Network


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

Hotspot


Dive into the research topics where Peter van Sluis is active.

Publication


Featured researches published by Peter van Sluis.


Cancer Cell | 2012

Hotspot Mutations in H3F3A and IDH1 Define Distinct Epigenetic and Biological Subgroups of Glioblastoma

Dominik Sturm; Hendrik Witt; Volker Hovestadt; Dong Anh Khuong-Quang; David T. W. Jones; Carolin Konermann; Elke Pfaff; Martje Tönjes; Martin Sill; Sebastian Bender; Marcel Kool; Marc Zapatka; Natalia Becker; Manuela Zucknick; Thomas Hielscher; Xiao Yang Liu; Adam M. Fontebasso; Marina Ryzhova; Steffen Albrecht; Karine Jacob; Marietta Wolter; Martin Ebinger; Martin U. Schuhmann; Timothy Van Meter; Michael C. Frühwald; Holger Hauch; Arnulf Pekrun; Bernhard Radlwimmer; Tim Niehues; Gregor Von Komorowski

Glioblastoma (GBM) is a brain tumor that carries a dismal prognosis and displays considerable heterogeneity. We have recently identified recurrent H3F3A mutations affecting two critical amino acids (K27 and G34) of histone H3.3 in one-third of pediatric GBM. Here, we show that each H3F3A mutation defines an epigenetic subgroup of GBM with a distinct global methylation pattern, and that they are mutually exclusive with IDH1 mutations, which characterize a third mutation-defined subgroup. Three further epigenetic subgroups were enriched for hallmark genetic events of adult GBM and/or established transcriptomic signatures. We also demonstrate that the two H3F3A mutations give rise to GBMs in separate anatomic compartments, with differential regulation of transcription factors OLIG1, OLIG2, and FOXG1, possibly reflecting different cellular origins.


PLOS ONE | 2008

Integrated Genomics Identifies Five Medulloblastoma Subtypes with Distinct Genetic Profiles, Pathway Signatures and Clinicopathological Features

Marcel Kool; Jan Koster; Jens Bunt; Nancy E. Hasselt; Arjan Lakeman; Peter van Sluis; Dirk Troost; Netteke Schouten-van Meeteren; Huib N. Caron; Jacqueline Cloos; Alan Mršić; Bauke Ylstra; Wieslawa A. Grajkowska; Wolfgang Hartmann; Torsten Pietsch; David W. Ellison; Steven C. Clifford; Rogier Versteeg

Background Medulloblastoma is the most common malignant brain tumor in children. Despite recent improvements in cure rates, prediction of disease outcome remains a major challenge and survivors suffer from serious therapy-related side-effects. Recent data showed that patients with WNT-activated tumors have a favorable prognosis, suggesting that these patients could be treated less intensively, thereby reducing the side-effects. This illustrates the potential benefits of a robust classification of medulloblastoma patients and a detailed knowledge of associated biological mechanisms. Methods and Findings To get a better insight into the molecular biology of medulloblastoma we established mRNA expression profiles of 62 medulloblastomas and analyzed 52 of them also by comparative genomic hybridization (CGH) arrays. Five molecular subtypes were identified, characterized by WNT signaling (A; 9 cases), SHH signaling (B; 15 cases), expression of neuronal differentiation genes (C and D; 16 and 11 cases, respectively) or photoreceptor genes (D and E; both 11 cases). Mutations in β-catenin were identified in all 9 type A tumors, but not in any other tumor. PTCH1 mutations were exclusively identified in type B tumors. CGH analysis identified several fully or partly subtype-specific chromosomal aberrations. Monosomy of chromosome 6 occurred only in type A tumors, loss of 9q mostly occurred in type B tumors, whereas chromosome 17 aberrations, most common in medulloblastoma, were strongly associated with type C or D tumors. Loss of the inactivated X-chromosome was highly specific for female cases of type C, D and E tumors. Gene expression levels faithfully reflected the chromosomal copy number changes. Clinicopathological features significantly different between the 5 subtypes included metastatic disease and age at diagnosis and histology. Metastatic disease at diagnosis was significantly associated with subtypes C and D and most strongly with subtype E. Patients below 3 yrs of age had type B, D, or E tumors. Type B included most desmoplastic cases. We validated and confirmed the molecular subtypes and their associated clinicopathological features with expression data from a second independent series of 46 medulloblastomas. Conclusions The new medulloblastoma classification presented in this study will greatly enhance the understanding of this heterogeneous disease. It will enable a better selection and evaluation of patients in clinical trials, and it will support the development of new molecular targeted therapies. Ultimately, our results may lead to more individualized therapies with improved cure rates and a better quality of life.


Nature | 2012

Dissecting the genomic complexity underlying medulloblastoma

David T. W. Jones; Natalie Jäger; Marcel Kool; Thomas Zichner; Barbara Hutter; Marc Sultan; Yoon-Jae Cho; Trevor J. Pugh; Volker Hovestadt; Adrian M. Stütz; Tobias Rausch; Hans-Jörg Warnatz; Marina Ryzhova; Sebastian Bender; Dominik Sturm; Sabrina Pleier; Huriye Cin; Elke Pfaff; Laura Sieber; Andrea Wittmann; Marc Remke; Hendrik Witt; Sonja Hutter; Theophilos Tzaridis; Joachim Weischenfeldt; Benjamin Raeder; Meryem Avci; Vyacheslav Amstislavskiy; Marc Zapatka; Ursula Weber

Medulloblastoma is an aggressively growing tumour, arising in the cerebellum or medulla/brain stem. It is the most common malignant brain tumour in children, and shows tremendous biological and clinical heterogeneity. Despite recent treatment advances, approximately 40% of children experience tumour recurrence, and 30% will die from their disease. Those who survive often have a significantly reduced quality of life. Four tumour subgroups with distinct clinical, biological and genetic profiles are currently identified. WNT tumours, showing activated wingless pathway signalling, carry a favourable prognosis under current treatment regimens. SHH tumours show hedgehog pathway activation, and have an intermediate prognosis. Group 3 and 4 tumours are molecularly less well characterized, and also present the greatest clinical challenges. The full repertoire of genetic events driving this distinction, however, remains unclear. Here we describe an integrative deep-sequencing analysis of 125 tumour–normal pairs, conducted as part of the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. Tetraploidy was identified as a frequent early event in Group 3 and 4 tumours, and a positive correlation between patient age and mutation rate was observed. Several recurrent mutations were identified, both in known medulloblastoma-related genes (CTNNB1, PTCH1, MLL2, SMARCA4) and in genes not previously linked to this tumour (DDX3X, CTDNEP1, KDM6A, TBR1), often in subgroup-specific patterns. RNA sequencing confirmed these alterations, and revealed the expression of what are, to our knowledge, the first medulloblastoma fusion genes identified. Chromatin modifiers were frequently altered across all subgroups. These findings enhance our understanding of the genomic complexity and heterogeneity underlying medulloblastoma, and provide several potential targets for new therapeutics, especially for Group 3 and 4 patients.


Nature | 2012

Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes

Jan J. Molenaar; Jan Koster; Danny A. Zwijnenburg; Peter van Sluis; Linda J. Valentijn; Ida van der Ploeg; Mohamed Hamdi; Johan van Nes; Bart A. Westerman; Jennemiek van Arkel; Marli E. Ebus; Franciska Haneveld; Arjan Lakeman; Linda Schild; Piet Molenaar; Peter Stroeken; Max M. van Noesel; Ingrid Øra; Evan E. Santo; Huib N. Caron; Ellen M. Westerhout; Rogier Versteeg

Neuroblastoma is a childhood tumour of the peripheral sympathetic nervous system. The pathogenesis has for a long time been quite enigmatic, as only very few gene defects were identified in this often lethal tumour. Frequently detected gene alterations are limited to MYCN amplification (20%) and ALK activations (7%). Here we present a whole-genome sequence analysis of 87 neuroblastoma of all stages. Few recurrent amino-acid-changing mutations were found. In contrast, analysis of structural defects identified a local shredding of chromosomes, known as chromothripsis, in 18% of high-stage neuroblastoma. These tumours are associated with a poor outcome. Structural alterations recurrently affected ODZ3, PTPRD and CSMD1, which are involved in neuronal growth cone stabilization. In addition, ATRX, TIAM1 and a series of regulators of the Rac/Rho pathway were mutated, further implicating defects in neuritogenesis in neuroblastoma. Most tumours with defects in these genes were aggressive high-stage neuroblastomas, but did not carry MYCN amplifications. The genomic landscape of neuroblastoma therefore reveals two novel molecular defects, chromothripsis and neuritogenesis gene alterations, which frequently occur in high-risk tumours.


The EMBO Journal | 2001

N‐myc enhances the expression of a large set of genes functioning in ribosome biogenesis and protein synthesis

Kathy Boon; Huib N. Caron; Ronald van Asperen; Linda J. Valentijn; Marie Christine Hermus; Peter van Sluis; Ilja Roobeek; Isabel Weis; P.A. Voûte; Manfred Schwab; Rogier Versteeg

The myc oncogenes are frequently activated in human tumors, but there is no comprehensive insight into the target genes and downstream cellular pathways of these transcription factors. We applied serial analysis of gene expression (SAGE) to identify targets of N‐myc in neuroblastomas. Analysis of 42 000 mRNA transcript tags in SAGE libraries of N‐myc‐ transfected and control neuroblastoma cells revealed 114 up‐regulated genes. The majority of these genes have a role in ribosome assembly and activity. Northern blot analysis confirmed up‐regulation of all tested transcripts. Induction was complete within 4 h after N‐myc expression. The large majority of the ribosomal proteins were induced, as well as genes controlling rRNA maturation. Cellular rRNA content was 45% induced. SAGE libraries and northern blot analysis confirmed up‐regulation of many of these genes in N‐myc‐amplified neuroblastomas. As N‐myc can functionally replace c‐myc, we analyzed whether N‐myc targets were induced by c‐myc as well. Approximately 40% of these N‐myc targets were up‐regulated in a c‐myc‐transfected melanoma cell line. These data suggest that myc genes function as major regulators of the protein synthesis machinery.


Nature Genetics | 2013

Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma

David T. W. Jones; Barbara Hutter; Natalie Jäger; Andrey Korshunov; Marcel Kool; Hans-Jörg Warnatz; Thomas Zichner; Sally R. Lambert; Marina Ryzhova; Dong Anh Khuong Quang; Adam M. Fontebasso; Adrian M. Stütz; Sonja Hutter; Marc Zuckermann; Dominik Sturm; Jan Gronych; Bärbel Lasitschka; Sabine Schmidt; Huriye Şeker-Cin; Hendrik Witt; Marc Sultan; Meryem Ralser; Paul A. Northcott; Volker Hovestadt; Sebastian Bender; Elke Pfaff; Sebastian Stark; Damien Faury; Jeremy Schwartzentruber; Jacek Majewski

Pilocytic astrocytoma, the most common childhood brain tumor, is typically associated with mitogen-activated protein kinase (MAPK) pathway alterations. Surgically inaccessible midline tumors are therapeutically challenging, showing sustained tendency for progression and often becoming a chronic disease with substantial morbidities. Here we describe whole-genome sequencing of 96 pilocytic astrocytomas, with matched RNA sequencing (n = 73), conducted by the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. We identified recurrent activating mutations in FGFR1 and PTPN11 and new NTRK2 fusion genes in non-cerebellar tumors. New BRAF-activating changes were also observed. MAPK pathway alterations affected all tumors analyzed, with no other significant mutations identified, indicating that pilocytic astrocytoma is predominantly a single-pathway disease. Notably, we identified the same FGFR1 mutations in a subset of H3F3A-mutated pediatric glioblastoma with additional alterations in the NF1 gene. Our findings thus identify new potential therapeutic targets in distinct subsets of pilocytic astrocytoma and childhood glioblastoma.


Nature Genetics | 2012

LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression

Jan J. Molenaar; Raquel Domingo-Fernández; Marli E. Ebus; Sven Lindner; Jan Koster; Ksenjia Drabek; Pieter Mestdagh; Peter van Sluis; Linda J. Valentijn; Johan van Nes; Marloes Broekmans; Franciska Haneveld; Richard Volckmann; Isabella Bray; Lukas C. Heukamp; Annika Sprüssel; Theresa Thor; Kristina Kieckbusch; Ludger Klein-Hitpass; Matthias Fischer; Jo Vandesompele; Alexander Schramm; Max M. van Noesel; Luigi Varesio; Franki Speleman; Angelika Eggert; Raymond L. Stallings; Huib N. Caron; Rogier Versteeg; Johannes H. Schulte

LIN28B regulates developmental processes by modulating microRNAs (miRNAs) of the let-7 family. A role for LIN28B in cancer has been proposed but has not been established in vivo. Here, we report that LIN28B showed genomic aberrations and extensive overexpression in high-risk neuroblastoma compared to several other tumor entities and normal tissues. High LIN28B expression was an independent risk factor for adverse outcome in neuroblastoma. LIN28B signaled through repression of the let-7 miRNAs and consequently resulted in elevated MYCN protein expression in neuroblastoma cells. LIN28B–let-7–MYCN signaling blocked differentiation of normal neuroblasts and neuroblastoma cells. These findings were fully recapitulated in a mouse model in which LIN28B expression in the sympathetic adrenergic lineage induced development of neuroblastomas marked by low let-7 miRNA levels and high MYCN protein expression. Interference with this pathway might offer therapeutic perspectives.


Nature | 2014

Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma.

Paul A. Northcott; C A Lee; Thomas Zichner; Adrian M. Stütz; Serap Erkek; Daisuke Kawauchi; David Shih; Volker Hovestadt; Marc Zapatka; Dominik Sturm; David T. W. Jones; Marcel Kool; Marc Remke; Florence M.G. Cavalli; Scott Zuyderduyn; Gary D. Bader; Scott R. VandenBerg; Lourdes Adriana Esparza; Marina Ryzhova; Wei Wang; Andrea Wittmann; Sebastian Stark; Laura Sieber; Huriye Seker-Cin; Linda Linke; Fabian Kratochwil; Natalie Jäger; Ivo Buchhalter; Charles D. Imbusch; Gideon Zipprich

Medulloblastoma is a highly malignant paediatric brain tumour currently treated with a combination of surgery, radiation and chemotherapy, posing a considerable burden of toxicity to the developing child. Genomics has illuminated the extensive intertumoral heterogeneity of medulloblastoma, identifying four distinct molecular subgroups. Group 3 and group 4 subgroup medulloblastomas account for most paediatric cases; yet, oncogenic drivers for these subtypes remain largely unidentified. Here we describe a series of prevalent, highly disparate genomic structural variants, restricted to groups 3 and 4, resulting in specific and mutually exclusive activation of the growth factor independent 1 family proto-oncogenes, GFI1 and GFI1B. Somatic structural variants juxtapose GFI1 or GFI1B coding sequences proximal to active enhancer elements, including super-enhancers, instigating oncogenic activity. Our results, supported by evidence from mouse models, identify GFI1 and GFI1B as prominent medulloblastoma oncogenes and implicate ‘enhancer hijacking’ as an efficient mechanism driving oncogene activation in a childhood cancer.


Nature Genetics | 2015

Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations

Thomas F. Eleveld; Derek A. Oldridge; Virginie Bernard; Jan Koster; Leo Colmet Daage; Sharon J. Diskin; Linda Schild; Nadia Bessoltane Bentahar; Angela Bellini; Mathieu Chicard; Eve Lapouble; Valérie Combaret; Patricia Legoix-Né; Jean Michon; Trevor J. Pugh; Lori S. Hart; JulieAnn Rader; Edward F. Attiyeh; Jun S. Wei; Shile Zhang; Arlene Naranjo; Julie M. Gastier-Foster; Michael D. Hogarty; Shahab Asgharzadeh; Malcolm A. Smith; Jaime M. Guidry Auvil; Thomas B. K. Watkins; Danny A. Zwijnenburg; Marli E. Ebus; Peter van Sluis

The majority of patients with neuroblastoma have tumors that initially respond to chemotherapy, but a large proportion will experience therapy-resistant relapses. The molecular basis of this aggressive phenotype is unknown. Whole-genome sequencing of 23 paired diagnostic and relapse neuroblastomas showed clonal evolution from the diagnostic tumor, with a median of 29 somatic mutations unique to the relapse sample. Eighteen of the 23 relapse tumors (78%) showed mutations predicted to activate the RAS-MAPK pathway. Seven of these events were detected only in the relapse tumor, whereas the others showed clonal enrichment. In neuroblastoma cell lines, we also detected a high frequency of activating mutations in the RAS-MAPK pathway (11/18; 61%), and these lesions predicted sensitivity to MEK inhibition in vitro and in vivo. Our findings provide a rationale for genetic characterization of relapse neuroblastomas and show that RAS-MAPK pathway mutations may function as a biomarker for new therapeutic approaches to refractory disease.


Cancer Cell | 2016

Atypical Teratoid/Rhabdoid Tumors Are Comprised of Three Epigenetic Subgroups with Distinct Enhancer Landscapes

Pascal Johann; Serap Erkek; Marc Zapatka; Kornelius Kerl; Ivo Buchhalter; Volker Hovestadt; David T. W. Jones; Dominik Sturm; Carl Hermann; Maia Segura Wang; Andrey Korshunov; Marina Rhyzova; Susanne Gröbner; Sebastian Brabetz; Lukas Chavez; Susanne Bens; Stefan Gröschel; Fabian Kratochwil; Andrea Wittmann; Laura Sieber; Christina Geörg; Stefan Wolf; Katja Beck; Florian Oyen; David Capper; Peter van Sluis; Richard Volckmann; Jan Koster; Rogier Versteeg; Andreas von Deimling

Atypical teratoid/rhabdoid tumor (ATRT) is one of the most common brain tumors in infants. Although the prognosis of ATRT patients is poor, some patients respond favorably to current treatments, suggesting molecular inter-tumor heterogeneity. To investigate this further, we genetically and epigenetically analyzed 192 ATRTs. Three distinct molecular subgroups of ATRTs, associated with differences in demographics, tumor location, and type of SMARCB1 alterations, were identified. Whole-genome DNA and RNA sequencing found no recurrent mutations in addition to SMARCB1 that would explain the differences between subgroups. Whole-genome bisulfite sequencing and H3K27Ac chromatin-immunoprecipitation sequencing of primary tumors, however, revealed clear differences, leading to the identification of subgroup-specific regulatory networks and potential therapeutic targets.

Collaboration


Dive into the Peter van Sluis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Koster

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Huib N. Caron

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alvin Chan

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marcel Kool

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

David T. W. Jones

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge