Network


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

Hotspot


Dive into the research topics where Sabine Schmidt is active.

Publication


Featured researches published by Sabine Schmidt.


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.


Cancer Cell | 2014

Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition

Marcel Kool; David T. W. Jones; Natalie Jäger; Paul A. Northcott; Trevor J. Pugh; Volker Hovestadt; Rosario M. Piro; L. Adriana Esparza; Shirley L. Markant; Marc Remke; Till Milde; Franck Bourdeaut; Marina Ryzhova; Dominik Sturm; Elke Pfaff; Sebastian Stark; Sonja Hutter; Huriye Şeker-Cin; Pascal Johann; Sebastian Bender; Christin Schmidt; Tobias Rausch; David Shih; Jüri Reimand; Laura Sieber; Andrea Wittmann; Linda Linke; Hendrik Witt; Ursula Weber; Marc Zapatka

Smoothened (SMO) inhibitors recently entered clinical trials for sonic-hedgehog-driven medulloblastoma (SHH-MB). Clinical response is highly variable. To understand the mechanism(s) of primary resistance and identify pathways cooperating with aberrant SHH signaling, we sequenced and profiled a large cohort of SHH-MBs (nxa0= 133). SHH pathway mutations involved PTCH1 (across all age groups), SUFU (infants, including germline), and SMO (adults). Children >3 years old harbored an excess of downstream MYCN and GLI2 amplifications and frequent TP53 mutations, often in the germline, all of which were rare in infants and adults. Functional assays in different SHH-MB xenograft models demonstrated that SHH-MBs harboring a PTCH1 mutation were responsive to SMO inhibition, whereas tumors harboring an SUFU mutation or MYCN amplification were primarily resistant.


Nature | 2014

Decoding the regulatory landscape of medulloblastoma using DNA methylation sequencing

Volker Hovestadt; David T. W. Jones; Simone Picelli; Wei Wang; Marcel Kool; Paul A. Northcott; Marc Sultan; Katharina Stachurski; Marina Ryzhova; Hans Jörg Warnatz; Meryem Ralser; Sonja Brun; Jens Bunt; Natalie Jäger; Kortine Kleinheinz; Serap Erkek; Ursula Weber; Cynthia C. Bartholomae; Christof von Kalle; Chris Lawerenz; Jürgen Eils; Jan Koster; Rogier Versteeg; Till Milde; Olaf Witt; Sabine Schmidt; Stephan Wolf; Torsten Pietsch; Stefan Rutkowski; Wolfram Scheurlen

Epigenetic alterations, that is, disruption of DNA methylation and chromatin architecture, are now acknowledged as a universal feature of tumorigenesis. Medulloblastoma, a clinically challenging, malignant childhood brain tumour, is no exception. Despite much progress from recent genomics studies, with recurrent changes identified in each of the four distinct tumour subgroups (WNT-pathway-activated, SHH-pathway-activated, and the less-well-characterized Group 3 and Group 4), many cases still lack an obvious genetic driver. Here we present whole-genome bisulphite-sequencing data from thirty-four human and five murine tumours plus eight human and three murine normal controls, augmented with matched whole-genome, RNA and chromatin immunoprecipitation sequencing data. This comprehensive data set allowed us to decipher several features underlying the interplay between the genome, epigenome and transcriptome, and its effects on medulloblastoma pathophysiology. Most notable were highly prevalent regions of hypomethylation correlating with increased gene expression, extending tens of kilobases downstream of transcription start sites. Focal regions of low methylation linked to transcription-factor-binding sites shed light on differential transcriptional networks between subgroups, whereas increased methylation due to re-normalization of repressed chromatin in DNA methylation valleys was positively correlated with gene expression. Large, partially methylated domains affecting up to one-third of the genome showed increased mutation rates and gene silencing in a subgroup-specific fashion. Epigenetic alterations also affected novel medulloblastoma candidate genes (for example, LIN28B), resulting in alternative promoter usage and/or differential messenger RNA/microRNA expression. Analysis of mouse medulloblastoma and precursor-cell methylation demonstrated a somatic origin for many alterations. Our data provide insights into the epigenetic regulation of transcription and genome organization in medulloblastoma pathogenesis, which are probably also of importance in a wider developmental and disease context.


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 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 Medicine | 2013

A largely random AAV integration profile after LPLD gene therapy

Christine Kaeppel; Stuart G Beattie; Raffaele Fronza; Richard van Logtenstein; Florence Salmon; Sabine Schmidt; Stephan Wolf; Ali Nowrouzi; Hanno Glimm; Christof von Kalle; Harald Petry; Daniel Gaudet; Manfred Schmidt

The clinical application of adeno-associated virus vectors (AAVs) is limited because of concerns about AAV integration–mediated tumorigenicity. We performed integration-site analysis after AAV1-LPLS447X intramuscular injection in five lipoprotein lipase–deficient subjects, revealing random nuclear integration and hotspots in mitochondria. We conclude that AAV integration is potentially safe and that vector breakage and integration may occur from each position of the vector genome. Future viral integration-site analyses should include the mitochondrial genome.


European Journal of Cancer | 2016

Next-generation personalised medicine for high-risk paediatric cancer patients – The INFORM pilot study

Barbara C. Worst; Cornelis M. van Tilburg; Gnana Prakash Balasubramanian; Petra Fiesel; Ruth Witt; Angelika Freitag; Miream Boudalil; Christopher Previti; Stephan Wolf; Sabine Schmidt; Sasithorn Chotewutmontri; Melanie Bewerunge-Hudler; Matthias Schick; Matthias Schlesner; Barbara Hutter; Lenka A. Taylor; Tobias Borst; Christian Sutter; Claus R. Bartram; Till Milde; Elke Pfaff; Andreas E. Kulozik; Arend von Stackelberg; Roland Meisel; Arndt Borkhardt; Dirk Reinhardt; Jan-Henning Klusmann; Gudrun Fleischhack; Stephan Tippelt; Uta Dirksen

The Individualized Therapy for Relapsed Malignancies in Childhood (INFORM) precision medicine study is a nationwide German program for children with high-risk relapsed/refractory malignancies, which aims to identify therapeutic targets on an individualised basis. In a pilot phase, reported here, we developed the logistical and analytical pipelines necessary for rapid and comprehensive molecular profiling in a clinical setting. Fifty-seven patients from 20 centers were prospectively recruited. Malignancies investigated included sarcomas (nxa0=xa025), brain tumours (nxa0=xa023), and others (nxa0=xa09). Whole-exome, low-coverage whole-genome, and RNA sequencing were complemented with methylation and expression microarray analyses. Alterations were assessed for potential targetability according to a customised prioritisation algorithm and subsequently discussed in an interdisciplinary molecular tumour board. Next-generation sequencing data were generated for 52 patients, with the full analysis possible in 46 of 52. Turnaround time from sample receipt until first report averaged 28xa0d. Twenty-six patients (50%) harbored a potentially druggable alteration with a prioritisation score of intermediate or higher (level 4 of 7). Common targets included receptor tyrosine kinases, phosphoinositide 3-kinase-mammalian target of rapamycin pathway, mitogen-activated protein kinase pathway, and cell cycle control. Ten patients received a targeted therapy based on these findings, with responses observed in some previously treatment-refractory tumours. Comparative primary relapse analysis revealed substantial tumour evolution as well as one case of unsuspected secondary malignancy, highlighting the importance of re-biopsy at relapse. This study demonstrates the feasibility of comprehensive, real-time molecular profiling for high-risk paediatric cancer patients. This extended proof-of-concept, with examples of treatment consequences, expands upon previous personalised oncology endeavors, and presents a model with considerable interest and practical relevance in the burgeoning era of personalised medicine.


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.


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.


Nature Medicine | 2014

Reply to: NGS library preparation may generate artifactual integration sites of AAV vectors.

Christine Kaeppel; Stuart G Beattie; Raffaele Fronza; Richard van Logtenstein; Florence Salmon; Sabine Schmidt; Stephan Wolf; Ali Nowrouzi; Hanno Glimm; Christof von Kalle; Harald Petry; Daniel Gaudet; Manfred Schmidt

Reply to: NGS library preparation may generate artifactual integration sites of AAV vectors

Collaboration


Dive into the Sabine Schmidt's collaboration.

Top Co-Authors

Avatar

Natalie Jäger

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Barbara Hutter

German Cancer Research Center

View shared research outputs
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

Marina Ryzhova

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

Stephan Wolf

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Volker Hovestadt

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christopher Previti

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge