Network


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

Hotspot


Dive into the research topics where Sebastian Brabetz is active.

Publication


Featured researches published by Sebastian Brabetz.


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.


Cancer Cell | 2016

HDAC and PI3K Antagonists Cooperate to Inhibit Growth of MYC-Driven Medulloblastoma

Yanxin Pei; Kun Wei Liu; Jun Wang; Alexandra Garancher; Ran Tao; Lourdes Adriana Esparza; Donna L. Maier; Yoko T. Udaka; Najiba Murad; Sorana Morrissy; Huriye Seker-Cin; Sebastian Brabetz; Lin Qi; Mari Kogiso; Simone Schubert; James M. Olson; Yoon-Jae Cho; Xiao-Nan Li; John R. Crawford; Michael L. Levy; Marcel Kool; Stefan M. Pfister; Michael D. Taylor; Robert J. Wechsler-Reya

Medulloblastoma (MB) is a highly malignant pediatric brain tumor. Despite aggressive therapy, many patients succumb to the disease, and survivors experience severe side effects from treatment. MYC-driven MB has a particularly poor prognosis and would greatly benefit from more effective therapies. We used an animal model of MYC-driven MB to screen for drugs that decrease viability of tumor cells. Among the most effective compounds were histone deacetylase inhibitors (HDACIs). HDACIs potently inhibit survival of MYC-driven MB cells in vitro, in part by inducing expression of the FOXO1 tumor suppressor gene. HDACIs also synergize with phosphatidylinositol 3-kinase inhibitors to inhibit tumor growth in vivo. These studies identify an effective combination therapy for the most aggressive form of MB.


International Journal of Cancer | 2016

Telomere dysfunction and chromothripsis

Aurélie Ernst; David T. W. Jones; Kendra Korinna Maass; Agata Rode; Katharina I. Deeg; Billy Michael Chelliah Jebaraj; Andrey Korshunov; Volker Hovestadt; Michael A. Tainsky; Kristian W. Pajtler; Sebastian Bender; Sebastian Brabetz; Susanne Gröbner; Marcel Kool; Frauke Devens; Jennifer Edelmann; Cindy Zhang; Pedro Castelo-Branco; Uri Tabori; David Malkin; Karsten Rippe; Stephan Stilgenbauer; Stefan M. Pfister; Marc Zapatka; Peter Lichter

Chromothripsis is a recently discovered form of genomic instability, characterized by tens to hundreds of clustered DNA rearrangements resulting from a single dramatic event. Telomere dysfunction has been suggested to play a role in the initiation of this phenomenon, which occurs in a large number of tumor entities. Here, we show that telomere attrition can indeed lead to catastrophic genomic events, and that telomere patterns differ between cells analyzed before and after such genomic catastrophes. Telomere length and telomere stabilization mechanisms diverge between samples with and without chromothripsis in a given tumor subtype. Longitudinal analyses of the evolution of chromothriptic patterns identify either stable patterns between matched primary and relapsed tumors, or loss of the chromothriptic clone in the relapsed specimen. The absence of additional chromothriptic events occurring between the initial tumor and the relapsed tumor sample points to telomere stabilization after the initial chromothriptic event which prevents further shattering of the genome.


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.


Oncotarget | 2017

Establishment and application of a novel patient-derived KIAA1549:BRAF-driven pediatric pilocytic astrocytoma model for preclinical drug testing

Florian Selt; Juliane Hohloch; Thomas Hielscher; Felix Sahm; David Capper; Andrey Korshunov; Diren Usta; Sebastian Brabetz; Johannes Ridinger; J Ecker; Ina Oehme; Jan Gronych; Viktoria Marquardt; David Pauck; Heidi Bächli; Charles D. Stiles; Andreas von Deimling; Marc Remke; Martin U. Schuhmann; Stefan M. Pfister; Tilman Brummer; David T. W. Jones; Olaf Witt; Till Milde

Pilocytic astrocytoma (PA) is the most frequent pediatric brain tumor. Activation of the MAPK pathway is well established as the oncogenic driver of the disease. It is most frequently caused by KIAA1549:BRAF fusions, and leads to oncogene induced senescence (OIS). OIS is thought to be a major reason for growth arrest of PA cells in vitro and in vivo, preventing establishment of PA cultures. Hence, valid preclinical models are currently very limited, but preclinical testing of new compounds is urgently needed. We transduced the PA short-term culture DKFZ-BT66 derived from the PA of a 2-year old patient with a doxycycline-inducible system coding for Simian Vacuolating Virus 40 Large T Antigen (SV40-TAg). SV40-TAg inhibits TP53/CDKN1A and CDKN2A/RB1, two pathways critical for OIS induction and maintenance. DNA methylation array and KIAA1549:BRAF fusion analysis confirmed pilocytic astrocytoma identity of DKFZ-BT66 cells after establishment. Readouts were analyzed in proliferating as well as senescent states, including cell counts, viability, cell cycle analysis, expression of SV40-Tag, CDKN2A (p16), CDKN1A (p21), and TP53 (p53) protein, and gene-expression profiling. Selected MAPK inhibitors (MAPKi) including clinically available MEK inhibitors (MEKi) were tested in vitro. Expression of SV40-TAg enabled the cells to bypass OIS and to resume proliferation with a mean doubling time of 45h allowing for propagation and long-term culture. Withdrawal of doxycycline led to an immediate decrease of SV40-TAg expression, appearance of senescent morphology, upregulation of CDKI proteins and a subsequent G1 growth arrest in line with the re-induction of senescence. DKFZ-BT66 cells still underwent replicative senescence that was overcome by TERT expression. Testing of a set of MAPKi revealed differential responses in DKFZ-BT66. MEKi efficiently inhibited MAPK signaling at clinically achievable concentrations, while BRAF V600E- and RAF Type II inhibitors showed paradoxical activation. Taken together, we have established the first patient-derived long term expandable PA cell line expressing the KIAA1549:BRAF-fusion suitable for preclinical drug testing.


Cancer Research | 2017

PDX-MI: Minimal Information for Patient-Derived Tumor Xenograft Models

Terrence F. Meehan; Nathalie Conte; Theodore C. Goldstein; Giorgio Inghirami; Mark A. Murakami; Sebastian Brabetz; Zhiping Gu; Jeffrey Wiser; Patrick Dunn; Dale A. Begley; Debra M. Krupke; Andrea Bertotti; Alejandra Bruna; Matthew H. Brush; Annette T. Byrne; Carlos Caldas; Amanda L. Christie; Dominic A. Clark; Heidi Dowst; Jonathan R. Dry; James H. Doroshow; Olivier Duchamp; Yvonne A. Evrard; Stephane Ferretti; Kristopher K. Frese; Neal C. Goodwin; Danielle Greenawalt; Melissa Haendel; Els Hermans; Peter J. Houghton

Patient-derived tumor xenograft (PDX) mouse models have emerged as an important oncology research platform to study tumor evolution, mechanisms of drug response and resistance, and tailoring chemotherapeutic approaches for individual patients. The lack of robust standards for reporting on PDX models has hampered the ability of researchers to find relevant PDX models and associated data. Here we present the PDX models minimal information standard (PDX-MI) for reporting on the generation, quality assurance, and use of PDX models. PDX-MI defines the minimal information for describing the clinical attributes of a patients tumor, the processes of implantation and passaging of tumors in a host mouse strain, quality assurance methods, and the use of PDX models in cancer research. Adherence to PDX-MI standards will facilitate accurate search results for oncology models and their associated data across distributed repository databases and promote reproducibility in research studies using these models. Cancer Res; 77(21); e62-66. ©2017 AACR.


Neuro-oncology | 2017

Preclinical drug screen reveals topotecan, actinomycin D, and volasertib as potential new therapeutic candidates for ETMR brain tumor patients

Christin Schmidt; Nil A. Schubert; Sebastian Brabetz; Norman Mack; Benjamin Schwalm; Jennifer A. Chan; Florian Selt; Christel Herold-Mende; Olaf Witt; Till Milde; Stefan M. Pfister; Andrey Korshunov; Marcel Kool

Background Embryonal tumor with multilayered rosettes (ETMR) is a rare and aggressive embryonal brain tumor that solely occurs in infants and young children and has only recently been recognized as a separate brain tumor entity in the World Health Organization classification for CNS tumors. Patients have a very dismal prognosis with a median survival of 12 months upon diagnosis despite aggressive treatment. The aim of this study was to develop novel treatment regimens in a preclinical drug screen in order to inform potentially more active clinical trial protocols. Methods We have carried out an in vitro and in vivo drug screen using the ETMR cell line BT183 and its xenograft model. Furthermore, we have generated the first patient-derived xenograft (PDX) model for ETMR and evaluated our top drug candidates in an in vitro drug screen using this model. Results BT183 cells are very sensitive to the topoisomerase inhibitors topotecan and doxorubicin, to the epigenetic agents decitabine and panobinostat, to actinomycin D, and to targeted drugs such as the polo-like kinase 1 (PLK1) inhibitor volasertib, the aurora kinase A inhibitor alisertib, and the mammalian target of rapamycin (mTOR) inhibitor MLN0128. In xenograft mice, monotherapy with topotecan, volasertib, and actinomycin D led to a temporary response in tumor growth and a significant increase in survival. Finally, using multi-agent treatment regimens of topotecan or doxorubicin combined with methotrexate and vincristine, the response in tumor growth and survival was further increased compared with mice receiving single treatments. Conclusions We have identified several promising candidates for combination therapies in future clinical trials for ETMR patients.


Cancer Cell | 2018

Aberrant ERBB4-SRC Signaling as a Hallmark of Group 4 Medulloblastoma Revealed by Integrative Phosphoproteomic Profiling

Antoine Forget; Loredana Martignetti; Stéphanie Puget; Laurence Calzone; Sebastian Brabetz; Daniel Picard; Arnau Montagud; Stéphane Liva; Alexandre Sta; Florent Dingli; Guillaume Arras; Jaime Rivera; Damarys Loew; Aurore Besnard; Joëlle Lacombe; Mélanie Pagès; Pascale Varlet; Christelle Dufour; Hua Yu; Audrey L. Mercier; Emilie Indersie; Anaïs Chivet; Sophie Leboucher; Laura Sieber; Kevin Beccaria; Michael Gombert; Frauke Meyer; Nan Qin; Jasmin Bartl; Lukas Chavez

The current consensus recognizes four main medulloblastoma subgroups (wingless, Sonic hedgehog, group 3 and group 4). While medulloblastoma subgroups have been characterized extensively at the (epi-)genomic and transcriptomic levels, the proteome and phosphoproteome landscape remain to be comprehensively elucidated. Using quantitative (phospho)-proteomics in primary human medulloblastomas, we unravel distinct posttranscriptional regulation leading to highly divergent oncogenic signaling and kinase activity profiles in groups 3 and 4 medulloblastomas. Specifically, proteomic and phosphoproteomic analyses identify aberrant ERBB4-SRC signaling in group 4. Hence, enforced expression of an activated SRC combined with p53 inactivation induces murine tumors that resemble group 4 medulloblastoma. Therefore, our integrative proteogenomics approach unveils an oncogenic pathway and potential therapeutic vulnerability in the most common medulloblastoma subgroup.


Journal of Clinical Oncology | 2018

Extensive Molecular and Clinical Heterogeneity in Patients With Histologically Diagnosed CNS-PNET Treated as a Single Entity: A Report From the Children’s Oncology Group Randomized ACNS0332 Trial

Eugene I. Hwang; Marcel Kool; Peter C. Burger; David Capper; Lukas Chavez; Sebastian Brabetz; Chris Williams-Hughes; Catherine A. Billups; Linda Heier; Alok Jaju; Jeff M. Michalski; Yimei Li; Sarah Leary; Tianni Zhou; Andreas von Deimling; David Jones; Maryam Fouladi; Ian F. Pollack; Amar Gajjar; Roger J. Packer; Stefan M. Pfister; James M. Olson

PURPOSE Children with histologically diagnosed high-risk medulloblastoma, supratentorial primitive neuroectodermal tumor of the CNS (CNS-PNET), and pineoblastoma (PBL) have had poor survival despite intensive treatment. We included these patients in this Childrens Oncology Group trial. Molecular profiling later revealed tumor heterogeneity that was not detectable at protocol inception. Enrollment of patients with CNS-PNET/PBL was subsequently discontinued, and outcomes for this part of the study are reported here. PATIENTS AND METHODS In this phase III, four-arm prospective trial, consenting children age 3-22 years with newly diagnosed CNS-PNET were randomly assigned (1:1) to receive carboplatin during radiation and/or adjuvant isotretinoin after standard intensive therapy. Primary outcome measure was event-free survival (EFS) in the intent-to-treat population. Molecular tumor classification was retrospectively completed using DNA methylation profiling. RESULTS Eighty-five participants with institutionally diagnosed CNS-PNETs/PBLs were enrolled. Of 60 patients with sufficient tissue, 31 were nonpineal in location, of which 22 (71%) represented tumors that were not intended for trial inclusion, including 18 high-grade gliomas (HGGs), two atypical teratoid rhabdoid tumors, and two ependymomas. Outcomes across tumor types were strikingly different. Patients with supratentorial embryonal tumors/PBLs exhibited 5-year EFS and overall survival of 62.8% (95% CI, 43.4% to 82.2%) and 78.5% (95% CI, 62.2% to 94.8%), respectively, whereas patients with molecularly classified HGG had EFS and overall survival of 5.6% (95% CI, 0% to 13.0%) and 12.0% (95% CI, 0% to 24.7%), respectively. Neither carboplatin, nor isotretinoin significantly altered outcomes for all patients. Survival for patients with HGG was similar to that of historic studies that avoid craniospinal irradiation and intensive chemotherapy. CONCLUSION For patients with CNS-PNET/PBL, prognosis is considerably better than previously assumed when molecularly confirmed HGGs are removed. Identification of molecular HGGs may spare affected children from unhelpful intensive treatment. This trial highlights the challenges of a histology-based diagnosis for pediatric brain tumors and indicates that molecular profiling should become a standard component of initial diagnosis.


Clinical Cancer Research | 2016

Abstract A25: Establishment of orthotopic patient-derived xenograft models of pediatric brain tumors – the Heidelberg experience

Norman Mack; Sebastian Brabetz; Xanthopolous Christina; David Sumerauer; Heidi Bächli; Arnulf Pekrun; Martin U. Schuhmann; Stefan M. Pfister; Olaf Witt; Till Milde; Marcel Kool

Solid tumors of the nervous system are the most common childhood cancers after leukemias. Although brain tumors are the leading cause of cancer-related mortality in children, there are not enough adequate model systems to study their biology. We therefore started a pediatric preclinical testing program in Heidelberg to generate orthotopic patient-derived xenograft (PDX) models for a large variety of pediatric brain tumors. Freshly dissected primary material from multiple centers is being sent to us immediately after surgical resections. One part of the tumor is being reserved for pathological and molecular analysis and the other part is being dissociated into a single cell suspension and injected into the brain of immunodeficient mice. After successful engraftment and passaging, extensive molecular characterization of the PDX tumor and the matching primary tumor are being performed. Thus far, we have injected 95 tumors: 36 low-grade gliomas (LGG), 23 medulloblastomas (MB), 13 ependymomas (EPN), 7 high-grade gliomas (HGG), 6 atypical teratoid rhaboid tumors (AT/RT), 3 meningeal tumors (MT), 3 embryonal tumors with multilayered rosettes (ETMR), 2 gliomatosis cerebri (GC), 1 dysembryoplastic neuroepithelial tumor (DNT) and 1 primitive neuroectodermal tumor (PNET). No engraftment was observed for any of the low-grade tumors (LGG, MT, DNT). For high-grade tumors we established initial engraftments of MB (5/23, 22%), EPN (5/13, 38%), HGG (2/7, 29%), AT/RT (2/6, 33%), ETMR (1/3, 33%) and PNET (1/1, 100%). 11 out of 16 (69%) established PDX models were already passaged at least twice in mice and can be used for preclinical experiments. We conclude that it is possible to generate preclinical models for most malignant pediatric brain tumor entities, but not for low-grade tumors using our current protocol. However, even for the malignant entities there seems to be a selection for only the most aggressive subtypes that successfully engraft. Therefore, due to the low engraftment rate of some tumor types, the rarity of pediatric brain tumors and the multitude of different subtypes, international collaborations are absolutely necessary in this field in order to generate and characterize a broad repertoire of PDX models for all pediatric brain tumor subtypes for preclinical testing. Citation Format: Norman L. Mack, Sebastian Brabetz, Florian Selt, Xanthopolous Christina, David Sumerauer, Heidi Bachli, Arnulf Pekrun, Martin U. Schuhmann, Stefan M. Pfister, Olaf Witt, Till Milde, Marcel Kool. Establishment of orthotopic patient-derived xenograft models of pediatric brain tumors – the Heidelberg experience. [abstract]. In: Proceedings of the AACR Special Conference: Patient-Derived Cancer Models: Present and Future Applications from Basic Science to the Clinic; Feb 11-14, 2016; New Orleans, LA. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(16_Suppl):Abstract nr A25.

Collaboration


Dive into the Sebastian Brabetz's collaboration.

Top Co-Authors

Avatar

Stefan M. Pfister

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

Till Milde

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

James M. Olson

Fred Hutchinson 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

David T. W. Jones

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lukas Chavez

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Xiao-Nan Li

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

David Capper

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Huriye Seker-Cin

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge