Princess Máxima Center

Sebastiaan van Heesch Group

Microprotein biology and immunotherapy target discovery in childhood cancer

Kép

Our Focus

At the Princess Máxima Center, Europe’s largest research hospital fully dedicated to childhood cancer, our lab specializes in advancing microprotein biology and immunotherapy target discovery. We develop and apply cutting-edge sequencing, proteomics, and computational methods to uncover and study the “dark genome” and proteome, with a focus on identifying novel human microproteins. These small proteins hold vast potential for understanding the mechanisms driving cancer and for developing innovative therapeutic targets.  

  

Central to our research is ribosome profiling (Ribo-seq), a powerful tool to investigate the translational landscape of cancer cells, revealing new insights into RNA usage and microprotein production. By integrating Ribo-seq with advanced genomic, transcriptomic, and proteomic approaches, we aim to uncover actionable targets for precision immunotherapies and cancer vaccines, benefiting both paediatric and adult patients.  

  

Additionally, we co-lead an international consortium to establish standards for identifying, annotating, and interpreting non-canonical open reading frames (ORFs) and newly discovered proteins. Our interdisciplinary approach bridges fundamental research and translational applications, paving the way for personalized treatments that align with the unique molecular profiles of individual patients.  

Main research questions:  

  

What are the roles of novel human microproteins in paediatric and adult cancers?  

How can these microproteins be harnessed as targets for cancer immunotherapies and vaccines?  

About Sebastiaan van Heesch

My Research

Sebastiaan van Heesch is a group leader at Princess Máxima Center for Pediatric Oncology, a junior Oncode Investigator at Oncode Institute, and vice chair of the Therapeutic Vaccines workstream at Oncode Accelerator - a Dutch nation-wide program that aims to accelerate the preclinical development of cancer vaccines. Trained at the Hubrecht Institute (PhD) and the MDC Berlin (postdoc), Sebastiaan has developed a passion for RNA systems biology, proteogenomics, and cancer immunotherapy. Within the Princess Máxima Center - Europe’s largest research hospital fully dedicated to battling childhood cancer, the van Heesch lab develops and applies advanced sequencing and computational methods to find and characterize new microproteins, find new cancer mechanisms, and select those microproteins that may serve as targets for immunotherapy.

Awards
  • 2023: NWO VIDI Award

  • 2019: DZHK Postdoc Excellence Award

  • 2015: NWO Rubicon Award

  • 2015: EMBO Long-Term Fellowship

Key Publications
  1. Hofman DA, Ruiz-Orera J, Yannuzzi I, Murugesan R, Brown A, Clauser KR, Condurat AL, van Dinter JT, Engels SAG, Goodale A, van der Lugt J, Abid T, Wang L, Zhou KN, Vogelzang J, Ligon KL, Phoenix TN, Roth JA, Root DE, Hubner N, Golub TR, Bandopadhayay P, van Heesch S, Prensner JR. Translation of non-canonical open reading frames as a cancer cell survival mechanism in childhood medulloblastoma. Molecular Cell (2024) Jan 261-276. e18. doi: https://doi.org/10.1016/j.molcel.2023.12.003

  2. Prensner JR, Abelin JG, Kok LW, Clauser KR, Mudge JM, Ruiz-Orera J, Bassani-Sternberg M, Deutsch EW, van Heesch S. What can Ribo-seq, immunopeptidomics, and proteomics tell us about the non-canonical proteome? Molecular Cellular Proteomics. (2023) Aug 10:100631. doi: https://doi.org/10.1016/j.mcpro.2023.100631

  3. Sandmann CL, Schulz JF, Ruiz-Orera J, Kirchner M, Ziehm M, Adami E, Marczenke M, Christ A, Liebe N, Greiner J, Schoenenberger A, Muecke MB, Liang N, Moritz RL, Sun Z, Deutsch EW, Gotthardt M, Mudge JM, Prensner JR, Willnow TE, Mertins P, van Heesch S, Hubner N. Evolutionary origins and interactomes of human, young microproteins and small peptides translated from short open reading frames. Molecular Cell (2023) Mar 16;83(6):994-1011.e18. doi: https://doi.org/10.1016/j.molcel.2023.01.023

  4. Mudge JM, Ruiz-Orera J, Prensner JR, Brunet MA, Calvet F, Jungreis I, Gonzalez JM, Magrane M, Martinez TF, Schulz JF, Yang YT, Albà MM, Aspden JL, Baranov PV, Bazzini AA, Bruford E, Martin MJ, Calviello L, Carvunis AR, Chen J, Couso JP, Deutsch EW, Flicek P, Frankish A, Gerstein M, Hubner N, Ingolia NT, Kellis M, Menschaert G, Moritz RL, Ohler U, Roucou X, Saghatelian A, Weissman JS, van Heesch S. Standardized annotation of translated open reading frames. Nature Biotechnology (2022) Jul;40(7):994-999. https://doi.org/10.1038/s41587-022-01369-0

  5. van Heesch S, Witte F, Schneider-Lunitz V, Schulz JF, Adami E, Faber AB, Kirchner M, Maatz H, Blachut S, Sandmann CL, Kanda M, Worth CL, Schafer S, Calviello L, Merriott R, Patone G, Hummel O, Wyler E, Obermayer B, Mücke MB, Lindberg EL, Trnka F, Memczak S, Schilling M, Felkin LE, Barton PJR, Quaife NM, Vanezis K, Diecke S, Mukai M, Mah N, Oh SJ, Kurtz A, Schramm C, Schwinge D, Sebode M, Harakalova M, Asselbergs FW, Vink A, de Weger RA, Viswanathan S, Widjaja AA, Gärtner-Rommel A, Milting H, Dos Remedios C, Knosalla C, Mertins P, Landthaler M, Vingron M, Linke WA, Seidman JG, Seidman CE, Rajewsky N, Ohler U, Cook SA, Hubner N. The Translational Landscape of the Human Heart. Cell (2019) 178(1):242-260.e29. https://doi.org/10.1016/j.cell.2019.05.010

Members

Sebastiaan van Heesch
Group leader
Alisa Potter    
Phd student
Ana Pinheiro Lopes    
Postdoc
Damon Hofman    
Phd student
Edwin Werf    
Technician
Emma Westerink    
Research analist
Ferhat Alkan    
Technician
Kazuma Nakatani    
Postdoc
Komal Yadav    
Research analist
Leron Kok    
Phd student
Luuk Broeils    
Phd student
Marina Reixachs Sole    
Phd student
Martina Jovancheva    
Phd student
Sanne Luijcx    
Technician
Simon Venneman    
Project manager
Ting Luo    
Reseach Analist
Wouter Makkinje    
Technician
 
Anne Rios Group

Cancer, immunotherapy development, high resolution 3D imaging, organoid technology, bio-engineering

Kép

Our Focus

The Rios group develops new model systems and tailored microscopy technologies to advance cancer research and find new leads for therapy improvement. A key focus lies on understanding the dynamic mode-of-action of cellular immunotherapy, a promising class of treatment that harvests the patient’s own immune system to fight cancer. These cellular therapies essentially are ‘living drugs’ and we argue that to exploit their full clinical potential we need to better understand their living mode-of-action. To achieve this, we developed a live cell imaging platform (BEHAV3D) designed to record the functional behaviour of these cells. In addition, we invest in the development of human model systems that not only capture the diverse tumour characteristics and varying treatment responses as observed in patients, but that we also try to further advance by adding key components of the tumour microenvironment. The direct environment of the tumour, and especially local immune cells, are known to significantly impact treatment outcomes. With these advanced models we try to map these often detrimental interactions between the local immune cells, the tumour cells and the cells used to treat cancer. Through these efforts we aim to find new therapeutic targets to maximize treatment outcomes.  

About Anne Rios

Name

Anne Rios

Position
Oncode Investigator at Princess Máxima Center
My Research

Visualize the unexpected’ is the driving force behind my science.

From the start of her scientific career, Anne developed an intense passion for imaging, which she applied during her PhD in developmental biology to unveil a novel developmental scenario by which migrating cells from adjacent tissues can trigger cell fate changes in muscle stem-cells (Nature, 2011). During her postdoc, she established a method to visualize intact tissue using 3D microscopic reconstruction (Neil Lawrence prize for most creative young scientist, 2016). The power of this method in driving scientific discovery is illustrated by the first in vivo evidence for bipotent stem cells in the mammary gland (Nature, 2014, Nature cell biology, 2017) and the unexpected finding of polyploid cells during lactation (Nature Communications, 2016). Moreover, it was optimized into a rapid and easy-to-use protocol for 3D imaging of entire organs, intact tumours and organoids (Cancer Cell, 2019 -highlighted Best of Cancer Cell 2019; Nature Biotechnology 2021; Nature Protocols 2019 & Nature Protocols, 2022). Combined with a novel multicolored single-cell 3D intravital imaging technique, this technique allowed for discovering a novel population of resident macrophages essential for maintaining breast homeostasis, as well as involved in breast cancer progression (Nature Cell Biology, 2020, Nature Protocols, 2021).

In 2017, Anne was recruited as a group leader to the Princess Máxima Center for Pediatric Oncology and head of the Princess Máxima Imaging Center. In 2019, she was also appointed Oncode junior principal investigator. She built a multidisciplinary team, with experts in bioengineering, imaging and computational science and together they study cancer biology and immunotherapy treatment response. Anne is a selected next generation member of the International Society for Stem Cell Research (ISSCR), a program committee member of the European Fight Kid Cancer funding agency and a founding member of the EMBO journal catalyst program. Anne received a European Research Council (ERC) Starting Grant and KWF high risk grant for her work on pediatric brain cancer, a CGC Young PI Award to study breast cancer dynamics and a St. Baldrick’s Robert J. Arceci Innovation Award recognizing her innovative approaches to tackle paediatric cancer research. In addition, Anne is a participating PI in multiple consortia dedicated to the next generation of T cell therapies, advancing organoid technology and further developing imaging and analytical frameworks to study and guide cells in their native environment. In 2021, she received the L’Oréal-UNESCO for Woman in Science Award and is now one of the founders of ACE (Authenticity in science for Community and Excellence) to promote women in science. In 2021, she was a honorary Fellow of NIAS-KNAW (Dutch royal Academy of Arts and Sciences) for 5 months, where she developed projects related to Art and Science. She has a strong passion for engaging the public (NWO Science Communication grant, 2002) and inspiring the future generation of scientists, which she aims to achieve through unique immersive displays of her group’s 3D imaging data.

Awards
  • 2022: NWO Science Communication Grant: An unexpected alliance to portray cancer.

  • 2022: Horizon Europe 2021 Marie Sklodowska-Curie Actions (MSCA) Cofund co-PI. From Caterpillar to BUTTERFLY: supporting transformation of DCs in a paediatric oncology network.

  • 2022: Zwaartekracht consortium grant Imagine. Innovative. Microscopy and Guidance of Cells in their naïve environment.

  • 2021: ODAS Foundation, consortium grant. Identify targets to modulate CAR-T cell functionality using advanced 3D co-culture and imaging technologies: Towards better survival of pediatric BCP-ALL patients.

  • 2021: KWF Consortium Grant. Targeting human cancer with the next generation of engineered immune cells: TEGs.

  • 2021: L’Oréal-UNESCO for Woman in Science Award

  • 2019: St. Baldrick’s Robert J. Arceci Innovation Award

  • 2018: ERC starting grant Deciphering and targeting the invasive nature of Diffuse Intrinsic Pontine Glioma

  • 2018: Villa Joep Optimizing neuroblastoma resection using fluorescent guided surgery

  • 2018: KWF high risk Unravelling the integration of pediatric high-grade glioma in the developing brain using state-of-the-art imaging

  • 2018: CGC young PI award Studying human breast cancer dynamics using multi-coloured lineage tracing

  • 2017: Cancer Genomic Center member

  • 2016: Lawrence Creative Prize Winner Medical Innovation Award

  • 2014: Walter and Eliza Hall Research Institute Post-Doctoral Award

  • 2013: Postdoctoral Fellowship co-funded by the National Breast Cancer Foundation and Cure Cancer Australia Foundation

  • 2011: Director’s Prize PhD award

Key Publications
  1. Van Ineveld R.L., Collot R., …, Rios A.C. Multispectral confocal 3D imaging of intact healthy and tumor tissue using mLSR-3D (Nature Protocols, Dec. 2022)

  2. Rios A.C. Resolving the spatial heterogeneity of cancer in 3D. (Nature Reviews Cancer, Tools of the Trade, Aug 2022)

  3. Dekkers J.F., Alieva M., …, Rios A.C. Behavioral-transcriptomic landscape of engineered T cells targeting human cancer organoids (Nature Biotechnology, Jul 2022)

  4. van Ineveld R.L., van Vliet E., Wehrens E.J., …, Rios A.C. 3D imaging for driving cancer discovery. Review. (EMBO J, Apr 2022)

  5. van Ineveld R.L., Kleinnijenhuis M., …, Rios A.C. Revealing the spatio-phenotypic patterning of cells in healthy and tumor tissues with mLSR-3D and STAPL-3D (Nature Biotechnology, Jun 2021).

  6. Dekkers J.F., …, Rios A.C. Long-term culture, genetic manipulation of human normal and breast cancer organoids. (Nature Protocols, Mar 2021).

  7. Dawson C.A., …, Rios A.C. & Visvader J.E. Intravital microscopy of dynamic single-cell behaviour in mouse mammary tissue. (Nature ProtocolsMar 2021).

  8. Wellens L.W., Deken M.M., …, Rios A.C. Anti-GD2-IRDye800CW as a targeted probe for fluorescence-guided surgery in neuroblastoma. (Scientific Reports, Oct 2020)

  9. Dawson C.A., …, Rios A.C. & Visvader J.E. Tissue-resident ductal macrophages survey the mammary epithelium and facilitate tissue remodelling. (Nature Cell Biology, May 2020).

  10. Dekkers J.F., …, Visvader J.E., Clevers H., Rios A.C. High-resolution three-dimensional imaging of fixed and cleared organoids. (Nature Protocols, Jun 2019)

  11. Rios A.C., …, Visvader J.E. Intraclonal plasticity in mammary tumors revealed through Large-scale Single-cell Resolution 3D imaging(Cancer Cell, Jun 2019

  12. Rios A.C., Clevers H. Imaging organoids: a bright future ahead. Review. (Nature Methods, Jan 2018)

  13. Rios A.C., Fu N.Y., Lindeman G.J. & Visvader J.E. In situ identification of bipotent stem cells in the mammary gland. (Nature, Feb 2014)

  14. · Rios A.C., Marcelle C. & Serralbo O. Gene loss-of-function and live imaging in chick embryos. (Methods Molecular Biology, 2012)

  15. Rios A.C., …, Marcelle C. Neural crest regulates myogenesis through the transient activation of NOTCH. (Nature, May 2011)

Members

Anne Rios
Group leader
Amber Wezenaar    
PhD Student
Amber Zeeman    
Technician
Britt Vervoort    
PostDoc
Celina Honhof    
Research Technician
Ellen Wehrens    
Senior researcher
Emma Bokobza    
Phd Student
Jessica Mazalo    
Postdoc
Mario Barrera Román    
Research Analyst
Marit Kruit    
Research Technician
Milo Molleson    
Computational analyst
Raphael Collot    
PhD student
Ravian van Ineveld    
Postdoc fellow
Rijndert Ariese    
Technician
Sam de Blank    
Bioinformatician
Uddeshya Pandey    
PhD student
  
René Medema Group

Cell Division and Cancer

Kép

Our Focus

The first aim of our group is to unravel the mechanisms that protect genetic integrity of a normal cell, and study how these mechanisms are compromised in cancer cells to allow them to evolve more rapidly. This capacity to rapidly evolve comes at a cost of having to adapt to a new genetic make-up, sometimes involves gains or losses of entire chromosomes, and we are very interested to understand how cells manage to adapt to this type of instability. This has allowed us to uncover several liabilities that can be exploited to target cancer cells, some of which are currently being tested clinically.  

In April 2024 my group and the group of Gerben Vader moved to the Princess Maxima Center, and we merged our groups. Coincident with this move we have also (further) developed 2 new research lines relevant to paediatric cancer. In the first we study the role of ecDNA, prominently present in various forms of difficult-to-treat paediatric cancers, and try to identify liabilities of ecDNA-containing cancers. In the second, we try to identify germ line-specific proteins that are expressed in paediatric cancers, as a means to identify proteins that can discriminate paediatric cancer cells from other somatic cells.  

About René Medema

My Research

Rene H. Medema started his career in 1989 in the lab of Hans Bos in Leiden as a PhD student working on signal transduction by p21ras. After his PhD he moved to the laboratory of Prof. Dr. R.A. Weinberg (Whitehead Institute, Cambridge, MA) for his postdoctoral training, working on cell cycle control. As an independent group leader he continued to study cell cycle control, first at the University Medical Center Utrecht (1995-2000) and subsequently at the Netherlands Cancer Institute (2001-2005). In 2005 he moved back to Utrecht to become professor in Experimental Oncology. In 2012, he was appointed director of research at the Netherlands Cancer Institute.

His group has made several key contributions to the general understanding of control of the cell cycle by Forkhead transcription factors, FoxO and FoxM1. In addition, work from his group has provided more insight on the role of motor proteins in spindle assembly and the consequences of chromosome missegregation on the genomic stability and viability of a tumor cell. His group was the first to show that recovery from a DNA damage-induced arrest is controlled by Polo-like kinase-1 in 2004, and his lab has since provided several major contributions to our understanding of the recovery process.

Awards
  • 2013: Appointed member of the European Academy of Cancer Sciences

  • 2013: Appointed member of the KNAW (Royal Netherlands Academy of Arts and Sciences)

  • 2012: Elected member of the Academia Europea

  • 2009: EMBO membership

  • 2004: VICI Laureate

Key Publications
  1. Friskes, A., Koob, L., Krenning, L., Severson, T.M., Koeleman, E.S., Vergara, X., Schubert, M., van den Berg, J., Evers, B., Manjón, A.G., Joosten, S., Kim, Y., Zwart, W., Medema, R.H. (2022). Double-strand break toxicity is chromatin context independent. Nucl. Acid Res. 50(17), 9930.

  2. Ferninga, F.M., Raaijmakers, J.A., Hadders, M.A., Vaarting, C., Macůrek, L., Heitink, L., Krenning, L., Medema, R.H. (2018). Persistent DNA repair intermediates induce senescence. Nat. Commun. 9(1), 1.

  3. Krenning, L., Feringa, F. M., Shaltiel, I.A., van den Berg, J., & Medema, R.H. (2014). Transient activation of p53 in G2 phase is sufficient to induce senescence. Molecular cell55(1), 59-72.

  4. Janssen, A., van der Burg, M., Szuhai, K., Kops, G. J., & Medema, R.H. (2011). Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science333(6051), 1895-1898.

  5. Macůrek, L., Lindqvist, A., Lim, D., Lampson, M.A., Klompmaker, R., Freire, R., ... & Medema, R.H. (2008). Polo-like kinase-1 is activated by aurora A to promote checkpoint recovery. Nature455(7209), 119.

Members

René Medema
Group leader, CSO
Amber Hondema    
PhD student
Anoek Friskes    
PhD student
Elsa Coolen    
PhD student
Gerben Vader    
Associate group leader
Lenno Krenning    
Research associate
Louise Janssen    
PhD student
Lukas Frank    
PostDoc
Marco Novais da Cruz    
PhD student
Maud Schoot Uiterkamp    
PhD student
Mila Ilic    
PhD student
Rob Klompmaker    
Lab Manager
Jan Hoeijmakers Group

DNA repair

Kép

Our Focus

Our group studies genomic instability and its consequences for cancer and aging: the main healthcare problems in all developed societies. DNA damage (DD) occurs continuously in every cell, at massive scale. It leads to mutations that initiate and fuel carcinogenesis including onset of therapy resistance frustrating effective cure. DD also triggers cell death, senescence and interferes with genome function causing functional decline and aging-related diseases. We discovered that DD is the main cause of aging and predicted that DNA-damaging chemo/radiotherapy would accelerate aging, later confirmed in cancer survivors. We also found that dietary restriction and fasting induce a remarkably powerful protective ‘survival response’, which suppresses growth and prioritizes resilience mechanisms, reduces DD, delays aging and protects from surgery-associated ischemia/reperfusion-injury and chemo/radiotherapy. We study underlying mechanisms using mouse models for human repair syndromes, generated by our own team, to fully understand this response, derive rational-based effective nutritional/pharmacological strategies, that promote healthy aging, improve cancer therapy and reduce its severe short/long-term side-effects in children/adults and hence significantly improve daily quality of life of all (ex-)cancer patients. Particular interest concerns cognitive decline as we found neurofunction to benefit disproportionally from such interventions. Importantly, these nutritional interventions are cheap, easily implementable and applicable to all (ex-)cancer patients, globally.  

About Jan Hoeijmakers

My Research

Starting from the molecular analysis of DNA repair in mammals in 1981 by cloning many human DNA repair genes, which enabled elucidation of the underlying mechanisms and resolved the basis of rare human repair syndromes, we embarked upon the generation of a large series of mouse repair mutants, which we found to display accelerated but fully bona fideaging. This allowed us to reveal time- and exposure-dependent accumulation of DNA damage as the main cause of systemic aging. Whereas DNA-damage-induced mutagenesis underlies carcinogenesis, accumulated DNA damage in proliferating cells also arrests cell cycle progression inducing senescence. In non-dividing cells accumulation of DNA damage was found to interfere with transcription, to lower and skew the transcriptional output, leading to cellular functional decline and cell death and hence aging of the soma. Intriguingly, we found that aging caused by DNA-damage-induced transcription stress, simultaneously triggered a potent and interesting anti-aging, anti-cancer ‘survival’ response, which prioritizes resilience above growth. Caloric restriction (CR), which delays aging, induces a similar response. Applying calorie restriction to progeroid repair mutant mice and patients strongly delayed accelerated aging and even dramatically improved neurofunction, partially reverting neurodegeneration.

As DNA damage is the main culprit for systemic aging, this implies that most genotoxic (anti-cancer) treatments including chemo- and radiotherapy are predicted to cause features of premature aging, consistent with epidemiological studies. Since ~90% of pediatric patients are cured for a large part by virtue of chemotherapy, and ex-patients have a long life expectancy, the long-term pro-aging side effects, which significantly affect well being and QoL, constitute one of the most pressing problems in (pediatric) oncology. However, as outlined above, nutritional interventions are able to delay aging and can trigger a potent ‘survival response’, which boosts resilience and defence systems. These ingredients form the basis of several new research directions with promising perspectives in oncology, surgery and neurodegeneration, which we currently pursue in the following research lines.

  1. The project aims to extend our understanding of the mechanism by which CR delays aging and exerts strong benefits in repair-deficient progeroid mice and patients, particularly improving neurofunction. The anti-aging effect of CR is still largely unknown. A very relevant observation we made is that CR reduces genome-wide transcription stress, indicating it lowers levels of DNA lesions. As DNA damage causes systemic aging, this finding reveals how CR delays aging, extends lifespan and even allows improved neurofunction. Particularly, we would like to know how CR reduces DNA damage and to get insight into the mechanism by which neurodegeneration is halted and in part even reverted, which is relevant for ex-(pediatric)cancer patients experiencing features of accelerated aging. These studies also aim to identify the components in food that are responsible for the benefits of CR and to search for effective CR-mimicking agents or treatments.

  2. The ‘survival’ response triggered by CR and accelerated aging induces potent protection against a wide range of stresses. We have shown that pre-treatment fasting (1-3 days water only) in mice induces an exceptionally strong protection against ischemia reperfusion injury (IRI) that occurs after clamping blood supply to kidney and liver. To see whether and to which extent 2-3 days pre-treatment fasting also reduces IRI and improves recovery associated with (oncological) surgery and organ transplantation in humans we have initiated two randomised controlled clinical trials:

    1. The FIURTT study, Fasting Intervention for children with Unilateral Renal Tumours to reduce Toxicity, and

    2. The FAST trial, Fasting before live kidney donation, effect on donor wellbeing and postoperative recovery.

    3. In addition, we started the KetoHeppy investigation, KETOgenic diet therapy in patients with HEPatocellular adenoma. Next to these clinical studies we are performing in vitrostudies at the level of cultured cells, organoids as well as human and mouse organotypical slices to examine the effect of CR-mimicking conditions on metabolism and stress resistance.

  3. To investigate whether pre-treatment fasting also induces resistance to the short- and long-term side effects of chemo- and radiotherapy we are examining the response of fasting/CR in healthy and PDX-harbouring mice on different types of anti-cancer treatment. Promising initial results have been obtained. This serves as a steppingstone to clinical trials in pediatric and possibly adult cancer patients. The in vitrosystems may also enable the development of a screening platform for CR mimetics or compounds/treatments which synergize with CR.

  4. Finally, our progeroid mouse repair mutants appeared superior models for Alzheimer’s disease and other dementias and displayed an extraordinary beneficial response to CR. This was even surpassed by applying CR to the first repair-deficient TTD patient, revealing that the mouse model is highly valid for humans. Moreover, we found that CR also alleviated DNA-damage-driven transcription stress in the brain of the mice, and preliminary findings indicate that CR diminished amyloid-like as well as other pathogenic protein aggregates, which spontaneously develop in our repair mutants. These observations may address a tremendous unmet medical need and provide the basis for research directed at better understanding the molecular mechanisms involved and translation to patients, including (ex-)pediatric patients suffering from cognitive decline.

In summary, our research aims to elucidate the surprisingly simple, globally-applicable, inexpensive potential of nutritional interventions on (accelerated) aging and on health and to pioneer their application in important areas in medicine, including surgery, organ transplantation, chemo- and radiotherapy and proteinopathies in common forms of dementia.

Awards
  • 2021: Official recognition of the Erasmus MC clinical expertise center for Rare Genome Instability Disorders, a multi-disciplinary clinic for children with DNA repair syndromes, initiated by Jan Hoeijmakers

  • 2020: Ammodo Research Team Award” to ‘Guardians and Caretakers of the Genome’ together with other members of the Erasmus Dept. Mol. Genet.

  • 2019: “EMGS Award” of the Environmental Mutagenesis and Genetics Soc. (Washington DC)

  • 2018: Knowledge Ambassador of the City of Rotterdam

  • 2017: International Olav Thon Foundation personal Award

  • 2017: Honorary TEFAF Oncology Chair of the Maastricht Univ. Medical Center

  • 2016: NVHG Galjaard Prize of the Netherlands Society of Human Genetics

  • 2016: Professor International Faculty, Cologne University (Cologne, Guest Professor)

  • 2016: Selected for the Nobel-Forum lecture at the Karolinska Institute

  • 2015: ERC PoC grant DEMENTIA European Research Council

  • 2014: Consulted by the Nobel Committee for the Nobel Prize in Chemistry for DNA repair (2014-2015)

  • 2013: Royal distinction Knight in the Order of the Dutch Lion for important scientific achievements in the area of cancer and aging research (2013).

  • 2012: Mendel Medal on the occasion of the 190th anniversary of Mendel’s birth

  • 2011: Koningin Wilhelmina Research Prize of the Dutch Cancer Society, for research on DNA damage response in prostate and urinary bladder cancer (2M€) (Leiden, 2011)

  • 2011: Academy Professor of the Royal Academy of Sciences of The Netherlands (KNAW), First Academy Professor new style in the broad domain of Beta sciences

  • 2011: Cancer Research Prize of the Charles Rudolph Brupbacher Stiftung for research on the role of genome stability in cancer and aging, shared with Bert Vogelstein

  • 2008: ERC Advanced Grant DamAge – Multi-disciplinary Sciences European Research Council

  • 2008: Seneca Medaille des Industrie-Clubs für Altensforschung Prize, for pioneering research on the molecular basis of aging (First awardee)

  • 2001: ‘Josephine Nefkens Prize’ for cancer research

  • 2000: Elected member of KNAW (section ‘Medicine’, dept. ‘Physics’)

  • 2000: ‘EC-Descartes’ Award for European collaboration on DNA repair

  • 2000: ‘Van Gogh’ Prize from the Dutch Science Organization

  • 2000: ‘Descartes-Huygens’ Award for French-Dutch scientific collaborations

  • 1999: ‘Spinoza’ Prize, most recognized prize of the Dutch Science Organization

  • 1995: The very prestigious 'Louis Jeantet' Prize for Medical Research in Europe for the entire work on DNA repair (Geneva, 1995)

  • 1995: EMBO

  • 1986: 'Snoo van t' Hoogerhuys' Prize (isolation of the first human DNA repair gene)

  • 1983: 'Harold Quintus Bosz' Prize (for the discovery of the molecular mechanism of antigenic variation in trypanosomes, PhD thesis)

Key Publications
  1. Hoeijmakers, J. H. (2001). Genome maintenance mechanisms for preventing cancer. nature411(6835), 366.

  2. De Boer, J., Andressoo, J. O., de Wit, J., Huijmans, J., Beems, R. B., van Steeg, H., ... & Hoeijmakers, J.H. (2002). Premature aging in mice deficient in DNA repair and transcription. Science, 296(5571), 1276-1279.

  3. Niedernhofer, L. J., Garinis, G. A., Raams, A., Lalai, A. S., Robinson, A. R., Appeldoorn, E., ... & Hoeijmakers, J.H. (2006). A new progeroid syndrome reveals that genotoxic stress suppresses the somatotroph axis. Nature, 444(7122), 1038.

  4. Marteijn, J. A., Lans, H., Vermeulen, W., & Hoeijmakers, J. H. (2014). Understanding nucleotide excision repair and its roles in cancer and ageing. Nature reviews Molecular cell biology, 15(7), 465.

  5. Vermeij, W. P., Dollé, M. T., Reiling, E., Jaarsma, D., Payan-Gomez, C., Bombardieri, C. R., ... & Hoeijmakers, J.H. (2016). Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature, 537(7620), 427.

  6. Schumacher, B., Pothof, J., Vijg, J., & Hoeijmakers, J.H. (2021). The central role of DNA damage in the ageing process. Nature, 592(7856), 695.

Members

Jan Hoeijmakers
Oncode Investigator
Alice Larsson    
Phd student
Anna Yakubovska    
Phd student
Carolina Konrdorfer Rangel    
Phd student
Chris Oudmaijer    
Arts-onderzoeker
Daphne Komninos    
PhD student
Irene van Dijken    
PhD student
Ivar van Galen    
PhD student
Kimberly Smit    
Research analyst
Rutger Ozinga    
Bio Informatician
Wilbert Vermeij    
Postdoc fellow
Willianne Vonk    
Senior Postdoc
Yvonne Rijksen    
Research-technician
Ziqin Tang    
PhD student
 
Jarno Drost Group

Molecular dissection of childhood solid tumors

Kép

Our Focus

Although survival rates for children with cancer have increased in recent decades, cancer is still the leading cause of disease-related deaths in children. Survivors suffer from side effects of the, in most cases, intensive treatment regimens. Hence, there is an urgent need to develop new therapies. However, therapeutic innovation is hampered by the lack of cell models representative of native tumour tissue. My lab pioneers the use of organoid technology for paediatric cancer research.   

  
Many childhood tumours originate in the developing fetes. They are likely caused by a block in processes driving lineage-specification and differentiation. In most cases, the cells from which the tumours originate are only present during short, specific time-windows in development, which makes it challenging to identify the processes initiating and driving tumorigenesis. We aim to identify the origin of childhood cancer and to increase our understanding of the processes that underpin their development. To this end, we take a multi-disciplinary approach making use of our unique in vitro models, access to unique patient material, in vivo orthotopic xenograft models, as well as state-of-the-art (single-cell) omics and lineage tracing technologies.  Ultimately, we aim to develop new, less toxic therapies to treat children with cancer.  

About Jarno Drost

My Research

Jarno Drost obtained his Master’s degree in Biomedical Sciences cum laude in 2005 from the Free University in Amsterdam. He received his PhD from the Erasmus University Rotterdam for his work on the identification of new tumor suppressor genes in the P53 pathway in the research group of Reuven Agami at the Netherlands Cancer Institute in Amsterdam.

Subsequently, Jarno joined the group of Hans Clevers at the Hubrecht Institute for his postdoctoral training where he exploited the organoid technology for cancer research. He developed colorectal cancer (CRC) progression models by introducing combinations of the most commonly mutated CRC genes in human small intestinal and colonic organoids using CRISPR/Cas9 genome editing. He used these models to study multistep tumorigenesis. For his work he received an NWO/Veni fellowship and the Dr. Patrick Hanlo Award for best postdoctoral researcher of the Hubrecht Institute.

In November 2016, he became a group leader at the Princess Máxima Center for pediatric oncology. His group studies the molecular alterations underlying pediatric solid tumors and uses novel pre-clinical model systems to identify therapeutic targets. He was granted a Bas Mulder young investigator award from the Dutch Cancer Society (KWF), the ERC Starting and NWO-Vidi grants, and the AACR St. Baldrick’s career development award for emerging leaders in the field of pediatric oncology.

Awards
  • 2021: NWO-Vidi grant

  • 2021: Achieved tenure at Princess Máxima Center for Pediatric Oncology

  • 2020: AACR St. Baldrick’s career development award

  • 2019: European Research Council (ERC) Starting Grant

  • 2017: Dr. Patrick Hanlo Award for best postdoctoral researcher of the Hubrecht Institute

  • 2016: Bas Mulder young investigator award from the Dutch Cancer Society (KWF)

  • 2014: NWO (Netherlands organization for scientific research) VENI award

Key Publications
  1. Meister, M.T., Groot Koerkamp, M.J.A., de Souza, T., Breunis, W.B., Frazer-Mendelewska, E., Brok, M., DeMartino, J., Manders, F., Calandrini, C., Kerstens, H.H.D., Janse, A., Dolman, M.E.M., Eising, S., Langenberg, K.P.S., van Tuil, M., Knops, R.R.G., van Scheltinga, S.T., Hiemcke-Jiwa, L.S., Flucke, U., Merks, J.H.M., van Noesel, M.M., Tops, B.B.J., Hehir-Kwa, J.Y., Kemmeren, P., Molenaar, J.J., van de Wetering, M., van Boxtel, R., Drost, J.#, Holstege, F.C.P#. Mesenchymal tumor organoid models recapitulate rhabdomyosarcoma subtypes. EMBO Molecular Medicine 2022 Aug 2; e16001. doi: 10.15252/emmm.202216001.

  2. Calandrini, C., van Hooff, S.R., Paassen, I., Ayyildiz, D., Derakhshan, S., Dolman, M.E.M., Langenberg, K.P.S., van de Ven, M., de Heus, C., Liv, N., Kool, M., de Krijger, R.R., Tytgat, G.A.M., van den Heuvel-Eibrink, M.M., Molenaar, J.J., Drost, J. Organoid-based drug screening reveals neddylation as therapeutic target for malignant rhabdoid tumors. Cell Reports 2021 Aug 24; 36(8):109568. doi: 10.1016/j.celrep.2021.109568.

  3. Young, M.D.*, Mitchell, T.J.*, Custers, L.*, Margaritis, T., Morales-Rodriguez, F., Kwakwa, K., Khabirova, E., Kildisiute, G., Oliver, T.R.W., de Krijger, R.R., van den Heuvel-Eibrink, M.M., Comitani, F., Piapi, A., Bugallo-Blanco, E., Thevanesan, C., Burke, C., Prigmore, E., Ambridge, K., Roberts, K., Vieira Braga, F.A., Coorens, T.H.H., Del Valle, I., Wilbrey-Clark, A., Mamanova, L., Stewart, G.D., Gnanapragasam, V.J., Rampling, D., Sebire, N., Coleman, N., Hook, L., Warren, A., Haniffa, M., Kool, M., Pfister, S.M., Achermann, J.C., He, X., Barker, R.A., Shlien, A., Bayraktar, O.A, Teichmann, S., Holstege, F.C., Meyer, K.B., Drost, J.#, Straathof, K.#, Behjati, S.#. Single cell derived mRNA signals across human kidney tumors. Nature Communications 2021 Jun 23; 12(1):3896. doi: 10.1038/s41467-021-23949-5.

  4. Custers, L.*, Khabirova, E.*, Coorens, T.H.H.*, Oliver, T.R.W., Calandrini, C., Young, M.D., Vieira Braga, F.A., Ellis, P., Mamanova, L., Segers, H., Maat, A., Kool, M., Hoving, E.W., van den Heuvel-Eibrink, M.M., Nicholson, J., Straathof, K., Hook, L., de Krijger, R.R., Trayers, C., Allinson, K., Behjati, S.#, Drost, J.#. Somatic mutations and single-cell transcriptomes reveal the root of malignant rhabdoid tumours. Nature Communications 2021 Mar 3; 12(1):1407. doi: 10.1038/s41467-021-21675-6.

  5. Calandrini, C.*, Schutgens, F.*, Oka, R., Margaritis, T., Candelli, T., Mathijsen, L., Ammerlaan, C., van Ineveld, R.L., Derakhshan, S., de Haan, S., Dolman, E., Lijnzaad, P., Custers, L., Begthel, H., Kerstens, H.H.D., Rookmaker, M., Verhaar, M., Tytgat, G.A.M., Kemmeren, P., de Krijger, R.R., Al-Saadi, R., Pritchard-Jones, K., Kool, M., Rios, A., van den Heuvel-Eibrink, M.M., Molenaar, J., van Boxtel, R., Holstege, F.C.P., Clevers, H., Drost, J. An organoid biobank for childhood kidney cancers that captures disease and tissue heterogeneity. Nature Communications 2020;11(1):1310. doi: 10.1038/s41467-020-15155-6.

Members

Jarno Drost
Oncode Investigator
Aleksandra Nenova    
PhD Student
Carla Rios Arceo    
PhD student
Charlotte Op 't Hoog    
Phd student
Giulia Perticari    
PhD student
Jiayou He    
PhD student
Marian Groot Koerkamp    
Research Technician
Mariel Brok    
Research technician
Marjolein Kes    
PhD student
Mark Dings    
PostDoc
Maroussia Ganpat    
PhD Student
Michael Meister    
Clinical Scientist
Nadia Anderson    
Technician
Rugile Januskeviciute    
PhD student
Sofia Doulkeridou    
Research Technician
   
Ruben van Boxtel Group

Cancer Etiology, Mutagenesis and Clonal Evolution

Kép

Our Focus

Our research group focuses on understanding the origins of cancer by decoding and recoding DNA. We address two main questions: 1) why do children develop cancer even though their cells are largely free from damage caused by aging, and 2) what processes cause the DNA mutations that drive carcinogenesis?  

  

To answer these questions, we analyse the DNA of individual cells isolated from patient samples. The DNA code can serve as an historical record, allowing us to trace back and study the moment cancer began. We have developed single-cell DNA analysis methods to decode these archives and uncover critical insights into the life history of cancer.  

  

In parallel, we use genetic modification technologies to recode DNA in stem cells, simulating cancer-driving mutations, or expose these cells to potential carcinogens to study their effects. Together, our work sheds light on preventable causes of cancer and provides unique insights into how cancer arises and sometimes evades treatment.  

About Ruben van Boxtel

My Research

The research group of Ruben van Boxtel has pioneered the development of experimental and computational approaches to study the genomes of human stem cells. Using these methods, his group showed that mutation accumulation varies across different tissues throughout human life. In 2017, Ruben was appointed as a group leader at the Princess Máxima Center for Pediatric Oncology, where he focusses on studying why children get leukemia and lymphoma, and what the genotoxic effects of treatment are in the hematopoietic system. Using in-depth mutational analyses, the group of Ruben has discovered mutational cancer signatures caused by a genotoxic gut bacterium and an antiviral drug.

Ruben received an NWO Vidi award in 2017 by the Netherlands Organization for Scientific Research, which is a prestigious personal Dutch grant for starting group leaders, for his research on studying genomic integrity of human stem cells throughout life. In addition, Ruben received an ERC Consolidator award by the European Research Council in 2019, which is the most prestigious grant for established young research leaders in Europe, to study the causes of therapy-related myeloid neoplasms in childhood cancer survivors. In the same year, he was selected as an Oncode Investigator, which a collective institute bringing together the best fundamental cancer researchers in the Netherlands. In 2022, Ruben received the NYSCF Robertson Stem Cell Investigator Award for his work on studying long-term toxicity in the hematopoietic system of childhood cancer survivors.

Awards
  • 2022: New York Stem Cell Foundation Robertson Stem Cell Investigator Award

  • 2022: Ammodo Science Award for ground-breaking research (team award with Omnes Pro Uno)

  • 2019: ERC Consolidator grant

  • 2019: Selected Oncode member

  • 2017: NWO VIDI Award

Key Publications
  1. Bertrums EJM, Rosendahl Huber AKM, de Kanter JK, Brandsma AM, van Leeuwen AJCN, Verheul M, van den Heuvel-Eibrink MM, Oka R, van Roosmalen MJ, de Groot-Kruseman HA, Zwaan CM, Goemans BF, van Boxtel R. Elevated mutational age in blood of children treated for cancer contributes to therapy-related myeloid neoplasms. Cancer Discovery (2022) 12:1860-1872.

  2. de Kanter JK, Peci F, Bertrums E, Rosendahl Huber A, van Leeuwen A, van Roosmalen MJ, Manders F, Verheul M, Oka R, Brandsma AM, Bierings M, Belderbos M, van Boxtel R. Antiviral treatment causes a unique mutational signature in cancers of transplantation recipients. Cell Stem Cell (2021) 28:1726-1739.e6.

  3. Brandsma AM, Bertrums EJM, van Roosmalen MJ, Hofman DA, Oka R, Verheul M, Manders F, Ubels J, Belderbos ME, van Boxtel R. Mutation signatures of pediatric acute myeloid leukemia and normal blood progenitors associated with differential patient outcomes. Blood Cancer Discovery (2021) 2:484-499.

  4. Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A, van Hoeck A, Wood HM, Nomburg J, Gurjao C, Manders F, Dalmasso G, Stege PB, Paganelli FL, Geurts MH, Beumer J, Mizutani T, van der Linden R, van Elst S; Genomics England Research Consortium, Top J, Willems RJL, Giannakis M, Bonnet R, Quirke P, Meyerson M, Cuppen E, van Boxtel R*,Clevers H*. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature (2020) 580:269-273. *co-corresponding authors

  5. Osorio FG, Rosendahl Huber A, Oka R, Verheul M, Patel SH, Karlijn Hasaart K, de la Fonteijne L, Varela I, Camargo FD, van Boxtel R. Somatic Mutations Reveal Lineage Relationships
    and Age-Related Mutagenesis in Human Hematopoiesis. Cell Reports (2018) 25: 2308-2316

  6. Blokzijl F, de Ligt J, Jager M, Sasselli V, Roerink S, Sasaki N, Huch M, Boymans S, Kuijk E, Prins P, Nijman IJ, Martincorena I, Mokry M, Wiegerinck CL, Middendorp S, Sato T, Schwank G, Nieuwenhuis EE, Verstegen MM, van der Laan LJ, de Jonge J, IJzermans JN, Vries RG, van de Wetering M, Stratton MR, Clevers H, Cuppen E, van Boxtel R. Tissue-specific mutation accumulation in human adult stem cells during life. Nature (2016) 538:260-264.

Members

Ruben van Boxtel
Group leader
Alexander Steemers 
PhD student

Anaïs van Leeuwen 
Phd student

Annemarie Rietman 
Scientific Research Coordinator

Diego Montiel González 
Bioinformatician
Bo Scherer    
PostDoc
Eirini Daskalaki 
PhD student
Elizaveta Kulaeva 
PhD student
Laurianne Trabut 
Research Technician
Lucca Derks 
Phd candidate

Maarten Geurts 
PostDoc

Mark van Roosmalen 
Bio informaticus

Mark Verheul 
Technician

Matteo Boretto 
PostDoc
Menno Tooren
PhD Student
Rico Hagelaar
Bioinformatician