Network


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

Hotspot


Dive into the research topics where Ivo Buchhalter is active.

Publication


Featured researches published by Ivo Buchhalter.


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.


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.


Nature Communications | 2015

A comprehensive assessment of somatic mutation detection in cancer using whole-genome sequencing

Tyler Alioto; Ivo Buchhalter; Sophia Derdak; Barbara Hutter; Matthew Eldridge; Eivind Hovig; Lawrence E. Heisler; Timothy Beck; Jared T. Simpson; Laurie Tonon; Anne Sophie Sertier; Ann Marie Patch; Natalie Jäger; Philip Ginsbach; Ruben M. Drews; Nagarajan Paramasivam; Rolf Kabbe; Sasithorn Chotewutmontri; Nicolle Diessl; Christopher Previti; Sabine Schmidt; Benedikt Brors; Lars Feuerbach; Michael Heinold; Susanne Gröbner; Andrey Korshunov; Patrick Tarpey; Adam Butler; Jonathan Hinton; David Jones

As whole-genome sequencing for cancer genome analysis becomes a clinical tool, a full understanding of the variables affecting sequencing analysis output is required. Here using tumour-normal sample pairs from two different types of cancer, chronic lymphocytic leukaemia and medulloblastoma, we conduct a benchmarking exercise within the context of the International Cancer Genome Consortium. We compare sequencing methods, analysis pipelines and validation methods. We show that using PCR-free methods and increasing sequencing depth to ∼100 × shows benefits, as long as the tumour:control coverage ratio remains balanced. We observe widely varying mutation call rates and low concordance among analysis pipelines, reflecting the artefact-prone nature of the raw data and lack of standards for dealing with the artefacts. However, we show that, using the benchmark mutation set we have created, many issues are in fact easy to remedy and have an immediate positive impact on mutation detection accuracy.


Nature | 2016

Active medulloblastoma enhancers reveal subgroup-specific cellular origins

Charles Y. Lin; Serap Erkek; Yiai Tong; Linlin Yin; Alexander J. Federation; Marc Zapatka; Parthiv Haldipur; Daisuke Kawauchi; Thomas Risch; Hans Jörg Warnatz; Barbara C. Worst; Bensheng Ju; Brent A. Orr; Rhamy Zeid; Donald R. Polaski; Maia Segura-Wang; Sebastian M. Waszak; David T. W. Jones; Marcel Kool; Volker Hovestadt; Ivo Buchhalter; Laura Sieber; Pascal Johann; Lukas Chavez; Stefan Gröschel; Marina Ryzhova; Andrey Korshunov; Wenbiao Chen; Victor V. Chizhikov; Kathleen J. Millen

Medulloblastoma is a highly malignant paediatric brain tumour, often inflicting devastating consequences on the developing child. Genomic studies have revealed four distinct molecular subgroups with divergent biology and clinical behaviour. An understanding of the regulatory circuitry governing the transcriptional landscapes of medulloblastoma subgroups, and how this relates to their respective developmental origins, is lacking. Here, using H3K27ac and BRD4 chromatin immunoprecipitation followed by sequencing (ChIP-seq) coupled with tissue-matched DNA methylation and transcriptome data, we describe the active cis-regulatory landscape across 28 primary medulloblastoma specimens. Analysis of differentially regulated enhancers and super-enhancers reinforced inter-subgroup heterogeneity and revealed novel, clinically relevant insights into medulloblastoma biology. Computational reconstruction of core regulatory circuitry identified a master set of transcription factors, validated by ChIP-seq, that is responsible for subgroup divergence, and implicates candidate cells of origin for Group 4. Our integrated analysis of enhancer elements in a large series of primary tumour samples reveals insights into cis-regulatory architecture, unrecognized dependencies, and cellular origins.


Nature | 2017

The whole-genome landscape of medulloblastoma subtypes

Paul A. Northcott; Ivo Buchhalter; A. Sorana Morrissy; Volker Hovestadt; Joachim Weischenfeldt; Tobias Ehrenberger; Susanne Gröbner; Maia Segura-Wang; Thomas Zichner; Vasilisa A. Rudneva; Hans-Jörg Warnatz; Nikos Sidiropoulos; Aaron H. Phillips; Steven E. Schumacher; Kortine Kleinheinz; Sebastian M. Waszak; Serap Erkek; David Jones; Barbara C. Worst; Marcel Kool; Marc Zapatka; Natalie Jäger; Lukas Chavez; Barbara Hutter; Matthias Bieg; Nagarajan Paramasivam; Michael Heinold; Zuguang Gu; Naveed Ishaque; Christina Jäger-Schmidt

Current therapies for medulloblastoma, a highly malignant childhood brain tumour, impose debilitating effects on the developing child, and highlight the need for molecularly targeted treatments with reduced toxicity. Previous studies have been unable to identify the full spectrum of driver genes and molecular processes that operate in medulloblastoma subgroups. Here we analyse the somatic landscape across 491 sequenced medulloblastoma samples and the molecular heterogeneity among 1,256 epigenetically analysed cases, and identify subgroup-specific driver alterations that include previously undiscovered actionable targets. Driver mutations were confidently assigned to most patients belonging to Group 3 and Group 4 medulloblastoma subgroups, greatly enhancing previous knowledge. New molecular subtypes were differentially enriched for specific driver events, including hotspot in-frame insertions that target KBTBD4 and ‘enhancer hijacking’ events that activate PRDM6. Thus, the application of integrative genomics to an extensive cohort of clinical samples derived from a single childhood cancer entity revealed a series of cancer genes and biologically relevant subtype diversity that represent attractive therapeutic targets for the treatment of patients with medulloblastoma.


Leukemia | 2015

The mutational pattern of primary lymphoma of the central nervous system determined by whole-exome sequencing

Inga Vater; M. Montesinos-Rongen; Matthias Schlesner; Andrea Haake; F. Purschke; R. Sprute; N. Mettenmeyer; I. Nazzal; I. Nagel; J. Gutwein; Julia Richter; Ivo Buchhalter; Robert B. Russell; Otmar D. Wiestler; Roland Eils; Martina Deckert; Reiner Siebert

To decipher the mutational pattern of primary CNS lymphoma (PCNSL), we performed whole-exome sequencing to a median coverage of 103 × followed by mutation verification in 9 PCNSL and validation using Sanger sequencing in 22 PCNSL. We identified a median of 202 (range: 139–251) potentially somatic single nucleotide variants (SNV) and 14 small indels (range: 7–22) with potentially protein-changing features per PCNSL. Mutations affected the B-cell receptor, toll-like receptor, and NF-κB and genes involved in chromatin structure and modifications, cell-cycle regulation, and immune recognition. A median of 22.2% (range: 20.0–24.7%) of somatic SNVs in 9 PCNSL overlaps with the RGYW motif targeted by somatic hypermutation (SHM); a median of 7.9% (range: 6.2–12.6%) affects its hotspot position suggesting a major impact of SHM on PCNSL pathogenesis. In addition to the well-known targets of aberrant SHM (aSHM) (PIM1), our data suggest new targets of aSHM (KLHL14, OSBPL10, and SUSD2). Among the four most frequently mutated genes was ODZ4 showing protein-changing mutations in 4/9 PCNSL. Together with mutations affecting CSMD2, CSMD3, and PTPRD, these findings may suggest that alterations in genes having a role in CNS development may facilitate diffuse large B-cell lymphoma manifestation in the CNS. This may point to intriguing mechanisms of CNS tropism in PCNSL.


Nature Medicine | 2016

Recurrent MET fusion genes represent a drug target in pediatric glioblastoma

Sebastian Bender; Jan Gronych; Hans-Jörg Warnatz; Barbara Hutter; Susanne Gröbner; Marina Ryzhova; Elke Pfaff; Volker Hovestadt; Florian Weinberg; Sebastian Halbach; Marcel Kool; Paul A. Northcott; Dominik Sturm; Lynn Bjerke; Thomas Zichner; Adrian M. Stütz; Kathrin Schramm; Bingding Huang; Ivo Buchhalter; Michael Heinold; Thomas Risch; Barbara C. Worst; Cornelis M. van Tilburg; Ursula Weber; Marc Zapatka; Benjamin Raeder; David Milford; Sabine Heiland; Christof von Kalle; Christopher Previti

Pediatric glioblastoma is one of the most common and most deadly brain tumors in childhood. Using an integrative genetic analysis of 53 pediatric glioblastomas and five in vitro model systems, we identified previously unidentified gene fusions involving the MET oncogene in ∼10% of cases. These MET fusions activated mitogen-activated protein kinase (MAPK) signaling and, in cooperation with lesions compromising cell cycle regulation, induced aggressive glial tumors in vivo. MET inhibitors suppressed MET tumor growth in xenograft models. Finally, we treated a pediatric patient bearing a MET-fusion-expressing glioblastoma with the targeted inhibitor crizotinib. This therapy led to substantial tumor shrinkage and associated relief of symptoms, but new treatment-resistant lesions appeared, indicating that combination therapies are likely necessary to achieve a durable clinical response.


Lancet Oncology | 2018

Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort

Sebastian M. Waszak; Paul A. Northcott; Ivo Buchhalter; Giles W. Robinson; Christian Sutter; Susanne N. Groebner; Kerstin Grund; Laurence Brugières; David T. W. Jones; Kristian W. Pajtler; A. Sorana Morrissy; Marcel Kool; Dominik Sturm; Lukas Chavez; Aurélie Ernst; Sebastian Brabetz; Michael Hain; Thomas Zichner; Maia Segura-Wang; Joachim Weischenfeldt; Tobias Rausch; Balca R. Mardin; Xin Zhou; Cristina Baciu; Christian Lawerenz; Jennifer A. Chan; Pascale Varlet; Lea Guerrini-Rousseau; Daniel W. Fults; Wieslawa A. Grajkowska

Summary Background Medulloblastoma is associated with rare hereditary cancer predisposition syndromes; however, consensus medulloblastoma predisposition genes have not been defined and screening guidelines for genetic counselling and testing for paediatric patients are not available. We aimed to assess and define these genes to provide evidence for future screening guidelines. Methods In this international, multicentre study, we analysed patients with medulloblastoma from retrospective cohorts (International Cancer Genome Consortium [ICGC] PedBrain, Medulloblastoma Advanced Genomics International Consortium [MAGIC], and the CEFALO series) and from prospective cohorts from four clinical studies (SJMB03, SJMB12, SJYC07, and I-HIT-MED). Whole-genome sequences and exome sequences from blood and tumour samples were analysed for rare damaging germline mutations in cancer predisposition genes. DNA methylation profiling was done to determine consensus molecular subgroups: WNT (MBWNT), SHH (MBSHH), group 3 (MBGroup3), and group 4 (MBGroup4). Medulloblastoma predisposition genes were predicted on the basis of rare variant burden tests against controls without a cancer diagnosis from the Exome Aggregation Consortium (ExAC). Previously defined somatic mutational signatures were used to further classify medulloblastoma genomes into two groups, a clock-like group (signatures 1 and 5) and a homologous recombination repair deficiency-like group (signatures 3 and 8), and chromothripsis was investigated using previously established criteria. Progression-free survival and overall survival were modelled for patients with a genetic predisposition to medulloblastoma. Findings We included a total of 1022 patients with medulloblastoma from the retrospective cohorts (n=673) and the four prospective studies (n=349), from whom blood samples (n=1022) and tumour samples (n=800) were analysed for germline mutations in 110 cancer predisposition genes. In our rare variant burden analysis, we compared these against 53 105 sequenced controls from ExAC and identified APC, BRCA2, PALB2, PTCH1, SUFU, and TP53 as consensus medulloblastoma predisposition genes according to our rare variant burden analysis and estimated that germline mutations accounted for 6% of medulloblastoma diagnoses in the retrospective cohort. The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MBSHH subgroup (20% in the retrospective cohort). These estimates were replicated in the prospective clinical cohort (germline mutations accounted for 5% of medulloblastoma diagnoses, with the highest prevalence [14%] in the MBSHH subgroup). Patients with germline APC mutations developed MBWNT and accounted for most (five [71%] of seven) cases of MBWNT that had no somatic CTNNB1 exon 3 mutations. Patients with germline mutations in SUFU and PTCH1 mostly developed infant MBSHH. Germline TP53 mutations presented only in childhood patients in the MBSHH subgroup and explained more than half (eight [57%] of 14) of all chromothripsis events in this subgroup. Germline mutations in PALB2 and BRCA2 were observed across the MBSHH, MBGroup3, and MBGroup4 molecular subgroups and were associated with mutational signatures typical of homologous recombination repair deficiency. In patients with a genetic predisposition to medulloblastoma, 5-year progression-free survival was 52% (95% CI 40–69) and 5-year overall survival was 65% (95% CI 52–81); these survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes. Interpretation Genetic counselling and testing should be used as a standard-of-care procedure in patients with MBWNT and MBSHH because these patients have the highest prevalence of damaging germline mutations in known cancer predisposition genes. We propose criteria for routine genetic screening for patients with medulloblastoma based on clinical and molecular tumour characteristics. Funding German Cancer Aid; German Federal Ministry of Education and Research; German Childhood Cancer Foundation (Deutsche Kinderkrebsstiftung); European Research Council; National Institutes of Health; Canadian Institutes for Health Research; German Cancer Research Center; St Jude Comprehensive Cancer Center; American Lebanese Syrian Associated Charities; Swiss National Science Foundation; European Molecular Biology Organization; Cancer Research UK; Hertie Foundation; Alexander and Margaret Stewart Trust; V Foundation for Cancer Research; Sontag Foundation; Musicians Against Childhood Cancer; BC Cancer Foundation; Swedish Council for Health, Working Life and Welfare; Swedish Research Council; Swedish Cancer Society; the Swedish Radiation Protection Authority; Danish Strategic Research Council; Swiss Federal Office of Public Health; Swiss Research Foundation on Mobile Communication; Masaryk University; Ministry of Health of the Czech Republic; Research Council of Norway; Genome Canada; Genome BC; Terry Fox Research Institute; Ontario Institute for Cancer Research; Pediatric Oncology Group of Ontario; The Family of Kathleen Lorette and the Clark H Smith Brain Tumour Centre; Montreal Childrens Hospital Foundation; The Hospital for Sick Children: Sonia and Arthur Labatt Brain Tumour Research Centre, Chief of Research Fund, Cancer Genetics Program, Garron Family Cancer Centre, MDTs Garron Family Endowment; BC Childhood Cancer Parents Association; Cure Search Foundation; Pediatric Brain Tumor Foundation; Brainchild; and the Government of Ontario.


bioRxiv | 2014

A comprehensive multicenter comparison of whole genome sequencing pipelines using a uniform tumor-normal sample pair

Ivo Buchhalter; Barbara Hutter; Tyler Alioto; Timothy Beck; Paul C. Boutros; Benedikt Brors; Adam Butler; Sasithorn Chotewutmontri; Robert E. Denroche; Sophia Derdak; Nicolle Diessl; Lars Feuerbach; Akihiro Fujimoto; Susanne Gröbner; Marta Gut; Nicholas J. Harding; Michael Heinold; Lawrence E. Heisler; Jonathan Hinton; Natalie Jäger; David Jones; Rolf Kabbe; Andrey Korshunov; John D. McPherson; Andrew Menzies; Hidewaki Nakagawa; Christopher Previti; Keiran Raine; Paolo Ribeca; Sabine Schmidt

As next-generation sequencing becomes a clinical tool, a full understanding of the variables affecting sequencing analysis output is required. Through the International Cancer Genome Consortium (ICGC), we compared sequencing pipelines at five independent centers (CNAG, DKFZ, OICR, RIKEN and WTSI) using a single tumor-blood DNA pair. Analyses by each center and with one standardized algorithm revealed significant discrepancies. Although most pipelines performed well for coding mutations, library preparation methods and sequencing coverage metrics clearly influenced downstream results. PCR-free methods showed reduced GC-bias and more even coverage. Increasing sequencing depth to ∼100x (two- to three-fold higher than current standards) showed a benefit, as long as the tumor:control coverage ratio remained balanced. To become part of routine clinical care, high-throughput sequencing must be globally compatible and comparable. This benchmarking exercise has highlighted several fundamental parameters to consider in this regard, which will allow for better optimization and planning of both basic and translational studies.


Acta Neuropathologica | 2018

Deep sequencing of WNT-activated medulloblastomas reveals secondary SHH pathway activation

J. Bryan Iorgulescu; Jessica Van Ziffle; Meredith Stevers; James P. Grenert; Boris C. Bastian; Lukas Chavez; Damian Stichel; Ivo Buchhalter; David Samuel; Theodore Nicolaides; Anuradha Banerjee; Sabine Mueller; Nalin Gupta; Tarik Tihan; Andrew W. Bollen; Paul A. Northcott; Marcel Kool; Stefan M. Pfister; Andrey Korshunov; Arie Perry; David A. Solomon

Author(s): Iorgulescu, J Bryan; Van Ziffle, Jessica; Stevers, Meredith; Grenert, James P; Bastian, Boris C; Chavez, Lukas; Stichel, Damian; Buchhalter, Ivo; Samuel, David; Nicolaides, Theodore; Banerjee, Anuradha; Mueller, Sabine; Gupta, Nalin; Tihan, Tarik; Bollen, Andrew W; Northcott, Paul A; Kool, Marcel; Pfister, Stefan; Korshunov, Andrey; Perry, Arie; Solomon, David A

Collaboration


Dive into the Ivo Buchhalter's collaboration.

Top Co-Authors

Avatar

David T. W. Jones

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Marcel Kool

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Andrey Korshunov

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan M. Pfister

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Marc Zapatka

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Barbara Hutter

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Dominik Sturm

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Paul A. Northcott

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Serap Erkek

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge