José A. Seoane is a cancer computational biologist with international experience in the UK and the US. His background is computer science (Ingeniero Técnico and Ingeniero en Informática at the University of A Coruña, UDC, Spain), and he was later trained in data science (Master in Statistical Learning and Data mining, UNED and Expert in Applied Statistical Methods, UNED). In 2012 he read his PhD dissertation at the University of A Coruña, supervised by Julian Dorado (UDC) and Fernando Martin, Institute of Health Carlos III (ISCIII), funded by a Xunta de Galicia Isabel Barreto Fellowship. His thesis, focused on different models of biomedical data integration, was supported by an Extraordinary PhD Award, Computer Science and Mathematics section, University of A Coruña.
In 2012 he started his first postdoc at the School of Social and Community Medicine, the University of Bristol, under the supervision of Tom Gaunt (Bristol Genetic Epidemiology Lab) and Colin Campbell (Intelligent Systems Lab), where he studied bioinformatics, genetics and genomics, as well as machine learning. During this time, he first authored two papers in the most important journals in bioinformatics, Bioinformatics and PLoS Computational Biology, and he started working with breast cancer datasets, which piqued his interest in cancer systems biology. He then subsequently moved to Christina Curtis’ lab at University of Southern California, Los Angeles, and later to Stanford University, Stanford (CA, USA), first as postdoc and later as Instructor.
During his time at Stanford University, he worked on cancer systems biology and cancer epigenetics, particularly on how chromatin remodeling units’ somatic alterations affects to cancer progression and drug resistance. Partnering with Gerald Crabtree (Stanford University and Howard Hughes Medical Institute Investigator), he proposed a project to follow up on previous findings reported by the Crabtree lab, showing that alteration in SWI/SNF complex alter TOP2A binding.
They hypothesized that since TOP2A is the major target of anthracyclines, modification of chromatin regulator genes could alter drug resistance in breast cancer. The project was funded by Susan G. Komen Foundation under Curtis and Crabtree’s mentorship, as well as others (DoD, PIs Curtis and Crabtree). These results were reported in Nature Medicine. During this time, he also participated in the international analysis team of some Cancer Genome Atlas projects (Esophageal, Gastrointestinal and ATAC-Seq). At Stanford, he had the opportunity to collaborate with top tier cancer researchers including Dennis Slammon (UCLA) and Mike Press (USC) (pioneers in the discovery of role of Her2 amplification in cancer), Carlos Caldas (Cambridge), Mike Snyder (Head of Genetics Department), Laura Attardi, Monte Winslow, Rob West, Calvin Kuo, Howard Chang, Julien Sage, Mike Bassik and Mike Clarke at Stanford. Results from these collaborations published in Nature (4), Science, Cancer Cell (2), PNAS, Molecular Cell, Cancer Discovery (2), JAMA Oncology, and Nature Genetics (2). In 2021 he was awarded with a Ramon y Cajal Fellowship, which supports his appointment as a Group Leader at the Vall d´Hebron Institute of Oncology (VHIO), as of May 2021.
Jose A. Seoane joined VHIO this year to set up the newly established Cancer Computational Biology Group. Our team leverages epi(genetic) cancer datasets to unmask the molecular mechanisms implicated in cancer initiation, progression, drug resistance and metastasis towards improved outcomes for patients.
We aim to advance insights into the role of chromatin regulatory elements in treatment response and metastasis, develop novel epigenetic synthetic lethality-based therapeutic options, improve patient stratification guided by multi-omics analysis, and seek out novel epigenetic biomarkers of treatment response.
We have previously shown how the genetic modifiers of chromatin structure are associated with chemotherapy resistance in breast cancer (Seoane et al. 2019)*. Our group aims to establish how these epigenetic alterations affect drug resistance and how epigenetic therapies can be used to target tumor suppressor genes.
We are also exploring how the machine learning-based integration of multi-omic datasets can facilitate the discovery of new cancer subgroups and biomarkers, as well as help to better predict treatment outcomes and drug response.
Our group has participated/participates in multiple international consortia including The Cancer Genome Atlas, the Human Tumor Atlas Network, Cancer Target Discovery and Development (CTD 2) Network, and, most recently, AURORA (metastatic breast cancer multi-omic cohort).
María Abad carried out her predoctoral studies at the Institute for Biomedical Research (IIB, CSIC-UAM, Madrid), during which time she identified the tumor suppressor ING1b as a critical regulator of cellular senescence. She was awarded her PhD in Biochemistry, Molecular Biology and Biomedicine by the Universidad Autónoma de Madrid in 2008.
In 2009, she joined Manuel Serrano’s lab as a Postdoctoral Fellow at the Spanish National Cancer Research Centre (CNIO - Madrid), where her research centered on stem cells and cellular reprogramming. During this period, she demonstrated for the first time that cellular reprograming in living organisms is feasible, and triggers the acquisition of totipotency features. This work, published in Nature, was considered a Notable Advance of the Year 2013 by Nature Medicine, receiving several prizes and coverage in various scientific journals.
After this first report, the group discovered an interesting crosstalk between tumor suppressors, damage-induced cellular senescence, and in vivo reprograming – the findings of which were recently published in Science.
In 2014, María joined a world leader in the field of tissue regeneration, E. Olson, at the UT Southwestern Medical Center (Dallas, USA), where she studied the role of cellular dedifferentiation in heart repair, and revealed the importance of Notch signaling pathway during cardiomyoctes reprogramming.
In October 2016, she set up her own laboratory and VHIO´s Cellular Plasticity & Cancer Group, focusing on the interplay between stem cells, cellular plasticity and cancer.
She has received several prizes and awards, including the prestigious Ramón y Cajal Grant for young investigators and a Fero Foundation Fellowship. Her research has also been awarded with grants from the Spanish Ministry of Economy and Competitiveness (MINECO), “La Caixa” Foundation (Health Research Program), and the Health Research Grant from Mutua Madrileña Foundation, among others.
We focus on the interplay between stress responses, cellular plasticity and cancer. Cellular plasticity is now recognized as a critical feature of cancer cells, enabling them to transit between different cellular states and promote tumor growth, disease progression after therapy, and metastasis.
Our group has previously reported that inducing dedifferentiation with the so-called Yamanaka factors can lead to the development of a variety of tumors. We have also demonstrated that tissue damage -the main driver of cancer- triggers the onset of cellular senescence which then induces dedifferentiation and the acquisition of stem cell properties in vivo.
These findings have important therapeutic implications given that chemotherapy and radiotherapy – cornerstones for the treatment of most cancers – could have the side effect of inducing stemness in non- stem cancer cells and, in turn, possibly contribute to tumor recurrence and cancer cell spread.
Our main objective is to advance insights into the mechanisms and players implicated in this process, with the ultimate goal of developing novel therapies based on the inhibition of cancer cell plasticity.
Recent results have demonstrated that some genomic regions, previously considered as non-coding (including lncRNAs), contain small open reading frames encoding for evolutionary conserved, unannotated microproteins. The few that have been identified to date assume key functions in elemental cellular processes, leading to a new level of complexity with major implications – from basic research to the clinical setting.
Over the past five years, our efforts have focused on identifying and characterizing novel cancer microproteins which could be novel actors in carcinogenesis. We have discovered five new cancer microproteins and have obtained compelling evidence in vitro and in vivo that four of them act as novel tumor suppressors, inducing cell cycle arrest, differentiation or inhibition of mesenchymal traits in cancer cells. In addition, using a peptidomics approach, we have identified a set of microproteins secreted by pancreatic tumors, either soluble or secreted in exosomes. These novel microproteins could be crucial cellular messengers for pancreatic cancer metastasis.
The identification of tumor-microproteins could be key to advancing insights into cancer physiopathology. Moreover, they could also serve as novel cancer biomarkers for the early detection of disease and patient stratification for tailored therapies, as well as therapeutic targets.
In 2021, we have continued to characterize our identified cancer microproteins and have published our work on pTUNAR, a microprotein that regulates neural differentiation through the modulation of calcium dynamics (Senís et al. 2021). In addition, we have contributed a book chapter on the role of Myc in in vivo reprogramming (Senís et al. 2021). Please see our paper pick for 2021 below.
Finally, we have embarked on a collaborative project to study therapy-induced tumoral senescence and its potential as a therapeutic target and prognostic biomarker, funded by the Asociación Española Contra el Cáncer - AECC (Spanish Association Against Cancer).
VHIO’s Chromatin Dynamics in Cancer Group is comprised of 2 postdocs (1 MD PhD, 1 PhD), 3 predocs, 1 Masters student, and 1 technician (with a 70% female:30% male ratio).
Our projects focus on chromatin dynamics, epigenetics, epithelial-to-mesenchymal transition (EMT) and cancer, are highly ambitious yet successful to-date thanks to close collaboration and connectivity between myself and my group members.
We provide multidisciplinary training and guidance to our PhD students, exposing them to a wide array of techniques and experimental systems throughout their studies. They also attend international meetings of excellence throughout the course of their training.
Our lab was previously located at the Parc de Recerca Biomèdica de Barcelona (PRBB) – a hub of several independent research centers. The proximity to other cutting-edge research institutes facilitated our interaction and collaboration with different groups – partnerships that we that we continue to uphold. Additionally, we still have access to all PRBB’s core facilities including the Confocal and the Ultrasequencing Units as well as the Animal, Proteomics, and Microarray Facilities.
Since Sandra Peiró’s incorporation at VHIO in 2016, her group has published two research papers and has established major collaborations with medical oncologists from the Vall d’Hebron University Hospital (HUVH). Thus, in addition to basic research, her team has initiated three different pre-clinical/translational studies in collaboration with medical oncologists from our Hospital, with financial support received from the ”la Caixa” Foundation, and the pharmaceutical companies BeiGene, Epigene, and Zenith.
Peiró’s group also has multidisciplinary expertise in molecular biology and genome-wide studies:
Our laboratory seeks to better understand how epigenetics and chromatin structure and dynamics affect cell behavior, with specific focus on cancer. Through our comprehensive studies, we aim to dissect the role of epigenetic changes in cancer, identify mechanisms of response and resistance to anti-cancer medicines, and explore new therapeutic opportunities.
Over the last few years, we have elucidated epigenetic changes during EMT and cancer progression, and discovered a new histone H3 modification (oxidized H3) enriched in heterochromatin that is implicated in chromatin condensation and the transition to a metastatic cell fate(Iturbide el al. 2015, Herranz et al. 2016, Cebrià-Costa et al. 2020)*. We have also discovered an important role for lamin B1 in the reorganization of 3D chromatin structure during EMT (Pascual-Reguant et al. 2018)**.
Dedicated to fully applying these insights to the epigenetic landscape and 3D structure during this malignant transformation, we have adopted chromosome conformation–based techniques together with ChIP-seq, ATAC-seq and RNA-seq. By combining these data with excellent computational and statistical tools in standard cancer models, such as cancer cell lines, and in a large and unique collection of patient-derived xenograft (PDX) models, we will continue to navigate this largely uncharted area which shows great promise in the early diagnosis of disease.
We are equally committed to describing the association of chromatin conformation modifications with the acquisition of malignant traits and evaluating the functional consequences of these developments in genes and pathways. Next steps will focus on deciphering how these alterations occur at the molecular level and more precisely identifying these putative culprits for future targeted therapy.
* Iturbide A, Pascual-Reguant L, Fargas L, Cebrià JP, Alsina B, García de Herreros A, Peiró S. LOXL2 Oxidizes Methylated TAF10 and Controls TFIID-Dependent Genes during Neural Progenitor Differentiation. Mol Cell. 2015 Jun 4;58(5):755-66.
Herranz N, Dave N, Millanes-Romero A, Pascual-Reguant L, Morey L, Díaz VM, Lórenz-Fonfría V, Gutierrez-Gallego R, Jerónimo C, Iturbide A, Di Croce L, García de Herreros A, Peiró S. Lysyl oxidase-like 2 (LOXL2) oxidizes trimethylated lysine 4 in histone H3. FEBS J. 2016 Dec;283(23):4263-4273.
Cebrià-Costa JP, Pascual-Reguant L, Gonzalez-Perez A, Serra-Bardenys G, Querol J, Cosín M, Verde G, Cigliano RA, Sanseverino W, Segura-Bayona S, Iturbide A, Andreu D, Nuciforo P, Bernado-Morales C, Rodilla V, Arribas J, Yelamos J, de Herreros AG, Stracker TH, Peiró S. LOXL2-mediated H3K4 oxidation reduces chromatin accessibility in triple-negative breast cancer cells. Oncogene. 2020 Jan;39(1):79-121.
** Pascual-Reguant L, Blanco E, Galan S, Le Dily F, Cuartero Y, Serra-Bardenys G, Di Carlo V, Iturbide A, Cebrià-Costa JP, Nonell L, de Herreros AG, Di Croce L, Marti-Renom MA, Peiró S. Lamin B1 mapping reveals the existence of dynamic and functional eurchromatin lamin B1 domains. Nat Commun. 2018 Aug 24;9(1):3420.
The two main goals of our laboratory are as follows:
Specifically:
Violeta Serra obtained her PhD in 2001 in the field of cellular aging and telomeres from Newcastle University (UK). She gained a Marie Curie Postdoctoral Fellowship, to study cancer chemotherapy resistance at the Humboldt University, Berlin (Germany). In 2004 she moved to the Spanish National Cancer Research Center (CNIO, Madrid), where she pursued her interest in understanding the role of intracellular kinases as potential drug targets in cancer.
In 2006 Violeta joined José Baselga’s group at the Vall d’Hebron Institute of Oncology (VHIO), to explore the mode of action and mechanisms of resistance to PI3K-pathway inhibitors. Her work has been pivotal in defining adaptive responses to these agents in breast cancer cells.
Since 2014 she has led VHIO´s Experimental Therapeutics Group, and continues to expand her research into targeted therapies in triple negative breast cancers – PARP inhibitors in particular.
To establish herself as an independent Principal Investigator, Violeta was awarded with two Instituto Salud Carlos III (ISCII) Project Grants, a Career Catalyst Research Grant from the Susan G. Komen Foundation and from the Breast Cancer Now Catalyst Program.
Violeta is the coordinator of a European project funded by the ERA PerMed program named RAD51predictand is a partner of the H2020 MESI-STRAT consortium.
She is also member of the American Association of Cancer Research (AACR), the European Association of Cancer Research, serves on the Editorial Board of Clinical Cancer Research, and is also an ad-hoc reviewer for multiple journals and Granting agencies.
Our group conducts bench-to-bedside preclinical research in breast cancer to advance insights into biomarkers of response to targeted therapies. To do so, we generate preclinical models including patient-derived xenografts (PDXs) and patient-derived cultures (PDCs) from breast cancer patient samples.
We have significantly contributed to the field of PI3K inhibitor resistance and continue to more deeply explore mechanisms of resistance to CDK4/6 inhibitors, FGFR inhibitors, AKT inhibitors and AR modulators (SARMs) in breast tumors.
Additionally, we are exploring the potential of a novel HER3-targeted antibody-drug conjugate (ADC) as a therapeutic strategy for advanced breast cancers that have developed resistance to the current standard of care treatments.
Using clinically relevant PDXs, we have provided data to further support that the loss of G1-cell cycle checkpoint control, such as mutation/loss of RB1 or CCND1- amplification, is associated with the lack of response to CDK4/6 blockade in estrogen receptor-positive breast cancer. Additionally, we generated a collection of PDXs containing FGFR amplification to study biomarkers of sensitivity to FGFR inhibitors; both pan-FGFR1-4 and Multi-targeted Tyrosine Kinase Inhibitors (MTKIs).
Encouraged by the early success of DNA damage repair inhibitors in germline BRCA1/2 mutated tumors, we initiated a project aimed at identifying response biomarkers of PARP inhibitors (PARPi) as well as other DNA damage repair inhibitors including those targeting WEE1 or ATR.
Our studies underpin the capacity of germline BRCA mutant tumors to recover DNA repair functionality and develop resistance to PARPi. We have developed an assay, the RAD51 test, which accurately identifies germline BRCA tumors that have restored DNA repair functionality and become resistant to these drugs. Importantly, this test also identifies tumors that are sensitive to PARPi through its alterations in DNA repair by homologous recombination beyond the germline BRCA condition. We filed a patent (EU application in 2017 and PCT in 2018), and we are currently validating the use of this test in tumor samples from breast, ovarian, and prostate cancer patients.
Finally, we are also investigating the effects of PARPi on the tumor immune environment. DNA repair-deficient tumors accumulate cytosolic DNA, which can elicit an innate immune signal (the STING pathway) and upregulate interferon-related genes, leading to lymphocytic infiltration and PD-L1 expression. We are testing the hypothesis that treatment of DNA repair-deficient tumors with PARPi elicits a DNA damage response, resulting in upregulation of PD-L1 that might limit the antitumor immune-mediated cytotoxicity by lymphocytes, but sensitizes to anti-PD-L1 treatments.
Our group works closely together with Cristina Saura’s Breast Cancer Group, and Judith Balmaña’s Hereditary Cancer Genetics Group. Reflective of VHIO’s purely multidisciplinary and translational approach, our research is also carried out in collaboration with other groups including VHIO’s Molecular Oncology Group, and Oncology Data Science – OdysSey Group, directed by Paolo Nuciforo and Rodrigo Dienstmann, respectively.
Our team has significantly advanced insights into the mode of action of novel targeted therapies, identified new response biomarkers, and developed a biomarker-based assay with potential clinical application. We have also demonstrated the efficacy of hypothesis-based drug combinations.
Upon completing his post-doctoral studies in 2004, Joan decided to establish his own group pursuing an independent research project. Since he believed that cancer can only be combated through combining basic and clinical research efforts, Joan sought to set up his laboratory as closely as possible to a clinical research unit in order to fluently interchange findings in basic research with the clinical ambit and hence carry out translational research. For this very reason, he chose to join a research institute linked to a hospital – the Vall d’Hebron Institute of Oncology (VHIO), which is both situated at the heart of the Vall d’Hebron University Hospital – one of the biggest hospitals in Spain, and also has a Medical Oncology Department with an outstanding clinical trial program and portfolio in solid tumors. VHIO therefore provided Joan with the optimal setting and environment through which to deliver on his objectives.
He was appointed as a Group Leader at VHIO and ICREA Research Professor in 2004, and decided to focus his research on glioblastoma as a model to study cancer. Specifically, Joan wanted to tackle one of the most important challenges in the treatment of cancer: tumor heterogeneity.
Cancer is a disease with two levels of heterogeneity, an intertumor heterogeneity (each patient has a different tumor) and an intratumor heterogeneity (cells within a tumor are different). Tumors from different patients are molecularly diverse and have different sensitivities to treatment. However, there is another level of complexity. Cells within a tumor are diverse with different genomic alterations or different states of differentiation. This has very important clinical implications since a treatment might target one cell type but not others – therefore being unable to cure.
Linked to the intratumoral heterogeneity concept, a subpopulation of undifferentiated cells with stem cell-like characteristics has been identified within the tumor mass. This pool of cells, called cancer-initiating cells (CICs) or cancer stem cells, are considered to be responsible for the initiation, recurrence and chemo- and radio-resistance of tumors. CICs are, therefore, crucial therapeutic targets and better understanding of the molecular mechanisms involved in this type of cells is still needed.
In order to study tumor heterogeneity, cancer must be studied as close as possible to the real tumor of the patient. In 2005, Joan set up a multidisciplinary team incorporating oncologists, neurosurgeons and pathologists. For the first time at the Vall d’Hebron University Hospital, he managed to establish a ‘circuit’ through which to obtain human specimens from the surgeons and pathologists to generate tumor models that recapitulate the characteristics of the human tumor of origin. Tumor samples were obtained 15 minutes after surgery and cell populations from the tumor such as CICs were studied.
Moreover, CICs are orthotopically inoculated in the brain of mice to generate tumors that recapitulate the characteristics of the original human tumor. Hence, Joan’s team regenerated patients’ tumors in mice to study the molecular characteristics of tumors and, more importantly, identify the optimal treatment tailored to the specificities of individual disease.
Using these models, Joan has generated important data in glioblastoma and cancer stem cells with important implications at the clinical level. He has three patents and an outstanding publication record with articles published in high impact journals including Cancer Cell, Nature Medicine, Cancer Discovery, and Nature Communications, among others, as corresponding author.
Joan’s team has identified therapeutic targets against glioblastoma; identified markers of response to treatment with anti-TGFβcompounds improving the design of clinical trials by providing markers to more precisely stratify patients; and developed novel strategies by liquid biopsies to characterize brain tumors.
This work epitomizes the essence of translational research.
Appointed as an ICREA Professor in 2004, Joan continues to serve as an Associate Professor of the Universitat Autònoma de Barcelona (UAB) – since 2008. He is a Member of the Executive Committee of the European Association for Cancer Research (EACR), as well as Member of the Neuro-oncology Committee of the Vall d’Hebron University Hospital (HUVH).
His contributions to the field have been recognized by various awards and honors including the Memorial Sloan Kettering Cancer Center (MSKCC) Research Fellow Award, and the Catalan Society of Biology Award. In 2007 Joan became an EMBO Young Investigator, and in 2009 he received both the Sabadell Bank Award and the Sociedad Española de Bioquímica y Biología Molecular Award. He was received a European Research Council (ERC) Starting Grant in 2008.
In 2011 he was appointed as Director of VHIO’s Translational Research Program, and in 2013 he was awarded with the Dr. Josef Steiner Cancer Research Award, also receiving the prestigious Doctores Diz Pintado National award as well as the Spanish periodical La Vanguardia´s prize for science in 2016.
Joan is also Founder of VHIO born spin-off Mosaic Biomedicals. Through this Company, one of his discoveries has already translated into anti-cancer agents that are being clinically tested in patients.
We study primary brain tumors and brain metastasis; some of the most aggressive of all cancers. Both glioblastoma and brain metastasis are dismal diseases with limited therapeutic options. Advancing progress in this field toward improving outcomes for these patients is therefore critical.
Our group is designing tools to monitor and characterize brain tumors, with specific focus on brain metastasis and central nervous system lymphomas. By analyzing immune cells in cerebrospinal fluid, we discovered that we could obtain predictive biomarkers of response to immunotherapy without having to perform a majorly invasive intracranial biopsy (Rubio-Perez et al. 2021). Moreover, we have shown that cell free circulating tumor DNA in cerebrospinal fluid facilitates the monitoring and early detection of tumor relapse in central nervous system lymphomas (Bobillo et al. 2021).
While no biomarker derived from liquid biopsy against these tumor types has yet been integrated into clinical practice, mounting evidence reported in the literature, including our findings, points to its efficacy in the real-time evaluation of malignant disease and potential to better inform and guide the therapeutic management of patients. Reflective of our work in the field, we were invited to help elaborate the EANO – ESMO Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with brain metastasis (Le Rhun et al. 2021). It has been a privilege for us to collaborate alongside other renowned scientists and clinicians specialized in brain metastasis to set out these European clinical guidelines for the management of this dismal disease.
We are as committed to advancing research into the role of the tumor microenvironment which, in the case of brain cancers, assumes a crucial role in cancer progression. Advancing insights into the tumor microenvironment promises powerful weaponry in combating cancer, regardless of heterogeneity.
By eliminating the niche where tumors reside and thrive should enable us to develop more effective anti-cancer compounds. In this respect, we have reported that the cytokine LIF assumes an essential role in the tumor microenvironment and is consequently a promising therapeutic target.
We are now testing a novel agent MSC-1 (now AZD0171), a LIF neutralizing antibody, developed by VHIO spin-off, Mosaic Biomedicals, founded by Joan Seoane in 2012. Mosaic Biomedicals was acquired by Medimmune/Astrazeneca in 2020. A phase II clinical trial (NCT04999969) to test MSC-1 initiated patient recruitment in 2021.
Joaquín Arribas completed his undergraduate studies in biochemistry at the Universidad Autónoma de Madrid in 1987, where he subsequently worked on the regulation of the catalytic activities of the proteasome and was awarded his PhD in Biology in 1991.
Supported by a fellowship from the Spanish Ministry of Education and Science, he joined the Memorial Sloan-Kettering Cancer Center, New York (USA), as a Postdoctoral Fellow to work with J. Massagué (1992-1996) on the proteolytic processing of transmembrane growth factors.
In 1997, he joined the Oncology Department of the Vall d'Hebron University Hospital (HUVH) in Barcelona as a Staff Scientist and he has since led VHIO's Growth Factors Group. In 2001, he was promoted to lead the Oncology Research Program and later in 2007, he was appointed as Research Professor of the Catalan Institution for Research and Advanced Studies (ICREA). Since 2010 he has served as Director of VHIO's Preclinical Research Program.
His research has been recognized through an EMBO Young Investigator Programme (YIP) Award, the Beckman Coulter Award for the Best Young Spanish Investigator in Biochemistry and Molecular Biology, and grants from the Breast Cancer Research Foundation (BCRF) and the Spanish Association Against Cancer (AECC).
Joaquín is member of the Spanish and American Societies of Biochemistry and Molecular Biology as well as the American Association for Cancer Research (AACR). He has presided over the Committee for the evaluation of cancer research projects of the Instituto Salud Carlos III (ISCIII), and has served on the Executive Committee of the Red Territorial de Investigación Cooperativa en Cáncer (RTICC); the former main cancer research consortium in Spain for which he also coordinated the Breast Cancer Program.
In 2017, he was appointed as Director of the CIBERONC, the Spanish Cancer Research Consortium. In January 2020he was appointed as Research Director of the Mar Health Park/ Hospital del Mar Institute of Medical Research (PSMAR/IMIM), Barcelona.
2021 has been the second year of conciliation with the contingent limitations caused by COVID. However, our group has managed to continue working on all our research lines and we have produced significant results.
We have advanced insights into new mechanisms of resistance to targeted therapies. Firstly, we have demonstrated the role of the transcription factor SLUG in resistance to therapies directed against the RAF-MEK1/2-ERK1/2 pathway in pancreatic cancer (Bilal et al. 2021) Secondly, we have described a novel mechanism of acquired cancer cell resistance to T cell bispecific antibodies and CAR T targeting HER2 through JAK2 down-modulation (Arenas et al. 2021; Martínez-Sabadell et al. 2021).
Our group has also collaborated with VHIO’s Sarcoma Translational Research Group, directed by César Serrano, in the study of E3 ubiquitin ligase Atrogin-1 as a mediator of adaptative resistance to KIT-targeted inhibition in gastrointestinal stromal tumor (García-Valverde et al. 2021).
Other collaborations have led to the following discoveries reported this year:
1) DNA hypomethylating agent (HMA) treatment can directly modulate the anti-tumor response and effector function of CD8+ T cells (Loo Yau et al. 2021), 2) the genetic activation of MAPK as a recurrent mechanism of anti-HER2 therapy resistance that may be effectively counteracted with MEK/ERK inhibitors (Smith E. et al. 2021), iii) CAF-derived NRG1 mediates trastuzumab resistance through HER3/AKT, which might be reverted by pertuzumab (Guardia et al. 2021), iv) RANK signaling increases after anti-HER2 therapy contributing to the emergence of resistance in HER2-positive breast cancer (Sanz-Moreno et al. 2021), and v) PI3K activation promotes resistance to eribulin in HER2-negative breast cancer (Gris-Oliver et al. 2021).
For all these studies published in 2021 see PI Paper Pick plus footnotes* below.
Regarding unpublished results:
In the generation and characterization of CARs against tumor specific antigens, we have produced a fourth-generation CAR T cell that has shown promising results in vitro and in vivo. Once we have validated its efficacy and safety, this therapy will be tested in a first phase clinical trial.
Our group has continued to pursue research into the role of senescent cells in tumor progression. We have obtained significant results on how this differs according to tumor stage. This project has received funding from the Breast Cancer Research Foundation (BCRF) for two years.
Ariadna Grinyo, a PhD student supported by a ”la Caixa” Foundation INPhINIT Doctoral Fellowship-Incoming, joined our group in 2021. Ariadna has previous experience as a researcher of immune response at the biotechnology company Integral Molecular in Philadelphia (PA, USA), and has started her thesis on the generation of fourth-generation CARs against solid tumors.
* García-Valverde A, Rosell J, Sayols S, Gómez-Peregrina D, Pilco-Janeta DF, Olivares-Rivas I, de Álava E, Maurel J, Rubió-Casadevall J, Esteve A, Gut M, Valverde C, Barretina J, Carles J, Demetri GD, Fletcher JA, Arribas J, Serrano C. E3 ubiquitin ligase Atrogin-1 mediates adaptive resistance to KIT-targeted inhibition in gastrointestinal stromal tumor. Oncogene. 2021 Dec;40(48):6614-6626.
Smith AE, Ferraro E, Safonov A, Morales CB, Lahuerta EJA, Li Q, Kulick A, Ross D, Solit DB, de Stanchina E, Reis-Filho J, Rosen N, Arribas J, Razavi P, Chandarlapaty S. HER2 + breast cancers evade anti-HER2 therapy via a switch in driver pathway. Nat Commun. 2021 Nov 18;12(1):6667.
Guardia C, Bianchini G, Arpí-LLucià O, Menendez S, Casadevall D, Galbardi B, Dugo M, Servitja S, Montero JC, Soria-Jiménez L, Sabbaghi M, Peña R, Madoz-Gúrpide J, Lloveras B, Lluch A, Eroles P, Arribas J, Pandiella A, Gianni L, Rojo F, Rovira A, Albanell J. Preclinical and Clinical Characterization of Fibroblast-derived Neuregulin-1 on Trastuzumab and Pertuzumab Activity in HER2-positive Breast Cancer. Clin Cancer Res. 2021 Sep 15;27(18):5096-5108.
Sanz-Moreno A, Palomeras S, Pedersen K, Morancho B, Pascual T, Galván P, Benítez S, Gomez-Miragaya J, Ciscar M, Jimenez M, Pernas S, Petit A, Soler-Monsó MT, Viñas G, Alsaleem M, Rakha EA, Green AR, Santamaria PG, Mulder C, Lemeer S, Arribas J, Prat A, Puig T, Gonzalez-Suarez E. RANK signaling increases after anti-HER2 therapy contributing to the emergence of resistance in HER2-positive breast cancer. Breast Cancer Res. 2021 Mar 30;23(1):42.
Gris-Oliver A, Ibrahim YH, Rivas MA, García-García C, Sánchez-Guixé M, Ruiz-Pace F, Viaplana C, Pérez-García JM, Llombart-Cussac A, Grueso J, Parés M, Guzmán M, Rodríguez O, Anton P, Cozar P, Calvo MT, Bruna A, Arribas J, Caldas C, Dienstmann R, Nuciforo P, Oliveira M, Cortés J, Serra V. PI3K activation promotes resistance to eribulin in HER2-negative breast cancer. Br J Cancer. 2021 Apr;124(9):1581-1591.
Laura Soucek graduated “Cum Laude” in Biological Sciences at the University La Sapienza, Rome (Italy) in 1996, defending her thesis on MYC dimerization specificity. At the same University, she then worked on the design of a MYC inhibitor – Omomyc – and was awarded her PhD in Genetics and Molecular Biology in 2001.
That same year, supported by a fellowship from the Italian National Research Center (CNR), she joined the Comprehensive Cancer Center of the University of California San Francisco (UCSF, USA), where she worked until 2006 as a Postdoctoral Fellow on mouse models of MYC driven tumorigenesis under the mentorship of Gerard Evan.
In 2006 she was promoted as Assistant Researcher, the Pathology Department of the University of California San Francisco (UCSF), where she worked on pre-clinically validating MYC inhibition as an anti-cancer therapeutic strategy. During this time, she was awarded various prestigious prizes including the AACR Future Leaders, New Directions Award for outstanding early-career scientists in cancer research, and the Bear Necessities Pediatric Cancer Foundation Award.
Laura joined the Vall d´Hebron Institute of Oncology (VHIO), Barcelona, in 2011 as Principal Investigator to lead the Models of Cancer Therapies Group. In Spain her research has been recognized through a Miguel Servet Programme grant, FERO Foundation Fellowships, a grant for Emerging Research Group of Catalunya from the Agency for Management of University and Research Grants (AGAUR), a BBVA Foundation grant in Biomedicine, a Retos de Colaboración grant from the Spanish Ministry of Economy, Industry and Competitiveness and two awards from the Instituto de Salud Carlos III (Institute of Health Carlos III, ISCIII) for a FIS – Fondo de Investigación en Salud (Health Research Fund). At international level, she was also recipient of a Worldwide Cancer Research grant and a European Research Council (ERC – European Commission) Consolidator, as well as two Proof-of-Concept grants within the framework of the Horizon 2020 Program.
Laura is a Scientific Editor for the journal Cancer Discovery, Review Editor for Frontiers in Oncology - Molecular and Cellular Oncology, and Reviewer for Anti-cancer Drugs, BBA (Biochimica et Biophysica Acta), BioDrugs, BioEssays, Cancer Discovery, Cancer Immunology and Immunotherapy, Cancer Immunology Research, Cancer Research, Cancer Research Communications, Cancer Treatment Reviews, Cancers, Cell Chemical Biology, Cell Death & Disease, Clinical Cancer Research, Communications Biology, Current Pharmaceutical Design, Developmental Dynamics, Drug Discovery Today, E-Biomedicine, eLIFE, Frontiers in Cell and Developmental Biology, Frontiers in Oncology, Gastroenterology Research and Practice, GEN Biotechnology, Gut, International Journal of Biological Sciences, IJEP (International Journal of Experimental Pathology), Journal of Cellular Biochemistry (JCB), Journal of Experimental and Clinical Cancer Research, JoVE Peer Reviewed Scientific Video Journal – Methods and Protocols, Medicinal Research Reviews, Molecular Biology and Evolution, Molecular Biology Reports, Molecular Oncology, Molecules, Nature, Nature Cell Biology, Nature Communications, Nature Medicine, Oncogene, Oncogenesis, “Oncology” books iConcept Press Ltd, Oncotarget, Peptide Science, Plos One, Science Advances, Science Translational Medicine, Scientific Reports, Therapeutic Advances in Chronic Disease, Trends in Cancer, and Tumor Biology.
Laura was appointed as a Research Professor at the prestigious Catalan Institution for Research and Advanced Studies (ICREA) in 2014. That same year, together with Marie-Eve Beaulieu – formerly a Postdoctoral Fellow of her group – she co-founded the VHIO and ICREA spin-off company Peptomyc S.L. In 2015, she was also appointed as an Associate Professor at the Universitat Autònoma de Barcelona (UAB).
Laura is also a member of the directive Scientific Committee of VHIO as well as the FERO Foundation.
Our group focuses on the pleiotropic and ubiquitous MYC oncoprotein, whose deregulation is implicated in almost all human cancers. The technical challenges of targeting nuclear transcription factors such as MYC –and the concern regarding potential side effects– had until recently precluded any preclinical validation of MYC inhibition as a possible therapeutic strategy.
Over the past few years, we have demonstrated in several mouse models that MYC inhibition has a dramatic therapeutic impact across several tumor types, with very mild and reversible side effects in normal tissue.
Encouraged by our results in mice, we are now interested in developing viable, non-toxic pharmacological options for MYC targeting in the clinic. To do so, we created a spin-off company, Peptomyc S.L., for the development of MYC-inhibiting peptides for cancer therapy. Our laboratory, in partnership with Peptomyc, is currently validating our novel approach against notoriously difficult-to-treat cancers that are resistant to standard treatments and in dire need of new therapeutic avenues (i.e., KRAS-driven Non-Small Cell Lung Cancer, glioblastoma, and metastatic triple negative breast cancer). Notably, the first Omomyc-derived compound, OMO-103, entered clinical trials Phase I/IIa in May 2021.
Our group has continued to contribute to cancer research in general and, more specifically, as a leader in the MYC field, by (co) authoring articles and reviews, exploring new aspects of MYC biology in different pathologies and summarizing efforts to develop a clinically viable MYC inhibitor.
Joaquin Mateo is a physician-scientist focused in the development of new therapeutic strategies towards a more precise care of advanced prostate cancer patients.
Joaquin completed his Medical Degree at the Universitat de Barcelona and completed his specialist training in Medical Oncology at the Catalan Institute of Oncology (ICO). He joined the Drug Development Unit at the Institute of Cancer Research – Royal Marsden NHS Foundation Trust in 2011 for a two-year fellowship under the supervision of Johann de Bono and Stan Kaye, where he worked on first-in-man clinical trials of several PARP inhibitors and compounds targeting the PI3-AKT-mTOR pathway. From 2013, he progressively focused on the design and development of clinical trials for castration-resistant prostate cancer (CRPC), completing his PhD in Cancer Genomics at Johann de Bono’s laboratory at the Institute of Cancer Research (UK). Towards the end of 2017, he joined VHIO to launch a new research group focused on advanced prostate cancer, following a bench-to-bedside-and-back research approach.
His main interest centers on the integration of tumor molecular characterization data into real-time clinical decision-making for patients through multidisciplinary tumor boards, as reflected by his involvement in the setting up of the molecular tumor boards at the Drug Development Unit and Prostate Cancer Targeted Therapy Unit at The Royal Marsden.
Joaquin is currently Chair of the European Society for Medical Oncology’s (ESMO) Precision Medicine Working Group, and led the development of the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT). First published in 2018, ESCAT is a classification system to implement genomic testing towards better guiding treatment decision making; in 2020, the group published the first Recommendations document from a medical society for the implementation of next-generation sequencing in clinical practice.
While completing his PhD in the University of London, in 2016, he received a Prostate Cancer Foundation Young Investigator Award to continue working on the development of PARP inhibitors, platinum-based therapies and companion predictive biomarkers for CRPC patients. Later, he received awards from the European Commission MSCA Fellowship program (2018) and the CRIS Cancer Foundation Clinical Talent Award program (2021).
His significant contribution to the field has already resulted in his (co) authorship of several manuscripts published in top-tier journals including The New England Journal of Medicine, Clinical Cancer Research, Cell, European Urology, and Cancer Discovery. He has received research funding from competitive grants from the Prostate Cancer Foundation, the US Department of Defense Medical Research Program, the Instituto de Salud Carlos III– Spanish Ministry of Science and Innovation, the Spanish Society for Medical Oncology (SEOM), Fundación FERO, and the European Commission’s H2020 research program. In 2020, he obtained one of the prestigious Asociación Española Contra el Cáncer - AECC (Spanish Association Against Cancer) LAB Awards for emerging groups and he is part of the awarded consortium in the GRUPOS COORDINADOS-AECC 2021 call.
As Principal Investigator of VHIO’s Prostate Cancer Translational Research Group, Joaquin leads research aimed at translating prostate cancer genotypes into phenotypes and a clinically-relevant classification of disease. He also seeks to build a precision medicine core for prostate cancer patients in his clinical practice at the Vall d’Hebron University Hospital (HUVH).
Our research model follows a bench-to-bedside-and-back approach. We have set up a platform for acquiring longitudinal samples from advanced prostate cancer patients, both tumor tissue as well as liquid biopsies, that can be used to investigate the evolving features of the disease, but also to generate patient-derived laboratory models of advanced prostate cancer that we can leverage to investigate new therapeutic strategies at the bench.
Moreover, the launch of investigator-initiated clinical trials is central to our research strategy, as a platform for correlative studies that can optimize the drug development pathway for prostate cancer patients. At present, we are the central laboratory for two academic multi-center clinical trials, as well as serve as the national repository for the IRONMAN registry; an important international effort to build a large bank of clinical data and biospecimens from metastatic prostate cancer (mPC) patients.
We aim to integrate insights in molecular biology, genomics, transcriptomics, computational sciences and clinical data towards developing precision medicine strategies for prostate cancer patients. To this end, our team comprises biomedical scientists with expertise in cancer biology, genomics and transcriptomics, bioinformatics and liquid biopsy, as well as medical oncologists and clinical data managers.
One of our main lines of research is to decipher how prostate cancers adapt to exposure to systemic therapies, particularly androgen-targeting agents, with a particular focus on the cell cycle and DNA damage response regulation and the emergence of quiescent and senescent phenotypes that can drive drug resistance. By studying in-vitro and in-vivo models, including patient-derived xenografts (PDX) from the patients participating in our clinical studies, we aim to understand how to target emergent phenotypes through drug combinations. These studies are currently funded by grants from the Spanish Ministry of Health, Fundación FERO (FERO Foundation), Asociación Española Contra el Cáncer - AECC (Spanish Association Against Cancer), as well as through collaborations with biopharmaceutical companies.
Our group also aims to pursue the molecular characterization of advanced prostate cancer, focusing on how tumors evolve in a heterogeneous manner as they become resistant to different treatments. We have therefore set up a genomics and transcriptomics platform in the lab to perform DNA and RNA profiling from patient biopsies. We are developing novel liquid biopsies assays that will enable us to study the disease though longitudinal samples. By exploiting publicly available genomics datasets from patients at different stages of disease, our computational scientists investigate how the genomic profile of disease changes over time. These studies are currently funded by grants from the US Department of Defense's (DoD) Congressionally Directed Medical Research Programs (CDMRP), the Spanish Ministry of Health, CRIS Cancer Foundation, AECC, Fundación FERO, and the ”la Caixa” Foundation.
Lastly, given that well-annotated correlative clinical data is crucial to establish the potential relevance of molecular data, our team maintains databases collecting outcome data for all patients who donate samples for our studies to perform subsequent correlative analysis.
To more rapidly advance the field, we believe in team science. We therefore participate in several different collaborations that pool resources and combine cross border expertise across several different research teams and groups.
In summary, our research integrates different modalities of data generated in the lab that can lead to the design of therapeutic interventions aimed at improving outcomes for prostate cancer patients.
I seek to pursue my academic career as a physician scientist focused on sarcoma translational research, with an emphasis on the biological understanding of sarcomas towards advancing drug development. To this end, our clinical-translational program is devoted to unravelling the intrinsic mechanisms of sensitivity and resistance to anticancer agents through high-throughput genomic and transcriptome studies, as well as developing cross-cutting therapeutic strategies.
In previous studies performed during my subspecialty training in sarcomas, I performed translational research that involved 1) demonstrating the molecular basis for the failure of TKIs in GIST; 2) developing preclinical evidence for an innovative therapeutic strategy against disease heterogeneity based on rapid alternation of TKIs with complementary inhibitory activity pattern, and 3) translating these insights into a phase Ib/II clinical trial.
Since my return to VHIO, my laboratory’s focus has extended beyond kinase signal transduction research and, with equal emphasis, we participate in highthroughput NGS studies to ultimately better understand the critical biologic mechanisms that lead to the heterogeneous behavior observed in the clinic when using different therapeutic agents.
We are also dedicated to developing liquid biopsy in sarcoma. Additionally, our current whole genome and transcriptome studies center on deciphering novel mechanisms of resistance and therapeutic vulnerabilities, irrespective of KIT genotype. Our team also leads translational research to reveal the biological mechanisms behind metastatic GIST patients with a long-term response to imatinib.
Finally, my clinical activities as a Medical Oncologist involve sarcoma patient care and drug development, thus applying basic discovery from the lab to the clinic for the benefit of these patients. In 2020, I was involved in the international teams that led to the FDA approval of ripretinib and avapritinib in GIST patients.
Sarcoma encompasses >70 entities of mesenchymal origin, constituting 1-2% of all cancers. From a biological perspective, sarcomas can be classified into two broad categories: sarcomas driven by simple genetic alterations, such as translocations or specific activating mutations; and tumors with complex and unbalanced genomic aberrations. Both include diverse sarcoma subtypes often with profound differences in their molecular makeup, course of disease and therapeutic approach.
We focus on the study of sarcomas with oncogenic dependency on specific drivers of disease. Among these, gastrointestinal stromal tumor (GIST) is the most common malignant mesenchymal neoplasm and constitutes a paradigmatic model for studying oncogene addiction and identifying structural and functional mechanisms for drug response and resistance.
Ongoing efforts aim at achieving a deeper biological understanding of GIST and other sarcomas in order to advance drug development. The heterogeneity of mechanisms of resistance represents one of the major challenges in improving outcomes for these patients. We therefore seek to identify crucial molecules and signaling mechanisms in GIST biology that can serve as therapeutic vulnerabilities.
Our group also continues to validate a core set of molecules that are co-regulated by KIT downstream pathways and identified through extensive whole transcriptome studies across several clinically representative human GIST models.
We also perform high-throughput genomic and transcriptomic studies to decipher the evolving patterns of resistance in GIST throughout the course of disease, as well as investigate liquid biopsy in sarcoma to provide robust evidence that will help to more precisely guide treatment decisions by plasma sequencing. In addition, the GISTomics project, a European initiative driven by our group, aims to advance insights into the landscape of GIST evolution.
Beyond GIST, our group has initiated new lines of research focused on other sarcoma subtypes, including muscle-derived sarcomas (leiomyosarcoma and rhabdomyosarcoma), angiosarcoma, and liposarcoma. Functional precision medicine is a major focus of research in these neoplasms.
Our aim is to have a true clinical impact by improving the daily treatment and care of our patients. We are proud to report that our Sarcoma Multidisciplinary Unit has been designated as an Expert Sarcoma Center of the European Reference Network ERN-EURACAN, and thus constitutes an optimal setting for translating cancer discovery into clinical benefits.
Héctor G. Palmer obtained his PhD in Biochemistry and Molecular Biology from the Universidad Autónoma de Madrid in 2001 for his work at the Instituto de Investigaciones Biomedicas – IIB (CSIC-UAM, Madrid) under the supervision of A. Muñoz. During this time his studies focused on the anti-tumoral capacity of vitamin D analogues on human colon cancer cells.
From 2001 – 2003 he continued to work at the IIB as a Post-Doctoral Fellow exploring the crosstalk between vitamin D receptor and the transcriptional repressor Snail in colon cancer.
In 2003 Héctor was awarded a Marie Curie Intra European Fellowship, and in 2004 he joined the London Research Institute-Cancer Research UK (LRI-CRUK) as a Post-Doctoral Fellow under the leadership of F. M. Watt, during which time he described the vitamin D receptor as a novel transcriptional effector of the Wnt pathway that controls stem cell fate in adult epidermis. He also discovered that the central role of the Wnt signalling in tumor initiation depends on VDR function, opening a new avenue for the use of Vitamin D-based therapies to prevent the development of cancer.
In 2008 Héctor returned to Spain to join the Vall d’Hebron Institute of Oncology (VHIO) as Principal Investigator of the Stem Cells & Cancer Group. His group focuses on the relevance of persistent tumor cells in cancer self-renewal, drug-resistance, relapse, and metastasis.
VHIO’s Stem Cells & Cancer Group studies the mechanisms that enable tumors to evade effective treatments and progress to advanced stages of disease.
We use multi-omics approaches to reveal unexpected alterations related to tumor and single cell phenotypes. Combining gene editing (CRISPR/Cas) with classical signaling biochemistry in cancer cell lines as well as genetically modified mice, patient-derived organoids (PDO) and xenografts (PDX), our group investigates the functional relevance of these newly identified alterations in patients' response to therapies.
We participate in a global multidisciplinary task force incorporating medical oncologists, surgeons, radiologists, and nurses. This strong collaboration aims to rapidly translate laboratory results to the clinic.
Main research lines include:
Tumor cell dormancy
The study of the intriguing biology of cancer cell dormancy as a driver of chemoresistance, formation of minimal residual disease, and disease relapse in patients.
We previously discovered a core epigenetic network that governs dormancy of tumor cells (Puig et al. 2018)*, and are now investigating the function of TET2, DPPA3 and other epigenetic and transcription factors governing dormancy in greater depth. Importantly, our group is rapidly progressing in developing drugs that modulate dormancy drivers including TET2, and defining novel biomarkers to detect chemo-resistant dormant tumor cells (DTC).
Response to target-directed drugs
We work in close collaboration with oncologists and pharmaceutical companies to identify molecular mechanisms responsible for the sensitivity or resistance to drugs blocking Wnt/beta-catenin, Notch, PI3K/AKT, EGFR/LGR5 or BRAF/MEK/ERK, oncogenic signals (Tenbaum et al. 2012; Puig et al. 2013; Capdevila et al. 2020)**.
Based on our discoveries, we are designing new prescreening tests for the genetic-guided enrolment of patients in clinical trials. Importantly, findings are helping to define new rational drug combinations to treat cancer patients with progressive disease.
Preclinical cancer models
Our group is also expanding and characterizing its PDX collections (CRC, neuroendocrine and peritoneal pseudomyxoma), and optimizing their use in evaluating drug efficacy and metastasis by orthotopic injection and live imaging (TC, PET and Echography).
Lastly, we are developing ambitious projects through the EurOPDX, PERSIST-SEQ, CRCelerate Consortia, co-founded by VHIO. These partnerships comprise all the main reference groups working with PDX, single cell sequencing and cancer models in Europe.
* Puig I, Tenbaum SP, Chicote I, Arqués O, Martínez-Quintanilla J, Cuesta-Borrás E, Ramírez L, Gonzalo P, Soto A, Aguilar S, Eguizabal C, Caratù G, Prat A, Argilés G, Landolfi S, Casanovas O, Serra V, Villanueva A, Arroyo AG, Terracciano L, Nuciforo P, Seoane J, Recio JA, Vivancos A, Dienstmann R, Tabernero J, Palmer HG. TET2 controls chemoresistant slow-cycling cancer cell survival and tumor recurrence. J Clin Invest. 2018 Aug 31;128(9):3887-3905.
**
Tenbaum SP, Ordóñez-Morán P, Puig I, Chicote I, Arqués O, Landolfi S, Fernández Y, Herance JR, Gispert JD, Mendizabal L, Aguilar S, Ramón y Cajal S, Schwartz S Jr, Vivancos A, Espín E, Rojas S, Baselga J, Tabernero J, Muñoz A, Palmer HG. β-catenin confers resistance to PI3K and AKT inhibitors and subverts FOXO3a to promote metastasis in colon cancer. Nat Med. 2012 Jun;18(6):892-901.
Puig I, Chicote I, Tenbaum SP, Arqués O, Herance JR, Gispert JD, Jimenez J, Landolfi S, Caci K, Allende H, Mendizabal L, Moreno D, Charco R, Espín E, Prat A, Elez ME, Argilés G, Vivancos A, Tabernero J, Rojas S, Palmer HG. A personalized preclinical model to evaluate the metastatic potential of patient-derived colon cancer initiating cells. Clin Cancer Res. 2013 Dec 15;19(24):6787-801.
Capdevila J, Arqués O, Hernández Mora JR, Matito J, Caratù G, Mancuso FM, Landolfi S, Barriuso J, Jimenez-Fonseca P, Lopez Lopez C, Garcia-Carbonero R, Hernando J, Matos I, Paolo N, Hernández-Losa J, Esteller M, Martínez-Cardús A, Tabernero J, Vivancos A, Palmer HG. Epigenetic EGFR Gene Repression Confers Sensitivity to Therapeutic BRAFV600E Blockade in Colon Neuroendocrine Carcinomas. Clin Cancer Res. 2020 Feb 15;26(4):902-909.
Josep Villanueva obtained his PhD in Biochemistry and Molecular Biology from the Universitat Autonoma de Barcelona in the year 2000.
In 2002 he accepted a postdoctoral fellowship to join Paul Tempst’s laboratory at the Memorial Sloan-Kettering Cancer Center (MSKCC), New York (USA), where he participated in a new clinical proteomics program aimed at the identification of serum cancer biomarkers. He was appointed as lead scientist in developing a unique automated platform for the measurement of peptides in serum using magnetic beads and a mass spectrometry read-out.
Following the ‘technology phase’, he collaborated with physicians and clinical chemists at MSKCC to demonstrate that a limited subset of serum peptides provides accurate class discrimination between patients with three types of solid tumors and healthy controls. Sequence analysis revealed that these peptides were generated by ‘cancer-type’-specific exopeptidase activities. He then developed and reduced to practice in vitro Exoprotease read-out assays for cancer diagnostic applications.
In 2006 he was promoted as Senior Staff Scientist. Focused on becoming an Independent Investigator, he started developing a longer-term project exploring targeted approaches for tumor biomarker discovery. During this time he developed novel methodology for the high-throughput proteomics profiling of cell-secreted inventories (the ‘secretome’) centered on biomarkers, which led to his first publication as senior author in 2009.
Josep’s postdoctoral work is widely recognized in the proteomics and biomarker discovery field. He first authored all ten clinical proteomic publications published from the Tempst laboratory. Furthermore, the platform that he developed was licensed to a biotech company – this is currently available as a commercial solution for proteomic biomarker discovery.
In 2009, he returned to Barcelona as Principal Investigator of VHIO’s Tumor Biomarkers Group. His research focuses on the discovery of new tumor-specific biomarkers and therapeutic targets using proteomic methodologies to improve cancer diagnostics and therapeutics.
Tumor cell communication with its microenvironment plays an important role in tumor initiation and progression. Cancer cells hijack the tumor microenvironment ecosystem via paracrine signaling to promote a pro-oncogenic microenvironment that is crucial for the development of primary and metastatic tumors.
We aim to characterize the mechanisms adopted by these cells to communicate amongst themselves as well as with their microenvironment during tumorigenesis, and exploit these data to advance biomarker and drug target discovery.
Our group’s working hypothesis is that cellular signaling pathways undergo alterations during the tumorigenesis process and that these changes are translated into differential protein secretion, which can also potentially be used to identify secreted markers. Furthermore, some of the differentially regulated proteins could be direct extracellular messengers of intracellular signaling pathways contributing to fundamental stages implicated in cancer initiation and disease progression, thus representing potential therapeutic targets.
Our methodological focus centers on profiling the secreted sub-proteome (‘secretome’) of cells by quantitative mass spectrometry. Most secreted proteins contain a signal peptide that directs their sorting to the extracellular space through the endoplasmic reticulum (ER)–Golgi secretory pathway. Strikingly, when secretome profiles are carefully produced and analyzed, they contain hundreds of theoretical intracellular proteins.
Recent reports showing intracellular proteins with alternative extracellular functions suggest that new protein functions associated with alternative subcellular localizations could be implicated in tumorigenesis.
In line with this notion, our recent efforts in the context of therapeutics and tumor invasion have led us to hypothesize that the characterization of non-classical protein secretion could lead to the development of novel anti-cancer therapies.
Over the last 13 years, Alena Gros’ research has focused on understanding and exploiting the naturally occurring T-cell response to treat cancer. Initially trained as a Biologist, she obtained a PhD in Genetics and did a 7- year postdoctoral fellowship working with Steven A. Rosenberg at the Surgery Branch, National Cancer Institute, US.
She has been directly involved in several clinical studies that enrolled patients to receive neoantigen-enriched or 4-1BB+ selected lymphocytes. Her most cited contribution in this field is the identification of PD-1 as a biomarker of tumor-reactive lymphocytes residing in the tumor (JCI 2014), and the use of this marker to develop noninvasive personalized T cell therapies from peripheral blood (Nature Med 2016, JCI 2019). She is an author of 40 peer-reviewed articles and inventor of over 7 patents related to the field of cancer immunotherapy. In 2016, she was awarded with a Miguel Servet I Fellowship and was recruited by VHIO, where she initiated her independent career leading the Tumor Immunology and Immunotherapy Group.
Her group members are international with diverse training backgrounds. Our research focuses on better understanding the immune response to cancer by identifying the tumor antigens recognized by lymphocytes, evaluating the contribution of identified tumor antigens to antitumor responses following treatment with immunotherapy, and studying the interaction between T cells and tumor cells. These research projects will collectively drive novel insights that will enable us to develop more effective T-cell therapies for patients with solid tumors.
The immune system can recognize and eliminate cancer. However, tumors evade the immune response through multiple mechanisms. Immunotherapies exploit the immune system to attack cancer. Clinical studies have shown that immune checkpoint inhibitors and T-cell-based therapies can mediate tumor regression in patients with metastatic disease. Thus, in addition to surgery, radiation therapy and chemotherapy, immunotherapy has become the fourth pillar of anti-cancer therapy.
Our group’s work has demonstrated that tumor-reactive T cells can frequently be detected circulating in the blood of cancer patients, regardless of the specific tumor type. Tumor-reactive lymphocytes can recognize cancer neoantigens, derived from mutated gene products, and these appear to play an important role in the clinical efficacy of cancer immunotherapies. Furthermore, we have reported biomarkers expressed preferentially on tumor and neoantigen-reactive lymphocytes residing in tumors and in circulation.
Thanks to the support received from the BBVA Foundation's Comprehensive Program of Cancer Immunotherapy & Immunology - CAIMI, as well as other funding agencies, we aim to leverage these biomarkers to reveal T-cell characteristics associated with the clinical efficacy of immune checkpoint blockade in patients treated at the Vall d’Hebron University Hospital (HUVH). In addition, we are currently developing tailored and minimally-invasive T-cell therapies targeting neoantigens derived from peripheral blood.
Through our group’s long-standing collaboration with Elena Garralda, Principal Investigator of VHIO’s Early Clinical Drug Development Group, and Director of our Research Unit for Molecular Therapy of Cancer (UTIM) – CaixaResearch, we recently received authorization from the Agencia Española de Medicamentos y Productos Sanitarios (AEMPS - Spanish Regulatory Agency) in May 2021 to initiate a phase I clinical trial to test the safety and tolerability of neoantigen-selected TILs.
In this clinical trial we use a highly personalized approach (see Figure) to screen for T-cell mediated recognition of mutated antigens using autologous antigen presenting cells that can process and present in all the potential human leukocyte antigen (HLA) restriction elements. In this pilot clinical study funded by the Instituto de Salud Carlos III – ISCIII (Carlos III Health Institute), we aim to treat up to 10 patients with epithelial cancers and melanoma refractory to standard therapies. By enriching for neoantigen-reactive lymphocytes, we hope to extend the efficacy of TIL therapy beyond melanoma.
In summary, our group focuses on better understanding the naturally occurring T-cell response to cancer and establishing ways to exploit these antitumor responses to develop more effective, powerful, and personalized immunotherapies against cancer.