Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Estíbaliz Miranda is active.

Publication


Featured researches published by Estíbaliz Miranda.


PLOS ONE | 2011

Frequent and Simultaneous Epigenetic Inactivation of TP53 Pathway Genes in Acute Lymphoblastic Leukemia

Amaia Vilas–Zornoza; Xabier Agirre; Vanesa Martín-Palanco; José I. Martín-Subero; Edurne San José-Enériz; Leire Garate; Sara Alvarez; Estíbaliz Miranda; Paula Rodriguez-Otero; José Rifón; Antonio Faustino Torres; María José Calasanz; Juan C. Cigudosa; Jose Roman-Gomez; Felipe Prosper

Aberrant DNA methylation is one of the most frequent alterations in patients with Acute Lymphoblastic Leukemia (ALL). Using methylation bead arrays we analyzed the methylation status of 807 genes implicated in cancer in a group of ALL samples at diagnosis (n = 48). We found that 154 genes were methylated in more than 10% of ALL samples. Interestingly, the expression of 13 genes implicated in the TP53 pathway was downregulated by hypermethylation. Direct or indirect activation of TP53 pathway with 5-aza-2′-deoxycitidine, Curcumin or Nutlin-3 induced an increase in apoptosis of ALL cells. The results obtained with the initial group of 48 patients was validated retrospectively in a second cohort of 200 newly diagnosed ALL patients. Methylation of at least 1 of the 13 genes implicated in the TP53 pathway was observed in 78% of the patients, which significantly correlated with a higher relapse (p = 0.001) and mortality (p<0.001) rate being an independent prognostic factor for disease-free survival (DFS) (p = 0.006) and overall survival (OS) (p = 0.005) in the multivariate analysis. All these findings indicate that TP53 pathway is altered by epigenetic mechanisms in the majority of ALL patients and correlates with prognosis. Treatments with compounds that may reverse the epigenetic abnormalities or activate directly the p53 pathway represent a new therapeutic alternative for patients with ALL.


Leukemia | 2012

Preclinical activity of LBH589 alone or in combination with chemotherapy in a xenogeneic mouse model of human acute lymphoblastic leukemia.

Amaia Vilas-Zornoza; Xabier Agirre; Gloria Abizanda; Cristina Moreno; Victor Segura; A De Martino Rodriguez; Edurne San José-Enériz; Estíbaliz Miranda; José I. Martín-Subero; Leire Garate; María J. Blanco-Prieto; J A García de Jalón; P Río; José Rifón; Juan C. Cigudosa; Jose A. Martinez-Climent; Jose Roman-Gomez; María José Calasanz; José Manuel Ribera; Felipe Prosper

Histone deacetylases (HDACs) have been identified as therapeutic targets due to their regulatory function in chromatin structure and organization. Here, we analyzed the therapeutic effect of LBH589, a class I–II HDAC inhibitor, in acute lymphoblastic leukemia (ALL). In vitro, LBH589 induced dose-dependent antiproliferative and apoptotic effects, which were associated with increased H3 and H4 histone acetylation. Intravenous administration of LBH589 in immunodeficient BALB/c-RAG2−/−γc−/− mice in which human-derived T and B-ALL cell lines were injected induced a significant reduction in tumor growth. Using primary ALL cells, a xenograft model of human leukemia in BALB/c-RAG2−/−γc−/− mice was established, allowing continuous passages of transplanted cells to several mouse generations. Treatment of mice engrafted with T or B-ALL cells with LBH589 induced an in vivo increase in the acetylation of H3 and H4, which was accompanied with prolonged survival of LBH589-treated mice in comparison with those receiving vincristine and dexamethasone. Notably, the therapeutic efficacy of LBH589 was significantly enhanced in combination with vincristine and dexamethasone. Our results show the therapeutic activity of LBH589 in combination with standard chemotherapy in pre-clinical models of ALL and suggest that this combination may be of clinical value in the treatment of patients with ALL.


Nature Communications | 2017

Discovery of first-in-class reversible dual small molecule inhibitors against G9a and DNMTs in hematological malignancies.

Edurne San José-Enériz; Xabier Agirre; Obdulia Rabal; Amaia Vilas-Zornoza; Juan A. Sánchez-Arias; Estíbaliz Miranda; Ana Ugarte; Sergio Roa; Bruno Paiva; Ander Estella-Hermoso de Mendoza; Rosa Alvarez; Noelia Casares; Victor Segura; José I. Martín-Subero; François-Xavier Ogi; Pierre Soule; Clara M. Santiveri; Ramón Campos-Olivas; Giancarlo Castellano; Maite Garcia Fernandez de Barrena; Juan R. Rodriguez-Madoz; María José García-Barchino; Juan José Lasarte; Matías A. Avila; Jose A. Martinez-Climent; Julen Oyarzabal; Felipe Prosper

The indisputable role of epigenetics in cancer and the fact that epigenetic alterations can be reversed have favoured development of epigenetic drugs. In this study, we design and synthesize potent novel, selective and reversible chemical probes that simultaneously inhibit the G9a and DNMTs methyltransferase activity. In vitro treatment of haematological neoplasia (acute myeloid leukaemia-AML, acute lymphoblastic leukaemia-ALL and diffuse large B-cell lymphoma-DLBCL) with the lead compound CM-272, inhibits cell proliferation and promotes apoptosis, inducing interferon-stimulated genes and immunogenic cell death. CM-272 significantly prolongs survival of AML, ALL and DLBCL xenogeneic models. Our results represent the discovery of first-in-class dual inhibitors of G9a/DNMTs and establish this chemical series as a promising therapeutic tool for unmet needs in haematological tumours.


Nature Communications | 2017

An in-silico approach to predict and exploit synthetic lethality in cancer metabolism

Iñigo Apaolaza; Edurne San José-Enériz; Luis Tobalina; Estíbaliz Miranda; Leire Garate; Xabier Agirre; Felipe Prosper; Francisco J. Planes

Synthetic lethality is a promising concept in cancer research, potentially opening new possibilities for the development of more effective and selective treatments. Here, we present a computational method to predict and exploit synthetic lethality in cancer metabolism. Our approach relies on the concept of genetic minimal cut sets and gene expression data, demonstrating a superior performance to previous approaches predicting metabolic vulnerabilities in cancer. Our genetic minimal cut set computational framework is applied to evaluate the lethality of ribonucleotide reductase catalytic subunit M1 (RRM1) inhibition in multiple myeloma. We present a computational and experimental study of the effect of RRM1 inhibition in four multiple myeloma cell lines. In addition, using publicly available genome-scale loss-of-function screens, a possible mechanism by which the inhibition of RRM1 is effective in cancer is established. Overall, our approach shows promising results and lays the foundation to build a novel family of algorithms to target metabolism in cancer.Exploiting synthetic lethality is a promising approach for cancer therapy. Here, the authors present an approach to identifying such interactions by finding genetic minimal cut sets (gMCSs) that block cancer proliferation, and apply it to study the lethality of RRM1 inhibition in multiple myeloma.


Journal of Medicinal Chemistry | 2018

Discovery of Reversible DNA Methyltransferase and Lysine Methyltransferase G9a Inhibitors with Antitumoral in Vivo Efficacy

Obdulia Rabal; Edurne San José-Enériz; Xabier Agirre; Juan A. Sánchez-Arias; Amaia Vilas-Zornoza; Ana Ugarte; Irene de Miguel; Estíbaliz Miranda; Leire Garate; Mario F. Fraga; Pablo Santamarina; Raúl F Pérez; Raquel Ordoñez; Elena Sáez; Sergio Roa; María José García-Barchino; Jose A. Martinez-Climent; Yingying Liu; Wei Wu; Musheng Xu; Felipe Prosper; Julen Oyarzabal

Using knowledge- and structure-based approaches, we designed and synthesized reversible chemical probes that simultaneously inhibit the activity of two epigenetic targets, histone 3 lysine 9 methyltransferase (G9a) and DNA methyltransferases (DNMT), at nanomolar ranges. Enzymatic competition assays confirmed our design strategy: substrate competitive inhibitors. Next, an initial exploration around our hit 11 was pursued to identify an adequate tool compound for in vivo testing. In vitro treatment of different hematological neoplasia cell lines led to the identification of molecules with clear antiproliferative efficacies (GI50 values in the nanomolar range). On the basis of epigenetic functional cellular responses (levels of lysine 9 methylation and 5-methylcytosine), an acceptable therapeutic window (around 1 log unit) and a suitable pharmacokinetic profile, 12 was selected for in vivo proof-of-concept ( Nat. Commun. 2017 , 8 , 15424 ). Herein, 12 achieved a significant in vivo efficacy: 70% overall tumor growth inhibition of a human acute myeloid leukemia (AML) xenograft in a mouse model.


Journal of Medicinal Chemistry | 2018

Detailed Exploration around 4-Aminoquinolines Chemical Space to Navigate the Lysine Methyltransferase G9a and DNA Methyltransferase Biological Spaces

Obdulia Rabal; Juan A. Sánchez-Arias; Edurne San José-Enériz; Xabier Agirre; Irene de Miguel; Leire Garate; Estíbaliz Miranda; Elena Sáez; Sergio Roa; Jose A. Martinez-Climent; Yingying Liu; Wei Wu; Musheng Xu; Felipe Prosper; Julen Oyarzabal

Epigenetic regulators that exhibit aberrant enzymatic activities or expression profiles are potential therapeutic targets for cancers. Specifically, enzymes responsible for methylation at histone-3 lysine-9 (like G9a) and aberrant DNA hypermethylation (DNMTs) have been implicated in a number of cancers. Recently, molecules bearing a 4-aminoquinoline scaffold were reported as dual inhibitors of these targets and showed a significant in vivo efficacy in animal models of hematological malignancies. Here, we report a detailed exploration around three growing vectors born by this chemotype. Exploring this chemical space led to the identification of features to navigate G9a and DNMT1 biological spaces: not only their corresponding exclusive areas, selective compounds, but also common spaces. Thus, we identified from selective G9a and first-in-class DNMT1 inhibitors, >1 log unit between their IC50 values, with IC50 < 25 nM (e.g., 43 and 26, respectively) to equipotent inhibitors with IC50 < 50 nM for both targets (e.g., 13). Their ADME/Tox profiling and antiproliferative efficacies, versus some cancer cell lines, are also reported.


