Network


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

Hotspot


Dive into the research topics where Daniel Hübschmann is active.

Publication


Featured researches published by Daniel Hübschmann.


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.


Cold Spring Harb Mol Case Stud | 2016

Integration of genomics and histology revises diagnosis and enables effective therapy of refractory cancer of unknown primary with PDL1 amplification

Stefan Gröschel; Martin Bommer; Barbara Hutter; Jan Budczies; David Bonekamp; Christoph Heining; Peter Horak; Martina Fröhlich; Sebastian Uhrig; Daniel Hübschmann; Christina Geörg; Daniela Richter; Nicole Pfarr; Katrin Pfütze; Stephan Wolf; Peter Schirmacher; Dirk Jäger; Christof von Kalle; Benedikt Brors; Hanno Glimm; Wilko Weichert; Albrecht Stenzinger; Stefan Fröhling

Identification of the tissue of origin in cancer of unknown primary (CUP) poses a diagnostic challenge and is critical for directing site-specific therapy. Currently, clinical decision-making in patients with CUP primarily relies on histopathology and clinical features. Comprehensive molecular profiling has the potential to contribute to diagnostic categorization and, most importantly, guide CUP therapy through identification of actionable lesions. We here report the case of an advanced-stage malignancy initially mimicking poorly differentiated soft-tissue sarcoma that did not respond to multiagent chemotherapy. Molecular profiling within a clinical whole-exome and transcriptome sequencing program revealed a heterozygous, highly amplified KRAS G12S mutation, compound-heterozygous TP53 mutation/deletion, high mutational load, and focal high-level amplification of Chromosomes 9p (including PDL1 [CD274] and JAK2) and 10p (including GATA3). Integrated analysis of molecular data and histopathology provided a rationale for immune checkpoint inhibitor (ICI) therapy with pembrolizumab, which resulted in rapid clinical improvement and a lasting partial remission. Histopathological analyses ruled out sarcoma and established the diagnosis of a poorly differentiated adenocarcinoma. Although neither histopathology nor molecular data were able to pinpoint the tissue of origin, our analyses established several differential diagnoses including triple-negative breast cancer (TNBC). We analyzed 157 TNBC samples from The Cancer Genome Atlas, revealing PDL1 copy number gains coinciding with excessive PDL1 mRNA expression in 24% of cases. Collectively, these results illustrate the impact of multidimensional tumor profiling in cases with nondescript histology and immunophenotype, show the predictive potential of PDL1 amplification for immune checkpoint inhibitors (ICIs), and suggest a targeted therapeutic strategy in Chromosome 9p24.1/PDL1-amplified cancers.


International Journal of Cancer | 2017

Precision Oncology Based on Omics Data: The NCT Heidelberg Experience

Peter Horak; Barbara Klink; Christoph Heining; Stefan Gröschel; Barbara Hutter; Martina Fröhlich; Sebastian Uhrig; Daniel Hübschmann; Matthias Schlesner; Roland Eils; Daniela Richter; Katrin Pfütze; Christina Geörg; Bettina Meißburger; Stephan Wolf; Angela Schulz; Roland Penzel; Esther Herpel; Martina Kirchner; Amelie Lier; Volker Endris; Stephan Singer; Peter Schirmacher; Wilko Weichert; Albrecht Stenzinger; Richard F. Schlenk; Evelin Schröck; Benedikt Brors; Christof von Kalle; Hanno Glimm

Precision oncology implies the ability to predict which patients will likely respond to specific cancer therapies based on increasingly accurate, high‐resolution molecular diagnostics as well as the functional and mechanistic understanding of individual tumors. While molecular stratification of patients can be achieved through different means, a promising approach is next‐generation sequencing of tumor DNA and RNA, which can reveal genomic alterations that have immediate clinical implications. Furthermore, certain genetic alterations are shared across multiple histologic entities, raising the fundamental question of whether tumors should be treated by molecular profile and not tissue of origin. We here describe MASTER (Molecularly Aided Stratification for Tumor Eradication Research), a clinically applicable platform for prospective, biology‐driven stratification of younger adults with advanced‐stage cancer across all histologies and patients with rare tumors. We illustrate how a standardized workflow for selection and consenting of patients, sample processing, whole‐exome/genome and RNA sequencing, bioinformatic analysis, rigorous validation of potentially actionable findings, and data evaluation by a dedicated molecular tumor board enables categorization of patients into different intervention baskets and formulation of evidence‐based recommendations for clinical management. Critical next steps will be to increase the number of patients that can be offered comprehensive molecular analysis through collaborations and partnering, to explore ways in which additional technologies can aid in patient stratification and individualization of treatment, to stimulate clinically guided exploratory research projects, and to gradually move away from assessing the therapeutic activity of targeted interventions on a case‐by‐case basis toward controlled clinical trials of genomics‐guided treatments.


Nature Communications | 2016

Spatial niche formation but not malignant progression is a driving force for intratumoural heterogeneity

Rouven Hoefflin; Bernd Lahrmann; Gregor Warsow; Daniel Hübschmann; Cathleen Spath; Britta Walter; Xin Chen; Luisa Hofer; Stephan Macher-Goeppinger; Yanis Tolstov; Nina Korzeniewski; Anette Duensing; Carsten Grüllich; Dirk Jäger; Sven Perner; Gita Schönberg; Joanne Nyarangi-Dix; Sanjay Isaac; Gencay Hatiboglu; Dogu Teber; Boris Hadaschik; Sascha Pahernik; Wilfried Roth; Roland Eils; Matthias Schlesner; Holger Sültmann; Markus Hohenfellner; Niels Grabe; Stefan Duensing

Intratumoural heterogeneity (ITH) is a major cause of cancer-associated lethality. Extensive genomic ITH has previously been reported in clear cell renal cell carcinoma (ccRCC). Here we address the question whether ITH increases with malignant progression and can hence be exploited as a prognostic marker. Unexpectedly, precision quantitative image analysis reveals that the degree of functional ITH is virtually identical between primary ccRCCs of the lowest stage and advanced, metastatic tumours. Functional ITH was found to show a stage-independent topological pattern with peak proliferative and signalling activities almost exclusively in the tumour periphery. Exome sequencing of matching peripheral and central primary tumour specimens reveals various region-specific mutations. However, these mutations cannot directly explain the zonal pattern suggesting a role of microenvironmental factors in shaping functional ITH. In conclusion, our results indicate that ITH is an early and general characteristic of malignant growth rather than a consequence of malignant progression.


Nature Communications | 2018

Integrative genomic and transcriptomic analysis of leiomyosarcoma

Priya Chudasama; Sadaf S. Mughal; Mathijs A. Sanders; Daniel Hübschmann; Inn Chung; Katharina I. Deeg; Siao-Han Wong; Sophie Rabe; Mario Hlevnjak; Marc Zapatka; Aurélie Ernst; Kortine Kleinheinz; Matthias Schlesner; Lina Sieverling; Barbara Klink; Evelin Schröck; Remco M. Hoogenboezem; Bernd Kasper; Christoph E. Heilig; Gerlinde Egerer; Stephan E. Wolf; Christof von Kalle; Roland Eils; Albrecht Stenzinger; Wilko Weichert; Hanno Glimm; Stefan Gröschel; Hans-Georg Kopp; Georg W. Omlor; Burkhard Lehner

Leiomyosarcoma (LMS) is an aggressive mesenchymal malignancy with few therapeutic options. The mechanisms underlying LMS development, including clinically actionable genetic vulnerabilities, are largely unknown. Here we show, using whole-exome and transcriptome sequencing, that LMS tumors are characterized by substantial mutational heterogeneity, near-universal inactivation of TP53 and RB1, widespread DNA copy number alterations including chromothripsis, and frequent whole-genome duplication. Furthermore, we detect alternative telomere lengthening in 78% of cases and identify recurrent alterations in telomere maintenance genes such as ATRX, RBL2, and SP100, providing insight into the genetic basis of this mechanism. Finally, most tumors display hallmarks of “BRCAness”, including alterations in homologous recombination DNA repair genes, multiple structural rearrangements, and enrichment of specific mutational signatures, and cultured LMS cells are sensitive towards olaparib and cisplatin. This comprehensive study of LMS genomics has uncovered key biological features that may inform future experimental research and enable the design of novel therapies.The molecular genetic landscape of leiomyosarcoma (LMS) is largely unknown. Here, the authors identify frequent DNA copy number alterations, whole-genome duplication, TP53 and RB1 inactivation, alternative telomere lengthening, and genomic imprints of defective DNA repair via homologous recombination as a potential therapeutic target in LMS patients.


Scientific Reports | 2018

Genomic features of renal cell carcinoma with venous tumor thrombus

Gregor Warsow; Daniel Hübschmann; Kortine Kleinheinz; Cathleen Nientiedt; Martina Heller; Laura Van Coile; Yanis Tolstov; Lukas Trennheuser; Kathrin Wieczorek; Carine Pecqueux; Claudia Gasch; Timur H. Kuru; Joanne Nyarangi-Dix; Gencay Hatiboglu; Dogu Teber; Sven Perner; Albrecht Stenzinger; Wilfried Roth; Boris Hadaschik; Sascha Pahernik; Dirk Jäger; Carsten Grüllich; Anette Duensing; Roland Eils; Matthias Schlesner; Holger Sültmann; Markus Hohenfellner; Stefan Duensing

A venous tumor thrombus (VTT) is a potentially lethal complication of renal cell carcinoma (RCC) but virtually nothing is known about the underlying natural history. Based on our observation that venous thrombi contain significant numbers of viable tumor cells, we applied multiregion whole exome sequencing to a total of 37 primary tumor and VTT samples including normal tissue specimens from five consecutive patients. Our findings demonstrate mutational heterogeneity between primary tumor and VTT with 106 of 483 genes (22%) harboring functional SNVs and/or indels altered in either primary tumor or thrombus. Reconstruction of the clonal phylogeny showed clustering of tumor samples and VTT samples, respectively, in the majority of tumors. However, no new subclones were detected suggesting that pre-existing subclones of the primary tumor drive VTT formation. Importantly, we found several lines of evidence for “BRCAness” in a subset of tumors. These included mutations in genes that confer “BRCAness”, a mutational signature and an increase of small indels. Re-analysis of SNV calls from the TCGA KIRC-US cohort confirmed a high frequency of the “BRCAness” mutational signature AC3 in clear cell RCC. Our findings warrant further pre-clinical experiments and may lead to novel personalized therapies for RCC patients.


Archive | 2011

Quantitative Approaches to Nuclear Architecture Analysis and Modelling

Daniel Hübschmann; Nikolaus Kepper; Christoph Cremer; Gregor Kreth

The spatial organisation of the genome in the cell nucleus has emerged as a key element to understand gene function. A wealth of molecular and microscopic information has been accumulated, resulting in a variety of – sometimes contradictory – models of nuclear architecture. So far, however, a large part of this structural information and in consequence also the models derived from them are ‘qualitative’. In this overview, a brief introduction will be given into quantitative experimental and modelling approaches to large scale nuclear genome architecture in human cells. As a biomedical application example, the use of a quantitative computer model of the 3D architecture allowed to explore different implications of nuclear structure on chromosomal aberrations. In addition, we shall present two novel examples for quantitative computer modelling: (1) The impact of SC 35 splicing domains on nuclear genome structure; (2) The dynamics of large scale nuclear genome structure in a Brownian motion model. Finally, we shall discuss some perspectives to extend quantitative nuclear structure analysis to the nanoscale.


Clinical Immunology | 2017

Newborn screening for severe combined immunodeficiency using a novel and simplified method to measure T-cell excision circles (TREC)

Laura Tagliaferri; Joachim B. Kunz; Margit Happich; Susanna Esposito; Thomas Bruckner; Daniel Hübschmann; Jürgen G. Okun; Georg F. Hoffmann; Ansgar Schulz; Judit Kappe; Carsten Speckmann; Martina U. Muckenthaler; Andreas E. Kulozik


Cancer Research | 2018

Abstract 4336: Integrative genomic and transcriptomic analysis of leiomyosarcoma

Priya Chudasama; Sadaf S. Mughal; Mathijs A. Sanders; Daniel Hübschmann; Inn Chung; Aurélie Ernst; Bernd Kasper; Hans-Georg Kopp; Sebastian Bauer; Karsten Rippe; Benedikt Brors; Marcus Renner; Peter Hohenberger; Claudia Scholl; Stefan Fröhling


Blood | 2018

IG-MYC-positive neoplasms with precursor B-cell phenotype are molecularly distinct from Burkitt lymphomas

Rabea Wagener; Cristina López; Kortine Kleinheinz; Julia Bausinger; Sietse M. Aukema; Inga Nagel; Umut H. Toprak; Julian Seufert; Janine Altmüller; Holger Thiele; Christof Schneider; Julia Kolarova; Jeongbin Park; Daniel Hübschmann; Eva Maria Murga Penas; Hans G. Drexler; Andishe Attarbaschi; Randi Hovland; Eigil Kjeldsen; Michael Kneba; Udo Kontny; Laurence de Leval; Peter Nürnberg; Ilske Oschlies; David Oscier; Brigitte Schlegelberger; Stephan Stilgenbauer; Wilhelm Wössmann; Matthias Schlesner; Birgit Burkhardt

Collaboration


Dive into the Daniel Hübschmann's collaboration.

Top Co-Authors

Avatar

Matthias Schlesner

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Albrecht Stenzinger

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Kortine Kleinheinz

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

Benedikt Brors

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk Jäger

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Hanno Glimm

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan Gröschel

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge