre-standardization cross-validation round, an overall concordance of 60.98% was obtained with a great variability in selected genes and conditions across laboratories. After consensus of relevant genes and optimization of quality parameters the overall concordance rose to 85.57% in the second cross-validation round. We show that a diagnostic network with harmonized next-generation sequencing analysis and reporting in seven experienced laboratories is feasible in the context of a scientific group.
-
Autores: Polverelli, N. (Autor de correspondencia); Mauff, K.; Kroger, N.; et al.Revista: AMERICAN JOURNAL OF HEMATOLOGYISSN: 0361-8609 Vol.96 N° 1 2021 págs. 69 - 79ResumenThe role of spleen size and splenectomy for the prediction of post-allogeneic hematopoietic stem cell transplant (allo-HCT) outcome in myelofibrosis remains under debate. In EBMT registry, we identified a cohort of 1195 myelofibrosis patients transplanted between 2000-2017 after either fludarabine-busulfan or fludarabine-melphalan regimens. Overall, splenectomy was performed in 202 (16.9%) patients and its use decreased over time (28.3% in 2000-2009 vs 14.1% in 2010-2017 period). By multivariate analysis, splenectomy was associated with less NRM (HR 0.64, 95% CI 0.44-0.93, P = .018) but increased risk of relapse (HR 1.43, 95% CI 1.01-2.02, P = .042), with no significant impact on OS (HR 0.86, 95% CI 0.67-1.12, P = .274). However, in subset analysis comparing the impact of splenectomy vs specific spleen sizes, for patients with progressive disease, an improved survival was seen in splenectomised subjects compared to those patients with a palpable spleen length >= 15 cm (HR 0.44, 95% CI 0.28-0.69, P < .001), caused by a significant reduction in NRM (HR 0.26, 95% CI 0.14-0.49, P < .001), without significantly increased relapse risk (HR 1.47, 95% CI 0.87-2.49, P = .147). Overall, despite the possible biases typical of retrospective cohorts, this study highlights the potential detrimental effect of massive splenomegaly in transplant outcome and supports the role of splenectomy for myelofibrosis patients with progressive disease and large splenomegaly.
-
Autores: Grigorian-Shamagian, L.; Sanz-Ruiz, R.; Climent, A.; et al.Revista: CARDIOVASCULAR RESEARCHISSN: 0008-6363 Vol.117 N° 6 2021 págs. 1428 - 1433ResumenGreat expectations have been set around the clinical potential of regenerative and reparative medicine in the treatment of cardiovascular diseases [i.e. in particular, heart failure (HF)]. Initial excitement, spurred by encouraging preclinical data, resulted in a rapid translation into clinical research. The sobering outcome of the resulting clinical trials suggests that preclinical testing may have been insufficient to predict clinical outcome. A number of barriers for clinical translation include the inherent variability of the biological products and difficulties to develop potency and quality assays, insufficient rigour of the preclinical research and reproducibility of the results, manufacturing challenges, and scientific irregularities reported in the last years. The failure to achieve clinical success led to an increased scrutiny and scepticism as to the clinical readiness of stem cells and gene therapy products among clinicians, industry stakeholders, and funding bodies. The present impasse has attracted the attention of some of the most active research groups in the field, which were then summoned to analyse the position of the field and tasked to develop a strategy, to re-visit the undoubtedly promising future of cardiovascular regenerative and reparative medicine, based on lessons learned over the past two decades. During the scientific retreat of the ESC Working Group on Cardiovascular Regenerative and Reparative Medicine (CARE) in November 2018, the most relevant and timely research aspects in regenerative and/or reparative medicine were presented and critically discussed, with the aim to lay out a strategy for the future development of the field. We report herein the main ideas and conclusions of that meeting.
-
Autores: Calvo Arnedo, Isabel; Sharma, S.; Paulo, J. A.; et al.Revista: ISCIENCEISSN: 2589-0042 Vol.24 N° 11 2021 págs. 103338ResumenThe target of Rapamycin complex1 (TORC1) senses and integrates several environmental signals, including amino acid (AA) availability, to regulate cell growth. Folliculin (FLCN) is a tumor suppressor (TS) protein in renal cell carcinoma, which paradoxically activates TORC1 in response to AA supplementation. Fewtractable systems formodeling FLCN-as a TS are available. Here, we characterize the FLCNcontaining complex in Schizosaccharomyces pombe ( called BFC) and show that BFC augments TORC1 repression and activation in response to AA starvation and supplementation, respectively. BFC co-immunoprecipitates V-ATPase, a TORC1 modulator, and regulates its activity in an AA-dependent manner. BFC genetic and proteomic networks identify the conserved peptide transmembrane transporter Ptr2 and the phosphoribosylformylglycinamidine synthase Ade3 as new AA-dependent regulators of TORC1. Overall, these data ascribe an additional repressive function to Folliculin in TORC1 regulation and reveal S. pombe as an excellent system for modeling the AA-dependent, FLCN-mediated repression of TORC1 in eukaryotes.
-
Autores: Perez-Amill, L.; Suñe, G.; Antoñana-Vildosola, A.; et al.Revista: HAEMATOLOGICA-THE HEMATOLOGY JOURNALISSN: 0390-6078 Vol.106 N° 1 2021 págs. 173 - 184ResumenMultiple myeloma is a prevalent and incurable disease, despite the development of new and effective drugs. The recent development of chimeric antigen receptor (CAR)-T cell therapy has shown impressive results in the treatment of patients with relapsed or refractory hematological B cell malignancies. In the recent years, B-cell maturation antigen (BCMA) has appeared as a promising antigen to target using a variety of immuno-therapy treatments including CART cells, for MM patients. To this end, we generated clinical-grade murine CART cells directed against BCMA, named ARI2m cells. Having demonstrated its efficacy, and in an at-tempt to avoid the immune rejection of CART cells by the patient, the single chain variable fragment was humanized, creating ARI2h cells. ARI2h cells demonstrated comparable in vitro and in vivo efficacy to ARI2m cells, and superiority in cases of high tumor burden disease. In terms of inflammatory response, ARI2h cells showed a lower TNF¿ production and lower in vivo toxicity profile. Large-scale expansion of both ARI2m and ARI2h cells was efficiently conducted following Good Manufacturing Practice guidelines, obtaining the target CART cell dose required for treatment of multiple myeloma patients. Moreover, we demonstrate that soluble BCMA and BCMA released in vesicles impacts on CAR-BCMA activity. In sum-mary, this study sets the bases for the implementation of a clinical trial (EudraCT code: 2019-001472-11) to study the efficacy of ARI2h cell treatment for multiple myeloma patients.
-
Autores: Isidro, I. A. (Autor de correspondencia); Vicente, P.; Pais, D. A. M.; et al.Revista: BIOTECHNOLOGY AND BIOENGINEERINGISSN: 0006-3592 Vol.118 N° 9 2021 págs. 3610 - 3617ResumenHepatocyte-like cells derived from human-induced pluripotent stem cells (hiPSC-HLC) are expected to have important applications in drug screening and regenerative medicine. However, hiPSC-HLC are difficult to produce on a large-scale to obtain relevant numbers for such applications. The aim of this study was to implement a novel integrated strategy for scalable production of hiPSC-HLC and demonstrate the applicability of dielectric spectroscopy to monitor hiPSC expansion/differentiation processes. We cultured hiPSC as three-dimensional (3D) aggregates in stirred-tank bioreactors (STB) operated in perfusion with an in situ capacitance probe. Dissolved oxygen concentration and dilution rate were controlled along the process and after 5 days of cell expansion, the hepatic differentiation was integrated in sequential steps for 28 days. The hiPSC were able to grow as 3D aggregates and the expression of hepatic markers and albumin production after differentiation confirmed that hepatocyte differentiation improved when compared to 2D culture. These hiPSC-HLC exhibited functional characteristics of hepatocytes including glycogen storage and drug metabolization capacity. Our results also show a good correlation between the cell permittivity measured online and the aggregate biovolume measured by standard offline methods, demonstrating for the first time the potential of dielectric spectroscopy to monitor hiPSC expansion and differentiation in STB.
-
Autores: Zawislak, A. (Autor de correspondencia); Wozniak, K.; Aguirre Ena, Xabier; et al.Revista: INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTHISSN: 1661-7827 Vol.18 N° 21 2021 págs. 11483ResumenBackground: Non-syndromic cleft lip with/without cleft palate (NSCL/P) is a common congenital condition with a complex aetiology reflecting multiple genetic and environmental factors. Single nucleotide polymorphisms (SNPs) in ABCA4 have been associated with NSCL/P in several studies, although there are some inconsistent results. This study aimed to evaluate whether two SNPs in ABCA4, namely rs4147811 and rs560426, are associated with NSCL/P occurrence in the Polish population. Methods: The study included 627 participants: 209 paediatric patients with NSCL/P and 418 healthy newborn controls. DNA was isolated from the saliva of NSCL/P patients and from umbilical cord blood in the controls. Genotyping of rs4147811 and rs560426 was performed using quantitative PCR. Results: The rs4147811 (AG genotype) SNP in ABCA4 was associated with a decreased risk of NSCL/P (odds ratio (OR) 0.57; 95% confidence interval (CI) 0.39-0.84; p = 0.004), whereas the rs560426 (GG genotype) SNP was associated with an increased risk of NSCL/P (OR 2.13; 95% CI 1.31-3.48; p = 0.002). Limitations: This study-based on the correlation between single genetic variants and the occurrence of different phenotypes-might have limited power in detecting relevant, complex inheritance patterns. ORs are often low to moderate when investigating the association of single genes with the risk of a complex trait. Another limitation was the small number of available NSCL/P samples. Conclusions: The results suggest that genetic variations in ABCA4 are important risk markers of NSCL/P in the Polish population. Further investigation in a larger study group is warranted.</p>
-
Autores: Rabal Gracia, María Obdulia; San José Enériz, Edurne; Aguirre Ena, Xabier; et al.Revista: JOURNAL OF MEDICINAL CHEMISTRYISSN: 0022-2623 Vol.64 N° 6 2021 págs. 3392 - 3426ResumenConcomitant inhibition of key epigenetic pathways involved in silencing tumor suppressor genes has been recognized as a promising strategy for cancer therapy. Herein, we report a first-in-class series of quinoline-based analogues that simultaneously inhibit histone deacetylases (from a low nanomolar range) and DNA methyltransferase-1 (from a mid-nanomolar range, IC50 < 200 nM). Additionally, lysine methyltransferase G9a inhibitory activity is achieved (from a low nanomolar range) by introduction of a key lysine mimic group at the 7-position of the quinoline ring. The corresponding epigenetic functional cellular responses are observed: histone-3 acetylation, DNA hypomethylation, and decreased histone-3 methylation at lysine-9. These chemical probes, multitarget epigenetic inhibitors, were validated against the multiple myeloma cell line MM1.S, demonstrating promising in vitro activity of 12a (CM-444) with GI(50) of 32 nM, an adequate therapeutic window (>1 log unit), and a suitable pharmacokinetic profile. In vivo, 12a achieved significant antitumor efficacy in a xenograft mouse model of human multiple myeloma.
-
Autores: Parra-Salinas, I. (Autor de correspondencia); Bermudez, A.; Lopez-Corral, L.; et al.Revista: CLINICAL TRANSPLANTATIONISSN: 0902-0063 Vol.35 N° 5 2021 págs. e14255ResumenTreatment of steroid-refractory chronic graft-versus-host disease (cGVHD) is a challenge. Here, we describe a retrospective analysis of 66 patients with steroid-refractory cGVHD treated with imatinib (starting dose of 100 mg in 70% of patients; maximum dose of 100-200 mg in 74%). Most patients had multi-organ involvement (>= 2 organs, 83%), with the most affected being skin (85%), oral mucosa (55%), eyes (42%), and lungs (33%). The overall response rate was 41% (21 partial and three complete responses). The organ with the best response rate was the skin (46%), followed by gastrointestinal tract (43%), liver (41%), the oral mucosa (36%), eyes (29%), and lungs (18%). Imatinib led to steroid tapering in 17/38 patients. Twenty-five (38%) patients experienced imatinib-related adverse events, comprising extra-hematologic toxicity (n = 24, 36%) and hematologic toxicity (n = 6, 9%). No cases of grade 4-5 toxicity were reported. The main causes of imatinib discontinuation were treatment failure (52%) and toxicity (9%). After a median follow-up of 41 months, the 3-year overall survival was 81%, with no difference between imatinib responders and non-responders. These real-life results show that imatinib is safe and has moderate efficacy in patients with heavily pre-treated cutaneous sclerotic cGVHD; however, activity against lung cGVHD is very limited.
-
Autores: Garcia-Gomez, A. (Autor de correspondencia); Li, T.; de la Calle-Fabregat, C.; et al.Revista: NATURE COMMUNICATIONSISSN: 2041-1723 Vol.12 N° 1 2021 págs. 421ResumenMultiple myeloma (MM) progression and myeloma-associated bone disease (MBD) are highly dependent on bone marrow mesenchymal stromal cells (MSCs). MM-MSCs exhibit abnormal transcriptomes, suggesting the involvement of epigenetic mechanisms governing their tumor-promoting functions and prolonged osteoblast suppression. Here, we identify widespread DNA methylation alterations of bone marrow-isolated MSCs from distinct MM stages, particularly in Homeobox genes involved in osteogenic differentiation that associate with their aberrant expression. Moreover, these DNA methylation changes are recapitulated in vitro by exposing MSCs from healthy individuals to MM cells. Pharmacological targeting of DNMTs and G9a with dual inhibitor CM-272 reverts the expression of hypermethylated osteogenic regulators and promotes osteoblast differentiation of myeloma MSCs. Most importantly, CM-272 treatment prevents tumor-associated bone loss and reduces tumor burden in a murine myeloma model. Our results demonstrate that epigenetic aberrancies mediate the impairment of bone formation in MM, and its targeting by CM-272 is able to reverse MBD. Mesenchymal stromal cells (MSCs) have been shown to support multiple myeloma (MM) development. Here, MSCs isolated from the bone marrow of MM patients are shown to have altered DNA methylation patterns and a methyltransferase inhibitor reverts MM-associated bone loss and reduces tumour burden in MM murine models.
-
Autores: Jagannath, S. (Autor de correspondencia); Lin, Y.; Goldschmidt, H.; et al.Revista: BLOOD CANCER JOURNALISSN: 2044-5385 Vol.11 N° 6 2021 págs. 116ResumenPatients with relapsed and refractory multiple myeloma (RRMM) who are triple-class exposed (to an immunomodulatory agent, proteasome inhibitor, and anti-CD38 antibody) have limited treatment options and there is no standard of care. Idecabtagene vicleucel (ide-cel, bb2121), a BCMA-directed CAR T-cell therapy, demonstrated efficacy in triple-class exposed RRMM patients in the KarMMa trial (NCT03361748). In this retrospective study (KarMMa-RW), patient-level data from triple-class exposed RRMM patients were merged into a single data model and compared with KarMMa using trimmed stabilized inverse probability of treatment weighting. Endpoints included overall response rate (ORR; primary), rate of very good partial response or better (>= VGPR), progression-free survival (PFS), and overall survival (OS). Of 1949 real-world triple-class exposed RRMM patients, 190 received subsequent (index) line of therapy and met KarMMa eligibility criteria (Eligible RRMM cohort). With a median follow-up of 13.3 months in KarMMa and 10.2 months in Eligible RRMM, ORR, and >= VGPR were significantly improved in KarMMa versus Eligible RRMM (ORR, 76.4% vs 32.2%; >= VGPR, 57.9% vs 13.7%; both P < 0.0001) as were PFS (11.6 vs 3.5 months; P = 0.0004) and OS (20.2 vs 14.7 months; P = 0.0006). This study demonstrated that ide-cel significantly improved responses and survival compared with currently available therapies in triple-class exposed RRMM.
-
Autores: Valcárcel, L. V.; Amundarain, A.; Kulis, M.; et al.Revista: LEUKEMIAISSN: 0887-6924 Vol.35 N° 10 2021 págs. 3012 - 3016ResumenClinical and genetic risk factors are currently used in multiple myeloma (MM) to stratify patients and to design specific therapies. However, these systems do not capture the heterogeneity of the disease supporting the development of new prognostic factors. In this study, we identified active promoters and alternative active promoters in 6 different B cell subpopulations, including bone-marrow plasma cells, and 32 MM patient samples, using RNA-seq data. We find that expression initiated at both regular and alternative promoters was specific of each B cell subpopulation or MM plasma cells, showing a remarkable level of consistency with chromatin-based promoter definition. Interestingly, using 595 MM patient samples from the CoMMpass dataset, we observed that the expression derived from some alternative promoters was associated with lower progression-free and overall survival in MM patients independently of genetic alterations. Altogether, our results define cancer-specific alternative active promoters as new transcriptomic features that can provide a new avenue for prognostic stratification possibilities in patients with MM.
-
Autores: Feliu, J.; Panizo Santos, Carlos Manuel; Marcos Jubilar, María; et al.Libro: La clínica y el laboratorio2019 págs. 661 - 708
-
Autores: Ezponda Itoiz, Teresa; Alkorta Aranburu, Gorka; Prosper Cardoso, Felipe; et al.Título: NGS data analysis processLibro: Principles of Nutrigenetics and Nutrigenomics: Fundamentals of Individualized NutritionISSN: 9780128045725; 9780128045879 2019 págs. 33 - 39ResumenThe study of nutrigenetics and nutrigenomics requires an identification of the genetic background of an organism to determine how this affects nutrient metabolism and the effects that the nutrients have on it. Advances in sequencing technologies have caused a shift from gene-based to genome-wide analyses that have provided information previously unsuspected and unavailable. Although these techniques are commonly used, they are still evolving and susceptible to improvement: therefore, some potential biases need to be considered regarding their performance and use. In this chapter, we describe the most common techniques used to study nutrigenetics and nutrigenomics, focusing on key steps to be followed to obtain reliable results.
Proyectos desde 2018
-
Título: DeSarrollO de terapias CAR-T innovadoras para el trAtamiento de Tumores Hematológicos y Sólidos. (SOCRATHeS)Código de expediente: 0011-1411-2022-000053Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2022 GN PROYECTOS ESTRATEGICOS DE I+D 2022-2025Fecha de inicio: 01-05-2022Fecha fin: 30-12-2024Importe concedido: 371.489,00€Otros fondos: -
-
Título: Bioingeniería avanzada para el desarrollo del tejido cardiaco y su aplicación al estudio y detección de cardiotoxicidadCódigo de expediente: 0011-1411-2022-000071Investigador principal: MANUEL MARIA MAZO VEGA.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2022 GN PROYECTOS ESTRATEGICOS DE I+D 2022-2025Fecha de inicio: 03-04-2022Fecha fin: 30-12-2024Importe concedido: 196.436,13€Otros fondos: -
-
Título: Terapias avanzadasCódigo de expediente: RD21/0017/0009Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2021 AES Redes de Investigación cooperativa orientadas a resultados (RICORS)Fecha de inicio: 01-01-2022Fecha fin: 31-12-2024Importe concedido: 86.316,00€Otros fondos: Fondos FEDER
-
Título: CARDIOPRINT_Biofabricación avanzada multifunción en 3D para la generación de tejido cardíaco terapéuti co a escala humana diseñado por ordenador.Código de expediente: PLEC2021-008127Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: AGENCIA ESTATAL DE INVESTIGACIONConvocatoria: 2021 AEI Proyectos de I+D+i en líneas estratégicasFecha de inicio: 01-12-2021Fecha fin: 31-12-2024Importe concedido: 203.867,00€Otros fondos: -
-
Título: Bioingenieria personalizada para el tratamiento de las enfermedades cardiovasculares. Estudio de la implicación del géneroCódigo de expediente: 0011-1383-2022-000015(PC020-21-022 BIOGEN)Investigador principal: MANUEL MARIA MAZO VEGA.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2022 GN Proyectos ColaborativosFecha de inicio: 01-12-2021Fecha fin: 30-11-2024Importe concedido: 211.870,50€Otros fondos: -
-
Título: New targets and designs to improve CAR-T cell based immunotherapy against pancreatic cancer (CarPanTu)Código de expediente: PLEC2021-008094Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: AGENCIA ESTATAL DE INVESTIGACIONConvocatoria: 2021 AEI Proyectos de I+D+i en líneas estratégicasFecha de inicio: 01-11-2021Fecha fin: 31-10-2024Importe concedido: 100.042,00€Otros fondos: Fondos MRR
-
Título: Biotecnología aplicada a la obtención de polímeros imprimibles para aplicaciones biomédicas a partir de subproductos de origen agroalimentario de Navarra (IMPRIMED)Código de expediente: 0011-1411-2021-000096Investigador principal: MANUEL MARIA MAZO VEGA.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2021 GN PROYECTOS ESTRATEGICOS DE I+D 2021-2024Fecha de inicio: 01-06-2021Fecha fin: 31-12-2023Importe concedido: 223.280,88€Otros fondos: -
-
Título: Aplicaciones del estudio multi-ómico de la microbiota al desarrollo de soluciones biotecnológicas innovadoras en el área de la salud (microBiomics)Código de expediente: 0011-1411-2021-000106Investigador principal: MARIA TERESA HERRAIZ BAYOD.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2021 GN PROYECTOS ESTRATEGICOS DE I+D 2021-2024Fecha de inicio: 15-04-2021Fecha fin: 30-11-2023Importe concedido: 366.577,17€Otros fondos: -
-
Título: Papel de los mecanimso de regulación trascripcional en la patogenia y el tratamiento de los SMDCódigo de expediente: PI20/01308Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2020 AES Proyectos de investigaciónFecha de inicio: 01-01-2021Fecha fin: 31-12-2023Importe concedido: 275.880,00€Otros fondos: Fondos FEDER
-
Título: Alianza en Genómica Avanzada para el desarrollo de Terapias Personalizadas en NavarraCódigo de expediente: 0011-1411-2020-000010Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: GOBIERNO DE NAVARRAConvocatoria: FIMA 2020 GN PROYECTOS ESTRATEGICOS DE I+D 2020-2022Fecha de inicio: 17-06-2020Fecha fin: 30-11-2022Importe concedido: 725.480,08€Otros fondos: -
-
Título: Alianza en Genómica Avanzada para el desarrollo de Terapias Avanzadas en Navarra (AGATA)Código de expediente: 0011-1411-2020-000011Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2020 GN PROYECTOS ESTRATEGICOS DE I+D 2020-2022Fecha de inicio: 16-06-2020Fecha fin: 30-11-2022Importe concedido: 441.998,75€Otros fondos: -
-
Título: Transcriptional and gene regulatory networks in acute myeloid leukemia secondary to myelodysplastic syndromes: identification of novel therapeutic targetsCódigo de expediente: 0011-0537-2019-000001Investigador principal: NEREA BERASTEGUI ZUFIAURRE.Financiador: GOBIERNO DE NAVARRA / DPTO. EDUCACIÓN CULTURA Y TURISMOConvocatoria: FIMA GNE 2019 BECAS PREDOCTORALESFecha de inicio: 01-06-2020Fecha fin: 01-10-2023Importe concedido: 68.715,96€Otros fondos: -
-
Título: Metabolic vulnerabilities for personalized therapeutic approaches in acute myeloid leukemiaCódigo de expediente: 0011-2750-2019-000001Investigador principal: FELIPE LUIS PROSPER CARDOSO, FRANCISCO JAVIER PLANES PEDREÑO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2019 GN PERMEDFecha de inicio: 01-03-2020Fecha fin: 28-02-2023Importe concedido: 228.600,00€Otros fondos: -
-
Título: Desarrollo y evaluación de inmunoterapia celular mediante CART alogénicas para el tratamiento de la Leucemia Mieloide AgudaCódigo de expediente: 0011-1383-2020-000010 PC011Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2020 GN Proyectos ColaborativosFecha de inicio: 01-12-2019Fecha fin: 30-11-2022Importe concedido: 180.675,00€Otros fondos: -
-
Título: Caracterización bioinformática, del perfil transcriptómico y epigenético de células madre hematopoyéticas (CD34+). Integración de ambas tecnologías genómicas para identificar "gene regulatory networks"Código de expediente:Investigador principal: MARINA AINCIBURU FERNANDEZ.Financiador: GOBIERNO DE NAVARRA / DPTO. EDUCACIÓN CULTURA Y TURISMOConvocatoria: GNE 2018 BECAS PREDOCTORALESFecha de inicio: 01-04-2019Fecha fin: 08-09-2019Importe concedido: 68.718,00€Otros fondos: Fondos FEDER
-
Título: Desarrollo EStratégico de terapias CART para el tratamiento de Tumores Hematológicos y Sólidos (DESCARTHeS)Código de expediente: 0011-1411-2019-000072Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2019 GN PROYECTOS ESTRATEGICOS DE I+D 2019-2021Fecha de inicio: 01-04-2019Fecha fin: 30-11-2021Importe concedido: 164.695,50€Otros fondos: -
-
Título: Optimización de nuevos agentes epigenéticos para el tratamiento de la Leucemia Mieloide Aguda.Código de expediente: 0011-1383-2019-000006Investigador principal: MARIA OBDULIA RABAL GRACIA.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2019 - GN INDUSTRIA PROYECTOS CENTROS TECNOLOGICOS Y ORGANISMOS DE INVESTIGACIONFecha de inicio: 01-12-2018Fecha fin: 30-11-2019Importe concedido: 125.835,30€Otros fondos: -
-
Título: Terapia basada en RNA mediante quimeras aptámero-siRNAs contra lncRNAs en el mieloma múltiple. RTHALMYCódigo de expediente: SAF2017-92632-EXPInvestigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: MINISTERIO DE CIENCIA E INNOVACIÓNConvocatoria: 2017 - PROYECTOS EXPLORA CIENCIA Y EXPLORA TECNOLOGIAFecha de inicio: 01-11-2018Fecha fin: 31-12-2020Importe concedido: 84.700,00€Otros fondos: Fondos FEDER
-
Título: Identificación y estudio funcional de transcritos quiméricos no codificantes en el mieloma múltipleCódigo de expediente: FPU17/02733Investigador principal: ANE AMUNDARAIN IRAOLA.Financiador: MINISTERIO DE CIENCIA E INNOVACIÓNConvocatoria: 2017 - MINECO BECAS FPU 2017 - MINECO BECAS FPU 2017 - MINECO BECAS FPUFecha de inicio: 01-10-2018Fecha fin: 28-02-2023Importe concedido: 91.828,32€Otros fondos: Fondos FEDER
-
Título: Contratación Río HortegaCódigo de expediente: CM17/00046Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: AES 2017 RIO HORTEGAFecha de inicio: 13-04-2018Fecha fin: 12-04-2020Importe concedido: 56.732,00€Otros fondos: -
-
Título: MINERVA. Estudio genómico para la personalización del diagnóstico y el tratamiento de los pacientes con insuficiencia Cardíaca crónica y enfermedad renal crónica (Medicina cardIoreNal pERsonalizada en NaVArra)Código de expediente: 0011-1411-2018-000043Investigador principal: DOMINGO FRANCISCO JAVIER DIEZ MARTINEZ, DOMINGO FRANCISCO JAVIER DIEZ MARTINEZ.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2018- GN PROY. ESTRATEGICOS DE I+D 2018-2020Fecha de inicio: 01-04-2018Fecha fin: 30-11-2020Importe concedido: 1.001.241,69€Otros fondos: -
-
Título: GENEURONA. Implantación del diagnóstico genómico de la epilepsia y la migraña en NavarraCódigo de expediente: 0011-1411-2018-000044Investigador principal: ANA MARIA GARCIA OSTA.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2018- GN PROY. ESTRATEGICOS DE I+D 2018-2020Fecha de inicio: 01-04-2018Fecha fin: 30-11-2020Importe concedido: 476.239,53€Otros fondos: -
-
Título: Estudio genómico para la personalización del diagnóstico y el tratamiento de los pacientes con insuficiencia cardiaca crónica y enfermedad renal crónica (Medicina cardIoreNal pERsonalizada en NaVArra) (MINERVA)Código de expediente: 0011-1411-2018-000036Investigador principal: JUAN JOSE GAVIRA GOMEZ.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2018 GN PROYECTOS ESTRATEGICOS DE I+D 2018-2020Fecha de inicio: 01-04-2018Fecha fin: 30-11-2020Importe concedido: 97.237,60€Otros fondos: -
-
Título: Detección y validación funcional del trasncriptoma completo y análisis de genes esenciales y sintéticos letales en el Mieloma MúltipleCódigo de expediente: FI17/00297Investigador principal: LUIS VITORES VALCARCEL GARCIA.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2017 AYUDAS PREDOCTORALES DE FORMACION EN INVESTIGACION EN SALUD PFISFecha de inicio: 24-01-2018Fecha fin: 23-01-2022Importe concedido: 82.400,00€Otros fondos: -
-
Título: Estudio de la arquitectura genómica y transcripcional en SMDs como herramienta para la determinación de factores pronósticos y nuevas dianas terapeúticas.Código de expediente: PI17/00701Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: AES2017 PROYECTOS DE INVESTIGACIÓNFecha de inicio: 01-01-2018Fecha fin: 31-12-2020Importe concedido: 209.330,00€Otros fondos: Fondos FEDER
-
Título: Plataformas automáticas de producción de células CAR T para el tratamiento de leucemia B y linfoma BCódigo de expediente: RTC-2017-6578-1Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: MINISTERIO DE CIENCIA E INNOVACIÓNConvocatoria: 2017 MINECO RETOS COLABORACIÓNFecha de inicio: 01-01-2018Fecha fin: 31-10-2021Importe concedido: 100.985,84€Otros fondos: Fondos FEDER
-
Título: Ayuda del ISCIII Sara BorrellCódigo de expediente: CD17/00108Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: AES2017 SARA BORRELLFecha de inicio: 01-01-2018Fecha fin: 31-12-2020Importe concedido: 80.598,00€Otros fondos: -
-
Título: Tecnología de secuenciación de nueva generación (NGS) para optimizar la eficacia del diagnóstico y tratamiento en pacientes con tumores de alta mortalidad (DIANA: Diagnostico biomédico e Innovación Abierta en Navarra)Código de expediente: 0011-1411-2017-000028Investigador principal: MARIA JOSE CALASANZ ABINZANO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2017 GN ESTRATEGICOSFecha de inicio: 01-04-2017Fecha fin: 30-11-2019Importe concedido: 821.776,08€Otros fondos: -
-
Título: Tecnología de secuenciación de nueva generación (NGS) para optimizar la eficacia del diagnóstico y tratamiento en pacientes con tumores de alta mortalidad (DIANA: Diagnostico biomédico e Innovación Abierta en Navarra)Código de expediente: 0011-1411-2017-000030Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2017 GN ESTRATEGICOSFecha de inicio: 01-04-2017Fecha fin: 30-11-2019Importe concedido: 37.315,72€Otros fondos: -
-
Título: Detección y validación funcional de los RNAs largos no codificantes específicos de la célula plasmática en el Mieloma MúltipleCódigo de expediente: PI16/02024Investigador principal: XABIER AGUIRRE ENA.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2016 - PROYECTOS DE I+D EN SALUDFecha de inicio: 01-01-2017Fecha fin: 31-12-2019Importe concedido: 98.615,00€Otros fondos: Fondos FEDER
-
Título: Inhibición de la actividad metiltransferasa de G9a mediante moléculas pequeñas como estrategia terapéutica en tumores hematológicos.Código de expediente: FI16/00275Investigador principal: ARANTXA CARRASCO LEON.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2016 AYUDAS PREDOCTORALES DE FORMACION EN INVESTIGACION EN SALUD PFISFecha de inicio: 01-01-2017Fecha fin: 31-12-2020Importe concedido: 82.400,00€Otros fondos: Fondos FEDER
-
Título: Identificación de nuevas alteraciones en síndrome mielodisplásico en riesgo de progresión a leucemia mieloide aguda: biomarcadores y nuevas dianas terapéuticas mediante NGSCódigo de expediente: PI16/00159Investigador principal: MARTA FERNANDEZ MERCADO, MARIA JOSE CALASANZ ABINZANO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: AES2017 PROYECTOS DE INVESTIGACIÓNFecha de inicio: 01-01-2017Fecha fin: 30-06-2020Importe concedido: 122.815,00€Otros fondos: Fondos FEDER
-
Título: Detección, anotación y análisis del papel de los eRNAs en el Mieloma MúltipleCódigo de expediente: 40_2016Investigador principal: XABIER AGUIRRE ENA.Financiador: GOBIERNO DE NAVARRA. DEPARTAMENTO DE SALUDConvocatoria: 2016 PROYECTOS DE I+D EN SALUDFecha de inicio: 09-12-2016Fecha fin: 08-12-2019Importe concedido: 63.549,00€Otros fondos: Fondos FEDER
-
Título: Nanopartículas de esqualeno-adenosina para el tratamiento de la isquemia-reperfusión cardiacaCódigo de expediente: PCIN-2016-046Investigador principal: MARIA JOSE BLANCO PRIETO.Financiador: MINISTERIO DE CIENCIA E INNOVACIÓNConvocatoria: 2016 MINECO ACCIONES DE PROGRAMACIÓN CONJUNTA INTERNACIONALFecha de inicio: 01-05-2016Fecha fin: 31-12-2020Importe concedido: 98.000,00€Otros fondos: -
-
Título: Understanding primary hyperoxaluria type 1 towards the development of innovative therapeutic strategies (ERARE)Código de expediente: AC15/00036Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2015 AES ACCIONES COMPLEMENTARIASFecha de inicio: 01-01-2016Fecha fin: 31-12-2019Importe concedido: 99.946,00€Otros fondos: -
-
Título: Inhibición de la actividad metiltransferasa de G9a mediante moléculas pequeñas como estrategia terapéutica en tumores hematológicosCódigo de expediente: PI14/01867Investigador principal: FELIPE LUIS PROSPER CARDOSO.Financiador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: 2014 - PROYECTOS DE I+D EN SALUDFecha de inicio: 01-01-2015Fecha fin: 30-06-2018Importe concedido: 207.515,00€Otros fondos: Fondos FEDER
-
Título: SyLeNCe: Synapses between Leukaemia and its Neighbouring CellsCódigo de expediente: 837491Investigador principal: ISABEL CALVO ARNEDOFinanciador: COMISIÓN EUROPEAConvocatoria: H2020-EC-MSCA-IFFecha de inicio: 01-07-2020Fecha fin: 30-06-2022Importe concedido: 160.932,48€Otros fondos: -
-
Título: Elucidating transcriptional rewiring on hematological malignancies via computational methodsCódigo de expediente: 898356Investigador principal: MIKEL HERNAEZ ARRAZOLAFinanciador: COMISIÓN EUROPEAConvocatoria: H2020-EC-MSCA-IFFecha de inicio: 01-03-2020Fecha fin: 28-02-2022Importe concedido: 172.932,48€Otros fondos: -
-
Título: EPICA: Dual epigenetic targeting and immunotherapy to fight against cancer.Código de expediente: AC16/00041Investigador principal: FELIPE LUIS PROSPER CARDOSOFinanciador: INSTITUTO DE SALUD CARLOS IIIConvocatoria: TRANSCAN-2Fecha de inicio: 01-03-2017Fecha fin: 31-08-2020Importe concedido: 144.159,40€Otros fondos: Fondos FEDER
-
Título: DECALCódigo de expediente:Investigador principal: TERESA EZPONDA ITOIZFinanciador: GILEAD SCIENCES, S.L.Convocatoria: GILEAD 2015Fecha de inicio: 24-06-2016Fecha fin: 23-06-2018Importe concedido: 115.000,00€Otros fondos: -
-
Título: Identification of the epigenomic and transcriptomic alterations in the transformation of benign monoclonal gammapathies to symptomatic multiple myeloma.Investigador principal: FELIPE LUIS PROSPER CARDOSOFinanciador: FUNDACION BBVAConvocatoria: 2021 Fundación BBVA Proyectos de investigaciónFecha de inicio: 01-07-2022Fecha fin: 30-06-2024Importe concedido: 149.984,50€
-
Título: Gene Regulatory NETworks in NORmal and MALignant Hematopoiesis- Identification and Targeting (GR-NET NORMAL-HITInvestigador principal: FELIPE LUIS PROSPER CARDOSOFinanciador: FUNDACIÓN BANCARIA LA CAIXAConvocatoria: 2020 FD La Caixa Health ResearchFecha de inicio: 01-12-2020Fecha fin: 30-11-2023Importe concedido: 411.250,00€
-
Título: Deciphering the role of transcription factors as chromatin modulators and drivers of lymphoid neoplasmInvestigador principal: XABIER AGUIRRE ENAFinanciador: FUNDACIO "LA MARATO DE TV3"Convocatoria: FIMA 2019 FD LA MARATÓ TV3 PROYECTOS DE INVESTIGACIÓNFecha de inicio: 30-07-2020Fecha fin: 29-07-2023Importe concedido: 101.875,00€
-
Título: Caracterización del transcriptoma y vías reguladoras génicas en el eje envejecimiento-hematopoyesis clonal-síndromes mielodisplásicos como herramienta para la identificación de nuevas dianas terapéuticasInvestigador principal: TERESA EZPONDA ITOIZFinanciador: ASOCIACION ESPAÑOLA CONTRA EL CANCERConvocatoria: 2019 AECC Investigador, 2019 AECC InvestigadorFecha de inicio: 01-12-2019Fecha fin: 30-11-2023Importe concedido: 200.000,00€
-
Título: A systems biology approach to reveal the niche composition and connectivity in aging and malignant transformation at the single cell levelInvestigador principal: ITZIAR CENZANO ARMENDARIZFinanciador: ASOCIACION ESPAÑOLA CONTRA EL CANCERConvocatoria: FIMA 2019 Predoctorales AECC, FIMA 2019 Predoctorales AECCFecha de inicio: 01-12-2019Fecha fin: 30-11-2023Importe concedido: 91.530,54€
-
Título: SUBCONTRACTING RNA SEQInvestigador principal: FELIPE LUIS PROSPER CARDOSOFecha de inicio: 20-06-2019Fecha fin: 20-06-2020Importe: 0Otros fondos: -
-
Título: GENOMICS OF 5q- SYNDROME AND INMUNE RESIST TO REVLIMIDInvestigador principal: FELIPE LUIS PROSPER CARDOSOFecha de inicio: 13-05-2019Fecha fin: 30-09-2021Importe: 0Otros fondos: -
-
Título: Development of non-genotoxic conditioning regimens for hematopoietic stem cell transplantation in Ataxia Telangiectasia.Investigador principal: FELIPE LUIS PROSPER CARDOSOFinanciador: Asocición Española Familia Ataxia TelangiectasiaConvocatoria: 2017 AEFATFecha de inicio: 24-04-2018Fecha fin: 24-04-2021Importe concedido: 150.000,00€
-
Título: Investigación y explotación de la función de microRNAs aberrantemente expresados en linfomas B difusos de células grandesInvestigador principal: FELIPE LUIS PROSPER CARDOSOFinanciador: ASOCIACION ESPAÑOLA CONTRA EL CANCERConvocatoria: 2017 AECC PostdoctoralesFecha de inicio: 01-05-2017Fecha fin: 31-07-2019Importe concedido: 56.250,00€
-
Título: Analysis of functional role of the IncRNAs in multiple myelomaInvestigador principal: FELIPE LUIS PROSPER CARDOSOFinanciador: GILEAD SCIENCES, S.L.Convocatoria: 2016 GILEADFecha de inicio: 20-01-2017Fecha fin: 20-01-2019Importe concedido: 49.967,79€