Scientific Reports | 2017

In-silico gene essentiality analysis of polyamine biosynthesis reveals APRT as a potential target in cancer

Jon Pey; Edurne San José-Enériz; Maria C. Ochoa; Iñigo Apaolaza; Pedro de Atauri; Angel Rubio; Xabier Cendoya; Estíbaliz Miranda; Leire Garate; Marta Cascante; Arkaitz Carracedo; Xabier Agirre; Felipe Prosper; Francisco J. Planes

Constraint-based modeling for genome-scale metabolic networks has emerged in the last years as a promising approach to elucidate drug targets in cancer. Beyond the canonical biosynthetic routes to produce biomass, it is of key importance to focus on metabolic routes that sustain the proliferative capacity through the regulation of other biological means in order to improve in-silico gene essentiality analyses. Polyamines are polycations with central roles in cancer cell proliferation, through the regulation of transcription and translation among other things, but are typically neglected in in silico cancer metabolic models. In this study, we analysed essential genes for the biosynthesis of polyamines. Our analysis corroborates the importance of previously known regulators of the pathway, such as Adenosylmethionine Decarboxylase 1 (AMD1) and uncovers novel enzymes predicted to be relevant for polyamine homeostasis. We focused on Adenine Phosphoribosyltransferase (APRT) and demonstrated the detrimental consequence of APRT gene silencing on different leukaemia cell lines. Our results highlight the importance of revisiting the metabolic models used for in-silico gene essentiality analyses in order to maximize the potential for drug target identification in cancer.


PLOS ONE | 2017

Reversible dual inhibitor against G9a and DNMT1 improves human iPSC derivation enhancing MET and facilitating transcription factor engagement to the genome

Juan R. Rodriguez-Madoz; Edurne San José-Enériz; Obdulia Rabal; Natalia Zapata-Linares; Estíbaliz Miranda; Saray Rodriguez; Angelo Porciuncula; Amaia Vilas-Zornoza; Leire Garate; Victor Segura; Elizabeth Guruceaga; Xabier Agirre; Julen Oyarzabal; Felipe Prosper; Austin J. Cooney

The combination of defined factors with small molecules targeting epigenetic factors is a strategy that has been shown to enhance optimal derivation of iPSCs and could be used for disease modelling, high throughput screenings and/or regenerative medicine applications. In this study, we showed that a new first-in-class reversible dual G9a/DNMT1 inhibitor compound (CM272) improves the efficiency of human cell reprogramming and iPSC generation from primary cells of healthy donors and patient samples, using both integrative and non-integrative methods. Moreover, CM272 facilitates the generation of human iPSC with only two factors allowing the removal of the most potent oncogenic factor cMYC. Furthermore, we demonstrated that mechanistically, treatment with CM272 induces heterochromatin relaxation, facilitates the engagement of OCT4 and SOX2 transcription factors to OSKM refractory binding regions that are required for iPSC establishment, and enhances mesenchymal to epithelial transition during the early phase of cell reprogramming. Thus, the use of this new G9a/DNMT reversible dual inhibitor compound may represent an interesting alternative for improving cell reprogramming and human iPSC derivation for many different applications while providing interesting insights into reprogramming mechanisms.


Blood | 2014

Inhibition of the Methyltransferase G9a with Small Molecules As a New Therapeutic Strategy for Treatment of Hematological Malignancies

Edurne San José-Enériz; Xabier Agirre; Obdulia Rabal; Sergio Roa; Amaia Vilas-Zornoza; Estíbaliz Miranda; Ana Ugarte; Juan A. Sánchez-Arias; Bruno Paiva; Jesús F. San Miguel; Jose A. Martinez-Climent; Julen Oyarzabal; Felipe Prosper


Blood | 2011

Preclinical Activity of LBH589 Alone or in Combination with Chemotherapy in a Xenogeneic Mouse Model of Human Acute Lymphoblastic Leukemia

Xabier Agirre; Amaia Vilas-Zornoza; Gloria Abizanda; Cristina Moreno; Victor Segura; Alba de Martino Rodríguez; Edurne San José-Enériz; Estíbaliz Miranda; José I. Martín-Subero; Leire Garate; María J. Blanco-Prieto; José A. García de Jalón; José Rifón; Jose A. Martinez-Climent; Juan C. Cigudosa; Jose Roman-Gomez; María José Calasanz; Josep-Maria Ribera; Felipe Prosper

Collaboration


Dive into the Estíbaliz Miranda's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge