Goedgekeurde dataverzoeken
De data in de Hartwig Medical Database stellen wij beschikbaar voor wetenschappelijk onderzoek voor verbetering van de zorg voor toekomstige patiënten. Sinds 2017 zijn er al meer dan 300 dataverzoeken van onderzoekgroepen uit de hele wereld goedgekeurd.
Hieronder vindt u korte samenvattingen van de goedgekeurde dataverzoeken per jaar, de naam van de hoofdaanvrager en het instituut waarbij zij zijn aangesloten (op het moment dat het dataverzoek werd goedgekeurd).
Chantal Guillemette, CHU de Québec Research Center - Université Laval, Canada
Taichiro Nonaka, Louisiana State University Health Sciences Center - Shreveport, United States of America
Giovanni Capranico, University of Bologna, Italy
Sebastiaan van Heesch, Prinses Maxima Centrum, the Netherlands
John Martens, Erasmus MC, the Netherlands
Jüri Reimand, Ontario Institute for Cancer Research, Canada
Sergey Nikolaev, Gustave Roussy Cancer Campus, France
Harmen van de Werken, Erasmus MC, the Netherlands
Steven Gallinger, Ontario Institute for Cancer Research, Canada
Toni Celià-Terrassa, Hospital del Mar Research Institute, Spain
César Serrano, Vall d'Hebron Institute of Oncology (VHIO), Spain
Mia Petljak, New York University Grossman School of Medicine, United States of America
Francesca Ciccarelli, Queen Mary, University of London, United Kingdom
Luis Zapata, The Institute of Cancer Research, United Kingdom
Paul Verhagen, Erasmus MC, the Netherlands
Samra Turajlic, The Francis Crick Institute, United Kingdom
Felix Dietlein, Chidren's Hospital Boston, USA
- Cancer is a complex disease, characterized by a wide spectrum of genome abnormalities. However, emerging data indicates that genomic drivers may not stand alone as the cause of cancer. Rather, it is becoming clear that the interaction with the patient immune system is critical for development of advanced cancer.
- Using HMF data on metastatic cancer, we will characterize the immune microenvironment, and develop measures of biological age, to investigate how it impacts cancer evolution and treatment response.
- Understanding the relevance of the patient immune response on cancer development and progression may enable novel therapeutic interventions.
Nicolai Birkbak, Aarhus University, Denmark
Ivo Gut, Centro Nacional de Análisis Genómico, Spain
Roelof Koster, Stichting Het Nederlands Kanker Instituut – Antoni van Leeuwenhoek Ziekenhuis, the Netherlands
Fátima Al-Shahrour, Spanish National Cancer Research Center (CNIO), Spain
Roelof Koster, Stichting Het Nederlands Kanker Instituut – Antoni van Leeuwenhoek Ziekenhuis, the Netherlands
Matthew Coelho, Wellcome Sanger Institute, Cambridgeshire, United Kingdom
Metastatic castrate-resistant prostate cancer (mCRPC) is generally insensitive to immune checkpoint inhibitor therapy (ICIs), partly due to its relatively low tumor mutation burden. Up to 5% of patients with mCRPC show a microsatellite unstable (MSI-H), hypermutated phenotype that is more sensitive to ICIs. While studies have demonstrated the clinical efficacy of ICIs among patients with MSI-H solid tumors, data on the efficacy of ICIs in mCRPC specifically is limited to small cohort studies with limited follow up data. The efficacy of ICIs may differ between tumor types due to differences in underlying tumor biology. In this retrospective multicenter study, we aim to improve our insight into the efficacy of anti-PD-(L)1 ICI therapy in MSI-H mCRPC by combining data from the DRUP trial with data from other centres in and outside of Europe.
Niven Mehra, Radboudumc, the Netherlands
Sander Bins, Erasmus MC, the Netherlands
The approval of anti-EGFR treatments for metastatic colorectal cancer significantly improved patient survival. However, drug resistance remains a significant challenge. Limited biomarkers are available to predict patient responses, leaving many without benefits. This project aims to identify genetic and transcriptomic alterations predicting anti-EGFR therapy response using available sequencing data. We aim to validate preliminary findings suggesting intrinsic resistance in CRC tumors is associated with the CDX1/CDX2-GUCY2C axis loss. Secondary findings will also be considered for further validation using our laboratory data. This research aims to enhance patient selection, offering improved therapeutic options for non-responders, potentially increasing the number of beneficiaries.
Saskia Wilting, Erasmus MC, The Netherlands
Sangwoo Kim, Yonsei University College of Medicine, South Korea
Sehrish Kanwal, University of Melbourne Centre for Cancer Research (UMCCR), Australia
Marcin Imielinski, New York University Grossman School of Medicine, United States of America
Maria Anisimova, Institute of Computational Life Sciences, ZHAW, Switserland
Yinxiu Zhan, Istituto Europeo di Oncologia, Italy
Maria Cartolano, University Hospital Cologne, Germany
Matthew Meyerson, Dana Farber Cancer Institute, USA
Joris van de Haar, Netherlands Cancer Institute, the Netherlands
Johannes Brägelmann, University Hospital Cologne, Germany
Jessica Zucman, Cordeliers Research Center, France
Rameen Beroukhim, Dana Farber Cancer Institute, United States of America
We built a prognostic model based on metabolic profile of patients with head and neck squamous cell carcinoma. We need external cohort to test for our developed model.
Vito Carlo Caponio, University of Foggia, Italia
Marc Attiyeh, Cedars-Sinai Medical Center, USA
Floris Barthel, The Translational Genomics Research Institute (TGen), USA
Stefan Loipfinger, Universitair Medisch Centrum Groningen, the Netherlands
Prasanta Dey, Rutgers Cancer Institute of New Jersey, USA
Dan Higgens, Memorial Sloan Kettering Cancer Center, USA
Robin Mjelle, Norwegian university of science and technology, Norway
Jinhyuk Bhin, Yonsei University College of Medicine, South Korea
Francisco Real, Spanish National Cancer Research Center (CNIO), Spain
Based on our DR-196 data request for Cancer of Unknown Primary (CUP) we predicted the primary tumor of multiple patients to be Lung, Gallbladder, and Bile Duct. We wish to compare the WGS and RNA-seq data of the predicted primary CUP tissue with known primary counterparts directly, to unravel specific but also general CUP biology.
Harmen van de Werken, Erasmus MC, the Netherlands
Eliezer van Allen, Dana Farber Cancer Institute, USA
Roel Verhaak, Yale University, United States of America
Francisco Martínez-Jiménez, Vall d'Hebron Institute of Oncology (VHIO), Spain
Nicholas DeVito, Duke University Medical Center, Unites States
Maximilian Haist, Universitätsmedizin Mainz der Johannes-Gutenberg Universität Mainz, Deutschland
Geoff Macintyre, Spanish National Cancer Research Center (CNIO), Spain
Ignacio Flores, Centro de Biología Molecular Severo Ochoa (CBMSO), Spain
Michaël Noë, Stichting Het Nederlands Kanker Instituut – Antoni van Leeuwenhoek Ziekenhuis, the Netherlands
Felix Dietlein, Chidren's Hospital Boston, USA
John Martens, Erasmus MC, the Netherlands
Emre Kocakavuk, University Hospital Essen, Germany
Sarah Aitken, University of Cambridge, United Kingdom
Gaetano Gargiulo, Max Delbrueck Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC)
Hannah Carter, University of California San Diego, USA
Leonie Smeenk, Erasmus MC, the Netherlands
Jeroen de Ridder, Universtair Medisch Centrum Utrecht (UMCU), the Netherlands
Ana Vivancos, Vall d'Hebron Institute of Oncology (VHIO), Spain
Maria Casanova, Spanish National Cancer Research Center (CNIO), Spain
Kim Monkhorst, Nederlands Kanker Instituut – Antoni van Leeuwenhoek, the Netherlands
Balázs Győrffy, Research Centre for Natural Sciences, Hungary
Lixing Yang, University of Chicago, USA
Pawel Zawadzki, Adam Mickiewicz University in Poznań, Poland
Klaus Elenius, Turun Yliopisto, Finland
Charles Swanton, The Francis Crick Institute, United Kingdom
Joost Boormans, Erasmus MC, The Netherlands
Teodora Radonic, VU Medical Center, Amsterdam UMC, the Netherlands
Mark van Opijnen, Haaglanden Medisch Centrum, the Netherlands
Tineke Buffart, VU Medical Center, Amsterdam UMC, the Netherlands
Winan van Houdt, Netherlands Cancer Institute, the Netherlands
Bianca Mostert, Erasmus MC, the Netherlands
Many viruses that commonly infect humans have been found to fuse cells. Evidence suggests that the products of cell fusion can retain some characteristics of the original cells, but that they can also develop malignant characteristics, accelerate cancer progression, and facilitate metastasis. Using whole genome sequencing data we want to identify tumours in which virus-induced cell-cell fusion may have played a role, and potentially contributed to metastasis. This will lead to novel insights into the origins of metastasis, and may be used to identify patients who are at risk of virus-induced cell-cell fusion mediated metastasis.
David Wedge, University of Manchester, United Kingdom
Carlos Caldas, Cancer Research UK Cambridge Institute, United Kingdom
Arul Chinnaiyan, University of Michigan, USA
Ekta Khurana, Weill Medical College of Cornell University, United States of America
Jeanine Roodhart, Universtair Medisch Centrum Utrecht (UMCU), the Netherlands
Sohail Tavazoie, Rockefeller University, United States
Jeffrey van Putten, Hartwig Medical Foundation, The Netherlands
Every year >14,000 patients are diagnosed with cutaneous squamous cell carcinoma (cSCC) and > 35,000 new patients with basal cell carcinoma (BCC). cSCCs metastasize in ~2% of patients and BCCs rarely metastasize. We will investigate if predictive information about metastatic risk is present in the primary tumours. We will 1) characterize the genomic alterations in cSCC/BCC metastatic samples and 2) confirm that genes that were previously identified to be related to the metastatic potential of cSCC/BCC are also mutated in metastatic HMF samples. The previously identified genes will be extended with the new genomic alterations and validated in another cohort.
Loes Zandwijk - Hollestein, Erasmus MC, The Netherlands
Neeltje Steeghs, Netherlands Cancer Institute, the Netherlands
The observed incidence of cancer biomarkers can influence resource investment into key areas such as drug discovery and companion diagnostic technology. The list of clinically relevant cancer biomarkers is growing, however a systematic description of the incidence of these biomarkers is currently lacking. Understanding the prevalence of these biomarkers in late-stage cancer can be especially relevant since many companion diagnostics are utilized in the treatment of stage III and IV cancers.
By using multiple publicly available knowledge management resources that describe clinically relevant alterations, we aim to characterize the frequency of these biomarkers and contextualize their associations within the Hartwig dataset.
Eivind Hovig, University of Oslo, Norge
Patients with Lynch Syndrome (LS) carry a mutation in MLH1, MSH2 (EPCAM), MSH6 or PMS2 and as a result have a genetic predisposition to develop colorectal cancer and endometrial cancer. Recent studies demonstrated that the clinical presentation of LS patients varies depending on which of these genes is affected, though the underlying mechanisms are incompletely understood. By analyzing the DNA of tumors of LS patients, we aim to identify and describe the pathogenic pathways responsible for this clinical variation, with the ultimate goal to develop subgroup-specific guidelines for the diagnosis, surveillance and treatment of patients with LS.
Maartje Nielsen, Leiden University Medical Center, the Netherlands
Jeroen de Ridder, Universtair Medisch Centrum Utrecht (UMCU), the Netherlands
We wish to study the non-coding RNA genes and their somatic mutations in cancer. We have analysed the somatic mutations in ncRNAs from the PCAWG international project and wish to study the data from the Hartwig database to robustly validate our previous findings. We propose that the results will add novel markers for patients' stratification and management.
Stefano Volinia, University of Ferrara, Italy
Cancer develops by the accumulation of mutations in the DNA of individual cells. In many patients, DNA repair mechanisms are often perturbed early in cancer development, leading to an increased mutation rate and faster development of cancer. However, repair deficiencies can also make the cancer cells more susceptible to certain types of treatments. Better understanding and detection of DNA repair deficiencies may thus help improve the treatment of cancer patients.
Using the cancer genomics data from Hartwig Medical Foundation, we aim to characterize the pattern of DNA deficiencies across different cancer types. We further aim to improve the detection of DNA repair deficiencies through their effect on mutation rates and patterns across the genomes of cancer cells. Finally, we aim further clarify the relationship between DNA repair deficiencies and cancer treatment efficacy.
Jakob Skou Pedersen, Aarhus University, Denmark
Recent advances in melanoma treatments have greatly improved patient survival. However, many patients still do not benefit, and we do not fully understand why some patients respond to treatment, while others do not. We have a cohort of melanoma patients with DNA and clinical data, that may help us understand this better. We aim to combine our study with other melanoma datasets from Hartwiig and other sources, to boost the strength and numbers of our study so that we will be better able to answer these questions.
Samra Turajlic, The Francis Crick Institute, United Kingdom
Tumors from children with underlying DNA damage response or DNA repair defects often carry somatic mutational footprints that can aid their identification. Furthermore, relapses or second tumors in these patients may present with specific signatures that arise due to treatment related damage, particularly in DNA-repair deficient tumors. For the detection and analysis of these signatures in childhood tumors, we request the data from the pan-cancer study on metastatic solid tumors (PMID 31748536) that contain repair and therapy associated signatures that will be used to increase power in signature extraction and detailed analysis of mutational signature extraction.
Roland Kuiper, Prinses Maxima Centrum, the Netherlands
Piotr Kozłowski, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poland
Cancer of unknown primary (CUP) has a worldwide incidence of 3-5% of all cancers diagnoses. The clinical problem of this group of patients involves long diagnostic period, limited treatment options (anti-cancer treatment is largely based on primary origin) and poor survival. Whole genome sequencing (WGS) helps these patients by both predicting the primary origin and finding all DNA alterations linked with targeted therapy. The Hartwig prediction algorithm of the tissue site of primary origin (CUPPA) is based on a reference dataset of tumors in the Hartwig database. We aim to improve the prediction algorithm by refining the classification of tumors within the reference dataset towards a more clinically relevant setup.
Petur Snaebjornsson, Netherlands Cancer Institute, the Netherlands
In recent years the treatment of patients with lung cancer has improved tremendously with the introduction of immunotherapy. This therapy helps the patient’s own immune system to attack the tumor cells. Unfortunately, not all patients benefit from this treatment. We want to better understand the mechanisms used by the tumor cells to escape the effect of immunotherapy. By improving our knowledge of these mechanisms, we hope to contribute to new cancer therapies in the future.
Willemijn Theelen, Netherlands Cancer Institute, The Netherlands
Sarcoma is a rare and complex cancer with poor prognosis and survival. While existing genetic studies in cancer have focused on common cancer types such as Breast cancer, we have endeavoured to uncover the underlying genetics of sarcoma by establishing the International Sarcoma Kindred Study (ISKS) consortium. The goal is to better inform standard clinical care of sarcoma. We have performed whole genome sequencing on 1,644 ISKS cases, 3,611 healthy controls and have developed a novel methodology to determine key genetic pathways implicated in sarcoma. Our analysis has revealed novel genetic signatures which will be validated in an independent study.
David Thomas, Garvan Institute of Medical Research, Australia
Camilla Krakstad, University of Bergen, Norway
Nicholas DeVito, Duke University Medical Center, United States
Biliary tract cancer (BTC) is a malignancy of the bile ducts (also called cholangiocarcinoma) and gallbladder. BTC is a rare malignancy with a poor prognosis. Surgical resection is the only treatment with a chance on long-term overall survival but unfortunately only the minority of the patients qualify for resection because of locally advanced or metastatic disease at presentation. For these patients gemcitabine / cisplatin palliative chemotherapy is the standard treatment. Treatments with agents directed to DNA changes (targeted therapy) did not show better survival in different studies because of the heterogeneous nature of BTC. To improve the treatment of advanced BTC, a more comprehensive overview of the genomic changes in advanced BTC is required. lf we can identify subgroups of patients with different prognosis based on genomic changes, new clinical studies could be initiated to study a specific treatment in one or more of these subgroup of patients.
Heinz-Josef Klümpen, Amsterdam UMC locatie AMC, the Netherlands
Nick van Es, Amsterdam UMC locatie AMC, the Netherlands
Hoon Kim, Sungkyunkwan University, South Korea
Nuria Lopez-Bigas, Fundació Institut de Recerca Biomèdica (IRB Barcelona), Spain
Immune cells in the microenvironment of cancers impact disease progression and response to both conventional therapies and immune checkpoint blockers. Accumulating evidence indicates that genomic instability impacts the anti-tumor immune response; while microsatellite instability and mutational burden are associated with an inflamed immune microenvironment, chromosomal instability seems to be associated with immune cell exclusion. To explore this we aim to use genomic (copy numbers, somatic mutations) and microenvironmental (RNA) data to find interactions between genomic instability and the immune response, specifically using a novel measure of chromosomal instability that we recently developed (Van Dijk et al, 2021). Furthermore we aim to explore the feasibility of genomic and microenvironmental measures to predict the course of the disease and calculate associations with clinical parameters such as patient survival. Furthermore, we aim to identify groups of patients based on characteristics of genomic instability, and chromosomal instability (CIN) in particular, that respond better or worse to immune-activating agents.
Sarah Derks, VU Medical Center, Amsterdam UMC, the Netherlands
Fran Supek, Fundació Institut de Recerca Biomèdica (IRB Barcelona, Spain
DNA damage response (DDR) network is critical in maintaining genome integrity. The contribution of the DNA damage tolerance (DDT) system in this network is poorly defined. In this project we strive to delinate mutational profiles to specific defects in the DRR/DDT system.
Heinz Jacobs, Netherlands Cancer Institute, the Netherlands
Wilbert Zwart, Netherlands Cancer Institute, the Netherlands
Wilbert Zwart, Netherlands Cancer Institute, the Netherlands
Wilbert Zwart, Netherlands Cancer Institute, the Netherlands
Juan Garaycoechea, Hubrecht Institute, The Netherlands
Serena Nik-Zainal, University of Cambridge Clinical School, UK
Joaquin Mateo, Vall d'Hebron Institute of Oncology (VHIO), Spain
Barend Sikkema, Erasmus MC, The Netherlands
Gavin Ha, FRED HUTCHINSON CANCER RESEARCH CENTER, United States of America
Andre Bergman, Netherlands Cancer Institute, the Netherlands
Anton Henssen, Charité - Universitätsmedizin Berlin, Germany
Arne van Hoeck, Universtair Medisch Centrum Utrecht (UMCU), the Netherlands
Sushant Kumar, University Health Network, Canada
1. Mutational signatures are patterns of mutations that inform us of the mutational process underlying tumorigenesis. We will investigate the mutational signatures caused by chemotherapeutics, to better understand how chemotherapy affects cancer cells.
2. We will use all the sequencing data from tumors that were treated with chemotherapeutics prior to surgery to discover mutational signatures.
3. Mutations induced by chemotherapy can play a role in how cancers develop resistance to the chemotherapy, therefore understanding where those mutations are likely to farm will he|p us understand how resistance could develop. This knowledge can guide us in avoiding/overcoming resistance to chemotherapy.
Steve Rozen, National University of Singapore, acting through its Duke-NUS Medical School, Singapore
Philippe Hupe, Institut Curie, France
Andrea Sottoriva, Human Technopole, Italy
Peter ten Dijke, Leiden University Medical Center, The Netherlands
Lixing Yang, University of Chicago, USA
Remco van Doorn, Leiden University Medical Center, The Netherlands
Roel Verhaak, The Jackson Laboratory, USA
Anna Poetsch, Technische Universität Dresden, Germany
Christina Curtis, Stanford University School of Medicine, USA
Susan Rotenberg, Queens College of The City University of New York, United States of America
There is no cure for metastatic breast cancer, but there are still many treatment options that may prolong life with reasonable to good preservation of quality of life. To identify suitable treatment options, a tissue biopsy of the metastatic lesion is necessary. Such biopsy is however a burden for the patient, depending on the anatomic site, and may not be representative of other metastatic lesions. Alternatively, a “liquid” biopsy, i.e. blood could be used to find viable treatment options based on circulating tumor DNA. This is now technically feasible and associated with minimal discomfort for the patient. Comparative studies between such liquid and solid biopsies are needed to evaluate whether liquid biopsies are in fact a good alternative for tissue biopsies in metastatic breast cancer patients. Using HMF sequencing data from metastatic tissue lesions (“solid” biopsies), combined with our own analyses of paired blood samples obtained through the Center for Personalised Cancer Treatment (“liquid” biopsies), we will assess whether liquid biopsies are a proper alternative for solid biopsies in metastatic breast cancer patients to identify suitable treatment options.
Cathy Moelans , Universtair Medisch Centrum Utrecht (UMCU), the Netherlands
Joachim Weischenfeldt, Rigshospitalet, Denmark
Colorectal cancer (CRC) ranks as the second most lethal cancer and the third most prevalent malignant tumor worldwide. Despite of continuous development of a new targeted therapies and well defined molecular targets of potential sensitivity, the mechanisms associated with resistance events are poorly understood. Therefore, we will perform whole-genome sequencing data analyses on CRC patients genomes downloaded from Hartwig Medical Foundation and will identify a set of individual genomic features, underlying tumor drug sensitivity and/or resistance. Obtained data will be used to develop a clinical predictor that will help patient stratification toward anti-cancer therapies, improving long-term clinical outcomes.
Pawel Zawadzki, Adam Mickiewicz University in Poznań, Poland
Nick Turner, The Institute of Cancer Research, United Kingdom
One out of 8 women will develop breast cancer (BC) in her lifetime and BC remains the main cause of cancer-related mortality for women in industrialized countries. Approximately 20–30% of patients with early stage BC will develop metastases. Liver metastases are present in ~50% of all metastatic BC patients. There is a clinical need to understand the heterogeneity in outcomes in patients with liver metastatic BC and to have biomarkers to guide the local and systemic treatment strategy for these patients. Here, we will perform an in-depth molecular characterization of liver metastases from BC patients. The ultimate goal is to refine existing and identify new potential treatment avenues for these patients with liver metastatic BC.
Christine Desmedt, Katholieke Universiteit Leuven, for the purposes of this Agreement represented by KU Leuven Research & Development, Belgium
Patients with carcinoma of unknown primary (CUP) are patients with proven metastases but no primary tumour found. These patients have a very poor prognosis of only 2-3 months median survival.
Until now only little research on this patient population is performed in the Netherlands. By improving our knowledge of this entity and comparing it to metastasised known primaries, we hope to improve our current insights.
Ultimately this will lead to identifying those patients who could benefit from currently available therapies and improve the survival of these treated patients. Moreover, preventing unnecessary treatment will improve the quality of life of those who will not benefit from anticancer therapy.
Yes van de Wouw, Viecuri Medisch Centrum, the Netherlands
Mariska Bierkens, Netherlands Cancer Institute, the Netherlands
Lincoln Stein, Ontario Institute for Cancer Research, Canada
Kim Monkhorst, Netherlands Cancer Institute, the Netherlands
Jeanine Roodhart Universtair Medisch Centrum Utrecht (UMCU) the Netherlands
Andrew Stubbs Erasmus MC the Netherlands
Bianca Mostert Erasmus MC the Netherlands
Cristina Rodriguez-Antona, Spanish National Cancer Research Center (CNIO), Spain
Robin Mjelle, Norwegian university of science and technology, Norway
Ivo Gut Centro Nacional de Análisis Genómico (CNAG-CRG) Spain
Niven Mehra Radboudumc the Netherlands
Ian Tomlinson Cancer Research Centre, University of Edinburgh United Kingdom
Yardena Samuels Weizmann Institute of Science Israel
Leendert Looijenga Prinses Maxima Centrum the Netherlands
Pawel Zawadzki Adam Mickiewicz University in Poznań Poland
David Quigley UCSF USA
Itamar Simon Hebrew University Israel
Martijn Lolkema Erasmus MC the Netherlands
Tom van Wezel Leiden University Medical Center the Netherlands
Daniel Zingg Netherlands Cancer Institute the Netherlands
Nick Turner The Institute of Cancer Research United Kingdom
Robin Mjelle Norwegian university of science and technology Norway
Eduard Porta Pardo Josep Carreras Leukaemia Research Institute Spain
David Torrents Barcelona Supercomputing Center Spain
Benjamin Haibe-Kains University Health Network Canada
Rob Knight University of California San Diego USA
Manish Gala Massachusetts General Hospital USA
Alexander Eustace Dublin City University Ireland
Carlos Caldas Cancer Research UK Cambridge Institute United Kingdom
Ruben van Boxtel Prinses Maxima Centrum Netherlands
Chris Barnes University College London United Kingdom
Petur Snaebjornsson Netherlands Cancer Institute the Netherlands
Donate Weghorn Centre for Genomic Regulation (CRG) Spain
Daniel Zingg Netherlands Cancer Institute the Netherlands
Sean Benson Netherlands Cancer Institute the Netherlands
Martijn Lolkema Erasmus MC the Netherlands
Claudio Toma Centro de Biología Molecular Severo Ochoa (CBMSO) Spain
Petur Snaebjornsson Netherlands Cancer Institute the Netherlands
Erik Jan Dubbink Erasmus MC the Netherlands
Johan Lindberg Karolinska Instutet Sweden
Pawel Zawadzki Adam Mickiewicz University in Poznań Poland
Colin Semple The University Court of the University of Edinburgh United Kingdom
Peter van Loo The Francis Crick Institute United Kingdom
Wilbert Zwart Netherlands Cancer Institute the Netherlands
Bart Westerman VU Medical Center, Amsterdam UMC the Netherlands
Joost Boormans Erasmus MC the Netherlands
Andrea Sottoriva The Institute of Cancer Research United Kingdom
Niven Mehra Radboudumc the Netherlands
Matti Nykter Tampere University Finland
Trevor Graham Queen Mary, University of London United Kingdom
Wei Li University of California, Irvine USA
Eliezer Van Allen Dana-Farber Cancer Institute USA
Eliezer Van Allen Dana-Farber Cancer Institute USA
Niven Mehra Radboudumc the Netherlands
Neeltje Steeghs Netherlands Cancer Institute the Netherlands
Binzhi Qian The University of Edinburgh, QMRI United Kingdom
Joachim Aerts Erasmus MC The Netherlands
Jüri Reimand Ontario Institute for Cancer Research Canada
Alessandro Romanel University of Trento Italy
Steven Jones BC Cancer, Part of the Provincial Health Services Authority Canada
Daniel Zingg Netherlands Cancer Institute the Netherlands
Irene Santi Erasmus University Rotterdam The Netherlands
Nicholas McGranahan University College London United Kingdom
Stefano Lise The Institute of Cancer Research United Kingdom
Nicolai Juul Birkbak Aarhus University Denmark
Bianca Mostert Erasmus MC The Netherlands
Astrid van der Veldt Erasmus MC The Netherlands
Carlos Caldas University of Cambridge United Kingdom
Michiel van der Heijden NKI-AvL the Netherlands
Edison Liu The Jackson Laboratory USA
Johannes Koster University Hospital Essen Germany
Christina Curtis Stanford University School of Medicine USA
Liudmila Kodach NKI-AvL the Netherlands
Edwin Cuppen University Medical Center Utrecht the Netherlands
Hanneke van Laarhoven Amsterdam UMC the Netherlands
Harry Groen UMCG the Netherlands
Harry Groen UMCG the Netherlands
Remond Fijneman NKI-AvL the Netherlands
Remond Fijneman NKI-AvL the Netherlands
Heinz-Josef Klümpen Amsterdam UMC the Netherlands
Wilbert Zwart NKI/AvL the Netherlands
Nuria Lopez Bigas IRB Barcelona Spain
Sergey Nikolaev Gustave Roussy Cancer Campus France
Wilbert Zwart NKI/AvL the Netherlands
Steve Rozen Duke-NUS Medical School Singapore
Onno Kranenburg University Medical Center Utrecht the Netherlands
Martijn Lolkema Erasmus MC the Netherlands
Joaquin Mateo Vall d’Hebron Institute of Oncology Spain
Paweł Zawadzki Adam Mickiewicz University Poland
Paweł Zawadzki Adam Mickiewicz University Poland
Laurens van der Vlier Princes Maxima Centrum the Netherlands
Andre Bergman NKI/AvL the Netherlands
Astrid van der Veldt Erasmus MC the Netherlands
John Martens, NKI/AvL,Marjanka Schmidt, Erasmus MC NKI/AvL / Erasmus MC the Netherlands
Filip de Vos University Medical Center Utrecht the Netherlands
Andre Bergman NKI/AvL the Netherlands
Mariette Labots Amsterdam UMC the Netherlands
Harry Büller Amsterdam UMC the Netherlands
Astrid van der Veldt Erasmus MC the Netherlands
Charles Swanton The Francis Crick Institute United Kingdom
Emile Voest NKI/AvL the Netherlands
John Martens, NKI/AvL,Marjanka Schmidt, Erasmus MC NKI/AvL / Erasmus MC the Netherlands
Hans Clevers Hubrecht Institute Utrecht the Netherlands
Martijn Lolkema Erasmus MC the Netherlands
Margot Tesselaar NKI/AvL the Netherlands
Fran Supek IRB Barcelona Spain
John Martens Erasmus MC the Netherlands
Maria Jakobsdottir (before David Wedge) University of Oxford United Kingdom
Jeroen de Ridder UMC Utrecht the Netherlands
Kathleen Marchal Ghent University (UGhent) Belgium
Victor Guryev UMC Groningen the Netherlands
Joachim Aerts Erasmus MC the Netherlands
Peter van Loo The Francis Crick Institute United Kingdom
Filip De Vos UMC Utrecht the Netherlands
Sean Grimmond University of Melbourne Australia
Saskia M. Wilting Erasmus MC the Netherlands
Roel Verhaak The Jackson Laboratory USA
Emile Voest NKI/AvL the Netherlands
Ivo Gut Centro Nacional de Análisis Genómico Spain
Paul Spellman Oregon Health & Science University USA
Andrea Sottoriva The Institute of Cancer Research United Kingdom
Eliezer van Allen Dana-Farber Cancer Institute USA
Jakob Skou Pedersen Aarhus University Denmark
Casper van Eijck Erasmus MC the Netherlands
Heinz Jacobs NKI/AvL the Netherlands
Edwin Cuppen UMC Utrecht the Netherlands
John Martens Erasmus MC the Netherlands
Emile Voest NKI/AvL the Netherlands
Emile Voest NKI/AvL the Netherlands
Harmen van der Werken Erasmus MC the Netherlands
Bauke Ylstra Amsterdam UMC the Netherlands
Wilbert Zwart NKI/AvL the Netherlands
Bianca Mostert Erasmus MC the Netherlands
David Wedge University of Oxford United Kingdom
David Thomas Garvan Institute Medical Research Australia
Bianca Mostert Erasmus MC the Netherlands
Joost Boormans Erasmus MC the Netherlands
Martijn Lolkema Erasmus MC the Netherlands
Eric Wiemer Erasmus MC the Netherlands
Stefan Sleijfer Erasmus MC the Netherlands
John Martens Erasmus MC the Netherlands
Nuria López-Bigas IRB Barcelona Spain
Emile Voest NKI/AvL the Netherlands
Serena Nik-Zainal University of Cambridge United Kingdom
Paul van Diest UMC Utrecht the Netherlands
Edwin Cuppen UMC Utrecht the Netherlands
Edwin Cuppen UMC Utrecht the Netherlands
Nuria López-Bigas IRB Barcelona Spain
Remond Fijneman NKI/AvL the Netherlands
Louisa Hoes NKI/AvL the Netherlands
John Martens Erasmus MC the Netherlands
Michiel van der Heijden NKI/AvL the Netherlands
Henk Verheul Amsterdam UMC locatie Vumc the Netherlands
Martijn Lolkema Erasmus MC the Netherlands
Edwin Cuppen UMC Utrecht the Netherlands
Emile Voest NKI/AvL the Netherlands
Joachim Aerts Erasmus MC the Netherlands
Emile Voest NKI/AvL the Netherlands
Neeltje Steeghs NKI/AvL the Netherlands
Hans van Snellenberg Hartwig Medical Foundation the Netherlands
Edwin Cuppen (previous Wigard Kloosterman) UMC Utrecht the Netherlands
Louisa Hoes (previous Fleur Weeber) NKI/AvL the Netherlands
René Bernards NKI/AvL the Netherlands
Edwin Cuppen (previous Ruben van Boxtel) UMC Utrecht the Netherlands
Deze complete DNA-test maakt de individuele behandeling mogelijk en beperkt daarmee onwenselijke bijwerkingen.