Fundación BBVA - Severo Ochoa VHIO International Masters Internship Programme 

VHIO launches the annual  Master Internship Programme for excellent and motivated students that wish to do the Master Thesis @ VHIO. Apply now to boost your scientific career in a Translational Research Center of Excellence.

In this call, we do offer 6 internships for international life sciences Master students. Deadline 8th of December 2023

Decide which Masters Project fits better your research interests
and APPLY through our online application portal
Project
Supervisor(s)
Group
Towards improved patient selection in borderline-resectable pancreatic ductal adenocarcinomas via liquid biopsy and radiomics integration Tian Tian, MD. PhD Noncolorectal Gastrointenstinal Cancer Translation Research Group
Unlocking the Potential: Investigating Senescent Cell Metabolism to Combat Cancer Mate Maus, PhD Aging and Cancer Research Group
Transcriptomic signatures in metastatic breast cancer José Antonio Seoane, PhD Cancer Computational Biology Group
Generation of murine tumor cells expressing human HER2 and p95HER2 for the characterization of murine CAR T Cells Joaquín Arribas López, M.D PhD Growth Factors Group
Identification of composite genomic biomarkers of upfront resistance to Immune CheckPoint Inhibitors across human cancers Fran Martínez-Jiménez, PhD Computational Immunogenomics Group
Investigating the mechanism of action of Omomyc-based MYC inhibitors Laura Soucek, PhD Models of Cancer Therapies Group
Personalizing Radiotherapy: Unraveling Germline Genetics and MicroRNAs for Enhanced Tumor Control Lara Nonell, PhD | Sara Gutiérrez-Enríquez, PhD Bioinformatics Unit | Hereditari Cancer Genetics Group
Understanding genomic and genetic determinants of tumor evolution upon driver kinase inhibition César Serrano, M.D. PhD Sarcoma Translational Research Group
Discovery of biomarkers and clonal evolution analysis in tumors under immunotherapy using longitudinal tumors and plasma ctDNA Rodrigo Toledo, PhD Biomarkers and Clonal Dynamics Group

Candidates should fulfil the following eligibility criteria at the time of the call deadline:

  • Master students in Life Sciences or related subjects (e.g., Bioinformatics, Mathematics, Medicine, Biochemistry, Chemistry, Physics, etc) with an excellent academic record and a strong commitment to scientific research
  • Enrolled in the Master Programme of their choice. Candidates may not have received a master’s degree at the time of the Internship
  • High level of English
  • Availability to do a student placement/internship agreement through the correspondent University

Selected candidates will receive a monthly stipend of 650€ for five months during the first semester of 2024 ( Jan/Feb – Jun/Jul). The fellows will sign an agreement between the University and VHIO which will include insurance coverage.
All fellows will have access to an exceptional scientific environment, including outstanding equipment and facilities, together with a wide range of training opportunities, including access to seminars and some training workshops.

Interested candidates should apply via the VHIO online form including the following information:

  • Personal and academic data, indicating the master in which the applicant is enrolled
  • BSc certified academic record
  • Letter of motivation, highlighting your interest towards a specific Masters project
  • Brief summary of previous research experience (if any)
  • Applicants should select up to two research groups/projects

Eligible applications will be assessed based on the information provided in the application form by an internal evaluation committee. Short-listed candidates might be invited for an interview with the group leader who has shown interest in the applicant.

  • Call opening: 3 November 2023
  • Call deadline: 8 December 2023
  • Selection process: Early January 2024
  • Incorporation: Early February 2024
Days
Hours
Minutes
Seconds

PROJECT DESCRIPTION

Towards improved patient selection in borderline-resectable pancreatic ductal adenocarcinomas via liquid biopsy and radiomics integration | Tian Tian, MD. PhD

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with limited curative options. In cases of borderline-resectable PDAC, patient selection for neoadjuvant therapy and surgery is challenging, and assessing treatment response remains problematic. To address these issues, we investigate the role of extracellular vesicle (EV) DNA mutational profiles in PDAC plasma samples, in conjunction with radiomic patterns, to improve patient selection. Our objectives include characterizing EV profiles, determining mutational profiles of EV-DNAs, and correlating these profiles with patient radiomic data. This study offers a promising avenue for more effective patient selection in neoadjuvant therapy for borderline PDACs, potentially improving outcomes in the battle against this aggressive cancer.

Unlocking the Potential: Investigating Senescent Cell Metabolism to Combat Cancer | Mate Maus, PhD

Aging is intricately linked to the accumulation of senescent cells, which, in turn, foster inflammation, disrupt tissue repair, and suppress the body’s anti-tumor defenses, creating thereby a cancer-permissive environment. Recent insights reveal that senescent cells, through their senescence-associated secretory phenotype (SASP), play a pivotal role in driving tumorigenesis and fueling cancer relapse. Senescence is triggered when cells encounter irreparable DNA damage, whether due to aging, injury, or other insults. Senescent cells exhibit profound alterations in their metabolism, and our recent research has unveiled that these changes are at the heart of their SASP. This project sets out to dissect the mechanisms that link the metabolic profile of senescent cells to their pathological phenotype. If you’re ready to make a difference in the fight against cancer by exploring these research questions, consider joining us. For more details, visit https://www.mauslab.org

Transcriptomic signatures in metastatic breast cancer | José Antonio Seoane, PhD

Most of the breast cancer transcriptomic signatures developed to categorize breast cancer tumors have been developed using primary breast cancer patients. In this project we will evaluate whether the transcriptomic signatures developed for primary breast cancer are also valid for metastatic breast cancer. In order to do this, we will use computational methods and public available data, together with different algorithms for calling the subgroups and signatures. This is a fully computational project and requires bioinformatics or computer science expertise.

Generation of murine tumor cells expressing human HER2 and p95HER2 for the characterization of murine CAR T Cells | Joaquin Arribas, PhD

Our study will investigate whether murine Chimeric Antigen Receptor (CAR) T cells designed to target human HER2 and/or p95HER2 can effectively eliminate murine E0771 tumor cells expressing HER2/p95HER2. The project’s objectives include transducing E0771 murine cells with human p95HER2 and HER2 constructs to create cell lines with expression of HER2/p95HER2. Flow cytometry and Western blot techniques will be used to quantify protein expression. Murine CAR T cells, specifically targeting HER2 and/or p95HER2, will be developed and tested in in vitro cytotoxicity assays. In addition, transduced E0771 cells will be implanted in C57BL/6 mice to monitor tumor growth allowing for further evaluation of the in vivo cytotoxicity of murine CAR T cells. This project will shed light on the potential of murine CAR T cells to target murine tumor cells expressing human HER2/p95HER2, which is essential for posterior safety analyses of the therapy.

Identification of composite genomic biomarkers of upfront resistance to Immune CheckPoint Inhibitors across human cancers | Fran Martínez-Jiménez, PhD

Immune Checkpoint Inhibitors (ICIs) have transformed the treatment of certain high mutation burden tumors, but response rates are still suboptimal for the majority of them. Identifying biomarkers for ICI resistance is therefore crucial to optimize treatment regimes for patients that are unlikely to respond. This study aims to leverage state-of-the art data analysis and machine learning techniques to identify composite biomarkers of upfront resistance to immunotherapy treatment across cancer types. Implementing these biomarkers is a feasible way to enhance the quality of life for patients who do not respond positively to ICI treatment.

Investigating the mechanism of action of Omomyc-based MYC inhibitors | Laura Soucek, PhD

MYC is a critical player in cancer development and progression, making it a compelling target for therapeutic interventions in cancer. The recombinant mini-protein Omomyc, a dominant negative of MYC, is the first ever direct MYC inhibitor to successfully complete a Phase 1 clinical trial. To continue expanding our knowledge about the intricacies of MYC inhibition (MYCi) in different contexts, we will take advantage of our preclinical RNA-seq data to hypothesise on the mechanistic aspect of MYCi, with a primary emphasis on senescence-related changes. Furthermore, we are initiating the characterisation of a new therapeutic approach based on the delivery of mRNA designed to inhibit MYC. Making use of cell culture assays and a reporter system, we will screen for the best dose regimen. Additionally, we will determine the half-life of the expressed proteins resulting from the mRNA transcription and their subcellular localisation. Ultimately, if time permits, we will evaluate their biodistribution and therapeutic efficacy using xenograft and syngeneic mouse models of ovarian cancer.

Personalizing Radiotherapy: Unraveling Germline Genetics and MicroRNAs for Enhanced Tumor Control | Lara Nonell, PhD | Sara Gutiérrez-Enríquez, PhD

The variability in radiotherapy benefits among patients underscores the need for robust predictive markers to guide treatment and prevent disease recurrence. The role of germline genetic variation in modulating radiotherapy outcomes remains poorly understood. Our main goal is to personalize radiotherapy by investigating the germline genetic susceptibility underlying tumour response to radiotherapy.    The specific objectives are: (i) to assess the impact of germline genetic variation on radiotherapy efficacy through a genome-wide association study (GWAS) generated with an Illumina OncoArray available from 3600 Breast Cancer and Prostate Cancer patients; (ii) to study the whole blood microRNA expression profile in relation to radiotherapy efficacy using RNA sequencing data, and (iii) to create a risk score associated with radiotherapy efficacy. In addition, microRNA-eQTL analysis will be performed to assess the impact of genetic variants in miRNA expression. This project will be performed by exploiting the dataset from two previously funded European projects.

Understanding genomic and genetic determinants of tumor evolution upon driver kinase inhibition. | César Serrano, M.D. PhD

There is still an incomplete understanding of tumor cell evolution resulting from the selective pressures exerted by targeted inhibition of critical oncogenic drivers. Besides the intrinsic biological complexity, this is largely due to the paucity of reliable laboratory models mirroring the clinical reality.  Gastrointestinal stromal tumor (GIST) is the most common neoplasm of mesenchymal origin featured by the oncogenic activation of KIT or PDGFRA receptor tyrosine kinases. How these tumors progress to the five targeted therapies currently approved? To answer this question, and in the context of an ongoing multicohort- and multiomic-driven research project, we are generating and characterizing novel cellular models in two directions: a) determination of the numerical and structural patterns of chromosomal instability as well as their evolution in the context of kinase inhibition; b) generation of a clinically-representative repertoire of the subclonal variety identified in GIST patients so far. Multiple classical and cutting-edge techniques will be used for this purpose.

Discovery of biomarkers and clonal evolution analysis in tumors under immunotherapy using longitudinal tumors and plasma ctDNA | Rodrigo Toledo, PhD

The Biomarkers and Clonal Dynamics Lab at the Vall d’Hebron Institute of Oncology (VHIO), led by Dr. Rodrigo Toledo, focuses on developing new biomarkers for cancer by investigating the clonal dynamics of tumors using longitudinal liquid biopsy of patients during oncological treatment. In 2023, the lab published a paper in the journal Nature Medicine describing a new biomarker for predicting response to FDA-approved targeted in colorectal cancer. Our ultimate goal now is cancer immunotherapy. The lab is currently looking for highly motivated Master’s and PhD students to join their team. The ideal candidates will have a strong background in bioinformatics, computational biology, mathematics, or a related field. The students will have the opportunity to work on cutting-edge research projects using multi-omics data from tumor and plasma cell-free DNA samples of a large cohort of patients with metastatic tumors treated with immune-checkpoint inhibitors.

Privacy Preferences
When you visit our website, it may store information through your browser from specific services, usually in form of cookies. Here you can change your privacy preferences. Please note that blocking some types of cookies may impact your experience on our website and the services we offer.