Network


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

Hotspot


Dive into the research topics where Naiara Martínez-Vélez is active.

Publication


Featured researches published by Naiara Martínez-Vélez.


Neuro-oncology | 2016

Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51

Enric Xipell; Tomás Aragón; Naiara Martínez-Vélez; Beatriz Vera; Miguel Angel Idoate; Juan J. Martínez-Irujo; Antonia García Garzón; Marisol Gonzalez-Huarriz; Arlet M. Acanda; Chris Jones; Frederick F. Lang; Juan Fueyo; Candelaria Gomez-Manzano; Marta M. Alonso

BACKGROUND Endoplasmic reticulum (ER) stress results from protein misfolding imbalance and has been postulated as a therapeutic strategy. ER stress activates the unfolded protein response which leads to a complex cellular response, including the upregulation of aberrant protein degradation in the ER, with the goal of resolving that stress. O(6)-methylguanine DNA methyltransferase (MGMT), N-methylpurine DNA glycosylase (MPG), and Rad51 are DNA damage repair proteins that mediate resistance to temozolomide in glioblastoma. In this work we sought to evaluate whether ER stress-inducing drugs were able to downmodulate DNA damage repair proteins and become candidates to combine with temozolomide. METHODS MTT assays were performed to evaluate the cytotoxicity of the treatments. The expression of proteins was evaluated using western blot and immunofluorescence. In vivo studies were performed using 2 orthotopic glioblastoma models in nude mice to evaluate the efficacy of the treatments. All statistical tests were 2-sided. RESULTS Treatment of glioblastoma cells with ER stress-inducing drugs leads to downregulation of MGMT, MPG, and Rad51. Inhibition of ER stress through pharmacological treatment resulted in rescue of MGMT, MPG, and Rad51 protein levels. Moreover, treatment of glioblastoma cells with salinomycin, an ER stress-inducing drug, and temozolomide resulted in enhanced DNA damage and a synergistic antitumor effect in vitro. Of importance, treatment with salinomycin/temozolomide resulted in a significant antiglioma effect in 2 aggressive orthotopic intracranial brain tumor models. CONCLUSIONS These findings provide a strong rationale for combining temozolomide with ER stress-inducing drugs as an alternative therapeutic strategy for glioblastoma.


Journal of Bone and Mineral Research | 2014

The oncolytic adenovirus Δ24-RGD in combination with cisplatin exerts a potent anti-osteosarcoma activity

Naiara Martínez-Vélez; Enric Xipell; Patricia Jauregui; Marta Zalacain; Lucía Marrodán; Carolina Zandueta; Beatriz Vera; Leire Urquiza; Luis Sierrasesúmaga; Mikel San Julián; Gemma Toledo; Juan Fueyo; Candelaria Gomez-Manzano; Wensceslao Torre; Fernando Lecanda; Ana Patiño-García; Marta M. Alonso

Osteosarcoma is the most common malignant bone tumor in children and adolescents. The presence of metastases and the lack of response to conventional treatment are the major adverse prognostic factors. Therefore, there is an urgent need for new treatment strategies that overcome both of these problems. Our purpose was to elucidate whether the use of the oncolytic adenovirus Δ24‐RGD alone or in combination with standard chemotherapy would be effective, in vitro and in vivo, against osteosarcoma. Our results showed that Δ24‐RGD exerted a potent antitumor effect against osteosarcoma cell lines that was increased by the addition of cisplatin. Δ24‐RGD osteosarcoma treatment resulted in autophagy in vitro that was further enhanced when combined with cisplatin. Of importance, administration of Δ24‐RGD and/or cisplatin, in novel orthotopic and two lung metastatic models in vivo resulted in a significant reduction of tumor burden meanwhile maintaining a safe toxicity profile. Together, our data underscore the potential of Δ24‐RGD to become a realistic therapeutic option for primary and metastatic pediatric osteosarcoma. Moreover, this study warrants a future clinical trial to evaluate the safety and efficacy of Δ24‐RGD for this devastating disease.


Clinical Cancer Research | 2016

The Oncolytic Adenovirus VCN-01 as Therapeutic Approach Against Pediatric Osteosarcoma

Naiara Martínez-Vélez; Enric Xipell; Beatriz Vera; Arlet Acanda de la Rocha; Marta Zalacain; Lucía Marrodán; Marisol Gonzalez-Huarriz; Gemma Toledo; Manel Cascallo; Ramon Alemany; Ana Patiño; Marta M. Alonso

Purpose: Osteosarcoma is the most common malignant bone tumor in children and adolescents. Despite aggressive chemotherapy, more than 30% of patients do not respond and develop bone or lung metastasis. Oncolytic adenoviruses engineered to specifically destroy cancer cells are a feasible option for osteosarcoma treatment. VCN-01 is a replication-competent adenovirus specifically engineered to replicate in tumors with a defective RB pathway, presents an enhanced infectivity through a modified fiber and an improved distribution through the expression of a soluble hyaluronidase. The aim of this study is to elucidate whether the use of VCN-01 would be an effective therapeutic strategy for pediatric osteosarcoma. Experimental Design: We used osteosarcoma cell lines established from patients with metastatic disease (531MII, 678R, 588M, and 595M) and a commercial cell line (143B). MTT assays were carried out to evaluate the cytotoxicity of VCN-01. Hexon assays were used to evaluate the replication of the virus. Western blot analysis was performed to assess the expression levels of viral proteins and autophagic markers. The antitumor effect of VCN-01 was evaluated in orthotopic and metastatic osteosarcoma murine animal models. Results: This study found that VCN-01, a new generation genetically modified oncolytic adenovirus, administered locally or systemically, had a potent antisarcoma effect in vitro and in vivo in mouse models of intratibial and lung metastatic osteosarcoma. Moreover, VCN-01 administration showed a safe toxicity profile. Conclusions: These results uncover VCN-01 as a promising strategy for osteosarcoma, setting the bases to propel a phase I/II trial for kids with this disease. Clin Cancer Res; 22(9); 2217–25. ©2015 AACR.


PLOS ONE | 2016

Characterization of the Antiglioma Effect of the Oncolytic Adenovirus VCN-01

Beatriz Vera; Naiara Martínez-Vélez; Enric Xipell; Arlet Acanda de la Rocha; Ana Patiño-García; Javier Saez-Castresana; Marisol Gonzalez-Huarriz; Manel Cascallo; Ramon Alemany; Marta M. Alonso

Despite the recent advances in the development of antitumor therapies, the prognosis for patients with malignant gliomas remains dismal. Therapy with tumor-selective viruses is emerging as a treatment option for this devastating disease. In this study we characterize the anti-glioma effect of VCN-01, an improved hyaluronidase-armed pRB-pathway-selective oncolytic adenovirus that has proven safe and effective in the treatment of several solid tumors. VCN-01 displayed a significant cytotoxic effect on glioma cells in vitro. In vivo, in two different orthotopic glioma models, a single intra-tumoral administration of VCN-01 increased overall survival significantly and led to long-term survivors free of disease.


Journal of bone oncology | 2017

Oncolytic adenoviruses as a therapeutic approach for osteosarcoma: A new hope

Marc García-Moure; Naiara Martínez-Vélez; Ana Patiño-García; Marta M. Alonso

Osteosarcoma is the most common bone cancer among those with non-hematological origin and affects mainly pediatric patients. In the last 50 years, refinements in surgical procedures, as well as the introduction of aggressive neoadjuvant and adjuvant chemotherapeutic cocktails, have increased to nearly 70% the survival rate of these patients. Despite the initial therapeutic progress the fight against osteosarcoma has not substantially improved during the last three decades, and almost 30% of the patients do not respond or recur after the standard treatment. For this group there is an urgent need to implement new therapeutic approaches. Oncolytic adenoviruses are conditionally replicative viruses engineered to selectively replicate in and kill tumor cells, while remaining quiescent in healthy cells. In the last years there have been multiple preclinical and clinical studies using these viruses as therapeutic agents in the treatment of a broad range of cancers, including osteosarcoma. In this review, we summarize some of the most relevant published literature about the use of oncolytic adenoviruses to treat human osteosarcoma tumors in subcutaneous, orthotopic and metastatic mouse models. In conclusion, up to date the preclinical studies with oncolytic adenoviruses have demonstrated that are safe and efficacious against local and metastatic osteosarcoma. Knowledge arising from phase I/II clinical trials with oncolytic adenoviruses in other tumors have shown the potential of viruses to awake the patient´s own immune system generating a response against the tumor. Generating osteosarcoma immune-competent adenoviruses friendly models will allow to better understand this potential. Future clinical trials with oncolytic adenoviruses for osteosarcoma tumors are warranted.


PLOS ONE | 2016

Analysis of SOX2-Regulated Transcriptome in Glioma Stem Cells

Arlet M. Acanda de la Rocha; Hernando Lopez-Bertoni; Elizabeth Guruceaga; Marisol Gonzalez-Huarriz; Naiara Martínez-Vélez; Enric Xipell; Juan Fueyo; Candelaria Gomez-Manzano; Marta M. Alonso

Introduction Glioblastoma is the most malignant brain tumor in adults and is associated with poor survival despite multimodal treatments. Glioma stem-like cells (GSCs) are cells functionally defined by their self-renewal potential and the ability to reconstitute the original tumor upon orthotopic implantation. They have been postulated to be the culprit of glioma chemo- and radio-resistance ultimately leading to relapse. Understanding the molecular circuits governing the GSC compartment is essential. SOX2, a critical transcription regulator of embryonic and neural stem cell function, is deregulated in GSCs however; the precise molecular pathways regulated by this gene in GSCs remain poorly understood. Results We performed a genome-wide analysis of SOX2-regulated transcripts in GSCs, using a microarray. We identified a total of 2048 differentially expressed coding transcripts and 261 non-coding transcripts. Cell adhesion and cell-cell signaling are among the most enriched terms using Gene Ontology (GO) classification. The pathways altered after SOX2 down-modulation includes multiple cellular processes such as amino-acid metabolism and intercellular signaling cascades. We also defined and classified the set of non-coding transcripts differentially expressed regulated by SOX2 in GSCs, and validated two of them. Conclusions We present a comprehensive analysis of the transcriptome controlled by SOX2 in GSCs, gaining insights in the understanding of the potential roles of SOX2 in glioblastoma.


OncoImmunology | 2018

Intratumoral injection of activated B lymphoblast in combination with PD-1 blockade induces systemic antitumor immunity with reduction of local and distal tumors

Mario Martínez Soldevilla; Helena Villanueva; Naiara Martínez-Vélez; Daniel Meraviglia-Crivelli; Marta M. Alonso; Javier Cebollero; Ashwathi P Menon; Montserrat Puigdelloses; Fernando Pastor

ABSTRACT In spite of the success of PD-1 blocking antibodies in the clinic their benefits are still restricted to a small fraction of patients. Immune-desert tumors and/or the highly immunosuppressive tumor milieu might hamper the success of PD-1/PD-L1 blocking therapies into a broader range of cancer patients. Although still under debate, there is a cumulative body of evidence that indicates B tumor-infiltrating lymphocytes are a good prognostic marker in most types of cancer, especially in those that form ectopic lymphoid tissue structures. Taking this into account, we reason that the adoptive transfer of activated B lymphoblasts (ABL) in the tumor could be a feasible therapeutic approach to shift the immunosuppressive tumor microenvironment into an immune-permissive one. In this work we show the antitumor effect of ABL therapy in two different tumor models: colon carcinoma (CT26) and melanoma (B16/F10). The ABL transfer in the most relevant non-immunogenic B16/F10 melanoma model depicts synergism with anti-PD-1 antibody therapy. Furthermore, systemic antitumor immunity was detected in mice treated with PD-1 antibody/ABL combination which was able to reach distal metastatic lesions.


Archive | 2018

Oncolytic Virotherapy for Gliomas

Naiara Martínez-Vélez; Candelaria Gomez-Manzano; Juan Fueyo; Ana Patiño-García; Marta M. Alonso

Abstract Gliomas are the most common and aggressive brain tumors and despite advances in their therapeutic management they remain fatal. Radically, different therapeutic approaches are needed to treat this aggressive disease. Oncolytic viruses that intrinsically kill cancer cells or that have been genetically engineered to specifically destroy this population are becoming a viable therapeutic options for gliomas. A number of preclinical studies have demonstrated the efficacy of oncolytic viruses as a therapeutic strategy for gliomas. Those studies have led to a number of clinical trials using oncolytic virus for glioma treatment. In this chapter, we review the different viruses (adenovirus, herpes virus, parvovirus, poliovirus, reovirus, newcastle disease virus, retrovirus, and measles viruses) that have been evaluated preclinically and clinically for the treatment of glioma.


Neuro-oncology | 2018

The aberrant splicing of BAF45d links splicing regulation and transcription in glioblastoma

Guillermo Aldave; Marisol González-Huarriz; Angel Rubio; Juan Pablo Romero; Datta Ravi; Belén Miñana; Mar Cuadrado-Tejedor; Ana García-Osta; Roeland Verhaak; Enric Xipell; Naiara Martínez-Vélez; Arlet Acanda de la Rocha; Montserrat Puigdelloses; Marc García-Moure; Miguel Marigil; Jaime Gállego Pérez-Larraya; Oskar Marín-Béjar; Maite Huarte; Maria Stella Carro; Roberto Ferrarese; Cristobal Belda-Iniesta; Angel Ayuso; Ricardo Prat-Acín; Fernando Pastor; Ricardo Díez-Valle; Sonia Tejada; Marta M. Alonso

Background Glioblastoma, the most aggressive primary brain tumor, is genetically heterogeneous. Alternative splicing (AS) plays a key role in numerous pathologies, including cancer. The objectives of our study were to determine whether aberrant AS could play a role in the malignant phenotype of glioma and to understand the mechanism underlying its aberrant regulation. Methods We obtained surgical samples from patients with glioblastoma who underwent 5-aminolevulinic fluorescence-guided surgery. Biopsies were taken from the tumor center as well as from adjacent normal-appearing tissue. We used a global splicing array to identify candidate genes aberrantly spliced in these glioblastoma samples. Mechanistic and functional studies were performed to elucidate the role of our top candidate splice variant, BAF45d, in glioblastoma. Results BAF45d is part of the switch/sucrose nonfermentable complex and plays a key role in the development of the CNS. The BAF45d/6A isoform is present in 85% of over 200 glioma samples that have been analyzed and contributes to the malignant glioma phenotype through the maintenance of an undifferentiated cellular state. We demonstrate that BAF45d splicing is mediated by polypyrimidine tract-binding protein 1 (PTBP1) and that BAF45d regulates PTBP1, uncovering a reciprocal interplay between RNA splicing regulation and transcription. Conclusions Our data indicate that AS is a mechanism that contributes to the malignant phenotype of glioblastoma. Understanding the consequences of this biological process will uncover new therapeutic targets for this devastating disease.


Cancers | 2018

Tune Up In Situ Autovaccination against Solid Tumors with Oncolytic Viruses

Teresa Nguyen; Naze G. Avci; Dong Ho Shin; Naiara Martínez-Vélez; Hong Jiang

With the progress of immunotherapy in cancer, oncolytic viruses (OVs) have attracted more and more attention during the past decade. Due to their cancer-selective and immunogenic properties, OVs are considered ideal candidates to be combined with immunotherapy to increase both specificity and efficacy in cancer treatment. OVs preferentially replicate in and lyse cancer cells, resulting in in situ autovaccination leading to adaptive anti-virus and anti-tumor immunity. The main challenge in OV approaches is how to redirect the host immunity from anti-virus to anti-tumor and optimize the clinical outcome of cancer patients. Here, we summarize the conceptual updates on oncolytic virotherapy and immunotherapy in cancer, and the development of strategies to enhance the virus-mediated anti-tumor immune response, including: (1) arm OVs with cytokines to modulate innate and adaptive immunity; (2) combining OVs with immune checkpoint inhibitors to release T cell inhibition; (3) combining OVs with immune co-stimulators to enhance T cell activation. Future studies need to be enforced on developing strategies to augment the systemic effect on metastasized tumors.

Collaboration


Dive into the Naiara Martínez-Vélez's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juan Fueyo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Candelaria Gomez-Manzano

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Arlet Acanda de la Rocha

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge