26 results on '"Huriye Seker-Cin"'
Search Results
2. Supplementary Figures and Legends from Functional Precision Medicine Identifies New Therapeutic Candidates for Medulloblastoma
- Author
-
Robert J. Wechsler-Reya, Jill P. Mesirov, Marcel Kool, Pablo Tamayo, Stefan M. Pfister, Eliezer M. Van Allen, Michael L. Levy, John R. Crawford, Denise Malicki, Shareef A. Nahas, David P. Dimmock, Terence C. Wong, Matija Snuderl, Iris Reyes, James M. Olson, Xiao-Nan Li, Yoon-Jae Cho, Till Milde, Kristiina Vuori, Michael E. Berens, Jacob J. Henderson, Patricia A. Baxter, Yuchen Du, Mari Kogiso, Lin Qi, Jonas Ecker, Jonathan Serrano, Susanne Gröbner, Brendan Reardon, Huriye Seker-Cin, Darren Finlay, Yoko T. Udaka, Sameerah Wahab, Silvia K. Tacheva-Grigorova, Lianne Q. Chau, Alexandra Garancher, James Jensen, Sebastian Brabetz, Edwin F. Juarez, and Jessica M. Rusert
- Abstract
Supplementary Figures 1-5
- Published
- 2023
3. Data from Functional Precision Medicine Identifies New Therapeutic Candidates for Medulloblastoma
- Author
-
Robert J. Wechsler-Reya, Jill P. Mesirov, Marcel Kool, Pablo Tamayo, Stefan M. Pfister, Eliezer M. Van Allen, Michael L. Levy, John R. Crawford, Denise Malicki, Shareef A. Nahas, David P. Dimmock, Terence C. Wong, Matija Snuderl, Iris Reyes, James M. Olson, Xiao-Nan Li, Yoon-Jae Cho, Till Milde, Kristiina Vuori, Michael E. Berens, Jacob J. Henderson, Patricia A. Baxter, Yuchen Du, Mari Kogiso, Lin Qi, Jonas Ecker, Jonathan Serrano, Susanne Gröbner, Brendan Reardon, Huriye Seker-Cin, Darren Finlay, Yoko T. Udaka, Sameerah Wahab, Silvia K. Tacheva-Grigorova, Lianne Q. Chau, Alexandra Garancher, James Jensen, Sebastian Brabetz, Edwin F. Juarez, and Jessica M. Rusert
- Abstract
Medulloblastoma is among the most common malignant brain tumors in children. Recent studies have identified at least four subgroups of the disease that differ in terms of molecular characteristics and patient outcomes. Despite this heterogeneity, most patients with medulloblastoma receive similar therapies, including surgery, radiation, and intensive chemotherapy. Although these treatments prolong survival, many patients still die from the disease and survivors suffer severe long-term side effects from therapy. We hypothesize that each patient with medulloblastoma is sensitive to different therapies and that tailoring therapy based on the molecular and cellular characteristics of patients' tumors will improve outcomes. To test this, we assembled a panel of orthotopic patient-derived xenografts (PDX) and subjected them to DNA sequencing, gene expression profiling, and high-throughput drug screening. Analysis of DNA sequencing revealed that most medulloblastomas do not have actionable mutations that point to effective therapies. In contrast, gene expression and drug response data provided valuable information about potential therapies for every tumor. For example, drug screening demonstrated that actinomycin D, which is used for treatment of sarcoma but rarely for medulloblastoma, was active against PDXs representing Group 3 medulloblastoma, the most aggressive form of the disease. Functional analysis of tumor cells was successfully used in a clinical setting to identify more treatment options than sequencing alone. These studies suggest that it should be possible to move away from a one-size-fits-all approach and begin to treat each patient with therapies that are effective against their specific tumor.Significance:These findings show that high-throughput drug screening identifies therapies for medulloblastoma that cannot be predicted by genomic or transcriptomic analysis.
- Published
- 2023
4. Analysis of Rare Fusions in Nsclc: Genomic Architecture and Clinical Implications
- Author
-
Huriye Seker-Cin, Timothy Tay Kwang Yong, Daniel Kazdal, Klaus Kluck, Markus Ball, Olaf Neumann, Hauke Winter, Felix J. Herth, Claus-Peter Heußel, Rajkumar Savai, Peter Schirmacher, Michael Thomas, Jan Budczies, Michael Allgäuer, Petros Christopoulos, Albrecht Stenzinger, and Anna-Lena Volckmar
- Published
- 2023
5. Targeting rare and non-canonical driver variants in NSCLC – An uncharted clinical field
- Author
-
Eugen Rempel, Peter Horak, Roland Penzel, Anna-Lena Volckmar, Claus Peter Heußel, Michael Allgäuer, Michael Thomas, Regine Brandt, Huriye Seker-Cin, Mark Kriegsmann, Volker Endris, Jan Budczies, Olaf Neumann, Jonas Leichsenring, Tilman Brummer, Felix J.F. Herth, Petros Christopoulos, M. Faehling, Jürgen Fischer, Daniel Kazdal, Albrecht Stenzinger, Martina Kirchner, Julia Glade, Tilmann Bochtler, Hauke Winter, Peter Schirmacher, Stefan Fröhling, and Hannah Goldschmid
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,Thoracic Oncology ,Internal medicine ,ROS1 ,medicine ,Humans ,Precision Medicine ,Lung cancer ,business.industry ,High-Throughput Nucleotide Sequencing ,Protein-Tyrosine Kinases ,medicine.disease ,Clinical trial ,030104 developmental biology ,Non canonical ,Precision oncology ,030220 oncology & carcinogenesis ,Mutation ,Molecular targets ,business ,Systematic search - Abstract
Objectives Implementation of tyrosine kinase inhibitors (TKI) and other targeted therapies was a main advance in thoracic oncology with survival gains ranging from several months to years for non-small-cell lung cancer (NSCLC) patients. High-throughput comprehensive molecular profiling is of key importance to identify patients that can potentially benefit from these novel treatments. Material and Methods Next-generation sequencing (NGS) was performed on 4500 consecutive formalin-fixed, paraffin-embedded specimens of advanced NSCLC (n = 4172 patients) after automated extraction of DNA and RNA for parallel detection of mutations and gene fusions, respectively. Results and Conclusion Besides the 24.9 % (n = 1040) of cases eligible for approved targeted therapies based on the presence of canonical alterations in EGFR exons 18–21, BRAF, ROS1, ALK, NTRK, and RET, an additional n = 1260 patients (30.2 %) displayed rare or non-canonical mutations in EGFR (n = 748), BRAF (n = 135), ERBB2 (n = 30), KIT (n = 32), PIK3CA (n = 221), and CTNNB1 (n = 94), for which targeted therapies could also be potentially effective. A systematic literature search in conjunction with in silico evaluation identified n = 232 (5.5 %) patients, for which a trial of targeted treatment would be warranted according to available evidence (NCT level 1, i.e. published data showing efficacy in the same tumor entity). In conclusion, a sizeable fraction of NSCLC patients harbors rare or non-canonical alterations that may be associated with clinical benefit from currently available targeted drugs. Systematic identification and individualized management of these cases can expand applicability of precision oncology in NSCLC and extend clinical gain from established molecular targets. These results can also inform clinical trials.
- Published
- 2021
6. Early Development of Ubiquitous Acanthocytosis and Extravascular Hemolysis in Lung Cancer Patients Receiving Alectinib
- Author
-
Julia Kunz, Christiane Wiedemann, Heidrun Grosch, Katharina Kriegsmann, Stefanie Gryzik, Julia Felden, Michael Hundemer, Huriye Seker-Cin, Miriam Stenzinger, Albrecht Leo, Albrecht Stenzinger, Michael Thomas, and Petros Christopoulos
- Subjects
Cancer Research ,ALK+ NSCLC ,alectinib ,anemia ,hemolysis ,acanthocytosis ,Oncology - Abstract
Alectinib is a standard initial treatment for patients with advanced anaplastic lymphoma kinase (ALK) rearranged non-small-cell lung cancer (NSCLC). The current study analyzed a prospective cohort of 24 consecutive alectinib-treated patients and controls in order to comprehensively characterize longitudinal erythrocyte changes under treatment with ALK inhibitors. Upon starting alectinib, all examined patients developed reticulocytosis and abnormal erythrocyte morphology with anisocytosis and a predominance of acanthocytes (64% of red blood cells on average, range 36–100%) in the peripheral blood smear within approximately 2 weeks. Changes were accompanied by a gradual reduction in Eosin-5-maleimide (EMA) binding, which became pathologic (
- Published
- 2022
- Full Text
- View/download PDF
7. Genomic architecture of FGFR2 fusions in cholangiocarcinoma and its implication for molecular testing
- Author
-
Olaf Neumann, Timothy C. Burn, Michael Allgäuer, Markus Ball, Martina Kirchner, Thomas Albrecht, Anna-Lena Volckmar, Susanne Beck, Volker Endris, Hannah Goldschmid, Ulrich Lehmann, Huriye Seker-Cin, Sebastian Uhrig, Stephanie Roessler, Jan Budczies, Stefan Fröhling, Thomas Longerich, Alex H. Wagner, Arndt Vogel, Peter Schirmacher, Albrecht Stenzinger, and Daniel Kazdal
- Subjects
Cholangiocarcinoma ,Cancer Research ,Bile Ducts, Intrahepatic ,Oncology ,Bile Duct Neoplasms ,Molecular Diagnostic Techniques ,Humans ,Genomics ,Receptor, Fibroblast Growth Factor, Type 2 - Abstract
Background Cholangiocarcinoma (CCA) is a primary malignancy of the biliary tract with a dismal prognosis. Recently, several actionable genetic aberrations were identified with significant enrichment in intrahepatic CCA, including FGFR2 gene fusions with a prevalence of 10–15%. Recent clinical data demonstrate that these fusions are druggable in a second-line setting in advanced/metastatic disease and the efficacy in earlier lines of therapy is being evaluated in ongoing clinical trials. This scenario warrants standardised molecular profiling of these tumours. Methods A detailed analysis of the original genetic data from the FIGHT-202 trial, on which the approval of Pemigatinib was based, was conducted. Results Comparing different detection approaches and displaying representative cases, we described the genetic landscape and architecture of FGFR2 fusions in iCCA and show biological and technical aspects to be considered for their detection. We elaborated parameters, including a suggestion for annotation, that should be stated in a molecular diagnostic FGFR2 report to allow a complete understanding of the analysis performed and the information provided. Conclusion This study provides a detailed presentation and dissection of the technical and biological aspects regarding FGFR2 fusion detection, which aims to support molecular pathologists, pathologists and clinicians in diagnostics, reporting of the results and decision-making.
- Published
- 2022
8. The immune microenvironment in EGFR- and ERBB2-mutated lung adenocarcinoma
- Author
-
Klaus Kluck, Michael Allgäuer, Stefan Fröhling, Michael Thomas, Hannah Goldschmid, Olaf Neumann, Nikolaj Frost, Petros Christopoulos, Volker Endris, Regine Brandt, Harland S. Winter, Martina Kirchner, Jan Budczies, Albrecht Stenzinger, Daniel Kazdal, Martin Reck, Thomas Muley, S. Perner, Julia Glade, Anna-Lena Volckmar, Huriye Seker-Cin, Mark Kriegsmann, Peter Schirmacher, and Roland Penzel
- Subjects
Cancer Research ,Lung Neoplasms ,Receptor, ErbB-2 ,Adenocarcinoma of Lung ,Histone Deacetylases ,Exon ,Immune system ,Carcinoma, Non-Small-Cell Lung ,medicine ,Tumor Microenvironment ,Humans ,EGFR exon 20 insertion ,Epidermal growth factor receptor ,Lung cancer ,Original Research ,Tumor microenvironment ,immunosuppression ,biology ,business.industry ,ERBB2 exon 20 insertion ,RNA-Binding Proteins ,medicine.disease ,lung adenocarcinoma ,Gene expression profiling ,ErbB Receptors ,Oncology ,Cancer research ,biology.protein ,Quality of Life ,Adenocarcinoma ,business ,Tyrosine kinase - Abstract
Background Targeted therapies have improved survival and quality of life for patients with non-small-cell lung cancer with actionable driver mutations. However, epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 gene (HER2, also known as ERBB2) exon 20 insertions (Ex20mut) are characterized by a poor response to currently approved tyrosine kinase inhibitors and immunotherapies. The underlying immune biology is not well understood. Materials and methods We carried out messenger RNA expression profiling of lung adenocarcinomas (ADCs) with ERBB2 (n = 19) and EGFR exon 20-insertion mutations (n = 13) and compared these to tumors with classical EGFR mutations (n = 40, affecting EGFR exons 18, 19 or 21) and EGFR/ERBB2 mutation-negative lung ADC (EGFR/ERBB2wt, n = 26) focusing on immunologically relevant transcripts. Tumor-infiltrating immune cells were estimated from gene expression profiles. Results Cytotoxic cells were significantly lower in EGFR-mutated tumors regardless of the affected exon, while Th1 cells were significantly lower in EGFR-Ex20mut compared to EGFR/ERBB2wt tumors. We assessed the differentially expressed genes of ERBB2-Ex20mut and EGFR-Ex20mut tumors compared to EGFR-Ex18/19/21mut and EGFR/ERBB2wt tumors. Of these, the genes GUSB, HDAC11, IFNGR2, PUM1, RASGRF1 and RBL2 were up-regulated, while a lower expression of CBLC, GBP1, GBP2, GBP4 and MYC was observed in all three comparison groups. The omnibus test revealed 185 significantly (FDR = 5%) differentially expressed genes and we found these four most significant gene expression changes in the study cohort: VHL and JAK1 were overexpressed in ERBB2-Ex20mut and EGFR-Ex20mut tumors compared to both EGFR-Ex18/19/21mut and EGFR/ERBB2wt tumors. RIPK1 and STK11IP showed the highest expression in ERBB2-Ex20mut tumors. Conclusions Targeted gene expression profiling is a promising tool to read out the characteristics of the tumor microenvironment from routine diagnostic lung cancer biopsies. Significant immune reactivity and specific immunosuppressive characteristics in ERBB2-Ex20mut and EGFR-Ex20mut lung ADC with at least some degree of immune infiltration support further clinical evaluation of immune-modulators as partners of immune checkpoint inhibitors in such tumors., Highlights • Gene expression profiling to characterize the tumor microenvironment is feasible using diagnostic lung cancer biopsies. • EGFR exon 20-mutated tumors show a higher expression of VHL and an immunologic ‘colder’ phenotype than EGFR/ERBB2wt tumors. • ERBB2 exon 20-mutated tumors show an overexpression of RIPK1 and STK11IP and a reduction of cytotoxic natural killer cells. • Drugs targeting these alterations are potential partners of checkpoint blockade in exon 20-mutated non-small-cell lung cancer.
- Published
- 2021
9. Assigning evidence to actionability: An introduction to variant interpretation in precision cancer medicine
- Author
-
Jan Budczies, Laura Gieldon, Olaf Neumann, Stefan Fröhling, Marcus Renner, Christian P. Schaaf, Daniel Kazdal, Nicola Dikow, Peter Horak, Michael Allgäuer, Anna-Lena Volckmar, Christoph E. Heilig, Huriye Seker-Cin, Regine Brandt, Simon Kreutzfeldt, Veronica Teleanu, Roland Penzel, Jonas Leichsenring, Volker Endris, Albrecht Stenzinger, Hannah Goldschmid, Peter Schirmacher, Carolin Ploeger, and Martina Kirchner
- Subjects
Cancer Research ,Functional evaluation ,Scope (project management) ,Interpretation (philosophy) ,Computational Biology ,Computational biology ,Genomics ,Biology ,Molecular biomarkers ,Functional annotation ,Cancer Medicine ,Neoplasms ,Genetics ,Tumor board ,Humans ,Clinical significance ,Precision Medicine - Abstract
Modern concepts in precision cancer medicine are based on increasingly complex genomic analyses and require standardized criteria for the functional evaluation and reporting of detected genomic alterations in order to assess their clinical relevance. In this article, we propose and address the necessary steps in systematic variant evaluation consisting of bioinformatic analysis, functional annotation and clinical interpretation, focusing on the latter two aspects. We discuss the role and clinical application of current variant classification systems and point out their scope and limitations. Finally, we highlight the significance of the molecular tumor board as a platform for clinical decision-making based on genomic analyses.
- Published
- 2021
10. Functional precision medicine identifies new therapeutic candidates for medulloblastoma
- Author
-
Eliezer M. Van Allen, Terence C. Wong, Jessica M. Rusert, Susanne Gröbner, Silvia K. Tacheva-Grigorova, Kristiina Vuori, Jill P. Mesirov, Robert J. Wechsler-Reya, Darren Finlay, Jonas Ecker, Denise M. Malicki, Shareef Nahas, David Dimmock, Brendan Reardon, James M. Olson, Till Milde, Sameerah Wahab, Matija Snuderl, Jonathan Serrano, Mari Kogiso, Huriye Seker-Cin, Patricia Baxter, Stefan M. Pfister, Michael J. Levy, Yoon Jae Cho, Jacob J. Henderson, Yuchen Du, James Jensen, Alexandra Garancher, Edwin F. Juarez, Michael E. Berens, Iris Reyes, Sebastian Brabetz, Xiao-Nan Li, Marcel Kool, Yoko T. Udaka, Pablo Tamayo, Lianne Q. Chau, John R. Crawford, and Lin Qi
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Disease ,Transcriptome ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,Medicine ,Precision Medicine ,Child ,Exome sequencing ,Cancer ,media_common ,Pediatric ,Tumor ,Single Nucleotide ,Genomics ,Gene Expression Regulation, Neoplastic ,Pharmaceutical Preparations ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Dactinomycin ,Sarcoma ,Development of treatments and therapeutic interventions ,Biotechnology ,Drug ,Pediatric Research Initiative ,medicine.medical_specialty ,Pediatric Cancer ,media_common.quotation_subject ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Polymorphism, Single Nucleotide ,Article ,Cell Line ,03 medical and health sciences ,Rare Diseases ,Internal medicine ,Cell Line, Tumor ,Exome Sequencing ,Genetics ,Animals ,Humans ,Genetic Testing ,Oncology & Carcinogenesis ,Polymorphism ,Cerebellar Neoplasms ,Medulloblastoma ,Neoplastic ,business.industry ,Human Genome ,Neurosciences ,Precision medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,Brain Disorders ,High-Throughput Screening Assays ,Brain Cancer ,Gene expression profiling ,Orphan Drug ,Good Health and Well Being ,030104 developmental biology ,Gene Expression Regulation ,Mutation ,Inbred NOD ,Generic health relevance ,business - Abstract
Medulloblastoma is among the most common malignant brain tumors in children. Recent studies have identified at least four subgroups of the disease that differ in terms of molecular characteristics and patient outcomes. Despite this heterogeneity, most patients with medulloblastoma receive similar therapies, including surgery, radiation, and intensive chemotherapy. Although these treatments prolong survival, many patients still die from the disease and survivors suffer severe long-term side effects from therapy. We hypothesize that each patient with medulloblastoma is sensitive to different therapies and that tailoring therapy based on the molecular and cellular characteristics of patients' tumors will improve outcomes. To test this, we assembled a panel of orthotopic patient-derived xenografts (PDX) and subjected them to DNA sequencing, gene expression profiling, and high-throughput drug screening. Analysis of DNA sequencing revealed that most medulloblastomas do not have actionable mutations that point to effective therapies. In contrast, gene expression and drug response data provided valuable information about potential therapies for every tumor. For example, drug screening demonstrated that actinomycin D, which is used for treatment of sarcoma but rarely for medulloblastoma, was active against PDXs representing Group 3 medulloblastoma, the most aggressive form of the disease. Functional analysis of tumor cells was successfully used in a clinical setting to identify more treatment options than sequencing alone. These studies suggest that it should be possible to move away from a one-size-fits-all approach and begin to treat each patient with therapies that are effective against their specific tumor. Significance: These findings show that high-throughput drug screening identifies therapies for medulloblastoma that cannot be predicted by genomic or transcriptomic analysis.
- Published
- 2020
11. A biobank of patient-derived pediatric brain tumor models
- Author
-
Volker Hovestadt, Paul A. Northcott, Kyle Pedro, Sebastian Brabetz, Susanne Gröbner, Gnana Prakash Balasubramanian, Stefan M. Pfister, Huriye Seker-Cin, Sarah Leary, Peter Lichter, Karina Bloom, Madison W. Nakamoto, Jan Koster, Andrey Korshunov, Stacey Hansen, Joyoti Dey, Andrew D. Strand, Marcel Kool, Bonnie Cole, Fiona Pakiam, Emily J. Girard, Norman Mack, David T.W. Jones, Lukas Chavez, Sally Ditzler, Benjamin Schwalm, James M. Olson, Oncogenomics, and CCA - Imaging and biomarkers
- Subjects
0301 basic medicine ,Male ,Genomics ,medicine.disease_cause ,Pediatrics ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Child ,Biological Specimen Banks ,Mutation ,business.industry ,Brain Neoplasms ,Cancer ,General Medicine ,DNA Methylation ,medicine.disease ,Biobank ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,3. Good health ,030104 developmental biology ,Child, Preschool ,DNA methylation ,Cancer research ,Pediatric Brain Tumor ,Female ,business - Abstract
Brain tumors are the leading cause of cancer-related death in children. Genomic studies have provided insights into molecular subgroups and oncogenic drivers of pediatric brain tumors that may lead to novel therapeutic strategies. To evaluate new treatments, better preclinical models adequately reflecting the biological heterogeneity are needed. Through the Children’s Oncology Group ACNS02B3 study, we have generated and comprehensively characterized 30 patient-derived orthotopic xenograft models and seven cell lines representing 14 molecular subgroups of pediatric brain tumors. Patient-derived orthotopic xenograft models were found to be representative of the human tumors they were derived from in terms of histology, immunohistochemistry, gene expression, DNA methylation, copy number, and mutational profiles. In vivo drug sensitivity of targeted therapeutics was associated with distinct molecular tumor subgroups and specific genetic alterations. These models and their molecular characterization provide an unprecedented resource for the cancer community to study key oncogenic drivers and to evaluate novel treatment strategies.
- Published
- 2018
12. Non-random aneuploidy specifies subgroups of pilocytic astrocytoma and correlates with older age
- Author
-
László Bognár, Sébastien Brunet, David T.W. Jones, Andrey Korshunov, Geneviève Bourret, Denise Bechet, Dong-Anh Khuong-Quang, Jose-Luis Montes, Nicolas De Jay, Noha Gerges, Pierre Lepage, Huriye Seker-Cin, Tenzin Gayden, Tony Kwan, V. Peter Collins, Uri Tabori, Margret Shirinian, Werner Paulus, M Kool, Stefan M. Pfister, Adam M. Fontebasso, Hendrik Witt, Karine Jacob, Barbara Hutter, Jean-Pierre Farmer, Peter Hauser, Almos Klekner, Damien Faury, Jeffrey Atkinson, Nada Jabado, Steffen Albrecht, Alexandre Montpetit, Sally R. Lambert, Miklós Garami, and Martin Hasselblatt
- Subjects
Adult ,Male ,Proto-Oncogene Proteins B-raf ,Brain tumor ,Aneuploidy ,Astrocytoma ,Real-Time Polymerase Chain Reaction ,Klinikai orvostudományok ,medicine.disease_cause ,BRAF ,Cohort Studies ,Young Adult ,MDM2 ,Biomarkers, Tumor ,medicine ,Humans ,RNA, Messenger ,Receptor, Fibroblast Growth Factor, Type 1 ,aneuploidy ,pilocytic astrocytoma ,Young adult ,Child ,Neoplasm Staging ,Mutation ,biology ,Pilocytic astrocytoma ,Brain Neoplasms ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,aging ,Age Factors ,Proto-Oncogene Proteins c-mdm2 ,Orvostudományok ,Cell cycle ,Prognosis ,medicine.disease ,Oncology ,biology.protein ,Cancer research ,Mdm2 ,Female ,PLK2 ,Research Paper - Abstract
Pilocytic astrocytoma (PA) is the most common brain tumor in children but is rare in adults, and hence poorly studied in this age group. We investigated 222 PA and report increased aneuploidy in older patients. Aneuploid genomes were identified in 45% of adult compared with 17% of pediatric PA. Gains were non-random, favoring chromosomes 5, 7, 6 and 11 in order of frequency, and preferentially affecting non-cerebellar PA and tumors with BRAF V600E mutations and not with KIAA1549-BRAF fusions or FGFR1 mutations. Aneuploid PA differentially expressed genes involved in CNS development, the unfolded protein response, and regulators of genomic stability and the cell cycle (MDM2, PLK2),whose correlated programs were overexpressed specifically in aneuploid PA compared to other glial tumors. Thus, convergence of pathways affecting the cell cycle and genomic stability may favor aneuploidy in PA, possibly representing an additional molecular driver in older patients with this brain tumor.
- Published
- 2015
13. PM-09ESTABLISHMENT AND MOLECULAR CHARACTERIZATION OF PATIENT-DERIVED XENOGRAFT MODELS OF PEDIATRIC BRAIN TUMORS
- Author
-
Susanne Gröbner, Kyle Pedro, Stefan M. Pfister, Marcel Kool, Huriye Seker-Cin, Robert J. Wechsler-Reya, Jessica M. Rusert, Volker Hovestadt, Paul A. Northcott, Sebastian Brabetz, James M. Olson, Xiao-Nan Li, Karina Bloom, Till Milde, and David T.W. Jones
- Subjects
Ependymoma ,Medulloblastoma ,Cancer Research ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease ,Bioinformatics ,Primary tumor ,Gene expression profiling ,Oncology ,Atypical teratoid rhabdoid tumor ,Cancer research ,medicine ,Neurology (clinical) ,Abstracts from the 3rd Biennial Conference on Pediatric Neuro-Oncology Basic and Translational Research ,Exome sequencing - Abstract
Brain tumors are the leading cause of cancer-related mortality in childhood. Based on the many recent genomic studies, we know now that multiple molecular subtypes of brain cancers exist that are not only biologically but also clinically highly distinct. These findings may lead to novel treatment strategies. For this we need better preclinical models that correctly reflect the proper tumor (sub)type. Orthotopic patient-derived xenograft (PDX) models generated by intracranial injection of primary patient material into the brain of NOD scid gamma (NSG) mice offer the unique possibility to test novel substances in primary patient tissue in an in vivo environment. Prior to drug selection and testing, extensive molecular characterizations are needed to learn about targetable oncogenic drivers in each model. Therefore, we aim to generate a large repertoire of PDX models reflecting the many different molecular subtypes of pediatric brain cancer. For each established PDX model, we perform DNA methylation profiling, gene expression profiling, low-coverage whole genome sequencing and whole exome sequencing and compare these data with the matching primary tumor. Thus far, as a collaborative effort between different laboratories, we have established and fully characterized 34 PDX models from 1 atypical teratoid rhabdoid tumor (ATRT), 4 ependymoma, 9 glioblastoma, 18 medulloblastoma, and 2 primitive neuroectodermal tumors (PNET). Molecular analysis of all available PDX models identified a clear overrepresentation of most aggressive tumors such as models characterized by MYC- or MYCN amplification. Other, less aggressive cancers, like Wnt medulloblastoma, are underrepresented. For in vivo imaging during treatment of PDX models we created luciferase labeled PDX sublines. Our data demonstrate that PDX models retain characteristics of the primary human tumors from which they were derived. These reagents provide an unprecedented resource to study tumor biology and pave the way for improving treatment strategies of malignant pediatric brain tumors.
- Published
- 2015
14. PDTB-23. CHEMI-GENOMIC ANALYSIS OF PATIENT-DERIVED XENOGRAFTS TO IDENTIFY PERSONALIZED THERAPIES FOR MEDULLOBLASTOMA
- Author
-
Robert J. Wechsler-Reya, James Jensen, Alexandra Garancher, Mari Kogiso, Sebastian Brabetz, Huriye Seker-Cin, Yoko T. Udaka, Pablo Tamayo, Stefan M. Pfister, Yoon Jae Cho, Simone Schubert, Michael J. Levy, Lin Qi, Marcel Kool, Lourdes Adriana Esparza, Xiao-Nan Li, John R. Crawford, Jill P. Mesirov, James M. Olson, Till Milde, and Jessica M. Rusert
- Subjects
Medulloblastoma ,Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Neurology (clinical) ,business ,medicine.disease - Published
- 2016
15. Abstract 1935: Molecular characterization of orthotopic patient-derived xenograft models of pediatric brain tumors
- Author
-
Xiao-Nan Li, Sarah Leary, Kyle Pedro, Robert J. Wechsler-Reya, Huriye Seker-Cin, Sebastian Brabetz, Marcel Kool, Madison T. Wise, Florian Selt, Till Milde, Jessica M. Rusert, Andy Strand, Olaf Witt, James M. Olson, David T.W. Jones, Stefan M. Pfister, and Susanne Gröbner
- Subjects
Cancer Research ,Oncology ,Pediatric brain ,business.industry ,Cancer research ,Medicine ,business ,Tumor xenograft - Abstract
[Introduction: Solid tumors of the nervous system are the most common childhood cancers after leukemias. Even though we might be able to cure more and more patients, survivors still severely suffer long-term from the intensive treatments. Therefore, new treatment strategies are urgently needed. Orthotopic patient-derived xenograft (PDX) models are an excellent platform for biomarker and preclinical drug development. However, the rarity of pediatric brain tumors and the multitude of different sub entities hinder the generation of large collection of PDX models of specific entities within single institutions. In order to generate an overview about existing PDX models in the community, we started collecting established PDX models from various centers all over the world and performed extensive molecular characterization to precisely determine the distinct molecular subgroup and constellation of genetic alterations for each PDX model, and thus identify its targetable oncogenic drivers. Material and Methods: PDX models were established and maintained by dissociating tumor material into a single cell suspension and then orthotopically injecting it into the brain of immunodeficient animals. All PDX models and matching primary tumors (if available) have been analyzed by whole-exome and low-coverage whole-genome sequencing, as well as DNA methylation and gene expression profiling at the German Cancer Research Center (DKFZ). Results and Discussion: Thus far, we have collected and characterized 70 established PDX models from 6 ATRTs, 8 ependymomas, 16 high-grade gliomas, 38 medulloblastomas, and 2 CNS-PNETs. PDX models always retain their molecular subtype and in the vast majority of cases also the mutations and copy number alterations when compared to their primary tumors. Only in rare cases do we observe additional aberrations, which most likely represent outgrowths of subclones from the primary tumor. Analysis of our entire cohort identified an overrepresentation of the most aggressive tumor subtypes, but also subtypes which have not been available for preclinical testing before due to lack of genetically engineered mouse models or suitable cell lines, such as Group 4 medulloblastoma. Based on our current analysis, the PDX models within the community are not yet covering the entire heterogeneity within the patient population. As a follow up, we aim to make these models and data accessible in a user-friendly manner so that the community can use them for preclinical research. Conclusion: PDX models of pediatric brain tumors are very rare. Our molecular characterization allows researchers all over the world to find the right models for their specific scientific question. Therefore, this work will provide an unprecedented resource to study tumor biology and pave the way for improving treatment strategies for children with malignant brain tumors. Citation Format: Sebastian Brabetz, Susanne N. Gröbner, Huriye Seker-Cin, Florian Selt, Till Milde, David T. Jones, Madison T. Wise, Jessica M. Rusert, Kyle Pedro, Andy Strand, Olaf Witt, Sarah E. Leary, Xiao-Nan Li, Robert J. Wechsler-Reya, James M. Olson, Stefan M. Pfister, Marcel Kool. Molecular characterization of orthotopic patient-derived xenograft models of pediatric brain tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1935. doi:10.1158/1538-7445.AM2017-1935
- Published
- 2017
16. TMOD-05. MOLECULAR CHARACTERIZATION OF ORTHOTOPIC PATIENT-DERIVED XENOGRAFT MODELS OF PEDIATRIC BRAIN TUMORS AND THEIR USE IN PRECLINICAL EXPERIMENTS
- Author
-
Lukas Chavez, Till Milde, Jones Dtw, Susanne N. Groebner, Olaf Witt, Xiao-Nan Li, Robert J. Wechsler-Reya, M Kool, Huriye Seker-Cin, Sebastian Brabetz, Sarah Leary, James Olson, Norman Mack, S Pfister, and Christin Schmidt
- Subjects
Abstracts ,Cancer Research ,Text mining ,Oncology ,business.industry ,Pediatric brain ,Cancer research ,Medicine ,Neurology (clinical) ,business ,Tumor xenograft - Abstract
Genomic studies have shown that multiple molecular subtypes of pediatric brain tumors exist that are biologically and clinically highly distinct. These findings ask for novel subtype specific treatments. To develop these we need more and better preclinical models that correctly reflect the proper tumor (sub)type. Orthotopic patient-derived xenograft (PDX) models generated by intracranial injection of primary patient material into the brain of NSG mice offer the unique possibility to test novel substances in primary patient tissue in an in vivo environment. Prior to drug selection and testing, extensive molecular characterizations of PDX and matching primary tumor/blood (DNA methylation, DNA sequencing, and gene expression) are needed to see how the PDX represents the original disease and to learn about targetable oncogenic drivers in each model. In collaboration with several groups around the world we have generated and fully characterized thus far 75 PDX models reflecting 15 distinct subtypes of pediatric brain cancer. PDX models always retain their molecular subtype and in the vast majority of cases also mutations and copy number alterations compared to matching primary tumors. Most aggressive tumors, harboring MYC(N) amplifications, are overrepresented in the cohort, but also subtypes which have not been available for preclinical testing before due to lack of genetically engineered mouse models or suitable cell lines, such as Group 4 medulloblastoma, are included. All models and corresponding molecular data will become available for the community for preclinical research. Examples of such preclinical experiments will be presented. PDX models of pediatric brain tumors are still quite rare. Our repertoire of PDX models and corresponding molecular characterizations allow researchers all over the world to find the right models for their specific scientific questions. It will provide an unprecedented resource to study tumor biology and pave the way for improving treatment strategies for children with malignant brain tumors.
- Published
- 2017
17. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma
- Author
-
Robert J. Wechsler-Reya, Fabian Kratochwil, Ursula D. Weber, Gideon Zipprich, Stephan Wolf, Florence M.G. Cavalli, Adrian M. Stütz, Scott Zuyderduyn, Roland Eils, Peter Lichter, Wei Wang, Charles D. Imbusch, Dominik Sturm, Stefan Wiemann, Guido Reifenberger, Stefan Rutkowski, Martin Ebinger, Wolfram Scheurlen, Hans-Jörg Warnatz, Lourdes Adriana Esparza, Marc Remke, Thomas Zichner, Andrea Wittmann, Martin Mynarek, Eszter Turányi, David Sumerauer, Martin U. Schuhmann, Marcel Kool, Emma Sandén, Chris Lawerenz, Marc Zapatka, Rogier Versteeg, Stefan M. Pfister, Benedikt Brors, Daisuke Kawauchi, Jan O. Korbel, Peter Siesjö, Jan Koster, Volker Hovestadt, Catherine A. Lee, Peter Hauser, David Shih, Cynthia C. Bartholomae, Peter van Sluis, Karel Zitterbart, Giles W. Robinson, Michael D. Taylor, Jörg Felsberg, Torsten Pietsch, Paul A. Northcott, Huriye Seker-Cin, Marie-Laure Yaspo, Benjamin Raeder, Jaroslav Sterba, Katja von Hoff, Richard J. Gilbertson, Sabine Schmidt, David T.W. Jones, Marina Ryzhova, Scott R. VandenBerg, Anna Darabi, Andreas von Deimling, Andrey Korshunov, Thomas Risch, Olaf Witt, Ivo Buchhalter, Laura Sieber, Richard Volckmann, Sebastian Stark, Natalie Jäger, Andreas E. Kulozik, Gary D. Bader, Amar Gajjar, Nicolle Diessl, Serap Erkek, H. Leighton Grimes, Jürgen Eils, Linda Linke, Christof von Kalle, Other departments, Oncogenomics, and CCA -Cancer Center Amsterdam
- Subjects
Somatic cell ,Childhood cancer ,Genomics ,Biology ,Mutually exclusive events ,Article ,03 medical and health sciences ,0302 clinical medicine ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Enhancer ,030304 developmental biology ,Medulloblastoma ,Oncogene Activation ,0303 health sciences ,Multidisciplinary ,Mechanism (biology) ,Oncogenes ,medicine.disease ,3. Good health ,DNA-Binding Proteins ,Repressor Proteins ,Enhancer Elements, Genetic ,030220 oncology & carcinogenesis ,Genomic Structural Variation ,Cancer research ,Transcription Factors - Abstract
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 intertumoural heterogeneity of medulloblastoma, identifying four distinct molecular subgroups. Group 3 and Group 4 subgroup medulloblastomas account for the majority of 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 protooncogenes, GFI1 and GFI1B. Somatic structural variants juxtapose GFI1/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.
- Published
- 2014
18. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy
- Author
-
Stephen C. Mack, Kenneth Aldape, Karin M. Muraszko, James T. Rutka, Livia Garzia, Scott L. Pomeroy, Wiesława Grajkowska, Caretha L. Creasy, Till Milde, Marcel Kool, Joanna J. Phillips, Pedro Castelo-Branco, Martin Hirst, Jan Koster, Andrey Korshunov, David Malkin, Mathieu Lupien, Theophilos Tzaridis, Xiaochong Wu, Xiaoyang Zhang, Peter Lichter, Peter B. Dirks, Loretta Lau, Sharad K. Verma, Sergio L. Pereira, Andreas E. Kulozik, Gary D. Bader, Adrian M. Dubuc, K. Nethery-Brokx, Luca Massimi, Rosario M. Piro, Uri Tabori, T. Van Meter, Jennifer Zuccaro, Hendrik Witt, Ian D. Clarke, Martin Sill, David T.W. Jones, Sonja Hutter, Khalida Wani, Scott Zuyderduyn, Marina Ryzhova, L. Gu, Eric Bouffet, Mark Barszczyk, T. J. Pugh, Patrick Sin-Chan, Renee Head, Sameer Agnihotri, Roland Eils, Paul Kongkham, Adrian M. Stütz, Daniel W. Fults, John Peacock, Kelsey C. Bertrand, Sebastian Bender, Olaf Witt, Pascal Johann, Paul A. Northcott, Boleslaw Lach, Thomas Zichner, Sebastian Stark, Annie Huang, Marc Remke, Natalie Jäger, Huriye Seker-Cin, Cynthia Hawkins, Yuan Yao, Marco Gallo, Kory Zayne, Xin Wang, Claudia C. Faria, Volker Hovestadt, Xing Fan, Nalin Gupta, Michael D. Taylor, Stefan M. Pfister, Swneke D. Bailey, Yoon Jae Cho, Florence M.G. Cavalli, William A. Weiss, Marco A. Marra, Jan O. Korbel, Stephen W. Scherer, Nada Jabado, Stephen S. Roberts, A. von Deimling, David Shih, Vijay Ramaswamy, Graduate School, Oncogenomics, and Other departments
- Subjects
Regulation of gene expression ,Multidisciplinary ,CpG Island Methylator Phenotype ,NF-kappa B ,Transcription Factor RelA ,Proteins ,Hindbrain ,Biology ,Article ,Epigenesis, Genetic ,Cell Transformation, Neoplastic ,CpG site ,Ependymoma ,Immunology ,DNA methylation ,Cancer research ,Animals ,Humans ,Pediatric ependymoma ,CpG Islands ,Female ,Epigenetics ,Epigenomics ,Signal Transduction - Abstract
Ependymomas are common childhood brain tumours that occur throughout the nervous system, but are most common in the paediatric hindbrain. Current standard therapy comprises surgery and radiation, but not cytotoxic chemotherapy as it does not further increase survival. Whole-genome and whole-exome sequencing of 47 hindbrain ependymomas reveals an extremely low mutation rate, and zero significant recurrent somatic single nucleotide variants. Although devoid of recurrent single nucleotide variants and focal copy number aberrations, poor-prognosis hindbrain ependymomas exhibit a CpG island methylator phenotype. Transcriptional silencing driven by CpG methylation converges exclusively on targets of the Polycomb repressive complex 2 which represses expression of differentiation genes through trimethylation of H3K27. CpG island methylator phenotype-positive hindbrain ependymomas are responsive to clinical drugs that target either DNA or H3K27 methylation both in vitro and in vivo. We conclude that epigenetic modifiers are the first rational therapeutic candidates for this deadly malignancy, which is epigenetically deregulated but genetically bland.
- Published
- 2014
19. Targeting H3.3 mutated pediatric high grade gliomas – A high throughput screening approach to identify novel therapeutic agents
- Author
-
N Truffaux, Huriye Seker-Cin, Dominik Sturm, S Stark, J Lewis, Till Milde, S Pfister, J Grill, Andreas E. Kulozik, Olaf Witt, Peter Lichter, Sebastian Bender, and Pascal Johann
- Subjects
business.industry ,Pediatrics, Perinatology and Child Health ,Medicine ,Computational biology ,business ,Throughput (business) - Published
- 2013
20. HDAC and PI3K Antagonists Cooperate to Inhibit Growth of MYC-Driven Medulloblastoma
- Author
-
Alexandra Garancher, Sorana Morrissy, Jun Wang, Simone Schubert, Lourdes Adriana Esparza, Ran Tao, Donna L. Maier, Mari Kogiso, Huriye Seker-Cin, Stefan M. Pfister, Yanxin Pei, Kun Wei Liu, James M. Olson, Yoon Jae Cho, Yoko T. Udaka, Michael J. Levy, Sebastian Brabetz, John R. Crawford, Marcel Kool, Najiba Murad, Lin Qi, Robert J. Wechsler-Reya, Xiao-Nan Li, and Michael D. Taylor
- Subjects
0301 basic medicine ,Cancer Research ,FOXO1 ,Inbred C57BL ,Phosphatidylinositol 3-Kinases ,Mice ,Genes, Tumor Suppressor ,Cancer ,Phosphoinositide-3 Kinase Inhibitors ,Pediatric ,Forkhead Transcription Factors ,3. Good health ,Oncology ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Tumor Suppressor ,Biotechnology ,Pediatric Research Initiative ,Combination therapy ,Tumor suppressor gene ,Pediatric Cancer ,Oncology and Carcinogenesis ,Biology ,Article ,Histone Deacetylases ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Rare Diseases ,In vivo ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,Cerebellar Neoplasms ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Medulloblastoma ,Animal ,Cell growth ,Neurosciences ,Cell Biology ,medicine.disease ,Brain Disorders ,Brain Cancer ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Genes ,Disease Models ,Cancer research ,Histone deacetylase - Abstract
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 (HDACI). HDACI potently inhibit survival of MYC-driven MB cells in vitro, in part by inducing expression of the FOXO1 tumor suppressor gene. HDACI also synergize with phosphatidylinositol 3-kinase inhibitors (PI3KI) to inhibit tumor growth in vivo. These studies identify an effective combination therapy for the most aggressive form of MB.
- Published
- 2013
21. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma
- Author
-
Till Milde, Sonja Hutter, Cynthia C. Bartholomae, Peter van Sluis, Camelia M. Monoranu, Sabine Schmidt, Martin Hasselblatt, Richard Volckmann, Chris Lawerenz, Adrian M. Stütz, Marina Ryzhova, David T.W. Jones, Marc Sultan, Benedikt Brors, Paul A. Northcott, Damien Faury, Marcel Kool, Charles D. Imbusch, S. Radomski, Sally R. Lambert, Huriye Seker-Cin, V. Peter Collins, Bärbel Lasitschka, Martin Ebinger, Olaf Witt, Yoon Jae Cho, Sebastian Bender, Jan O. Korbel, Stephan Wolf, Jeremy Schwartzentruber, Dominik Sturm, Hans-Jörg Warnatz, Keith L. Ligon, Barbara Hutter, Andrey Korshunov, Andreas Unterberg, Dong Anh Khuong Quang, Marc Zapatka, Nada Jabado, Christel Herold-Mende, Jan Gronych, Rogier Versteeg, Jörg Felsberg, Catherine L. Worth, Martin U. Schuhmann, Jan Koster, Marie-Laure Yaspo, Peter Lichter, Matthias A. Karajannis, Benjamin Raeder, Scott L. Pomeroy, Matthias Schlesner, Wolfram Scheurlen, Mark W. Kieran, Meryem Ralser, Ursula D. Weber, Hans Lehrach, Adam M. Fontebasso, Thomas Zichner, Andreas E. Kulozik, Beate Winkler, Sebastian Stark, Elke Pfaff, Natalie Jäger, Jacek Majewski, Andreas von Deimling, Hendrik Witt, Christof von Kalle, Charles G. Eberhart, Volker Hovestadt, Michael D. Taylor, Roland Eils, Guido Reifenberger, Marc Zuckermann, Other departments, Oncogenomics, and CCA -Cancer Center Amsterdam
- Subjects
Mutation ,Pilocytic astrocytoma ,Astrocytoma ,Disease ,Biology ,medicine.disease ,Bioinformatics ,medicine.disease_cause ,Article ,nervous system diseases ,Fusion gene ,PTPN11 ,Genetics ,medicine ,Cancer research ,Childhood Glioblastoma ,ddc:610 ,Gene ,neoplasms - Abstract
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.
- Published
- 2013
22. Abstract A07: Molecular characterization of patient-derived xenograft models of pediatric brain tumors
- Author
-
Marcel Kool, Xiao-Nan Li, Norman Mack, Sebastian Brabetz, David T.W. Jones, Kyle Pedro, Huriye Seker-Cin, Susanne Gröbner, Robert J. Wechsler-Reya, Till Milde, Volker Hovestadt, Karina Bloom, Stefan M. Pfister, Jessica M. Rusert, James M. Olson, and Madison T. Wise
- Subjects
Cancer Research ,Basic science ,Brain tumor ,Chromosome ,Cancer ,Biology ,Bioinformatics ,medicine.disease ,Primary tumor ,Gene expression profiling ,Oncology ,Gene expression ,DNA methylation ,Cancer research ,medicine - Abstract
Recent genomic studies have revealed multiple molecular subtypes of pediatric brain cancers that are not only biologically but also clinically distinct. In order to develop novel treatment strategies for these often fatal diseases we need more preclinical models like orthotopic patient-derived xenograft (PDX) models that correctly reflect the many different tumor types. Prior to drug selection and testing, extensive molecular characterizations are needed to precisely assign a distinct molecular subgroup to each PDX model and to learn about its targetable oncogenic drivers. In an international effort we aim to characterize a large repertoire of PDX models reflecting the many different molecular subtypes of pediatric brain cancer. Thus far, we have collected and characterized 64 established PDX models from 6 atypical teratoid rhabdoid tumors (AT/RT), 7 ependymomas (EPN), 16 high-grade gliomas (HGG), 32 medulloblastomas (MB), and 3 primitive neuroectodermal tumors (PNET). All PDX models and their matching primary tumors (if available) are analyzed by whole-exome and low-coverage whole-genome sequencing, as well as DNA methylation and gene expression profiling. The DNA methylation and gene expression data showed that PDX models always cluster together with their respective brain tumor reference samples and in most cases very close to their matching primary tumor. Tumor subtype-specific oncogenic lesions could be detected by both sequencing technologies. By comparing PDX models to their primary tumor, we showed that they retain the molecular subtype, mutations and copy number alterations. Only in rare cases we observed additional aberrations in PDX models such as chromothriptic events in one MB (Group 3) or chromosome 1q gain in one EPN (posterior fossa subtype A). However, these additional aberrations are typical for this tumor subtype and were therefore most likely already present in the primary lesion at a subclonal level. Analysis of our entire cohort identified a overrepresentation of the most aggressive tumor subtypes, but also subtypes which have not been available for preclinical testing before due to lack of genetically engineered mouse models or suitable cell lines, such as Group 4 MBs. Our molecular characterizations of PDX models provide an unprecedented resource to study tumor biology and pave the way for improving treatment strategies of malignant pediatric brain tumors. Citation Format: Sebastian Brabetz, Huriye Seker-Cin, Susanne N. Gröbner, Norman L. Mack, Volker Hovestadt, David T. W. Jones, Till Milde, Madison T. Wise, Jessica M. Rusert, Kyle Pedro, Karina Bloom, Xiao-Nan Li, Robert J. Wechsler-Reya, James M. Olson, Stefan M. Pfister, Marcel Kool. Molecular characterization of patient-derived xenograft models of pediatric brain tumors. [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 A07.
- Published
- 2016
23. Abstract B37: Chemi-genomic analysis of patient-derived xenografts to identify personalized therapies for medulloblastoma
- Author
-
Xiao-Nan Li, John R. Crawford, Marcel Kool, Yoko T. Udaka, Till Milde, Michael J. Levy, Lourdes A Esparza, James M. Olson, Simone Schubert, Sebastian Brabetz, Jessica M. Rusert, Mari Kogiso, Huriye Seker-Cin, Robert J. Wechsler-Reya, Lin Qi, Stefan M. Pfister, Alexandra Garancher, and Yoon Jae Cho
- Subjects
Medulloblastoma ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Cancer ,Disease ,medicine.disease ,Bioinformatics ,Gene expression profiling ,Regimen ,Internal medicine ,DNA methylation ,Medicine ,business ,Exome ,Epigenomics - Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. Even with an intensive regimen of surgery, radiation and chemotherapy, one-third of patients still die from their disease. Moreover, survivors suffer devastating side effects including cognitive deficits, endocrine disorders and an increased incidence of secondary cancers later in life. Thus, more effective and less toxic therapies are desperately needed. Recent genomic analyses have identified 4 major subgroups of MB—WNT, SHH, Group 3 and Group 4—that differ in terms of mutations, gene expression profiles and patient outcomes. Despite this heterogeneity, all MB patients currently receive the same therapy. To identify novel therapies for each subgroup of MB, we have assembled a diverse panel of patient-derived xenograft (PDX) lines. These lines, established by orthotopic transplantation of tumor cells obtained from surgery, recapitulate the properties of patients' tumors more accurately than cultured cell lines. We are using these PDX lines to screen small molecule libraries and identify compounds that can inhibit tumor growth and survival. To date we have completed screening of 18 lines, including 10 representing Group 3 MB, the most aggressive and lethal form of the disease. Among the ~7800 compounds tested, we have found 20 that are effective against the majority of Group 3 PDX lines. Ongoing studies are focused on validating the activity of these compounds against additional Group 3 lines and moving the most promising ones forward into in vivo efficacy studies. Similar approaches will be pursued for each of the other subgroups of MB. Drug response data will also be compared with genomic and epigenomic data (whole exome and low coverage whole genome DNA sequencing, DNA methylation analysis, and gene expression profiling) to identify biomarkers of drug responsiveness and key pathways that may be exploited for therapy. Based on these studies, we hope to move away from a one-size-fits-all approach, and begin to treat each patient with therapies that are likely to be effective against their tumor. Citation Format: Jessica M. Rusert, Alexandra Garancher, Yoko T. Udaka, Sebastian Brabetz, Lourdes A. Esparza, Huriye Seker-Cin, Lin Qi, Mari Kogiso, Simone Schubert, Till Milde, Yoon-Jae Cho, Xiao-Nan Li, James M. Olson, John R. Crawford, Michael L. Levy, Marcel Kool, Stefan M. Pfister, Robert J. Wechsler-Reya. Chemi-genomic analysis of patient-derived xenografts to identify personalized therapies for medulloblastoma. [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 B37.
- Published
- 2016
24. PCM-16MOLECULAR CHARACTERIZATION OF ORTHOTOPIC PATIENT-DERIVED XENOGRAFT MODELS OF PEDIATRIC BRAIN TUMORS
- Author
-
Volker Hovestadt, Kyle Pedro, David T.W. Jones, Xiao-Nan Li, Till Milde, Marcel Kool, Sebastian Brabetz, Susanne N. Groebner, Olaf Witt, Madison T. Wise, Robert J. Wechsler-Reya, Karina Bloom, Norman Mack, Florian Selt, Sarah Leary, Stefan M. Pfister, Jessica M. Rusert, Huriye Seker-Cin, and James M. Olson
- Subjects
Medulloblastoma ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Cancer ,medicine.disease ,Primary tumor ,Pre-clinical development ,Abstracts ,Pediatric brain ,Internal medicine ,DNA methylation ,medicine ,Biomarker (medicine) ,Neurology (clinical) ,business ,Tumor xenograft - Abstract
PCM-16. MOLECULAR CHARACTERIZATION OF ORTHOTOPIC PATIENT-DERIVED XENOGRAFT MODELS OF PEDIATRIC BRAIN TUMORS Sebastian Brabetz1, Susanne N. Groebner1, Huriye Seker-Cin1, Norman L. Mack1, Volker Hovestadt1, David T. W. Jones1, Florian Selt1,2, Till Milde1,2, Madison T. Wise3, Jessica M.Rusert4, Kyle Pedro3, Karina Bloom3, Olaf Witt1,2, Sarah E. Leary3, Xiao-Nan Li5, Robert J. Wechsler-Reya4, James M. Olson3, Stefan M. Pfister1,2, and Marcel Kool1; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany; Center for Individualized Pediatric Oncology (ZIPO) and Pediatric Brain Tumors, Department of Pediatric Oncology, University Hospital and National Center for Tumor Diseases (NCT), Heidelberg, Germany; Fred Hutchinson Cancer Research Center and Seattle Children’s Hospital, Seattle, WA, USA; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA; Baylor College of Medicine, Houston, TX, USA Orthotopic patient-derived xenograft (PDX) models are an excellent platform for biomarker and preclinical drug development. Prior to drug selection and testing, extensive molecular characterization is needed to precisely determine the distinct molecular subgroup and constellation of genetic alterations for each PDX model, and thus identify its targetable oncogenic drivers. In an international effort we have characterized a large repertoire of PDX models reflecting many different molecular subtypes of pediatric brain tumors and assessed inter-tumoral heterogeneity within these subtypes. Thus far, we have collected and characterized 70 established PDX models from 6 ATRTs, 8 ependymomas, 16 high-grade gliomas, 38 medulloblastomas, and 2 CNS-PNETs. All PDX models and matching primary tumors (if available) have been analyzed by whole-exome and low-coverage wholegenome sequencing, as well as DNA methylation and gene expression profiling.PDXmodels always retain theirmolecular subtypeand in thevastmajority of cases also the mutations and copy number alterations when compared to their primary tumors. Only in rare cases do we observe additional aberrations, which most likely represent outgrowths of subclones from the primary tumor. Analysis of our entire cohort identified an overrepresentation of the most aggressive tumor subtypes, but also subtypes which have not been available for preclinical testing before due to lack of genetically engineered mouse models or suitable cell lines, such as Group 4 medulloblastoma. Our molecular characterization of PDX models will provide an unprecedented resource to study tumorbiologyandpave theway for improving treatment strategies forchildren with malignant brain tumors. Neuro-Oncology 18:iii139–iii144, 2016. doi:10.1093/neuonc/now080.16 #The Author(s) 2016. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.
- Published
- 2016
25. Genome Sequencing of SHH Medulloblastoma Predicts Genotype-Related Response to Smoothened Inhibition
- Author
-
Olaf Witt, Sebastian Stark, Volker Hovestadt, Rosario M. Piro, Arie Perry, Wolfram Scheurlen, Natalie Jäger, Michael D. Taylor, Tobias Rausch, Robert J. Wechsler-Reya, Cynthia Cowdrey, Andrey Korshunov, V. Peter Collins, Jan Koster, David Shih, Marcel Kool, Ulrich Schüller, Linda Linke, Silvia Hofer, Nada Jabado, Till Milde, Trevor J. Pugh, Andreas von Deimling, Sebastian Bender, Scott L. Pomeroy, Hendrik Witt, Dominik Sturm, Guillaume Bergthold, Christof von Kalle, Mark W. Kieran, Jüri Reimand, Rogier Versteeg, Sabine Schmidt, Marc Zapatka, Marc Remke, Rameen Beroukhim, Andrea Wittmann, Pascal Johann, Huriye Seker-Cin, Andreas Unterberg, Rainer König, Laura Sieber, Benedikt Brors, Andreas E. Kulozik, Gary D. Bader, François Doz, Richard Volckmann, Olivier Delattre, Christin Schmidt, Stephan Wolf, John R. Crawford, Gerald A. Grant, Shirley L. Markant, Paul A. Northcott, Martin G. McCabe, Peter Lichter, David T.W. Jones, Martin Hasselblatt, Sonja Hutter, Ursula D. Weber, Cynthia C. Bartholomae, Peter van Sluis, L. Adriana Esparza, Roland Eils, Guido Reifenberger, Marina Ryzhova, Christel Herold-Mende, Stefan M. Pfister, Yoon Jae Cho, Elke Pfaff, Sydney Croul, Jörg Felsberg, Chris Lawerenz, Franck Bourdeaut, Jan O. Korbel, Gelareh Zadeh, Other departments, Oncogenomics, and CCA -Cancer Center Amsterdam
- Subjects
Male ,Cancer Research ,Pyridines ,Mice, SCID ,medicine.disease_cause ,Germline ,Receptors, G-Protein-Coupled ,DEAD-box RNA Helicases ,Mice ,Phosphatidylinositol 3-Kinases ,Mice, Inbred NOD ,Child ,Promoter Regions, Genetic ,Telomerase ,Oncogene Proteins ,Genetics ,N-Myc Proto-Oncogene Protein ,Mutation ,High-Throughput Nucleotide Sequencing ,Nuclear Proteins ,Smoothened Receptor ,Patched-1 Receptor ,Biphenyl compound ,Oncology ,Child, Preschool ,embryonic structures ,Female ,Signal Transduction ,Adult ,Patched Receptors ,animal structures ,Adolescent ,DNA Copy Number Variations ,Molecular Sequence Data ,Kruppel-Like Transcription Factors ,Receptors, Cell Surface ,Zinc Finger Protein Gli2 ,Biology ,Article ,Young Adult ,GLI2 ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Cerebellar Neoplasms ,Medulloblastoma ,Base Sequence ,Gene Expression Profiling ,Biphenyl Compounds ,Infant ,Cell Biology ,medicine.disease ,Repressor Proteins ,Have You Seen? ,PTCH1 ,Drug Resistance, Neoplasm ,Cancer research ,Tumor Suppressor Protein p53 ,Smoothened ,Proto-Oncogene Proteins c-akt ,Neoplasm Transplantation - Abstract
SummarySmoothened (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 (n = 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.
- Full Text
- View/download PDF
26. Reduced H3K27me3 and DNA Hypomethylation Are Major Drivers of Gene Expression in K27M Mutant Pediatric High-Grade Gliomas
- Author
-
S. Chen, Andreas von Deimling, Kristian Helin, Jonas W. Højfeldt, Simone Schubert, Christoph Plass, David T.W. Jones, Dominik Sturm, Olaf Witt, V. Peter Collins, Michelle Monje, Jochen Meyer, Jan Gronych, Till Milde, Nada Jabado, David Castel, Marina Ryzhova, Stefan M. Pfister, Anitha Ponnuswami, Peter Lichter, Yoon Jae Cho, Sebastian Bender, Martin U. Schuhmann, Paul A. Northcott, Marc Zapatka, Chris Jones, Yujie Tang, Huriye Seker-Cin, Wei Wang, Pascal-David Johann, Martin Ebinger, Andrey Korshunov, Camelia M. Monoranu, Anders Lindroth, Stéphanie Puget, Marcel Kool, Volker Hovestadt, Bernhard Radlwimmer, Nathalene Truffaux, Jacques Grill, and Sebastian Stark
- Subjects
Cancer Research ,Transcription, Genetic ,Molecular Sequence Data ,Mutant ,Bisulfite sequencing ,Mutation, Missense ,macromolecular substances ,Methylation ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Brain Stem Neoplasms ,Humans ,Child ,Genes, Dominant ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,biology ,Brain Neoplasms ,EZH2 ,Polycomb Repressive Complex 2 ,Cell Biology ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Molecular biology ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Histone ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,biology.protein ,Glioblastoma ,Protein Processing, Post-Translational ,Chromatin immunoprecipitation ,Protein Binding ,Transcription Factors ,DNA hypomethylation - Abstract
SummaryTwo recurrent mutations, K27M and G34R/V, within histone variant H3.3 were recently identified in ∼50% of pHGGs. Both mutations define clinically and biologically distinct subgroups of pHGGs. Here, we provide further insight about the dominant-negative effect of K27M mutant H3.3, leading to a global reduction of the repressive histone mark H3K27me3. We demonstrate that this is caused by aberrant recruitment of the PRC2 complex to K27M mutant H3.3 and enzymatic inhibition of the H3K27me3-establishing methyltransferase EZH2. By performing chromatin immunoprecipitation followed by next-generation sequencing and whole-genome bisulfite sequencing in primary pHGGs, we show that reduced H3K27me3 levels and DNA hypomethylation act in concert to activate gene expression in K27M mutant pHGGs.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.