Network


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

Hotspot


Dive into the research topics where Péter Hauser is active.

Publication


Featured researches published by Péter Hauser.


Nature | 2012

Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma

Jeremy Schwartzentruber; Andrey Korshunov; Xiao Yang Liu; David T. W. Jones; Elke Pfaff; Karine Jacob; Dominik Sturm; Adam M. Fontebasso; Dong Anh Khuong Quang; Martje Tönjes; Volker Hovestadt; Steffen Albrecht; Marcel Kool; André Nantel; Carolin Konermann; Anders M. Lindroth; Natalie Jäger; Tobias Rausch; Marina Ryzhova; Jan O. Korbel; Thomas Hielscher; Péter Hauser; Miklós Garami; Almos Klekner; László Bognár; Martin Ebinger; Martin U. Schuhmann; Wolfram Scheurlen; Arnulf Pekrun; Michael C. Frühwald

Glioblastoma multiforme (GBM) is a lethal brain tumour in adults and children. However, DNA copy number and gene expression signatures indicate differences between adult and paediatric cases. To explore the genetic events underlying this distinction, we sequenced the exomes of 48 paediatric GBM samples. Somatic mutations in the H3.3-ATRX-DAXX chromatin remodelling pathway were identified in 44% of tumours (21/48). Recurrent mutations in H3F3A, which encodes the replication-independent histone 3 variant H3.3, were observed in 31% of tumours, and led to amino acid substitutions at two critical positions within the histone tail (K27M, G34R/G34V) involved in key regulatory post-translational modifications. Mutations in ATRX (α-thalassaemia/mental retardation syndrome X-linked) and DAXX (death-domain associated protein), encoding two subunits of a chromatin remodelling complex required for H3.3 incorporation at pericentric heterochromatin and telomeres, were identified in 31% of samples overall, and in 100% of tumours harbouring a G34R or G34V H3.3 mutation. Somatic TP53 mutations were identified in 54% of all cases, and in 86% of samples with H3F3A and/or ATRX mutations. Screening of a large cohort of gliomas of various grades and histologies (n = 784) showed H3F3A mutations to be specific to GBM and highly prevalent in children and young adults. Furthermore, the presence of H3F3A/ATRX-DAXX/TP53 mutations was strongly associated with alternative lengthening of telomeres and specific gene expression profiles. This is, to our knowledge, the first report to highlight recurrent mutations in a regulatory histone in humans, and our data suggest that defects of the chromatin architecture underlie paediatric and young adult GBM pathogenesis.


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.


Nature Genetics | 2014

Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma

Adam M. Fontebasso; Simon Papillon-Cavanagh; Jeremy Schwartzentruber; Hamid Nikbakht; Noha Gerges; Pierre‑Olivier Fiset; Denise Bechet; Damien Faury; Nicolas De Jay; Lori A. Ramkissoon; Aoife Corcoran; David T. W. Jones; Dominik Sturm; Pascal Johann; Tadanori Tomita; Stewart Goldman; Mahmoud Nagib; Liliana Goumnerova; Daniel C. Bowers; Jeffrey R. Leonard; Joshua B. Rubin; Tord D. Alden; Samuel R. Browd; J. Russell Geyer; Sarah Leary; George I. Jallo; Kenneth Cohen; Nalin Gupta; Michael D. Prados; Anne Sophie Carret

Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease.


Journal of Clinical Oncology | 2007

Molecular Profiling Identifies Prognostic Subgroups of Pediatric Glioblastoma and Shows Increased YB-1 Expression in Tumors

Damien Faury; André Nantel; Sandra E. Dunn; Marie Christine Guiot; Takrima Haque; Péter Hauser; Miklós Garami; László Bognár; Zoltán Hanzély; Pawel P. Liberski; Enrique López-Aguilar; Elvis Terci Valera; Luis G. Tone; Anne Sophie Carret; Rolando F. Del Maestro; Martin Gleave; Jose Luis Montes; Torsten Pietsch; Stephen Albrecht; Nada Jabado

PURPOSE Pediatric glioblastoma (pGBM) is a rare, but devastating brain tumor. In contrast to GBM in adults (aGBM), little is known about the mechanisms underlying its development. Our aim is to gain insight into the molecular pathways of pGBM. MATERIALS AND METHODS Thirty-two pGBM and seven aGBM samples were investigated using biochemical and transcriptional profiling. Ras and Akt pathway activation was assessed through the phosphorylation of downstream effectors, and gene expression profiles were generated using the University Health Network Human 19K cDNA arrays. Results were validated using real-time polymerase chain reaction and immunohistochemistry and compared with existing data sets on aGBM. RESULTS There are at least two subsets of pGBM. One subset, associated with Ras and Akt pathway activation, has very poor prognosis and exhibits increased expression of genes related to proliferation and to a neural stem-cell phenotype, similar to findings in aggressive aGBM. This subset was still molecularly distinguishable from aGBM after unsupervised and supervised analysis of expression profiles. A second subset, with better prognosis, is not associated with activation of Akt and Ras pathways, may originate from astroglial progenitors, and does not express gene signatures and markers shown to be associated with long-term survival in aGBM. Both subsets of pGBM show overexpression of Y-box-protein-1 that may help drive oncogenesis in this tumor. CONCLUSION Our work, the first study of gene expression profiles in pGBM, provides valuable insight into active pathways and targets in a cancer with minimal survival, and suggests that these tumors cannot be understood exclusively through studies of aGBM.


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.


Journal of Clinical Oncology | 2013

Subgroup-Specific Prognostic Implications of TP53 Mutation in Medulloblastoma

Nataliya Zhukova; Vijay Ramaswamy; Marc Remke; Elke Pfaff; David Shih; Dianna Martin; Pedro Castelo-Branco; Berivan Baskin; Peter N. Ray; Eric Bouffet; André O. von Bueren; David Jones; Paul A. Northcott; Marcel Kool; Dominik Sturm; Trevor J. Pugh; Scott L. Pomeroy; Yoon-Jae Cho; Torsten Pietsch; Marco Gessi; Stefan Rutkowski; László Bognár; Almos Klekner; Byung Kyu Cho; Seung Ki Kim; Kyu Chang Wang; Charles G. Eberhart; Michelle Fèvre-Montange; Maryam Fouladi; Pim J. French

PURPOSE Reports detailing the prognostic impact of TP53 mutations in medulloblastoma offer conflicting conclusions. We resolve this issue through the inclusion of molecular subgroup profiles. PATIENTS AND METHODS We determined subgroup affiliation, TP53 mutation status, and clinical outcome in a discovery cohort of 397 medulloblastomas. We subsequently validated our results on an independent cohort of 156 medulloblastomas. RESULTS TP53 mutations are enriched in wingless (WNT; 16%) and sonic hedgehog (SHH; 21%) medulloblastomas and are virtually absent in subgroups 3 and 4 tumors (P < .001). Patients with SHH/TP53 mutant tumors are almost exclusively between ages 5 and 18 years, dramatically different from the general SHH distribution (P < .001). Children with SHH/TP53 mutant tumors harbor 56% germline TP53 mutations, which are not observed in children with WNT/TP53 mutant tumors. Five-year overall survival (OS; ± SE) was 41% ± 9% and 81% ± 5% for patients with SHH medulloblastomas with and without TP53 mutations, respectively (P < .001). Furthermore, TP53 mutations accounted for 72% of deaths in children older than 5 years with SHH medulloblastomas. In contrast, 5-year OS rates were 90% ± 9% and 97% ± 3% for patients with WNT tumors with and without TP53 mutations (P = .21). Multivariate analysis revealed that TP53 status was the most important risk factor for SHH medulloblastoma. Survival rates in the validation cohort mimicked the discovery results, revealing that poor survival of TP53 mutations is restricted to patients with SHH medulloblastomas (P = .012) and not WNT tumors. CONCLUSION Subgroup-specific analysis reconciles prior conflicting publications and confirms that TP53 mutations are enriched among SHH medulloblastomas, in which they portend poor outcome and account for a large proportion of treatment failures in these patients.


British Journal of Cancer | 2009

Duplication of 7q34 is specific to juvenile pilocytic astrocytomas and a hallmark of cerebellar and optic pathway tumours

Karine Jacob; Steffen Albrecht; C. Sollier; Damien Faury; E. Sader; Alexandre Montpetit; D. Serre; Péter Hauser; Miklós Garami; László Bognár; Zoltán Hanzély; Jose Luis Montes; Jeffrey Atkinson; Jean Pierre Farmer; Eric Bouffet; Cynthia Hawkins; Uri Tabori; Nada Jabado

Background:Juvenile pilocytic astrocytomas (JPA), a subgroup of low-grade astrocytomas (LGA), are common, heterogeneous and poorly understood subset of brain tumours in children. Chromosomal 7q34 duplication leading to fusion genes formed between KIAA1549 and BRAF and subsequent constitutive activation of BRAF was recently identified in a proportion of LGA, and may be involved in their pathogenesis. Our aim was to investigate additional chromosomal unbalances in LGA and whether incidence of 7q34 duplication is associated with tumour type or location.Methods and results:Using Illumina-Human-Hap300-Duo and 610-Quad high-resolution-SNP-based arrays and quantitative PCR on genes of interest, we investigated 84 paediatric LGA. We demonstrate that 7q34 duplication is specific to sporadic JPA (35 of 53 – 66%) and does not occur in other LGA subtypes (0 of 27) or NF1-associated-JPA (0 of 4). We also establish that it is site specific as it occurs in the majority of cerebellar JPA (24 of 30 – 80%) followed by brainstem, hypothalamic/optic pathway JPA (10 of 16 – 62.5%) and is rare in hemispheric JPA (1 of 7 – 14%). The MAP-kinase pathway, assessed through ERK phosphorylation, was active in all tumours regardless of 7q34 duplication. Gain of function studies performed on hTERT-immortalised astrocytes show that overexpression of wild-type BRAF does not increase cell proliferation or baseline MAPK signalling even if it sensitises cells to EGFR stimulation.Conclusions and interpretation:Our results suggest that variants of JPA might arise from a unique site-restricted progenitor cell where 7q34 duplication, a hallmark of this tumour-type in association to MAPK-kinase pathway activation, potentially plays a site-specific role in their pathogenesis. Importantly, gain of function abnormalities in components of MAP-Kinase signalling are potentially present in all JPA making this tumour amenable to therapeutic targeting of this pathway.


