Idoia Blanco-Luquin
University College London
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Idoia Blanco-Luquin.
OncoImmunology | 2014
Therese Liechtenstein; Noemi Perez-Janices; Idoia Blanco-Luquin; Cleo Goyvaerts; Julia Katharina Schwarze; Inès Dufait; Alessio Lanna; Mark De Ridder; David Guerrero-Setas; Karine Breckpot; David Escors
Efficacious antitumor vaccines strongly stimulate cancer-specific effector T cells and counteract the activity of tumor-infiltrating immunosuppressive cells. We hypothesised that combining cytokine expression with silencing programmed cell death ligand 1 (PD-L1) could potentiate anticancer immune responses of lentivector vaccines. Thus, we engineered a collection of lentivectors that simultaneously co-expressed an antigen, a PD-L1-silencing shRNA, and various T cell-polarising cytokines, including interferon γ (IFNγ), transforming growth factor β (TGFβ) or interleukins (IL12, IL15, IL23, IL17A, IL6, IL10, IL4). In a syngeneic B16F0 melanoma model and using tyrosinase related protein 1 (TRP1) as a vaccine antigen, we found that simultaneous delivery of IL12 and a PD-L1-silencing shRNA was the only combination that exhibited therapeutically relevant anti-melanoma activities. Mechanistically, we found that delivery of the PD-L1 silencing construct boosted T cell numbers, inhibited in vivo tumor growth and strongly cooperated with IL12 cytokine priming and antitumor activities. Finally, we tested the capacities of our vaccines to counteract tumor-infiltrating myeloid-derived suppressor cell (MDSC) activities ex vivo. Interestingly, the lentivector co-expressing IL12 and the PD-L1 silencing shRNA was the only one that counteracted MDSC suppressive activities, potentially underlying the observed anti-melanoma therapeutic benefit. We conclude that (1) evaluation of vaccines in healthy mice has no significant predictive value for the selection of anticancer treatments; (2) B16 cells expressing xenoantigens as a tumor model are of limited value; and (3) vaccines which inhibit the suppressive effect of MDSC on T cells in our ex vivo assay show promising and relevant antitumor activities.
OncoImmunology | 2013
David Escors; Therese Liechtenstein; Noemi Perez-Janices; Julia Schwarze; Ines Dufait; Cleo Goyvaerts; Alessio Lanna; Frederick Arce; Idoia Blanco-Luquin; Grazyna Kochan; David Guerrero-Setas; Karine Breckpot
Since dendritic cells operate as professional antigen-presenting cells (APCs) and hence are capable of jumpstarting the immune system, they have been exploited to develop a variety of immunotherapeutic regimens against cancer. In the few past years, myeloid-derived suppressor cells (MDSCs) have been shown to mediate robust immunosuppressive functions, thereby inhibiting tumor-targeting immune responses. Thus, we propose that the immunomodulatory activity of MDSCs should be carefully considered for the development of efficient anticancer immunotherapies.
Virus Research | 2013
Therese Liechtenstein; Noemi Perez-Janices; Christopher Bricogne; Alessio Lanna; Ines Dufait; Cleo Goyvaerts; Roberta Laranga; Antonella Padella; Frederick Arce; Mehdi Baratchian; Natalia Ramirez; Natalia Lopez; Grazyna Kochan; Idoia Blanco-Luquin; David Guerrero-Setas; Karine Breckpot; David Escors
Our work over the past eight years has focused on the use of HIV-1 lentiviral vectors (lentivectors) for the genetic modification of dendritic cells (DCs) to control their functions in immune modulation. DCs are key professional antigen presenting cells which regulate the activity of most effector immune cells, including T, B and NK cells. Their genetic modification provides the means for the development of targeted therapies towards cancer and autoimmune disease. We have been modulating with lentivectors the activity of intracellular signalling pathways and co-stimulation during antigen presentation to T cells, to fine-tune the type and strength of the immune response. In the course of our research, we have found unexpected results such as the surprising immunosuppressive role of anti-viral signalling pathways, and the close link between negative co-stimulation in the immunological synapse and T cell receptor trafficking. Here we review our major findings and put them into context with other published work.
Oncotarget | 2015
Maria Gato-Cañas; Xabier Martínez de Morentin; Idoia Blanco-Luquin; Joaquín Fernández-Irigoyen; Isabel Zudaire; Therese Liechtenstein; Hugo Arasanz; Teresa Lozano; Noelia Casares; A. Chaikuad; Stefan Knapp; David Guerrero-Setas; David Escors; Grazyna Kochan; Enrique Santamaría
Myeloid-derived suppressor cells (MDSCs) differentiate from bone marrow precursors, expand in cancer-bearing hosts and accelerate tumor progression. MDSCs have become attractive therapeutic targets, as their elimination strongly enhances anti-neoplastic treatments. Here, immature myeloid dendritic cells (DCs), MDSCs modeling tumor-infiltrating subsets or modeling non-cancerous (NC)-MDSCs were compared by in-depth quantitative proteomics. We found that neoplastic MDSCs differentially expressed a core of kinases which controlled lineage-specific (PI3K-AKT and SRC kinases) and cancer-induced (ERK and PKC kinases) protein interaction networks (interactomes). These kinases contributed to some extent to myeloid differentiation. However, only AKT and ERK specifically drove MDSC differentiation from myeloid precursors. Interfering with AKT and ERK with selective small molecule inhibitors or shRNAs selectively hampered MDSC differentiation and viability. Thus, we provide compelling evidence that MDSCs constitute a distinct myeloid lineage distinguished by a “kinase signature” and well-defined interactomes. Our results define new opportunities for the development of anti-cancer treatments targeting these tumor-promoting immune cells.
Proteomics | 2016
Maria Gato; Idoia Blanco-Luquin; Maribel Zudaire; Xabier Martínez de Morentin; Estela Pérez-Valderrama; Aintzane Zabaleta; Grazyna Kochan; David Escors; Joaquín Fernández-Irigoyen; Enrique Santamaría
Myeloid‐derived suppressor cells (MDSCs) are a heterogeneous population of cells that are defined by their myeloid origin, immature state, and ability to potently suppress T‐cell responses. They regulate immune responses and the population significantly increases in the tumor microenvironment of patients with glioma and other malignant tumors. For their study, MDSCs are usually isolated from the spleen or directly of tumors from a large number of tumor‐bearing mice although promising ex vivo differentiated MDSC production systems have been recently developed. During the last years, proteomics has emerged as a powerful approach to analyze MDSCs proteomes using shotgun‐based mass spectrometry (MS), providing functional information about cellular homeostasis and metabolic state at a global level. Here, we will revise recent proteome profiling studies performed in MDSCs from different origins. Moreover, we will perform an integrative functional analysis of the protein compilation derived from these large‐scale proteomic studies in order to obtain a comprehensive view of MDSCs biology. Finally, we will also discuss the potential application of high‐throughput proteomic approaches to study global proteome dynamics and post‐translational modifications (PTMs) during the differentiation process of MDSCs that will greatly boost the identification of novel MDSC‐specific therapeutic targets to apply in cancer immunotherapy.
Oncotarget | 2017
Esperanza Martín-Sánchez; Saioa Mendaza; Ane Ulazia-Garmendia; Iñaki Monreal-Santesteban; Idoia Blanco-Luquin; Alicia Córdoba; Francisco Vicente-Garcia; Noemi Perez-Janices; David Escors; Diego Megías; Paula Lopez-Serra; Manel Esteller; Jose Juan Illarramendi; David Guerrero-Setas
The CHL1 gene encodes a cell-adhesion molecule proposed as being a putative tumour-suppressor gene in breast cancer (BC). However, neither the underlying molecular mechanisms nor the clinical value of CHL1 downregulation in BC has been explored. The methylation status of three CpG sites in the CHL1 promoter was analysed by pyrosequencing in neoplastic biopsies from 142 patients with invasive BC and compared with that of non-neoplastic tissues. We found higher CHL1 methylation levels in breast tumours than in non-neoplastic tissues, either from mammoplasties or adjacent-to-tumour, which correlated with lower levels of protein expression in tumours measured by immunohistochemistry. A panel of five BC cell lines was treated with two epigenetic drugs, and restoration of CHL1 expression was observed, indicating in vitro dynamic epigenetic regulation. CHL1 was silenced by shRNA in immortalized but non-neoplastic mammary cells, and enhanced cell proliferation and migration, but not invasion, were found by real-time cell analysis. The prognostic value of CHL1 hypermethylation was assessed by the log-rank test and fitted in a Cox regression model. Importantly, CHL1 hypermethylation was very significantly associated with shorter progression-free survival in our BC patient series, independent of age and stage (p = 0.001). In conclusion, our results indicate that CHL1 is downregulated by hypermethylation and that this epigenetic alteration is an independent prognostic factor in BC.
Pathology International | 2015
Idoia Blanco-Luquin; Rosa Guarch; Amaya Ojer; Noemi Perez-Janices; Esperanza Martín-Sánchez; Sergio Maria-Ruiz; Iñaki Monreal-Santesteban; Laura Blanco-Fernandez; Eduardo Pernaut-Leza; David Escors; David Guerrero-Setas
Cervical cancer is the third most common cancer in women worldwide. The hypermethylation of P16, TSLC‐1 and TSP‐1 genes was analyzed in squamous cell carcinomas (SCC), cervical intraepithelial lesions (CIN) and adenocarcinomas (ADC) of the uterine cervix (total 181 lesions). Additionally human papillomavirus (HPV) type, EPB41L3, RASSF1 and RASSF2 hypermethylation were tested in ADC and the results were compared with those obtained previously by our group in SCC. P16, TSLC‐1 and TSP‐1 hypermethylation was more frequent in SCCs than in CINs. These percentages and the corresponding ones for EPB41L3, RASSF1 and RASSF2 genes were also higher in SCCs than in ADCs, except for P16. The presence of HPV in ADCs was lower than reported previously in SCC and CIN. Patients with RASSF1A hypermethylation showed significantly longer disease‐free survival (P = 0.015) and overall survival periods (P = 0.009) in ADC patients. To our knowledge, this is the first description of the EPB41L3 and RASSF2 hypermethylation in ADCs. These results suggest that the involvement of DNA hypermethylation in cervical cancer varies depending on the histological type, which might contribute to explaining the different prognosis of patients with these types of tumors.
Melanoma management | 2017
Hugo Arasanz; Alejandra Lacalle; Maria José Lecumberri; Ángela Fernández de Lascoiti; Idoia Blanco-Luquin; Maria Gato-Cañas; Leyre Pérez-Ricarte; Miren Zuazo; Grazyna Kochan; David Escors
Immunotherapy has radically transformed the management of metastatic malignant melanoma. Ipilimumab, a CTLA-4-targeted monoclonal antibody, was the first immunotherapeutic drug to reach a survival benefit compared with traditional chemotherapy. PD-1 targeted therapies, pembrolizumab and nivolumab, have demonstrated, in recent clinical trials, to be even more effective and safer. PD-1 and CTLA-4 blockade combination appears to improve the outcomes achieved so far, although increasing toxicity. However, many questions concerning the optimal timing of administration or the most adequate sequence of treatment are yet to be answered.
Clinical Epigenetics | 2017
Esperanza Martín-Sánchez; Saioa Mendaza; Ane Ulazia-Garmendia; Iñaki Monreal-Santesteban; Alicia Córdoba; Francisco Vicente-Garcia; Idoia Blanco-Luquin; Susana de la Cruz; Ana Aramendia; David Guerrero-Setas
BackgroundCadherin-like protein 22 (CDH22) is a transmembrane glycoprotein involved in cell-cell adhesion and metastasis. Its role in cancer is controversial because it has been described as being upregulated in colorectal cancer, whereas it is downregulated in metastatic melanoma. However, its status in breast cancer (BC) is unknown. The purpose of our study was to determine the molecular status and clinical value of CDH22 in BC.ResultsWe observed by immunohistochemistry that the level of CDH22 expression was lower in BC tissues than in their matched adjacent-to-tumour and non-neoplastic tissues from reduction mammoplasties. Since epigenetic alteration is one of the main causes of gene silencing, we analysed the hypermethylation of 3 CpG sites in the CDH22 promoter by pyrosequencing in a series of 142 infiltrating duct BC cases. CDH22 was found to be hypermethylated in tumoral tissues relative to non-neoplastic mammary tissues. Importantly, this epigenetic alteration was already present in adjacent-to-tumour tissues, although to a lesser extent than in tumoral samples. Furthermore, CDH22 gene regulation was dynamically modulated in vitro by epigenetic drugs. Interestingly, CDH22 hypermethylation in all 3 CpG sites simultaneously, but not expression, was significantly associated with shorter progression-free survival (p = 0.015) and overall survival (p = 0.021) in our patient series. Importantly, CDH22 hypermethylation was an independent factor that predicts poor progression-free survival regardless of age and stage (p = 0.006).ConclusionsOur results are the first evidence that CDH22 is hypermethylated in BC and that this alteration is an independent prognostic factor in BC. Thus, CDH22 hypermethylation could be a potential biomarker of poor prognosis in BC.
Immunotherapy | 2016
Maria Gato-Cañas; Hugo Arasanz; Idoia Blanco-Luquin; Estíbaliz Glaría; Virginia Arteta-Sanchez; Grazyna Kochan; David Escors
Immunotherapies are achieving clinical success for the treatment of many cancers. However, it has taken a long time to exploit the potential of the immune system for the treatment of human cancers. We cannot forget that this has been the consequence of very extensive work in basic research in preclinical models and in human patients. Thus, it is rather hard to compile all of it while giving a comprehensive view on this subject. Here we have attempted to give an overall perspective in immunotherapy of melanoma. A brief overview on current therapies is provided, followed by adoptive cell therapies. Gene engineering strategies to improve these therapies are also explained, finishing with therapies based on interference with immune checkpoint pathways.