Journal of Clinical Oncology | 2014

Cytogenetic Prognostication Within Medulloblastoma Subgroups

David Shih; Paul A. Northcott; Marc Remke; Andrey Korshunov; Vijay Ramaswamy; Marcel Kool; Betty Luu; Yuan Yao; Xin Wang; Adrian Dubuc; Livia Garzia; John Peacock; Stephen C. Mack; Xiaochong Wu; Adi Rolider; A. Sorana Morrissy; Florence M.G. Cavalli; David T. W. Jones; Karel Zitterbart; Claudia C. Faria; Ulrich Schüller; Leos Kren; Toshihiro Kumabe; Teiji Tominaga; Young Shin Ra; Miklós Garami; Péter Hauser; Jennifer A. Chan; Shenandoah Robinson; László Bognár

PURPOSE Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. PATIENTS AND METHODS Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. RESULTS Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. CONCLUSION Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials.


Nature Genetics | 2014

Fusion of TTYH1 with the C19MC microRNA cluster drives expression of a brain-specific DNMT3B isoform in the embryonal brain tumor ETMR

Claudia L. Kleinman; Noha Gerges; Simon Papillon-Cavanagh; Patrick Sin-Chan; Albena Pramatarova; Dong Anh Khuong Quang; Véronique Adoue; Stephan Busche; Maxime Caron; Haig Djambazian; Amandine Bemmo; Adam M. Fontebasso; Tara Spence; Jeremy Schwartzentruber; Steffen Albrecht; Péter Hauser; Miklós Garami; Almos Klekner; László Bognár; Jose Luis Montes; Alfredo Staffa; Alexandre Montpetit; Pierre Bérubé; Magdalena Zakrzewska; Krzysztof Zakrzewski; Pawel P. Liberski; Zhifeng Dong; Peter M. Siegel; Thomas F. Duchaine; Christian Perotti

Embryonal tumors with multilayered rosettes (ETMRs) are rare, deadly pediatric brain tumors characterized by high-level amplification of the microRNA cluster C19MC. We performed integrated genetic and epigenetic analyses of 12 ETMR samples and identified, in all cases, C19MC fusions to TTYH1 driving expression of the microRNAs. ETMR tumors, cell lines and xenografts showed a specific DNA methylation pattern distinct from those of other tumors and normal tissues. We detected extreme overexpression of a previously uncharacterized isoform of DNMT3B originating at an alternative promoter that is active only in the first weeks of neural tube development. Transcriptional and immunohistochemical analyses suggest that C19MC-dependent DNMT3B deregulation is mediated by RBL2, a known repressor of DNMT3B. Transfection with individual C19MC microRNAs resulted in DNMT3B upregulation and RBL2 downregulation in cultured cells. Our data suggest a potential oncogenic re-engagement of an early developmental program in ETMR via epigenetic alteration mediated by an embryonic, brain-specific DNMT3B isoform.


Cancer Cell | 2017

Intertumoral Heterogeneity within Medulloblastoma Subgroups

Florence M.G. Cavalli; Marc Remke; Ladislav Rampasek; John Peacock; David Shih; Betty Luu; Livia Garzia; Jonathon Torchia; Carolina Nör; A. Sorana Morrissy; Sameer Agnihotri; Yuan Yao Thompson; Claudia M. Kuzan-Fischer; Hamza Farooq; Keren Isaev; Craig Daniels; Byung Kyu Cho; Seung Ki Kim; Kyu Chang Wang; Ji Yeoun Lee; Wieslawa A. Grajkowska; Marta Perek-Polnik; Alexandre Vasiljevic; Cécile Faure-Conter; Anne Jouvet; Caterina Giannini; Amulya A. Nageswara Rao; Kay Ka Wai Li; Ho Keung Ng; Charles G. Eberhart

While molecular subgrouping has revolutionized medulloblastoma classification, the extent of heterogeneity within subgroups is unknown. Similarity network fusion (SNF) applied to genome-wide DNA methylation and gene expression data across 763 primary samples identifies very homogeneous clusters of patients, supporting the presence of medulloblastoma subtypes. After integration of somatic copy-number alterations, and clinical features specific to each cluster, we identify 12 different subtypes of medulloblastoma. Integrative analysis using SNF further delineates group 3 from group 4 medulloblastoma, which is not as readily apparent through analyses of individual data types. Two clear subtypes of infants with Sonic Hedgehog medulloblastoma with disparate outcomes and biology are identified. Medulloblastoma subtypes identified through integrative clustering have important implications for stratification of future clinical trials.

Collaboration


Dive into the Péter Hauser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
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
Top Co-Authors

Avatar

David T. W. Jones

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrey Korshunov

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge